The Journal of Physiology - medicine.mcgill.ca

7
J Physiol 599.2 (2021) pp 389–395 389 The Journal of Physiology EDITORIAL Neurotransmitter-gated ion channels, still front and centre stage Derek Bowie Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, H3G 1Y6, Canada Email: [email protected] Edited by: Kim Barrett Neurotransmitter-gated ion channels are critical for the normal hardwiring of neuro- nal circuits but also fine tune synaptic strength during periods of sustained patterned activity and altered homeostasis. Ionotropic glutamate receptors (iGluRs) and nicotinic acetylcholine receptors trans- mit the vast majority of fast excitatory signalling in the developing and adult CNS whereas almost all inhibitory neuro- transmission is mediated by GABA A and glycine receptors. The study of neurotransmitter-gated ion channels has undergone unprecedented advances in recent years with the convergence of several scientific disciplines on this important research topic. Structural biology has emerged as a leading approach to under- Correction made on 12 January 2021, after first online publication: The article has been updated to include the Kamalova, A & Nakagawa T. (2021) reference. standing ion channel function and drug action, and recent advances in genetics have permitted different families of neurotransmitter-gated ion channels to be assigned to distinct roles in neuronal health and disease. To explore this advanced area of physio- logy, The Journal of Physiology sponsored back-to-back 2-day conferences on the downtown campus of McGill University in Montréal during the summer of 2019. The 2019 Ionotropic Glutamate Receptor Retreat or iGluRetreat was organized by Dr David Stellwagen (McGill) and myself and follows on a tradition since 2013 of holding the conference at a different university campus in North America (https: //www.ion-channelconferences.org/). The iGluRetreat brought together 25 speakers whose work is at the forefront of this rapidly developing field of study (Fig. 1). The 2-day International Society of Neuro- chemistry (ISN) satellite conference on ‘Neurotransmitter-gated ion-channels in health and disease’ was organized by Dr Katherine Roche (NIH) and myself and brought together a unique group of leading researchers from across the globe to showcase the most recent advances in the study of excitatory and inhibitory receptor synapses (Fig. 2). The first day of the satellite conference focused on the complexity of the ion channel signalling complex and how structural studies have brought new advances in drug design and therapy. The conference’s second day examined the emerging roles of neurotransmitter-gated ion channels in plasticity mechanisms and their involvement in neuronal circuits and CNS disease. This issue of The Journal of Physiology brings together eight timely review articles that capture some of the ideas and discussions that arose during these 4 days of exciting and intense scientific discourse. Dr Lonnie Wollmuth and colleagues from Stony Brook University discussed their recent work on disease-causing mutations that disrupt the functional properties of NMDA-type ionotropic glutamate receptors (iGluRs) (Amin et al. 2021). The Wollmuth lab has spent the past two decades mapping out the structure–function properties of wild-type NMDA- and AMPA-type iGluRs. In particular, they have pinpointed the unique roles fulfilled by different subunits and evolutionarily conserved domains, such as DRPEER (Watanabe et al. 2002) and SYTANLAAF (Jatzke et al. 2003), in channel gating and ion permeation as well as identifying the role of the transmembrane region in channel tetramerization (Gan et al. 2015). This work has provided a useful framework in which to understand the impact of disease-causing mutations on NMDARs which, in some cases, are able to curtail the time course of channel activation whilst diminishing Ca 2+ transport through the pore (Amin et al. 2018). Given the key role of NMDARs in neurodevelopment and plasticity (Constantine-Paton et al. 1990; Figure 1. Photograph of the speakers and attendees at the 2019 iGluRetreat conference that was organized by Drs David Stellwagen (McGill) and Derek Bowie (McGill) on the downtown campus of McGill University © 2020 The Authors. The Journal of Physiology © 2020 The Physiological Society DOI: 10.1113/JP280800

Transcript of The Journal of Physiology - medicine.mcgill.ca

J Physiol 599.2 (2021) pp 389–395 389

TheJourn

alofPh

ysiology

ED ITOR IAL

Neurotransmitter-gated ionchannels, still front and centrestage

Derek BowieDepartment of Pharmacology andTherapeutics, McGill University, Montréal,Québec, H3G 1Y6, CanadaEmail: [email protected] by: Kim Barrett

Neurotransmitter-gated ion channels arecritical for the normal hardwiring of neuro-nal circuits but also fine tune synapticstrength during periods of sustainedpatterned activity and altered homeostasis.Ionotropic glutamate receptors (iGluRs)and nicotinic acetylcholine receptors trans-mit the vast majority of fast excitatorysignalling in the developing and adultCNS whereas almost all inhibitory neuro-transmission is mediated by GABAAand glycine receptors. The study ofneurotransmitter-gated ion channelshas undergone unprecedented advances inrecent years with the convergence of severalscientific disciplines on this importantresearch topic. Structural biology hasemerged as a leading approach to under-

Correction made on 12 January 2021, afterfirst online publication: The article hasbeen updated to include the Kamalova, A &Nakagawa T. (2021) reference.

standing ion channel function and drugaction, and recent advances in geneticshave permitted different families ofneurotransmitter-gated ion channels tobe assigned to distinct roles in neuronalhealth and disease.To explore this advanced area of physio-logy, The Journal of Physiology sponsoredback-to-back 2-day conferences on thedowntown campus of McGill Universityin Montréal during the summer of 2019.The 2019 Ionotropic Glutamate ReceptorRetreat or iGluRetreat was organized byDr David Stellwagen (McGill) and myselfand follows on a tradition since 2013of holding the conference at a differentuniversity campus in North America (https://www.ion-channelconferences.org/). TheiGluRetreat brought together 25 speakerswhose work is at the forefront of thisrapidly developing field of study (Fig. 1).The 2-day International Society of Neuro-chemistry (ISN) satellite conference on‘Neurotransmitter-gated ion-channelsin health and disease’ was organized byDr Katherine Roche (NIH) and myselfand brought together a unique group ofleading researchers from across the globe toshowcase the most recent advances in thestudy of excitatory and inhibitory receptorsynapses (Fig. 2). The first day of the satelliteconference focused on the complexity ofthe ion channel signalling complex andhow structural studies have brought newadvances in drug design and therapy. Theconference’s second day examined the

emerging roles of neurotransmitter-gatedion channels in plasticity mechanisms andtheir involvement in neuronal circuits andCNS disease. This issue of The Journal ofPhysiology brings together eight timelyreview articles that capture some of theideas and discussions that arose duringthese 4 days of exciting and intense scientificdiscourse.Dr Lonnie Wollmuth and colleaguesfrom Stony Brook University discussedtheir recent work on disease-causingmutations that disrupt the functionalproperties of NMDA-type ionotropicglutamate receptors (iGluRs) (Aminet al. 2021). The Wollmuth lab has spentthe past two decades mapping out thestructure–function properties of wild-typeNMDA- and AMPA-type iGluRs. Inparticular, they have pinpointed the uniqueroles fulfilled by different subunits andevolutionarily conserved domains, suchas DRPEER (Watanabe et al. 2002) andSYTANLAAF (Jatzke et al. 2003), inchannel gating and ion permeation as wellas identifying the role of the transmembraneregion in channel tetramerization (Ganet al. 2015). This work has provided a usefulframework in which to understand theimpact of disease-causing mutations onNMDARs which, in some cases, are able tocurtail the time course of channel activationwhilst diminishing Ca2+ transport throughthe pore (Amin et al. 2018). Given the keyrole of NMDARs in neurodevelopment andplasticity (Constantine-Paton et al. 1990;

Figure 1. Photograph of the speakers and attendees at the 2019 iGluRetreat conference that wasorganized by Drs David Stellwagen (McGill) and Derek Bowie (McGill) on the downtown campus ofMcGill University

© 2020 The Authors. The Journal of Physiology © 2020 The Physiological Society DOI: 10.1113/JP280800

390 Editorial J Physiol 599.2

Nicoll, 2017), the negative impact of thesemutations can be readily appreciated. Intheir review, Amin and colleagues arguethat the study of disease-causing mutationsnot only provides insight into howdefects inNMDAR function and assembly contributeto neurological disease, but also expandsour understanding of the inner workings ofNMDARs. The authors also acknowledgethat many disease mutations are foundat sites in the NMDAR that have yet tobe tied to a particular functional role. Anongoing challenge is to understand howboth gain- and loss-of-function mutationsin the NMDAR (Myers et al. 2019) can leadto neurological disease with many of thesame cardinal features.Dr David MacLean, his graduate studentMatthew Rook from Rochester University,USA and Dr Maria Musgaard fromthe University of Ottawa, Canada pre-sented work on a collaborative project ofacid-sensing ion channels (ASICs), whichhave been the focus of intense researchactivity in recent years (Rook et al. 2021).ASICs are distinguished from most otherligand-gated ion channels by their trimericsubunit stoichiometry which differs fromthe more conventional tetrameric orpentameric stoichiometry associated withiGluRs and GABAA receptors, respectively,at central synapses. First identified throughearly forays into using the concentrationclamp technique, ASICs respond to rapidmillisecond changes in extracellular pHby evoking a rapidly decaying, cationic

membrane conductance in freshly iso-lated sensory neurons (Krishtal, 2015).Since then, ASICs have been shown tobe expressed throughout the body in thecentral and peripheral nervous systemswhere they have been linked to learningand memory, pain sensation, fear andanxiety, substance abuse, and cell death.Perhaps most surprisingly, ASICs are ableto respond to rapid and brief changesin extracellular acidification (MacLean &Jayaraman, 2016)much like the rapid gatingproperties of AMPARs in response to l-Gluat auditory synapses (Joshi & Wang, 2002).Their importance to cell physiology andimplication in numerous disease states hasplaced the spotlight on ASICs and a need tobetter understand their structure–functioncharacteristics, which is the focus of thereview. An immediate challenge is tryingto understand how proton binding elicitsconformational changes in the ASICchannel structure to access the open anddesensitized states. Structural studies haveshown that each ASIC subunit consistsof a large extracellular domain, two trans-membrane helices, and short intracellular Nand C termini (Baconguis et al. 2014). Theextracellular domain has been proposed tocontain the proton binding site, although, asnoted by the authors, its role in the processof channel activation and desensitizationis still debated. Aided by recent full-lengthstructures of ASIC channels (e.g. Yoderet al. 2018), Rook and colleagues haverecently developed a working model of how

ASIC1 channels enter into and out of thedesensitized state(s) using a combination offast perfusion electrophysiology, moleculardynamic simulations and crosslinkingexperiments (Rook et al. 2020). Their workunexpectedly highlights the importance ofthe β11-12 linker connecting the proposedextracellular proton binding pocket withtransmembrane helices as being a keyregulator of channel desensitization.Despite all these advances, the authorsreflect on the challenges in studying gatingevents promoted by proton binding butoffer some insights for the way forward forthis important area of ion channel biology.Dr Anthony Koleske and graduate student

Juliana Shaw from Yale University alsodiscussed the role of NMDARs andquestioned exactly how they couple tothe cell’s cytoskeleton (Shaw & Koleske,2021). The idea that NMDARs are linked tothe cytoskeleton was first appreciated fromexperiments showing that the inactivationof NMDARs, which occurs due to thetransport of Ca2+ through the channelpore, could be blocked by agents that pre-vent actin depolymerization (Rosenmund& Westbrook, 1993). This finding linkingchannel activity to the cell’s architecturalproteins was further supported by workshowing that NMDARs were, in fact,mechanosensitive (Paoletti & Ascher,1994). Direct biochemical evidence wasestablished by using latrunculin-A toinduce actin depolymerization, whichdispersed NMDARs from synaptic sites

Figure 2. The speakers and attendees at the 2019 ISN satellite conference on ‘Neurotransmitter-gatedion-channels in health and disease’ that was organized by Drs Katherine Roche (NIH) and Derek Bowie(McGill) on the downtown campus of McGill University

© 2020 The Authors. The Journal of Physiology © 2020 The Physiological Society

J Physiol 599.2 Editorial 391

(Allison et al. 1998) through its inter-action with α-actinin-2, which binds to thecytoplasmic tails of GluN1 and GluN2BNMDAR subunits (Wyszynski et al. 1997).In their review article, Shaw and Koleskedescribe the mechanisms by which actinregulates the functional properties ofdifferent voltage- and ligand-gated channelfamilies with an emphasis on the regulationof NMDARs (Shaw & Koleske, 2021).Previous work from the Koleske lab hasestablished the importance of dynamicactin networks in regulating NMDARfunction at central synapses in Noonansyndrome (Levy et al. 2018), a multi-systemdisorder characterized by developmentaldelay and learning difficulties (Robertset al. 2013). Noonan syndrome is causedby hyperactivating mutations in the SHP2tyrosine phosphatase which disrupt thephosphorylated tyrosine binding site onthe GluN2B cytoplasmic tail for the Nck2adaptor protein. This phosphorylationevent is detrimental in Noonan syndromesince it disrupts the partnership of Nck2with N-WASp, which activate actin branchnucleation via the Arp2/3 complex(Levy et al. 2018). The untethering ofNck2 selectively impairs GluN2B NMDARfunctionality at Schaffer collateral–CA1neuron excitatory synapses inducingdeficits in long-term potentiation (Levyet al. 2018). The authors conclude byspeculating on why NMDARs may coupleto the cytoskeleton via actin, suggestingthat the interaction may be importantin the gating properties of NMDARs.Since alterations in the intrinsic disorderof the cytoplasmic tail of NMDARs, byphosphorylation for example, impactchannel functionality (Choi et al. 2011,2013), it is clear that more work is neededto better understand how these molecularevents impact neuronal communicationand ultimately contribute to CNS disease.The review article by Drs KatherineRoche and Sehoon Won from theNational Institutes of NeurologicalDisorders and Stroke in Bethesda, MDhighlights the important balancing roleof striatal-enriched tyrosine phosphatase61 (STEP61) at glutamatergic synapses(Won & Roche, 2021). The strength ofsignalling at glutamatergic synapses isstrongly regulated by phosphorylationevents driven by the actions of a numberof serine and tyrosine kinases (Chen &Roche, 2007). The NMDAR, in particular,is targeted by a family of src tyrosinekinases, which includes Src and Fyn, which

upregulate channel activity and contributeto long-term potentiation in the CA1 regionof the hippocampus (Salter & Kalia, 2004).Tyrosine phosphatases, such as STEP, arethus important in regulating the balanceat central synapses. The most abundantisoform in the brain is STEP61, whichhas garnered a lot of attention due to itsimplicated roles in neurological disease(Goebel-Goody et al. 2012). STEP61 hasa number of synaptic targets includingtyrosine kinase Fyn and NMDA- andAMPA-type ionotropic glutamate receptors(Won et al. 2016, 2019). Fyn and Srckinases phosphorylate NMDARs on severaltyrosine residues in the cytoplasmic tail ofthe GluN2A and GluN2B subunits. Amongthese tyrosine residues, Y1472 is adjacentto the PSD-95 binding site, which whenphosphorylated inhibits clathrin-mediatedendocytosis of GluN2B-containingNMDARs. STEP61 targets GluN2BY1472 for dephosphorylation and inter-nalization. Although AMPARs have shortercytoplasmic tails, phosphorylation anddephosphorylation events by numerouskinases and phosphatases similarlyregulate receptor trafficking, endocytosisand synaptic plasticity. For example,phosphorylation of Y876 in the cyto-plasmic tail of the GluA2 subunit disruptsGRIP1/2 binding leaving PICK1 bound,which promotes internalization of AMPARs(Hayashi & Huganir, 2004). STEP61 wasrecently shown by mass spectrometry tobind directly to the cytoplasmic tails ofthe GluA2 and A3 subunits but not theGluA1 subunit. Accordingly, expressionof GluA2 and GluA3 subunits at synapsesis increased in STEP-KO mouse brainwhereas STEP61 overexpression reduces thesynaptic expression and responsiveness ofAMPARs (Won et al. 2019). The authorsconclude by reviewing the evidence linkingan upregulation in the expression of STEP61in cognitive impairment associated withageing reported in several rodent, rhesusmonkey and human studies suggestingthat STEP61 inhibitors, such as TC-2153(Xu et al. 2014), may be a valuable form oftreatment.The article by Dr Teru Nakagawa andgraduate student Aichurok Kamalovafrom Vanderbilt University provides atimely review of the multifaceted natureof the AMPA receptor–auxiliary subunitcomplex (Kamalova & Nakagawa, 2021).An early cryo-EM study gave us our firstglimpse of the complex nature by whichtransmembrane AMPA receptor auxiliary

proteins (TARPs) affect receptor functionby bracing the transmembrane domainsof the ion channel pore region (Nakagawaet al. 2005). Since then, several labs haveprovided an even greater picture of theseinteractions by studying full-length homo-and heteromeric structures of the AMPARin complex with γ 2 and γ 8 TARPs, germcell-specific gene 1-like protein (GSG1L;Greger et al. 2017; Chen & Gouaux, 2019;Twomey et al. 2019) and more recently,in association with cornichon-3 (CNIH3;Nakagawa, 2019). It is becoming clear thatnative AMPARs co-assemble with a numberof different auxiliary subunit families,which has made it imperative to betterunderstand how these associations affectbrain function. The review focuses on theemerging evidence suggesting that differentfamilies of auxiliary subunits interact withAMPARs in different ways, explainingtheir distinct effects on ion channel gatingand ion-permeation. The slower gatingbehaviour of the AMPAR–TARP complexcan be explained by TARP interactionswith the KGK site (Dawe et al. 2016) of theligand-binding domain of AMPARs as wellas residues located in the transmembraneregions (Ben-Yaacov et al. 2017; Hawkenet al. 2017). GSG1L has a similar overallarchitecture to TARPs and it is thoughtto interact with AMPARs in a similarmanner (Twomey et al. 2017), yet it lacksan extracellular helix found in TARPsand possesses a larger β1–β2 loop. Theauthors argue that these distinctions explainwhy TARPs preferentially slow entry intodesensitization whereas GSG1L slowsrecovery, although the exact details remainto be resolved. The recent structure ofthe AMPAR–CNIH3 complex reveals thatcornichons contain four transmembraneregions, not three as originally proposed,and lack the extensive extracellular domainsthat are so critical for TARP and GSG1Lfunction (Nakagawa, 2019). Consequently,the modulatory effects of CNIHs thatslow AMPAR channel gating and relievepolyamine channel block are presumablymediated by residues in the transmembranedomain. Details are still emerging, but itmay suggest that the structural events thatregulate channel gating and ion permeationare perhaps functionally coupled. What isclear is that the coming years will provide afuller understanding of the structural andfunctional nature of the AMPAR–auxiliarysubunit complex and how it shapes thephysiology of neuronal circuits and disruptsthem in neurological disease.

© 2020 The Authors. The Journal of Physiology © 2020 The Physiological Society

392 Editorial J Physiol 599.2

Drs Jakob von Engelhardt and EricJacobi from the Johannes GutenbergUniversity Mainz, Germany also discussthe importance of the AMPAR–auxiliarysubunit complex but focus on its criticalrole in shaping the strength and fidelityof synaptic communication as well asits impact on the signalling capacity ofneuronal circuits (Jacobi & von Engelhardt,2021). AMPARs co-assemble with threemajor auxiliary subunit families, includingmembers of the claudin family (TARPsγ 2-5 and γ 7-8 as well as GSG1L), theCKAMP family (CKAMP39, -44, -52 and-59) and the cornichons (CNIH2 and -3),most of which facilitate the traffickingand synaptic localization of AMPARs inneurons. Knockout mice or overexpressionstudies reveal that auxiliary proteinsalso impact the gating and permeationproperties of native AMPARs. The TARP,CKAMP and CNIH families are, however,differentially expressed in the mammalianbrain (Schwenk et al. 2014) suggesting thatsignalling in different brain regions maybe finely tuned by their expression pattern.For example, TARP γ 8 and CNIH2 are pre-dominantly expressed in the hippocampus,cortex and striatum (Schwenk et al. 2014),with CKAMP44 and GSG1L primarilyexpressed in glutamatergic neurons of thecortex (Zeisel et al. 2018). The authorsdiscuss how most auxiliary subunitsincrease the strength of glutamatergicsynapses through a number of mechanismsthat include increasing channel densityand augmenting open channel probabilityand/or its unitary conductance. Theexception to this is GSG1L, which has beenshown to decrease channel density andunitary conductance in the cerebellumand hippocampus (McGee et al. 2015;Gu et al. 2016). The von Engelhardt labhas provided compelling evidence tosupport the important role of CKAMP44 ingoverning short-term plasticity of granulecells in the dentate gyrus (von Engelhardtet al. 2010; Khodosevich et al. 2014)and relay neurons in the dorsal lateralgeniculate nucleus (Chen et al. 2018), withother groups similarly establishing theimportance of CKAMP52 in CA1 neuronsof the hippocampus andPurkinje cells of thecerebellum (Klaassen et al. 2016; Peter et al.2020). The review concludes by discussingthe extensive literature on the importantrole fulfilled by TARP γ 2 and γ 8 inlong-term potentiation in the hippocampusand the paucity of information on howAMPAR auxiliary subunits affect homeo-

static plasticity. The greater understandingof the AMPAR–auxiliary subunit complexand the role it plays in neuronal circuits andultimately in human behaviour and diseasemay lead to breakthrough strategies of drugdesign that target the complex and notsimply the channel pore forming subunits(Rosenbaum et al. 2020).Drs Melanie Woodin and Jessica Presseyfrom the University of Toronto reviewthe unexpectedly diverse mechanismsby which native kainate-type (KARs)ionotropic glutamate receptors signal atdifferent synapses in the hippocampus(Pressey & Woodin, 2021). Unlike thecanonical view of AMPARs and NMDARs,it has been much more difficult to assigngeneral guiding principles to how KARssignal at central synapses. A major break-through was the appreciation that theselective, non-competitive block by severalGYKI compounds of the much largerAMPAR response revealed the smalleramplitude, slower decaying responses ofsynaptic KARs (Paternain et al. 1995).With this research tool at hand and withgenetic knockout strategies, it has beenpossible to pinpoint KARs to both thepre- and postsynaptic sides of excitatoryglutamatergic and inhibitory GABAergicsynapses (Contractor et al. 2011). Asoutlined by the authors, a significantsurprise has been the understanding thatKARs not only signal via an ionotropicpathway but also regulate the activity ofvoltage-gated potassium (Melyan et al.2002) and calcium (Rodriguez-Moreno &Lerma, 1998) channels via a metabotropicG-protein coupled pathway. The Woodinlab has added further complexity to theKAR signalling profile by linking theiractivity to that of the neuron-specificK+–Cl− co-transporter, KCC2 (Pre-ssey et al. 2017). Since KCC2 sets thechloride equilibrium potential in matureneurons (Kaila et al. 2014), it ultimatelydetermines the strength of GABAergicneurotransmission. The indirect regulatoryeffect of KARs on KCC2 is mediated bythe KAR auxiliary protein, Neto2, whichnot only binds and regulates the gatingand permeation properties of KARs butalso binds directly to KCC2 (Ivakineet al. 2013; Mahadevan et al. 2014). Theimportance of this interaction can beappreciated from the behavioural analysisof Neto2 KO mice, which have a reducedthreshold for seizures through the lossof KCC2 and diminished GABAergicneurotransmission (Mahadevan et al.

2015). Whether dysfunction of the newlydiscovered KAR–Neto2–KCC2 complexplays a role in CNS disease awaits to beuncovered.Last but certainly not least, Drs Stephen

Glasgow, Ed Ruthazer and Tim Kennedyfrom the Montreal Neurological Institute ofMcGill University provide a comprehensivereview of their work highlighting thesignificant role of netrin-1 in synapticplasticity and memory consolidation inthe adult brain (Glasgow et al. 2021).Netrins were originally identified aschemoattractant guidance cues duringembryogenesis that direct cell and axonmigration (Kennedy & Tessier-Lavigne,1995). However, netrins continue to beexpressed by neurons into adulthoodopening the question of whether theyadopt different roles in mature neuronalcircuits. The authors open their reviewby noting that many of the signallingcascades that are activated during changesin synaptic plasticity in the adult brainare similar to the cascade of signallingevents triggered by chemotropic axonguidance cues during development. Thefirst clue to understanding this puzzle wasthe observation that genetic deletion ofthe canonical receptor for netrin-1, thesingle-pass transmembrane protein calleddeleted in colorectal cancer (DCC) resultsin the loss of hippocampal long-termpotentiation and impairment of spatialand recognition memory (Horn et al.2013). Their next findings revealed thatthe ligand, Netrin-1, promotes excitatorysynaptogenesis between cortical neuronsby initiating synapse assembly and contactpoints, which increases the efficacy ofexcitatory synaptic transmission (Goldmanet al. 2013). More details of this mechanismemerged with the finding that membranedepolarization and NMDAR activation atcentral synapses promotes the secretionof netrin-1 from dendrites (Glasgowet al. 2018). In agreement with theirearlier findings with DCC deletion, theauthors show that netrin-1 expression byhippocampal CA1 pyramidal neurons isrequired forNMDAR-dependent long-termpotentiation (Glasgow et al. 2018). In fact,the exogenous application of netrin-1 isenough to trigger the potentiation of CA1neuron glutamatergic synapses with theinsertion of GluA1-containing AMPARsmediated through a rise in cytosolic Ca2+(Glasgow et al. 2018). This surprising yetrational alignment between molecularevents in embryogenesis and the signalling

© 2020 The Authors. The Journal of Physiology © 2020 The Physiological Society

J Physiol 599.2 Editorial 393

and structural processes that give rise tolong-term potentiation in the adult brainreveals a remarkable economy in how theneurons are able to find different uses forprocesses with quite distinct purposes.Much is still to be uncovered about the roleof netrin-1 in the adult brain though morerecent work highlighting its role in spatialmemory formation (Wong et al. 2019) andthe specific pre- and postsynaptic role ofits receptor, DCC (Glasgow et al. 2020),suggest that a fuller understanding may beforthcoming.It has been more than three decades sincethe first cloning studies began to reveal themolecular identity of the neurotransmitterreceptors that populate glutamatergic andGABAergic synapses in the mammalianbrain (Schofield et al. 1987; Hollmannet al. 1989). Since then, genetic knockoutand knockin studies have helped pinpointthe distribution and synaptic roles ofiGluRs and GABAA receptors and refinean understanding of their roles in synaptictransmission and plasticity mechanisms.With many full-length homomeric andheteromeric structures of pore-formingsubunits iGluRs and GABAA receptorsreported in the last decade (Sobolevsky et al.2009; Miller & Aricescu, 2014), our nextsteps will be to understand the structuralbiology of the synapse in its entirety. Thisis needed if we are to appreciate howauxiliary subunits and signalling proteins,such as kinases and phosphatases, coupleto ion channels and regulate excitatoryand inhibitory neurotransmission inthe brain. The cell- and region-specificexpression pattern of auxiliary subunits inthe developing and adult brain (Zeisel et al.2018) also offers an opportunity for moretargeted neuropharmacology that exploitstheir unique protein–protein interactionsto develop drugs with more specificity andfewer side effects (Rosenbaum et al. 2020).Together with the strident advances insystems biology, the next decade looks setto provide for the first time a comprehensiveunderstanding of how molecular events atthe synapse and within neuronal circuitsgive rise to complex animal and humanbehaviour and go awry in neurologicaldisease.

References

Allison DW, Gelfand VI, Spector I & CraigAM (1998). Role of actin in anchoring post-synaptic receptors in cultured hippocampalneurons: differential attachment of NMDA

versus AMPA receptors. J Neurosci 18,2423–2436.

Amin JB, Leng X, Gochman A, Zhou HX &Wollmuth LP (2018). A conserved glycineharboring disease-associated mutations permitsNMDA receptor slow deactivation and highCa2+ permeability. Nat Commun 9, 3748.

Amin JB, Moody GR & Wollmuth LP(2021). From bedside-to-bench: Whatdisease-associated variants are teaching usabout the NMDA receptor. J Physiol 599,397–416.

Baconguis I, Bohlen CJ, Goehring A, JuliusD & Gouaux E (2014). X-ray structureof acid-sensing ion channel 1-snake toxincomplex reveals open state of a Na+-selectivechannel. Cell 156, 717–729.

Ben-Yaacov A, Gillor M, Haham T, Parsai A,Qneibi M & Stern-Bach Y (2017). Molecularmechanism of AMPA receptor modulation byTARP/Stargazin. Neuron 93, 1126–1137.e4.

Chen BS & Roche KW (2007). Regulationof NMDA receptors by phosphorylation.Neuropharmacology 53, 362–368.

Chen S & Gouaux E (2019). Structure andmechanism of AMPA receptor–auxiliaryprotein complexes. Curr Opin Struct Biol 54,104–111.

Chen X, Aslam M, Gollisch T, Allen K & vonEngelhardt J (2018). CKAMP44 modulatesintegration of visual inputs in the lateralgeniculate nucleus. Nat Commun 9, 261.

Choi UB, Kazi R, Stenzoski N, WollmuthLP, Uversky VN & Bowen ME (2013).Modulating the intrinsic disorder in thecytoplasmic domain alters the biologicalactivity of the N-methyl-D-aspartate-sensitiveglutamate receptor. J Biol Chem 288,22506–22515.

Choi UB, Xiao S, Wollmuth LP & Bowen ME(2011). Effect of Src kinase phosphorylationon disordered C-terminal domain ofN-methyl-D-aspartic acid (NMDA) receptorsubunit GluN2B protein. J Biol Chem 286,29904–29912.

Constantine-Paton M, Cline HT & DebskiE (1990). Patterned activity, synapticconvergence, and the NMDA receptor indeveloping visual pathways. Annu Rev Neuro-sci 13, 129–154.

Contractor A, Mulle C & Swanson GT (2011).Kainate receptors coming of age: milestonesof two decades of research. Trends Neurosci34, 154–163.

Dawe GB, Musgaard M, Aurousseau MRP,Nayeem N, Green T, Biggin PC & BowieD (2016). Distinct structural pathwayscoordinate the activation of AMPAreceptor-auxiliary subunit complexes. Neuron89, 1264–1276.

Gan Q, Salussolia CL & Wollmuth LP (2015).Assembly of AMPA receptors: mechanismsand regulation. J Physiol 593, 39–48.

Glasgow SD, Labrecque S, Beamish IV,Aufmkolk S, Gibon J, Han D, Harris SN,Dufresne P, Wiseman PW, McKinney RA,

Seguela P, De Koninck P, Ruthazer ES &Kennedy TE (2018). Activity-dependentnetrin-1 secretion drives synaptic insertionof GluA1-containing AMPA receptors in thehippocampus. Cell Rep 25, 168–182.e6.

Glasgow SD, Ruthazer ES & Kennedy TE(2021). Guiding synaptic plasticity: novelroles for netrin-1 in synaptic plasticity andmemory formation in the adult brain. JPhysiol 599, 493–505.

Glasgow SD, Wong EW, Thompson-Steckel G,Marcal N, Seguela P, Ruthazer ES & KennedyTE (2020). Pre- and post-synaptic roles forDCC in memory consolidation in the adultmouse hippocampus. Mol Brain 13, 56.

Goebel-Goody SM, Baum M, PaspalasCD, Fernandez SM, Carty NC, KurupP & Lombroso PJ (2012). Therapeuticimplications for striatal-enriched proteintyrosine phosphatase (STEP) in neuro-psychiatric disorders. Pharmacol Rev 64,65–87.

Goldman JS, Ashour MA, Magdesian MH,Tritsch NX, Harris SN, Christofi N, ChemaliR, Stern YE, Thompson-Steckel G, GrisP, Glasgow SD, Grutter P, Bouchard JF,Ruthazer ES, Stellwagen D & KennedyTE (2013). Netrin-1 promotes excitatorysynaptogenesis between cortical neurons byinitiating synapse assembly. J Neurosci 33,17278–17289.

Greger IH, Watson JF & Cull-Candy SG (2017).Structural and functional architecture ofAMPA-type glutamate receptors and theirauxiliary proteins. Neuron 94, 713–730.

Gu X, Mao X, Lussier MP, Hutchison MA,Zhou L, Hamra FK, Roche KW & LuW (2016). GSG1L suppresses AMPAreceptor-mediated synaptic transmission anduniquely modulates AMPA receptor kineticsin hippocampal neurons. Nat Commun 7,10873.

Hawken NM, Zaika EI & Nakagawa T (2017).Engineering defined membrane-embeddedelements of AMPA receptor induces opposinggating modulation by cornichon 3 andstargazin. J Physiol 595, 6517–6539.

Hayashi T & Huganir RL (2004). Tyrosinephosphorylation and regulation of the AMPAreceptor by SRC family tyrosine kinases. JNeurosci 24, 6152–6160.

Hollmann M, O’Shea-Greenfield A, RogersSW & Heinemann S (1989). Cloning byfunctional expression of a member of theglutamate receptor family. Nature 342,643–648.

Horn KE, Glasgow SD, Gobert D, Bull SJ,Luk T, Girgis J, Tremblay ME, McEachernD, Bouchard JF, Haber M, Hamel E,Krimpenfort P, Murai KK, Berns A, DoucetG, Chapman CA, Ruthazer ES & Kennedy TE(2013). DCC expression by neurons regulatessynaptic plasticity in the adult brain. Cell Rep3, 173–185.

Ivakine EA, Acton BA, Mahadevan V, OrmondJ, Tang M, Pressey JC, Huang MY, Ng

© 2020 The Authors. The Journal of Physiology © 2020 The Physiological Society

394 Editorial J Physiol 599.2

D, Delpire E, Salter MW, Woodin MA &McInnes RR (2013). Neto2 is a KCC2 inter-acting protein required for neuronal Cl−regulation in hippocampal neurons. Proc NatlAcad Sci U S A 110, 3561–3566.

Jacobi E & von Engelhardt J (2021). Modulationof information processing by AMPA receptorauxiliary subunits. J Physiol 599, 471–483.

Jatzke C, Hernandez M & Wollmuth LP (2003).Extracellular vestibule determinants of Ca2+influx in Ca2+-permeable AMPA receptorchannels. J Physiol 549, 439–452.

Joshi I & Wang LY (2002). Developmentalprofiles of glutamate receptors and synaptictransmission at a single synapse in the mouseauditory brainstem. J Physiol 540, 861–873.

Kaila K, Price TJ, Payne JA, PuskarjovM & Voipio J (2014). Cation-chloridecotransporters in neuronal development,plasticity and disease. Nat Rev Neurosci 15,637–654.

Kamalova A & Nakagawa T (2021). AMPAreceptor structure and auxiliary subunits. JPhysiol 599, 453–469.

Kennedy TE & Tessier-Lavigne M (1995).Guidance and induction of branch formationin developing axons by target-deriveddiffusible factors. Curr Opin Neurobiol 5,83–90.

Khodosevich K, Jacobi E, Farrow P, SchulmannA, Rusu A, Zhang L, Sprengel R, MonyerH & von Engelhardt J (2014). Coexpressedauxiliary subunits exhibit distinct modulatoryprofiles on AMPA receptor function. Neuron83, 601–615.

Klaassen RV, Stroeder J, Coussen F, HafnerAS, Petersen JD, Renancio C, SchmitzLJ, Normand E, Lodder JC, Rotaru DC,Rao-Ruiz P, Spijker S, Mansvelder HD,Choquet D & Smit AB (2016). Shisa6 trapsAMPA receptors at postsynaptic sites andprevents their desensitization during synapticactivity. Nat Commun 7, 10682.

Krishtal O (2015). Receptor for protons: Firstobservations on acid sensing ion channels.Neuropharmacology 94, 4–8.

Levy AD, Xiao X, Shaw JE, Sudarsana DeviSP, Katrancha SM, Bennett AM, GreerCA, Howe JR, Machida K & Koleske AJ(2018). Noonan syndrome-associated SHP2dephosphorylates GluN2B to regulate NMDAreceptor function. Cell Rep 24, 1523–1535.

MacLean DM & Jayaraman V (2016).Acid-sensing ion channels are tuned tofollow high-frequency stimuli. J Physiol 594,2629–2645.

Mahadevan V, Dargaei Z, Ivakine EA,Hartmann AM, Ng D, Chevrier J, OrmondJ, Nothwang HG, McInnes RR & WoodinMA (2015). Neto2-null mice have impairedGABAergic inhibition and are susceptible toseizures. Front Cell Neurosci 9, 368.

Mahadevan V, Pressey JC, Acton BA, UvarovP, Huang MY, Chevrier J, Puchalski A, LiCM, Ivakine EA, Airaksinen MS, Delpire E,McInnes RR & Woodin MA (2014). Kainate

receptors coexist in a functional complexwith KCC2 and regulate chloride homeo-stasis in hippocampal neurons. Cell Rep 7,1762–1770.

McGee TP, Bats C, Farrant M & Cull-CandySG (2015). Auxiliary subunit GSG1L acts tosuppress calcium-permeable AMPA receptorfunction. J Neurosci 35, 16171–16179.

Melyan Z, Wheal HV & Lancaster B (2002).Metabotropic-mediated kainate receptorregulation of IsAHP and excitability inpyramidal cells. Neuron 34, 107–114.

Miller PS & Aricescu AR (2014). Crystalstructure of a human GABAA receptor.Nature 512, 270–275.

Myers SJ, Yuan H, Kang JQ, Tan FCK, TraynelisSF & Low CM (2019). Distinct roles ofGRIN2A and GRIN2B variants in neuro-logical conditions. F1000Res 8, 1940.

Nakagawa T (2019). Structures of the AMPAreceptor in complex with its auxiliary subunitcornichon. Science 366, 1259–1263.

Nakagawa T, Cheng Y, Ramm E, Sheng M&Walz T (2005). Structure and differentconformational states of native AMPAreceptor complexes. Nature 433, 545–549.

Nicoll RA (2017). A brief history of long-termpotentiation. Neuron 93, 281–290.

Paoletti P & Ascher P (1994). Mechano-sensitivity of NMDA receptors in culturedmouse central neurons. Neuron 13, 645–655.

Paternain AV, Morales M & Lerma J (1995).Selective antagonism of AMPA receptorsunmasks kainate receptor-mediatedresponses in hippocampal neurons. Neuron14, 185–189.

Peter S, Urbanus BHA, Klaassen RV, Wu B,Boele HJ, Azizi S, Slotman JA, HoutsmullerAB, Schonewille M, Hoebeek FE, Spijker S,Smit AB & De Zeeuw CI (2020). AMPARauxiliary protein SHISA6 facilitates purkinjecell synaptic excitability and proceduralmemory formation. Cell Rep 31, 107515.

Pressey JC, Mahadevan V, KhademullahCS, Dargaei Z, Chevrier J, Ye W, HuangM, Chauhan AK, Meas SJ, Uvarov P, Air-aksinen MS & Woodin MA (2017). A kainatereceptor subunit promotes the recycling ofthe neuron-specific K+-Cl− co-transporterKCC2 in hippocampal neurons. J Biol Chem292, 6190–6201.

Pressey JC & Woodin MA (2021). Kainatereceptor regulation of synaptic inhibition inthe hippocampus. J Physiol 599, 485–492.

Roberts AE, Allanson JE, Tartaglia M & GelbBD (2013). Noonan syndrome. Lancet 381,333–342.

Rodriguez-Moreno A & Lerma J (1998). Kainatereceptor modulation of GABA releaseinvolves a metabotropic function. Neuron20, 1211–1218.

Rook ML, Musgaard M & MacLean DM(2021). Coupling structure with functionin acid-sensing ion channels: challengesin pursuit of proton sensors. J Physiol 599,417–430.

Rook ML, Williamson A, Lueck JD, MusgaardM & Maclean DM (2020). β11-12 linker iso-merization governs acid-sensing ion channeldesensitization and recovery. Elife 9, e51111.

Rosenbaum MI, Clemmensen LS, Bredt DS,Bettler B & Stromgaard K (2020). Targetingreceptor complexes: a new dimension in drugdiscovery. Nat Rev Drug Discov 19, 884–901.

Rosenmund C & Westbrook GL (1993).Calcium-induced actin depolymerizationreduces NMDA channel activity. Neuron 10,805–814.

Salter MW & Kalia LV (2004). Src kinases: ahub for NMDA receptor regulation. Nat RevNeurosci 5, 317–328.

Schofield PR, Darlison MG, Fujita N, BurtDR, Stephenson FA, Rodriguez H, RheeLM, Ramachandran J, Reale V & GlencorseTA, et al. (1987). Sequence and functionalexpression of the GABA A receptor shows aligand-gated receptor super-family. Nature328, 221–227.

Schwenk J, Baehrens D, Haupt A, Bildl W,Boudkkazi S, Roeper J, Fakler B & Schulte U(2014). Regional diversity and developmentaldynamics of the AMPA-receptor proteome inthe mammalian brain. Neuron 84, 41–54.

Shaw JE & Koleske AJ (2021). Functional inter-actions of ion channels with the actin cyto-skeleton: does coupling to dynamic actinregulate NMDA receptors? J Physiol 599,431–441.

Sobolevsky AI, Rosconi MP & Gouaux E(2009). X-ray structure, symmetry andmechanism of an AMPA-subtype glutamatereceptor. Nature 462, 745–756.

Twomey EC, Yelshanskaya MV, GrassucciRA, Frank J & Sobolevsky AI (2017).Structural bases of desensitization in AMPAreceptor-auxiliary subunit complexes. Neuron94, 569–580.e5.

Twomey EC, Yelshanskaya MV & Sobolevsky AI(2019). Structural and functional insights intotransmembrane AMPA receptor regulatoryprotein complexes. J Gen Physiol 151,1347–1356.

von Engelhardt J, Mack V, Sprengel R,Kavenstock N, Li KW, Stern-Bach Y, SmitAB, Seeburg PH & Monyer H (2010).CKAMP44: a brain-specific proteinattenuating short-term synaptic plasticityin the dentate gyrus. Science 327, 1518–1522.

Watanabe J, Beck C, Kuner T, Premkumar LS& Wollmuth LP (2002). DRPEER: a motif inthe extracellular vestibule conferring highCa2+ flux rates in NMDA receptor channels.J Neurosci 22, 10209–10216.

Won S, Incontro S, Li Y, Nicoll RA & RocheKW (2019). The STEP61 interactome revealssubunit-specific AMPA receptor binding andsynaptic regulation. Proc Natl Acad Sci U S A116, 8028–8037.

Won S, Incontro S, Nicoll RA & Roche KW(2016). PSD-95 stabilizes NMDA receptorsby inducing the degradation of STEP61. ProcNatl Acad Sci U S A 113, E4736–E4744.

© 2020 The Authors. The Journal of Physiology © 2020 The Physiological Society

J Physiol 599.2 Editorial 395

Won S & Roche KW (2021). Regulation ofglutamate receptors by striatal-enrichedtyrosine phosphatase 61 (STEP61). J Physiol599, 443–451.

Wong EW, Glasgow SD, Trigiani LJ, ChitsazD, Rymar V, Sadikot A, Ruthazer ES, HamelE & Kennedy TE (2019). Spatial memoryformation requires netrin-1 expression byneurons in the adult mammalian brain. LearnMem 26, 77–83.

Wyszynski M, Lin J, Rao A, Nigh E, Beggs AH,Craig AM & Sheng M (1997). Competitivebinding of alpha-actinin and calmodulin tothe NMDA receptor. Nature 385, 439–442.

Xu J, Chatterjee M, Baguley TD, BrouilletteJ, Kurup P, Ghosh D, Kanyo J, Zhang Y,Seyb K, Ononenyi C, Foscue E, AndersonGM, Gresack J, Cuny GD, Glicksman MA,Greengard P, Lam TT, Tautz L, Nairn AC,Ellman JA & Lombroso PJ (2014). Inhibitorof the tyrosine phosphatase STEP reversescognitive deficits in a mouse model ofAlzheimer’s disease. PLoS Biol 12, e1001923.

Yoder N, Yoshioka C & Gouaux E (2018).Gating mechanisms of acid-sensing ionchannels. Nature 555, 397–401.

Zeisel A, Hochgerner H, Lonnerberg P,Johnsson A, Memic F, van der Zwan J,Haring M, Braun E, Borm LE, La MannoG, Codeluppi S, Furlan A, Lee K, SkeneN, Harris KD, Hjerling-Leffler J, ArenasE, Ernfors P, Marklund U & Linnarsson S(2018). Molecular architecture of the mousenervous system. Cell 174, 999–1014.e22.

Additional information

Competing interests

No competing interests declared.

Author contributions

Sole author.

Funding

The author received funding from CIHR: FRN162317 and FRN 142431.

Keywords

AMPA receptor, ASIC channels, ion channels,GABA receptor, kainate receptor, NMDAreceptor, synapse, synaptic plasticity

© 2020 The Authors. The Journal of Physiology © 2020 The Physiological Society