COMMENTARY

Kv2 potassium channels meet VAP COMMENTARY

Elizabeth Wen Suna,b,c,d and Pietro De Camillia,b,c,d,1

A defining characteristic of eukaryotic cells is the presence of distinct intracellular membrane-bound A compartments. Much research has focused on the functional interconnection of these organelles via membrane traffic. A flurry of recent studies, however, has brought to center stage the important role of interorganelle communication independent of vesic- ular transport and mediated by direct contacts (1–3). At these sites, membranes are tethered to each other by –protein or protein–lipid interactions not leading to fusion. These contacts play a variety of func- tions, including regulation of ion fluxes across mem- Plasma Membrane Endoplasmic Reticulum ER-PM contact sites (<30nm) branes and transport of lipids between participating organelles. In PNAS, Johnson et al. (4) provide yet an- cytosolic VAP interactors other unexpected example of direct communication B Kv2 channels B’ VAP (A/B) PM B’ between two membranes: the binding of the plasma Mitochondrion Kv2 membrane (PM)-localized major delayed-rectifier voltage- PM + gated K channels, Kv2.1 and Kv2.2, to VAMP-associated Endosome C’

protein (VAP), an integral of the endo- Lipid droplet N’

plasmic reticulum (ER). VAP Kv2.1 and Kv2.2 (KCNB1 and KCNB2) channels RyR are very abundant in the brain, where they play a major MVB

role in neuronal excitability. They form large clusters in Golgi PRC region ER the PM of neuronal cell bodies, proximal dendrites, and ER Lipid transfer axon initial segments, and such clusters are at sites – where the PM is tightly apposed to the ER (5). In the Fig. 1. ER PM contacts and role of VAP as a tethering protein at the surface of the ER. (A) A 3D reconstruction from a focused ion beam–SEM image stack of the ER in clusters, the cytosolic tails of the channels are in a phos- the cortical region of the cell body of a mouse brain neuron. ER cisternae closely phorylated state and the clusters disperse within mi- apposed to the PM are shown in red (adapted with permission from ref. 8). (Scale nutes due to calcium-dependent dephosphorylation bar: 400 nm.) (B) Schematic cartoon highlighting VAP-dependent interactions at – upon excessive excitatory stimulation or exposure to the ER surface and the coclustering of VAP with Kv2 channels at ER PM contacts. (B′) The cartoon highlights binding of a VAP dimer to the PRC region in the noxious conditions, such as hypoxia (6). While an amino C-terminal portion of Kv2 channels and to a lipid transport protein. A RyR is acid stretch, referred to as the PRC (proximal restriction also shown. Only one subunit of the tetrameric Kv2 channels and two subunits and clustering), was known to be required for clustering of the RyR are shown for simplicity. (7), how the PRC mediates clustering remained unclear. The ER is the most abundant intracellular mem- branous organelle. It comprises a system of tubules cellular membranes (Fig. 1) (8). Contacts between the and cisterns that are continuous with each other and ER and the PM were first observed in muscle, where populate every cell compartment, including axon they are the basis for contraction cou- endings and dendritic spines in neurons (8). Its mem- pling via a direct interaction between PM voltage- + branes make focal and tight apposition with all other dependent Ca2 channels and the ryanodine receptors

aDepartment of Neuroscience, Yale University School of Medicine, New Haven, CT 06510; bDepartment of Cell Biology, Yale University School of Medicine, New Haven, CT 06510; cKavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06510; and dHoward Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510 Author contributions: E.W.S. and P.D.C. wrote the paper. The authors declare no conflict of interest. Published under the PNAS license. See companion article on page E7331. 1To whom correspondence should be addressed. Email: [email protected]. Published online July 17, 2018.

www.pnas.org/cgi/doi/10.1073/pnas.1810059115 PNAS | July 31, 2018 | vol. 115 | no. 31 | 7849–7851 Downloaded by guest on October 2, 2021 + (RyR): that is, the Ca2 release channels of the ER. However, it more of its serines, providing a possible explanation for why subsequently became clear that ER–PM contacts are present in the clustering of Kv2 channels is critically dependent on their all cells and may have a variety of functions. For example, ER–PM being in the phosphorylated state. Supporting this possibility, + contacts mediate store-operated Ca2 entry, a housekeeping pro- serines within the PRC (S583, S586, and S589) had been previ- + cess in which binding of the ER protein STIM1 (a sensor of Ca2 ously shown to be required for clustering (7), although a precise + concentration in the ER lumen) to the PM Ca2 channel Orai trig- validation of this mode of binding will require structural studies. A + gers Ca2 influx. Another function is lipid transport, or countertran- puzzling question left open by these studies is what controls the sport, via that contain lipid transport modules and also clustering of Kv2 channels at axon initial segments, because the tether the two participating membranes (1). This transport helps PRC region of Kv2, and thus the interaction with VAP, is dispens- control the lipid composition of the PM and allows homeostatic able for this localization (13). adaptations of its lipid bilayer in response to acute perturbations. While the study by Johnson et al. (4) provides an explanation ER–PM appositions are particularly large in neurons, where for the clustering of Kv2 channels at ER–PM contacts in neuronal micrometer-sized ER cisternae (subsurface/hypolemmal cisternae), often with a very narrow lumen (thin ER), cover about 10% of the PM of cell bodies (Fig. 1A) (8). Smaller contacts are also present in Collectively, Johnson et al.’s experiments dendrites, axons, and axon terminals. demonstrate the causality of Kv2 channels The localization of Kv2 clusters at ER–PM contacts suggested their direct or indirect interactions with ER proteins. To identify binding to VAP for their accumulation, and thus such partners, Johnson et al. (4) capitalize on proximity labeling clustering, at ER–PM contact sites. with biotin using APEX-based methodology (8). They appended APEX to the cytosolic region of AMIGO, an integral plasma mem- brane protein closely associated with the α-subunits of the Kv2 chan- perikarya and dendrites, the physiological significance of this nels, and then searched for biotinylated proteins by streptavidin localization, and more specifically of the interaction with VAP, labeling. A prominently biotinylated protein of 33 kDa was identified, remains an open question. Clustered Kv2 channels have a leading Johnson et al. (4) to hypothesize that VAP (also known as different threshold for activation than nonclustered channels VAP33), an abundant ER protein, could be the interactor (9). Johnson (14), raising the possibility that binding to VAP may control et al. (4) tested and proved this hypothesis through expression and their voltage sensitivity. Alternatively, as VAP proteins dimerize coexpression of tagged wild-type and mutant Kv2 channels and and further oligomerize (10), they could recruit other proteins VAP proteins, as well as through knockdown experiments in neu- to help modulate channel function indirectly. They may do so rons and HEK293 cells. Collectively, Johnson et al.’s experiments by affecting the focal lipid composition of the PM, as a large demonstrate the causality of Kv2 channels binding to VAP for their fraction of client proteins for VAP are implicated in lipid accumulation, and thus clustering, at ER–PM contact sites (Fig. 1B′). transport and metabolism and the function of Kv2 channels is – VAP is the collective name for two very similar small dimeric regulated by PI(4,5)P2 (9, 15). For example, Kv2.1-enriched ER proteins expressed throughout evolution from yeast (Scs2 and PM contacts that face C-type presynaptic nerve terminals are Scs22) to humans (VAPA and VAPB), and comprising an N-terminal also enriched in phospholipase C (PLC)-coupled muscarinic (MSP) module followed by a coiled–coil region acetylcholine receptors (16), thus making plausible a potential and a C-terminal hydrophobic sequence that anchors it to the ER. coupling between PI(4,5)P2 metabolism and Kv2.1 channel The surface of the MSP domain harbors a positively charged function. Clearly, many other scenarios are possible, as a groove that functions as the binding site for the so-called FFAT recent comprehensive analysis of the VAP “interactome” motif [two phenylalanine (F) in an acidic tract] (9, 10). Dozens of revealed many proteins with roles not obviously linked to lipid proteins with this motif or its variants have been identified and for dynamics (9). many of them the interaction with VAP has been experimentally VAP-induced recruitment of the ER to the Kv2 channels in the confirmed (9). Thus, VAP serves as an anchor at the surface of the PM may also have roles independent of VAP itself. In Purkinje + ER for many cytosolic proteins, and participates in bridging two cells, another class of K channels, the BK channels (large- + membranes when the FFAT motif-containing partner protein is in conductance Ca2 -activated potassium channels), is concen- turn anchored via a lipid-binding module or a transmembrane re- trated in regions of the PM closely apposed to the ER (17). + gion to another membrane (Fig. 1 B and B′). The importance of VAP The association of K channels with the ER may provide a func- + for cell physiology is underscored by the striking perturbation of tional link between these channels and Ca2 dynamics. This may lipid homeostasis and intracellular organization occurring in VAPA occur either through the presence of RyRs or of voltage- + and VAPB double KO cells (11) and by the embryonic lethality of dependent PM Ca2 (CaV1.2) channels at the same ER–PM con- VAPA KO mice (12). While VAP-dependent contacts between the tacts (14, 18). Because a large fraction of the Kv2 channels are ER and all other cellular membranes, including the PM, have been thought to be in a nonconducting state, and more so the pop- reported (1), a direct binding of VAP to a PM protein had never ulation of the channels accumulated at ER–PM contacts, it is also been previously identified. possible that these channels may have a nonconducting role As Johnson et al. (4) show, neither Kv2.1 nor Kv2.2 contains a that impacts some function of the ER, or of ER-anchored pro- canonical FFAT motif. However, the authors note that an amino teins. Because the voltage-sensing portion of the channels is not acid sequence reminiscent of this motif is present within the PRC affected by their clustering (14), voltage-dependent conforma- stretch of their cytosolic region (Fig. 1B′). In the stretch, the sec- tional changes of the Kv2 channels may affect proteins at the ond phenylalanine (F) is replaced by an isoleucine (I). Addition- PM–ER interface. ally, the acidic amino acids, which typically characterize this Perturbation of the interaction between Kv2 channels and VAP motif, are not present. Johnson et al. propose that the acidic may be involved in disease. Mutations that truncate the channels charge is conferred to the PRC by of one or upstream of the PRC motif, but do not perturb voltage-dependent

7850 | www.pnas.org/cgi/doi/10.1073/pnas.1810059115 Sun and De Camilli Downloaded by guest on October 2, 2021 function, result in developmental delay (19). Furthermore, In conclusion, while the precise functions of Kv2.1 and Kv2.2 mutations of VAPB that disrupt its FFAT motif binding properties channels concentrated at ER–PM contact sites remain elusive, the result in a dominant familial form of amyotrophic lateral sclerosis knowledge of the mechanism responsible for this localization (20). Given the multiplicity of VAP interactors, clinical manifestations opens new possibilities for future investigation. may be the collective results of many perturbations, but it will be of great interest to determine if loss of Kv2 binding may have a role Note Added in Proof. A manuscript reporting similar findings by in disease. another group (21) is in press in the Journal of Neuroscience.

1 Salvador-Gallego R, Hoyer MJ, Voeltz GK (2017) SnapShot: Functions of endoplasmic reticulum membrane contact sites. Cell 171:1224–1224.e1. 2 Saheki Y, De Camilli P (2017) Endoplasmic reticulum-plasma membrane contact sites. Annu Rev Biochem 86:659–684. 3 Levine T, Loewen C (2006) Inter-organelle membrane contact sites: Through a glass, darkly. Curr Opin Cell Biol 18:371–378. 4 Johnson B, et al. (2018) Kv2 potassium channels form endoplasmic reticulum/plasma membrane junctions via interaction with VAPA and VAPB. Proc Natl Acad Sci USA 115:E7331–E7340. 5 Trimmer JS (1991) Immunological identification and characterization of a delayed rectifier K+ channel polypeptide in rat brain. Proc Natl Acad Sci USA 88:10764–10768. 6 Misonou H, et al. (2004) Regulation of ion channel localization and phosphorylation by neuronal activity. Nat Neurosci 7:711–718. 7 Lim ST, Antonucci DE, Scannevin RH, Trimmer JS (2000) A novel targeting signal for proximal clustering of the Kv2.1 K+ channel in hippocampal neurons. Neuron 25:385–397. 8 Wu Y, et al. (2017) Contacts between the endoplasmic reticulum and other membranes in neurons. Proc Natl Acad Sci USA 114:E4859–E4867. 9 Murphy SE, Levine TP (2016) VAP, a versatile access point for the endoplasmic reticulum: Review and analysis of FFAT-like motifs in the VAPome. Biochim Biophys Acta 1861:952–961. 10 Kaiser SE, et al. (2005) Structural basis of FFAT motif-mediated ER targeting. Structure 13:1035–1045. 11 Dong R, et al. (2016) Endosome-ER contacts control actin nucleation and retromer function through VAP-dependent regulation of PI4P. Cell 166:408–423. 12 McCune BT, et al. (2017) Noroviruses co-opt the function of host proteins VAPA and VAPB for replication via a phenylalanine-phenylalanine-acidic-tract-motif mimic in nonstructural viral protein NS1/2. MBio 8:e00668-e17. 13 Jensen CS, et al. (2017) Trafficking of Kv2.1 channels to the axon initial segment by a novel nonconventional secretory pathway. J Neurosci 37:11523–11536. 14 O’Connell KMS, Loftus R, Tamkun MM (2010) Localization-dependent activity of the Kv2.1 delayed-rectifier K+ channel. Proc Natl Acad Sci USA 107:12351–12356. 15 Delgado-Ram´ırez M, et al. (2018) Regulation of Kv2.1 channel inactivation by phophatidylinositol 4,5-bisphosphate. Sci Rep 8:1769. 16 Muennich EAL, Fyffe REW (2004) Focal aggregation of voltage-gated, Kv2.1 subunit-containing, potassium channels at synaptic sites in rat spinal motoneurones. J Physiol 554:673–685. 17 Kaufmann WA, et al. (2009) Large-conductance calcium-activated potassium channels in Purkinje cell plasma membranes are clustered at sites of hypolemmal microdomains. J Comp Neurol 515:215–230. 18 Kirmiz M, et al. (2018) Organizing neuronal ER-PM junctions is a conserved nonconducting function of Kv2 plasma membrane ion channels. bioRxiv, 10.1101/ 296731. 19 de Kovel CGF, et al. (2017) Neurodevelopmental disorders caused by de novo variants in KCNB1 genotypes and phenotypes. JAMA Neurol 74:1228–1236. 20 Nishimura AL, et al. (2004) A mutation in the vesicle-trafficking protein VAPB causes late-onset spinal muscular atrophy and amyotrophic lateral sclerosis. Am J Hum Genet 75:822–831. 21 Kirmiz M, Vierra NC, Palacio S, Trimmer JS, Identification of VAPA and VAPB as Kv2 channel-interacting proteins defining endoplasmic reticulum-plasma membrane junctions in mammalian brain neurons. J Neurosci, in press.

Sun and De Camilli PNAS | July 31, 2018 | vol. 115 | no. 31 | 7851 Downloaded by guest on October 2, 2021