Quick viewing(Text Mode)

The Diversity of Calcium Sensor Proteins in the Regulation of Neuronal Function

The Diversity of Calcium Sensor Proteins in the Regulation of Neuronal Function

Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

The Diversity of Sensor in the Regulation of Neuronal Function

Hannah V. McCue, Lee P. Haynes, and Robert D. Burgoyne

The Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Crown Street, Liverpool L69 3BX, United Kingdom Correspondence: [email protected]

Calcium signaling in as in other cell types mediates changes in expression, , development, survival, and cell death. However, neuronal Ca2þ signaling processes have become adapted to modulate the function of other important pathways including axon outgrowth and changes in synaptic strength. Ca2þ plays a key role as the trigger for fast neuro- transmitter release. The ubiquitous Ca2þ sensor is involved in various aspects of neuronal regulation. The mechanisms by which changes in intracellular Ca2þ concentration in neurons can bring about such diverse responses has, however, become a topic of wide- spread interest that has recently focused on the roles of specialized neuronal Ca2þ sensors. In this article, we summarize in release, the neur- onal roles of calmodulin, and the functional significance of the NCS and the CaBP/ calneuron families of neuronal Ca2þ sensors.

alcium signaling in many cell types can processes have been shown to be dependent Cmediate changes in , cell upon the particular route of Ca2þ entry into growth, development, survival, and cell death. the cell. It has long been known that the physio- However, neuronal processes logical outcome from a change in [Ca2þ]i have become adapted to modulate the function depends on its location, amplitude, and dura- of important pathways in the , including tion. The importance of location becomes neuronal survival, axon outgrowth, and changes even more pronounced in neurons because of in synaptic strength. Changes in the concentra- their complex and extended morphologies. tion of intracellular free Ca2þ ([Ca2þ]i) are [Ca2þ]i also regulates neuronal development essential for the transmission of information and neuronal survival (Spitzer 2006). In addi- through the nervous system as the trigger for tion, modifications to Ca2þ signaling pathways neurotransmitter release at . In addi- have been suggested to underlie various neuro- tion, alterations in [Ca2þ]i can lead to a wide pathological disorders (Braunewell 2005; Ber- range of different physiological changes that ridge 2010). can modify neuronal functions over time scales Highly localized Ca2þ elevations (Augustine of milliseconds through tens of minutes to et al. 2003) formed following Ca2þ entry though days or longer (Berridge 1998). Many of these voltage-gated Ca2þ channels (VGCCs) lead to

Editors: Martin Bootman, Michael J. Berridge, James W. Putney, and H. Llewelyn Roderick Additional Perspectives on Calcium Signaling available at www.cshperspectives.org Copyright # 2010 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a004085 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085

1 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

H.V. McCue, L.P. Haynes, and R.D. Burgoyne

synaptic with the presynaptic role of synaptotagmins in neurotransmitter membrane and thereby allow neurotransmitter release has been the subject of intense investiga- release within less than a millisecond. Differ- tions, which have been extensively reviewed ently localized and timed Ca2þ signals can, for (Chapman 2008; Rizo and Rosenmund 2008; example, result in changes to the properties of Sudhof and Rothman 2009) and so only a brief the VGCCs (Catterall and Few 2008) or lead to outline is given here. Synaptotagmins bind changes in gene expression (Bito et al. 1997). Ca2þ with relatively low affinity (Kd . 10 mM) Postsynaptic Ca2þ signals arising from activa- through their two C2 domains (C2A and C2B) tion of NMDA receptors give rise to two impor- (Shao et al. 1998; Fernandez et al. 2001), which tant processes in , long term are functional in many but not all synaptotag- potentiation (LTP) and long term depression min isoforms. Ca2þ binding by C2 domains (LTD). LTP and LTD are examples of the way requires coordination of Ca2þ by both the synaptic transmission can change synaptic effi- protein and membrane lipids and this lipid cacy and are thought to be important in modu- interaction is a key aspect for its function. In lating learning and memory. Importantly, the I, the C2A and C2B domains Ca2þ signals that bring about either LTP or (Fig. 1) bind three and two Ca2þ ions, respec- LTD differ only in their timing and duration. tively (Shao et al. 1998; Fernandez et al. 2001). LTP is triggered by Ca2þ signals on the micro- It is now well established that synaptotagmin I molar scale for shorter durations, whereas LTD is a key sensor for evoked, synchronous neuro- is triggered by changes in [Ca2þ]i on the nano- transmitter release in many classes of neurons molar scale for longer durations (Yang et al. (Fernandez-Chacon et al. 2001). Structure– 1999). Specific Ca2þ signals are likely to be function studies based on expression of specific decoded by different Ca2þ sensor proteins. mutants have been carried out in mice, worms, These are proteins that undergo a conforma- and flies. For example, disruption of Ca2þ bind- tional change on Ca2þ binding and then interact ing to the C2B domain of synaptotagmin I has with and regulate various target proteins. Among those Ca2þ sensors that are important for neuronal function are the synaptotagmins that control neurotransmitter release (Chap- man 2008), the ubiquitous EF-hand contain- ing sensor calmodulin that has many neuronal roles, and the more recently discovered neuronal EF-hand containing proteins, including the (NCS) protein (Bur- goyne 2007) and the calcium-binding protein (CaBP)/calneuron (Haeseleer et al. 2002) fam- ilies. We will briefly review synaptotagmins and the neuronal functions of calmodulin but concentrate on the NCS and CaBP families of Ca2þ sensors.

SYNAPTOTAGMINS AND NEUROTRANSMITTER RELEASE Synaptotagmins are transmembrane proteins Figure 1. Structures of the C2A and C2B domains of synaptotagmin I. The structures show the isolated C2 mostly found associated with synaptic and domains in their Ca2þ-loaded state with the bound secretory vesicles. There are multiple known Ca2þ ions shown in green. The coordinates for the isoforms of synaptotagmin (Craxton 2004) of structures for the C2A and C2B domains come which synaptotagmin I is the best studied. The from the PDB files 1BYN and 1K5W, respectively.

2 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

Calcium Sensor Proteins in Neuronal Function

been shown to have a more deleterious effect NEURONAL FUNCTIONS OF CALMODULIN than disruption of Ca2þ binding to its C2A domain (Mackler et al. 2002; Robinson et al. Calmodulin is a ubiquitously expressed Ca2þ 2002). The details of exactly how it triggers exo- -binding protein that can bind four Ca2þ ions cytosis and the function of other syntaptotag- through its four EF-hand domains (Chattopad- min isoforms remain controversial. Membrane hyaya et al. 1992). This protein has been highly fusion requires the pairing and interaction of conserved throughout evolution, is found in all so-called SNARE proteins on vesicle and target , and is 100% identical across all ver- membranes (Sollner et al. 1993). These can tebrates at the amino acid level. It is involved in assemble into a SNARE complex that may the regulation of many essential physiological form the minimal fusion machinery. For synap- processes including cell , , tic vesicle and neuroendocrine exocytosis, the cytoskeletal assembly, and modulation of intra- SNARE proteins are SNAP-25, 1, and cellular Ca2þ concentrations. The first two EF- . In the case of regulated exocy- hands of calmodulin form an amino-terminal tosis, such as in neurotransmitter release, ves- globular domain that is joined by a flexible icle fusion is tightly regulated and requires a linker to a highly homologous carboxy-termi- Ca2þ signal for activation. Ca2þ entry through nal region encompassing the third and fourth VGCCs leading to Ca2þ elevation in local EF-hands. The carboxy-terminal pair of EF- microdomains close to the mouth of the Ca2þ hands has a much higher affinity for Ca2þ channel is able to trigger very rapid (within than the amino-terminal pair, which allows less than 1 ms) fusion of synaptic vesicles. Syn- the two domains to behave independently at aptotagmin can bind to both syntaxin and varying Ca2þ concentrations (Tadross et al. SNAP-25, and fast neurotransmitter release 2008). The highly flexible linker between the requires synaptotagmin (Geppert et al. 1994) two domains can be bent dramatically upon probably prebound to assembled or partially binding to target proteins (Fig. 2) and is an assembled SNARE complexes (Schiavo et al. essential property of calmodulin, which permits 1997; Rickman et al. 2006) so that Ca2þ- this protein to interact with a large and diverse induced interaction with can array of interacting partners. The significant occur rapidly (Xue et al. 2008). It is still under conformational changes on binding to its tar- debate how important synaptotagmin is in gets (Fallon et al. 2005) can increase its affinity vesicle docking (de Wit et al. 2009) and how it for Ca2þ. acts at the plasma membrane in fusion itself Calmodulin is present in brain at high con- (Tang et al. 2006; Hui et al. 2009). Synaptotag- centrations (up to 100 mM). In addition to min could act as a brake on fusion that is its more general functions, calmodulin also has relieved on Ca2þ binding or have a positive a series of specific roles in transducing Ca2þ sig- role in membrane fusion (Chicka et al. 2008). nals in neurons, including, for example, in the A recent focus has been on the combined role regulation of glutamate receptors (O’Connor of synaptotagmin and another SNARE interact- 1999), ion channels (Saimi and Kung 2002), ing protein in timing and proteins in signaling pathways such as neu- fusion (Sudhof and Rothman 2009), but much ronal synthase, and it can affect syn- still remains to be learnt about the molecular aptic plasticity (Lisman et al. 2002; Xia and basis of its function. While synaptotagmin is a Storm 2005). One key direct function of calmo- key sensor for evoked neurotransmitter release, dulin is in regulating the activity of VGCCs by an alternative C2-domain containing protein, binding to channel subunits (Catterall and Few Doc2b, has been identified as a Ca2þ sensor for 2008). Ca2þ-binding to VGCC-associated cal- spontaneous neurotransmitter release (Groffen modulin can have a range of effects on channel et al. 2010). Like synaptotagmin, Doc2b ap- function, including mediating Ca2þ-dependent pears to function via interaction with SNARE facilitation or Ca2þ-inactivation (Lee et al. 2000; complexes. DeMaria et al. 2001; Catterall and Few 2008;

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 3 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

H.V. McCue, L.P. Haynes, and R.D. Burgoyne

Figure 2. Comparison of the structures of Ca2þ-loaded calmodulin and yeast frequenin with and without bound target peptides. The structures at the top are of Ca2þ-bound calmodulin alone (PDB 1CLL) or in a complex with 2þ the IQ-like domain of the Cav1.2 Ca -channel a-subunit (PDB 2F3Z). The structures at the bottom are of the Ca2þ-bound yeast frequenin (Frq1) alone (PDB 1FPW) or in a complex with the binding domain from Pik1 (PDB 2JU0). In each of the complexes, the target peptide is shown in yellow.

Liu et al. 2010). Calmodulin is also constitu- Ca2þ/calmodulin-dependent (CaMKs) tively associated with and regulates opening of and . CaMKs contribute to a num- Ca2þ-activated potassium channels (Xia 1998; ber of regulatory pathways involving, for ex- Schumacher et al. 2001) and other types of ample, of AMPA receptors potassium channels (Wen and Levitan 2002). (Barria et al. 1997) and the nuclear transcrip- Twoother major modes of action of calmodulin tion factor CREB (Deisseroth et al. 1998). Cal- are exerted indirectly through its target proteins modulin also positively regulates presynaptic

4 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

Calcium Sensor Proteins in Neuronal Function

release probability and this is mediated via acti- closely related (Sanchez-Gracia et al. vation of CaMKII (Pang et al. 2010). The Ca2þ- 2010). Although initially thought to be neu- activated calcineurin can dephos- ronal specific (Nef et al. 1995), NCS-1 has phorylate a wide range of neuronal proteins, also been identified in Saccharomyces cerevisiae leading to direct effects and effects through (Hendricks et al. 1999). After this first appear- changes in gene transcription following activa- ance of NCS-1 in yeast, there has been a steady tion of the NFAT and its increase in the diversity of this family through- translocation into the nucleus. Calcineurin has out evolution, which roughly correlates with also been implicated, for example, in synaptic increasing organism complexity. Five classes of plasticity (Malleret et al. 2001; Xia and Storm NCS proteins have now been identified in 2005). Although many aspects of neuronal higher organisms termed classes A-E (Burgoyne function are known to be regulated by calmodu- 2007). Class A contains NCS-1, which is present lin, proteins related to calmodulin have been in yeast and all higher organisms. Class B con- discovered in recent years, which are enriched sists of the visinin-like proteins (VSNLs), which or expressed exclusively in neurons. Duplication appear first in Caenorhabditis elegans. Classes C and diversification of the calmodulin gene may and D evolved with the appearance of fish and have given rise to these neuronal calcium sens- comprise and the guanylyl-cyclase- ing proteins so that they can carry out specific activating proteins (GCAPs), respectively. Fin- neuronal functions in higher organisms. ally, class E contains the Kþ channel-interacting proteins (KChIPs), which are found in insects and evolutionary subsequent species (Burgoyne NCS PROTEIN FAMILY 2004). have a single NCS-1, five Whereas calmodulin is ubiquitously expressed, VSNL proteins (, d,and the expression of other calcium sensing proteins VILIPs1-3), a single recoverin, three GCAPs, can be restricted to particular tissues and cell and four KChIPs. Expression of the recoverins types. A good example of this is the neuronal and GCAPs is restricted to the retina, whereas calcium sensor (NCS) family of proteins, which the rest of the NCS family is found in varied are primarily expressed in neurons or retinal neuronal populations (Burgoyne 2007). Al- photoreceptors. The NCS family of proteins is though localization and expression studies have related to calmodulin but have distinct proper- proven difficult because of cross-reactivity of ties that allow them to carry out nonredundant antibodies, it has been established that certain roles that do not overlap with the functions of neurons express several or all of the NCS pro- calmodulin. Members of the NCS protein teins, but in general, the expression profile for family have been implicated in the regulation each of the NCS proteins is unique (Paterlini of neurotransmitter release, regulation of cell- et al. 2000; Rhodes et al. 2004). This suggests surface receptors and ion channels, control of that despite the high be- gene transcription, cell growth, and survi- tween the proteins, each is likely to perform dis- val (Burgoyne 2007). The NCS proteins are tinct functions in specific cell types (Burgoyne encoded by a family of 14 genes in mammals and Weiss 2001). with greater diversity stemming from alterna- Unlike calmodulin, not all EF-hands are tive splicing of transcripts from a number of functional in the NCS proteins and the most the genes. All NCS gene products harbor four amino-terminal EF-hand is unable to bind EF-hand motifs and display limited similarity Ca2þ in all family members. In the case of (,20%) to calmodulin (Burgoyne 2004). recoverin and KChIP1, only two of its four NCS-1 is the most widely expressed of the EF-hand motifs are functional in Ca2þ bind- NCS proteins and is thought to be the primor- ing (Burgoyne et al. 2004; Burgoyne 2007). dial NCS protein. The protein was first discov- Unlike the dumbbell structure of calmodulin, ered (as frequenin) in the NCS proteins are compact and globular (Pongs et al. 1993), where there are two very when Ca2þ-bound and they undergo limited

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 5 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

H.V. McCue, L.P. Haynes, and R.D. Burgoyne

conformational change on binding to their tar- motifs, which have differing cation specificities. get proteins (Ames et al. 2006; Pioletti et al. It has been suggested that under resting condi- 2þ 2006; Strahl et al. 2007; Wang et al. 2007) tions when [Ca ]i is low (0.1 mM), EF2 and (Fig. 2). NCS proteins also differ from calmo- EF3 are Mgþþ bound, whereas EF4 is a Ca2þ dulin in that many have motifs that allow mem- specific binding site and remains vacant. In brane association. KChIP1 and all the members the Mgþþ bound state, NCS-1 adopts a confor- of classes A–D are N-myristoylated, whereas mation, which reduces exposure of hydropho- certain KChIP2, KChIP3, and KChIP4 isoforms bic regions. This may be important in the harbor palmitoylation motifs. In some cases, the prevention of nonspecific interactions in the membrane association conferred by these moi- absence of a specific Ca2þ-signal. In the pres- 2þ 2þ eties is dynamically regulated by Ca binding ence of elevated [Ca ]i, EF2 and EF3 become when a sequestered mysristoyl chain becomes Ca2þ-occupied, simultaneously followed by solvent-exposed following a Ca2þ-driven shift Ca2þ binding to EF4 (Aravind et al. 2008). in conformation as originally described for The Mgþþ bound form of NCS-1 has a fivefold recoverin (Ames et al. 1997). VSNL proteins lower affinity for Ca2þ than the Mgþþ-free/ 2þ 2þ þþ are also cytosolic at resting [Ca ]i but localize Ca -free apo-form. This implies that Mg to the plasma membrane or Golgi complex binding permits significant modulation of upon Ca2þ elevation (O’Callaghan et al. 2002; NCS-1 and is important in fine tuning its Ca2þ- Spilker et al. 2002; O’Callaghan et al. 2003b). sensing properties (Aravind et al. 2008; Mikhay- Each of the NCS proteins displays distinct sub- lova et al. 2009). cellular localizations, which are in part deter- Much current understanding concerning mined by additional interactions with specific the function of NCS-1 derives from overexpres- phosphoinositides mediated by basic amino- sion or knockout studies. Overexpression in terminal residues immediately proximal to the Drosophila caused a frequency-dependent facil- site of acylation (O’Callaghan et al. 2003a; itation of neurotransmitter release (Pongs et al. O’Callaghan et al. 2005). 1993) and its importance for neurotransmis- NCS proteins are multifunctional regulators sions has been confirmed by knockout of the of various proteins involved in processes rang- two Drosophila frequenin genes (Dason et al. ing from trafficking and modula- 2009). In Xenopus, overexpression caused en- tion to gene transcription (Burgoyne 2004), hanced spontaneous and evoked transmission at and the function of NCS-1 in particular has neuromuscular junctions (Olafsson et al. 1995) been intensively studied. NCS-1, the primordial and over-expression was also found to increase NCS protein, is highly evolutionarily conserved, Ca2þ-dependent exocytosis of dense core gran- retaining 59% identity with its yeast ortholog, ules in PC12 cells (McFerran et al. 1998) and to frequenin. It displays a high Ca2þ-binding affin- enhance associative learning and memory in ity and is able to respond to small fluctuations Caenorhabditis elegans (Gomez et al. 2001; Hil- 2þ in [Ca ]i. NCS-1 is amino-terminally myris- fiker 2003). toylated and is constitutively associated with Knockout of NCS-1 (Frq1) in the yeast Sac- membranes including plasma and Golgi mem- charomyces cerevisiae is lethal because of its branes (O’Callaghan et al. 2002), although in requirement for the activation of Pik1, one of some cell lines, NCS-1 has been found to be par- the two yeast phosphatidylinositol-4 kinases tially cytosolic (de Barry et al. 2006) and it is (PI4Ks) (Hendricks et al. 1999). NCS-1 can able to rapidly exchange between membrane also interact with the mammalian Golgi and cytosolic pools (Handley et al. 2010). In PI4KIIIb and enhances its activity three- to contrast to all other NCS family members, 10-fold (Taverna et al. 2002; Haynes et al. NCS-1 is not specific and is expressed 2005; de Barry et al. 2006). The interaction in neuroendocrine cells (McFerran et al. 1998) with Golgi-associated PI4KIIIb suggests that it and at low levels in several nonneuronal cell may regulate secretion through the modulation types. NCS-1 has three functional EF-hand of phosphatidylinositol-dependent trafficking

6 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

Calcium Sensor Proteins in Neuronal Function

steps (Hendricks et al. 1999; Zhao et al. 2001; specific for NCS-1 (Haynes et al. 2006). Various Haynes et al. 2005). In support of this, NCS-1 studies have implicated NCS-1 in the regulation has also been demonstrated to associate with an- of VGCCs (Weiss et al. 2000; Tsujimoto et al. other PI4KIIIb regulator ARF1, a small GTPase 2002; Dason et al. 2009) but there is as yet no critical to multiple trafficking steps in mam- evidence for a direct interaction. Interestingly, malian cells (Haynes et al. 2005; Haynes et al. in Drosophila, the effects of NCS-1 on both neu- 2007). rotransmission and nerve-terminal growth can Knockout of NCS-1 in other organisms is be explained by a functional interaction with not lethal but does generate specific phenotypes. the VGCC cacophony, which is related to the In Dictyostelium discoideum, loss of NCS-1 func- mammalian P/Q-type VGCCs (Dason et al. tion alters developmental rate (Coukell et al. 2009). In contrast, in this study, there was no 2004) and in C. elegans results in impaired learn- evidence for an essential functional interaction ing and memory (Gomez et al. 2001). Knock- with the fly PI4KIIIb ortholog four wheel drive. downof one of the twoNCS-1 genes in zebrafish, The example of NCS-1 illustrates how ncs-1, prevents formation of the semicircular evolutionary pressures have fine-tuned Ca2þ canals of the inner ear (Blasiole et al. 2005). sensors to carry out specialized neuronal func- The signaling pathway involving NCS-1, ARF1, tions. The individual properties of NCS-1 allow and PI4KIIIb (Haynes et al. 2005) modulates this protein to localize to discrete domains the secretion of components important for the within the cell and interact with distinct target development of the vestibular apparatus of the proteins under conditions of Ca2þ stimulation, inner ear (Petko et al. 2009). Knockdown of which would not activate the archetypal Ca2þ NCS-1 or expression of a dominant–negative sensor, calmodulin. Although NCS-1 has been inhibitor based on an EF-hand mutation (Weiss found not to be neuronal specific and may et al. 2000) disrupted the induction of long- carry out more generalized functions con- term depression in rat cortical neurons (Jo et al. served through evolution in organisms from 2008). yeast onwards, further adaptive mutations Many different binding partners have been have given rise to many more members of the identified for NCS-1 (Haynes et al. 2006; Hay- NCS protein family, each tasked with dedicated nes et al. 2007) (Fig. 3) and in some cases these functions. interactions overlap with those of calmodulin Much less is known about the VSNL or class (Schaad et al. 1996). This overlap using in vitro B proteins, although these appear to modulate binding assays may not be physiologically various pathways such as meaningful because of substantial lower affin- cyclic nucleotide and MAPK signaling (Braune- ity of NCS-1 for known calmodulin targets well and Klein-Szanto 2009). VILIP-1 has been (Schaad et al. 1996; Fitzgerald et al. 2008). found to regulate a class of purinergic receptors NCS-1 has a higher affinity for calcium than (Chaumont et al. 2008). They have been shown calmodulin and therefore may preferentially to have effects on gene expression and are also interact with certain binding partners when involved in traffic of proteins to the plasma the amplitude of a Ca2þ-signal falls below the membrane (Lin et al. 2002; Brackmann et al. threshold for activation of calmodulin. For 2005). Hippocalcin has been suggested to be example, both calmodulin and NCS-1 have involved as a Ca2þsensor in long term depres- been shown to interact with and desensitize sion in hippocampal neurons (Palmer et al. dopamine D2 receptors but are likely to mediate 2005) and shows a Ca2þ/myristoyl switch for 2þ their effects at different [Ca ]i (Kabbani et al. translocation within such neurons (Markova 2002; Woods et al. 2008). Functional analyses et al. 2008). It has also been implicated in pro- have confirmed that NCS-1 is a regulator of tection from neuronal (Mercer et al. D2 receptors and that this function modulates 2000; Korhonen et al. 2004). learning in mice (Saab et al. 2009). Other Recoverin is expressed exclusively in the NCS-1 target proteins (Fig. 3) appear to be retina and is believed to have a role in light

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 7 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

H.V. McCue, L.P. Haynes, and R.D. Burgoyne

MLK2 α4β2 Nic R NAIP – + – P2Y2R – GC-D + GC-B GC-E Hippo VILIP-1 GluR6 – + + + + – + + GC-A CAPS δ – Neuro + Alsin GRK2 D2R + – – + + + + IL1RAPL β + – TGF R1 NCS-1 + + – Kv4.2 + + S100β 2+ + + Ca -dependent + AP2 PICK1 + + 2+ AP1 Jacob – Ca -independent – + – – β – – PI4KIII ARF1 + MAP1/LC3 CaBP – – CaV1.2 + – + V-ATPase PDE VILIP-2 IP3R – CaV2.1 TRPC5 + – – – +

+ Calc1neurin Calmodulin

Calmodulin-specific targets

Figure 3. An interaction map showing protein–protein interactions made by some NCS proteins and CaBP1 compared to calmodulin. Known protein interactions for CABP1, hippocalcin, NCS-1, neurocalcin d,VILIP1, and VILIP2. Links indicate where these target proteins have also been found to interact with calmodulin. It is also indicated whether these interactions require the Ca2þ-bound form of the protein or not.

and can enhance visual sensitivity Klenshin et al. 1995). The function of recoverin (Polans et al. 1996; Sampath et al. 2005). Recov- has been controversial and this hypothesis may erin is found primarily in rod and cone cells of be oversimplified. Discrepancies have been 2þ the retina (Yamagata et al. 1990; Dizhoor et al. noted regarding the [Ca ]i required for rho- 1991). Recoverin was predicted to prolong the dopsin interaction, which may lie lifetime of photolyzed by inhibiting outside normal physiological limits but anal- its phosphorylation by to ysis of recoverin knockout mice have shown extend the light response (Chen et al. 1995; changes in photoresponses consistent with a

8 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

Calcium Sensor Proteins in Neuronal Function

physiological role in inhibition of rhodopsin Burns et al. 2002; Howes et al. 2002; Pennesi kinase (Makino et al. 2004). et al. 2003). They are unusual in that they acti- The structure of recoverin has been exten- vate GCs when in their Ca2þ-free form but sively studied by X-ray crystallography and become inhibitors of GCs at higher Ca2þ con- NMR studies to interrogate its structure in its centrations (Dizhoor and Hurley 1996). GCAP3 Ca2þ-bound and -free forms (Flaherty et al. is expressed in cone cells, whereas GCAP1 and 1993; Ames et al. 1995; Tanaka et al. 1995; GCAP2 are expressed in rod cells, and despite Ames et al. 1997; Ames et al. 2002; Weiergraber GCAP1 and GCAP2 having the same function et al. 2003). Recoverin is composed of two dis- in the same cell type, the two proteins have dif- tinct domains connected through a bent linker ferent Ca2þ binding affinities for GC activation. and forms a compact structure in the absence This means that both proteins are required for of Ca2þ. Unlike other NCS proteins, recoverin GC activation over the full physiological Ca2þ has only two functional EF-hand motifs. Upon concentration range, thus maximizing the dy- binding of Ca2þ, the amino-terminal domain namic range of GC activity (Koch 2006). The comprising EF-1 and EF-2 rotates through GCAPs are an example of how calcium sensors 458 relative to the carboxy-terminal domain have become adapted to increase the dynamic driving extrusion of its buried myristoyl group Ca2þ sensitivity of important regulatory mech- to permit association with membranes and anisms in specialized cell types (Palczewski et al. revealing a hydrophobic surface, which can me- 2004). diate interaction with the target protein rho- KChIPs have been found to associate with dopsin kinase (Ames et al. 2006). The residues transient voltage-gated potassium channels of involved in the interaction of the myristoyl the Kv4 family (An et al. 2000) and can stimu- group with the hydrophobic pocket are also late their traffic to the plasma membrane (Has- conserved in the other members of the NCS demir et al. 2005). Four KChIP genes and a large family, however not all of the other family number of expressed splice variants are present members display this Ca2þ/myristoyl switch in mammals (Pruunsild and Timmusk 2005). (O’Callaghan et al. 2002; Stephen et al. 2007). Knockout of KChIP1 has revealed a potential NCS-1 and KChIP1 expose a similar hydropho- role in the GABAergic inhibitory system (Xiong bic surface upon Ca2þ-binding, which could et al. 2009). The KChIPs are expressed predom- be similarly important for target interactions inantly in the brain but KChIP2 is also expressed (Bourne et al. 2001; Scannevin et al. 2004; Zhou in the heart, and knockout of KChIP2 causes a et al. 2004b; Pioletti et al. 2006). In contrast, complete loss of calcium-dependent transient other NCS proteins are able to interact with outward potassium currents and susceptibility certain binding proteins in the absence of Ca2þ to ventricular tachycardia (Kuo et al. 2001). and therefore Ca2þ-driven exposure of a hydro- KChIP3 is also known as DREAM or , phobic surface cannot be the sole mechanism and has documented roles in transcriptional by which these proteins bind to effectors. Al- regulation (Carrion et al. 1999; Mellstrom and though extensive structural characterization of Naranjo 2001) and in the processing of preseni- recoverin may go some way to inform an un- lins and amyloid precursor protein, which are derstanding of the general conserved structures important in the pathogenesis of Alzheimer’s of members of the NCS family, subtle dif- disease (Buxbaum et al. 1998; Jo et al. 2004). ferences in “active” surface residues of the indi- Despite KChIP3 being implicating in three vidual proteins gives rise to their ability to quite different functions, it is likely that these interact specifically with awide range of binding are all physiologically relevant. KChIP3 knock- partners. out mice show reduced responses in acute GCAPs are the only known activators of ret- pain models because of changes in prodynor- inal guanylyl cyclases (GCs) (Palczewski et al. phin synthesis (Cheng et al. 2002), decreased b- 2004) and are known to be physiological regula- amyloid production, and physiological defects tors of light adaptation (Mendez et al. 2001; consistent with changes to the Kv4 channels

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 9 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

H.V. McCue, L.P. Haynes, and R.D. Burgoyne

2þ (Lilliehook et al. 2003). Although many of the changes in [Ca ]i would depend on which KChIPs and their isoforms may have overlapp- populations of calcium binding proteins are ing functions, some differences between them activated under particular conditions (Bur- are beginning to emerge (Holmqvist et al. goyne and Weiss 2001). As mentioned previ- 2002; Venn et al. 2008). ously, the individual expression patterns and In support of key roles for the NCS family in subcellular localization of each of the NCS pro- higher organisms, a number of recent studies teins is also likely to represent a key factor in have implicated these proteins in the patholog- their specific roles in neuronal . ical progression of human neurological diseases The characteristic amino-terminal myristoyla- in addition to the potential link with Alz- tion or palmitoylation modifications, which heimer’s disease via the interaction of KChIP3 allow these proteins to associate with mem- with presenilins. VILIP1 may have a role in Alz- branes, may spatially partition them to relevant heimer’s disease because of an association with subcellular sites within the cell, leading to a amyloid plaques in diseased (Schnurra faster and more efficient response to particular et al. 2001). NCS-1 has been found to be Ca2þ signals. Specific physiological outcomes up-regulated in patients with schizophrenia will be determined by their distinct target pro- and bipolar disorders (Koh et al. 2003) and teins. The various members of the NCS family also interacts with interleukin-1 acces- arose at points in evolution corresponding to sory protein-like (IL1RAPL), a protein, which increasing neuronal sophistication in higher when mutated results in X-linked mental retar- animals. As such, these proteins represent an dation (Bahi et al. 2003). The effects conferred example of how the properties of calcium bind- by NCS proteins in all of these diseases would ing proteins have been fine-tuned to act in spe- appear to be dependent on the up- or down- cific neuronal signaling pathways. regulation of their expression. As yet few genetic links have been established between NCS pro- CaBP Family teins and the aforementioned diseases, suggest- ing epigenetic effects may be responsible. One The CaBPs are a relatively recently discovered idea is that epigenetic mediated alterations in family of EF-hand containing Ca2þ-binding NCS protein function may contribute to cogni- proteins, which are only found in vertebrates tive impairments observed in neurodegenera- (Haeseleer et al. 2000). They represent another tive states. Targeting of NCS protein function example of a diverse family of Ca2þ-sensors through this novel route may offer a future ther- capable of regulating discrete processes in the apeutic approach for such diseases (Braunewell nervous systems of higher organisms. The 2005). CaBPs share sequence homology with calmo- The NCS protein family has evolved to carry dulin and also display a similar structural out specialized neuronal functions that are sep- arrangement of EF-hand motifs. Each of the arate to those of calmodulin. When attempt- CaBPs has four EF hands, although, like the ing to decipher precisely why these proteins are NCS proteins, they display different patterns of particularly well adapted for carrying out func- EF-hand inactivation (Fig. 4). In CaBPs 1–5, tions in neurons, it is therefore relevant to com- the second EF-hand motif is inactive with the pare their properties to those of calmodulin. Of exception of CaBP3, which also has an inactive note is their approximately 10-fold higher affin- EF-1 motif. CaBP3 is believed to represent a ity for Ca2þ when compared to calmodulin. pseudogene as only the mRNA has been de- This higher affinity would allow the NCS tected in cells and as yet no protein product proteins to be activated at much lower Ca2þ has been found (Haeseleer et al. 2000). Twopro- concentrations and, in combination with calm- teins were named CaBP7 and CaBP8 (Haeseleer odulin, extends the dynamic range over which et al. 2002), but bioinformatic analysis is more Ca2þ can regulate neuronal processes. In this consistent with them being a conserved and dis- way, responses to very slight or more dramatic tinct subfamily of CaBPs (McCue et al. 2010).

10 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

Calcium Sensor Proteins in Neuronal Function

EF1 EF2 EF3 EF4 Calmodulin

EF1 EF2 EF3 EF4 Caldendrin

EF1 EF2 EF3 EF4 CaBP1/2

EF1 EF2 EF3 EF4 CaBP3

EF1 EF2 EF3 EF4 CaBP4/5

EF1 EF2 EF3 EF4 Calneurons 1/2

Figure 4. Schematic diagram showing the domain structure of calmodulin and members of the CaBP/calneuron protein family. Active EF-hand motifs are shown in red and inactive EF-hand motifs are shown in pink. Compared to calmodulin, the CaBPs have an extended linker region between their first EF-hand pair and their second EF-hand pair (shown in black). CaBP1 and CaBP2 have an N- site (shown in blue). CaBP1 and CaBP2 have alternative splice sites at their N-terminus, which give rise to long and short isoforms (shown in orange). Calneurons 1 and 2 possess a 38 amino acid extension at their C-terminus (shown in purple).

We will therefore refer to them by their alterna- the Golgi and also displays some cytosolic local- tive names, calneuron 2 and calneuron 1, re- ization, whereas CaBP1-Short localizes most spectively (Wu et al. 2001; Mikhaylova et al. prominently to the plasma membrane and to 2006). The calneurons, by contrast to the CaBPs, Golgi structures (Haeseleer et al. 2000; McCue have a different pattern of EF-hand inactivation et al. 2009). Alternative splicing of the CaBP1 with active EF-hands 1 and 2 and inactive EF- genegeneratesathirdproteinproduct,caldendrin hands 3 and 4 (Mikhaylova et al. 2006). The (Seidenbecher et al. 1998). This splice isoform is CaBPs also differ from calmodulin in that their significantly larger than either CaBP1-Long or central a helical linker domain connecting the CaBP1-Short because of an amino-terminal carboxy- and amino-terminal EF-hand pairs extension, but caldendrin mRNA lacks the exon is extended by four amino acid residues. This required for N-myristoylation and as a result the has been suggested to allow these proteins to protein displays a markedly different subcellular interact with unique targets (Haeseleer et al. localization to its shorter relatives. 2000). N-terminal acylation is important in the A major difference compared with calmo- localization of some CaBPs, but the calneurons dulin is the ability of CaBP 1 and 2 and calneu- appear to be targeted via a different mechanism. rons 1 and 2 to target to specific cellular Like CaBP1 and CaBP2, calneurons 1 and 2 membranes (McCue et al. 2009). CaBP 1 and 2 localize to internal membranes that colabel are amino-terminally myristoylated, which per- with Golgi-specific markers and also to vesicu- mits localization to the plasma membrane and lar structures (McCue et al. 2009; Mikhaylova Golgi apparatus (Haeseleer et al. 2000; Haynes et al. 2009). Calneurons 1 and 2 do not possess et al. 2004). The precise amino-terminal se- an amino-terminal myristoylation motif and quence to which the myristoyl group is attached differ from the rest of the CaBP family because is also important in the targeting of these two of a 38-amino acid extension at their carboxyl proteins, as exemplified by the long and short terminus. Analysis of this sequence revealed a splice isoforms generated from their genes, predicted C-terminal transmembrane domain which show subtle differences in their localiza- with a cytosolic amino terminus. The carboxy- tion. CaBP1-Long localizes predominantly to terminal domain resembles tail-anchor motifs

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 11 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

H.V. McCue, L.P. Haynes, and R.D. Burgoyne

and directly localizes calneurons 1 and 2 to same target molecules. For instance, both membranes particularly of the trans-Golgi net- CaBP1 and calmodulin bind to L-type Ca2þ work (McCue et al. 2009). channels with calmodulin causing Ca2þ-in- To date, only the structure of CaBP1-Short duced channel closure but CaBP1 promoting has been solved (Wingard et al. 2005; Li et al. channel opening (Zhou et al. 2004a; Zhou 2009). This structural information may provide et al. 2005). Both calmodulin and CaBP1 also insight into the structures of the rest of the regulate inositol 1,4,5-trisphosphate receptors CaBPs. Analogy to calmodulin would suggest (IP3Rs) (Yang et al. 2002; Haynes et al. 2004; that the CaBPs should adopt a dumbbell like Kasri et al. 2004) with CaBP1 binding the type tertiary conformation consisting of an amino- IIP3R with 100-fold higher affinity than calm- terminal domain containing EF-1 and EF-2 odulin. This high affinity binding may result and a carboxy-terminal domain containing from the exposure of a distinct hydrophobic EF-3 and EF-4, connected by a central linker. patch revealed in the carboxyl terminus of NMR analysis revealed that CaBP1 does in- CaBP1 upon Ca2þ-binding (Haynes et al. deed have two independent, noninteracting do- 2004; Li et al. 2009). This unique surface hydro- mains, joined by a flexible linker (Wingard et al. phobicity profile is likely to be important for the 2005). Investigation into the effects of Mgþþ specialization of CaBP1 function in the brain and Ca2þ binding has shown that as predicted and retina, and the existence of splice isoforms the second EF hand of CaBP1 is incapable of is also likely to further fine-tune the actions of binding divalent cations. EF-3 and EF-4 bind this Ca2þ sensor. The differing expression pat- to both Mgþþ and to Ca2þ, whereas EF-1 is terns, subcellular targeting mechanisms, and thought to be constitutively Mgþþ bound. Ca2þ binding properties of the various mem- Binding to either Mgþþ or Ca2þ induces dis- bers of the CaBP protein family would allow tinct conformations of this protein. Mgþþ them to carry out highly specialized regulatory binding results in a global conformational roles modulating important Ca2þ-channels in change, whereas Ca2þ binding results in only a the central nervous system. localized change in EF-3 and EF-4. These two The majority of studies to date on CaBP1 conformational states may allow CaBP1 to have examined the functions of the longest interact with different target molecules driven splice isoform caldendrin and it is not yet clear by the ratio of Mgþþ to Ca2þ (Wingard et al. whether the other splice isoforms of CaBP1 can 2005; Li et al. 2009). The Mgþþ bound form carry out the same functions. CaBP1-Long and of CaBP1 is similar to that of apo-calmodulin, -Short have been found to have roles in the reg- but the Ca2þ bound form appears markedly dif- ulation of various Ca2þ-channels including P/ ferent. This is perhaps unsurprising as neither Q-type (CaV2.1) channels (Lee et al. 2002), of the amino-terminal EF-hands of CaBP1 L-type (CaV1.2) channels (Zhou et al. 2005; bind to Ca2þ under saturating conditions and Cui et al. 2007), TRPC5 channels (Kinoshita- þþ only EF-1 binds to Mg . This results in a con- Kawada et al. 2005), and IP3Rs (Yang et al. stitutively closed conformation of the amino- 2002), which they apparently inhibit (Haynes terminal domain, whereas the carboxy-terminal et al. 2004; Kasri et al. 2004). The interaction domain can switch to an open conformation of CaV2.1 with CaBP1 appears to rely acutely upon Ca2þ binding to EF-3 and EF-4. Compar- upon amino-terminal myristoylation. Wild ison of the carboxy-terminal domain with that type, myristoylated, CaBP1-Long enhances of calmodulin, however, still reveals differences channel inactivation and shifts the activation in exposed hydrophobic residues thought to range to more depolarizing voltages (Lee et al. mediate target interactions (Wingard et al. 2002). An N-myristoylation mutant, however, 2005). was unable to mediate these effects and instead The structural differences between calmo- modulated channels in a similar fashion to dulin and CaBP1 may go some way to explain- calmodulin (Few et al. 2005). Differential mod- ing how they impose differing effects on the ulation of L-type channels depending on the

12 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

Calcium Sensor Proteins in Neuronal Function

splice isoform of CaBP1 has also been observed. and suppress calcium-dependent inactivation CaBP1-Short has been shown to completely of CaV1.2 channels (Rieke et al. 2008). inhibit inactivation of CaV1.2 channels (Zhou CaBP4 is the most extensively characterized et al. 2005), but caldendrin causes a more mod- of the CaBP family. It is expressed in the retina, est suppression and signals through a different where it localizes to synaptic terminals and has set of molecular determinants (Tippens and also been detected in auditory inner hair cells. Lee 2007). This suggests that the subcellular CaBP4 modulates voltage gated Ca2þ-channels localization of CaBP1 splice variants is impor- and directly associates with the carboxyl termi- tant for their function and there are likely to nus of the CaV1.4 a1 pore-forming subunit, be individual roles for each protein. Interactions shifting the activation range of the channel to of caldendrin with other types of proteins have more hyperpolarized voltages in transfected also been reported, such as its interaction with cells (Haeseleer et al. 2004). CaBP4 has also light chain 3 of MAP1A/B, a microtubule been shown to eliminate Ca2þ-dependent inac- cytoskeletal protein (Seidenbecher et al. 2004), tivation of CaV1.3 channels, which is likely to be and an interaction with myo1c, a member of important in the modulation of these channels the -1 family of motor proteins (Tang in inner hair cells, where Ca2þ-dependent inac- et al. 2007). Finally, a role for caldendrin in tivation is weak or absent, probably allowing NMDA receptor (NMDAR) signaling has been the audition of sustained sounds (Yang et al. reported via an interaction with a novel neuro- 2006). A stronger inhibitory effect has been nal protein, Jacob. Upon extrasynaptic NMDAR noted for CaBP1, however, suggesting that activation, Jacob translocates to the nucleus to CaBP4 may not be the key Ca2þ sensor involved influence CREB activity, resulting in the strip- in this process (Cui et al. 2007). The function of ping of synaptic contacts and an associated sim- CaBP4 is modulated by C z in the plification of dendritic architecture. Synaptic retina, with increased CaBP4 phosphorylation 2þ NMDAR mediated synpatodendritic [Ca ]i in light-adapted tissue. Phosphorylation pro- 2þ elevation induces caldendrin binding to Jacob, longs Ca currents through CaV1.3 channels inhibiting nuclear trafficking and maintaining and suggests that light-stimulated phosphoryla- dendritic organization. This interaction repre- tion of CaBP4 might help to regulate presynap- sents a novel mechanism of to nucleus tic Ca2þ signals in photoreceptors (Lee et al. communication and highlights the important 2007). CaBP4 has also been implicated in neu- roles of CaBP family members in the mamma- rotransmitter release at synaptic terminals lian central nervous system (Dieterich et al. because of its interaction with unc119, a synap- 2008). tic photoreceptor protein important for neuro- Little information is available concerning transmitter release and maintenance of the the function of CaBP2 apart from in vitro stud- nervous system (Haeseleer 2008). Knockout of ies suggesting that it might stimulate CaMK CaBP4 results in mice with abnormalities in activity (Cui et al. 2007). Initially, CaBP2 was retinal function, where rod bipolar responses detected exclusively in the retina, although it are approximately 100 times lower than those has also been identified in auditory inner hair observed in wild-type animals (Haeseleer et al. cells (Cui et al. 2007). CaBP5 was also detected 2004). in inner hair cells as well as in the retina, but in The functions of calneurons 1 and 2 have contrast to CaBP2, was found to have a modest only recently begun to be investigated in detail. inhibitory effect on the inactivation of CaV1.3 Both have been found to inhibit the activity of 2þ channels in transfected cells (Cui et al. 2007). PI4KIIIb at low or resting [Ca ]i. Overexpres- Little is known about the functions of CaBP5, sion of the proteins was also found to inhibit but knockout mice displayed reduced sensitiv- Golgi-to-plasma membrane trafficking, caused ity of retinal ganglion cells to light responses, enlargement of the trans-Golgi network (TGN), implicating CaBP5 in phototransduction path- and reduced the number of Piccolo-Bassoon ways. CaBP5 was also found to interact with positive transport vesicles. A molecular switch

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 13 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

H.V. McCue, L.P. Haynes, and R.D. Burgoyne

for the production of phosphoinositides at the underlie cone-rod synaptic disorders (Littink TGN is thought to be created by the opposing et al. 2009). roles of NCS-1 and calneurons 1 or 2. At ele- vated Ca2þ levels, NCS-1 preferentially binds to PI4KIIIb over the calneurons, activating CONCLUDING REMARKS the enzyme to drive enhanced TGN-to-plasma It has become increasingly clear that a full un- membrane trafficking (Mikhaylova et al. derstanding of how specific aspects of neuronal 2009). Patch clamping experiments have shown function are regulated in response to spatially that over-expressed calneuron 1 can inhibit N- 2þ 2þ and temporally distinct Ca signals will require type Ca -channel currents in 293T cells and a detailed knowledge of the Ca2þ sensors this inhibition was not observed with a trun- involved. Some of these, like synaptotagmin, cated calneuron 1 lacking its hydrophobic C- are specialized for particular neuronal func- terminus, suggesting normal localisation is tions, whereas others such as calmodulin may important in carrying out this function (Shih be involved in multiple cellular processes. In et al. 2009). recent years, much has been learnt about the CaBPs have been directly or indirectly im- functions of the NCS family of Ca2þ sensors, plicated in multiple neuronal diseases. Post- although the functions of some of the family mortem brains of chronic schizophrenics have members are still unknown. Nor is it clear lower numbers of caldendrin-immunoreactive what the significance is of the multiple genes neurons, which express the protein at a much and splice variants of these proteins. The CaBPs higher level. This loss of caldendrin in some have so far been much less studied and much neurons and up-regulation in others is likely remains to be learnt about the functions of to profoundly change synapto-dendritic sig- each of these sensors. Further advances will nalling in schizophrenic patients (Bernstein require new insights into the molecular targets et al. 2007). Changes in the distribution of cal- of each of the Ca2þ sensors, the molecular basis dendrin have also been observed in kainate- for their regulation of these targets, and more induced epileptic seizures in rats. Caldendrin detailed dissection of the functional roles of translocates to the postsynaptic density only each protein in identified neurons. in rats that suffered epileptic seizures and may implicate the protein in the pathophysiology of the disease (Smalla et al. 2003). CaBP4 func- ACKNOWLEDGMENTS tion has been convincingly linked to disease and mutations in this gene generate defects HVM was supported by a Wellcome Trust Prize in retinal function. Knockout of CaBP4 was PhD Studentship. shown to cause a phenotype similar to that of incomplete congenital stationary night blind- REFERENCES ness patients (Haeseleer et al. 2004) and muta- tions in CaBP4 can cause autosomal recessive Ames JB, Hamashima N, Molchanova T. 2002. Structure night blindness (Zeitz et al. 2006). Patients and calcium-binding studies of a recoverin mutant (E85Q) in an allosteric intermediate state. Biochemistry with mutations in the CaBP4 gene have been 41: 5776–5787. identified, which display congenital stationary Ames JB, Ishima R, Tanaka T, Gordon JI, Stryer L, Ikura M. night blindness. However, some patients with 1997. Molecular mechanics of calcium-myristoyl mutations display different phenotypes (Zeitz switches. Nature 389: 198–202. Ames JB, Levay K, Wingard JN, Lusin JD, Slepak VZ. 2006. et al. 2006). In particular, a novel homozy- Structural basis for calcium-induced inhibition of rho- gous nonsense mutation has been reported in dopsin kinase by recoverin. J Biol Chem 281: 37237– two siblings that resulted in severely reduced 37245. cone function but only negligible effects on Ames JB, Tanaka T, Ikura M, Stryer L. 1995. Nuclear mag- netic resonance evidence for Ca2þ-induced extrusion rod function (Littink et al. 2009). It appears, of the myristoyl group of recoverin. J Biol Chem 270: therefore, that genetic mutations in CaBP4 30909–30913.

14 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

Calcium Sensor Proteins in Neuronal Function

An WF, Bowlby MR, Bett M, Cao J, Ling HP, Mendoza G, Burgoyne RD, O’Callaghan DW, Hasdemir B, Haynes LP, Hinson JW, Mattsson KI, Strassle BW, Trimmer JS, TepikinAV.2004. Neuronal calcium sensor proteins: Mul- et al. 2000. Modulation of A-type potassium channels titalented regulators of neuronal function. Trends Neuro- by a family of calcium sensors. Nature 403: 553–556. sci 27: 203–209. Aravind P, Chandra K, Reddy PP, Jeromin A, Chary KV, Burns ME, Mendez A, Chen J, Baylor DA. 2002. Dynamics of Sharma Y. 2008. Regulatory and structural EF-hand cyclic AMP synthesis in retinal rods. Neuron 36: 81–91. 2þ motifs of neuronal calcium sensor-1: Mg modulates Buxbaum JD, Choi EK, Luo YX, Lilliehook C, Crowley AC, 2þ 2þ Ca binding, Ca -induced conformational changes, Merriam DE, Wasco W. 1998. Calsenilin: A calcium- and equilibrium unfolding transitions. J Mol Biol 376: binding protein that interacts with the presenilins and 1100–1115. regulates the levels of a presenilin fragment. Nature Med- Augustine GJ, Santamaria F, Tanaka K. 2003. Local calcium icine 4: 1177–1181. signaling in neurons. Neuron 40: 331–346. Carrion AM, Link WA, Ledo F, Mellstrom B, Naranjo JR. Bahi N, Friocourt G, Carrie´ A, Graham ME, WeissJL, Chafey 1999. DREAM is a Ca2þ-regulated transcriptional P,Fauchereau F, Burgoyne RD, Chelly J. 2003. IL1 recep- repressor. Nature 398: 80–84. tor accessory protein like, a protein involved in X-linked Catterall WA,Few AP.2008. regulation and mental retardation, interacts with Neuronal Calcium presynaptic plasticity. Neuron 59: 882–901. Sensor-1 and regulates exocytosis. Human Mol Genetics Chapman ER. 2008. How does synaptotagmin trigger neu- 12: 1415–1425. rotransmitter release? Annu Rev Biochem 77: 615–641. Barria A, Muller D, Derkach V, Griffith LC, Soderling TR. Chattopadhyaya R, Meador WE, Means AR, Quiocho FA. 1997. Regulatory phosphorylation of AMPA-type gluta- 1992. Calmodulin structure refined at 1.7 A resolution. mate receptors by CaM-KII during long-term potentia- J Mol Biol 228: 1177–1192. tion. Science 276: 2042–2045. Chaumont S, Compan V,Toulme E, Richler E, Housley GD, Bernstein HG, Sahin J, Smalla KH, Gundelfinger ED, Rassendren F,Khakh BS. 2008. Regulation of P2X2 recep- Bogerts B, Kreutz MR. 2007. A reduced number of cort- tors by the neuronal calcium sensor VILIP1. Sci Signal 1: ical neurons show increased Caldendrin protein levels in ra8. chronic schizophrenia. Schizophr Res 96: 246–256. Chen CK, Inglese J, Lefkowitz RJ, Hurley JB. 1995. Ca2þ- Berridge MJ. 1998. Neuronal calcium signalling. Neuron 21: dependent interaction of recoverin with rhodopsin kin- 13–26. ase. J Biol Chem 270: 18060–18066. Berridge MJ. 2010. Calcium hypothesis of Alzheimer’s dis- Cheng H-YM, Pitcher GM, Laviolette SR, Whishaw IQ, ease. Pflugers Arch 459: 441–449. Tong KI, Kockeritz LK, Wada T, Joza NA, Crackower M, 2þ Bito H, Deisseroth K, Tsien RW.1997. Ca -dependent reg- Goncalves J, et al. 2002. DREAM is a critical transcrip- ulation in neuronal gene expression. Current Opinion in tional repressor for pain modulation. Cell 108: 31–43. Neurobiol 7: 419–429. Chicka MC, Hui E, Liu H, Chapman ER. 2008. Synaptotag- Blasiole B, Kabbani N, Boehmler W,Thisse B, Thisse C, Can- min arrests the SNARE complex before triggering fast, field V, Levenson R. 2005. Neuronal calcium sensor-1 efficient membrane fusion in response to Ca2þ. Nat gene ncs-1 is essential for semicircular canal formation Struct Mol Biol 15: 827–835. in zebrafish inner ear. J Neurobiol 64: 285–297. Coukell B, Cameron A, Perusini S, Shim K. 2004. Disruption Bourne Y, Dannenberg J, Pollmann V, Marchot P, Pongs O. of the NCS-1/frequenin-related ncsA gene in Dictyoste- 2001. Immunocytochemical localisation and crystal lium discoideum accelerates development. Develop structure of human frequenin (neuronal calcium sensor Growth Differ 46: 449–458. 1). J Biol Chem 276: 11949–11955. Craxton M. 2004. Synaptotagmin gene content of the Brackmann M, Schuchmann S, Anand R, Braunewell KH. sequenced genomes. BMC Genomics 5: 43. 2þ 2005. Neuronal Ca sensor protein VILIP-1 affects Cui G, Meyer AC, Calin-Jageman I, Neef J, Haeseleer F, cGMP signalling of guanylyl cyclase B by regulating 2þ Moser T,Lee A. 2007. Ca2þ-binding proteins tune Ca - clathrin-dependent receptor recycling in hippocampal feedback to Cav1.3 channels in mouse auditory hair cells. neurons. J Cell Sci 118: 2495–2505. J Physiol 585: 791–803. Braunewell K-H. 2005. The darker side of Ca2þ signaling by Dason JS, Romero-Pozuelo J, Marin L, Iyengar BG, Klose neuronal Ca2þ-sensor proteins: From Alzheimer’s dis- MK, Ferrus A, Atwood HL. 2009. Frequenin/NCS-1 ease to cancer. Trends Pharmacol Sci 26: 345–351. and the Ca2þ-channel {a} 1-subunit co-regulate synaptic Braunewell KH, Klein-Szanto AJ. 2009. Visinin-like proteins transmission and nerve-terminal growth. J Cell Sci 122: (VSNLs): Interaction partners and emerging functions in 4109–4121. 2þ signal transduction of a subfamily of neuronal Ca de Barry J, Janoshazi A, Dupont JL, Procksch O, Chasserot- -sensor proteins. Cell Tissue Res 335: 301–316. Golaz S, Jeromin A, Vitale N. 2006. Functional implica- Burgoyne RD. 2004. The neuronal calcium-sensor proteins. tion of neuronal calcium sensor-1 and PI4 kinase-b Biochim Biophys Acta 1742: 59–68. interaction in regulated exocytosis of PC12 cells. J Biol Burgoyne RD. 2007. Neuronal calcium sensor proteins: Chem 281: 18098–18111. 2þ Generating diversity in neuronal Ca signalling. Nat de Wit H, Walter AM, Milosevic I, Gulyas-Kovacs A, Riedel Rev Neurosci 8: 182–193. D, Sorensen JB, Verhage M. 2009. Synaptotagmin-1 Burgoyne RD, Weiss JL. 2001. The neuronal calcium sensor docks secretory vesicles to syntaxin-1/SNAP-25 acceptor family of Ca2þ-binding proteins. Biochem J 353: 1–12. complexes. Cell 138: 935–946.

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 15 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

H.V. McCue, L.P. Haynes, and R.D. Burgoyne

Deisseroth K, Heist EK, Tsien RW. 1998. Translocation of Haeseleer F. 2008. Interaction and colocalization of CaBP4 calmodulin to the nucleus supports CREB phosphoryla- and Unc119 (MRG4) in photoreceptors. Investigative tion in hippocampal neurons. Nature 392: 198–202. Ophthalmol Visual Sci 49: 2366–2375. DeMaria CD, Soong TW, Alselkhan BA, Alvania RS, Yue Haeseleer F, Imanishi Y, Maeda T, Possin DE, Maeda A, Lee 2þ DT. 2001. Calmodulin bifurcates the local Ca2þ signal A, Rieke F, Palczewski K. 2004. Essential role of Ca - 2þ that modulates P/Q-type Ca channels. Nature 411: binding protein 4, a Cav1.4 channel regulator in photore- 484–489. ceptor synaptic function. Nature Neurosci 7: 1079–1087. Dieterich DC, Karpova A, Mikhaylova M, Zdobnova I, Haeseleer F, Imanishi Y, Sokal I, Filipek S, Palczewski K. Konig I, Landwehr M, Kreutz M, Smalla KH, Richter K, 2002. Calcium-binding proteins: Intracellular sensors Landgraf P, et al. 2008. Caldendrin-Jacob: A protein from the calmodulin superfamily. Biochem Biophys Res liaison that couples NMDA receptor signalling to the Comm 290: 615–623. nucleus. PLoS Biol 6: e34. Haeseleer F, Sokal I, Verlinde CLMJ, Erdjument-Bromage H, Tempst P, Pronin AN, Benovic JL, Fariss RN, Dizhoor AM, Hurley JB. 1996. Inactivation of EF-hands Palczewski K. 2000. Five members of a novel Ca2þ bind- makes GCAP-2 (p24) a constitutive activator of photore- ing protein (CABP) subfamily with similarity to calmo- ceptor guanulyl cyclase by preventing a Ca2þ-induced dulin. J Biol Chem 275: 1247–1260. “activator-to-inhibitor” transition. J Biol Chem 271: 19346–19350. Handley MT,Lian LY,Haynes LP,Burgoyne RD. 2010. Struc- tural and functional deficits in a Neuronal Calcium Dizhoor AM, Ray S, Kumar S, Niemi G, Spencer M, Brolley Sensor-1 mutant identified in a case of Autistic Spectrum D, Walsh KA, Philipov PP, Hurley JB, Stryer L. 1991. Disorder. PLoS ONE 5: e10534. Recoverin: A calcium sensitive activator of retinal rod Hasdemir B, Fitzgerald DJ, Prior IA, Tepikin AV, Burgoyne . Science 251: 915–918. RD. 2005. Traffic of Kv4 Kþ channels mediated by Fallon JL, Halling DB, Hamilton SL, Quiocho FA. 2005. KChIP1 is via a novel post-ER vesicular pathway. J Cell Structure of calmodulin bound to the hydrophobic IQ Biol 171: 459–469. domain of the cardiac Ca(v)1.2 calcium channel. Struc- Haynes LP, Tepikin AV, Burgoyne RD. 2004. Calcium Bind- ture 13: 1881–1886. ing Protein 1 is an inhibitor of agonist-evoked, inositol Fernandez-Chacon R, Konigstorfer A, Gerber SH, Garcia J, 1,4,5-trisphophate-mediated calcium signalling. J Biol- Matos MF, Stevens CF, Brose N, Rizo J, Rosenmund C, Chem 279: 547–555. Sudhof TC. 2001. Synaptotagmin I functions as a calcium Haynes LP,Thomas GMH, Burgoyne RD. 2005. Interaction regulator of release probability. Nature 410: 41–49. of neuronal calcium sensor-1 and ARF1 allows bidirec- Fernandez I, Arac D, Ubach J, Gerber SH, Shin O-h, Gao Y, tional control of PI(4) kinase and TGN-plasma mem- Anderson RGW, Sudhof TC, Rizo J. 2001. Three- brane traffic. J Biol Chem 280: 6047–6054. dimensional structure of the synaptotagmin 1 C2B- Haynes LP,Fitzgerald DJ, Wareing B, O’Callaghan DW,Mor- domain: Synaptotagmin 1 as a binding gan A, Burgoyne RD. 2006. Analysis of the interacting machine. Neuron 32: 1057–1069. partners of the neuronal calcium-binding proteins Few AP, Lautermilch NJ, Westenbroek RE, Scheuer T, Cat- L-CaBP1, hippocalcin, NCS-1 and neurocalcin. Proteo- mics 6: 1822–1832. terall WA. 2005. Differential regulation of Cav2.1 chan- nels by calcium binding protein 1 and visinin-like Haynes LP,Sherwood MW,Dolman NJ, Burgoyne RD. 2007. protein-2 requires N-terminal myristoylation. J Neurosci Specificity, promiscuity and localization of ARF protein 25: 7071–7080. interactions with NCS-1 and phosphatidylinositol-4 kinase-IIIb. Traffic 8: 1080–1092. Fitzgerald DJ, Burgoyne RD, Haynes LP. 2008. Neuronal calcium sensor proteins are unable to modulate NFAT Hendricks KB, Wang BQ, Schnieders EA, Thorner J. 1999. activation in mammalian cells. Biochim Biophys Acta Yeast homologue of neuronal frequenin is a regulator of 1780: 240–248. phosphatidylinositol-4-OH kinase. Nature Cell Biology 1: 234–241. Flaherty KM, Zoulya S, Stryer L, McKay DB. 1993. 3-Dimensional structure of recoverin, a calcium sensor Hilfiker S. 2003. Neuronal calcium sensor-1: A multifunc- tional regulator of secretion. Biochem Soc Trans 31: in vision. Cell 75: 709–716. 828–832. Geppert M, Goda Y, Hammer RE, Li C, Rosahl TW, Stevens Holmqvist MH, Cao J, Hernandez-Pineda R, Jacobson MD, CF, Sudhof TC. 1994. Synaptotagmin I: A major Ca2þ Carroll KI, Sung MA, Betty M, Ge P, Gilbride KJ, Brown sensor for transmitter release at a central synapse. Cell ME, et al. 2002. Elimination of fast inactivation in Kv4 79: 717–727. A-type potassium channels by an auxiliary subunit Gomez M, De Castro E, Guarin E, Sasakura H, Kuhara A, domain. Proc Natl Acad Sci USA 99: 1035–1040. 2þ Mori I, Bartfai T, Bargmann CI, Nef P.2001. Ca signal- Howes KA, Pennesi ME, Sokal I, Church-Kopish J, Schmidt ling via the neuronal calcium sensor-1 regulates associa- B, Margolis D, Frederick JM, Rieke F, Palczewski K, Wu tive learning and memory in C.elegans. Neuron 30: SM, et al. 2002. GCAP1 rescues rod photoreceptor 241–248. response in GCAP1/GCAP2 knockout mice. EMBO Jour- Groffen AJ, Martens S, Arazola RD, Cornelisse LN, Lozovaya nal 21: 1545–1554. N, de Jong AP,Goriounova NA, Habets RL, Takai Y,Borst Hui E, Johnson CP,YaoJ, Dunning FM, Chapman ER. 2009. JG, et al. 2010. Doc2b is a high-affinity Ca2þ sensor Synaptotagmin-mediated bending of the target mem- for spontaneous neurotransmitter release. Science 327: brane is a critical step in Ca(2þ)-regulated fusion. Cell 1614–1618. 138: 709–721.

16 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

Calcium Sensor Proteins in Neuronal Function

Jo J, Heon S, Kim MJ, Son GH, Park Y,Henley JM, Weiss JL, Lin L, Jeanclos EM, Treuil M, Braunewell K-H, Gundelfinger Sheng M, Collingridge GL, Cho K. 2008. Metabotropic ED, Anand R. 2002. The calcium sensor protein visinin- glutamate receptor-mediated LTD involves two interact- like protein-1 modulates the surface expression and ing Ca2þ sensors, NCS-1 and PICK1. Neuron 60: 1095– agonist sensitivity of the a4b2 nicotinic acetylcholine 1111. receptor. J Biol Chem 277: 41872–41878. Jo D-G, Jang J, Kim B-J, Lundkvist J, Jung Y-K. 2004. Over- Lisman J, Schulman H, Cline H. 2002. The molecular basis expression of calsenilin enhances g-secretase activity. of CaMKII function in synaptic and behavioural mem- Neuroscience letters 378: 59–64. ory. Nat Rev Neurosci 3: 175–190. Kabbani N, Negyessy L, Lin R, Goldman-Rakic P,Levenson Littink KW, van Genderen MM, Collin RW, Roosing S, de R. 2002. Interaction with the neuronal calcium sensor Brouwer AP, Riemslag FC, Venselaar H, Thiadens AA, NCS-1 mediates desensitization of the D2 dopamine Hoyng CB, Rohrschneider K, et al. 2009. A novel homo- receptor. J Neurosci 22: 8476–8486. zygous nonsense mutation in CABP4 causes congenital cone-rod synaptic disorder. Investigative Ophthalmol Vis- Kasri NN, Holmes AM, Bultynck G, Parys JB, Bootman MD, ual Sci 50: 2344–2350. Rietdorf K, Missiaen L, McDonald F,Smedt HD, Conway Liu X, Yang PS, Yang W, Yue DT. 2010. Enzyme-inhibitor- SJ, et al. 2004. Regulation of InsP receptor activity by 3 like tuning of Ca2þ channel connectivity with calmodu- neuronal Ca2þ-binding proteins. EMBO J 23: 1–10. lin. Nature 463: 968–972. Kinoshita-Kawada M, Tang J, Xiao R, Kaneko S, Foskett JK, 2þ Mackler JM, Drummond JA, Loewen CA, Robinson IM, Zhu MX. 2005. Inhibition of TRPC5 channels by Ca Reist NE. 2002. The C2B Ca2þ-binding motif of synapto- binding protein 1 in Xenopus . Pflugers Arch tagmin is required for synaptic transmission in vivo. 450: 345–354. Nature 418: 340–344. Klenshin VA,Calvert PD, Bownds MD. 1995. Inhibition of Makino CL, Dodd RL, Chen J, Burns ME, Roca A, Simon rhodopsin kinase by recoverin. J Biol Chem 270: MI, Baylor DA. 2004. Recoverin regulates light- 16147–16152. dependent phosphodiesterase activity in retinal rods. J Koch K-W. 2006. GCAPs, the classical neuronal calcium Gen Physiol 123: 729–741. 2þ sensors in the retina. A Ca -relay model of guanylate Malleret G, Haditsch U, Genoux D, Jones MW, Bliss TV, cyclase activation. Calcium Binding Proteins 1: 3–6. VanhooseAM, WeitlaufC, Kandel ER, Winder DG, Man- Koh PO, Undie AS, Kabbani N, Levenson R, Goldman- suy IM. 2001. Inducible and reversible enhancement of Rakic PS, Lidow MS. 2003. Up-regulation of neuronal learning, memory, and long-term potentiation by genetic calcium sensor-1 (NCS-1) in the prefrontal cortex of inhibition of calcineurin. Cell 104: 675–686. schizophrenic and bipolar patients. Proc Natl Acad Sci Markova O, Fitzgerald D, Stepanyuk A, Dovgan A, Cherkas 100: 313–317. V, Tepikin A, Burgoyne RD, Belan P. 2008. Hippocalcin Korhonen L, Hansson I, Kukkonen JP,Brannvall K, Kobaya- signaling via site-specific translocation in hippocampal shi M, Takamatsu K, Lindholm D. 2004. Hippocalcin neurons. Neuroscience letters 442: 152–157. protects against caspase-12-induced and age-dependent McCue HV,Burgoyne RD, Haynes LP.2009. Membrane tar- neuronal degeneration. Mol Cell Neurosci 28: 85–95. geting of the EF-hand containing calcium-sensing pro- Kuo H-C, Cheng C-F, Clark RB, Lin JJ-C, Lin JL-C, Hoshi- teins CaBP7 and CaBP8. Biochem Biophys Res Commun jima M, Nguyen-Tran VTB, Gu Y,Ikeda Y,Chu P-H, et al. 380: 825–831. 2001. A defect in the Kv channel-interacting protein 2 McCue HV, Haynes LP, Burgoyne RD. 2010. Bioinformatic analysis of CaBP/calneuron proteins reveals a family of (KChIP2) gene leads to a complete loss of Ito and confers 2þ susceptibility to ventricular tachycardia. Cell 107: highly conserved vertebrate Ca -binding proteins. 801–813. BMC Res Notes 3: 118. Lee A, Scheuer T, Catterall WA. 2000. Ca2þ/calmodulin- McFerran BW,Graham ME, Burgoyne RD. 1998. NCS-1, the dependent facilitation and inactivation of P/Q-type mammalian homologue of frequenin is expressed in 2þ chromaffin and PC12 cells and regulates neurosecre- Ca channels. J Neurosci 20: 6830–6838. tion from dense-core granules. J Biol Chem 273: Lee A, Jimenez A, Cui G, Haeseleer F.2007. Phosphorylation 22768–22772. of the Ca2þ-binding protein CaBP4 by Mellstrom B, Naranjo JR. 2001. Ca2þ-dependent transcrip- zeta in photoreceptors. J Neurosci 27: 12743–12754. tional repression and derepression: DREAM, a direct Lee A, Westenbroek RE, Haeseleer F, Palczewski K, Scheuer effector. Seminars Cell Develop Biol 12: 59–63. T, Catterall WA. 2002. Differential modulation of 2þ Mendez A, Burns ME, Sokal I, Dizhoor AM, Baehr W, Palc- Cav2.1 channels by calmodulin and Ca -binding pro- zewski K, Baylor DA, Chen J. 2001. Role of guanylate tein 1. Nature Neurosci 5: 210–217. cyclase-activating proteins (GCAPs) in setting the flash Li C, Chan J, Haeseleer F, Mikoshiba K, Palczewski K, Ikura sensitivity of rod photoreceptors. Proc Natl Acad Sci 98: M, Ames JB. 2009. Structural insights into Ca2þ- 9948–9953. dependent regulation of inositol 1,4,5-trisphosphate Mercer WA,Korhonen L, Skoglosa Y, Olssen P-A, Kukknen receptors by CaBP1. J Biol Chem 284: 2472–2481. JP, Lindholm D. 2000. NAIP interacts with hippocalcin Lilliehook C, Bozdagi O, Yao J, Gomez-Ramirez M, Zaidi and protects neurons against calcium-induced cell death NF, Wasco W, Gandy S, Santucci AC, Haroutunian V, through caspase-3-dependent and -independent path- Huntley GW,et al. 2003. Altered Ab formation and long- ways. EMBO J 19: 3597–3607. term potentiation in a calsenilin knock-out. J Neurosci Mikhaylova M, Reddy PP,Munsch T,Landgraf P,Suman SK, 23: 9097–9106. Smalla KH, Gundelfinger ED, Sharma Y, Kreutz MR.

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 17 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

H.V. McCue, L.P. Haynes, and R.D. Burgoyne

2009. Calneurons provide a calcium threshold for trans- reveals a cross-shaped octamer. Nature Struct Mol Biol 13: Golgi network to plasma membrane trafficking. Proc Natl 987–995. Acad Sci 106: 9093–9098. Polans A, Baehr W,Palczewski K. 1996. Turnedon by Ca2þ! Mikhaylova M, Sharma Y, Reissner C, Nagel F, Aravind P, The physiology and pathology of Ca2þ-binding proteins Rajini B, Smalla KH, Gundelfinger ED, Kreutz MR. in the retina. Trends Neurosci 19: 547–554. 2þ 2006. Neuronal Ca signaling via caldendrin and cal- Pongs O, Lindemeier J, Zhu XR, Theil T, Endelkamp D, neurons. Biochim Biophys Acta 1763: 1229–1237. Krah-Jentgens I, Lambrecht H-G, Koch KW, Schwemer Nef S, Fiumelli H, de Castro E, Raes M-B, Nef P.1995. Iden- J, Rivosecchi R, et al. 1993. Frequenin - A novel calcium- tification of a neuronal calcium sensor (NCS-1) possibly binding protein that modulates synaptic efficacy in the involved in the regulation of receptor phosphorylation. J Drosophila nervous system. Neuron 11: 15–28. Receptor Signal Trans 15: 365–378. Pruunsild P, Timmusk T. 2005. Structure, alternative splic- O’Callaghan DW,TepikinAV,Burgoyne RD. 2003b. Dynam- ing, and expression of the human and mouse KCNIP 2þ ics and calcium-sensitivity of the Ca -myristoyl switch gene family. Genomics 86: 581–593. protein hippocalcin in living cells. J Cell Biol 163: Rhodes KJ, Carroll KI, Sung MA, Doliveira LC, Monaghan 715–721. MM, Burke SL, Strassle BW, Buchwalder L, Menegola O’Callaghan DW, Haynes LP, Burgoyne RD. 2005. High- M, Cao J, et al. 2004. KChIPs and Kv4asubunitsSA as inte- affinity interaction of the N-terminal myristoylation gral components of A-type potassium channels in mam- motif of the neuronal calcium sensor protein hippocalcin malian brain. J Neurosci 24: 7903–7915. with phosphatidylinositol 4,5-bisphosphate. Biochem J Rickman C, Jime´nez JL, Graham ME, Archer DA, Soloviev 391: 231–238. M, Burgoyne RD, Davletov B. 2006. Conserved pre- O’Callaghan DW, Hasdemir B, Leighton M, Burgoyne RD. fusion protein assembly in regulated exocytosis. Mol 2003a. Residues within the myristoylation motif deter- 2þ Biol Cell 17: 283–294. mine intracellular targeting of the neuronal Ca sensor 2þ protein KChIP1 to post-ER transport vesicles and traffic Rieke F, Lee A, Haeseleer F. 2008. Characterization of Ca - of Kv4 Kþ channels. J Cell Sci 116: 4833–4845. binding protein 5 knockout mouse retina. Investigative Ophthalmol Visual Sci 49: 5126–5135. O’Callaghan DW, Ivings L, Weiss JL, Ashby MC, TepikinAV, Burgoyne RD. 2002. Differential use of myristoyl groups Rizo J, Rosenmund C. 2008. Synaptic vesicle fusion. Nat on neuronal calcium sensor proteins as a determinant of Struct Mol Biol 15: 665–674. spatio-temporal aspects of Ca 2þ-signal transduction. J Robinson IM, Ranjan R, Schwarz TL. 2002. Synaptotagmins BiolChem 277: 14227–14237. I and IV promote transmitter release independently of 2þ O’Connor V, El Far O, Bofill-Cardona E, Nanoff C, Freiss- Ca binding in the C2A domain. Nature 418: 336–340. muth M, Karschin A, Airas JM, Betz H, Boehm S. 1999. Saab BJ, Georgiou J, Nath A, Lee FJ, Wang M, Michalon A, Calmodulin dependence of presynaptic metabotropic Liu F, Mansuy IM, Roder JC. 2009. NCS-1 in the dentate glutamate receptor signalling. Science 286: 1180–1184. gyrus promotes exploration, synaptic plasticity, and Olafsson P,WangT,Lu B. 1995. Molecular cloning and func- rapid acquisition of . Neuron 63: tional characterisation of the Xenopus Ca2þ binding pro- 643–656. tein frequenin. Proc Natl Acad Sci 92: 8001–8005. Saimi Y, Kung C. 2002. Calmodulin as an ion channel sub- Palczewski K, Sokal I, Baehr W. 2004. Guanylate unit. Ann Rev Physiol 64: 289–311. cyclase-activating proteins: Structure, function and Sampath A, Strissel KJ, Elias R, Arshavsky VY, McGinnis JF, diversity. Biochem Biophys Res Comm 322: 1123–1130. Rieke F, Hurley JB. 2005. Recoverin improves rod- Palmer CL, Lim W, Hastie PG, Toward M, Korolchuk VI, mediated vision by enhancing signal transmission in Burbidge SA, Banting G, Collingridge GL, Isaac JT, Hen- the mouse retina. Neuron 46: 413–420. ley JM. 2005. Hippocalcin functions as a calcium sensor Sanchez-Gracia A, Romero-Pozuelo J, Ferrus A. 2010. Two in hippocampal LTD. Neuron 47: 487–494. frequenins in Drosophila: unveiling the evolutionary his- Pang ZP, Cao P, Xu W, Sudhof TC. 2010. Calmodulin con- tory of an unusual neuronal calcium sensor (NCS) dupli- trols synaptic strength via presynaptic activation of calm- cation. BMC Evolutionary Biol 10: 54. odulin kinase II. J Neurosci 30: 4132–4142. Scannevin RH, Wang K-W, Jow F, Megules J, Kospco DC, Paterlini M, Revilla V, Grant AL, Wisden W. 2000. Expres- Edris W, Carroll KC, Lu Q, Xu W, Xu Z, et al. 2004. sion of the neuronal calcium sensor protein family in Two N-terminal domains of Kv4 Kþ channels regulate the rat brain. Neurosci 99: 205–216. binding to and modulation by KChIP1. Neuron 41: Pennesi ME, Howes KA, Baehr W, Wu SM. 2003. Guanylate 587–598. cyclase-activating protein (GCAP) 1 rescues cone recov- Schaad NC, De Castro E, Nef S, Hegi S, Hinrichsen R, ery kinetics in GCAP1/GCAP2 knockout mice. Proc Martone ME, Ellisman MH, Sikkink R, Sygush J, Nef Natl Acad Sci 100: 6783–6788. P. 1996. Direct modulation of calmodulin targets by the Petko JA, Kabbani N, Frey C, Woll M, Hickey K, Craig M, neuronal calcium sensor NCS-1. Proc Natl Acad Sci 93: Canfield VA,Levenson R. 2009. Proteomic and functional 9253–9258. analysis of NCS-1 binding proteins reveals novel signal- Schiavo G, Stenbeck G, Rothman JE, Sollner TH. 1997. ing pathways required for inner ear development in Binding of the synaptic vesicle v-SNARE, synaptotagmin, zebrafish. BMC Neurosci 10: 27. to the plasma membrane t-SNARE, SNAP-25 can explain Pioletti M, Findeisen F, Hura GL, Minor DL. 2006. Three- docked vesicles at neurotoxin-treated synapses. Proc Natl dimensional structure of the KChIP1–Kv4.3 T1 complex Acad Sci 94: 997–1001.

18 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

Calcium Sensor Proteins in Neuronal Function

Schnurra I, Bernstein H-G, Riederer P, Braunewell K-H. Tanaka T, Ames JB, Harvey TS, Stryer L, Ikura M. 1995. 2001. The neuronal calcium sensor protein VILIP-1 is Sequestration of the membrane targeting myristoyl associated with amyloid plaques and promotes cell death group of recoverin in the calcium-free state. Nature and tau phosphorylation in vitro: A link between calcium 376: 444–447. sensors and alzheimers disease? Neurobiology Disease 8: Tang N, Lin T,YangJ, Foskett JK, Ostap EM. 2007. CIB1 and 900–909. CaBP1 bind to the myo1c regulatory domain. J Muscle Schumacher MA, Rivard AF, Bachinger HP, Adelman JP. Res Cell Motility 28: 285–291. 2þ 2001. Structure of the gating domain of a Ca -activated Tang J, Maximov A, Shin OH, Dai H, Rizo J, Sudhof TC. þ 2þ K channel complexed with Ca /calmodulin. Nature 2006. A complexin/synaptotagmin 1 switch controls 410: 1120–1124. fast synaptic vesicle exocytosis. Cell 126: 1175–1187. Seidenbecher CI, Landwehr M, Smalla K-H, Kreutz M, Diet- Taverna E, Francolini M, Jeromin A, Hilfiker S, Roder J, Rosa erich DC, Zuschratter W, Reissner C, Hammarback JA, P.2002. Neuronal calcium sensor 1 and phosphatidylino- Bockers TM, Gundelfinger ED, et al. 2004. Caldendrin sitol 4-OH kinase b interact in neuronal cells and are but not calmodulin binds to light chain 3 of MAP1A/ translocated to membranes during nucleotide-evoked B: An association with the microtubule exocytosis. J Cell Sci 115: 3909–3922. highlighting exclusive binding partners for neuronal 2þ Tippens AL, Lee A. 2007. Caldendrin, a neuron-specific Ca -sensor proteins. J Mol Biol 336: 957–970. modulator of Cav/1.2 (L-type) Ca2þ channels. J Biol Seidenbecher CI, Langnaese K, Sanmarti-Vila L, Boeckers Chem 282: 8464–8473. TM, Smalla K-H, Sabel BA, Garner CC, Gundelfinger Tsujimoto T, Jeromin A, Satoh N, Roder JC, Takahashi T. ED, Kreutz MR. 1998. Caldendrin, a novel neuronal 2002. Neuronal calcium sensor 1 and activity-dependent calcium-binding protein confined to the somato- facilitation of P/Q-type calcium channel currents at pre- dendritic compartment. J Biol Chem 273: 21324–21331. synaptic nerve terminals. Science 295: 2276–2279. Shao X, Fernandez I, Sudhof TC, Rizo J. 1998. Solution 2þ 2þ Venn N, Haynes LP, Burgoyne RD. 2008. Specific effects of structures of the Ca -free and Ca -bound C2A domain / / 2þ KChIP3 calsenilin DREAM but not KChIPs1, 2 and 4 of synaptotagmin I: Does Ca induce a conformational on calcium signalling and regulated secretion in PC12 change? Biochem 37: 16106–16115. cells. Biochem J 413: 71–80. Shih PY, Lin CL, Cheng PW, Liao JH, Pan CY. 2009. Cal- Wang H, Yan Y, Liu Q, Huang Y, Shen Y, Chen L, Chen Y, 2þ neuron I inhibits Ca channel activity in bovine chro- Yang Q, Hao Q, Wang K, et al. 2007. Structural basis maffin cells. Biochem Biophys Res Commun 388: for modulation of Kv4 Kþ channels by auxiliary KChIP 549–553. subunits. Nat Neurosci 10: 32–39. Smalla K-H, Seidenbecher CI, Tischmeyer W,Schicknick H, Weiergraber OH, Senin II, Philippov PP, Granzin J, Koch Wyneken U, Bockers TM, Gundelfinger ED, Kreutz MR. K-W. 2003. Impact of N-terminal myristoylation on the 2003. Kainate-induced epileptic seizures induce a recruit- Ca2þ-dependent conformational transition in recoverin. ment of caldendrin to the postsynaptic density in rat J Biol Chem 278: 22972–22979. brain. Mol Brain Res 116: 159–162. WeissJL, Archer DA, Burgoyne RD. 2000. NCS-1/frequenin Sollner T, Whiteheart SW,Brunner M, Erdjument-Bromage functions in an autocrine pathway regulating Ca2þ chan- H, Geromanos S, Tempst P, Rothman JE. 1993. SNAP nels in bovine adrenal chromaffin cells. J Biol Chem 275: receptors implicated in vesicle targeting and fusion. 40082–40087. Nature 362: 318–324. WenH, Levitan IB. 2002. Calmodulin is an auxiliary subunit Spilker C, Dresbach T, Braunewell K-H. 2002. Reversible of KCNQ2/3 potassium channels. J Neurosci 22: translocation and activity-dependent localisation of the 7991–8001. calcium-myristoyl switch protein VILIP-1 to different Wingard JN, Chan J, Bosanac I, Haeseleer F, Palczewski K, membrane compartments in living hippocampal neu- Ikura M, Ames JB. 2005. Structural analysis of Mg2þ rons. J Neurosci 22: 7331–7339. and Ca2þ binding to CaBP1, a neuron-specific regulator Spitzer NC. 2006. Electrical activity in early neuronal devel- of calcium channels. J Biol Chem 280: 37461–37470. opment. Nature 444: 707–712. Woods AS, Marcellino D, Jackson SN, Franco R, Ferre S, Stephen R, Bereta G, Golczak M, Palczewski K, Sousa MC. Agnati LF, Fuxe K. 2008. How calmodulin interacts 2007. Stabilizing function for myristoyl group revealed with the adenosine A(2A) and the dopamine D(2) recep- by the crystal structure of a neuronal calcium sensor, gua- tors. J Proteome Res 7: 3428–3434. nylate cyclase-activating protein 1. Structure 15: Wu YQ, Lin X, Liu CM, Jamrich M, Shaffer LG. 2001. Iden- 1392–1402. tification of a human brain-specific gene, calneuron 1, a Strahl T,Huttner IG, Lusin JD, Osawa M, King D, Thorner J, new member of the calmodulin superfamily. Mol Genet Ames JB. 2007. Structural insights into activation of Metab 72: 343–350. phosphatidylinositol 4-kinase (pik1) by yeast frequenin Xia Z, Storm DR. 2005. The role of calmodulin as a signal (Frq1). J Biol Chem 282: 30949–30959. integrator for synaptic plasticity. Nat Rev Neurosci 6: Sudhof TC, Rothman JE. 2009. Membrane fusion: Grap- 267–276. pling with SNARE and SM proteins. Science 323: Xia X-M, Fakler B, Rivard A, Wayman G, Johnson-Pais T, 474–477. Keen JE, Ishii T, Hirschberg B, Bond CT, Lutsenko S, Tadross MR, Dick IE, Yue DT.2008. Mechanism of local and Maylie J, Adelman JP.1998. Mechanisms of calcium gat- global Ca2þ sensing by calmodulin in complex with a ing in small conductance calcium activated potassium Ca2þ channel. Cell 133: 1228–1240. channels. Nature 395: 503–507.

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 19 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

H.V. McCue, L.P. Haynes, and R.D. Burgoyne

Xiong H, Xia K, Li B, Zhao G, Zhang Z. 2009. KChIP1: A Zeitz C, Kloeckener-Gruissem B, Forster U, Kohl S, Magyar potential modulator to GABAergic system. Acta Biochim I, Wissinger B, Matyas G, Borruat FX, Schorderet DF, Biophys Sin (Shanghai) 41: 295–300. Zrenner E, et al. 2006. Mutations in CABP4, the gene 2þ Xue M, Ma C, Craig TK, Rosenmund C, Rizo J. 2008. The encoding the Ca -binding protein 4, cause autosomal Janus-faced nature of the C(2)B domain is fundamental recessive night blindness. Am J Hum Genet 79: 657–667. for synaptotagmin-1 function. Nat Struct Mol Biol 15: Zhao X, Varnai P, Tuymetovna G, Balla A, Toth ZE, Oker- 1160–1168. Blom C, Roder J, Jeromin A, Balla T. 2001. Interaction Yamagata K, Goto K, Kuo CH, Kondo H, Miki N. 1990. Vis- of neuronal calcium sensor-1 (NCS-1) with phosphati- inin: A novel calcium binding protein expressed in retianl dylinositol 4-kinase b stimulates lipid kinase activity cone cells. Neuron 4: 469–476. and affects membrane trafficking in COS-7 cells. J Biol YangS-N, TangY-G, Zucker RS. 1999. Selective induction of Chem 276: 40183–40189. 2þ LTPand LTD by postsynaptic [Ca ]i elevation. J Neuro- Zhou H, Kim S-A, Kirk EA, Tippens AL, Sun H, Haeseleer F, physiol 81: 781–787. Lee A. 2004a. Ca2þ-binding protein-1 facilitates and forms a postsynaptic complex with Cav1.2 (L-type) Yang PS, Alseikhan BA, Hiel H, Grant L, Mori MX, Yang W, 2þ Fuchs PA, Yue DT. 2006. Switching of Ca2þ-dependent Ca channels. J Neurosci 24: 4698–4708. inactivation of Ca(V)1.3 channels by calcium binding Zhou H, Yu K, McCoy KL, Lee A. 2005. Molecular mecha- 2þ proteins of auditory hair cells. J Neurosci 26: 10677– nism for divergent regulation of Cav1.2 Ca channels 10689. by calmodulin and Ca2þ-binding protein-1. J BiolChem Yang J, McBride S, Mak D-OD, Vardi N, Palczewski K, Hae- 280: 29612–29619. seleer F,Foskett JK. 2002. Identification of a family of cal- Zhou W, Qian Y, Kunjilwar K, Pfaffinger PJ, Choe S. 2004b. cium sensors as protein ligands of Structural insights into the functional interaction receptor Ca2þ release channels. Proc Natl Acad Sci 99: of KChIP1 with shal-type Kþ channels. Neuron 41: 7711–7716. 573–586.

20 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a004085 Downloaded from http://cshperspectives.cshlp.org/ on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press

The Diversity of Calcium Sensor Proteins in the Regulation of Neuronal Function

Hannah V. McCue, Lee P. Haynes and Robert D. Burgoyne

Cold Spring Harb Perspect Biol 2010; doi: 10.1101/cshperspect.a004085 originally published online July 28, 2010

Subject Collection

For additional articles in this collection, see http://cshperspectives.cshlp.org/cgi/collection/

Copyright © 2010 Cold Spring Harbor Laboratory Press; all rights reserved