You are on page 1of 12

MICROSCOPY RESEARCH AND TECHNIQUE 49:14 –25 (2000)

Drosophila Larval Neuromuscular Junction:


Molecular Components and Mechanisms
Underlying Synaptic Plasticity
YOUNG HO KOH,1,2 L. SIAN GRAMATES,1,3 AND VIVIAN BUDNIK1–3*
1
Biology Department, University of Massachusetts, Amherst, Massachusetts 01003
2
Neuroscience and Behavior Program, University of Massachusetts, Amherst, Massachusetss 01003
3
Program for Molecular, Cellular and Developmental Biology, University of Massachusetts, Amherst, Massachusetts 01003

KEY WORDS: discs large; CAMKII; MAGUKs; in vitro system; synapse assembly; PKA; second
messengers; retrograde signals; postsynaptic protein translation; gene regula-
tion
ABSTRACT Understanding the mechanisms that mediate synaptic plasticity is a primary goal
of molecular neuroscience. The Drosophila larval neuromuscular junction provides a particularly
useful model for investigating the roles of synaptic components in both structural and functional
plasticity. The powerful molecular genetics of this system makes it possible to uncover new synaptic
components and signaling molecules, as well as their function in the intact organism. Together with
the mouse hippocampus and Aplysia dissociated cell culture, the Drosophila larval neuromuscular
junction has been among the most valuable model systems for examining the molecular and cellular
basis of neuronal plasticity. Microsc. Res. Tech. 49:14 –25, 2000. © 2000 Wiley-Liss, Inc.

INTRODUCTION and, therefore, to allow for synaptic plasticity, is not


A chemical synapse is organized into a presynaptic fully known.
terminal, containing synaptic vesicles and neurotrans- Most studies aimed at unraveling the mechanisms
mitter release machinery, and a postsynaptic element underlying synaptic plasticity in the mammalian cen-
that is characterized by the presence of dense clusters tral nervous system (CNS) have been performed using
of neurotransmitter receptor and ion-channel com- in vitro systems, in cultured cells, brain slices, or bio-
plexes. The pre- and postsynaptic elements are sepa- chemical assays (e.g., Fields and Itoh, 1996; Kryl et al.,
rated by a synaptic cleft across which neuroactive mol- 1999; Niethammer et al., 1999). Most in vivo studies
ecules and ions can diffuse (reviewed by Garner and have been limited to immunocytochemical descriptions
Kindler, 1996). If a signal is to be transmitted effec- of protein localization (e.g., Burgin et al., 1990; Furui-
tively across the synaptic cleft, a precise localization of chi et al., 1993). Therefore, the analysis of plasticity in
ion channels and neurotransmitter receptors at both the whole organism has been greatly enhanced by the
the pre- and the postsynaptic elements is critical (re- use of model systems such as the fruit fly and Aplysia.
viewed in Budnik, 1996; Gramates and Budnik, The Drosophila larval neuromuscular junction
1999a). (NMJ) is particularly advantageous to the investiga-
Synaptic plasticity, the process by which connections tion of synaptic plasticity. The most favorable features
between a neuron and its target are modified, is be- of this model system include its powerful genetics, an-
lieved to be the basic mechanism underlying learning atomical and electrophysiological accessibility, and a
and memory. Studies using mammalian and inverte- well-defined synapse ultrastructure. These features fa-
brate models show that there are many common mech- cilitate the discovery of synaptic constituents and the
anisms involved in different categories of learning and functional analysis of these proteins at identified syn-
memory, and that these mechanisms are evolutionarily apses.
conserved from lower invertebrate to higher mamma- Although the Drosophila NMJ is simpler than the
lian systems (reviewed by Bailey et al., 1996; Milner et mammalian CNS, both in regard to synaptic connec-
al., 1998). For example the cAMP and Ca2⫹-dependent tions and diversity of neurons, it shows striking con-
signal transduction pathways appear to be common servation of many key synaptic molecules identified in
elements in every form of learning and memory among mammals. Amongst these conserved components are
animal species (reviewed by Abel et al., 1998; Dubnau Discs-large (DLG), the Drosophila Membrane-Associ-
and Tully, 1998). ated Guanylate Kinase (MAGUK) homologous to PSD-
Studies of synaptic plasticity in the mammalian hip- 95/SAP90 family proteins (Budnik et al., 1996; Guan et
pocampus (Frey and Morris, 1997), Aplysia dissociated
cell cultures (Martin et al., 1997a), and the Drosophila
neuromuscular junction (Davis and Goodman, 1998) Contract grant sponsor: NIH; Contract grant numbers: RO1 NS3007, RO1
indicate that the minimal unit of synaptic plasticity NS37061, F31 NS10861-01; Contract grant sponsor: Human Frontiers Grant.
*Correspondence to: Vivian Budnik, Biology Department, Morrill (South),
may be a single synapse. However, the full array of University of Massachusetts, Amherst MA 01003.
components required to make a functional synapse, E-mail: vbudnik@bio.umass.edu

© 2000 WILEY-LISS, INC.


REGULATION OF SYNAPTIC COMPONENTS 15
al., 1996; Lahey et al., 1994; Tejedor et al., 1997; et al., 1996; Kurdyak et al., 1994; reviewed by Gra-
Thomas et al., 1997); Fasciclin II (FasII), the structural mates and Budnik, 1999a).
homologue of neuronal cell adhesion molecules
(NCAMs) (Schuster et al., 1996b; Thomas et al., 1997; SHALL WE GATHER AT THE SYNAPSE?
Zito et al., 1997); Drosophila Ca2⫹/calmodulin depen- MAGUKS AS ORGANIZERS OF
dent kinase II (CaMKII) (Griffith et al., 1993; Koh et GLUTAMERGIC SYNAPSES
al., 1999); and Drosophila cAMP dependent protein MAGUKs of the PSD-95 family have been shown to
kinase (PKA) (reviewed by Davis et al., 1995, 1998). be important organizers of multiprotein complexes in
Recent reviews (Budnik, 1996; Gramates and Bud- both mammalian CNS excitatory synapses (reviewed
nik, 1999a; Keshishian et al., 1993) have concentrated by Garner and Kindler, 1996; Kennedy, 1997) and at
on the assembly and maturation of synapses. In this the Drosophila larval NMJ (reviewed by Budnik, 1996;
review, we will integrate recent findings using the Dro- Gramates and Budnik, 1999a). MAGUKs contain three
sophila NMJ with studies of the mammalian brain and types of protein-protein interaction modules, one or
Aplysia dissociated cell cultures to address the ques- more PDZ domains, a Src homology 3 (SH3) domain
tion of how synaptic components implicated in devel- (Woods and Bryant, 1993), and a guanylate kinase-like
opment and synaptic plasticity are regulated. (GuK) domain (Woods and Bryant, 1991). Some
Synaptic plasticity is a dynamic process that is reg- MAGUKs and MAGUK-related proteins also contain
ulated at a number of different levels. Specific synaptic additional functional regions, such as a calmodulin
components are activated or deactivated, assembled in binding site and a CaM kinase-like N-terminal in the
a spatially restricted cluster or segregated into differ- cases of CamGUK (Dimitratos et al., 1997), CASK
ent cell compartments, brought into proximity to or (Hata et al., 1996) and Lin-2 (Hoskins et al., 1996)
released from the junctional membrane or cytoskele- (Fig. 1).
ton, synthesized or degraded. Regulatory signals are An ever-growing array of neuronal MAGUK binding
required for the translation of mRNA into newly syn- partners have been identified, as summarized in Fig-
thesized proteins, or to signal the genetic machinery in ure 1. Recent findings implicate PSD-95 family
the nucleus to activate or deactivate transcription of MAGUKs in the structural and functional regulation of
particular genes. synaptic plasticity via interaction with receptors (re-
viewed by Sheng, 1996), ion channels (reviewed by
TAKE ME OUT TO THE BOUTONS: A BRIEF Garcia et al., 1998; Sheng, 1996), cell adhesion mole-
OVERVIEW OF THE DROSOPHILA LARVAL cules (Irie et al., 1997; Thomas et al., 1997; Zito et al.,
NEUROMUSCULAR JUNCTION 1997), protein kinases (Koh et al., 1999; Tezuka et al.,
1999), or via regulation of retrograde signaling path-
The Drosophila larval body wall musculature con- ways (Brenman et al., 1996; Jaffrey et al., 1998) or
sists of a segmentally repeated pattern of 30 muscles changes in GTPase activity (Chen at al., 1998; Kim et
innervated by about 40 motorneurons. The axons of al., 1998). The existence of at least three different pro-
these motorneurons branch onto the surface of the tein-protein interaction motifs in MAGUKs and the
muscle fibers forming a synaptic arbor composed of a identification of many of their binding partners suggest
series of varicosities connected by thin axonal pro- that MAGUKs may serve to assemble and to anchor
cesses. These varicosities, or synaptic boutons, fall into specific synaptic components to allow for efficient reg-
three classes (reviewed in Gramates and Budnik, ulation of signal transduction pathways.
1999a; Rheuben, et al., 1999). The major class of bou- The first MAGUK binding partners identified were
tons (type I) are glutamatergic, and are enveloped by Shaker-type K⫹ channels (Kim et al., 1995) and
the subsynaptic reticulum (SSR), an elaborately lay- NMDA-type glutamate receptor subunits (Kim et al.,
ered structure formed from the muscle junctional mem- 1996; Kornau et al., 1995; Muller, et al., 1996). These
brane (Atwood et al., 1993; Jia et al., 1993). All 30 body transmembrane proteins were shown to become clus-
wall muscles are innervated by type I glutamatergic tered in heterologous cells upon transfection with
boutons. Subsets of muscles are additionally inner- mammalian MAGUKs PSD-95 and chapsyn-110 (Kim
vated by type II or type III boutons, which contain et al., 1995, 1996; Niethammer et al., 1998). These
octopamine or peptide neurotransmitters as well as studies demonstrated that the first and second PDZ
glutamate (Anderson et al., 1988; Cantera and Nassel, domains of the MAGUK bind the cytoplasmic C-termi-
1992; Gorczyca et al., 1993; Monastriani et al., 1995; nal S/TXV motifs of the binding partners (Doyle et al.,
Zhong and Pena, 1995). 1996; Kim et al., 1995; Kornau et al., 1995; reviewed by
Muscles 6 and 7, which have been a particular focus Budnik, 1996; Gramates and Budnik, 1999a; Sheng,
of NMJ research, are innervated exclusively by type I 1996). Similarly, the third PDZ domain of PSD-95 was
boutons. Extensive ultrastructural analyses on these found to interact with the cytoplasmic C-terminal
boutons reveal that they can be subdivided into type I S/TXV motif of neuroligin (Irie et al., 1997). Neuroligin
small (Is) and type I big (Ib) by size, the extent of is a postsynaptic transmembrane protein that func-
folding of the postsynaptic membrane or SSR, and elec- tions as a heterophilic cell adhesion molecule, binding
trophysiological properties (Atwood et al., 1993; Jia et tightly to the extracellular domain of presynaptic-neur-
al., 1993; Kurdyak et al., 1994). These two bouton exin in a Ca2⫹-dependent reaction (Nguyen and Sud-
subtypes show different functional properties; type Is hof, 1997). The intracellular C-terminal of ␤-neurexin
boutons have larger stimulation thresholds and excita- binds to the PDZ domain of CASK, a MAGUK-related
tory junctional currents (EJCs) of larger amplitude, protein containing an N-terminal similar to CaM ki-
while more pronounced short-term facilitation is ob- nase, in addition to PDZ, SH3, and GuK domains (Hata
served at type Ib boutons (Atwood et al., 1997; Budnik et al., 1996). This intricate multiprotein complex span-
16 Y.H. KOH ET AL.

suggesting that they are complexed with PSD-95 (Te-


zuka et al., 1999). These data indicate that PSD-95
brings receptors and kinases together for proper signal
transmission.
Changes in synaptic plasticity are accompanied by
changes in synapse morphology; axons form new
branches, new boutons are formed and stabilized or
retracted. Such alterations in cell morphology are nec-
essarily accompanied by cytoskeletal rearrangements.
In the mammalian synapse, the postsynaptic density
(PSD) is heavily enriched in cytoskeletal elements. Re-
cent experiments have raised the intriguing possibility
that PSD-95 may function in the rearrangement of the
PSD cytoskeleton, acting indirectly through its binding
partners. For example, the microtubule binding protein
CRIPT (Cysteine-rich interactor of PDZ three) also
binds to the third PDZ domain of PSD-95 (Niethammer
et al., 1998). CRIPT colocalizes with PSD-95 in the
postsynaptic density of brain synapses, and can be
coimmunoprecipitated with antisera to PSD-95 and tu-
bulin. If both proteins are co-transfected into heterolo-
gous cells, CRIPT redistributes PSD-95 to the microtu-
bule-based reticular network (Niethammer et al.,
1998).
PSD-95 also forms a complex with the Rho effector
Citron in hippocampal GABAergic neurons (Zhang et
al., 1999) and thalamic excitatory neurons (Furu-
yashiki et al., 1999); as with CRIPT, this interaction is
Fig. 1. Top: Protein-protein interaction modules of PSD-95 family
MAGUKs. Examples include DLG (Woods and Bryant, 1991), SAP97 mediated by the third PDZ domain (Zhang et al., 1999).
(Muller et al., 1995), PSD-95 (Cho et al, 1992; Kistner et al., 1993), The Rho family of small GTPases is known to act on
SAP102 (Muller et al., 1996), and Chapsyn-110 (Kim et al., 1996). rearrangements of the actin-based cytoskeleton, e.g.,
Middle: Schematic diagram of MAGUK-related proteins containing a during cytokinesis (Madaule et al., 1998), axonal out-
CaMKII-like domain, including CASK (Hata et al., 1996), CamGUK
(Dimitratos et al., 1997), and Lin-2 (Hoskins et al., 1996). Bottom: growth, and cell migration (Furuyashiki et al., 1999;
Selected binding partners of PSD-95 family MAGUKs, including Zipkin et al., 1997). Citron interacts with the GTP-
Shaker-type K⫹ channels (Kim et al., 1995), Fasciclin II (Thomas et bound form of Rho (Madaule et al., 1995). The interac-
al., 1997; Zito et al., 1997), NMDA-type glutamate receptor subunit tions of PSD-95 with proteins known to be involved in
(Kornau et al., 1995; Kim, et al., 1996; Muller, et al., 1996), nNOS
(Brenman et al., 1996), SynGAP (Kim et al., 1998; Chen et al., 1998), cytoskeleton dynamics suggest that synaptic MAGUKs
CRIPT (Niethammer et al. 1998), Citron (Furuyashiki et al., 1999; could have a regulatory relationship with cytoskeletal
Zhang et al., 1998), neuroligin (Irie et al., 1997), kainate receptor elements of the postsynaptic density.
(Garcia et al., 1998), band 4.1 (Lue et al., 1996), GKAP (Kim et al., Nearly all of these observations have been made
1997; Naisbett, et al., 1997; Takeuchi, et al., 1997) and GUKHolder
(Gramates and Budnik, 1999b). using biochemical assays, cell culture, or other in vitro
methods, with in vivo studies largely limited to the
determination of patterns of protein distribution via
immunocytochemistry. Very few mammalian MAGUK
ning the cell membranes of both the pre-and postsyn- mutants exist to assess their in vivo function. So far,
aptic cells provides an insight into how MAGUKs may only the generation of PSD-95 knockout mice, which
serve to organize pre- and postsynaptic elements into express a truncated form of the protein, has been re-
the precise arrangement required for efficient trans- ported (Migaud et al., 1999). These mice exhibit en-
mission of signals across the synaptic cleft, clustering hanced long-term potentiation and severe impairment
receptors and ion channels, and the association of these of spatial learning and memory (Migaud et al., 1999).
clusters with the synaptic cytoskeleton. However, there are no significant changes in synapse
MAGUKs have also been implicated in an important number or in clustering of NMDA NR1 subunits in the
reversible regulatory mechanism of synaptic transmis- hippocampus, possibly due to genetic redundancy of
sion, the phosphorylation of ion-channels or receptors. MAGUKs in the mammalian CNS (Migaud et al.,
Fyn, a member of the Src family of protein tyrosine 1999).
kinases, phosphorylates the NR2A subunit of NMDA In the fly, however, direct demonstrations of the clus-
receptors in heterologously transfected cells. fyn mu- tering function of MAGUKs have proved feasible. The
tant mice exhibit a reduced level of NR2A phosphory- Drosophila MAGUK Discs-large (DLG) is required both
lation, indicating that Fyn is likely to have an in vivo pre- and postsynaptically for synaptic targeting and
regulatory role (Tezuka et al., 1999). Deletion analysis clustering of Shaker-type K⫹ channels and the cell
indicates that the SH2 domain of Fyn binds to the third adhesion molecule FasII (Tejedor et al., 1997; Thomas
PDZ domain of PSD-95, and that this association is et al., 1997; reviewed by Budnik, 1996; Gramates and
independent of tyrosine phosphorylation. A number of Budnik, 1999a). Mutations deleting all but the first two
Src family protein tyrosine kinases, including Src, Yes, PDZ domains of DLG induce the ectopic clustering of
Lyn, and Fyn, are coimmunoprecipitated with PSD-95, Shaker-type K⫹ channels (Tejedor et al., 1997). Muta-
REGULATION OF SYNAPTIC COMPONENTS 17
tions with low levels of DLG result in mislocalization of cles of Drosophila larvae; mutations that delete one of
both Shaker-type K⫹ channels (Tejedor et al., 1997) the receptors are homozygous viable (Petersen et al.,
and FasII (Thomas et al., 1997) over muscles. Ultra- 1997). DGluRIIA, but not DGluRIIB, has an optimal
structural changes in pre- and postsynptic elements of consensus PKA phosphorylation site, thus making it a
dlg mutants are also observed at the EM level. In possible substrate for PKA regulation (Petersen et al.,
hypomorphic dlg alleles, the number of active zones 1997). Unlike wild type larvae, mutants lacking DGlu-
within presynaptic terminals is significantly increased RIIA do not show decreased quantal size in response to
(Thomas et al., 1997), while the elaborate membrane bath application of Sp-cAMPS, a nonhydrolyzable
folding of the postsynaptic SSR is substantially re- cAMP analog known to increase PKA activity (Davis et
duced (Budnik et al., 1996; Koh et al., 1999; Lahey et al., 1998). These data strongly suggest that PKA may
al., 1994). The increased number of active zones, as regulate the activity of DGluRIIA. The PKA-dependent
well as an increase in bouton size, seen in severe dlg reduction in quantal size is accompanied developmen-
hypomorphic alleles, is similar to the phenotype seen in tally by an increase in quantal content, indicating the
hyperexcitability mutants affecting K⫹ channels, such presence of a retrograde signal regulating presynaptic
as Shaker and ether á gógo (Jia et al., 1993), and is release of glutamate (Davis et al., 1998).
accompanied by an increase in synaptic efficacy (Bud-
nik et al., 1996). Both increased activity and decreased NO as a Potential Retrograde Signal at the
DLG expression result in decreased levels of FasII at Neuromuscular Junction
the synapse, which may explain this phenotype (Schus- The change in axon arborization induced by selective
ter et al., 1996b; Thomas et al., 1997). Evidence sugg- overexpression of the cell adhesion molecule FasII also
gests that decreased levels of FasII, or of the Aplysia points to the existence of a retrograde signal from
FasII homolog ApCAM, result in synapse growth (see postsynaptic muscle to presynaptic motorneuron
Ca2⫹/Calmodulin Kinase Dependent Phosphorylation (Davis and Goodman, 1998). When Fas II is overex-
of Synaptic Components) (Abel et al., 1998; Bailey and pressed in body wall muscles 6 and 13, they become
Kandel, 1993; Koh et al., 1999; Schuster et al., 1996b; hyperinnervated by RP3 or RP1 and RP5 motor neu-
Thomas et al., 1997). rons, while their companion muscles 7 and 12, which
normally share innervation by the same motor neuron,
RETURN TO SENDER: SECOND MESSENGER have reduced innervation. The dramatic increase in
PATHWAYS AND RETROGRADE SIGNALS AT bouton number on muscles 6 and 13 is compensated by
THE SYNAPSE a decrease in the amount of neurotransmitter released
cAMP-Dependent Pathway per bouton. Likewise, quantal size undergoes a com-
The cAMP-dependent second messenger pathway is pensatory increase at the muscles with reduced inner-
believed to be intimately involved in activity-depen- vation (Davis and Goodman, 1998). These differential
dent changes in synaptic plasticity. Mutations of com- changes in synaptic efficacy between hyper- and hypo-
ponents of this pathway have a profound effect on innervated muscle indicate a muscle-to-motor neuron
functional and structural synaptic plasticity of the lar- retrograde regulation at specific presynaptic terminals.
val NMJ, affecting the degree of branching of axon Several candidates have been suggested to poten-
terminals, bouton number, number of active zones per tially act as retrograde signalling molecules (reviewed
bouton, and number of vesicles released per nerve im- in Harish and Poo, 1992; Nguyen and Lichtman, 1996).
pulse (Davis et al., 1995; Griffith and Greenspan, 1993; Of these, an attractive candidate for this retrograde
Malenka et al., 1989; Wang et al., 1994; Zhong and Wu, signal is nitric oxide (NO), which has been implicated
1991; Zhong et al., 1992; reviewed in Budnik, 1996; in retrograde signaling in the mammalian hippocam-
Gramates and Budnik, 1999a; Hannan and Zhong, pus (O’Dell, et al., 1991; reviewed in Hawkins et al.,
1999). 1994, 1998) as well as in snail motor neurons (Park et
cAMP-dependent protein kinase A (PKA) is one of al., 1998). NO, synthesized by neuronal NO synthase
the multitudinous proteins regulated by cAMP. Inac- (nNOS), has been shown to activate soluble guanylyl
tive PKA exists as a tetramer, made up of two catalytic cyclase and lead to formation of cyclic GMP (reviewed
and two regulatory subunits. When the regulatory sub- in Bredt and Snyder, 1992). Mammalian nNOS is en-
units are bound by cAMP, the catalytic subunits are riched postsynaptically, where its activation is tightly
released as active monomers (Coffino et al., 1976). coupled to NMDA receptor-mediated Ca2⫹ influx.
There is some evidence that PKA phosphorylation of nNOS contains a PDZ domain, and interacts with the
postsynaptic glutamate receptors has a regulatory ef- MAGUKs PSD-95 or PSD-93 through a PDZ-PDZ in-
fect on functional synaptic plasticity. Overexpression of teraction (Brenman et al., 1996), thus physically as
wild type PKA catalytic or regulatory subunits in Dro- well as functionally linking nNOS to the NMDA recep-
sophila larval muscles decreases quantal size, as mea- tor. If expression of PSD-95 is suppressed in cultured
sured by the amplitude of miniature excitatory junc- cortical neurons, nNOS expression and function is un-
tional currents (mEJCs) or miniature excitatory junc- affected, but Ca2⫹-activated NO production is blocked,
tional potentials (mEJPs). Conversely, muscle-specific confirming that the physical association of NMDA re-
transgenic expression of a defective regulatory subunit ceptors with nNOS is critical to the functional associ-
in which the cAMP binding site is mutated has the ation (Sattler et al., 1999). This association appears to
opposite effect; quantal size is increased (Davis et al., be regulated by CAPON (carboxy terminal PDZ ligand
1998). These effects are not seen in mutants that lack of nNOS), which interacts with the nNOS PDZ domain
muscle-specific glutamate receptor IIA (DGluRIIA) via a C-terminal S/TXV–like motif (Jaffrey et al., 1998).
(Davis et al., 1998). DGluRIIA (Schuster et al., 1991) is PSD-95/nNOS clusters are lost in transfected cultured
one of two glutamate receptors expressed in the mus- cells overexpressing transgenic CAPON, which directly
18 Y.H. KOH ET AL.

competes with PSD-95 for binding of the nNOS PDZ Greenberg, 1991; English and Sweatt, 1996, 1997).
domain (Jaffrey et al., 1998). Ras-GTPases block this cascade by hydrolyzing the
There is evidence that NO may also function at the Ras-bound GTP (Downward et al., 1990; Trahey et al.,
Drosophila NMJ. An NO-sensitive guanylyl cyclase is 1988; Vogel et al., 1988; Wigler, 1990). Activation of
expressed in a stage-specific manner in the RP3 motor NMDA receptors is known to activate the MAP kinase
neuron, which innervates larval muscles 6 and 7, sug- signal transduction pathway (Bading and Greenberg,
gesting that an NO/cGMP signalling system could exist 1991; English and Sweatt, 1996, 1997). The PSD-95-
(Wildemann and Bicker, 1999a). Application of the NO mediated association of CaMKII and SynGAP with
donor sodium nitroprusside (SNP) to the larval NMJ NMDA receptors could establish a link between the
induces an increase in cGMP immunoreactivity in the NMDA receptor-mediated rise in Ca2⫹ and the activa-
boutons and neuronal cell bodies of presynaptic nerve tion of the MAP kinase pathway (Chen et al., 1998).
terminals, but not in the postsynaptic muscle. This can CaMKII is equally important at fly central and NMJ
be blocked by specific inhibitors of soluble guanylyl synapses where, much as in the mammalian central
cyclase (Wildemann and Bicker, 1999b). Furthermore, synapse, it is believed to be a major factor in synaptic
NO donors and membrane permeant cGMP analogues plasticity (Griffith et al., 1994; Koh et al., 1999). The
cause vesicle release at the NMJ (Wildeman and Drosophila CaMKII (DCaMKII) gene shows highest
Bicker, 1999b). However, there is no direct evidence homology to the mammalian-CaMKII subunit (Cho et
that larval muscles are the source of NO release. Mus- al., 1992), which is exclusively expressed in the mam-
cle-specific expression of Drosophila NOS has not yet malian hippocampus (Mayford et al., 1996). It has been
been detected by immunocytochemistry (Wildemann proposed that CaMKII can transduce transient synap-
and Bicker, 1999b). dNOS, unlike mammalian nNOS, tic activity to persistent synaptic structural and func-
does not have a PDZ domain (Regulski and Tully, tional changes via phosphorylation of its substrates
1995), and hence may not be able to interact directly (Lisman, 1994). The activities of the mammalian-
with DLG as nNOS interacts with PSD-95 (Brenman et CaMKII and DCaMKII are similarly regulated in a
al., 1996). complex manner by Ca2⫹/calmodulin and by autophos-
phorylation. This process has been demonstrated in
Ca2ⴙ/Calmodulin Kinase Dependent molecular-genetic experiments, in both mammals and
Phosphorylation of Synaptic Components flies, in which mutant forms of the molecules are over-
Ca2⫹/calmodulin-dependent protein kinase II (CaM expressed in specific tissues. In the presence of Ca2⫹/
kinase II) is one of the most abundant proteins in the calmodulin CaMKII autophosphorylates serine 287
postsynaptic density of the mammalian central syn- (286 in ␣-CaMKII) to produce a Ca2⫹/calmodulin-inde-
apse, although it is also expressed presynaptically. It pendent kinase. Mutation of serine 287 to alanine
has been suggested that it may play critical roles in (CaMKIIT287A) prevents autophosphorylation and re-
long-term potentiation (Mayford et al., 1996). In cul- sults in kinase activity that is exclusively dependent on
tured hippocampal neurons, transgenic CaMKII be- Ca2⫹/calmodulin (Mayford et al., 1996; Meyer et al.,
comes dissociated from filamentous actin in response to 1992; Wang et al., 1998). In contrast, changing serine
NMDA receptor activation by glutamate. Unbound 287 to aspartic acid (CaMKIIT287D) mimics autophos-
CaMKII is then translocated to the postsynaptic den- phorylation, so that the kinase becomes constitutively
sity of dendritic terminals, a process that requires the active, even in the absence of Ca2⫹/calmodulin (May-
binding of calmodulin. Transgenic CaMKII is colocal- ford et al., 1996; Wang et al., 1998). It is possible to
ized with PSD-95 immunoreactivity at the postsynap- inhibit CaMKII activity by overexpressing alanine
tic density, suggesting a possible functional or struc- CaM kinase II inhibitory peptide (ala), which competes
tural interaction between MAGUKs and CaMKII for its catalytic site, thus reducing kinase activity (Grif-
(Shen and Meyer, 1999). Interestingly, calmodulin fith and Greenspan, 1993; Wang et al., 1994).
binds near the SH3 domain of the human MAGUK Studies at the Drosophila larval NMJ show that
NE-dlg/SAP-102 in a Ca2⫹-dependent manner, and DCaMKII has an important role in the regulation of
NE-dlg/SAP 102 interacts in vitro with the GuK do- synapse structure and function via its interaction with
main of PSD-95 in the presence of Ca2⫹ and calmodulin DLG. DCaMKII appears to be expressed both pre- and
(Masuko et al., 1999). Thus, the interaction of neuronal postsynaptically at the larval NMJ where it partially
MAGUKs may be modulated by Ca2⫹/calmodulin. co-localizes with DLG (Koh et al., 1999). Antibodies to
A likely CaMKII substrate is SynGAP, another com- DCaMKII coimmunoprecipitate DLG from larval body
ponent of the postsynaptic density. p135 SynGAP is a wall muscle extracts (Koh et al., 1999). Persistent
synaptic Ras-GTPase activating protein. It interacts in DCaMKII activation via overexpression of the consti-
vitro with all three PDZ domains of PSD-95 and SAP- tutively active form at the NMJ induces abnormal
102 by its C-terminal S/TXV motif. Moreover, SynGAP NMJ morphology that is similar to the phenotype seen
colocalizes with PSD-95 at excitatory synapses (Chen in dlg mutant alleles. These abnormal phenotypes in-
et al., 1998; Kim et al., 1998). CaMKII-dependent phos- clude increased bouton size and abnormal NMJ ar-
phorylation of p135 SynGAP inhibits its Ras-GTPase borization pattern as well as mislocalization of FasII
activating activity (Chen et al., 1998). The GTP-bound (Koh et al., 1999). The striking similarity in NMJ mu-
form of Ras recruits Raf kinase to the cell membrane, tant phenotypes is also seen at the ultrastructural
where it can then initiate a phosphorylation cascade level. EM analysis reveals that the length of the SSR,
leading to activation of mitogen-activated protein which is dependent on DLG levels (Budnik et al., 1996),
(MAP) kinase (Cobb and Goldsmith, 1995). MAP ki- is significantly decreased compared to wild type con-
nase has been suggested to be intimately involved in trols. Further, the number of active zones and size of
the induction of long-term potentiation (Bading and boutons is increased, with values similar to those seen
REGULATION OF SYNAPTIC COMPONENTS 19
atively low levels of signal at the NMJ (Koh et al.,
1999). Thus, CaMKII appears to regulate DLG function
by affecting its synaptic localization through phosphor-
ylation (Koh et al., 1999). As phosphorylated DLG is
dissociated from the synaptic terminals, FasII and
other DLG binding partners could then be free to move
away from the NMJ. Intriguingly, X-ray crystallogra-
phy of the third PDZ domain of DLG shows that a 7
amino acid stretch contains both the sequence RGNS,
the CaMKII phosphorylation consensus site, and the
sequence GLG, a motif conserved in all MAGUK PDZ
domains, which is the site to which the C-terminal
S/TXV COOH motifs of MAGUK binding partners bind
(Doyle et al., 1996). Thus, phosphorylation of serine 48
by DCaMKII may alter binding affinities between the
first PDZ domain and the C-terminal S/TXV motifs of
FasII or Shaker-type K⫹ channels.
Fig. 2. Dynamic regulation of DLG at synapses by CaMKII depen-
dent phosphorylation. Increased Ca2⫹ concentration at the postsyn- During larval development, the muscle fibers of
aptic cell activates CaMKII. Activated CaMKII then phosphorylates growing fly larvae increase exponentially in size; syn-
serine 48 of PDZ1 of DLG. Phosphorylated DLG dissociates from the aptic arbors continuously expand to keep pace (Gorc-
synaptic membrane. DLG-binding synaptic components, such as zyca et al., 1993; Guan et al., 1996; Keshishian et al.,
FasII and Shaker, could subsequently be free to move away from
synaptic complexes. Reproduced from Koh et al. (1999) with permis- 1993). Structural plasticity is affected by neuronal ac-
sion of the publisher. tivity levels and by cell adhesion (Budnik et al., 1990;
Schuster et al., 1996a,b). Hyperexcitable mutants that
have increased electrical activity levels have larger and
more elaborately branched NMJs with more boutons
in dlg mutant alleles. Interestingly, when DCaMKII than do wild type larvae (Budnik et al, 1990). A similar
activity is reduced by expression of ala, an opposite phenotype has been shown to be mediated by the level
phenotype is observed—the length of the SSR increases of FasII present at the synapse (Schuster et al., 1996b;
compared to wild type control. Consistent with these Thomas et al., 1997; Zito et al., 1997). FasII mutants
EM results, DLG immunoreactivity at NMJs also de- that have a very low level of FasII expression exhibit
creases by constitutive DCaMKII activation and in- poorly elaborated NMJs with few boutons, while mu-
creased by partial inhibition of DCaMKII activity (Koh tants with a moderate reduction of FasII expression
et al., 1999). These observations suggest that CaMKII show an expansion of the axonal arbor (Schuster et al.,
and DLG are involved in similar aspects of synapse 1996a). These observations suggest a model in which
development, and that CaMKII may regulate DLG synapses are stabilized by the presence of high levels of
function. cell adhesion molecules, but can expand when this
A possible mechanism for this regulation is that restriction is attenuated; partial disassembly of exist-
CaMKII may affect the degree of association of DLG ing stable synapses permits the assembly of new syn-
with the synaptic membrane/cytoskeleton (Fig. 2). The aptic connections.
first PDZ domain of all MAGUKs contains a conserved The regulation of DLG function by CaMKII provides
CaMKII phosphorylation consensus site that appears a potential mechanism by which synaptic activity af-
to be a target for CaMKII. In vitro, this site (serine 48) fects synaptic plasticity. The dissociation of CaMKII-
is phosphorylated by CaMKII, and this phosphoryla- phosphorylated DLG from the synaptic complex allows
tion can be prevented by substituting serine 48 to ala- FasII to diffuse away from the synapse, thus permit-
nine or aspartic acid (Koh et al., 1999). A demonstra- ting the activity-dependent expansion of the NMJ (Koh
tion that phosphorylation of serine 48 is required in et al., 1999).
vivo for regulation of DLG localization at the NMJ was There are several alternative ways by which activity
obtained by using three different enhanced green fluo- may regulate NMJ development. For example overex-
rescence protein (eGFP)-tagged DLG constructs, with pression of frequenin, a member of the Neuronal Cal-
or without point mutations of serine 48. These trans- cium Sensor family, also alters NMJ terminal morphol-
genic DLG forms were expressed in both the pre- and ogy (Angaut-Petit et al., 1998). Whether this effect is
postsynaptic cells in dlg mutant larvae (Koh et al., mediated through the pathways described above is cur-
1999). In one mutant construct (eGFP-DLGS48D) rently not known.
serine 48 was substituted by aspartic acid, mimicking CaMKII appears to be expressed at both pre-and
CaMKII phosphorylation; in the other (eGFP- postsynaptic sites, in vertebrates (reviewed in Green-
DLGS48A) serine 48 was substituted by alanine, pre- gard et al., 1993; Turner et al., 1999) as well as in flies
venting CaMKII-dependent phosphorylation. When (Koh et al., 1999), where it interacts with a number of
wild type eGFP-DLG was expressed, eGFP signal was molecules. In flies, one such substrate is Slowpoke
concentrated at the NMJ, and a low level of fluores- binding protein (Slob). Drosophila Slowpoke (dSlo) is a
cence was observed at extrasynaptic regions. In con- Ca2⫹-dependent potassium channel with a consider-
trast, when eGFP-DLGS48A was expressed, GFP fluo- ably extended carboxy terminal domain (Adelman et
rescence was restricted to the NMJ. However, expres- al., 1992; Atkinson et al., 1991). Slob was identified in
sion of eGFP-DLGS48D resulted in GFP signal a yeast two-hybrid screen using this long carboxy ter-
throughout muscle cytoplasm and membrane, and rel- minal as bait (Schopperle et al., 1998). Using Slob as
20 Y.H. KOH ET AL.

bait, another screen identified the ␨ isoform of 14-3-3, a (Crino et al., 1998), the NMDAR1 subunit (Gazzaley et
protein that regulates presynaptic functions of the al., 1997), and the neurotrophic factors TrkB and
NMJ (Broadie et al., 1997), as a Slob binding protein BDNF (Tongiorgi et al., 1997). The localization of
(Zhou et al., 1999). Binding of the two proteins is me- mRNA at dendritic terminals raises two questions:
diated by a pair of serine-containing motifs in Slob, and How are specific mRNAs translocated to the dendritic
abolished by mutation of the second serine in each of terminals; and what are the mechanisms of local pro-
these motifs. Further, CaMKII-mediated phosphoryla- tein translation at dendritic terminals?
tion of Slob, which dynamically regulates the interac- Insight into the first question can be found in studies
tion of Slob and 14-3-3, is also prevented by mutation of of RNA binding proteins, which have been intensely
the two Slob serines (Zhou et al., 1999). investigated for their roles in embryonic development
Immunocytochemistry shows that 14-3-3, dSlo, and (reviewed by Bashirullah et al., 1998). Specialized lo-
Slob colocalize at presynaptic terminals. dSlo channel calization of mRNAs is required for the proper tempo-
activity is changed when wild type Slob and 14-3-3 are rospatial development of embryos. Restricted spatial
co-expressed; however, this effect is absent when 14- localization of mRNA is achieved by cis-acting RNA
3-3 is co-expressed with the Slob serine mutant. These elements present in the transcript itself, usually lo-
results indicate that presynaptic dSlo/Slob/14-3-3 exist cated in the 3’-untranslated region (UTR) of the tran-
in a regulatory protein complex, mediated by CaMKII- script but sometimes within the 5’-UTR or in the coding
dependent phosphorylation of Slob (Zhou et al., 1999). regions, and a variety of trans-acting factors. Many of
I HEARD IT THROUGH THE AXON: SIGNALS these trans-acting factors are RNA-binding proteins
FROM THE SYNAPSE TO NUCLEUS AND that interact with the transcript (reviewed by Ba-
VICE VERSA shirullah et al., 1998). In some cases, the RNA binding
Local Protein Synthesis at the proteins may regulate mRNA translation and localiza-
Postsynaptic Sites tion at the dendritic terminal by forming mRNA-pro-
tein complexes. For example, BC1 RNA has a 62 nu-
In contrast to short-term synaptic plasticity, long- cleotide dendritic targeting element within its 5’-UTR
term synaptic plasticity requires structural and func- (Muslimov et al., 1997), which has a Testis-Brain RNA
tional changes dependent upon new protein synthesis binding protein (TB-RNB)-binding Y-element (Wu et
(Abel at al., 1998). Several hypotheses, which are not al., 1997).
mutually exclusive, have been suggested to explain Other transcripts, such as Arc mRNA, exhibit activ-
how long-term synaptic plasticity might be accom- ity-dependent localization to activated dendritic termi-
plished.
nals in the presence of inhibitors of protein synthesis,
The synaptic tagging hypothesis is based on the ob-
suggesting that the signal for dendritic localization
servation that inhibition of protein synthesis at den-
exists in the transcript itself. Therefore, newly synthe-
dritic terminals abolishes long-term potentiation (Frey
sized RNA binding proteins are unnecessary for appro-
and Morris, 1997) and facilitation (Martin et al.,
1997a). In this model, newly synthesized proteins from priate mRNA targeting (Steward et al., 1998). Thus,
the neuronal soma are transported down the axon and there appear to exist multiple mRNA translocation
then captured into specific synaptic terminals marked mechanisms.
by synaptic tags (reviewed in Frey and Morris, 1998). Once the mRNA has reached the synapse, it needs to
The nature of these tags has not yet been defined; be translated into proteins required to mediate long-
candidate mechanisms include a change in the geome- term changes in synaptic plasticity. This brings us to
try of the tagged synapse (Rusakov et al., 1995) or the the second question: what are the mechanisms of local
phosphorylation of an early LTP-associated kinase (Os- protein translation at dendritic terminals? The regula-
ten et all, 1996). An alternative hypothesis is that tory mechanisms involved in synaptic mRNA transla-
synthesis of the necessary proteins occurs locally at the tion are as yet poorly understood, although regulation
post-synaptic terminal. This hypothesis is supported of the translation of ␣-CaMKII mRNA in dendritic
by several lines of evidence, including synaptic local- terminals of the mammalian brain has begun to be
ization of several mRNAs and tRNAs (Tiedge and Bro- characterized (Wu et al., 1998). One of the mechanisms
sius, 1996), the existence of polyribosome-like struc- known to be involved is polyadenylation binding pro-
tures and membrane organelles containing Golgi tein (CPEB)-dependent polyadenylation of the 3’-UTR
markers within dendritic terminals (reviewed by of ␣-CaMKII mRNA at dendritic terminals. CPEB-de-
Huang, 1999; Steward, 1997; Tiedge et al., 1999), and pendent regulation of mRNA translation requires the
the rapid increase of CaMKII protein immunoreactiv- presence of a pair of CPE binding sequences upstream
ity at the dendritic terminals within 30 minutes follow- of polyadenylation sequences within 3’-UTR (Wu et al.,
ing repeated tetanic stimulation (Ouyang et al., 1998). 1998).
An impressive array of mRNAs has been found to Local regulation of mRNA translation at the Droso-
have dendritic localization. These include the high mo- phil a larval NMJ is a new area of inquiry, and has not
lecular weight forms of microtubule-associated protein yet been intensively investigated. However, many syn-
2 (MAP2a/b) (Garner et al., 1988), the ␣-CaMKII sub- aptic proteins show polarized localization at postsyn-
unit (Burgin et al., 1990), the inositol 1,4,5-trisphos- aptic elements, suggesting that such regulation may
phate receptor type 1 (Furuichi et al., 1993), neurogra- well exist. Among these are DLG, which is highly en-
nin (Landry et al., 1994), the cytoskeleton-associated riched at type I synaptic boutons (Lahey et al., 1994);
protein Arc (Link et al., 1995; Lyford et al., 1995), the Shaker-type K⫹ channels, partially colocalized with
noncoding short RNA of polymerase III transcript BC1 DLG at postsynaptic sites (Tejedor et al., 1997); FasII,
(Muslimov et al., 1997), the transcription factor CREB concentrated on synaptic borders (Thomas et al., 1997);
REGULATION OF SYNAPTIC COMPONENTS 21
translocate into the nucleus (Martin et al., 1997b). The
expression of immediate early genes, needed for new
protein synthesis after a neuron has been repeatedly
stimulated, has been shown to be regulated by CREB
transcription factors (Bailey et al., 1996; reviewed by
Abel et al., 1998). The injection of antibodies to MAPK
or inhibitors of MAPK kinase into the soma of cultured
Aplysia sensory cells prevents long-term facilitation,
without changing either basal transmission or short-
term facilitation (Martin et al., 1997b). A regulatory
link between MAPK and CREB could explain this re-
sult; CREB2 has a MAPK phosphorylation consensus
site, making it a potential substrate for activated
MAPK that has been translocated into the nucleus
(Martin et al., 1997b). CREB2 dimerizes with CREB1,
blocking CREB1-mediated transcription. MAPK phos-
phorylation of CREB2 may induce the dissociation of
CREB2 from CREB1, permitting CREB1 to form ho-
modimers, which could then be phosphorylated by the
activated PKA catalytic subunit and finally bind to the
cAMP response element (CRE) sites of immediate early
Fig. 3. Polyribosome-like structures are observed within the SSR.
Cross-section of a type Ib bouton showing the presynaptic terminal (b) genes. One of the immediate early gene products, the
with an active zone (AZ), and the postsynaptic SSR. Inset: High C/EBP transcription factor, may be phosphorylated by
magnification view of the boxed region. Arrowheads indicate polyri- a Rsk protein kinase and then begin late gene transla-
bosome-like structures resembling a short string of beads (Koh, un- tion (Martin et al., 1997b; reviewed in Impey et al.,
published data).
1999).
Long-term changes in synaptic plasticity can be
directly observed in Aplysia dissociated cell culture.
and Dorsal and Cactus, enriched postsynaptically Activated MAPK is found in the dendritic terminals
(Cantera et al., 1999). of Aplysia neurons (Bailey et al., 1997). One of its
However, it is distinctly possible that only a small known substrates is the Aplysia cell adhesion mole-
number of synaptic proteins, if any, are translated cule (ApCAM), a structural homologue of mamma-
within the SSR. Studies into the nature of localization lian NCAM and Drosophila FasII (Mayford et al.,
signals for synaptic components such as PSD-95 (Cra- 1992), which has a MAPK consensus site within the
ven et al., 1999) and metabotropic glutamate receptors C-terminal PEST sequence. ApCAM at the neuronal
(Stowell and Craig, 1999) in primary neuronal cultures cell surface is internalized when this site is phos-
indicate that certain protein domains or amino acid phorylated (Bailey et al., 1997). Binding between the
motifs are necessary for the targeting of a protein to pre- and postsynptic elements is weakened by the
postsynaptic sites. downregulation of ApCAM from sensory neuronal
The only evidence in favor of localized postsynaptic surfaces, allowing the expansion of synapses or ar-
protein translation at the fly NMJ is the existence of borizations after long-term facilitation. Consistent
polyribosome-like structures within the SSR, as re- observations have been seen in the Drosophila larval
vealed by EM examination of the larval NMJ (Fig. 3; NMJ. Mutations in dunce, the gene for phosphodies-
Koh and Budnik, unpublished data). Although localiza- terase II, and/or the potassium channel genes ether á
tion of mRNA of specific synaptic proteins at postsyn- gógo or Shaker, induce hyperexcitability (Budnik et
aptic sites has not yet been investigated, these data al., 1990; Ganetzky and Wu, 1983), accompanied by
indicate that some synaptic components may be trans- decreased levels of FasII (Schuster et al., 1996b) and
lated within the SSR. synapse overgrowth (Budnik et al., 1990; Schuster et
al., 1996b; Zhong et al., 1992). However, it has not
Regulation of Gene Expression yet been established that MAPK plays a role in syn-
From Synaptic Terminals aptic plasticity at the Drosophila larval NMJ.
The establishment of long-term memory is predi- In primary cultures of mammalian hippocampal neu-
cated upon the expression of specific genes, and thus rons, CREB is able to carry signals directly from the
requires the transmission of signals from the synapse synapse to the nucleus. CREB mRNA exists within
to the nucleus. Although it is not yet clear which sig- dendritic terminals, where it is translated into protein,
nals are involved in this process, several candidates and can be phosphorylated (Crino et al., 1998). Labeled
have recently emerged, including PKA (reviewed in CREB microinjected into dendritic terminals is rapidly
Abel et al., 1998), MAPK (reviewed in Impey et al., transported into the nucleus (Crino et al., 1998). Thus,
1999), CREB (Crino et al., 1998), and Nuclear Factor CREB protein synthesized in dendrites could poten-
kappa-B (NF␬B) (reviewed by O’Neil and Kaltschmidt, tially directly affect gene expression, serving as a mes-
1997). senger from dendrite to nucleus.
Stimulation of NMDA receptors is known to result in In flies, Dorsal, a member of the Rel family of tran-
activation of PKA and MAPK in the mammalian hip- scription factors, has been detected at the larval NMJ
pocampus (reviewed in Abel at al., 1998), and the acti- (Cantera et al., 1999). Rel proteins are involved in
vated forms of these proteins have been shown to many different cellular, molecular and physiological
22 Y.H. KOH ET AL.

processes such as immune response (reviewed by Hult- it has in synaptic plasticity. The finding of polyribo-
mark, 1994), inflammation, oncogenesis, apoptosis (re- some-like structures within the SSR indicates that one
viewed by Baeuerle and Henkel, 1994), and embryonic function of the SSR may be post-transcriptional regu-
development (reviewed by Belvin and Anderson, 1996). lation of synaptic proteins. Which mRNAs are localized
Dorsal, along with Relish and Dif, is one of three within the SSR, as well as what complement of post-
known Drosophila Rel proteins. All three are homo- transcriptional machineries can be found there to
logues of the mammalian transcription factor nuclear translate and process those messages, is an area of
factor kappa B (NF␬B) (Dushay et al., 1996; Ip et al., inquiry that has scarcely begun.
1993; Steward, 1987). Dorsal function is inhibited by The interplay of molecular and genetic approaches
Cactus, the Drosophila homologue of Inhibitor kappa B available in the fruit fly may provide the key to resolv-
(I␬B) (Geisler et al., 1992; Kidd, 1992). It has been ing this question, and other aspects of regulation of the
proposed that NF␬B and I␬B play important roles in biochemical and molecular components involved in
the mammalian nervous system, and may be involved synaptic plasticity.
in NMDA receptor-mediated synaptic plasticity (Guer-
rini et al., 1995; reviewed by O’Neil and Kaltschmidt, ACKNOWLEDGMENTS
1997). We thank Ulrich Thomas and Michael Gorczyca for
Intriguingly, both Dorsal and Cactus proteins are permission to cite their unpublished results. We thank
strongly enriched in the SSR of type I boutons, with a Michael Gorczyca, Ulrich Thomas, Laurie Quysner,
much lower level of distribution in the cytoplasm and Mary Packard, and David Epstein for their invaluable
nuclei of muscle fibers (Cantera et al., 1999). The ab- comments on this manuscript. L. Sian Gramates is
sence of Dorsal protein leads to abnormal synaptic supported by an NIH predoctoral fellowship (F31
morphology and nuclear localization of Cactus, as well NS10861-01).
as malformations of the body wall muscles and nuclear
hypotrophy (Cantera et al., 1999). These results sug- REFERENCES
gest that Rel family proteins and their I␬B inhibitors Abel T, Martin KC, Bartsch D, Kandel ER. 1998. Memory suppressor
may act as a synapse-to-nucleus signal mediating syn- genes: inhibitory constraints on the storage of long term memory.
Science 279:338 –341.
aptic plasticity. Adelman JP, Shen KZ, Kavanaugh MP, Warren RA, Wu YN Lagrutta
A Bond, CT, North RA. 1992. Calcium-activated potassium chan-
SOMEWHERE OVER THE SYNAPSE: nels expressed from cloned complementary DNA. Neuron 9:209 –
CONCLUDING REMARKS AND FUTURE 216.
DIRECTIONS Anderson MS, Halpern ME, Keshishian H. 1988. Identification of the
neuropeptide transmitter proctolin in Drosophila larval character-
Studies using the Drosophila neuromuscular junc- ization of fiber-specific neuromuscular endings. J Neurosci 8:242–
tion have shown it to be a fruitful model system for 255.
dissecting the workings of synapses. The existence of Angaut-Petit D, Toth P, Rogero O, Faille L, Tejedor FJ, Ferŕus A.
1998. Enhanced neurotransmitter release is associated with reduc-
evolutionarily conserved synaptic components and sig- tion of neuronal branching in a Drosophila mutant overexpresssing
nal transduction pathways in an easily genetically ma- frequenin. Eur J Neurosci 10:423– 434.
nipulated organism make this system an invaluable Atkinson NS, Robertson GA, Ganetzky B. 1991. A component of cal-
tool for investigating the cellular and molecular mech- cium-activated potassium channels encoded by the Drosophila slo
locus. Science 253:551–555.
anisms underlying synaptic plasticity and learning and Atwood HL, Govind CK. and Wu C-F. 1993. Differential ultrastruc-
memory in higher organisms. Further, this system can ture of synaptic terminals on ventral longitudinal muscles in larval
be used to discern the role of mammalian synaptic Drosophila. J Neurobiol 24:1009 –1044.
proteins important in pathological conditions, but Atwood HL, Karunanithi S, Georgiou J, Charlton MP. 1997. Strength
of synaptic transmission at neuromuscular junctions of crustaceans
whose normal function is as yet obscure. For example, and insects in relation to calcium entry. Invert Neurosci 3:81– 87.
amyloid precursor protein (APP), which is cleaved to Bading H, Greenberg ME. 1991. Stimulation of protein tyrosine phos-
yield Alzheimer’s disease-associated ␤-amyloid, has a phorylation by NMDA receptor activation. Science 253:912–914.
Drosophila homologue, amyloid precursor protein-like Baeuerle PA, Henkel T 1994. Function and activation of NF-B in the
immune system. Annu Rev Immunol 12:141–179.
protein (APPL) (Luo et al., 1990). Immunocytochemical Bailey CH, Kandel ER. 1993. Structural changes accompanying mem-
analysis of NMJs of APPL deficient larvae, together ory storage. Ann Rev Physiol 55:397– 426.
with immunocytochemical and ultrastructural analysis Bailey CH, Bartsch D, Kandel ER. 1996. Toward a molecular defini-
of NMJs of larvae in which full-length or truncated tion of long-term memory storage. Proc Natl Acad Sci 93:13445–
13452.
Appl constructs were neurally expressed, indicate that Bailey CH, Kaang BK, Chen M, Martin KC, Lim CS, Casadio A,
APPL may influence synapse formation (Torroja et al., Kandel ER. 1997. Mutation in the phosphorylation sites of MAP
1999). Kinase blocks learning related internalization of apCAM in Aplysia
Here we have reviewed the analysis of an important sensory neurons. Neuron 18:913–924.
Bashirullah A, Cooperstock RL, Lipshitz HD. 1998. RNA localization
synaptic organizer, DLG, a molecule first identified at in development. Annu Rev Biochem 67:335–394.
vertebrate synapses but whose functional significance Belvin MP, Anderson KV. 1996. A conserved signaling pathway: the
in the intact organism was uncovered by the genetic Drosophila Toll-Dorsal pathway. Annu Rev Cell Dev Biol 12:393–
approaches possible in the fly. Genetic analysis has 416.
Bredt DS, Snyder SH. 1992. Nitric oxide, a novel neuronal messenger.
also allowed us understand the role played by CaMKII Neuron, 8:3–11.
in one of the mechanisms by which the localization of Brenman JE, Bredt DS. 1996. Synaptic signaling by nitric oxide. Curr
DLG is regulated. Whether this mechanism is also Opin Neurobiol 7:374 –378.
employed for vertebrate MAGUKs is a question await- Brenman JE, Chao DS, Gee SH, McGee AW, Craven SE, Santillano
DR, Wu Z, Huang F, Xia H, Peters MF, Froehner SC, Bredt DS.
ing future studies. 1996. Interaction of nitric oxide synthase with the postsynaptic
A particularly intriguing and long-standing question density protein PSD–95 and 1-syntrophin mediated by PDZ do-
has been the role of the SSR, and what, if any, function mains. Cell 84:757–767.
REGULATION OF SYNAPTIC COMPONENTS 23
Broadie K, Rushton E, Skoulakis EM.C, Davis RL. 1997. Leonardo, a Frey U, Morris RGM. 1998. Synaptic tagging: implications for late
Drosophila 14 –3–3 protein involved in learning, regulates presyn- maintenance of hippocampal long term potentiation. Trends Neu-
aptic function. Neuron 19:391– 402. rosci 21:181–188.
Budnik V. 1996. Synapse maturation and structural plasticity at Furuichi T, Samon-Chazottes D, Fujino I, Yamada N, Hasegawa M,
Drosophila neuromuscular junctions. Curr Opin Neurobiol 6:858 – Miyaki A, Yoshikawa S, Guenet J-L, Mikoshiba K. 1993. Wide
867. spread expression of inositol 1,4,5-triphosphate receptor type 1 gene
Budnik V, Zhong Y, Wu C -F. 1990. Morphological plasticity of motor (Insp3r1. in the mouse central nervous system. Receptors Chan-
axons in Drosophila mutants with altered excitability. J Neurosci nels, 1:11–24.
10:3754 –3768. Furuyashiki T, Fujisawa K, Fujita A, Madaule P, Uchino S, Mishina
Budnik V, Koh YH, Guan B, Hartmann B, Hough C, Woods D, Gorc- M, Bito H, Narumiya S 1999. Citron, a Rho-target, interacts with
zyca M. 1996. Regulation of synapse structure and function by the PSD–95/SAP–90 at glutamatergic synapses in the thalamus. J Neu-
Drosophila tumor suppressor gene dlg. Neuron 17:627– 640. rosci 19:109 –118.
Burgin KE, Waxham MN, Rickling S, Westgate SA, Mobley WC, Kelly Ganetzky B, Wu C-F. 1983. Neurogenetic analysis of potassium cur-
PT. 1990. In situ hybridization histochemistry of Ca2⫹/calmodulin- rents in Drosophila: synergistic effects on neuromuscular transmis-
dependent protein kinase in developing rat brain. J Neurosci 10: sion in double mutants. J Neurogenet 1:17–28.
1788 –1798. Garcia EP, Mehta S, Blair LA.C, Wells DG, Shang J, Fukushima, T,
Cantera R, Nassel DR. 1992. Segmental peptidergic innervation of Fallon JR, Garner C, Marshall J. 1998. SAP90 binds and clusters
abdominal targets in larval and adult dipteran insects revealed kainate receptors causing incomplete desensitization. Neuron 21:
with an antiserum against leucokinin I. Cell Tissue Res 269:459 – 727–739.
471. Garner C, Kindler S. 1996. Synaptic proteins and the assembly of the
Cantera R, Kozolva T, Barillas-Mury C, Kafatos FC. 1999. Muscle postsynaptic apparatus. Trends Cell Biol 6:429 – 433.
structure and innervation are affected by loss of dorsal in the fruit Garner CC, Tucker RP, Matus A. 1988. Selective localization of mes-
fly Drosophila melanogaster. Mol Cell Neurosci 13:131–141. senger RNA for the cytoskeletal protein MAP2 in dendrites. Nature
Chen HJ, Rojas-Soto M, Oguni A, Kennedy MB. 1998. A synaptic 336:674 – 677.
Ras-GTPase activating protein (p135 SynGAP) inhibited by CaM Gazzaley, A.H, Benson DL, Huntley GW, Morrison JH. 1997. Differ-
kinase II. Neuron 20:895–904. ential subcellular regulation of NMDAR1 protein and mRNA in
Cho K-O Wall JB, Pugh PC, Ito M, Muller SA, Kennedy MB. 1991. The dendrites of dentate gyrus granule cells after perforant path tran-
␣-subunit of type II Ca2⫹/calmodulin dependent protein kinase is section. J Neurosci 17:2006 –2017.
highly conserved in Drosophila. Neuron 7:439 – 450. Geisler R, Bergmann A, Hiromi Y, Nusslein-Volhard C. 1992. cactus,
Cho K-O, Hunt CA, Kennedy MB. 1992. The rat brain postsynaptic a gene involved in dorsoventral pattern formation of Drosophila, is
density fraction contains a homolog of the Drosophila discs-large related to the I kappa B gene family of vertebrates. Cell 71:613–
tumor suppressor protein. Neuron 9:929 –942. 621.
Cobb MH, Goldsmith EJ. 1995. How MAPKinases are regulated. Gorczyca M, Augart C, Budnik V. 1993. Insulin-like receptor and
J Biol Chem 270:14843–14846. insulin-like peptide are localized at neuromuscular junctions in
Coffino P, Bourne HR, Friedrich U, Hochman J, Insel PA, Lemaire I, Drosophila. J. Neurosci, 3692–3704.
Melmon, K.L, Tomkins GM. 1976. Molecular mechanisms of cyclic Gramates LS, Budnik V. 1999a. Assembly and maturation of the
AMP action: a genetic approach. Recent Prog Horm Res 32:669 – Drosophila larval neuromuscular junction. In: Budnik V, Gramates
684. LS, editors. Neuromuscular junctions in Drosophila. San Diego:
Craven SE, El-Husseini AE, Bredt DS. 1999. Synaptic targeting of the Academic Press, Int Rev Neurobiol 43:93–117.
postsynaptic density protein PSD–95 mediated by lipid and protein Gramates LS, Budnik V. 1999b. Genetics Society of America Annual
motifs. Neuron 22:497–509. Drosophila Research Conference 40:a190 (Abstract).
Crino P, Khodakhah K, Becker K, Ginsberg S, Hemby S, Eberwine J. Greengard P, Valtora F, Czernik AJ, Benfenati F. 1993. Synaptic
1998. Presence and phosphorylation of transcription factors in de- vesicle phosphoproteins and regulation of synaptic function. Sci-
veloping dendrites. Proc Natl Acad Sci 95:2313–2318. ence 259:780 –785.
Davis GW, Goodman CS. 1998. Synapse-specific control of synaptic Griffith LC, Greenspan RJ. 1993. The diversity of calcium/calmodulin-
efficacy at the terminals of a single neuron. Nature 392:82– 86. dependent protein kinase II isoforms in Drosophila is generated by
Davis GW, DiAntonio A, Petersen SA, Goodman CS. 1998. Postsyn- alternative splicing of a single gene. J Neurochem 61:1534 –1537.
aptic PKA controls quantal size and reveals a retrograde signal that Griffith LC, Verselis LM, Aitken KM, Kyriacou CP, Danho Wand
regulates presynaptic transmitter release in Drosophila. Neuron Greenspan RJ. 1994. Inhibition of calcium/calmodulin-dependent
20:305–315. protein kinase in Drosophila disrupts behavioral plasticity. Neuron
Davis RL, Cherry J, Dauwalder B, Han PL, Skoulakis E 1995. The 10:501–509.
cyclic AMP system and Drosophila learning. Mol Cell Biochem Guan B, Hartmann B, Koh YH, Gorczyca M, Budnik V. 1996. The
149/150:271–278. Drosophila tumor suppressor gene, dlg, is involved in structural
Dimitratos SD, Woods DF, Bryant PJ. 1997. Camguk Lin–2, and plasticity at a glutamatergic synapse. Curr Biol 6:695–706.
CASK: novel membrane-associated guanylate kinase homologs that Guerrini L, Blasi F, Denis-Donini S. 1995. Synaptic activation of
also contain CaM kinase domains. Mech Dev 63:127–30. NF-kappa B by glutamate in cerebellar granule neurons in vitro.
Downward J, Riehl R, Wu L, Weinberg RA. 1990. Identification of a Proc Natl Acad Sci 92:9077–9081.
nucleotide exchange-promoting activity for p21ras. Proc Natl Acad Hannan F, Zhong Y. 1999. Second messenger systems underlying
Sci 87:5998 – 6002. plasticity at the neuromuscular junction. In: Budnik V, Gramates
Doyle DA, Lee A, Lewis J, Kim E, Sheng M, MacKinnon R. 1996. LS, editors. Neuromuscular junction in Drosophila. San Diego: Ac-
Crystal structures of a complexed and peptide-free membrane pro- ademic Press, Int Rev Neurobiol 43:119 –138.
tein-binding domain: molecular basis of peptide recognition by PDZ. Harish OE, Poo MM. 1992. Retrograde modulation at developing
Cell 85:1067–1076. neuromuscular synapses: involvement of G protein and arachidonic
Dubnau J, Tully T. 1998. Gene discovery in Drosophila: new insight acid cascade. Neuron 9:1201–1209.
for learning and memory. Ann Rev Neurosci 21:407– 444. Hata Y, Butz S, Sudhof TC. 1996. CASK: a novel dlg/PSD95 homolog
Dushay MS, Asling B, Hultmark D. 1996. Origins of immunity: Rel- with an N-terminal calmodulin-dependent protein kinase domain
ish, a compound rel-like gene in the antibacterial defense of Dro- identified by interaction with neurexins. J Neurosci 16:2488 –2494.
sophila. Proc Natl Acad Sci 93:10343–10347. Hawkins RD, Zhuo M, Arancio O. 1994. Nitric oxide and carbon
English JD, Sweatt JD. 1996. Activation of p42 mitogen-activated monoxide as possible retrograde messengers in hippocampal long-
protein kinase in hippocampal long term potentiation. J Biol Chem term potentiation. Neurobiology 25:652– 65.
271:24329 –24332. Hawkins RD, Son H, Arancio O. 1998. Nitric oxide as a retrograde
English JD, Sweatt JD. 1997. A requirement for the mitogen-acti- messenger during long-term potentiation in hippocampus. Prog
vated protein kinase cascade in hippocampal long term potentia- Brain Res 118:155–172.
tion. J Biol Chem 272:19103–19106. Hoskins R, Hajnal AF, Harp SA, Kim SK. 1996. The C. elegans vulval
Fields RD, Itoh K. 1996. Neural cell adhesion molecules in activity- induction gene lin–2 encodes a member of the MAGUK family of cell
dependent development and synaptic plasticity. Trends Neurosci junction proteins. Development, 122:97–111.
19:473– 480 Huang EP. 1999. Synaptic plasticity: regulated translation in den-
Frey U, Morris RGM. 1997. Synaptic tagging and long-term potenti- drites. Curr Biol 9: R168-R170.
ation. Nature 385:533–536. Hultmark D. 1994. Ancient relationship. Nature 367:116 –117.
24 Y.H. KOH ET AL.

Impey S, Obrietan K, Storm DR. 1999. Making new connections: Role Madaule P, Eda M, Watanabe N, Fujisawa K, Matsuoka T, Bito H,
of ERK/MAP Kinase signaling in neuronal plasticity. Neuron 23: Ishizaki T, Narumiya S. 1998. Role of citron kinase as a target of the
11–14. small GTPase Rho in cytokinesis. Nature 394:491– 494.
Ip YT, Reach M, Engstrom Y, Kadalayil L, Cai H, Gonzales-Crespo S, Malenka RC, Kauer JA, Perkel DJ, Mauk MD, Kelly PT, Nicoll RA,
Tatei K, Levine M. 1993. Dif, a dorsal related gene that mediates an Waxham MN. 1989. An essential role for postsynaptic calmodulin
immune response in Drosophila. Cell 75:753–763. and protein kinase activity in long-term potentiation. Nature 340:
Irie M, Hata Y, Takeuchi M, Ichtchenko K, Toyoda A, Hirao K, Takai 554 –557.
Y, Rosahl TW, Sudhof TC. 1997. Binding of neuroligins to PSD–95. Martin KC, Casadio A, Zhu H, Yaping E, Rose JC, Kandel ER. 1997a.
Science 277:1511–1515. Synapse specific, long term facilitation of Aplysia sensory to motor
Jaffrey SR, Snowman AM, Eliasson MJ, Cohen NA, Snyder SH. 1998. synapses: A function for local protein synthesis in memory storage.
CAPON: a protein associated with neuronal nitric oxide synthase Cell 91:927–938.
that regulates its interactions with PSD95. Neuron 20:115–124. Martin KC, Michael D, Rose JC, Barad M, Casadio A, Zhu H, Kandel
Jia X, Gorczyca M, Budnik V. 1993. Ultrastructure of neuromuscular ER. 1997b. MAP Kinase translocates into the nucleus of the pre-
junctions in Drosophila: comparison of wild-type and mutants with synaptic cell and is required for long term facilitation in Aplysia.
increased excitability. J Neurobiol 24:1025–1044. Neuron 18:899 –912.
Kennedy M. 1997. The postsynaptic density at glutamatergic syn- Masuko N, Makino K, Kuwahara H, Fukunaga K, Sudo T, Araki N,
apses. Trends Neurosci 20:264 –268. Yamamoto N, Yamada Y, Miyamoto E, Saya H. 1999. Tissue-spe-
Keshishian H, Chiba A, Chang TN, Halfon MS, Harkins EW, Jarecki cific membrane-associated guanylate kinase protein, with calmod-
J, Wang L, erson M, Cash S, Halpern ME, Johansen J. 1993. ulin and PSD–95/SAP90 a possible regulatory role in molecular
Cellular mechanisms governing synaptic development in Drosoph- clustering at synaptic sites. J Biol Chem 274:5872–5790.
ila melanogaster. J Neurobiol 24:757–787. Mayford M, Barzilai A, Keller F, Schacher S, Kandel ER. 1992. Mod-
Kidd S. 1992. Characterization of the Drosophila cactus locus and ulation of an NCAM-related adhesion molecule with long-term syn-
analysis of interactions between cactus and dorsal proteins. Cell aptic plasticity in Aplysia. Science 272:1020 –1023.
71:623– 635. Mayford M, Bach ME, Kandel ER.1996. CaMKII function in the
Kim E, Niethammer M, Rothschild A, Jan YN, Sheng M. 1995. Clus- nervous system explored from a genetic perspective. Cold Spring
tering of Shaker-type K⫹ channels by interaction with a family of Harbor Symp Quant Biol LXI:219 –224.
membrane-associated guanylate kinases. Nature 378:85– 88. Meyer T, Hanson PI, Stryer L, Schulman H. 1992. Calmodulin trap-
Kim E, Cho KO, Rothschild A, Sheng M. 1996. Heteromultimerization ping by calcium-calmodulin-dependent protein kinase. Science 256:
and NMDA receptor-clustering activity of Chapsyn–110, a member 1199 –1202.
of the PSD–95 family of proteins. Neuron 17:103–113. Migaud M, Charlesworth P, Dempster M, Webster LC, Watabe AM,
Kim E, Naisbitt S, Hsueh YP, Rao A, Rothschild A, Craig AM, Sheng Makhinson M, He Y, Ramsay MF, Morris RG.M, Morrison JH,
M. 1997. GKAP, a novel synaptic protein that interacts with the O’Dell TJ, Grant SGN. 1999. Enhanced long-term potentiation and
guanylate kinase-like domain of the PSD–95/SAP90 family of chan- impaired learning in mice with mutant postsynaptic density–95
nel clustering molecules. J Cell Biol 136:669 – 678. protein. Nature 396:433– 439.
Kim JH, Liao D, Lau L-F, Huganir R 1998. SynGAP: a synaptic Milner B, Squire LR, Kandel ER. 1998. Cognitive neuroscience and
RasGAP that associates with the PSD–95/SAP90 protein family. the study of memory. Neuron 20:445– 468.
Neuron 20:683– 691. Monastriani M, Gorczyca M, Rapus J, Eckert M, White K, Budnik V.
Kistner U, Wenzel BM, Veh RW, Cases-Langhoff C, Garner AM,
1995. Octopamine immunoreactivity in the fruit fly Drosophila
Appeltauer U, Voss B, Gundelfinger ED, Garner CC. 1993. SAP90,
melanogaster. J Comp Neurol 356:275–287.
a rat presynaptic protein related to the product of the Drosophila
Muller BM, Kistner U, Veh RW, Cases-Langhoff C, Becker B, Gun-
tumor suppressor gene dlg-A. J Biol Chem 268:4580 – 4583.
delfinger ED, Garner CC. 1995. Molecular characterization and
Koh YH, Popova G, Thomas U, Griffith LC, Budnik V. 1999. Regula-
spatial distribution of SAP97, a novel presynaptic protein homolo-
tion of DLG localization at synapses by CaMKII-dependent phos-
phorylation. Cell 98:353–363. gous to SAP90 and the Drosophila discs-large tumor suppressor
Kornau HC, Shenker LT, Kennedy MB, Seeburg PH. 1995. Domain protein. J Neurosci 15:2354 –2366.
interaction between NMDA receptor subunits and the postsynaptic Muller BM. Kistner U, Kindler S, Chung WJ, Kuhlendahl S, Fenster
density protein PSD-95. Science 269:1737–1740. SD, Lau LF, Veh RW Huganir RL, Gundelfinger ED, Garner CC.
Kryl D, Yacoubian T, Haapasalo A, Castren E, Lo D, Barker PA. 1999. 1996. SAP102, a novel postsynaptic protein that interacts with
Subcellular localization of full-length and truncated Trk receptor NMDA receptor complexes in vivo. Neuron 17:255–265.
isoforms in polarized neurons and epithelial cells. J Neurosci 19: Muslimov IA, Santi E, Homel P, Perini S, Higgins D, Tiedge H. 1997.
5823–5833. RNA transport in dendrites: a cis-acting targeting element is con-
Kurdyak P, Atwood H L, Stewart BA, Wu C-F. 1994. Differential tained within neuronal BC1 RNA. J Neurosci 17:4722– 4733.
physiology and morphology of motor axons to ventral longitudinal Naisbitt S, Kim E, Weinberg RJ, Rao A, Yang FC, Craig AM, Sheng
muscles in larval Drosophila. J Comp Neurol 350:463– 472. M. 1997. Characterization of guanylate kinase-associated protein, a
Lahey T, Gorczyca M, Jia X, Budnik V. 1994. The Drosophila tumor postsynaptic density protein at excitatory synapses that interacts
suppressor gene dlg is required for normal synaptic bouton struc- directly with postsynaptic density–95/synapse-associated protein
ture. Neuron 13:823– 835. 90. J Neurosci 17:5687–5696.
Landry CF, Watson JB, Kashima T, Campagnoni AT. 1994. Cellular Nguyen QT, Lichtman JW. 1996. Mechanism of synapse disassembly
influences on RNA sorting in neurons and glia: an in situ hybrid- at the developing neuromuscular junction. Curr Opin Neurobiol
ization histochemical study. Mol Brain Res 27:1–11. 6:104 –112.
Link W, Konietzko U, Kauselmann G, Krug M, Schwanke B, Frey U, Nguyen T, Sudhof TC. 1997. Binding properties of neuroligin 1 and
Kuhl D. 1995. Somatodendritic expression of an immediate early neurexin 1beta reveal function as heterophilic cell adhesion mole-
gene is regulated by synaptic activity. Proc Natl Acad Sci 92:5734 – cules. J Biol Chem 272:26032–26039.
5738 Niethammer M, Valtschanoff JG, Kapoor TM, Allison DW, Weinberg
Lisman J. 1994. The CaM kinase II hypothesis for the storage of TM, Craig AM, Sheng M. 1998. CRIPT, a novel postsynaptic protein
synaptic memory. Trends Neurosci 17:406 – 412. that binds to the third PDZ domain of PSD–95/SAP90. Neuron
Lue RA, Brandin E, Chan EP, Branton D. 1996. Two independent 20:693–707.
domains of hDlg are sufficient for subcellular targeting: the PDZ1–2 O’Dell TJ, Hawkins RD, Kandel ER, Arancio O. 1991. Tests of the
conformational unit and an alternatively spliced domain. J Cell Biol roles of two diffusible substances in long-term potentiation: evi-
135:1125–1137. dence for nitric oxide as a possible early retrograde messenger. Proc
Luo L, Martin-Morri LE, White K. 1990. Identification, secretion, and Natl Acad Sci 88:11285–11289.
neural expression of APPL, a Drosophila protein similar to human O’Neill LA, Kaltschmidt. C. 1997. NF-kappa B: a crucial transcription
amyloid protein precursor. J Neurosci 10:3849 –3861. factor for glial and neuronal cell function. Trends Neurosci 20:252–
Lyford GL, Yamagata K, Kaufmann WE, Barnes CA, Sanders LK, 258.
Copeland NG, Gilbert DJ, Jenkins NA, Lanahan AA, Worley PF. Osten P, Valsamis L, Harris A, Sacktor TC. 1996. Protein synthesis-
1995. Arc, a growth factor and activity-regulated gene, encodes a dependent formation of protein kinase Mzeta in long-term potenti-
novel cytoskeleton-associated protein that is enriched in neuronal ation. J Neurosci 16:2444 –2451.
dendrites. Neuron 15:433– 445. Ouyang Y, Kantor D, Harris KM, Schuman EM, Kennedy MB. 1997.
Madaule P, Furuyashiki T, Reid T, Ishizaki T, Watanabe G, Morii N, Visualization of the distribution of autophosphorylated calcium/
Narumiya S. 1995. A novel partner for the GTP-bound forms of rho calmodulin-dependent protein kinase II after tetanic stimulation in
and rac. FEBS Lett 377:243–248. the CA1 area of the hippocampus. J Neurosci 14:5416 –5427.
REGULATION OF SYNAPTIC COMPONENTS 25
Park JH, Straub VA, O’Shea M. 1998. Anterograde signaling by nitric Tiedge H, Brosius J 1996. Translational machinery in hippocampal
oxide: characterization and in vitro reconstitution of an identified neurons in culture. J Neurosci 16: 7171–7181.
nitrergic synapse. J Neurosci 18:5463–5476. Tongiorgi E, Righi M, Cattaneo A. 1997. Activity-dependent dendritic
Petersen S,A, Fetter RD, Noordermeer JN, Goodman CS, DiAntonio targeting of BDNF and TrkB mRNAs in hippocampal neurons.
A. 1997. Genetic analysis of glutamate receptors in Drosophila J Neurosci 17:9492–9505.
reveals a retrograde signal regulating presynaptic transmitter re- Torroja L, Packard M, Gorczyca M, White K, Budnik V. 1999. The
lease. Neuron 19:1237–1248. Drosophila beta-amyloid precursor protein homolog promotes syn-
Regulski M, Tully T. 1995. Molecular and biochemical characteriza- apse differentiation at the neuromuscular junction. J Neurosci 19:
tion of dNOS: A Drosophila Ca2⫹/calmodulin-dependent nitric oxide 7793–7803.
synthase. Proc Natl Acad Sci 92: 9072–9076. Trahey M, Wong G, Halenbeck R, Rubinfeld B, Martin GA, Ladner M,
Rheuben MB, Yoshihara M, Kidokoro Y. 1999. Ultrastructural corre- Long CM, Crosier WJ, Watt K, Koths K, McCormick F. 1988.
lates of neuromuscular junction development. In: Budnik V, Gra- Molecular cloning of two types of GAP complementary DNA from
mates LS, editors. Neuromuscular junctions in Drosophila. San human placenta. Science 242:1697–1700.
Diego: Academic Press, Int Rev Neurobiol 43:69 –92. Turner KM, Burgoyne RD, Morgan A. 1999. Protein phosphorylation
Rusakov DA, Stewart MG, Sojka M, Richter-Levin G, Bliss TV. 1995. and the regulation of synaptic membrane traffic. TINS 22:459 – 464.
Dendritic spines form ’collars’ in hippocampal granule cells. Neu- Vogel US, Dixon RA, Schaber MD, Diehl RE, Marshall MS, Scolnick
roreport 6:1557–1561. EM, Sigal IS, Gibbs JB. 1988. Cloning of bovine GAP and its
Sattler R, Xiong Z, Lu W, Hafner M, MacDonald J F, Tymianski M. interaction with oncogenic ras p21. Nature 335:90 –93.
1999. Specific coupling of NMDA receptor activation to nitric oxide Wang J, Renger JJ, Griffith LC, Greenspan RJ, Wu C-F. 1994. Con-
neurotoxicity by PSD–95 Protein. Science 284:1845–1848. comitant alteration of physiological and developmental plasticity in
Schopperle WM, Holmqvist MH, Zhou Y, Wang J, Wang Z, Griffith Drosophila CaMKII inhibited synapses. Neuron 13:1373–1384.
LC, Keselman I, Kusinitz F, Dagan D, Levitan IB. 1998. Slob, a Wang Z Palmer G, Griffith LC. 1998. Regulation of Drosophila Ca2⫹/
novel protein that interacts with the Slowpoke calcium-dependent calmodulin-dependent protein kinase II by autophosphorylation an-
potassium channel. Neuron 20:565–573. alyzed by site-directed mutagenesis. J Neurochem 71:378 –387.
Schuster CM, Ultsch A, Schloss P, Cox JA, Schmitt B, Betz H. 1991. Wigler MH. 1990. Oncoproteins. GAPs in understanding Ras. Nature
Molecular cloning of an invertebrate glutamate receptor subunit 346:696 – 697.
expressed in Drosophila muscle. Science 254:112–114. Wildemann B, Bicker G. 1999a. Nitric oxide and cyclic GMP induce
Schuster C.M, Davis GW, Fetter RD, Goodman CS. 1996a. Genetic vesicle release at Drosophila neuromuscular junction. J Neurobiol
dissection of structural and functional components of synaptic plas- 39:337–346.
ticity. I. Fasciclin II controls synaptic stabilization and growth. Wildemann B, Bicker G 1999b) Developmental expression of nitric
Neuron 17:641– 654 oxide/cyclic GMP synthesizing cells in the nervous system of Dro-
Schuster C.M, Davis GW, Fetter RD, Goodman CS. 1996b. Genetic sophila melanogaster. J Neurobiol 38:1–15.
dissection of structural and functional components of synaptic plas- Woods DF, Bryant PJ. 1991. The discs-large tumor suppressor gene of
ticity. II. Fasciclin II controls presynaptic structural plasticity. Drosophila encodes a guanylate kinase homolog localized at septate
Neuron 17:655– 667. junctions. Cell 66:451– 464.
Shen K, Meyer T. 1999. Dynamic control of CaMKII translocation and Woods DF, Bryant PJ. 1993. ZO1 DlgA and PSD–95/SAP90: homolo-
localization in hippocampal neurons by NMDA receptor stimula- gous proteins in tight, septate and synaptic cell junctions. Mech Dev
tion. Science 284:162–166. 44:85– 89.
Sheng M. 1996. PDZs and receptor/channel clustering: rounding up Wu X -Q, Gu W, Meng X, Hecht NB. 1997. The RNA-binding protein
the latest suspects. Neuron 17:575–578. TB-RBP, is the mouse homologue of translin, a recombination pro-
Steward O. 1997. mRNA localization in neurons: a multipurpose tein associated with chromosomal translocations. Proc Natl Acad
mechanism? Neuron 18:9 –12. Sci 94:5640 –5645.
Steward O, Wallace C S, Lyford G L, Worley P F. 1998. Synaptic Wu L, Wells D, Tay J, Mendis D, Abbott M-A, Barnitt A, Quinlan E,
activation causes the mRNA for the IEG arc to localize selectively Heynen A, Fallon JR, Richter JD. 1998. CPEB-mediated cytoplas-
near activated postsynaptic sites on dendrites. Neuron 21:741–751. mic polyadenylation and the regulation of experience-dependent
Steward R 1987. Dorsal, an embryonic polarity gene in Drosophila, is translation of CaMKII mRNA at synapses. Neuron 21:1129 –1139.
homologous to the vertebrate proto-oncogene, c-rel. Science 238: Zhang W, Vazquez L, Apperson M, Kennedy MB. 1999. Citron binds
692– 694. to PSD–95 at glutamatergic synapses on inhibitory neurons in the
Stowell JN, Craig AM. 1999. Axon/dendrite targeting of metabotropic hippocampus. J Neurosci 19:96 –108.
glutamate receptors by their cytoplasmic carboxy-terminal do- Zhong Y, Pena L. 1995. A novel synaptic transmission mediated by a
mains. Neuron 22:525–536. PACAP-like neuropeptide in Drosophila. Neuron 15:2354 –2366.
Takeuchi M, Hata Y, Hirao K, Toyoda A, Irie M, Takai Y. 1997. Zhong Y, Wu C-F. 1991. Altered synaptic plasticity in Drosophila
SAPAPs. A family of PSD–95/SAP90-associated proteins localized memory mutants with a defective cyclic AMP cascade. Science,
at postsynaptic density. J Biol Chem 272:11943–11951. 251:198 –201.
Tejedor FJ, Bokhari A, Rogero O, Gorczyca M, Zhang J, Kim E, Sheng Zhong Y, Budnik V, Wu C-F. 1992. Synaptic plasticity in Drosophila
M and Budnik V. 1997. Essential role for dlg in synaptic clustering memory and hyperexcitable mutants: role of cAMP cascade. J Neu-
of Shaker K⫹ channels in vivo. J Neurosci 17:152–159. rosci 12:644 – 651.
Tezuka T, Umemori H, Akiyama T, Nakanishi S, Yamamoto T. 1999. Zhou Y, Schopperle WM, Murrey H, Jaramillo A, Dagan D, Griffith
PSD–95 promotes Fyn-mediated tyrosine phosphorylation of the LC, Levitan IB. 1999. A dynamically regulated 14 –3–3 Slob, and
N-methyl-D-aspartate receptor subunit NR2A. Proc Natl Am Sci Slowpoke potassium channel complex in Drosophila presynaptic
96:435– 440. nerve terminals. Neuron 22:809 – 818.
Thomas U, Kim E, Kuhlendahl S, Koh YH, Gundelfinger ED, Sheng Zipkin ID, Kindt RM, Kenyon CJ. 1997. Role of new Rho family
M, Garner CC, Budnik V. 1997. Synaptic clustering of the cell member in cell migration and axon guidance in C. elegans. Cell
adhesion molecule Fasciclin II by Discs-Large and its role in the 90:883– 894.
regulation of presynaptic structure. Neuron 19:787–799. Zito K, Fetter RD, Goodman CS, Isacoff EY. 1997. Synaptic clustering
Tiedge H, Bloom FE, Richter D 1999. RNA, whither goest thou? of Fasciclin II and Shaker: Essential targeting sequences and role of
Science, 283:186 –187. Dlg. Neuron 19:1007–1016.

You might also like