You are on page 1of 21

INVITED REVIEW ABSTRACT: The release of neurotransmitter from neurons represents one

of the pivotal events in synaptic transmission. Neurotransmitters are re-


leased from synaptic vesicles in presynaptic neurons in response to neural
activity, diffuse across the synaptic cleft, and bind specific receptors in order
to bring about changes in postsynaptic neurons. Some of the molecular
processes that govern neurotransmitter release are now becoming better
understood. The steps involved can be broken down into two partially over-
lapping presynaptic cycles, the neurotransmitter cycle and the synaptic
vesicle cycle. The neurotransmitter cycle involves transmitter biosynthesis,
storage, reuptake, and degradation. The synaptic vesicle cycle involves tar-
geting to the nerve terminal, docking, fusion, endocytosis, and recycling.
Biochemical and structural studies have yielded important insight into our
understanding of each of these two cycles. Further, both pharmacological
and genetic interference with either of these cycles results in profound al-
terations in synaptic transmission and behavior, demonstrating the crucial
role of neurotransmitter release.
© 2001 John Wiley & Sons, Inc. Muscle Nerve 24: 581–601, 2001

MOLECULAR MECHANISMS OF
NEUROTRANSMITTER RELEASE
EDWARD A. FON, MD, FRCP(C),1 and ROBERT H. EDWARDS, MD2

1
Centre for Neuronal Survival, Montreal Neurological Institute, McGill University,
3801 University Street, Montreal, Quebec H3A 2B4, Canada
2
Departments of Neurology and Physiology, UCSF School of Medicine,
San Francisco, California, USA

Accepted 1 December 2000

Chemical signaling between neurons and their tar- diffuses across the synaptic cleft and transduces the
get cells mediates information processing in the ner- physiological signal by binding to postsynaptic recep-
vous system. Neurotransmitter released from a pre- tors. The receptors thus determine the nature of the
synaptic neuron in response to neural activity physiological signal. Classical neurotransmitters
such as ␥-aminobutyric acid (GABA), glutamate, and
acetylcholine (ACh) activate ion channels and hence
Abbreviations: ACh, acetylcholine; AChE, acetylcholinesterase; ALS, mediate fast synaptic transmission. In contrast, neu-
amyotrophic lateral sclerosis; AP, clathrin adaptor protein; ATP, adeno- romodulators, such as monoamines and peptides (as
sine triphosphate; BNPI, brain-specific sodium-dependent inorganic
phosphate cotransporter; CaMK, Ca2+/calmodulin-dependent protein ki- well as GABA, glutamate, and ACh), activate G-
nase; CAPS, Ca2+-dependent activator of secretion; cDNA, complemen- protein–coupled receptors, which activate second
tary DNA; ChAT, choline acetyltransferase; CHT1, choline transporter 1;
COMT, catechol-o-methyl transferase; CSP, cysteine string protein; DA, messengers and act on a much longer time scale.
dopamine; DAT, plasma membrane DA transporter; DAG, diacylglycerol; Chemical neurotransmission is particularly well
DBH, dopamine ␤-hydroxylase; EAAT, excitatory amino acid transporter;
ER, endoplasmic reticulum; GABA, ␥-aminobutyric acid; GAD, glutamic suited for the complex computation performed by
acid decarboxylase; GAT, plasma membrane GABA transporter; GDI, the nervous system. Compared to other modes of
guanine nucleotide dissociation inhibitor; GTP, guanosine triphosphate;
5-HT, serotonin; LDCV, large, dense-cored vesicle; LTP, long-term poten- communication, such as electrical transmission, syn-
tiation; MAO, monoamine oxidase; MPP+, 1-methyl-4-phenylpyridinium; aptic transmission allows greater flexibility and regu-
MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; NE, norepinephrine;
NET, plasma membrane NE transporter; NSF, N-ethylmaleimide-sensitive lation. Indeed, the signal produced by chemical neu-
factor; PKA, cAMP-dependent protein kinase A; PKC, protein kinase C; PI, rotransmission decays very little and can vary in
phosphatidylinositol; PITP, phosphatidylinositol transfer protein; SERT,
plasma membrane 5-HT transporter; SLMVs, synaptic-like microvesicles; intensity, in speed, in the effect on membrane po-
SN1, System N transporter; SNARE, SNAP receptor; ␣-SNAP, soluble NSF tential, and in the modulation of second messenger
attachment protein; SV, synaptic vesicle; TGN, trans-Golgi network; TH,
tyrosine hydroxylase; VAChT, vesicular ACh transporter; VGAT, vesicular pathways. This flexibility contributes to synaptic plas-
GABA transporter; VMAT, vesicular monoamine transporter ticity and, at many synapses, prior activity influences
Key words: synaptic transmission; exocytosis; synaptic vesicles; trans-
porters; quantal release synaptic function, providing a mechanism for learn-
Correspondence to: E.A. Fon; e-mail: ted@mni.lan.mcgill.ca ing and memory. However, appropriate synaptic
© 2001 John Wiley & Sons, Inc. function requires the precise coupling of transmitter

Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001 581


release to neural activity. The biochemical processes centrations of neurotransmitter relative to the cyto-
that mediate neurotransmitter release are thus cru- plasm. Because classical transmitters are made in the
cial for information processing. cytoplasm, SV filling requires active transport from
Insight into the mode of neurotransmitter re- the cytoplasm. Further, the high rates of exocytosis
lease initially derived from the work of Bernard Katz observed at many synapses require a constant infu-
on the frog neuromuscular junction. Katz showed sion of cytoplasmic transmitter to maintain SV fill-
that release occurs in packets or quanta containing ing. Although biosynthesis produces the various
several thousand molecules of transmitter.114 It has transmitters de novo, plasma membrane reuptake
since become clear that synaptic release in the cen- systems recycle the released transmitter and the re-
tral nervous system is also quantal in nature.108 Fur- cycled transmitter generally contributes much more
ther, one quantum corresponds to the release of a to SV filling than newly synthesized transmitter. In
single synaptic vesicle (SV) filled with neurotransmit- addition, transmitter degradation by metabolic en-
ter.99 SVs accumulate neurotransmitter to high con- zymes and their removal from the extracellular space
centrations133 and cluster in close proximity to the by reuptake systems both terminate signaling.
sites of release at active zones in the presynaptic The SV cycle involves targeting vesicles to the
nerve terminal.35,223 Neural activity then causes the nerve terminal where docking, fusion, endocytosis,
exocytotic fusion of SVs with the plasma membrane, and recycling occur. High rates of exocytosis require
resulting in transmitter release. the efficient regeneration of SVs at the nerve termi-
Neurotransmitter release involves two partially nal. Long distances from the cell body make it im-
overlapping cycles, the neurotransmitter cycle and possible to replenish released SVs by retrieval of
the SV cycle (Fig. 1).269 The neurotransmitter cycle their constituents to the Golgi complex. Rather, SVs
involves transmitter biosynthesis, storage, reuptake, recycle locally, in the nerve terminal. The local re-
and degradation. SVs contain extremely high con- cycling nonetheless involves sorting of SV proteins

FIGURE 1. A model illustrating the relationship between the neurotransmitter (nt) cycle and the synaptic vesicle (sv) cycle at the nerve
terminal. The nt cycle involves transmitter biosynthesis, storage, reuptake, and degradation (red arrows). The sv cycle involves targeting
vesicles to the nerve terminal where docking, fusion, endocytosis, and recycling occur (green arrows). High rates of exocytosis require
the efficient packaging of neurotransmitter into SVs (vesicular neurotransmitter transporters, shown as blue triangles) as well as rapid
recycling of SVs at the nerve terminal, either directly from the plasma membrane or through an endosomal intermediate. The local
recycling involves sorting of SV proteins from plasma membrane proteins, clathrin-mediated endocytosis (blue), as well as docking of SVs
at the plasma membrane. Regulated fusion occurs in response to locally elevated Ca2+ entering the nerve terminal through voltage-gated
Ca2+ channels (orange), which cluster near the site of vesicle fusion.

582 Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001


from plasma membrane and other proteins, as well fers substantially for distinct classes of neurotrans-
as the docking of SVs at the plasma membrane and mitters, reflecting differences in their mode of ac-
regulated fusion in response to an intracellular sig- tion. Classical neurotransmitters such as ACh,
nal, usually elevated Ca2+. Importantly, pharmaco- GABA, and glutamate recycle locally in the nerve
logical or genetic interference with either the neu- terminal, where they accumulate in SVs. SVs un-
rotransmitter or SV cycle profoundly alters synaptic dergo exocytosis within a specialized domain of the
transmission and behavior, demonstrating the im- plasma membrane overlying the synapse known as
portance of each for signaling by neurons. Molecu- the active zone, consistent with the role of classical
lar and biochemical studies have now begun to illu- transmitters in rapid, precise computational signal-
minate the cellular mechanisms involved in these ing. In contrast, neural peptides are synthesized as
two cycles. larger precursor proteins in the lumen of the endo-
plasmic reticulum (ER). They transit through the
THE NEUROTRANSMITTER CYCLE Golgi complex and sort directly from the trans-Golgi
Modulation of the neurotransmitter cycle can im- network (TGN) into large, dense-cored vesicles
pact neurotransmitter release and signaling in two (LDCVs, or secretory granules in endocrine cells)
important ways. First, it can affect signaling qualita- where they undergo proteolytic processing into ma-
tively by determining the site and the mode of neu- ture, biologically active peptides. Like SVs, LDCVs
rotransmitter release. Second, it can exert a pro- undergo regulated exocytosis in response to elevated
found influence on the magnitude of signaling by cytoplasmic Ca2+. However, unlike SVs, LDCVs fuse
regulating the quantity of neurotransmitter available with the plasma membrane at virtually any site in the
for release. For each neurotransmitter, analogous, cell body or dendrite as well as the nerve terminal
albeit distinct, sets of proteins carry out the distinct and hence are not restricted to an active zone. The
steps in the cycle (Table 1). This diversity allows flex- exocytosis of LDCVs also requires stronger stimula-
ibility in how release is regulated for each of the tion than the release of SVs and takes place over a
neurotransmitters. The diversity contrasts to the rela- longer time scale, consistent with the role of neural
tively conserved processes implicated in the SV cycle, peptides as modulators of synaptic activity rather
which are similar not only in different types of neu- than as fast neurotransmitters. Monoamines appear
rons but also for distinct organelle fusion and bud- to represent a special case because, like other clas-
ding events within the same cell. sical transmitters, their synthesis occurs in the cyto-
plasm, but they undergo storage and release from
Site and Mode of Neurotransmitter Release. No LDCVs as well as SVs. Indeed, monoamine release
feature more clearly defines the phenotype of a neu- from these two different compartments presumably
ron than the identity of its neurotransmitter. Trans- subserves distinct roles in signaling.64
mitter identity depends on the expression of specific
biosynthetic, storage, reuptake, and degradation ma- Amount of Neurotransmitter Release. Changes in
chinery. The localization of this machinery within the rate of transmitter biosynthesis, storage, reup-
neurons in turn determines the site and mode of take, and degradation each have the potential to in-
neurotransmitter release. Indeed, the machinery dif- fluence extracellular neurotransmitter concentra-

Table 1. Proteins involved in the neurotransmitter cycle.

Biosynthesis Vesicular uptake Storage vesicle Reuptake Degradation

Monoamines
Norepinephrine D␤H VMAT2 LDCVs NET MAO and COMT
Dopamine TH VMAT2 SVs and LDCVs DAT MAO and COMT
Serotonin TPH VMAT2 SVs SERT MAO
Acetylcholine ChAT VAChT SVs CHT1 AChE
Amino acids
GABA GAD65, 67 VGAT SVs GAT GABA transaminase
Glutamate Glutaminase VGLUT1 SVs EAATs Glutamine synthetase
Neural peptides ER and Golgi None LDCVs None Proteases

AChE, acetylcholinesterase; ChAT, choline acetyltransferase; CHT1, choline transporter 1; COMT, catechol-o-methyltransferase; DAT, dopamine
transporter; D ␤H, dopamine ␤-hydroxylase; EAATs, excitatory amino acid transporters; ER, endoplasmic reticulum; GAD, glutamic acid decarboxylase;
GAT, GABA transporter; LDCVs, large, dense-cored vesicles; MAO, monoamine oxidase; NET, norepinephrine transporter; SERT, serotonin transporter;
SVs, synaptic vesicles; TH, tyrosine hydroxylase; TPH, tryptophan hydroxylase; VAChT, vesicular acetylcholine transporter; VGAT, vesicular GABA
transporter; VGLUT1, vesicular glutamate transporter; VMAT2, vesicular monoamine transporter 2.

Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001 583


tions, and hence signaling. Until recently, however, ylase (GAD), which occurs in two distinct isoforms in
the role of these activities in regulating transmitter the mammalian brain, GAD65 and GAD67. Al-
release has remained unclear. Indeed, it had been though GAD65 is by far the most abundant isoform,
widely held that the amount of neurotransmitter re- GAD67 appears responsible for most GABA biosyn-
leased from individual vesicles vastly exceeds the thesis.212 GABA levels remain normal in GAD65
amount required to saturate postsynaptic receptors, knockout mice but fall to 10% of wild-type litter-
precluding a regulatory role for this machinery. On mates in GAD67 knockout animals. Nonetheless,
the other hand, recent experiments suggest that re- GAD65 associates with brain membranes through an
ceptors at central synapses are often not saturated by N-terminal domain and appears more tightly regu-
single release events.140 Similarly, at the developing lated than GAD67, which occurs throughout the cy-
neuromuscular junction, increasing SV filling with toplasm of inhibitory neurons. It has therefore been
ACh increases the amplitude of quantal events re- suggested that GAD67 contributes to GABA synthesis
corded from the muscle.220 Further, the amount of for general metabolic activity, such as flux through
neurotransmitter stored in individual SVs has gener- the tricarboxylic acid cycle, whereas GAD65 contrib-
ally been considered invariant. However, recent utes to synaptic transmission. Indeed, GAD65 knock-
work demonstrating the variable neurotransmitter out mice do not display major behavioral defects but
content of individual vesicles, even within the same rather have seizures and increased anxiety,112,113
terminal, challenges this view.131 Individual steps in presumably related to a reduction in GABA re-
the neurotransmitter cycle may therefore have an lease.238 Interference with GAD in humans also im-
important influence, not only on the site and mode pairs synaptic transmission with important conse-
of transmitter release, but also on the amount of
quences on health. Indeed, about 90% of patients
neurotransmitter released. The observed effects on
with stiff-person syndrome have autoantibodies di-
postsynaptic signaling support the significance of
rected against GAD, resulting in impaired GABAer-
this form of regulation.
gic transmission and continuous axial muscle con-
tractions.33,213,214
Neurotransmitter Biosynthesis. Monoamines.
Acetylcholine. Choline acetyltransferase (ChAT),
Regulation of the biosynthetic pathway influences
the enzyme responsible for ACh synthesis, also asso-
monoamine release. In mice, disruption of the gene
ciates with membranes in the nerve terminal.259 De-
encoding tyrosine hydroxylase, the rate-limiting en-
tergent extraction shows that the membrane associa-
zyme in catecholamine biosynthesis, results in
midgestational lethality, illustrating the critical role tion depends on amphipathic rather than ionic or
of transmitter release for survival.267 In addition, ty- covalent interactions.167 Further, ChAT may specifi-
rosine hydroxylase undergoes intricate control at the cally localize to SVs,37 suggesting a mechanism to
transcriptional and posttranslational levels, includ- couple ACh biosynthesis with storage, although the
ing modulation by cofactors, feedback inhibition of physiological significance of the membrane associa-
the enzyme by catecholamine products, and, impor- tion remains unclear.
tantly, phosphorylation.120 Indeed, phosphorylation
by multiple protein kinases appears to couple tyro- Neurotransmitter Storage. Large, Dense-Cored
sine hydroxylase activity with synaptic activity as mea- Vesicles. Neural peptides and classical neurotrans-
sured by autoreceptor stimulation.268 Activation of mitters differ in their site of storage. As noted pre-
D2-like dopamine receptors in particular inhibits ad- viously, the storage of neural peptides occurs in
enylcyclase, reduces the activity of cAMP-dependent LDCVs (or secretory vesicles in endocrine cells).
protein kinase A (PKA), and hence reduces the Precursors to the neural peptides translocate into
phosphorylation of tyrosine hydroxylase, which nor- the lumen of the endoplasmic reticulum during
mally serves to increase enzyme activity. In addition, translation and hence already reside within the lu-
the enzyme, dopamine ␤-hydroxylase (DBH), which men of the secretory pathway. After transit through
converts dopamine (DA) to norepinephrine (NE), the Golgi complex, the proteins sort selectively into
resides inside secretory vesicles. NE biosynthesis LDCVs through unknown mechanisms prior to regu-
therefore requires prior DA transport into vesicles, lated exocytosis. Cleavage of the precursors by endo-
and the targeting of this and other biosynthetic en- proteases such as furin and prohormone convertases
zymes to secretory vesicles presumably contributes to may promote sorting to LDCVs or the regulated (as
their role in neurotransmitter release. opposed to constitutive) secretory pathway.153 In-
GABA. The conversion of glutamic acid to deed, it has been proposed that sorting to LDCVs
GABA requires the enzyme, glutamic acid decarbox- involves interactions with lumenal granule proteins

584 Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001


or membrane lipids42,119 rather than with a cytoplas- amine transport also predict polytopic membrane
mic sorting machinery that mediates most other proteins with 12 transmembrane domains and de-
membrane-trafficking events within the cell. fine a new family of proteins that includes the ve-
Synaptic Vesicles. In contrast to neural peptides, sicular ACh transporter (VAChT)66,184 as well as the
classical neurotransmitters are synthesized in the cy- the nonneural VMAT1 and neuronal VMAT2.65,134
toplasm and therefore need to be packaged into the Interestingly, reserpine inhibits both VMATs,
lumen of SVs prior to release. Since neurotransmit- whereas tetrabenazine inhibits primarily VMAT2,169
ter concentrations within the lumen of neurosecre- possibly accounting for the more substantial side-
tory vesicles can approach the molar range, storage effects of reserpine.
requires active transport across the membrane of the Genetic manipulation in mice indicates that very
vesicle. Indeed, previous biochemical and pharma- little redundancy exists at the step of vesicular trans-
cological studies have identified four distinct trans- port. VMAT2 knockout mice move very little spon-
port activities expressed by native brain synaptic taneously, do not feed, and die within a few days
vesicles — for monoamines, ACh, GABA, and gluta- after birth.75,230,251 Neurons cultured from the mid-
mate.133 All of these activities rely on the proton brain of these mice are unable to package mono-
electrochemical gradient generated by a vacuolar amines into synaptic vesicles and therefore cannot
H+–adenosine triphosphatase (ATPase) that pumps release the transmitter in response to neural activ-
protons into the vesicles. In particular, transport in- ity.75 Further, cultured midbrain neurons from
volves the exchange of lumenal protons for cytoplas- VMAT2+/− mice that express half the wild-type levels
mic neurotransmitter. Drugs that interfere with ve- of VMAT2 protein, store and release half as much
sicular transport also have the potential to modulate dopamine as wild-type animals,75 indicating that
neurotransmitter storage and release, and thus influ- VMAT2 limits monoamine storage and release.
ence behavior. Indeed, reserpine and tetrabenazine, Overexpression of VMAT2 and VAChT in cultured
which directly inhibit vesicular monoamine trans- neurons also increases quantal size by several-
port, have profound effects on behavior by depleting fold,175,220 suggesting that vesicular transport limits
the pool of stored monoamines, resulting in a clini- release even under physiological conditions.181
cal state resembling depression and Parkinson’s dis- Overexpression increases the frequency of release
ease.76,83 Recent cloning of the proteins responsible events as well, suggesting an increase in the number
for vesicular monoamine, ACh, GABA and gluta- of vesicles containing transmitter. The expression of
mate transport activities now allows a molecular vesicular transport activity may thus determine not
analysis of their role in transmitter release. only the amount of neurotransmitter stored per
VMATs and VAChT. Selection in the potent par- vesicle but also the number of filled vesicles available
kinsonian neurotoxin, 1-methyl-4-phenylpyridinium for release.
(MPP+; the active metabolite of 1-methyl-4-phenyl- Expression of vesicular transport proteins on
1,2,3,6-tetrahydropyridine [MPTP]), led to identifi- different types of storage vesicles has the potential
cation of the first vesicular neurotransmitter trans- to influence the site and mode of release as well as
porter, vesicular monoamine transporter 1 the amount of transmitter released. Classic studies
(VMAT1). Both VMAT1 and VMAT2 protect against have suggested that rat pheochromocytoma PC12
MPP+ by sequestering the toxin, like normal mono- cells release monoamines from LDCVs and ACh
amine substrates, inside secretory vesicles and away from synaptic-like microvesicles (SLMVs),14 suggest-
from its primary site of action in the mitochon- ing the differential localization of VMAT2 and
dria.135 Interestingly, the sequence indicates similar- VAChT. Indeed, VAChT occurs mostly on SVs in the
ity to a family of bacterial antibiotic resistance pro- brain,81,254 whereas VMAT2 occurs on both SVs and
teins that extrude antibiotics and also act as proton LDCVs.158–160 In PC12 cells, VMATs reside almost
exchangers that show sensitivity to reserpine.134 The exclusively on LDCVs, whereas VAChT localizes pref-
VMATs thus appear to have evolved from ancient erentially to SLMVs.132,254 Since LDCVs do not re-
detoxification mechanisms and may have two dis- lease at a specialized active zone, and release more
tinct roles, one in transmitter release and the other slowly and in response to different stimuli than SVs,
in detoxification. The VMATs may indeed protect the localization of the transport proteins presumably
against an endogenous toxin, such as the normal contributes to important differences in the mode of
neurotransmitter dopamine, which is quite toxic to signaling by monoamines and ACh. The strong se-
many cells in culture. A defect in VMAT function quence similarity between VMAT2 and VAChT has
may thus predispose one to Parkinson’s disease. The further enabled the identification of critical regions
sequences of the cDNAs encoding vesicular mono- responsible for the differential targeting.246 Indeed,

Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001 585


acidic residues upstream of dileucine motifs present tions both as an inorganic phosphate transporter
in the cytoplasmic C-termini of both proteins appear when it is translocated to the plasma membrane after
important for the differential localization to LDCVs exocytosis and as a vesicular glutamate transporter
and SVs.233 Interestingly, VAChT also undergoes when internalized on SVs.
phosphorylation on a residue upstream of its dileu-
cine motif, and this modification redirects VAChT to Neurotransmitter Reuptake. The reuptake of clas-
LDCVs,117 indicating the potential to regulate the sical neurotransmitters across the plasma membrane
site of transmitter storage and hence its role in sig- serves to remove extracellular transmitter from the
naling. synaptic cleft and thereby terminate signaling.
VGAT and Related Transporters. Although the Plasma membrane transport proteins, which medi-
transport of all classical transmitters into SVs de- ate reuptake of distinct neurotransmitters, can be
pends on a proton electrochemical gradient, the classified into two large families.145 Both rely on the
transport of monoamines and ACh depends primar- Na+ gradient across the cell membrane to drive up-
ily on the chemical component ⌬pH, whereas the take, but plasma membrane monoamine and GABA
transport of glutamate and other amino acids de- transporters (DAT, NET, SERT, and GAT) also re-
pends to a greater extent on the electrical compo- quire Cl− for transport (Na+/Cl−-dependent trans-
nent ⌬␺ It is thus not surprising to find that the porters).157 The localization of this family of trans-
sequence of a cDNA encoding vesicular GABA trans- porters on the presynaptic neuron allows reuptake
port (VGAT) showed no sequence similarity to the to replenish stores by recycling transmitter released
VMATs or to VAChT. 147 Originally identified by exocytosis.110 In contrast, plasma membrane
through molecular genetic analysis in C. elegans, transporters for the excitatory amino acid transmit-
VGAT encodes a novel protein with 11 predicted ters glutamate and aspartate (excitatory amino acid
transmembrane domains. In addition to GABAergic transporters, or EAATs) require K+, instead of Cl−,
neurons, it resides in glycinergic neurons and ap- for transport (Na+/ K+-dependent transporters) and,
pears to recognize glycine as well as GABA.48 VGAT in most cases, do not occur on presynaptic neurons,
also defines a novel family of membrane proteins precluding a direct role in neurotransmitter recy-
that includes the classically described amino acid cling.203
transport Systems N and A (see later). Monoamine Transport. Interference with plasma
Vesicular Glutamate Transporter. The uptake of membrane DA transport with psychostimulants such
glutamate into SVs is mediated by a previously char- as cocaine and amphetamines has profound effects
acterized brain-specific sodium-dependent inorganic on behavior.4,204 Indeed, cocaine inhibits the reup-
phosphate cotransporter (BNPI).16 BNPI was ini- take of catecholamines and hence prolongs its ac-
tially thought to influence glutamate biosynthesis by tion on the postsynaptic neuron,12 whereas amphet-
regulating glutaminase, the enzyme that converts amines promote flux reversal by DAT, which
glutamine to glutamate. Indeed, the glutaminase iso- increases extracellular dopamine.74 Genetic disrup-
form occurring in neurons depends exquisitely on tion of the DAT gene in mice mimics the behavioral
intracellular inorganic phosphate levels. BNPI could phenotype of cocaine use, with dramatic locomotor
therefore regulate glutamate levels indirectly by me- hyperactivity.82 Interestingly, these animals show a
diating phosphate uptake into neurons, which in dramatic reduction in total brain dopamine levels
turn could activate glutaminase. Interestingly, how- despite the high extracellular concentrations.110
ever, BNPI resides largely on SVs, where it presum- This suggests that reuptake through DAT may nor-
ably cannot function in inorganic phosphate up- mally contibute more DA to the intracellular pool
take.15 Further, a selective defect in glutamate than even de novo synthesis, highlighting its impor-
release by the C. elegans BNPI homolog eat-4 has also tant role in signaling. These transporters also have
suggested a more direct role for BNPI in glutamate the potential to modulate synaptic efficacy depend-
release.123 Indeed, measurements show that BNPI ing on the level of transporter expression at the cell
(renamed vesicular glutamate transporter 1, or surface of neurons. In the case of SERT, DAT, and
VGLUT1) mediates glutamate uptake into vesicles NET, protein kinase C (PKC)-mediated phosphory-
with kinetics and bioenergetics very similar to endog- lation promotes internalization from the cell sur-
enous brain vesicular glutamate transport activity. In face, thereby reducing neurotransmitter reuptake
addition, VGLUT1 exhibits a conductance for chlo- into cells.7,27,57,178 For SERT, both PKC-mediated
ride that is blocked by glutamate, a feature also re- phosphorylation and sequestration were inhibited by
ported for glutamate transport into brain SVs.16 It serotonin, but not inhibitors such as cocaine and
remains possible nonetheless that VGLUT1 func- antidepressants, suggesting a homeostatic mecha-

586 Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001


nism for regulation of extracellular monoamine con- membrane of astrocytes.49 Like VGAT, it acts as a
centrations.177 proton exchanger but also mediates the Na + -
Excitatory Amino Acid Transport. The plasma dependent uptake of glutamine and apparently
membrane transport of the excitatory amino acids corresponds to classically described amino acid
glutamate and aspartate differs in important ways transport System N.116,232 Under physiological cir-
from the transport of monoamines and GABA. The cumstances, however, the System N transporter
EAATs also depend on Na+ but not Cl− to drive re- (SN1) more likely mediates the efflux of glutamine
uptake.145,203 Further, they catalyze the inward trans- from astrocytes and hence contributes to the gluta-
location of a proton in exchange for cytoplasmic K+ mine–glutamate cycle. Also, another protein closely
coupled to the uptake of glutamate.264 In addition to related to SN1, encoding System A, occurs on neu-
providing an extremely strong driving force for glu- rons and may mediate the uptake of glutamine by
tamate uptake, this ionic dependence presumably these cells that is also required for the glutamine–
reflects the function of a class of proteins entirely glutamate cycle.180,247
distinct from the monoamine and GABA transport-
ers. Interestingly, the EAATs also exhibit a gluta- Neurotransmitter Degradation. Acetylcholine. Deg-
mate-activated chloride conductance uncoupled radation can also influence extracellular neurotrans-
from transport activity that would serve to hyperpo- mitter levels and synaptic transmission. Acetylcholin-
larize the neuron.69,219,249 The postsynaptic location esterase (AChE), the enzyme that hydrolyzes ACh, is
of neuronal EAATs (EAAT3 and EAAT4 isoforms) the target of drugs used in the diagnosis and treat-
would enable them to act as glutamate receptors as ment of myasthenia gravis. In myasthenia, autoanti-
well as transport proteins, and the uncoupled chlo- bodies directed against acetylcholine receptors at
ride conductance appears to have a physiological the neuromuscular junction impair synaptic trans-
role in phototransduction. 68,124,218 In contrast, mission. Drugs that inhibit AChE prolong the half-
EAAT1 and 2 reside on astrocytes, where they ac- life of ACh in the neuromuscular junction and
count for the bulk of glutamate reuptake in the thereby partially rescue the synaptic defect in myas-
brain.191,234 Importantly, this localization is also con- thenia.67,150 AChE occurs in multiple forms, classi-
sistent with recent work suggesting a more direct fied as either globular (G) or asymmetric (A), each
role for astrocytes in glutamatergic synaptic transmis- of which can be membrane-bound or soluble.54 At
sion.248 In addition, removal of glutamate from the certain synapses, AChE binds to the plasma mem-
extracellular space by EAAT1 and 2 appears to help brane through a glycophospholipid anchor. At oth-
shape the postsynaptic response and protect against ers, such as the neuromuscular junction, the asym-
excitotoxicity. 20,21,61 Indeed, disruption of the metric (A12) form containing a collagen tail
EAAT2 gene in mice produces seizures and hippo- covalently attached to complexes containing 12
campal degeneration.234 In amyotrophic lateral scle- globular subunits, binds to the synaptic basal lamina
rosis (ALS), aberrant RNA processing of EAAT2 re- rather than the plasma membrane.193 This localiza-
duces transporter levels in affected brain regions, tion involves a very tight association with extracellu-
presumably resulting in excitotoxic injury and motor lar matrix proteins such as perlacan and dystoglycan
neuron degeneration.28,127 Of all the EAATs, how- and mirrors the clustering of ACh receptors at the
ever, only EAAT5 appears to reside on the presyn- neuromuscular junction. Localization of AChE to
aptic nerve terminal,9,176 but this isoform occurs this site couples degradation with neurotransmitter
only on photoreceptors. Thus, the EAATs do not release, and may also contribute to regulation. The
appear to have a direct role in the recycling of glu- recycling of ACh thus differs from other classical
tamate in the same way that DAT contributes to do- transmitters, which undergo reuptake at the nerve
pamine recycling. Rather, synaptically released glu- terminal. In contrast to direct reuptake of the
tamate is taken up by glia, converted to glutamine, transmitter, the Na+-dependent uptake of choline
and then delivered back to the neuron for conver- produced by AChE is mediated by CHT1, a novel
sion to glutamate.53,88,139,165,237 Inhibition of glial protein unrelated to other neurotransmitter trans-
glutamine synthetase and neuronal glutaminase re- porters.162
duces glutamate release, supporting a role for this Monoamines. In contrast to ACh, monoamine
cycle in synaptic transmission.192 However, the pro- degradation occurs at cytoplasmic as well as extracel-
teins responsible for glutamine export from glia and lular sites. Monoamine oxidase (MAO), an enzyme
uptake by neurons have not been identified until localized to the outer membrane of mitochondria,
recently. Interestingly, a novel protein related to the mediates the cytoplasmic metabolism of mono-
vesicular GABA transporter occurs on the plasma amines and also produces hydrogen peroxide.209 In-

Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001 587


hibition of the principal neuronal isoform MAO-A fused membrane and a second round of exocytosis
enhances monoamine signaling, particularly for se- can take as little as 20 seconds.25,194,195 Membrane
rotonin (5-HT). Genetic disruption of MAO-A in retrieval and SV biogenesis must therefore occur
mice leads to high 5-HT levels and causes aggressive very efficiently. Retrieval and translocation to the ac-
behavior and defects in cortical development.38 tive zone also appear to undergo regulation196 that
Monoamine levels do, however, eventually decline to contributes to synaptic plasticity and perhaps learn-
near wild-type in MAO-A knockout mice, presumably ing and memory.141 Long-term potentiation (LTP)
due to expression later in life of the glial isoform, in the mossy fiber terminals of region CA3 in the
MAO-B. Under normal circumstances, MAO-B pre- hippocampus involves presynaptic changes, and the
dominantly degrades catecholamines rather than genetic manipulation of transmitter release inhibits
5-HT. MAO-B also activates the parkinsonian toxin mossy fiber LTP.39
MPTP by converting it to MPP+, and either pharma- Our understanding of the SV cycle has evolved
cological inhibition or genetic interuption of rapidly over the past two decades. The purification
MAO-B completely prevents MPTP toxicity.85,122 of SVs has allowed identification of many protein
However, MAO-B is present in glia and therefore can constituents223 and these proteins form complexes
degrade only the dopamine released from neurons. that have suggested possible functions (Table 2). For
If cytoplasmic dopamine contributes to the patho- example, association of an SV protein with a plasma
genesis of Parkinson’s disease, inhibiting MAO-B membrane protein suggests that these proteins me-
would have little protective effect, consistent with the diate SV docking and fusion.35,216 The discovery that
results of recent large-scale clinical studies.210 Since botulinum and tetanus toxins inhibit neurotransmit-
it is expressed by dopamine neurons, MAO-A may ter release by specifically cleaving proteins in this
have a much larger role, but MAO-A may actually complex has supported a physiological role for these
protect against dopamine toxicity by preventing the interactions.189,199 In contrast to the clostridial tox-
spontaneous, nonenzymatic oxidation of dopamine ins that inhibit release, the black widow spider
that yields more highly reactive products.63 In addi- venom toxin, ␣-latrotoxin, promotes massive neuro-
tion, catecholamines are degraded by catechol-o- transmitter release by Ca2+-dependent and -indepen-
methyltransferase (COMT) which, like AChE, occurs dent mechanisms.97,164 Two presynaptic ␣-latrotoxin
in the synaptic cleft.155 Thus, catecholamine metabo- receptors, neurexin and latrophilin/CIRL, appear to
lism may occur outside as well as inside neurons. The mediate these effects and may also control neuro-
redundancy in metabolic pathways suggests that the transmitter release under physiological circum-
cell may require multiple mechanisms to protect stances.118,226 To understand the function of these
against monoamine and in particular dopamine tox- novel proteins, their genes have been disrupted in
icity. organisms ranging from C. elegans and D. melanogas-
ter to mice and the phenotypes characterized by elec-
THE SYNAPTIC VESICLE CYCLE trophysiological methods.260 In addition, similar ge-
Synaptic transmission depends on the efficient coor- netic and biochemical approaches have also
dination of multiple events, including SV fusion, re- reconstituted various other steps in the SV cycle.73
trieval, and docking prior to a second round of
fusion. SV fusion involves highly conserved mecha- Targeting and Docking. The Readily Releasable Pool.
nisms responsible for essentially all the fusion events Tightly regulated exocytosis requires the localization
that contribute to membrane trafficking in eukary- of SVs to the nerve terminal and, in particular, to the
otic organisms.19,190 However, SV exocytosis differs active zone. The active zone aligns precisely with the
from many other forms of membrane fusion in terms synaptic cleft and the postsynaptic density, a mem-
of its regulation. SV fusion depends on an elevation brane specialization enriched in neurotransmitter
in cytoplasmic Ca2+ mediated by voltage-sensitive receptors.99,100 At the active zone, a few SVs are in
Ca2+ channels.156 Indeed, the regulation of exocyto- direct contact with the plasma membrane, a state
sis by neural activity accounts for the morphological known as docked.170 Docked vesicles are irreversibly
accumulation of SVs and has enabled their purifica- bound to the cell surface but not necessarily compe-
tion.107 SV fusion also exhibits a high degree of spa- tent for fusion.98,187 Invasion of the nerve terminal
tial specificity, with exocytosis essentially restricted to by action potentials activates voltage-sensitive Ca2+
the active zone. Further, high rates of exocytosis re- channels and the resulting increase in cytoplasmic
quire the retrieval of SVs locally, in the nerve termi- Ca2+ causes fusion of a small fraction of the docked
nal. At both the neuromuscular junction and central vesicles. In contrast, application of hypertonic su-
synapses, the time from exocytosis to retrieval of the crose results in fusion all docked vesicles, indepen-

588 Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001


Table 2. Proteins involved in the synaptic vesicle cycle.

Protein Function

Targeting and docking Synapsins Phosphoproteins linking SVs to the actin cytoskeleton
Piccolo and bassoon Presynaptic cytomatrix proteins; role in active zone assembly
Sec6/8 complex Mammalian exocyst homolog; defines the site of the active zone
Fusion Synaptobrevin v-SNARE
SNAP-25 t-SNARE
Syntaxin t-SNARE
NSF ATPase; dissociates SNARE complex
␣-SNAP NSF binding protein
Modulation and Ca2+ dependence Rab3a Small GTPase; regulates SV fusion via multiple effectors
Uso1/ p115 Rab effector; modifies lipids; “tethers” SVs
nSec-1 Inhibits SNARE assembly by binding syntaxin; Rab effector
Munc13 C1 and C2 domain protein; required for neurotransmitter release
N, P/Q-type Ca2+ channels Bind SNAP-25 and Syntaxin; occur near the site of SV fusion
Synaptotagmin C2 domain SV protein; Ca2+ sensor
SV2 Transporter; inhibits SV fusion
CAPS Facilitates LDCV exocytosis
Endocytosis and recycling Clathrin Coats nascent endocytic vesicles and induces their invagination
AP-2 Adaptor protein recruits clathrin to the plasma membrane
Dynamin GTPase; “pinches” the neck of endocytic vesicles
Amphiphysin Binds AP-2 and clathrin; recruits dynamin to endocytic vesicles
Synaptojanin Binds amphiphysin and endophilin; inositol 5-phosphatase
Endophilin SH3 domain protein; lysophosphatidic acid arachidonate transferase
AP-3 Adaptor protein for SV recycling from endosomal intermediates

dent of Ca2+, and this population of SVs is known as neurons, the homologous sec6/8 complex localizes
the readily releasable pool.188 to the synaptic plasma membrane.104 However, the
The Reserve Pool. In addition to the docked exocyst localizes at this site prior to the development
vesicles, a large number of SVs cluster over the syn- of a presynaptic element, and regresses with the
aptic cleft. Although not physically docked, these maturation of the nerve terminal.95 The exocyst may
vesicles aggregate over the active zone as a result of therefore define the subsequent site of the active
interactions with the actin cytoskeleton through pro- zone, but does not appear to be directly involved in
teins such as synapsins and the enormous cytomatrix the SV cycle.
proteins piccolo and bassoon.101,239 Genetic dis-
ruption of synapsins in mice causes seizures and a Fusion. SNAREs. Regulated fusion with the
decrease in short-term synaptic plasticity, most plasma membrane is the central event in the SV
likely due to a reduction in the reserve pool of cycle. A little more than 10 years ago, a number of
SVs.126,186 Interestingly, stimulation at physiological synaptic proteins were identified that together
frequencies does not release SVs from this reserve formed a stable complex, known as the SNARE (for
pool.173 However, it remains unclear whether re- SNAP receptor). The complex consists of a vesicle-
cycled SVs have a higher likelihood of release associated SNARE (v-SNARE), synaptobrevin (or
than vesicles in the reserve pool.195 The clustering of VAMP),240 and two target or plasma membrane
SVs over the active zone may also undergo regula- SNAREs (t-SNAREs), SNAP-25166 and syntaxin.17 In-
tion that contributes to synaptic plasticity. Indeed, deed, v- and t-SNAREs form extremely stable protein
synapsin binding to SVs is negatively regulated by complexes resistant to detergents that require boil-
PKA and Ca2+/calmodulin-dependent protein ki- ing to dissociate.94 It was also shown that the ATPase,
nases (CaMKs).41,103,221 N-ethylmaleimide-sensitive factor (NSF), and its as-
The Exocyst. Additional mechanisms also appear sociated protein, soluble NSF attachment protein (␣-
to restrict SV docking and fusion to the nerve termi- SNAP), both previously implicated in membrane fu-
nal active zone. In yeast, polarized secretion involves sion, specifically bind the SNARE complex. 217
a large protein complex, known as the exocyst, Further, NSF-dependent ATP hydrolysis dissociated
which appears to function as a landmark for recruit- the complex, possibly serving as a trigger initiating
ing secretory vesicles to the site of exocytosis.236 In membrane fusion.215

Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001 589


SNAREs and Fusion. The SNARE hypothesis ex-
tends to the fusion of many other membranes in the
cell, such as those involved in endoplasmic reticu-
lum (ER) to Golgi transport and transport to the
lysosome. Distinct v- and t-SNAREs occur in these
different compartments, suggesting that they confer
the specificity required for appropriate membrane
fusion. Also, SNARE proteins mediate membrane
trafficking and secretion in yeast as well as mamma-
lian cells,18 indicating the generality of the SNARE
hypothesis. However, the precise role of the SNARE
complex remains unclear and it has been proposed
to function in SV docking, fusion, or both.215 De-
spite the identification of different v- and t-SNAREs
on different compartments, they appear to form
complexes promiscuously, raising questions about a
role in the specificity of docking.263 Indeed, disrup-
FIGURE 2. The molecular targets of tetanus and botulinum toxins
tion of the SNARE complex with clostridial toxins
are proteins in the SNARE complexes for SV exocytosis. Tetanus
does not prevent docking106 and morphological toxin cleaves synaptobrevin, whereas, of the seven classes of
docking is not prevented in Drosophila mutants lack- botulinum toxins, four also cleave synaptobrevin, two cleave
ing syntaxin.30 Further, recent observations have led SNAP-25, and the remaining one cleaves syntaxin.
to a revision in the role of the ATPase NSF as a
fusion protein.223 Indeed, NSF-mediated disruption
The conserved residues contributed by each of the
of the SNARE complex after docking neither trig-
helices create an ionic layer within the bundle, pre-
gers nor inhibits fusion, but rather induces a state of
sumably contributing to the high affinity of the com-
membrane hemifusion that does not involve the re-
plex. As type II membrane proteins, the N-termini of
lease of vesicle contents.244 Other work using the
all the SNARE proteins protrude into the cytoplasm
Drosophila temperature-sensitive NSF mutant comatose
and the parallel arrangement suggests that the heli-
suggests that NSF disassembles the SNARE complex
ces “zipper up,” driving the SV and plasma mem-
after fusion in order to retrieve v- but not t-SNAREs
to the synaptic vesicle.129
Clostridial Toxins and SNARE Structure. The ac-
tions of tetanus and botulinum toxins further dem-
onstrate the importance of SNARE complexes for SV
exocytosis.199 These clostridial toxins are composed
of disulfide-linked heavy and light chains. The heavy
chains mediate translocation of the toxins into the
cytoplasm, where the reducing environment sepa-
rates the two chains. The free light chains then func-
tion as endoproteases that inhibit exocytosis by se-
lectively cleaving a SNARE protein at a single site
(Fig. 2). Tetanus toxin cleaves synaptobrevin,
whereas, of the seven classes of botulinum toxins,
four also cleave synaptobrevin, two cleave SNAP-25,
and the remaining one cleaves syntaxin. In addition,
FIGURE 3. A model of the synaptic fusion complex as it joins the
structural studies have recently begun to indicate
membranes of the synaptic vesicle and plasma membrane,
how these cleavage events destabilize the SNARE based on the crystal structure of the SNARE complex. Although
complex and thereby inhibit neurotransmitter re- originally proposed to interact in an anti-parallel manner, which
lease.105,229,261 The crystal structure of the SNARE would suggest a role in docking, v- and t-SNAREs actually ap-
complex also supports a specific role in fusion. Al- pear to form a parallel helix bundle. The N-termini of all the
SNARE proteins protrude into the cytoplasm and the parallel ar-
though originally proposed to interact in an antipa-
rangement suggests that the helices “zipper up,” driving the SV
rallel manner, which would suggest a role in dock- and plasma membranes together. Indeed, the strain placed on
ing, this work shows that v- and t-SNAREs actually the parallel-bound SNAREs may drive membranes already in
form a four-helix bundle in parallel (Fig. 3).89,128,229 close proximity with one another to fuse.

590 Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001


branes together. Indeed, the strain placed on the rons, a number of rab effectors have been identified,
parallel bound SNAREs may drive membranes al- including rabphilin and RIM, but their precise role
ready in close proximity with one another to fuse. in transmitter release remains unknown.80,84
Further supporting a direct role for the SNAREs in Ca2+ Channels and SVs. Synaptic vesicle exocyto-
fusion, the addition of a SNARE peptide to the com- sis differs from most other forms of membrane fu-
plex cleaved by a clostridial toxin directly triggers sion in its speed and responsiveness to Ca2+. In the
fusion,51 and the v- and t-SNAREs reconstituted into Lambert–Eaton myasthenic syndrome, an autoim-
separate artificial membranes suffice to drive fusion mune disorder of neuromuscular transmission in
in vitro.253 However, fusion in this system occurs at a which antibodies are directed against P/Q-type volt-
rate much slower than observed in native mem- age-dependent Ca2+ channels, quantal release of
branes, suggesting modulation by other factors. ACh is reduced.198,262 Interference with Ca2+ chan-
nel function appears to impair the tight coupling of
Modulation and Ca2+ Dependence. Rab Proteins. neural activity with Ca2+ entry into the presynaptic
Rab proteins regulate membrane trafficking at a neuron. Indeed, neurotransmitter release occurs
number of different sites within the cell. They be- within 1 ms of Ca2+ entry into the nerve terminal,
long to the ras superfamily of small guanosine tri- suggesting that a significant fraction of the docked
phosphatases (GTPases) and, like the SNAREs, mul- SVs exist in a metastable, possibly hemifused, state.34
tiple, distinct isoforms occur on different membrane Proximity to the Ca2+ entry sites also contributes to
compartments.84 In neurons, the rab3a isoform pre- the speed of release. SV fusion actually has a rela-
dominates on SVs.80 Similar to other rabs, rab3a tively low sensitivity to Ca2+ in the 50–100-µmol/L
cycles between a soluble and membrane-bound state. range, but vesicle docking occurs in close proximity
In the GTP-bound form, rab3a associates with SVs. to the Ca2+ channels. SV exocytosis thus does not
After hydrolysis of the GTP to GDP, rab3a dissociates depend on diffusely elevated cytoplasmic Ca2+ but
from the vesicles, a process dependent on the pro- responds primarily to local increases in Ca2+ near the
tein, guanine nucleotide dissociation inhibitor active zone.
(GDI).242 It is likely that GTP hydrolysis serves as a Ca2+ Channels and SNAREs. Both syntaxin and
timer that monitors the completion of one reaction SNAP-25 bind to the N- and P/Q-type voltage-
in the SV cycle before initiation of the next, but the dependent Ca2+ channels known to be responsible
role remains unclear. Rab3a knockout mice show an for transmitter release, indicating a link between
initial increase in neurotransmitter release, suggest- Ca2+ entry and SNARE complex formation.40 Fur-
ing that rab3a initially acts to inhibit exocytosis.79 ther, syntaxin and SNAP-25 appear to inactivate Ca2+
Intriguingly, the knockouts also appear defective in a channels when expressed individually, but not when
presynaptic form of LTP39 and exhibit a profound they are assembled into a SNARE complex.26,266 This
depression in neurotransmitter release upon repeti- suggests that free t-SNAREs inhibit Ca2+ entry, but
tive stimulation, suggesting a depletion of the re- vesicle docking relieves this inhibition, restricting SV
serve pool of SVs.77 Indeed, alterations in rab func- fusion to the sites of Ca2+ entry. Recent work also
tion redistribute SVs away from the nerve terminal in suggests that Ca2+ entry through P/Q-type channels
C. elegans.161 influences syntaxin levels by regulating transcription
Rab Effectors. Consistent with this role in modu- via mechanisms involving internal Ca2+ stores and
lation of SV targeting, a yeast rab homolog binds to phosphorylation.227 An unusual SV protein known
the sec6/8 exocyst complex, possibly contributing to as the cysteine string protein (CSP) also associates
reversible tethering of vesicles to the plasma mem- with Ca2+ channels and may provide an additional
brane.87 Interestingly, yeast rabs also interact with link between SV docking and channel activa-
the soluble factor, Uso1/p115, to promote reversible tion.146,179,243 Interestingly, CSP also contains a DnaJ
vesicle attachment in preparation for SNARE com- domain that suggests a role in protein folding, but
plex formation.36 These fibrous proteins appear to the function of the protein remains unclear.
correspond to the “string” structures observed by Synaptotagmin. How do SVs sense a local in-
electron microscopy, which may also tether vesicles crease in Ca2+ and use this to drive exocytosis? The
to the plasma membrane.163 Uso1/p115 has also rapid kinetics of fusion suggest that a conforma-
been characterized as a CTP–phosphocholine- tional or electrostatic change in a Ca2+ sensor, rather
cytidyltransferase-enhancing factor, suggesting that than a large-scale rearrangement of proteins, trig-
at least one rab effector modifies lipids.72 In yeast, gers the final stages of fusion. Both genetic and bio-
rabs may also contribute to the dissociation of inhibi- chemical evidence further implicate the SV protein
tory nsec-1 from the t-SNARE, syntaxin.138 In neu- synaptotagmin I as such a Ca2+ sensor.80 Synap-

Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001 591


totagmin I belongs to a family of integral membrane also interacts with SV2,200 a synaptic vesicle protein
proteins expressed predominantly in neurons, with homology to transporters but as yet unknown
where they localize to SVs and other neurosecretory function.13,71 Interestingly, SV2 knockout mice show
vesicles such as LDCVs that also undergo regulated a Ca2+-sensitive facilitation of neurotransmitter re-
exocytosis.125 The data suggest that synaptotagmin lease, suggesting that SV2 may itself negatively regu-
does not constitute part of the basic fusion machin- late fusion by binding Ca2+ or pumping excess pre-
ery, an idea further supported by the absence of synaptic Ca2+ into SVs.56,109 In one model, Ca2+ may
synaptotagmin isoforms in yeast. Importantly, synap- simply shift the affinity of interactions made by syn-
totagmin I knockout mice lack the fast component aptotagmin. At low Ca2+ concentrations, synaptotag-
of Ca2+-triggered neurotransmitter release, but show min binds SV2, which might inhibit fusion. With
no abnormality in the slower asynchronous phase of Ca2+ entry, synaptotagmin dimerizes via its C2B do-
Ca2+-independent release.78 Synaptotagmin mutants main and interaction of the C2A domain with syn-
also show increased spontaneous release of SVs, sug- taxin may organize and promote the assembly of
gesting that synaptotagmin normally acts to inhibit SNARE proteins. The C2A domain also appears to
transmitter release, but can be overriden by in- penetrate the bilayer in a Ca2+-dependent fashion,
creased Ca2+.29,62 Consistent with this, overexpres- which may promote fusion directly.44 Interestingly,
sion of synaptotagmin in neuromuscular cultures de- synaptotagmin IV, an isoform with a substitution in a
creases spontaneous release, but increases Ca2+- highly conserved residue of the C2A domain that
dependent paired-pulse facilitation.152 abolishes its ability to bind membranes in response
C2 Domains. The cytoplasmic region of synap- to Ca2+, is upregulated during chronic depolariza-
totagmins contains two C2 domains (originally iden- tion or seizure activity. By assembling with synapto-
tified in PKC) that bind to phospholipids and Ca2+ tagmin I, synaptotagmin IV may prevent the com-
with varying affinity.183,224 The affinity of synaptotag- plex from penetrating the lipid bilayer, reduce
min I for Ca2+ clearly corresponds to the known, evoked neurotransmitter release, and perhaps con-
relatively low sensitivity of SV fusion to Ca2+. The tribute to a novel mechanism of synaptic plasticity.
crystal structure of one of the C2 domains in synap- However, many proteins in the nerve terminal in
totagmin I (C2A) suggests that it binds three Ca2+ addition to synaptotagmin could serve as the Ca2+
ions with the expected affinities.228,241 Further, Ca2+ sensor for exocytosis. The calcium-binding protein,
binding seems to induce electrostatic changes that calmodulin, has been shown to promote Ca 2+ -
may act as a switch to drive exocytosis.206,207 Synap- regulated exocytosis.222 More recently, two studies in
totagmin has thus been proposed to act as the Ca2+ yeast have suggested that both calmodulin and pro-
sensor for SV exocytosis.59 The existence of multiple tein phosphatase 1 are required for a late step in
synaptotagmin isoforms with widely varying sensitiv- constitutive membrane trafficking.171,172 In addi-
ity to Ca2+ may also contribute to the differences in tion, a number of proteins in the nerve terminal
sensitivity to Ca2+ observed between different popu- contain C2 domains, and thus have the same poten-
lations of neurosecretory vesicles. Although they tial as synaptotagmin to serve as Ca2+ sensors.
generally require more stimulation to induce fusion Other C2-Domain–Containing Proteins. Several
than SVs, LDCVs have an extremely high sensitivity proteins involved in synaptic transmission contain
to Ca2+. They presumably require more stimulation, C2 domains that may also bind Ca2+.183 For example,
because, unlike SVs, LDCVs do not reside in close the evolutionarily conserved neuron-specific verte-
proximity to the sites of Ca2+ entry. Rather, LDCVs brate ortholog of the C. elegans Unc-13 (Munc-13),
sense diffuse elevations in cytoplasmic Ca2+, and sev- binds syntaxin and contains both C1 and C2 do-
eral synaptotagmin isoforms show the high affinity mains.24,32,144,197 Interestingly, the C1 rather than
for Ca2+ that corresponds to physiological observa- the C2 domain of Munc-13 appears critical for a late
tions of LDCV exocytosis. However, it remains un- step in SV fusion and the C1 domain binds the sec-
clear how synaptotagmin contributes to the exocyto- ond messenger, diacylglycerol (DAG), as well as
sis of SVs. phorbol esters.115 Both DAG and phorbol esters in-
Function of Synaptotagmin. The two C2 domains duce the translocation of Munc-13 to the plasma
may act in very different ways to promote regulated membrane, and Munc-13 mediates the sensitivity of
exocytosis. The C2A domain interacts with syntaxin neurotransmitter release to phorbol esters.23 Impor-
as well as phospholipids, and this may promote SV tantly, recent reports have shown that C. elegans, Dro-
docking as well as fusion. The C2B domain is also sophila, and mouse Unc-13 mutants all have dramatic
predicted to bind Ca2+, but this reaction stimulates reductions in both action potential-evoked and
synaptotagmin dimerization.43,225 The C2B domain spontaneous, Ca 2+ -independent release. 8,10,182

592 Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001


Munc-13 may also link G-protein–coupled neuro- tion may nonetheless contribute to SV exocytosis,
transmitter receptors with neurotransmitter release. possibly through the involvement of a protein dis-
ACh and serotonin receptors couple to G proteins tinct from Unc-31.256 In addition, it is appears that
that activate the production of DAG. C. elegans mu- SV recycling prominently involves modulation by
tants in the corresponding receptors show differ- both Ca2+ and lipids.
ences from wild-type in neurotransmitter release via
the DAG/Unc-13 pathway.121,151 Together with a role Endocytosis and Recycling. Clathrin and AP-2.
in coupling neurotransmitter release to receptor sig- How does the nerve terminal maintain a relatively
naling, the requirement for both evoked and spon- constant surface area despite high rates of exocyto-
taneous release suggests that Munc-13 may have a sis? After exocytosis, empty SVs are rapidly endocy-
central role in SV fusion. The synaptic proteins tosed, a process dependent on the formation of a
DOC2, rabphilin, and RIM also contain C2 domains, clathrin coat on nascent endocytotic vesicles. Cla-
but their role in transmitter release remains less thrin presumably initiates invagination after recruit-
clear. Further, Ca2+-dependent regulation may occur ment to the plasma membrane by the clathrin adap-
via proteins without C2 domains such as the Ca2+- tor protein AP-2.55,142 According to one model, Ca2+
dependent activator of secretion (CAPS), a phospha- signals endocytosis as well as exocytosis, and Ca2+
tidylinositol 4,5-bisphosphate-binding protein that is does appear to be required for internalization.96 Al-
selectively required for catecholamine secretion though it remains unclear how Ca2+ promotes SV
from LDCVs but not glutamate release from SVs (dis- endocytosis, it is of interest that synaptotagmin, the
cussed in what follows).64 putative Ca2+ sensor for exocytosis, has also been
Large, Dense-Cored Vesicles. The release of neu- implicated in endocytosis. Synaptotagmin mutants
ral peptides from LDCVs shares certain similarities indeed show reduced numbers of SVs, implicating
with release of classical transmitters from SVs, but the protein in endo- as well as exocytosis, but the SV
also exhibits a number of important differences.143 depletion could also result from increased, unregu-
Like SVs, LDCVs contain the v-SNARE, synaptobre- lated fusion.111 Importantly, synaptotagmin binds
vin, and use the same t-SNAREs, syntaxin and SNAP- AP-2, and thereby provides a mechanism to link Ca2+
25.258 LDCVs also contain synaptotagmin I, although signaling with clathrin-mediated endocytosis.45,90,265
different isoforms may be responsible for the high The simple appearance of SV proteins, such as syn-
sensitivity of LDCV fusion to Ca2+.125 However, aptotagmin or synaptobrevin, on the plasma mem-
LDCV exocytosis appears to involve several mecha- brane may thus activate the endocytic machinery.
nisms distinct from those involved in SV release. In Dynamin. The late phase of endocytosis re-
particular, an elegant biochemical analysis of LDCV quires dynamin, which binds to the nascent vesicles
fusion has indicated two distinct phases.92 The first at the margin of the clathrin coat.201 At nonpermis-
phase involves a priming step that requires ATP and sive temperature, the Drosophila temperature-
probably follows SV docking at the plasma mem- sensitive dynamin mutant shibire rapidly becomes
brane. The factors responsible for this step include paralyzed and electron microscopy has shown a
the phosphatidylinositol (PI) transfer protein marked depletion of SVs.50,174,245 Dynamin func-
(PITP), PI-4-kinase, and PI(4-P)-5-kinase.91,93,255 tions as a GTPase and blocking dynamin-mediated
Priming thus appears to involve sequential lipid GTP hydrolysis in a preparation of nerve terminals
modification that forms PI(4,5)-bisphosphate. Inter- induces the formation of a spiral collar around the
estingly, the second phase of LDCV exocytosis in- neck of vesicles invaginating from the plasma mem-
volves a triggering step that requires Ca2+. Purifica- brane.102,231 This suggests a role for dynamin as the
tion of the factor responsible for Ca2+-dependent “pinchase” involved in SV retrieval.252 Although re-
triggering has yielded the Ca2+-dependent activator cent work has questioned a direct mechanical role
of secretion protein (CAPS). CAPS is the vertebrate for dynamin as the pinchase, it nonetheless appears
ortholog of the C. elegans Unc-31 protein, which has to have a role in orchestrating endocytosis and the
been implicated in the release of neural peptides, recycling of SVs.205 In particular, dynamin may func-
serotonin, and perhaps ACh, supporting a role for tion by associating with a series of accessory endo-
CAPS in LDCV exocytosis.6,11,250 Further, CAPS is a cytic proteins such as amphiphysin, synaptojanin,
soluble protein that associates with LDCVs and the and endophilin.31,58,60,149 Disruption of the interac-
plasma membrane by binding to PI(4,5)-bisphos- tion with amphiphysin interupts the SV cycle at a
phate,137 the product of the priming reaction. CAPS position shortly after membrane invagination.211
does not bind to SVs or appear important for their Synaptojanin exhibits phosphotydilinositol 5-phos-
exocytosis.22,235 However, sequential lipid modifica- phatase activity that may reverse the lipid modifica-

Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001 593


tions involved in priming.148 Endophilin appears to nerve terminal, certain proteins occur only on sub-
function as a lysophosphatidic acid arachidonate populations further away from the synaptic cleft that
transfer protein, which may contribute to the nega- may correspond to the reserve pool. The functional
tive membrane curvature required at the margin of significance of these observations remains unclear,
the nascent endocytotic vesicle.202 but they indicate the potential for novel modes of
“Kiss-and-Run” versus Full Fusion. The local recy- signaling and regulation.
cling of SVs at the nerve terminal is another special-
ization of neurons, which obviates the need for a CONCLUSIONS
continuous supply of new SVs to be transported from Neurotransmitter release constitutes a central yet
the neuronal soma, thereby also enhancing effi- poorly understood aspect of synaptic transmission.
ciency. Indeed, SVs may be recycled in a number of Two distinct cycles involve the neurotransmitter it-
different ways. Instead of full exocytosis, SVs may self and the membrane components, including lip-
form a transient fusion pore, allowing transmitters to ids as well as proteins. Within each of these cycles,
diffuse out, and then recycle instantly by closing the additional cycles monitor passage through the re-
pore (“kiss-and-run” exocytosis).3,52 After transmit- quired biochemical reactions. Many of the proteins
ter release, closure of the pore would regenerate the involved in transmitter release have now been iden-
original SV, albeit without or with less transmitter. tified but it is still not understood how many of them
This “kiss-and-run” model of transmitter release has integrate into these cycles. Together with biochemi-
not been demonstrated directly for SVs, but the cal analysis, future work will address the physiologi-
study of LDCV exocytosis does support its existence. cal consequences of these cycles and contribute to
In particular, measurements of cell capacitance, our understanding of synaptic plasticity and neuro-
which provide a direct measure of plasma mem- psychiatric disease.
brane surface area, coupled with electrochemical
measurement of monoamine release from LDCVs by
REFERENCES
amperometry, indicate that a slow leak of transmitter
often precedes the bulk release from an LDCV.1,2 1. Albillos A, Dernick G, Horstmann H, Almers W, Alvarez de
This slow leak presumably begins with the “flicker- Toledo G, Lindau M. The exocytotic event in chromaffin
cells revealed by patch amperometry. Nature 1997;389:
ing” of a fusion pore and the leakage of soluble 509–512.
transmitter. However, the bulk of monoamine in an 2. Ales E, Tabares L, Poyato JM, Valero V, Lindau M, Alvarez de
LDCV cannot be released until the pore dilates Toledo G. High calcium concentrations shift the mode of
exocytosis to the kiss-and-run mechanism. Nat Cell Biol
enough to allow solubilization of the dense core, 1999;1:40–44.
followed by its diffusion into the extracellular me- 3. Alvarez de Toledo G, Fernandez-Chacon R, Fernandez JM.
dium.257 Also, solubilization and release of the core Release of secretory products during transient vesicle fusion.
Nature 1993;363:554–558.
appear to depend on presynaptic autoreceptor activ- 4. Amara SG, Sonders MS. Neurotransmitter transporters as
ity, suggesting that the fusion pore constitutes a ma- molecular targets for addictive drugs. Drug Alcohol Depend
jor locus for the regulation of transmitter release.5 1998;51:87–96.
5. Angleson JK, Cochilla AJ, Kilic G, Nussinovitch I, Betz WJ.
Since SVs are considerably smaller than LDCVs, it is Regulation of dense core release from neuroendocrine cells
more difficult to detect the exocytosis of an indi- revealed by imaging single exocytic events. Nat Neurosci
vidual SV, but SV fusion may involve flicker as well. 1999;2:440–446.
6. Ann K, Kowalchyk JA, Loyet KM, Martin TF. Novel Ca2+-
“Kiss-and-run” also greatly simplifies the recycling of binding protein (CAPS) related to UNC-31 required for
SVs, which are regenerated directly from the plasma Ca 2 + -activated exocytosis. J Biol Chem 1997;272:
membrane. 19637–19640.
7. Apparsundaram S, Schroeter S, Giovanetti E, Blakely RD.
Recycling and Biogenesis. In neurons, the regen- Acute regulation of norepinephrine transport: II. PKC-
eration of SVs directly from the plasma membrane modulated surface expression of human norepinephrine
also appears to predominate after full fusion.154 transporter proteins. J Pharmacol Exp Ther 1998;287:
744–751.
However, some SVs undergo a more complex indi-
8. Aravamudan B, Fergestad T, Davis WS, Rodesch CK, Broadie
rect recycling pathway that involves endosomal inter- K. Drosophila UNC-13 is essential for synaptic transmission.
mediates. Biochemical analysis has implicated the Nat Neurosci 1999;2:965–971.
adaptor protein AP-3 specifically in sorting certain 9. Arriza JL, Eliasof S, Kavanaugh MP, Amara SG. Excitatory
amino acid transporter 5, a retinal glutamate transporter
proteins to SVs through this indirect pathway.70,208 coupled to a chloride conductance. Proc Natl Acad Sci USA
This suggests either that multiple trafficking path- 1997;94:4155–4160.
ways converge on a single population of SVs, or that 10. Augustin I, Rosenmund C, Sudhof TC, Brose N. Munc13-1 is
essential for fusion competence of glutamatergic synaptic
multiple, distinct, populations of SVs exist. Interest- vesicles. Nature 1999;400:457–461.
ingly, although many proteins occur on all SVs in the 11. Avery L, Bargmann CI, Horvitz HR. The Caenorhabditis el-

594 Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001


egans unc-31 gene affects multiple nervous system-controlled family of C2-domain proteins. J Biol Chem 1995;270:25273–
functions. Genetics 1993;134:455–464. 25280.
12. Axelrod J, Whitby L, Hertting G. Effect of psychotropic 33. Brown P, Marsden CD. The stiff man and stiff man plus
drugs on the uptake of 3H-norepinephrine by tissues. Sci- syndromes. J Neurol 1999;246:648–652.
ence 1961;133:383–384. 34. Bruns D, Jahn R. Real-time measurement of transmitter re-
13. Bajjalieh SM, Peterson K, Shinghal R, Scheller RH. SV2, a lease from single synaptic vesicles. Nature 1995;377:62–65.
brain synaptic vesicle protein homologous to bacterial trans- 35. Calakos N, Scheller RH. Synaptic vesicle biogenesis, docking
porters. Science 1992;257:1271–1273. and fusion: a molecular description. Physiol Rev 1996;76:1–
14. Bauerfeind R, Regnier-Vigouroux A, Flatmark T, Huttner 29
WB. Selective storage of acetylcholine, but not catechol- 36. Cao X, Ballew N, Barlowe C. Initial docking of ER-derived
amines, in neuroendocrine synaptic-like microvesicles of vesicles requires Usolp and Yptlp but is independent of
early endosomal origin. Neuron 1993;11:105–121. SNARE proteins. EMBO J 1998;17:2156–2165.
15. Bellocchio EE, Hu H, Pohorille A, Chan J, Pickel VM, Ed- 37. Carroll PT. Membrane-bound choline-O-acetyltransferase in
wards RH. The localization of the brain-specific inorganic rat hippocampal tissue is associated with synaptic vesicles.
phosphate transporter suggests a specific presynaptic role in Brain Res 1994;633:112–118.
glutamatergic transmission. J Neurosci 1998;18:8648–8659. 38. Cases O, Seif I, Grimsby J, Gaspar P, Chen K, Pournin S,
16. Bellocchio EE, Reimer RJ, Fremeau RT Jr, Edwards RH. Up- Muller U, Aguet M, Babinet C, Shih JC, De Maeyer E. Ag-
take of glutamate into synaptic vesicles by an inorganic phos- gressive behavior and altered amounts of brain serotonin
phate transporter. Science 2000;289:957–960. and norepinephrine in mice lacking MAOA. Science
17. Bennett MK, Calakos N, Scheller RH. Syntaxin: a synaptic 1995;268:1763–1766.
protein implicated in docking of synaptic vesicles at presyn- 39. Castillo PE, Janz R, Sudhof TC, Tzounopoulos T, Malenka
aptic active zones. Science 1992;257:255–259. RC, Nicoll RA. Rab3A is essential for mossy fibre long-term
18. Bennett MK, Scheller RH. The molecular machinery for se- potentiation in the hippocampus. Nature 1997:388:590–593.
cretion is conserved from yeast to neurons. Proc Natl Acad 40. Catterall WA. Interactions of presynaptic Ca2+ channels and
Sci USA 1993;90:2559–2563. snare proteins in neurotransmitter release. Ann N Y Acad Sci
19. Bennett MK, Scheller RH. A molecular description of syn- 1999;868:144–159.
aptic vesicle membrane trafficking. Annu Rev Biochem 41. Ceccaldi PE, Grohovaz F, Benfenati F, Chieregatti E, Green-
1994;63:63–100. gard P, Valtorta F. Dephosphorylated synapsin I anchors syn-
aptic vesicles to actin cytoskeleton: an analysis by videomi-
20. Bergles DE, Diamond JS, Jahr CE. Clearance of glutamate
croscopy. J Cell Biol 1995;128:905–912.
inside the synapse and beyond. Curr Opin Neurobiol
42. Chanat E, Huttner WB. Milieu-induced, selective aggrega-
1999;9:293–298.
tion of regulated secretory proteins in the trans-Golgi net-
21. Bergles DE, Jahr CE. Synaptic activation of glutamate trans-
work. J Cell Biol 1991;115:1505–1519.
porters in hippocampal astrocytes. Neuron 1997;19:
43. Chapman ER, An S, Edwardson JM, Jahn R. A novel function
1297–1308.
for the second C2 domain of synaptotagmin. Ca2+-triggered
22. Berwin B, Floor E, Martin TF. CAPS (mammalian UNC-31) dimerization. J Biol Chem 1996;271:5844–5849.
protein localizes to membranes involved in dense-core 44. Chapman ER, Davis AF. Direct interaction of a Ca2+-binding
vesicle exocytosis. Neuron 1998;21:137–145. loop of synaptotagmin with lipid bilayers. J Biol Chem
23. Betz A, Ashery U, Rickmann M, Augustin I, Neher E, Sudhof 1998;273:13995–14001.
TC, Rettig J, Brose N. Munc13-1 is a presynaptic phorbol 45. Chapman ER, Desai RC, Davis AF, Tornehl CK. Delineation
ester receptor that enhances neurotransmitter release. Neu- of the oligomerization, AP-2 binding, and synprint binding
ron 1998;21:123–136. region of the C2B domain of synaptotagmin. J Biol Chem
24. Betz A, Okamoto M, Benseler F, Brose N. Direct interaction 1998;273:32966–32972.
of the rat unc-13 homologue Munc13-1 with the N terminus 46. Chapman ER, Hanson PI, An S, Jahn R. Ca2+ regulates the
of syntaxin. J Biol Chem 1997;272:2520–2526. interaction between synaptotagmin and syntaxin 1. J Biol
25. Betz WJ, Bewick GS. Optical analysis of synaptic vesicle re- Chem 1995;270:23667–23671.
cycling at the frog neuromuscular junction. Science 47. Chapman ER, Jahn R. Calcium-dependent interaction of the
1992;255:200–203. cytoplasmic region of synaptotagmin with membranes. Au-
26. Bezprozvanny I, Scheller RH, Tsien RW. Functional impact tonomous function of a single C2-homologous domain. J
of syntaxin on gating of N-type and Q-type calcium channels. Biol Chem 1994;269:5735–5741.
Nature 1995;378:623–626. 48. Chaudhry FA, Reimer RJ, Bellocchio EE, Danbolt NC, Osen
27. Blakely RD, Ramamoorthy S, Schroeter S, Quian Y, Appar- KK, Edwards RH, Storm-Mathisen J. The vesicular GABA
sundaram S, Galli A, DeFelice LJ. Regulated phosphoryla- transporter, VGAT, localizes to synaptic vesicles in sets of
tion and trafficking of antidepressant-sensitive serotonin glycinergic as well as GABAergic neurons. J Neurosci
transporter proteins. Biological Psychiatry 1998;3:278–281. 1998;18:9733–9750.
28. Bristol LA, Rothstein JD. Glutamate transporter gene expres- 49. Chaudhry FA, Reimer RJ, Krizaj D, Barber D, Storm-
sion in amyotrophic lateral sclerosis motor cortex. Ann Neu- Mathisen J, Copenhagen DR, Edwards RH. Molecular analy-
rol 1996;39:676–679. sis of system N suggests novel physiological roles in nitrogen
29. Broadie K, Bellen HJ, DiAntonio A, Littleton JT, Schwarz metabolism and synaptic transmission. Cell 1999;99:
TL. Absence of synaptotagmin disrupts excitation–secretion 769–780.
coupling during synaptic transmission. Proc Natl Acad Sci 50. Chen MS, Obar RA, Schroeder CC, Austin TW, Poodry CA,
USA 1994;91:663–673. Wadsworth SC, Valle RB. Multiple forms of dynamin are
30. Broadie K, Prokop A, Bellen HJ, O’Kane CJ, Schulze KL, encoded by shibire, a Drosophila gene involved in endocyto-
Sweeney ST. Syntaxin and synaptobrevin function down- sis. Nature 1991;351:583–586.
stream of vesicle docking in Drosophila. Neuron 51. Chen YA, Scales SJ, Patel SM, Doung YC, Scheller RH.
1995;15:663–673. SNARE complex formation is triggered by Ca2+ and drives
31. Brodin L, Low P, Shupliakov O. Sequential steps in clathrin- membrane fusion. Cell 1999;97:165–174.
mediated synaptic vesicle endocytosis. Curr Opin Neurobiol 52. Chow RH, von Ruden L, Neher E. Delay in vesicle fusion
2000;10:312–320. revealed by electrochemical monitoring of single secretory
32. Brose N, Hofmann K, Hata Y, Sudhof TC. Mammalian ho- events in adrenal chromaffin cells. Nature 1992;356:60–63.
mologues of Caenorhabditis elegans unc-13 gene define novel 53. Conti F, Minelli A. Glutamate immunoreactivity in rat cere-

Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001 595


bral cortex is reversibly abolished by 6-diazo-5-oxo- L - striatal homogenates: evidence for an exchange diffusion
norleucine (DON), an inhibitor of phosphate-activated glu- model. J Pharmacol Exp Ther 1979;208:203–209.
taminase. J Histochem Cytochem 1994;42:717–726. 75. Fon EA, Pothos EN, Sun B-C, Killeen N, Sulzer D, Edwards
54. Cooper JR, Bloom FE, Roth RH. Acetylcholine. In: Cooper RH. Vesicular transport regulates monoamine storage and
JR, Bloom FE, Roth RH, editors. The biochemical basis of release but is not essential for amphetamine action. Neuron
neuropharmacolgoy. New York: Oxford University Press; 1997;19:1271–1283.
1996. p 194–225. 76. Frize ED. Mental depression in hypertensive patients treated
55. Cremona O, De Camilli P. Synaptic vesicle endocytosis. Curr for long periods with high doses of reserpine. N Engl J Med
Opin Neurobiol 1997;7:323–330. 1954;251:1006–1008.
56. Crowder KM, Gunther JM, Jones TA, Hale BD, Zhang HZ, 77. Geppert M, Bolshakov VY, Siegelbaum SA, Takei K, De
Peterson MR, Scheller RH, Chavkin C, Bajjalieh SM. Abnor- Camilli P, Hammer RE, Sudhof TC. The role of Rab3A in
mal neurotransmission in mice lacking synaptic vesicle pro- neurotransmitter release. Nature 1994;369:493–497.
tein 2A (SV2A). Proc Natl Acad Sci USA 1999;96:15268– 78. Geppert M, Goda Y, Hammer RE, Li C, Rosahl TW, Stevens
15273. CF, Sudhof TC. Synaptotagmin I: a major Ca2+ sensor for
57. Daniels GM, Amara SG. Regulated trafficking of the human transmitter release at a central synapse. Cell 1994;79:
dopamine transporter. Clathrin-mediated internalization 717–727.
and lysosomal degradation in response to phorbol esters. J 79. Geppert M, Goda Y, Stevens CF, Sudhof TC. The small GTP-
Biol Chem 1999;274:35794–35801. binding protein Rab3A regulates a late step in synaptic
58. David C, McPherson PS, Mundigl O, de Camilli P. A role of vesicle fusion. Nature 1997;387:810–814.
amphiphysin in synaptic vesicle endocytosis suggested by its 80. Geppert M, Sudhof TC. RAB3 and synaptotagmin: the yin
binding to dynamin in nerve terminals. Proc Natl Acad Sci and yang of synaptic membrane fusion. Annu Rev Neurosci
USA 1996;93:331–335. 1998;21:75–95.
59. Davis AF, Bai J, Fasshauer D, Wolowick MJ, Lewis JL, Chap- 81. Gilmor ML, Nash NR, Roghani A, Edwards RH, Yi H, Hersch
man ER. Kinetics of synaptotagmin responses to Ca2+ and SM, Levey AI. Expression of the putative vesicular acetylcho-
assembly with the core SNARE complex onto membranes. line transporter in rat brain and localization in cholinergic
Neuron 1999;24:363–376. synaptic vesicles. J Neurosci 1996;16:2179–2190.
60. de Heuvel E, Bell AW, Ramjaun AR, Wong K, Sossin WS,
82. Giros B, Jaber M, Jones SR, Wightman RM, Caron MG. Hy-
McPherson PS. Identification of the major synaptojanin-
perlocomotion and indifference to cocaine and amphet-
binding proteins in brain. J Biol Chem 1997;272:8710–8716.
amine in mice lacking the dopamine transporter. Nature
61. Diamond JS, Jahr CE. Transporter buffer synaptically re- 1996;379:606–612.
leased glutamate on a submillisecond time scale. J Neurosci
83. Goldstein JM, Barnett A, Malick JB. The evaluation of anti-
1997;17:4672–4687.
parkinson drugs on reserpine-induced rigidity in rats. Eur J
62. DiAntonio A, Schwarz TL. The effect on synaptic physiology
Pharmacol 1975;33:183–188.
of synaptotagmin mutations in Drosophila. Neuron
1994;12:909–920. 84. Gonzalez LJ, Scheller RH. Regulation of membrane traffick-
ing: structural insights from a Rab/effector complex. Cell
63. Edwards RH. Neural degeneration and the transport of neu-
1999;96:755–758.
rotransmitters. Ann Neurol 1993;34:638–645.
64. Edwards RH. Neurotransmitter release: variations on a 85. Grimsby J, Toth M, Chen K, Kumazawa T, Klaidman L, Ad-
theme. Curr Biol 1998;8:R883–R885. ams JD, Karoum F, Gal J, Shih JC. Increased stress response
65. Erickson JD, Eiden LE, Hoffman BJ. Expression cloning of a and beta-phenylethylamine in MAOB-deficient mice. Nat
reserpine-sensitive vesicular monoamine transporter. Proc Genet 1997;17:206–210.
Natl Acad Sci USA 1992;89:10993–10997. 86. Grote E, Kelly RB. Endocytosis of VAMP is facilitated by a
66. Erickson JD, Varoqui H, Schafer MD, Modi W, Diebler MF, synaptic vesicle targeting signal. J Cell Biol 1996;132:
Weihe E, Rand J, Eiden LE, Bonner TI, Usdin TB. Func- 537–547.
tional identification of a vesicular acetylcholine transporter 87. Guo W, Roth D, Walch-Solimena C, Novick P. The exocyst is
and its expression from a “cholinergic” gene locus. J Biol an affector for Sec4p, targeting secretory vesicles to sites of
Chem 1994;269:21929–21932. exocytosis. EMBO J 1999;18:1071–1080.
67. Evoli A, Batocchi AP, Tonali P. A practical guide to the 88. Hamberger AC, Chiang GH, Nylen ES, Scheff SW, Cotman
recognition and management of myasthenia gravis. Drugs CW. Glutamate as a CNS transmitter. I. Evaluation of glucose
1996;52:662–670. and glutamine as percursors for the synthesis of preferen-
68. Fairman WA, Amara SG. Functional diversity of excitatory tially released glutamate. Brain Res 1979;168:513–530.
amino acid transporters: ion channel and transport modes. 89. Hanson PI, Roth R, Morisaki H, Jahn R, Heuser JE. Structure
Am J Physiol 1999;277:F481–F486. and conformational changes in NSF and its mmebrane re-
69. Fairman WA, Vandenberg RJ, Arriza JL, Kavanaugh MP, ceptor complexes visualized by quick-freeze/deep-etch elec-
Amara SG. An excitatory amino-acid transporter with prop- tron microscopy. Cell 1997;90:523–535.
erties of a ligand-gated chloride channel. Nature 90. Haucke V, De Camilli P. AP-2 recruitment to synaptotagmin
1995;375:599–603. stimulated by tyrosine-based endocytic motifs. Science
70. Faundez V, Horng JT, Kelly RB. A function for the AP3 coat 1999;285:1268–1271.
complex in synaptic vesicle formation from endosomes. Cell 91. Hay JC, Fisette PL, Jenkins GH, Fukami K, Takenawa T,
1998;93:423–432. Anderson RA, Martin TF. ATP-dependent inositide phos-
71. Feany MB, Lee S, Edwards RH, Buckley KM. The synaptic phorylation required for Ca2+-activated secretion. Nature
vesicle protein SV2 is a novel type of transmembrane trans- 1995;374:173–177.
porter. Cell 1992;70:861–867. 92. Hay JC, Martin TF. Resolution of regulated secretion into
72. Feldman DA,Weinhold PA. Cytidylyltransferase-binding pro- sequential MgATP-dependent and calcium-dependent
tein is identical to transcytosis-associated protein (TAP/ stages mediated by distinct cytosolic proteins. J Cell Biol
p115) and enhances the lipid activation of cytidylytransfer- 1992;119:139–151.
ase. J Biol Chem 1998.273:102–109. 93. Hay JC, Martin TF. Phosphatidylinositol transfer protein re-
73. Fernandez-Chacon R, Sudhof TC. Genetics of synaptic quired for ATP-dependent priming of Ca2+-activated secre-
vesicle function: toward the complete functional anatomy of tion. Nature 1993;366:572–575.
an organelle. Annu Rev Physiol 1999;61:753–776. 94. Hayashi T, McMahon H, Yamasaki S, Binz T, Hata Y, Sudhof
74. Fischer JF, Cho AK. Chemical release of dopamine from TC, Niemann H. Synaptic vesicle membrane fusion com-

596 Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001


plex: action of clostridial neurotoxins on assembly. EMBO J terization of the cysteine-rich region of the Caenorhabditis
1994;13:5051–5061. elegans protein Unc-13 as a high affinity phorbol ester recep-
95. Hazuka CD, Foletti DL, Hsu SC, Kee Y, Hopf FW, Scheller tor. Analysis of ligand-binding interactions, lipid cofactor
RH. The sec6/8 complex is located at neurite outgrowth and requirements, and inhibitor sensitivity. J Biol Chem
axonal synapse-assembly domains. J Neurosci 1999;19: 1995;270:10777–10783.
1324–1334. 116. Kilberg MS, Handlogten ME, Christensen HN. Characteris-
96. Henkel AW, Almers W. Fast steps in exocytosis and endocy- tics of an amino acid transport system in rat liver for gluta-
tosis studied by capacitance measurements in endocrine mine, asparagine, histidine, and closely related analogs. J
cells. Curr Opin Neurobiol 1996;6:350–357. Biol Chem 1980;255:4011–4019.
97. Henkel AW, Sankaranarayanan S. Mechanisms of alpha- 117. Krantz DE, Waites C, Oorschot V, Liu Y, Wilson RI, Tan PK,
latrotoxin action. Cell Tissue Res 1999;296:229–233. Klumperman J, Edwards RH. A phosphorylation site regu-
98. Hessler NA, Shirke AM, Malinow R. The probability of trans- lates sorting of the vesicular acetylcholine transporter to
mitter release at a mammalian central synapse. Nature dense core vesicles. J Cell Biol 2000;149:379–395.
1993;366:569–572. 118. Krasnoperov VG, Bittner MA, Beavis R, Kuang Y, Salnikow
99. Heuser JE. Review of electron microscopic evidence favour- KV, Chepurny OG, Little AR, Plotnikov AN, Wu D, Holz RW,
ing vesicle exocytosis as the structural basis for quantal re- Petrenko AG. ␣-Latrotoxin stimulates exocytosis by the in-
lease during synaptic transmission. Q J Exp Physiol teraction with a neuronal G-protein-coupled receptor. Neu-
1989;74:1051–1069. ron 1997;18:925–937.
100. Heuser JE, Reese TS. Structural changes after transmitter 119. Kromer A, Glombik MM, Huttner WB, Gerdes HH. Essential
release at the frog neuromuscular junction. J Cell Biol role of the disulfide-bonded loop of chromogranin B for
1981;88:564–580. sorting to secretory granules is revealed by expression of a
101. Hilfiker S, Pieribone VA, Czernik AJ, Kao HT, Augustine GJ, deletion mutant in the absence of endogenous granin syn-
Greengard P. Synapsins as regulators of neurotransmitter thesis. J Cell Biol 1998;140:1331–1346.
release. Phil Trans R Soc Lond B Biol Sci 1999;354:269–279. 120. Kumer SC, Vrana KE. Intricate regulation of tyrosine hy-
102. Hinshaw JE, Schmid SL. Dynamin self-assembles into rings droxylase activity and gene expression. J Neurochem
suggesting a mechanism for coated vesicle budding. Nature 1996;67:443–462.
1995;374:190–192. 121. Lackner MR, Nurrish SJ, Kaplan JM. Facilitation of synaptic
103. Hosaka M, Hammer RE, Sudhof TC. A phospho-switch con- transmission by EGL-30 Gqalpha and EGL-8 PLCbeta: DAG
trols the dynamic association of synapsins with synaptic binding to UNC-13 is required to stimulate acetylcholine
vesicles. Neuron 1999;24:377–387. release. Neuron 1999;24:335–346.
104. Hsu SC, Hazuka CD, Roth R, Foletti DL, Heuser J, Scheller 122. Langston JW, Irwin I, Langston EB, Forno LS. Pargyline pre-
RH. Subunit composition, protein interactions, and struc- vents MPTP-induced parkinsonism in primates. Science
tures of the mammalian brain sec6/8 complex and septin 1984;225:1480–1482.
filaments. Neuron 1998;20:1111–1122.
123. Lee RY, Sawin ER, Chalfie M, Horvitz HR, Avery L. EAT-4, a
105. Hua SY, Charlton MP. Activity-dependent changes in partial homolog of a mammalian sodium-dependent inorganic
VAMP complexes during neurotransmitter release. Nat Neu- phosphate cotransporter, is necessary for glutamatergic neu-
rosci 1999;2:1078–1083. rotransmission in Caenorhabditis elegans. J Neurosci
106. Hunt JM, Bommert K, Charlton MP, Kistner A, Habermann 1999;19:159–167.
E, Augustine GJ, Betz H. A post-docking role for synaptobre-
124. Lester HA, Cao Y, Mager S. Listening to neurotransmitter
vin in synaptic vesicle fusion. Neuron 1994;12:1269–1279.
transporters. Neuron 1996;17:807–810.
107. Huttner WB, Schiebler W, Greengard P, De Camilli P. Syn-
apsin I (protein I), a nerve terminal-specific phosphopro- 125. Li C, Ullrich B, Zhang JZ, Anderson RG, Brose N, Sudhof
tein. III. Its association with synaptic vesicles studied in a TC. Ca2+-dependent and -independent activities of neural
highly purified synaptic vesicle preparation. J Cell Biol and non-neural synaptotagmins. Nature 1995;375:594–599.
1983;96:1374–1388. 126. Li L, Chin LS, Shupliakov O, Brodin L, Sihra TS, Hvalby O,
108. Isaacson JS, Walmsley B. Counting quanta: direct measure- Jensen V, Zheng D, McNamara JO, Greengard P, Andersen
ments of transmitter release at a central synapse. Neuron P. Impairment of synaptic vesicle clustering and of synaptic
1995;15:875–884. transmission, and increased seizure propensity, in synapsin
109. Janz R, Goda Y, Geppert M, Missler M, Sudhof TC. SV2A and I-deficient mice. Proc Natl Acad Sci USA 1995;92:9235–9239.
SV2B function as redundant Ca2+ regulators in neurotrans- 127. Lin CL, Bristol LA, Jin L, Dykes-Hoberg M, Crawford T,
mitter release. Neuron 1999;24:1003–1016. Clawson L, Rothstein JD. Aberrant RNA processing in a neu-
110. Jones SR, Gainetdinov RR, Jaber M, Giros B, Wightman RM, rodegenerative disease: the cause for absent EAAT2, a glu-
Caron MG. Profound neuronal plasticity in response to in- tamate transporter, in amyotrophic lateral sclerosis. Neuron
activation of the dopamine transporter. Proc Natl Acad Sci 1998;20:589–602.
USA 1998;95:4029–4034. 128. Lin RC, Scheller RH. Structural organization of the synaptic
111. Jorgensen EM, Hartwieg E, Schuske K, Nonet ML, Jin Y, exocytosis core complex. Neuron 1997;19:1087–1094.
Horvitz HR. Defective recycling of synaptic vesicles in synap- 129. Littleton JT, Chapman ER, Kreber R, Garment MB, Carlson
totagmin mutants of Caenorhabditis elegans. Nature SD, Ganetzky B. Temperature-sensitive paralytic mutations
1995;378:196–199. demonstrate that synaptic exocytosis requires SNARE com-
112. Kash SF, Johnson RS, Tecott LH, Noebels JL, Mayfield RD, plex assembly and disassembly. Neuron 1998;21:401–413.
Hanahan D, Baekkeskov S. Epilepsy in mice deficient in the 130. Littleton JT, Serano TL, Rubin GM, Ganetzky B, Chapman
65-kDa isoform of glutamic acid decarboxylase. Proc Natl ER. Synaptic function modulated by changes in the ratio of
Acad Sci USA 1997;94:14060–14065. synaptotagmin I and IV. Nature 1999;400:757–760.
113. Kash SF, Tecott LH, Hodge C, Baekkeskov S. Increased anxi- 131. Liu G, Choi S, Tsien RW. Variability of neurotransmitter
ety and altered responses to anxiolytics in mice deficient in concentration and nonsaturation of postsynaptic AMPA re-
the 65-kDa isoform of glutamic acid decarboxylase. Proc ceptors at synapses in hippocampal cultures and slices. Neu-
Natl Acad Sci USA 1999;96:1698–1703. ron 1999;22:395–409.
114. Katz B. The release of neural transmitter substances. The 132. Liu Y, Edwards RH. Differential localization of vesicular ace-
Sherrington Lectures. Volume X. Springfield, IL: CC Thom- tylcholine and monoamine transporters in PC12 cells but
as; 1969. not CHO cells. J Cell Biol 1997;139:907–916.
115. Kazanietz MG, Lewin NE, Bruns JD, Blumberg PM. Charac- 133. Liu Y, Edwards RH. The role of vesicular transport proteins

Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001 597


in synaptic transmission and neural degeneration. Annu Rev 154. Murthy VN, Stevens CF. Synaptic vesicles retain their identity
Neurosci 1997;20:125–156. through the endocytic cycle. Nature 1998;392:497–501.
134. Liu Y, Peter D, Roghani A, Schuldiner S, Prive GG, Eisen- 155. Napolitano A, Cesura AM, Da Prada M. The role of mono-
berg D, Brecha N, Edwards RH. A cDNA that supresses MPP+ amine oxidase and catechol-O-methyltransferase in dopami-
toxicity encodes a vesicular amine transporter. Cell nergic neurotransmission. J Neural Transm 1995;45:35–45.
1992;70:539–551. 156. Neher E. Vesicle pools and Ca2+ microdomains: new tools
135. Liu Y, Roghani A, Edwards RH. Gene transfer of a reserpine- for understanding their roles in neurotransmitter release.
sensitive mechanism of resistance to N-methyl-4- Neuron 1998;20:389–399.
phenylpyridinium. Proc Natl Acad Sci USA 1992;89: 157. Nelson N. The family of Na+/Cl− neurotransmitter trans-
9074–9078. porters. J Neurochem 1998;71:1785–1803.
136. Liu Y, Schweitzer ES, Nirenberg MJ, Pickel VM, Evans CJ, 158. Nirenberg MJ, Chan J, Liu Y, Edwards RH, Pickel VM. Ul-
Edwards RH. Preferential localization of a vesicular mono- trastructural localization of the vesicular monoamine trans-
amine transporter to dense core vesicles in PC12 cells. J Cell porter-2 in midbrain dopaminergic neurons: potential sites
Biol 1994;127:1419–1433. for somatodendritic storage and release of dopamine. J Neu-
137. Loyet KM, Kowalchyk JA, Chaudhary A, Chen J, Prestwich rosci 1996;16:4135–4145.
GD, Martin TFJ. Specific binding of phosphatidylinositol 4,5- 159. Nirenberg MJ, Chan J, Liu Y, Edwards RH, Pickel VM. Ve-
bisphosphate to calcium-dependent activator protein for se- sicular monoamine transporter-2: immunogold localization
cretion (CAPS), a potential phosphoinositide effector pro- in striatal axons and terminals. Synapse 1997;26:194–198.
tein for regulated exocytosis. J Biol Chem 1998;273: 160. Nirenberg MJ, Liu Y, Peter D, Edwards RH, Pickel VM. The
8337–8343. vesicular monoamine transporter-2 is present in small syn-
138. Lupashin VV, Waters MG. t-SNARE activation through tran- aptic vesicles and preferentially localizes to large dense core
sient interaction with a rab-like guanosine triphosphatase. vesicles in rat solitary tract nuclei. Proc Natl Acad Sci USA
Science 1997;276:1255–1258. 1995;92:8773–8777.
139. Magistretti PJ, Pellerin L, Rothman DL, Shulman RG. En- 161. Nonet ML, Staunton JE, Kilgard MP, Fergestad T, Hartwieg
ergy on demand. Science 1999;283:496–497. E, Horvitz HR, Jorgensen EM, Meyer BJ. Caenorhabditis el-
140. Mainen ZF, Malinow R, Svoboda K. Synaptic calcium tran- egans rab-3 mutant synapses exhibit impaired function and
sients in single spines indicate that NMDA receptors are not are partially depleted of vesicles. J Neurosci 1997;17:8061–
saturated. Nature 1999;399:151–155. 8073.
162. Okuda T, Haga T, Kanai Y, Endou H, Ishihara T, Katsura I.
141. Malenka RC, Nicoll RA. Long-term potentiation — a decade
Identification and characterization of the high-affinity cho-
of progress? Science 1999;285:1870–1874.
line transporter. Nature Neurosci 2000;3:120–125.
142. Marsh M, McMahon HT. The structural era of endocytosis.
163. Orci L, Perrelet A, Rothman JE. Vesicles on strings: morpho-
Science 1999;285:215–220.
logical evidence for processive transport within the Golgi
143. Martin TF. The molecular machinery for fast and slow neu- stack. Proc Natl Acad Sci USA 1998;95:2279–2283.
rosecretion. Curr Opin Neurobiol 1994;4:626–632.
164. Orlova EV, Rahman MA, Gowen B, Volynski KE, Ashton AC,
144. Maruyama IN, Brenner S. A phorbol ester/diacylglycerol- Manser C, van Heel, Ushkaryov YA. Structure of alpha-
binding protein encoded by the unc-13 gene of Caenorhab- latrotoxin oligomers reveals that divalent cation-dependent
ditis elegans. Proc Natl Acad Sci USA 1991;88:5729–5733. tetramers form membrane pores. Nat Struct Biol 2000;7:48–
145. Masson J, Sagné C, Hamon M, Mestikawy SE. Neurotrans- 53.
mitter transporters in the central nervous system. Pharmacol 165. Ottersen OP, Zhang N, Walberg F. Metabolic compartmen-
Rev 1999;51:439–464. tation of glutamate and glutamine: morphological evidence
146. Mastrogiacomo A, Parsons SM, Zampighi GA, Jenden DJ, obtained by quantitative immunocytochemistry in rat cer-
Umbach JA, Gundersen CB. Cysteine string proteins: a po- ebellum. Neuroscience 1992;46:519–534.
tential link between synaptic vesicles and presynaptic Ca2+ 166. Oyler GA, Higgins GA, Hart RA, Battenberg E, Billingsley M,
channels. Science 1994;263:981–982. Bloom FE, Wilson MC. The identification of a novel synap-
147. McIntire SL, Reimer RJ, Schuske K, Edwards RH, Jorgensen tosomal-associated protein, SNAP-25, differentially ex-
EM. Identification and characterization of the vesicular pressed by neuronal subpopulations. J Cell Biol 1989;109:
GABA transporter. Nature 1997;389:870–876. 3039–3052.
148. McPherson PS, Garcia EP, Slepnev VI, David C, Zhang X, 167. Pahud G, Salem N, van de Goor J, Medilanski J, Pellegrinelli
Grabs D, Sossin WS, Bauerfeind R, Nemoto Y, De Camilli P. N, Eder-Colli L. Study of subcellular localization of mem-
A presynaptic inositol-5-phosphatase. Nature 1996;379: brane-bound choline acetyltransferase in Drosophila central
353–357. nervous system and its association with membranes. Eur J
149. Micheva KD, Kay BK, McPherson PS. Synaptojanin forms two Neurosci 1998;10:1644–1653.
separate complexes in the nerve terminal. Interactions with 168. Peng HB, Xie H, Rossi SG, Rotundo RL. Acetylcholinester-
endophilin and amphiphysin. J Biol Chem 1997;272:27239– ase clustering at the neuromuscular junction involves perle-
27245. can and dystroglycan. J Cell Biol 1999;145:911–921.
150. Millard CB, Broomfield CA. Anticholinesterases: medical ap- 169. Peter D, Jimenez J, Liu Y, Kim J, Edwards RH. The chromaf-
plications of neurochemical principles. J Neurochem 1995; fin granule and synaptic vesicle amine transporters differ in
64:1909–1918. substrate recognition and sensitivity to inhibitors. J Biol
151. Miller KG, Emerson MD, Rand JB. Goalpha and diacylglyc- Chem 1994;269:7231–7237.
erol kinase negatively regulate the Gq-alpha pathway in C. 170. Peters A, Palay SL, Webster HF. The fine structure of the
elegans. Neuron 1999;24:323–333. nervous system. New York: Oxford University Press; 1991.
152. Morimoto T, Wang XH, Poo MM. Overexpression of synap- 171. Peters C, Andrews PD, Stark MJ, Cesaro-Tadic S, Glatz A,
totagmin modulates short-term synaptic plasticity at devel- Podtelejnikov A, Mann M, Meyer A. Control of the terminal
oping neuromuscular junctions. Neuroscience 1998;82:969– step of intracellular membrane fusion by protein phospha-
978. tase 1. Science 1999;285:1084–1087.
153. Mowla SJ, Pareek S, Farhadi HF, Petrecca K, Fawcett JP, 172. Peters C, Mayer A. Ca2+ calmodulin signals the completion
Seidah NG, Morris SJ, Sossin WS, Murphy RA. Differential of docking and triggers a late step of vacuole fusion. Nature
sorting of nerve growth factor and brain-derived neuro- 1998;396:575–580.
trophic factor in hippocampal neurons. J Neurosci 1999;19: 173. Pieribone VA, Shupliakov O, Brodin L, Hilfiker-Rothenfluh
2069–2080. S, Czernik AJ, Greengard P. Distinct pools of synaptic

598 Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001


vesicles in neurotransmitter release. Nature 1995;375:493– Smith SJ. The kinetics of synaptic vesicle recycling measured
497. at single presynaptic boutons. Neuron 1993;11:713–724.
174. Poodry CA, Edgar L. Reversible alteration in the neuromus- 195. Ryan TA, Smith SJ. Vesicle pool mobilization during action
cular junctions of Drosophila melanogaster bearing a tem- potential firing at hippocampal synapses. Neuron 1995;14:
perature-sensitive mutation, shibire. J Cell Biol 1979;81:520– 983–989.
527. 196. Ryan TA, Ziv NE, Smith SJ. Potentiation of evoked vesicle
175. Pothos EN, Larsen KE, Krantz DE, Liu Y-J, Haycock JW, Set- turnover at individually resolved synaptic boutons. Neuron
lik W, Gershon MD, Edwards RH, Sulzer D. Synaptic vesicle 1996;17:125–134.
transporter expression regulates vesicle phenotype and 197. Sassa T, Harada S, Ogawa H, Rand JB, Maruyama IN, Ho-
quantal size. J Neurosci 2000;20:7297–7306. sono R. Regulation of the UNC-18–Caenorhabditis elegans syn-
176. Pow DV, Barnett NL. Developmental expression of excitato- taxin complex by UNC-13. J Neurosci 1999;19:4772–4777.
ry amino acid transporter 5: a photoreceptor and bipolar 198. Satoh Y, Hirashima N, Tokumaru H, Takahashi MP, Kang J,
cell glutamate transporter in rat retina. Neurosci Lett Viglione MP, Kim YI, Kirino Y. Lambert–Eaton syndrome
2000;280:21–24. antibodies inhibit acetylcholine release and P/Q-type Ca2+
177. Ramamoorthy S, Blakely RD. Phosphorylation and seques- channels in electric ray nerve endings. J Physiol (Lond)
tration of serotonin transporters differentially modulated by 1998;508:427–438.
psychostimulants. Science 1999;285:763–766. 199. Schiavo G, Rossetto O, Montecucco C. Clostridial neurotox-
178. Ramamoorthy S, Giovanetti E, Qian Y, Blakely RD. Phos- ins as tools to investigate the molecular events of neurotrans-
phorylation and regulation of antidepressant-sensitive sero- mitter release. Semin Cell Biol 1994;5:221–229.
tonin transporters. J Biol Chem 1998;273:2458–2466. 200. Schivell AE, Batchelor RH, Bajjalieh SM. Isoform-specific,
179. Ranjan R, Bronk P, Zinsmaier KE. Cysteine string protein is calcium-regulated interaction of the synaptic vesicle proteins
required for calcium secretion coupling of evoked neuro- SV2 and synaptotagmin. J Biol Chem 1996;271:27770–27775.
transmission in drosophila but not for vesicle recycling. J 201. Schmid SL, McNiven MA, De Camilli P. Dynamin and its
Neurosci 1998;18:956–964. partners: a progress report. Curr Opin Cell Biol 1998;10:
180. Reimer RJ, Chaudhry FA, Gray AT, Edwards RH. Amino acid 504–512.
transport system A resembles system N in sequence but dif- 202. Schmidt A, Wolde M, Thiele C, Fest W, Kratzin H, Podtelej-
fers in mechanism. Proc Natl Acad Sci USA 2000;97:7715– nikov AV, Witke W, Huttner WB, Soling HD. Endophilin I
7720. mediates synaptic vesicle formation by transfer of arachido-
181. Reimer RJ, Fon EA, Edwards RH. Vesicular neurotransmitter nate to lysophosphatidic acid. Nature 1999;401:133–141.
transport and the presynaptic regulation of quantal size. 203. Seal RP, Amara SG. Excitatory amino acid transporters: a
Curr Opin Neurobiol 1998;8:405–412. family in flux. Annu Rev Pharmacol Toxicol 1999;39:431–
182. Richmond JE, Davis WS, Jorgensen EM. UNC-13 is required 456.
for synaptic vesicle fusion in C. elegans. Nat Neurosci 204. Seiden LS, Sabol KE, Ricaurte GA. Amphetamine: effects on
1999;2:959–964. catecholamine systems and behavior. Annu Rev Pharmacol
183. Rizo J, Sudhof TC. C2-domains, structure and function of a Toxicol 1993;32:639–677.
universal Ca2+-binding domain. J Biol Chem 1998;273: 205. Sever S, Muhlberg AB, Schmid SL. Impairment of dynamin’s
15879–15882. GAP domain stimulates receptor-mediated endocytosis. Na-
184. Roghani A, Feldman J, Kohan SA, Shirzadi A, Gundersen ture 1999;398:481–486.
CB, Brecha N, Edwards RH. Molecular cloning of a putative 206. Shao X, Fernandez I, Sudhof TC, Rizo J. Solution structures
vesicular transporter for acetylcholine. Proc Natl Acad Sci of the Ca2+-free and Ca2+-bound C2A domain of synaptotag-
USA 1994;91:10620–10624. min I: does Ca2+ induce a conformational change? Biochem-
185. Roos J, Kelly RB. The endocytic machinery in nerve termi- istry 1998;37:16106–16115.
nals surrounds sites of exocytosis. Curr Biol 1999;9:1411– 207. Shao X, Li C, Fernandez I, Zhang X, Sudhof TC, Rizo J.
1414. Synaptotagmin-syntaxin interaction: the C2 domain as a
186. Rosahl TW, Spillane D, Missler M, Herz J, Selig DK, Wolff JR, Ca2+-dependent electrostatic switch. Neuron 1997;18:133–
Hammer RE, Malenka, RC, Suchof TC. Essential functions 142.
of synapsins I and II in synaptic vesicle regulation. Nature 208. Shi G, Faundez V, Roos J, Dell’Angelica EC, Kelly RB. Neu-
1995;375:488–493. roendocrine synaptic vesicles are formed in vitro by both
187. Rosenmund C, Clements JD, Westbrook GL. Nonuniform clathrin-dependent and clathrin-independent pathways. J
probability of glutamate release at a hippocampal synapse. Cell Biol 1998;143:947–955.
Science 1993;262:754–757. 209. Shih JC, Chen K, Ridd MJ. Monoamine oxidase: from genes
188. Rosenmund C, Stevens CF. Definition of the readily releas- to behavior. Annu Rev Neurosci 1999;22:197–217.
able pool of vesicles at hippocampal synapses. Neuron 210. Shoulson I. DATATOP: a decade of neuroprotective inquiry.
1996;16:1197–1207. Parkinson study group. Deprenyl and tocopherol antioxida-
189. Rossetto O, Schiavo G, Montecucco C, Poulain B, Deloye F, tive therapy of parkinsonism. Ann Neurol 1998;44(suppl):
Lozzi L, Shone CC. SNARE motif and neurotoxins. Nature S160–S166.
1994;372:415–416. 211. Shupliakov O, Low P, Grabs D, Gad H, Chen H, David C,
190. Rothman JE. Mechanisms of intracellular protein transport. Takei K, De Camilli P, Brodin L. Synaptic vesicle endocytosis
Nature 1994;372:55–63. impaired by disruption of dynamin-SH3 domain interac-
191. Rothstein JD, Dykes-Hoberg M, Pardo CA, Bristol LA, Jin L, tions. Science 1997;276:259–263.
Kuncl RW, Kanai Y, Hediger MA, Wang Y, Schielke JP. 212. Soghomonian J-J, Martin DL. Two isoforms of glutamate de-
Knockout of glutamate transporters reveals a major role for carboxylase: why? Trends Pharmocol Sci 1998;19:500–505.
astroglial transport in excitotoxicity and clearance of gluta- 213. Solimena M, De Camilli P. Autoimmunity to glutamic acid
mate. Neuron 1996;16:675–686. decarboxylase (GAD) in stiff-man syndrome and insulin-
192. Rothstein JD, Tabakoff B. Alteration of striatal glutamate dependent diabetes mellitus. Trends Neurosci 1991;14:452–
release after glutamine synthetase inhibition. J Neurochem 457.
1984;43:1438–1446. 214. Solimena M, Folli F, Aparisi R, Pozza G, De Camilli P. Au-
193. Rotundo RL, Rossi SG, Anglister L. Transplantation of quail toantibodies to GABA-ergic neurons and pancreatic beta
collagen-tailed acetylcholinesterase molecules onto the frog cells in stiff-man syndrome. N Engl J Med 1990;322:1555–
neuromuscular synapse. J Cell Biol 1997;136:367–374. 1560.
194. Ryan TA, Reuter H, Wendland B, Schweizer FE, Tsien RW, 215. Sollner T, Bennett MK, Whiteheart SW, Scheller RH, Roth-

Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001 599


man JE. A protein assembly–disassembly pathway in vitro 236. TerBush DR, Maurice T, Roth D, Novick P. The exocyst is a
that may correspond to sequential steps of synaptic vesicle multiprotein complex required for exocytosis in Saccharomy-
docking, activation, and fusion. Cell 1993;75:409–418. ces cerevisiae. EMBO J 1996;15:6483–6494.
216. Sollner T, Rothman JE. Neurotransmission: harnessing fu- 237. Thanki CM, Sugden D, Thomas AJ, Bradford HF. In vivo
sion machinery at the synapse. Trends Neurosci 1994;17: release from cerebral cortex of [14C]glutamate synthesized
344–348. from [U-14C]glutamine. J Neurochem 1983;41:611–617.
217. Sollner T, Whiteheart SW, Brunner M, Erdjument-Bromage 238. Tian N, Petersen C, Kash S, Baekkeskov S, Copenhagen D,
H, Geromanos S, Tempst P, Rothman JE. SNAP receptors Nicoll R. The role of the synthetic enzyme GAD65 in the
implicated in vesicle targeting and fusion. Nature 1993;362: control of neuronal gamma-aminobutyric acid release. Proc
318–324. Natl Acad Sci USA 1999;96:12911–12916.
218. Sonders MS, Amara SG. Channels in transporters. Curr 239. tom Dieck S, Sanmarti-Vila L, Langnaese K, Richter K, Kind-
Opin Neurobiol 1996;6:294–302. ler S, Soyke A, Wex H, Smalla KH, Kampf U, Franzer JT,
219. Sonders MS, Zhu S-J, Zahniser NR, Kavanaugh MP, Amara Stumm M, Garner CC, Gundelfinger ED. Bassoon, a novel
SG. Multiple ionic conductances of the human dopamine zinc-finger CAG/glutamine-repeat protein selectively local-
transporter: the actions of dopamine and psychostimulants. ized at the active zone of presynaptic nerve terminals. J Cell
J Neurosci 1997;17:960–974. Biol 1998;142:499–509.
220. Song H-j, Ming G-l, Fon E, Bellocchio E, Edwards RH, Poo 240. Trimble WS, Cowan DM, Scheller RH. VAMP-1: a synaptic
MM. Expression of a putative vesicular acetylcholine trans- vesicle-associated integral membrane protein. Proc Natl
porter facilitates quantal transmitter packaging. Neuron Acad Sci USA 1988;85:4538–4542.
1997;18:815–826. 241. Ubach J, Zhang X, Shao X, Sudhof TC, Rizo J. Ca2+ binding
221. Stefani G, Onofri F, Valtorta F, Vaccaro P, Greengard P, to synaptotagmin: how many Ca2+ ions bind to the tip of a
Benfenati F. Kinetic analysis of the phosphorylation- C2-domain? EMBO J 1998;17:3921–3930.
dependent interactions of synapsin I with rat brain synaptic 242. Ullrich O, Stenmark H, Alexandrov K, Huber LA, Kaibuchi
vesicles. J Physiol (Lond) 1997;504:501–515. K, Sasaki T, Takai Y, Zerial M. Rab GDP dissociation inhibi-
222. Steinhardt RA, Alderton JM. Calmodulin confers calcium tor as a general regulator for the membrane association of
sensitivity on secretory exocytosis. Nature 1982;295:154–155. rab proteins. J Biol Chem 1993;268:18143–18150.
223. Sudhof TC. The synaptic vesicle cycle: a cascade of protein– 243. Umbach JA, Saitoe M, Kidokoro Y, Gundersen CB. Attenu-
protein interactions. Nature 1995;375:645–653. ated influx of calcium ions at nerve endings of csp and shi-
224. Sudhof TC, Rizo J. Synaptotagmins: C2-domain proteins that bire mutant Drosophila. J Neurosci 1998;18:3233–3240.
regulate membrane traffic. Neuron 1996;17:379–388. 244. Ungermann C, Sato K, Wickner W. Defining the functions of
225. Sugita S, Hata Y, Sudhof TC. Distinct Ca2+-dependent prop- trans-SNARE pairs. Nature 1998;396:543–548.
erties of the first and second C2-domains of synaptotagmin I. 245. van der Bliek AM, Meyerowitz EM. Dynamin-like protein
J Biol Chem 1996;271:1262–1265. encoded by the Drosophila shibire gene associated with ve-
226. Sugita S, Khvochtev M, Sudhof TC. Neurexins are functional sicular traffic. Nature 1991;351:411–414.
alpha-latrotoxin receptors. Neuron 1999;22:489–496. 246. Varoqui H, Erickson JD. The cytoplasmic tail of the vesicular
227. Sutton KG, McRory JE, Guthrie H, Murphy TH, Snutch TP. acetylcholine transporter contains a synaptic vesicle target-
P/Q-type calcium channels mediate the activity-dependent ing signal. J Biol Chem 1998;273:9094–9098.
feedback of syntaxin-1A. Nature 1999;401:800–804. 247. Varoqui H, Zhu H, Yao D, Ming H, Erickson JD. Cloning and
228. Sutton RB, Davletov BA, Berghuis AM, Sudhof TC, Sprang functional identification of a neuronal glutamine trans-
SR. Structure of the first C2 domain of synaptotagmin I: a porter. J Biol Chem 2000;275:4049–4054.
novel Ca2+ phospholipid-binding fold. Cell 1995;80:929–938. 248. Vesce S, Bezzi P, Volterra A. The highly integrated dialogue
229. Sutton RB, Fasshauer D, Jahn R, Brunger AT. Crystal struc- between neurons and astrocytes in brain function. Sci Progr
ture of a SNARE complex involved in synaptic exocytosis at 1999;82:251–270.
2.4 A resolution. Nature 1998;395:347–353. 249. Wadiche JI, Amara SG, Kavanaugh MP. Ion fluxes associated
230. Takahashi N, Miner LL, Sora I, Ujike H, Revay RS, Kostic V, with excitatory amino acid transport. Neuron 1995;15:721–
Jackson-Lewis V, Przedborski S, Uhl GR. VMAT2 knockout 728.
mice: heterozygotes display reduced amphetamine-condi- 250. Walent JH, Porter BW, Martin TF. A novel 145 kd brain
tioned reward, enhanced amphetamine locomotion, and en- cytosolic protein reconstitutes Ca2+-regulated secretion in
hanced MPTP toxicity. Proc Natl Acad Sci USA 1997;94: permeable neuroendocrine cells. Cell 1992;70:765–775.
9938–9943. 251. Wang Y-M, Gainetdinov RR, Fumagalli F, Xu F, Jones SR,
231. Takei K, McPherson PS, Schmid SL, De Camilli P. Tubular Bock CB, Miller GW, Wightman RM, Caron MG. Knockout
membrane invaginations coated by dynamin rings are in- of the vesicular monoamine transporter 2 gene results in
duced by GTP-gamma S in nerve terminals. Nature 1995; neonatal death and supersensitivity to cocaine and amphet-
374:186–190. amine. Neuron 1997;19:1285–1296.
232. Tamarappoo BK, Raizada MK, Kilberg MS. Identification of 252. Warnock DE, Schmid SL. Dynamin GTPase, a force-gener-
a system N-like Na(+)-dependent glutamine transport activ- ating molecular switch. Bioessays 1996;18:885–893.
ity in rat brain neurons. J Neurochem 1997;68:954–960. 253. Weber T, Zemelman BV, McNew JA, Westermann B, Gmachl
233. Tan PK, Waites C, Liu Y, Krantz DE, Edwards RH. A leucine- M, Parlati F, Sollner TH, Rothman JE. SNAREpins: minimal
based motif mediates the endocytosis of vesicular mono- machinery for membrane fusion. Cell 1998;92:759–772.
amine and acetylcholine transporters. J Biol Chem 1998;273: 254. Weihe E, Tao-Cheng J-H, Schafer MK-H, Erickson JD, Eiden
17351–17360. LE. Visualization of the vesicular acetylcholine transporter
234. Tanaka K, Watase K, Manabe T, Yamada K, Watanabe M, in cholinergic nerve terminals and its targeting to a specific
Takahashi K, Iwama H, Nishikawa T, Ichihara N, Kikuchi T, population of small synaptic vesicles. Proc Natl Acad Sci USA
Okuyama S, Kawashima N, Hori S, Takimoto M, Wada K. 1996;93:3547–3552.
Epilepsy and exacerbation of brain injury in mice lacking 255. Wiedemann C, Schafer T, Burger MM. Chromaffin granule-
the glutamate transporter GLT-1. Science 1997;276:1699– associated phosphatidylinositol 4-kinase activity is required
1702. for stimulated secretion. EMBO J 1996;15:2094–2101.
235. Tandon A, Bannykh S, Kowalchyk JA, Banerjee A, Martin TF, 256. Wiedemann C, Schafer T, Burger MM, Sihra TS. An essential
Balch WE. Differential regulation of exocytosis by calcium role for a small synaptic vesicle-associated phosphatidylino-
and CAPS in semi-intact synaptosomes. Neuron 1998;21: sitol 4-kinase in neurotransmitter release. J Neurosci 1998;
147–154. 18:5594–602.

600 Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001


257. Wightman RM, Schroeder TJ, Finnegan JM, Ciolkowski EL, tions for membrane fusion specificity. J Biol Chem 1999;274:
Pihel K. Time course of release of catecholamines from in- 5649–5653.
dividual vesicles during exocytosis at adrenal medullary cells. 264. Zerangue N, Kavanaugh MP. Flux coupling in a neuronal
Biophys J 1995;68:383–390. glutamate transporter. Nature 1996;383:634–637.
258. Winkler H. Membrane composition of adrenergic large and 265. Zhang JZ, Davletov BA, Sudhof TC, Anderson RG. Synapto-
small dense cored vesicles and of synaptic vesicles: conse- tagmin I is a high affinity receptor for clathrin AP-2: impli-
quences for their biogenesis. Neurochem Res 1997;22:921– cations for membrane recycling. Cell 1994;78:751–760.
932. 266. Zhong H, Yokoyama CT, Scheuer T, Catterall WA. Recipro-
259. Wu D, Hersh LB. Choline acetyltransferase: celebrating its cal regulation of P/Q-type Ca2+ channels by SNAP-25, syn-
fiftieth year. J Neurochem 1994;62:1653–1663. taxin and synaptotagmin. Nature Neurosci 1999;2:939–941.
260. Wu MN, Bellen HJ. Genetic dissection of synaptic transmis- 267. Zhou QY, Quaife CJ, Palmiter RD. Targeted disruption of
sion in Drosophila. Curr Opin Neurobiol 1997;7:624–630. the tyrosine hydroxylase gene reveals that catecholamines
261. Xu T, Binz T, Niemann H, Neher E. Multiple kinetic com- are required for mouse fetal development. Nature 1995;
ponents of exocytosis distinguished by neurotoxin sensitiv- 374:640–643.
ity. Nature Neurosci 1998;1:192–200. 268. Zigmond RE, Schwarzschild MA, Rittenhouse AR. Acute
262. Xu YF, Hewett SJ, Atchison WD. Passive transfer of Lam- regulation of tyrosine hydroxylase by nerve activity and by
bert–Eaton myasthenic syndrome induces dihydropyridine neurotransmitters via phosphorylation. Annu Rev Neurosci
sensitivity of ICa in mouse motor nerve terminals. J Neuro- 1989;12:415–461.
physiol 1998;80:1056–1069. 269. Zucker RS, Kullmann DM, Bennett M. Release of neuro-
263. Yang B, Gonzalez LJ, Prekeris R, Steegmaier M, Advani RJ, transmitters. Fundamental neuroscience. San Diego, CA:
Scheller RH. SNARE interactions are not selective. Implica- Academic Press; 1999. p 155–192.

Mechanisms of Neurotransmitter Release MUSCLE & NERVE May 2001 601

You might also like