SlideShare a Scribd company logo
1 of 9
Download to read offline
Special Issue: Membrane Trafficking
Tethering the assembly of SNARE
complexes
WanJin Hong1,2
and Sima Lev3
1
School of Pharmaceutical Sciences, Xiamen University, Xiamen, People’s Republic of China
2
Institute of Molecular and Cell Biology, Singapore
3
Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel
The fusion of transport vesicles with their target mem-
branes is fundamental for intracellular membrane traf-
ficking and diverse physiological processes and is driven
by the assembly of functional soluble N-ethylmaleimide-
sensitive factor attachment protein receptor (SNARE)
complexes. Prior to fusion, transport vesicles are physi-
cally linked to their target membranes by various teth-
ering factors. Recent studies suggest that tethering
factors also positively regulate the assembly of function-
al SNARE complexes, thereby coupling tethering with
fusion events. This coupling is mediated, at least in part,
by direct physical interactions between tethering fac-
tors, SNAREs, and Sec1/Munc18 (SM) proteins. In this
review we summarize recent progress in understanding
the roles of tethering factors in the assembly of specific
and functional SNARE complexes driving membrane-
fusion events.
Membrane fusion
Membrane fusion along the secretory and endocytic path-
ways is mediated by a large group of membrane-bound
proteins known as SNAREs. All SNAREs contain a char-
acteristic coiled-coil SNARE motif of approximately 70
amino acids comprising heptad repeats and can function
on either target membranes (t-SNAREs) or transport vesi-
cles (v-SNAREs). The interaction of cognate v- and t-
SNAREs leads to the formation of a trans-SNARE complex
or SNAREpin, in which four SNARE motifs assemble as a
twisted parallel four-helix bundle that brings the opposing
membranes together and eventually catalyzes membrane
fusion [1,2]. Following fusion, the remaining SNARE com-
plex on the fused membranes is referred to as a cis-SNARE
complex, which undergoes disassembly catalyzed by
ATPase N-ethylmaleimide-sensitive fusion protein (NSF)
and its cofactor soluble NSF attachment protein (SNAP) [3]
to recycle the SNAREs for a new fusion event [4].
Pairing of distinct SNAREs and subsequent assembly of
functional SNAREpins are employed by different mem-
brane-trafficking events [5]. In many cases, a single
SNARE can be assembled into more than one SNAREpin
and thus can regulate several fusion events [6,7]. Hence,
the assembly of a functional SNAREpin is remarkably
important for both membrane fusion specificity and coor-
dinating different membrane-trafficking pathways. Recon-
stitution studies suggest that numerous SNARE
complexes can be formed in vitro, but only a few can
functionally drive membrane fusion [8,9]. Furthermore,
the assembly of a functional SNAREpin occurs in a step-
wise manner and is attributed to the formation of SNARE
complex intermediates, such as t-SNARE subcomplexes on
the target membranes that provide templates for v-
SNARE binding [7]. These findings raise several critical
questions. What are the mechanisms that regulate the
assembly of specific SNAREpins? How is the promiscuous
assembly of nonfunctional SNARE complexes prevented in
intact cells? How are the intermediate steps of the assem-
bly process regulated?
Numerous studies have shown that membrane-fusion
specificity is largely dependent on SNARE proteins, be-
cause different SNAREpins can catalyze distinct fusion
events [1,6]. Recent studies, however, suggest that both
SM proteins and tethering factors play an active role in
SNAREpin formation and thus contribute to the specificity
of the fusion as well as to its speed and high fidelity [10–
15]. Tethering factors appear to influence SNAREpin as-
sembly in several different ways including stabilization of
SNARE proteins or the entire SNARE complex, gathering
of t-SNAREs on the target membranes, or activating the
assembly process by interacting with SM proteins. The
direct physical interactions of tethering factors with
SNAREs and SM proteins mediate many of these func-
tions, as will be discussed in this review. Tethering factors
also interact with Rab GTPases and vesicle coats and are
believed to bridge the transport vesicles with their target
membranes [16,17], thereby functionally coupling tether-
ing with fusion events. This review summarizes recent
advances in understanding the role of tethering factors
in the assembly of functional SNARE complexes.
SNARE: general features, classification, and complexes
The SNARE hypothesis was first introduced in 1993 [3]
and suggested that a v-SNARE on the vesicle pairs with
cognate t-SNAREs on the target membrane to form a
complex that not only determines the specificity of the
Review
0962-8924/$ – see front matter
ß 2013 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tcb.2013.09.006
Corresponding author: Lev, S. (Sima.Lev@weizmann.ac.il).
Keywords: SNAREs; SM proteins; tethering factors; SNAREpin; membrane fusion.
Trends in Cell Biology, January 2014, Vol. 24, No. 1 35
fusion but also catalyzes the fusion process. Since then
many SNAREs have been identified, of which 38 are known
to exist in humans [6,18] (Figure 1). Most SNAREs contain
one SNARE motif, whereas four SNAREs (SNAP-23, SNAP-
25, SNAP-29/GS32, and SNAP-47) contain two SNARE
motifs [6,18–20] (Figure 1). Initially, SNAREs were classi-
fied as v- or t-SNAREs according to their subcellular locali-
zation. However, many SNAREs can be found on both
vesicles and target membranes; therefore, an alternative
classification based on the crystal structure of the synaptic
SNARE complex (Syntaxin [Stx]1A, synaptobrevin2, and
Synaptosome-associated protein [SNAP]-25B) has been for-
mulated [21]. The crystal structure revealed that an ionic
layer comprising an arginine (R) (contributed by synapto-
brevin) and three glutamine (Q) residues (contributed by
Stx1A and SNAP-25) is present in the central position of the
four-helix bundle. These findings led to the structural clas-
sification of R- and Q-SNAREs [22] and suggested that a
functional SNARE complex generally comprises three Q-
SNARE motifs and one R-SNARE motif. Most R-SNAREs
act as v-SNAREs and most Q-SNAREs act as t-SNAREs.
However, there are exceptions: the R-SNAREs Ykt6 and
Sec22B act as part of the t-SNARE subcomplexes, whereas
GS15, Bet1, and Slt1 are Q-SNAREs that function as v-
SNAREs. In addition to the SNARE motif, most SNAREs
contain a C-terminal hydrophobic transmembrane domain
(TM) (Figure 1). Seven of the 38 SNAREs do not have a TM
domain and instead associate with membranes by various
lipid modifications. SNAP-23, SNAP-25, SNAP-29,
SNAP47, Stx9/19, and Stx11 are palmitoylated at multiple
cysteineresidues, whereasYkt6 containsa C-terminalmotif
that mediates its prenylation (farnesylation) and subse-
quent palmitoylation [23]. Moreover, many SNAREs con-
tain N-terminal regulatory regions preceding the SNARE
motif (Figure 1). These N-terminal regions regulate impor-
tant properties of SNAREs. For example, the short N-ter-
minal peptide of Stx1, Stx5, and Stx16 interacts with SM
proteins [6]. The Habc region ofStx16folds intoa three-helix
bundle and intramolecularily interacts with the SNARE
motif, thereby preventing its assembly into a SNARE com-
plex [24]. Similarly, the longin domain of Vamp7, an
R-SNARE involved in both endocytic and secretory path-
ways, interacts with its SNARE motif via hydrophobic inter-
actions [25]. The N-terminal extension of VAMP4 contains a
dileucine motif and acidic clusters that mediate its recycling
fromtheendosometothetrans-Golginetwork(TGN)[26,27].
Because the formation of a functional SNAREpin is the
key driver for membrane fusion, identification of functional
SNARE complexes in vivo has been an important area of
SNARE research. The major SNARE complexes function-
ing in the secretory and endocytic pathways are shown in
Figure 2 [6]. As can be seen, most SNAREpins contain at
least one t-SNARE belonging to the Stx family, and a given
SNARE can participate in several distinct SNARE com-
plexes. For example, Stx5 is present in at least three
different SNAREpins regulating Golgi trafficking and thus
can be envisioned as the master SNARE of the Golgi
apparatus [28].
Regulation of SNAREpin assembly and function
Given that SNAREpins mediate fusion events, it is no
surprise that several mechanisms regulate their formation
and/or functionality. Below we discuss the role of SM
proteins and tethering factors in SNAREpin assembly.
SM proteins
SM proteins are evolutionarily conserved soluble periph-
eral membrane proteins of 60–90 kDa. Four major classes
of SM proteins have been identified in mammals: Sly1,
VPS45, VPS33 (VPS33A, VPS33B), and Munc18 (Munc18-
1, Munc18-2, and Munc18-3) [6,10]. SM proteins are con-
sidered key universal components of the fusion machinery.
They function in distinct intracellular transport steps and
physically interact with different SNARE proteins
(Figure 2) [7,10,29]. Sly1, for example, functions at the
endoplasmic reticulum (ER)–Golgi, Vps45 at the endo-
some–TGN, Vps33 at the endocytic/lysosomal system,
and Muc18 at the plasma membrane (Figure 2). In vitro
reconstitution studies convincingly showed that SM pro-
teins strongly accelerate the rate of SNARE-mediated
fusion and contribute to the specificity of various fusion
events [11,30,31]. Indeed, SM proteins are required for all
Stx1, 2, 3, 4, 5, 7, 12/13, 16, 17, 18Ha SNARE moƟfHb Hc TMN-Pep
VƟ1a, VƟ1b, GS27, GS28, Stx6, 8, 10Ha SNARE moƟfHb Hc TM
VAMP7, Sec22b, Sec22a, Sec22cSNARE moƟfLongin TM
Slt1/Use1, Sec20
SNARE moƟf TM VAMP1, 2, 3, 5, 8, GS15, Bet1
Ha SNARE moƟfHb HcN-Pep
SNARE moƟfLongin
VAMP4SNARE moƟfTGN S TM
SNARE moƟf TMUnknown
SNAP-23, 25, 29, 47 (mulƟple Cys for palmitoylaƟon)
Ykt6 (C-terminal prenylaƟon and palmitoylaƟon)
Stx9/19, 11 (mulƟple Cys for palmitoylaƟon)
SNARE moƟfSNARE MoƟf
TRENDS in Cell Biology
Figure 1. Domain organization of human soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs). The indicated SNAREs along with their
corresponding domains are shown. Abbreviation: TM, transmembrane domain.
Review Trends in Cell Biology January 2014, Vol. 24, No. 1
36
SNARE-mediated fusion events and recent studies on
Vps33 suggest that SM proteins promote the opening of
a fusion pore by enhancing the activity of a SNARE com-
plex [32].
SM proteins can bind individual SNAREs or SNARE
complexes and regulate SNAREpin assembly [10]. Sly1
binds to a short, evolutionarily conserved N-terminal pep-
tide of the mammalian t-SNAREs Stx5 and Stx18 and their
yeast homologs Sed5p and Ufe1p, respectively [33]. Vps45
similarly interacts with the N-terminal peptide of Stx16
(Tlg2p in yeast) and regulates endosome-to-TGN retro-
grade trafficking [34]. Interestingly, Vps45p also interacts
with the v-SNARE Snc2p, which participates in SNARE
complexes containing Tlg2p in yeast. Hence, Vps45p can
bind Tlg2p-containing SNARE complexes independent of
its Tlg2p binding [35], suggesting that SM proteins can
interact with their cognate SNAREs through distinct
mechanisms and at different stages in the SNARE assem-
bly/disassembly cycle. Consistent with the multiple modes
of binding, yeast Sec1p interacts with a fully assembled
exocytic SNARE complex [36]. Mammalian Munc18-1 pro-
tein, however, binds the t-SNARE Stx1 in a closed confor-
mation [37,38], in which an intramolecular interaction
between the N-terminal Habc domain of Stx1 and its
SNARE domain preclude its assembly into a SNARE
complex. Nevertheless, Munc18-1 stimulates SNARE-me-
diated fusion in vitro. It was proposed that Munc18-1
facilitates functional SNAREpin assembly by interacting
with the preassembled Stx1–SNAP25 t-SNARE subcom-
plex, which serves as a platform for VAMP2 binding [31].
The different modes of interaction between SM proteins
and SNAREs demonstrate the important regulatory roles
of SM proteins in SNAREpin assembly. Indeed, in vitro
binding assays suggested that the interaction of SM pro-
teins with SNAREs prevents the formation of non-physio-
logical SNARE complexes and stimulates specific SNARE
pairing [39]. For additional information on SM proteins
and their mechanisms of action, see these recent reviews
[40,41].
Tethering factors
Tethering factors are a large group of proteins or multiple-
protein complexes that link transport vesicles to their
cognate target membranes. As such, they regulate funda-
mental cellular processes and are implicated in various
diseases (Box 1). Tethering factors can be divided into two
major groups: homodimeric long coiled-coil proteins and
multisubunit tethering complexes (MTCs) [16,42–44].
Coiled-coil tethers are large, hydrophilic, dimeric proteins
comprising two globular heads connected by long coiled-
coil domains that can interact with vesicles over distances
of more than 200 nm [42]. MTCs, which contain three to ten
Bet1
Stx5
GS28
Ykt6
GS15
Stx5
GS28
Ykt6
Slt1
Stx18
Sec20
Sec22b
Vamp7
Stx7
VƟ1b
Stx8
Bet1
Stx5
GS27
Sec22b
ER
ERGIC
PM
cis-Golgi
Golgi stack
Vamp2
Stx1
SNAP-25
Vamp8
Stx4
SNAP-23
Recycling
endosome
TGN
MVB
Lysosome
Vamp2/3
Stx13
SNAP-25/29
Vamp8
Stx7
VƟ1b
Stx8
Early
endosome
Vamp4
Stx6
Stx16
VƟ1a
Munc18
Sly1
Vps45
Vps33
Vps33
Sly1
Sly1
B
TRENDS in Cell Biology
SynapƟc
vesicles
Secretory
granule
Figure 2. Soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes along the secretory and endocytic pathways in mammals. Schematic
summary of known mammalian SNARE complexes and their sites of action along the exocytic and endocytic pathways. Potential v-SNAREs are indicated in red; the Sec1/
Munc18 SM proteins for some SNARE complexes are marked in purple. Abbreviations: ER, endoplasmic reticulum; ERGIC, ER–Golgi intermediate compartment; MVBs,
multivesicular bodies; PM, plasma membrane; TGN, trans-Golgi network.
Review Trends in Cell Biology January 2014, Vol. 24, No. 1
37
subunits with an overall molecular weight of approximate-
ly 250–800 kDa, can interact with vesicles over much
shorter distances (up to 30 nm) [45].
Many long coiled-coil tethers including GM130, Golgin-
45, Golgin-97, Golgin-245, and p115 localize to the Golgi
apparatus and belong to the golgin family. The golgins
(approximately 20 members) are regulated by small
GTPases of the Rab and Arl families and function in
membrane–membrane and membrane–cytoskeleton teth-
ering at the Golgi apparatus [46]. Other long coiled-coil
tethers such as EEA1, Rabaptin-5, Rabenosyn-5, and
Rabip4 associate with endosomal compartments [44].
MTCs can be divided into two major classes: complexes
associated with tethering containing helical rods
(CATCHR) and Class C vacuolar protein-sorting (Vps)
complexes. The exocyst, DSL1, Conserved Oligomeric Gol-
gi (COG), and Golgi-associated retrograde protein (GARP)
(Figure 3A,C,D) belong to the CATCHR family, which
exhibits low sequence identity but conserved helical-bun-
dle structures [45]. Homotypic fusion and protein sorting
(HOPS) (Figure 3B) and class C core vacuole/endosome
tethering (CORVET) belong to the Class C Vps complexes
and regulate transport from the late Golgi to endosomal/
lysosomal compartments [47]. The multisubunit Transport
Protein Particle (TRAPP) complexes (I, II, and III) have
also been considered as tethering factors. However, in
contrast to other tethering factors that function as small
GTPase effectors, TRAPPs act as guanine nucleotide ex-
change factors (GEFs) for the Rab small GTPases [48].
TRAPPI, which is required for ER–Golgi transport, inter-
acts directly with vesicle coats, thereby coupling Rab acti-
vation with coat recognition and vesicular tethering
[49,50].
Tethering factors are associated with different intracel-
lular membranes via distinct mechanisms, including specif-
ic targeting motifs (such as the GRIP domain in several
golgins), phospholipid binding, and/or interactions with
small GTPases [46,51]. Most tethering factors are Rab or
Arl effectors that interact directly with small GTPases in
their GTP-bound form [51]. CORVET and EEA1 are Rab5
effectors, whereas HOPS is a Rab7 effector. Rab5 (Vps21p in
yeast) functions at the early endosome, whereas Rab7
(Ypt7p in yeast) functions at late endosomes and lysosomes
[52]. COG is a Rab1 (Ypt1p) and Rab6 (Ypt6p) effector
[53–56], whereas GARP interacts with small GTPases of
the Rab and Arl families (Ypt6p and Arl1p, respectively, in
Saccharomyces cerevisiae) [57]. The exocyst interacts with
the GTP-bound form ofthe Rab small GTPase Sec4p in yeast
and with the Rab11, Rab8, and Ral small GTPases in
mammals. The exocyst also interacts with Rho and Cdc42
small GTPases [58]. Comprehensive description of these
tethers and their structural features and subcellular distri-
bution has been published previously [42,45,51,59,60].
Although both MTCs and long coiled-coil tethers have
been implicated in vesicular tethering, coiled-coil tethers
are presumed to be involved in the initial, highly dynamic
stages of tethering, because they can form transient, re-
versible, and low-affinity interactions [44]. By contrast,
MTCs may regulate later events and be actively involved
in SNAREpin assembly. MTCs, which capture vesicles at
relatively short distances from their target membranes,
can interact simultaneously with different transport com-
ponents through their multiple subunits. These multiva-
lent interactions could integrate different trafficking
components and consequently couple vesicular tethering,
docking, and fusion events. The structural and functional
differences between long coiled-coil tethers and MTCs
suggest that coiled-coil tethers and MTCs could have
complementary roles and potentially cooperate during
vesicular tethering, docking, and fusion. Consistent with
this hypothesis, previous studies have shown direct inter-
actions between long coiled-coil tethers such as p115 or
Golgin 84 and the COG complex [61,62].
In addition to their established roles in vesicular cap-
turing and docking, tethering factors also positively regu-
late SNAREpin assembly by direct interaction with
components of the fusion machinery – SNAREs and SM
proteins [12–15]. These interactions could influence: (i) the
stability of certain SNAREs and/or their corresponding
complexes; (ii) the assembly of SNARE subcomplex inter-
mediates; and (iii) the speed and specificity of SNAREpin
assembly. A brief description of these interactions and
their physiological roles is given below.
Tethering factors interact with SNAREs and SM
proteins
Both coiled-coil tethers and MTCs interact directly
with different SNAREs. The coiled-coil tether p115, for
Box 1. Genetic mutations in MTCs, SNAREpin assembly,
and inherited diseases
In recent years, multiple mutations in genes encoding different
subunits of MTCs have been identified as the cause of severe
inherited diseases. For example, a single amino acid substitution
(L967N) in the carboxyl-terminal region of the Vps54 subunit of the
GARP complex was identified as the cause of defective spermiogen-
esis and a motor neuron disease in the ‘wobbler’ mouse [92].
Homozygous wobbler mice display unsteady (‘wobbly’) gait with
progressive muscle weakness, a phenotype resembling the motor
neuron degeneration observed in amyotrophic lateral sclerosis
(ALS). This phenotype is accompanied by the formation of
ubiquitinated TDP-43 inclusions [93]. It was proposed that the
L967N mutation reduces the level of functional GARP and conse-
quently its associated autophagic activity, thereby attenuating the
clearance of TDP-43 aggregates and facilitating motor neuron death
[57]. Likewise, point mutations within the human VPS33B gene, a
subunit of the HOPS complex, cause arthrogryposis, renal dysfunc-
tion, and cholestasis (ARC) syndrome (Online Mendelian Inheritance
in Man [OMIM] 208085). ARC is an autosomal-recessive, multi-
system disorder characterized by renal tubular abnormalities,
cholestasis, hypotonia, and platelet storage-pool deficiency [94]. It
was proposed that the VPS33B mutations cause abnormal assembly
of the entire HOPS complex and consequently impair SNAREpin
assembly and biogenesis and/or fusion of lysosome-related orga-
nelles. Strikingly, a direct influence on the assembly of Golgi
SNAREpins has been demonstrated in Cog7- and Cog8-deficient
fibroblasts derived from patients with congenital disorders of
glycosylation (CDGs) [95]. CDGs are a heterogeneous group of
inherited disorders characterized by pleiotropic glycosylation de-
fects [96]. In recent years, multiple mutations in the genes encoding
different COG subunits have been identified as the cause of CDG
[97–101]. These mutations affect the assembly of the COG complex,
thereby impairing intra-Golgi retrograde transport and conse-
quently the proper localization of Golgi glycosylation enzymes
[102]. Overall, these findings demonstrate the importance of
tethering factors for normal cell function and their critical physio-
logical roles.
Review Trends in Cell Biology January 2014, Vol. 24, No. 1
38
example, interacts with several Golgi SNAREs including
Stx5, GS28, GS15, Ykt6, rSec22, and GS27/membrin [13]
and the endosomal EEA1 tether interacts with Stx6 [63].
Different subunits of all MTCs except the TRAPP com-
plexes physically interact with various SNAREs both in
yeast and mammals (Table 1 in [42]).
Mechanistically, tethering factors can interact with
multiple structural motifs in SNAREs. For example, the
Tip20 and Sec39 subunits of the DSL1 complex, which
regulates Golgi-to-ER retrograde transport, interact di-
rectly with the N-terminal regulatory domains of the t-
SNAREs Use1 and Sec20 at the ER membrane (Figure 3C)
[64,65]. The Vps51 subunit of the GARP complex, which
regulates endosome-to-TGN retrograde transport, inter-
acts with the N-terminal Habc domain of Stx6 (Tlg1p in
yeast) [66,67] and the Vps53 and Vps54 subunits of GARP
interact with the SNARE motifs of the Golgi SNAREs Stx6,
Stx16, and Vamp4 (Figure 3A) [68,69]. Several subunits of
the octameric COG complex (Cog1–8), which regulates
intra-Golgi and endosome-to-Golgi retrograde transport,
also interact with the SNARE motifs of multiple Golgi
SNAREs (Figure 3A) [12,70,71]. These interactions with
SNARE motifs are mediated by N-terminal coiled-coil
domains found in the GARP and COG subunits [43,72].
Subunits of the HOPS complex, which is required for
multiple fusion events at the late endosome and homotypic
fusion of vacuoles in yeast [73–75], also interact directly
with SNAREs. The Vps33 subunit of the HOPS complex
binds the SNARE motifs of the t-SNAREs Vam3 and Vam7
as well as the SNARE motif of the v-SNARE Nyv1. The
Vps11, Vps16, and Vps18 subunits of HOPS bind the N-
terminal Habc domain of Vam3 and the PX domain of
Vam7 (Figure 3B) [76–78]. Vps33, which is also an integral
subunit of the CORVET complex, interacts directly with
endosomal SNAREs, including the yeast syntaxin Pep12p
[74]. Remarkably, Vps33 is an SM protein that functions as
Vps52
Vps53
Vps54
Vps51
Golgi
Stx5
GS28Ykt6
Stx16
VƟ1a
Vam
p4
Stx6
Sly1
p
Exocyst
PM
Sec9
Sso1/2
Sec4 (Rab)
Vesicle
Snc1/2
Sec1 Sec15
Sec10Sec6
Sec8 Exo84
Exo70
Sec5
Sec3
DSL1
Sec39
Dsl1
Tip20
ER
COP-I
Vesicle
Sec22
Ufe1
Use1
Sec20
HOPS
16
33 41
18
11
39
Rab/Ypt7
Vam3
Vam7
Vam3
Vam7
Nyv1
Stx6
Stx16
Vamp4
Rab/Ypt7
Vam
3
VƟ1
Vam
7
Nyv1
SNARE
abc
abc
COG
4
1
8
2
3
7
5
6
Vps45
GARP
(D)
(B)(A)
(C)
PX
t-SNAREs
TRENDS in Cell Biology
Figure 3. Multisubunit tethering complexes (MTCs) interact with soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) to couple vesicular
tethering with fusion. (A) Conserved Oligomeric Golgi (COG) and Golgi-associated retrograde protein (GARP) MTCs interact with multiple SNAREs on the Golgi
membranes. The eight subunits of the COG complex are organized in two structurally and functionally distinct lobes (light and dark blue) [103]. The Cog4, Cog6, and Cog7
subunits bind the SNARE domains of the indicated SNAREs. Cog4 also binds the Sec1/Munc18 (SM) proteins Sly1 and Vps45 [12,82]. The Vps52, Vps53, and Vps54 subunits
of GARP (purple) can be assembled into a core complex and localize to the Golgi in the absence of Vps51 [67]. The Vps51 subunit (pink) interacts with the N-terminal
regulatory Habc domain of Syntaxin (Stx)6 (Tlg1p in yeast) [66,67]. The N-terminal regions of human Vps53 and Vps54 bind to the SNARE motifs of Stx6, Stx16, and Vamp4
[68]. (B) The yeast homotypic fusion and protein-sorting (HOPS) complex via its Vps33 subunit (pink) interacts with the SNARE motifs (pink) of target membrane SNAREs (t-
SNAREs) Vam3 (Stx7 in mammals) and Vam7 (Stx8 in mammals) and the transport vesicle SNARE (v-SNARE) Nyv1. The Vps11, Vps16, and Vps18 subunits (green) interact
with the regulatory Habc and PX domains of the t-SNAREs Vam3 and Vam7 on the vacuole or lysosome membrane (green). Vps39 and Vps41 subunits (yellow) bind the
small Rab GTPase Ypt7 [52]. (C) A schematic presentation of the yeast DSL1 complex according to its crystal structure [64]. DSL1 interacts with the t-SNAREs Sec20 and
Use1 via its Tip20 and Sec39 subunits and induces t-SNARE gathering on the endoplasmic reticulum (ER) membrane. The Dsl1p subunit interacts with subunits of the COP-I
coat and tethers COP-I vesicles to the ER membranes [90]. (D) The yeast exocyst complex tethers secretory vesicles to the plasma membrane (PM). The Sec3 and Exo70
(pink) subunits interact with the PM via their positively charged residues, whereas the Sec15 subunit interacts with the Rab GTPase Sec4, which localizes on a secretory
vesicle. These interactions are required for vesicular tethering [104]. The interactions of the Sec6 subunit with the t-SNARE Sec9 (SNAP23 in mammals) and the SM protein
Sec1 and of the Sec3 subunit with Sso1/2 (Stx1 in mammals) regulate the assembly the Sec9–Sso1 t-SNARE complex that binds the v-SNARE Snc1/2 (VAMP3 in mammals)
as described in the text.
Review Trends in Cell Biology January 2014, Vol. 24, No. 1
39
one subunit of the HOPS and CORVET heterohexameric
complexes and mediates multiple interactions with
SNAREs [52]. The incorporation of an SM protein into
MTCs demonstrates the coordinated functions of tethers,
SM proteins, and SNAREs in regulating intracellular
membrane fusion. Consistent with this mode of action,
both the exocyst and the COG complex interact directly
with SM proteins and SNAREs.
The exocyst, an evolutionarily conserved complex im-
plicated in the tethering of secretory vesicles to the plasma
membrane, comprises eight subunits (Sec3, Sec5, Sec6,
Sec8, Sec10, Sec15, Exo70, and Exo84) [58]. The complex
is directly involved in the assembly of the Sec9–Sso1–Snc2
SNAREpin at the plasma membrane in yeast (Figure 3D).
Its Sec6 subunit binds the SM protein Sec1 and the t-
SNARE Sec9 via overlapping binding sites that prevent
the formation of a ternary Sec6–Sec9–Sec1 complex. Fur-
thermore, Sec6 interacts with Sec9 in the absence of other
exocyst subunits, but interacts with Sec1 in the presence of
exocyst subunits [79,80]. When Sec6 is found outside the
exocyst complex, its interaction with Sec9 inhibits the
interaction between Sec9 and its syntaxin partner Sso1
[79], thereby preventing the formation of a premature t-
SNARE complex at the plasma membrane. However, the
assembly of the exocyst complex leads to the release of Sec6
from Sec9, opening of the autoinhibitory domain of Sso1,
and concomitant recruitment of Sec1 for regulating vesic-
ular tethering and fusion events [81]. These observations
suggest that the interactions between subunits of MTCs,
SNAREs, and SM proteins are crucial for spatial and
temporal regulation of SNAREpin assembly.
Consistent with these observations, previous studies of
the COG complex suggest that the interaction between its
Cog4 subunit and the SM protein Sly1 is crucial for SNAR-
Epin assembly and membrane fusion at the Golgi mem-
branes. However, in this case a ternary complex between
Cog4, Sly1, and Stx5 can form due to distinct adjacent
binding sites for Sly1 and Stx5 at the N-terminal coiled-coil
domain of Cog4 [82]. The interaction of Cog4 with both Sly1
and Stx5 is required for the assembly of Stx5–GS28–Ykt6–
GS15 SNAREpin at the Golgi complex and consequently for
intra-Golgi retrograde transport [70,82]. Strikingly, Cog4
also interacts with the SM protein Vps45 and Stx16, which
regulate endosome-to-Golgi retrograde transport. These
interactions are also mediated by the N-terminal coiled-coil
domain of Cog4 and compete with the Sly1–Stx5 interac-
tions [12]. These observations suggest that the COG com-
plex employs similar mechanisms to regulate the assembly
of the two different Golgi SNAREpins (Stx5–GS28–Ykt6–
GS15 and Stx6–Stx16–Vti1a–VAMP4) and thereby may
coordinate their corresponding transport routes.
It seems that both COG and exocyst complexes are in-
volved in recruiting and/or orienting the SM proteins to a
partially assembled SNARE complex, and possibly stabiliz-
ing SM–SNARE interactions. These effects might enhance
direct interaction between the SM proteins and both the t-
and v-SNAREs of SNARE complex intermediates, thereby
facilitating the progression of fusion events [11].
The roles of tethering factor–SNARE interactions
Functionally, the interactions between tethering factors
and SNAREs can influence the assembly of SNARE
SNAREpin assembly
tSNAREs gathering
Vesicle
UncoaƟng
Vesiclular
fusion
SNARE complex
disassembly
Golgi
Sly1
GS15 VAMP4
Vps45
COG
Vesicle capture
UncoaƟng
Ykt6
GS28
Stx5
VƟ1a
Stx6
Stx16
TRENDS in Cell Biology
Figure 4. Roles of the Conserved Oligomeric Golgi (COG) complex in vesicular tethering and SNAREpin assembly at the Golgi apparatus. The COG complex via its different
subunits interacts with distinct t-SNAREs and SM proteins on the Golgi membranes and gathers cognate t-SNAREs and SM protein, thereby promoting the assembly of
specific ternary t-SNARE complexes. The COG complex also interacts with vesicle coats [56] and consequently promotes vesicular tethering. It is currently unknown at
which step COG dissociates from the assembled SNARE complex, but it possibly stabilizes the assembled complex [70]. The COG complex possibly employs similar
mechanisms to assemble two distinct fusogenic Golgi SNARE complexes (Stx5–GS28–Ykt6–GS15 and Stx6–Stx16–Vtia–VAMP4), thereby coordinating their associated
transport routes. We propose that the assembly of fusogenic SNARE complexes prevents promiscuous assembly of non-fusogenic t-SNARE interactions on the Golgi
membranes [12].
Review Trends in Cell Biology January 2014, Vol. 24, No. 1
40
complex intermediates, stabilize SNARE complex assem-
bly, and spatially and temporally organize fusion events at
specific membrane domains.
Many MTCs can interact simultaneously through their
different subunits with multiple SNAREs and gather spe-
cific SNAREs at specific membrane compartments. Sub-
units of the GARP, DSL1, and COG complexes, for
example, can simultaneously interact with specific t-
SNAREs and enhance t-SNARE subcomplex assembly
on the Golgi or ER membranes (Figure 3) [12,64,68,69].
The formation of t-SNARE subcomplexes represents an
intermediate step in SNAREpin assembly [7]. Hence,
SNARE gathering appears to be the major role of MTCs
in SNAREpin assembly (Figure 4). This gathering mediat-
ed by multivalent interactions not only facilitates the
assembly of fusogenic SNARE complexes that functionally
drive membrane fusion, but can also prevent the assembly
of non-fusogenic SNARE interactions in intact cells. Re-
cently it was shown that the Cog4 subunit positively
regulates the assembly of fusogenic SNARE complexes
at the Golgi complex, thereby preventing non-fusogenic
t-SNARE interactions [12]. The interaction of the Sec6
subunit of the exocyst with Sec9 also prevents the forma-
tion of the Sec9–Sso1 t-SNARE complex before the assem-
bly of the exocyst complex and recruitment of Sec1. This
role of MTCs may be a general mechanism that inhibits
promiscuous assembly of non-functional or premature
SNARE complexes in biological membranes. In vitro stud-
ies suggest that SM proteins may also prevent the assem-
bly of non-fusogenic SNAREs; the interaction of Sed5p
(Stx5 in mammals) with Sly1p, for example, prevents its
interactions with non-physiological SNAREs [39].
In addition to SNARE gathering, tethering factors can
stabilize SNARE proteins or SNARE complexes and can
thereby influence SNAREpin assembly. The Cog6 subunit,
for example, interacts directly with and stabilizes Stx6.
Depletion of theCog6 subunit leads to abnormal proteasomal
degradation of Stx6, thus impairing the assembly of Stx6–
Stx16–Vti1a–VAMP4 SNARE complex and consequently
endosome-to-Golgiretrograde trafficking [71]. Abnormal pro-
teasomal degradation of the Golgi SNAREs GS28 and GS15
wasalsoobservedincellsdepletedofotherCOGsubunits[83]
anda directinteractionbetween GS28 andCog4 was recently
observed [12]. These direct interactions between MTCs and
SNAREs might induce conformational changes in the
SNAREs that could protect them from degradation as was
previously proposed for SM proteins. Sly1, for example,
directly interacts with the ER t-SNARE Ufe1p, a yeast
homolog of Stx18, and protects it from ERAD degradation
[84],whereasVps45pinteractswithTlg2p,theyeasthomolog
of Stx16, and protects it from proteasomal degradation [85].
The HOPS complex employs a different mechanism to
stabilize trans-SNARE complexes: it inhibits their disas-
sembly by Sec17p/Sec18p (SNAP/NSF) [86]. The HOPS
complex has affinity for multiple vacuolar SNAREs and
can facilitate the formation of trans-SNARE complexes. Its
ability to interact with both t- and v-SNAREs (Figure 3B)
possibly inhibits the access of Sec17p and/or Sec18p and
consequently SNAREpin disassembly [87].
As discussed, tethers can induce SNARE gathering
through multivalent interactions, stabilize SNAREs or
SNARE complexes, or activate SNARE complex assembly
possibly through interaction with SM proteins [11,40].
However, it appears that SNARE–tether interactions have
bidirectional effects and, in some cases, the SNAREs influ-
ence the localization of the tethers. SNAREs, for instance,
can enhance the binding of HOPS to liposomes and possi-
bly stabilize the interaction of HOPS with the vacuole [88].
Likewise, the interaction of the DSL1 complex with ER
SNAREs is possibly required for its membrane association,
because DSL1 does not interact with the Rab small
GTPases that generally mediate membrane recruitment
[64,89]. Strikingly, DSL1 also interacts directly with COP-
I subunits [90] and HOPS interacts with the Apl5 subunit
of the AP-3 coat [91]. These interactions with both coats
and SNAREs are possibly required for coupling tethering
with fusion events.
Concluding remarks
Studies over the past few years strongly suggest that
tethering factors do not simply bridge two membrane
compartments, but rather are actively involved in fusion
and influence fusion specificity, efficiency, and speed. A
network of protein–protein interactions between tethers,
SNAREs, and/or SM proteins is the molecular basis for
their critical roles in SNAREpin assembly and conse-
quently membrane fusion. The ability of tethers to inter-
act with multiple components of the transport machinery,
either simultaneously or sequentially, enables them spa-
tially to regulate fusion events at specific membrane
domains. Multiple tethering factors exist in eukaryotic
cells; some regulate similar transport routes and even the
assembly of the same SNARE complexes. It remains un-
clear how these tethers function coordinately and why
there are so many factors with potentially overlapping
functions. For example, both GARP and COG regulate the
assembly of the Stx6–Stx16–Vti1a–VAMP4 SNARE com-
plex and consequently endosome-to-TGN retrograde
transport. In light of the described studies, it could be
that different tethers regulate the assembly of similar
SNARE complexes but concomitantly prevent the assem-
bly of distinct SNARE complexes or vice versa. This might
depend on the temporal localization of the tethering fac-
tors and their ability to interact with other cellular
SNAREs. Other aspects including the affinity of tethers
to their interacting SNAREs, the avidity of binding, or
even post-translational modifications in tethers and/or
SNAREs can influence SNAREpin assembly and/or its
disassembly. Collectively, these different aspects could
dynamically influence SNAREpin assembly and tempo-
rally and spatially control membrane-fusion events. Fur-
ther in vitro and in vivo studies on tether–SNARE–SM
interactions will shed light on the molecular mechanisms
that regulate vesicular fusion.
Acknowledgments
S.L. is the incumbent of the Joyce and Ben B. Eisenberg Chair of
Molecular Biology and Cancer Research. This work was supported by the
Binational Science Foundation (BSF), Grant No. 2011404.
References
1 Chen, Y.A. and Scheller, R.H. (2001) SNARE-mediated membrane
fusion. Nat. Rev. Mol. Cell Biol. 2, 98–106
Review Trends in Cell Biology January 2014, Vol. 24, No. 1
41
2 Hanson, P.I. et al. (1997) Neurotransmitter release – four years of
SNARE complexes. Curr. Opin. Neurobiol. 7, 310–315
3 Sollner, T. et al. (1993) SNAP receptors implicated in vesicle targeting
and fusion. Nature 362, 318–324
4 Jahn, R. et al. (2003) Membrane fusion. Cell 112, 519–533
5 Sollner, T.H. (2003) Regulated exocytosis and SNARE function
(review). Mol. Membr. Biol. 20, 209–220
6 Hong, W. (2005) SNAREs and traffic. Biochim. Biophys. Acta 1744,
120–144
7 Malsam, J. et al. (2008) Membrane fusion: SNAREs and regulation.
Cell. Mol. Life Sci. 65, 2814–2832
8 Parlati, F. et al. (2002) Distinct SNARE complexes mediating
membrane fusion in Golgi transport based on combinatorial
specificity. Proc. Natl. Acad. Sci. U.S.A. 99, 5424–5429
9 Parlati, F. et al. (2000) Topological restriction of SNARE-dependent
membrane fusion. Nature 407, 194–198
10 Sudhof, T.C. and Rothman, J.E. (2009) Membrane fusion: grappling
with SNARE and SM proteins. Science 323, 474–477
11 Shen, J. et al. (2007) Selective activation of cognate SNAREpins by
Sec1/Munc18 proteins. Cell 128, 183–195
12 Laufman, O. et al. (2013) The COG complex interacts with multiple
Golgi SNAREs and enhances fusogenic SNARE complexes assembly.
J. Cell Sci. 126, 1506–1516
13 Shorter, J. et al. (2002) Sequential tethering of Golgins and catalysis
of SNAREpin assembly by the vesicle-tethering protein p115. J. Cell
Biol. 157, 45–62
14 Kraynack, B.A. et al. (2005) Dsl1p, Tip20p, and the novel Dsl3(Sec39)
protein are required for the stability of the Q/t-SNARE complex at the
endoplasmic reticulum in yeast. Mol. Biol. Cell 16, 3963–3977
15 Sztul, E. and Lupashin, V. (2006) Role of tethering factors in secretory
membrane traffic. Am. J. Physiol. Cell Physiol. 290, C11–C26
16 Lupashin, V. and Sztul, E. (2005) Golgi tethering factors. Biochim.
Biophys. Acta 1744, 325–339
17 Cai, H. et al. (2007) Coats, tethers, Rabs, and SNAREs work together
to mediate the intracellular destination of a transport vesicle. Dev.
Cell 12, 671–682
18 Jahn, R. and Scheller, R.H. (2006) SNAREs – engines for membrane
fusion. Nat. Rev. Mol. Cell Biol. 7, 631–643
19 Holt, M. et al. (2006) Identification of SNAP-47, a novel Qbc-SNARE
with ubiquitous expression. J. Biol. Chem. 281, 17076–17083
20 Weimbs, T. et al. (1997) A conserved domain is present in different
families of vesicular fusion proteins: a new superfamily. Proc. Natl.
Acad. Sci. U.S.A. 94, 3046–3051
21 Sutton, R.B. et al. (1998) Crystal structure of a SNARE complex
involved in synaptic exocytosis at 2.4 A resolution. Nature 395,
347–353
22 Fasshauer, D. et al. (1998) Conserved structural features of the
synaptic fusion complex: SNARE proteins reclassified as Q- and R-
SNAREs. Proc. Natl. Acad. Sci. U.S.A. 95, 15781–15786
23 Fukasawa, M. et al. (2004) Localization and activity of the SNARE
Ykt6 determined by its regulatory domain and palmitoylation. Proc.
Natl. Acad. Sci. U.S.A. 101, 4815–4820
24 Misura, K.M. et al. (2000) Three-dimensional structure of the
neuronal-Sec1–syntaxin 1a complex. Nature 404, 355–362
25 Schafer, I.B. et al. (2012) The binding of Varp to VAMP7 traps VAMP7
in a closed, fusogenically inactive conformation. Nat. Struct. Mol. Biol.
19, 1300–1309
26 Zeng, Q. et al. (2003) The cytoplasmic domain of Vamp4 and Vamp5 is
responsible for their correct subcellular targeting: the N-terminal
extension of VAMP4 contains a dominant autonomous targeting
signal for the trans-Golgi network. J. Biol. Chem. 278, 23046–23054
27 Tran, T.H. et al. (2007) VAMP4 cycles from the cell surface to the
trans-Golgi network via sorting and recycling endosomes. J. Cell Sci.
120, 1028–1041
28 Malsam, J. and Sollner, T.H. (2013) Organization of SNAREs within
the Golgi stack. Cold Spring Harb. Perspect. Biol. 3, a005249
29 Gallwitz, D. and Jahn, R. (2003) The riddle of the Sec1/Munc-18
proteins – new twists added to their interactions with SNAREs.
Trends Biochem. Sci. 28, 113–116
30 Scott, B.L. et al. (2004) Sec1p directly stimulates SNARE-mediated
membrane fusion in vitro. J. Cell Biol. 167, 75–85
31 Rodkey, T.L. et al. (2008) Munc18a scaffolds SNARE assembly to
promote membrane fusion. Mol. Biol. Cell 19, 5422–5434
32 Pieren, M. et al. (2010) The SM protein Vps33 and the t-SNARE
H(abc) domain promote fusion pore opening. Nat. Struct. Mol. Biol. 17,
710–717
33 Yamaguchi, T. et al. (2002) Sly1 binds to Golgi and ER syntaxins via a
conserved N-terminal peptide motif. Dev. Cell 2, 295–305
34 Dulubova, I. et al. (2002) How Tlg2p/syntaxin 16 ‘snares’ Vps45.
EMBO J. 21, 3620–3631
35 Carpp, L.N. et al. (2006) The Sec1p/Munc18 protein Vps45p binds its
cognate SNARE proteins via two distinct modes. J. Cell Biol. 173,
927–936
36 Carr, C.M. et al. (1999) Sec1p binds to SNARE complexes and
concentrates at sites of secretion. J. Cell Biol. 146, 333–344
37 Yang, B. et al. (2000) nSec1 binds a closed conformation of syntaxin1A.
J. Cell Biol. 148, 247–252
38 Dulubova, I. et al. (1999) A conformational switch in syntaxin during
exocytosis: role of Munc18. EMBO J. 18, 4372–4382
39 Peng, R. and Gallwitz, D. (2002) Sly1 protein bound to Golgi syntaxin
Sed5p allows assembly and contributes to specificity of SNARE fusion
complexes. J. Cell Biol. 157, 645–655
40 Rizo, J. and Sudhof, T.C. (2012) The membrane fusion enigma:
SNAREs, Sec1/Munc18 proteins, and their accomplices – guilty as
charged? Annu. Rev. Cell Dev. Biol. 28, 279–308
41 Jahn, R. and Fasshauer, D. (2012) Molecular machines governing
exocytosis of synaptic vesicles. Nature 490, 201–207
42 Brocker, C. et al. (2010) Multisubunit tethering complexes and their
role in membrane fusion. Curr. Biol. 20, R943–R952
43 Whyte, J.R. and Munro, S. (2002) Vesicle tethering complexes in
membrane traffic. J. Cell Sci. 115, 2627–2637
44 Gillingham, A.K. and Munro, S. (2003) Long coiled-coil proteins and
membrane traffic. Biochim. Biophys. Acta 1641, 71–85
45 Yu, I.M. and Hughson, F.M. (2010) Tethering factors as organizers of
intracellular vesicular traffic. Annu. Rev. Cell Dev. Biol. 26, 137–156
46 Barr, F.A. and Short, B. (2003) Golgins in the structure and dynamics
of the Golgi apparatus. Curr. Opin. Cell Biol. 15, 405–413
47 Peterson, M.R. and Emr, S.D. (2001) The class C Vps complex
functions at multiple stages of the vacuolar transport pathway.
Traffic 2, 476–486
48 Jones, S. et al. (2000) The TRAPP complex is a nucleotide exchanger
for Ypt1 and Ypt31/32. Mol. Biol. Cell 11, 4403–4411
49 Sacher, M. et al. (2001) TRAPP I implicated in the specificity of
tethering in ER-to-Golgi transport. Mol. Cell 7, 433–442
50 Cai, H. et al. (2007) TRAPPI tethers COPII vesicles by binding the
coat subunit Sec23. Nature 445, 941–944
51 Angers, C.G. and Merz, A.J. (2011) New links between vesicle coats
and Rab-mediated vesicle targeting. Semin. Cell Dev. Biol. 22, 18–26
52 Balderhaar, H.J. and Ungermann, C. (2013) CORVET and HOPS
tethering complexes – coordinators of endosome and lysosome fusion.
J. Cell Sci. 126, 1307–1316
53 VanRheenen, S.M. et al. (1998) Sec35p, a novel peripheral membrane
protein, is required for ER to Golgi vesicle docking. J. Cell Biol. 141,
1107–1119
54 VanRheenen, S.M. et al. (1999) Sec34p, a protein required for vesicle
tethering to the yeast Golgi apparatus, is in a complex with Sec35p. J.
Cell Biol. 147, 729–742
55 Miller, V.J. et al. (2013) Molecular insights into vesicle tethering at
the Golgi by the conserved oligomeric Golgi (COG) complex and the
golgin TATA element modulatory factor (TMF). J. Biol. Chem. 288,
4229–4240
56 Suvorova, E.S. et al. (2002) The Sec34/Sec35p complex, a Ypt1p
effector required for retrograde intra-Golgi trafficking, interacts
with Golgi SNAREs and COPI vesicle coat proteins. J. Cell Biol.
157, 631–643
57 Bonifacino, J.S. and Hierro, A. (2011) Transport according to GARP:
receiving retrograde cargo at the trans-Golgi network. Trends Cell
Biol. 21, 159–167
58 Heider, M.R. and Munson, M. (2012) Exorcising the exocyst complex.
Traffic 13, 898–907
59 Oka, T. and Krieger, M. (2005) Multi-component protein complexes
and Golgi membrane trafficking. J. Biochem. 137, 109–114
60 Waters, M.G. and Pfeffer, S.R. (1999) Membrane tethering in
intracellular transport. Curr. Opin. Cell Biol. 11, 453–459
61 Sohda, M. et al. (2007) The interaction of two tethering factors, p115
and COG complex, is required for Golgi integrity. Traffic 8, 270–284
Review Trends in Cell Biology January 2014, Vol. 24, No. 1
42
62 Sohda, M. et al. (2010) Interaction of Golgin-84 with the COG complex
mediates the intra-Golgi retrograde transport. Traffic 11, 1552–1566
63 Simonsen, A. et al. (1999) The Rab5 effector EEA1 interacts directly
with Syntaxin-6. J. Biol. Chem. 274, 28857–28860
64 Ren, Y. et al. (2009) A structure-based mechanism for vesicle capture
by the multisubunit tethering complex Dsl1. Cell 139, 1119–1129
65 Tripathi, A. et al. (2009) Structural characterization of Tip20p and
Dsl1p, subunits of the Dsl1p vesicle tethering complex. Nat. Struct.
Mol. Biol. 16, 114–123
66 Conibear, E. et al. (2003) Vps51p mediates the association of the
GARP (Vps52/53/54) complex with the late Golgi t-SNARE Tlg1p.
Mol. Biol. Cell 14, 1610–1623
67 Siniossoglou, S. and Pelham, H.R. (2002) Vps51p links the VFT
complex to the SNARE Tlg1p. J. Biol. Chem. 277, 48318–48324
68 Perez-Victoria, F.J. and Bonifacino, J.S. (2009) Dual roles of
the mammalian GARP complex in tethering and SNARE
complex assembly at the trans-Golgi network. Mol. Cell. Biol.
29, 5251–5263
69 Perez-Victoria, F.J. et al. (2010) Ang2/fat-free is a conserved subunit of
the Golgi-associated retrograde protein complex. Mol. Biol. Cell 21,
3386–3395
70 Shestakova, A. et al. (2007) Interaction of the conserved oligomeric
Golgi complex with t-SNARE Syntaxin5a/Sed5 enhances intra-Golgi
SNARE complex stability. J. Cell Biol. 179, 1179–1192
71 Laufman, O. et al. (2011) The COG complex interacts directly with
Syntaxin 6 and positively regulates endosome-to-TGN retrograde
transport. J. Cell Biol. 194, 459–472
72 Whyte, J.R. and Munro, S. (2001) The Sec34/35 Golgi transport
complex is related to the exocyst, defining a family of complexes
involved in multiple steps of membrane traffic. Dev. Cell 1, 527–537
73 Rieder, S.E. and Emr, S.D. (1997) A novel RING finger protein
complex essential for a late step in protein transport to the yeast
vacuole. Mol. Biol. Cell 8, 2307–2327
74 Subramanian, S. et al. (2004) The Sec1/Munc18 protein, Vps33p,
functions at the endosome and the vacuole of Saccharomyces
cerevisiae. Mol. Biol. Cell 15, 2593–2605
75 Mima, J. et al. (2008) Reconstituted membrane fusion requires
regulatory lipids, SNAREs and synergistic SNARE chaperones.
EMBO J. 27, 2031–2042
76 Stroupe, C. et al. (2006) Purification of active HOPS complex reveals
its affinities for phosphoinositides and the SNARE Vam7p. EMBO J.
25, 1579–1589
77 Kramer, L. and Ungermann, C. (2011) HOPS drives vacuole fusion by
binding the vacuolar SNARE complex and the Vam7 PX domain via
two distinct sites. Mol. Biol. Cell 22, 2601–2611
78 Lobingier, B.T. and Merz, A.J. (2012) Sec1/Munc18 protein Vps33
binds to SNARE domains and the quaternary SNARE complex. Mol.
Biol. Cell 23, 4611–4622
79 Sivaram, M.V. et al. (2005) Dimerization of the exocyst protein Sec6p
and its interaction with the t-SNARE Sec9p. Biochemistry 44, 6302–
6311
80 Morgera, F. et al. (2012) Regulation of exocytosis by the exocyst
subunit Sec6 and the SM protein Sec1. Mol. Biol. Cell 23, 337–346
81 Munson, M. et al. (2000) Interactions within the yeast t-SNARE Sso1p
that control SNARE complex assembly. Nat. Struct. Biol. 7, 894–902
82 Laufman, O. et al. (2009) Direct interaction between the COG complex
and the SM protein, Sly1, is required for Golgi SNARE pairing. EMBO
J. 28, 2006–2017
83 Oka, T. et al. (2004) The COG and COPI complexes interact to control
the abundance of GEARs, a subset of Golgi integral membrane
proteins. Mol. Biol. Cell 15, 2423–2435
84 Braun, S. and Jentsch, S. (2007) SM-protein-controlled ER-associated
degradation discriminates between different SNAREs. EMBO Rep. 8,
1176–1182
85 Bryant, N.J. and James, D.E. (2001) Vps45p stabilizes the syntaxin
homologue Tlg2p and positively regulates SNARE complex formation.
EMBO J. 20, 3380–3388
86 Xu, H. et al. (2010) HOPS prevents the disassembly of trans-SNARE
complexes by Sec17p/Sec18p during membrane fusion. EMBO J. 29,
1948–1960
87 Collins, K.M. et al. (2005) Sec17p and HOPS, in distinct SNARE
complexes, mediate SNARE complex disruption or assembly for
fusion. EMBO J. 24, 1775–1786
88 Stroupe, C. et al. (2009) Minimal membrane docking requirements
revealed by reconstitution of Rab GTPase-dependent membrane
fusion from purified components. Proc. Natl. Acad. Sci. U.S.A. 106,
17626–17633
89 Meiringer, C.T. et al. (2011) The Dsl1 protein tethering complex is a
resident endoplasmic reticulum complex, which interacts with five
soluble NSF (N-ethylmaleimide-sensitive factor) attachment protein
receptors (SNAREs): implications for fusion and fusion regulation. J.
Biol. Chem. 286, 25039–25046
90 Andag, U. et al. (2001) The coatomer-interacting protein Dsl1p is
required for Golgi-to-endoplasmic reticulum retrieval in yeast. J. Biol.
Chem. 276, 39150–39160
91 Angers, C.G. and Merz, A.J. (2009) HOPS interacts with Apl5 at the
vacuole membrane and is required for consumption of AP-3 transport
vesicles. Mol. Biol. Cell 20, 4563–4574
92 Perez-Victoria, F.J. et al. (2010) Structural basis for the wobbler
mouse neurodegenerative disorder caused by mutation in the
Vps54 subunit of the GARP complex. Proc. Natl. Acad. Sci. U.S.A.
107, 12860–12865
93 Dennis, J.S. and Citron, B.A. (2009) Wobbler mice modeling motor
neuron disease display elevated transactive response DNA binding
protein. Neuroscience 158, 745–750
94 Gissen, P. et al. (2004) Mutations in VPS33B, encoding a regulator of
SNARE-dependent membrane fusion, cause arthrogryposis–renal
dysfunction–cholestasis (ARC) syndrome. Nat. Genet. 36, 400–404
95 Laufman, O. et al. (2013) Deficiency of the Cog8 subunit in normal and
CDG-derived cells impairs the assembly of the COG and Golgi SNARE
complexes. Traffic 14, 1065–1077
96 Jaeken, J. and Matthijs, G. (2007) Congenital disorders of
glycosylation: a rapidly expanding disease family. Annu. Rev.
Genomics Hum. Genet. 8, 261–278
97 Wu, X. et al. (2004) Mutation of the COG complex subunit gene COG7
causes a lethal congenital disorder. Nat. Med. 10, 518–523
98 Kranz, C. et al. (2007) COG8 deficiency causes new congenital
disorder of glycosylation type IIh. Hum. Mol. Genet. 16, 731–741
99 Foulquier, F. et al. (2007) A new inborn error of glycosylation due to a
Cog8 deficiency reveals a critical role for the Cog1–Cog8 interaction in
COG complex formation. Hum. Mol. Genet. 16, 717–730
100 Foulquier, F. et al. (2006) Conserved oligomeric Golgi complex subunit
1 deficiency reveals a previously uncharacterized congenital disorder
of glycosylation type II. Proc. Natl. Acad. Sci. U.S.A. 103, 3764–3769
101 Foulquier, F. (2009) COG defects, birth and rise! Biochim. Biophys.
Acta 1792, 896–902
102 Freeze, H.H. and Ng, B.G. (2011) Golgi glycosylation and human
inherited diseases. Cold Spring Harb. Perspect. Biol. 3, a005371
103 Ungar, D. et al. (2006) Retrograde transport on the COG railway.
Trends Cell Biol. 16, 113–120
104 He, B. and Guo, W. (2009) The exocyst complex in polarized
exocytosis. Curr. Opin. Cell Biol. 21, 537–542
Review Trends in Cell Biology January 2014, Vol. 24, No. 1
43

More Related Content

What's hot

Retinoblastoma family proteins New players in DNA repair by non-homologous
Retinoblastoma family proteins New players in DNA repair by non-homologousRetinoblastoma family proteins New players in DNA repair by non-homologous
Retinoblastoma family proteins New players in DNA repair by non-homologousMaciej Luczynski
 
An expanding arsenal of experimental methods yields an explosion of insights ...
An expanding arsenal of experimental methods yields an explosion of insights ...An expanding arsenal of experimental methods yields an explosion of insights ...
An expanding arsenal of experimental methods yields an explosion of insights ...V P
 
mechanism based inactivation by quinoneimine reactive metabolite
mechanism based inactivation by quinoneimine reactive metabolitemechanism based inactivation by quinoneimine reactive metabolite
mechanism based inactivation by quinoneimine reactive metaboliteSurendra Jatav
 
Perales and Bentley MolCell 2009
Perales and Bentley MolCell 2009Perales and Bentley MolCell 2009
Perales and Bentley MolCell 2009Roberto Perales PhD
 
Histone demethylase and it srole in cell biology review
Histone demethylase and it srole in cell biology reviewHistone demethylase and it srole in cell biology review
Histone demethylase and it srole in cell biology reviewChristopher Wynder
 
The mechanism of protein folding
The mechanism of protein foldingThe mechanism of protein folding
The mechanism of protein foldingPrasanthperceptron
 

What's hot (9)

Retinoblastoma family proteins New players in DNA repair by non-homologous
Retinoblastoma family proteins New players in DNA repair by non-homologousRetinoblastoma family proteins New players in DNA repair by non-homologous
Retinoblastoma family proteins New players in DNA repair by non-homologous
 
12 miller chap.3 lecture
12 miller chap.3 lecture12 miller chap.3 lecture
12 miller chap.3 lecture
 
solman_maja
solman_majasolman_maja
solman_maja
 
An expanding arsenal of experimental methods yields an explosion of insights ...
An expanding arsenal of experimental methods yields an explosion of insights ...An expanding arsenal of experimental methods yields an explosion of insights ...
An expanding arsenal of experimental methods yields an explosion of insights ...
 
mechanism based inactivation by quinoneimine reactive metabolite
mechanism based inactivation by quinoneimine reactive metabolitemechanism based inactivation by quinoneimine reactive metabolite
mechanism based inactivation by quinoneimine reactive metabolite
 
Perales and Bentley MolCell 2009
Perales and Bentley MolCell 2009Perales and Bentley MolCell 2009
Perales and Bentley MolCell 2009
 
Protein folding
Protein foldingProtein folding
Protein folding
 
Histone demethylase and it srole in cell biology review
Histone demethylase and it srole in cell biology reviewHistone demethylase and it srole in cell biology review
Histone demethylase and it srole in cell biology review
 
The mechanism of protein folding
The mechanism of protein foldingThe mechanism of protein folding
The mechanism of protein folding
 

Similar to Tethering the assembly of SNARE complexes by Sima Lev and WonJin Hong

Replisome-Cohesin Interfacing A Molecular Perspective.pdf
Replisome-Cohesin Interfacing A Molecular Perspective.pdfReplisome-Cohesin Interfacing A Molecular Perspective.pdf
Replisome-Cohesin Interfacing A Molecular Perspective.pdfAtiaGohar1
 
SPERM-EGG INTERACTIONS AND MAMMALIAN FERTILIZATION
SPERM-EGG INTERACTIONS AND MAMMALIAN FERTILIZATIONSPERM-EGG INTERACTIONS AND MAMMALIAN FERTILIZATION
SPERM-EGG INTERACTIONS AND MAMMALIAN FERTILIZATIONEarthjournal Publisher
 
Describe briefly the means by which the U2AF splicing factor was ide.pdf
Describe briefly the means by which the U2AF splicing factor was ide.pdfDescribe briefly the means by which the U2AF splicing factor was ide.pdf
Describe briefly the means by which the U2AF splicing factor was ide.pdfsktambifortune
 
Google Scholar CSD-cited 17700_PM4840_ v3
Google Scholar CSD-cited 17700_PM4840_ v3Google Scholar CSD-cited 17700_PM4840_ v3
Google Scholar CSD-cited 17700_PM4840_ v3Shengwen Calvin Li, PhD
 
Google Scholar CSD-cited 17700_PM4840_ v3
Google Scholar CSD-cited 17700_PM4840_ v3Google Scholar CSD-cited 17700_PM4840_ v3
Google Scholar CSD-cited 17700_PM4840_ v3Shengwen Calvin Li, PhD
 
Neuromuscular Junction Formation
Neuromuscular Junction FormationNeuromuscular Junction Formation
Neuromuscular Junction FormationQilong Wu
 
ConSurf_an_algorithmic_tool_for_the_iden
ConSurf_an_algorithmic_tool_for_the_idenConSurf_an_algorithmic_tool_for_the_iden
ConSurf_an_algorithmic_tool_for_the_idenRony Armon
 
Encyclopedia of biophysics
Encyclopedia of biophysicsEncyclopedia of biophysics
Encyclopedia of biophysicsSpringer
 
RNA_splicing_ppt.ppt
RNA_splicing_ppt.pptRNA_splicing_ppt.ppt
RNA_splicing_ppt.pptGizatAlmaw1
 
13-miller-chap-8-lecture (1).ppt
13-miller-chap-8-lecture (1).ppt13-miller-chap-8-lecture (1).ppt
13-miller-chap-8-lecture (1).pptMasihUllah11
 
Superfamilia-de-las-inmunoglobulinas.pdf
Superfamilia-de-las-inmunoglobulinas.pdfSuperfamilia-de-las-inmunoglobulinas.pdf
Superfamilia-de-las-inmunoglobulinas.pdfEdgarRamrezMartnez
 

Similar to Tethering the assembly of SNARE complexes by Sima Lev and WonJin Hong (20)

Cognitive neuroscience active zone
Cognitive neuroscience   active zoneCognitive neuroscience   active zone
Cognitive neuroscience active zone
 
Replisome-Cohesin Interfacing A Molecular Perspective.pdf
Replisome-Cohesin Interfacing A Molecular Perspective.pdfReplisome-Cohesin Interfacing A Molecular Perspective.pdf
Replisome-Cohesin Interfacing A Molecular Perspective.pdf
 
Intramembrane Proteases
Intramembrane ProteasesIntramembrane Proteases
Intramembrane Proteases
 
SPERM-EGG INTERACTIONS AND MAMMALIAN FERTILIZATION
SPERM-EGG INTERACTIONS AND MAMMALIAN FERTILIZATIONSPERM-EGG INTERACTIONS AND MAMMALIAN FERTILIZATION
SPERM-EGG INTERACTIONS AND MAMMALIAN FERTILIZATION
 
Smith_COSB_2015
Smith_COSB_2015Smith_COSB_2015
Smith_COSB_2015
 
Describe briefly the means by which the U2AF splicing factor was ide.pdf
Describe briefly the means by which the U2AF splicing factor was ide.pdfDescribe briefly the means by which the U2AF splicing factor was ide.pdf
Describe briefly the means by which the U2AF splicing factor was ide.pdf
 
Paper 1-Senthil
Paper 1-SenthilPaper 1-Senthil
Paper 1-Senthil
 
EMBO
EMBOEMBO
EMBO
 
Google Scholar CSD-cited 17700_PM4840_ v3
Google Scholar CSD-cited 17700_PM4840_ v3Google Scholar CSD-cited 17700_PM4840_ v3
Google Scholar CSD-cited 17700_PM4840_ v3
 
Google Scholar CSD-cited 17700_PM4840_ v3
Google Scholar CSD-cited 17700_PM4840_ v3Google Scholar CSD-cited 17700_PM4840_ v3
Google Scholar CSD-cited 17700_PM4840_ v3
 
Neuromuscular Junction Formation
Neuromuscular Junction FormationNeuromuscular Junction Formation
Neuromuscular Junction Formation
 
ConSurf_an_algorithmic_tool_for_the_iden
ConSurf_an_algorithmic_tool_for_the_idenConSurf_an_algorithmic_tool_for_the_iden
ConSurf_an_algorithmic_tool_for_the_iden
 
Encyclopedia of biophysics
Encyclopedia of biophysicsEncyclopedia of biophysics
Encyclopedia of biophysics
 
RNA_splicing_ppt.ppt
RNA_splicing_ppt.pptRNA_splicing_ppt.ppt
RNA_splicing_ppt.ppt
 
13-miller-chap-8-lecture (1).ppt
13-miller-chap-8-lecture (1).ppt13-miller-chap-8-lecture (1).ppt
13-miller-chap-8-lecture (1).ppt
 
IAS Final Report
IAS Final ReportIAS Final Report
IAS Final Report
 
srep34405
srep34405srep34405
srep34405
 
Cognitive neuroscience synapses
Cognitive neuroscience   synapsesCognitive neuroscience   synapses
Cognitive neuroscience synapses
 
Snrk in development and disease
Snrk in development and diseaseSnrk in development and disease
Snrk in development and disease
 
Superfamilia-de-las-inmunoglobulinas.pdf
Superfamilia-de-las-inmunoglobulinas.pdfSuperfamilia-de-las-inmunoglobulinas.pdf
Superfamilia-de-las-inmunoglobulinas.pdf
 

More from Sima Lev

Sima Lev The AXL PYK2 PKC axis as a nexus of stemness circuits in TNBC
Sima Lev  The AXL PYK2 PKC axis as a nexus of stemness circuits in TNBCSima Lev  The AXL PYK2 PKC axis as a nexus of stemness circuits in TNBC
Sima Lev The AXL PYK2 PKC axis as a nexus of stemness circuits in TNBCSima Lev
 
Sima Lev - Virtual Training
Sima Lev - Virtual TrainingSima Lev - Virtual Training
Sima Lev - Virtual TrainingSima Lev
 
Sima Lev - Edinburgh Breast Cancer Symposium
Sima Lev - Edinburgh Breast Cancer SymposiumSima Lev - Edinburgh Breast Cancer Symposium
Sima Lev - Edinburgh Breast Cancer SymposiumSima Lev
 
Animal Lectin Galectin-8 by Hadas Shatz-Azoulay, Yaron Vinik, Roi Isaac, Ulri...
Animal Lectin Galectin-8 by Hadas Shatz-Azoulay, Yaron Vinik, Roi Isaac, Ulri...Animal Lectin Galectin-8 by Hadas Shatz-Azoulay, Yaron Vinik, Roi Isaac, Ulri...
Animal Lectin Galectin-8 by Hadas Shatz-Azoulay, Yaron Vinik, Roi Isaac, Ulri...Sima Lev
 
Sima Lev: Non-Vesicular Lipid Transport by Lipid-Transfer Proteins and Beyond
Sima Lev: Non-Vesicular Lipid Transport by Lipid-Transfer Proteins and BeyondSima Lev: Non-Vesicular Lipid Transport by Lipid-Transfer Proteins and Beyond
Sima Lev: Non-Vesicular Lipid Transport by Lipid-Transfer Proteins and BeyondSima Lev
 
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human CancerSima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human CancerSima Lev
 
Sima Lev: MEETING OF THE BIOCHEMICAL SOCIETY
Sima Lev: MEETING OF THE BIOCHEMICAL SOCIETYSima Lev: MEETING OF THE BIOCHEMICAL SOCIETY
Sima Lev: MEETING OF THE BIOCHEMICAL SOCIETYSima Lev
 
Sima Lev | Tethering the SNAREpin in vesicular fusion
Sima Lev | Tethering the SNAREpin in vesicular fusionSima Lev | Tethering the SNAREpin in vesicular fusion
Sima Lev | Tethering the SNAREpin in vesicular fusionSima Lev
 
Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...
Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...
Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...Sima Lev
 
Prof. Sima Lev - The role of Nir proteins in phosphatidylinositol (PI) transp...
Prof. Sima Lev - The role of Nir proteins in phosphatidylinositol (PI) transp...Prof. Sima Lev - The role of Nir proteins in phosphatidylinositol (PI) transp...
Prof. Sima Lev - The role of Nir proteins in phosphatidylinositol (PI) transp...Sima Lev
 
Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...
Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...
Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...Sima Lev
 
Professor Sima Lev - Cell signaling in cancer development and metastasis
Professor Sima Lev - Cell signaling in cancer development and metastasisProfessor Sima Lev - Cell signaling in cancer development and metastasis
Professor Sima Lev - Cell signaling in cancer development and metastasisSima Lev
 
Prof. Sima Lev: Lipid transfer proteins in cancer biology & metastasis
Prof. Sima Lev: Lipid transfer proteins in cancer biology & metastasisProf. Sima Lev: Lipid transfer proteins in cancer biology & metastasis
Prof. Sima Lev: Lipid transfer proteins in cancer biology & metastasisSima Lev
 
Professor Sima Lev: Epithelial–Mesenchymal Transition: EMT
Professor Sima Lev: Epithelial–Mesenchymal Transition: EMTProfessor Sima Lev: Epithelial–Mesenchymal Transition: EMT
Professor Sima Lev: Epithelial–Mesenchymal Transition: EMTSima Lev
 
Prof. Sima Lev - Lipid transfer proteins in cancer biology & metastasis
Prof. Sima Lev - Lipid transfer proteins in cancer biology & metastasisProf. Sima Lev - Lipid transfer proteins in cancer biology & metastasis
Prof. Sima Lev - Lipid transfer proteins in cancer biology & metastasisSima Lev
 
Sima Lev: VAP-B and its role in Amyotrophic lateral sclerosis
Sima Lev: VAP-B and its role in Amyotrophic lateral sclerosis Sima Lev: VAP-B and its role in Amyotrophic lateral sclerosis
Sima Lev: VAP-B and its role in Amyotrophic lateral sclerosis Sima Lev
 

More from Sima Lev (16)

Sima Lev The AXL PYK2 PKC axis as a nexus of stemness circuits in TNBC
Sima Lev  The AXL PYK2 PKC axis as a nexus of stemness circuits in TNBCSima Lev  The AXL PYK2 PKC axis as a nexus of stemness circuits in TNBC
Sima Lev The AXL PYK2 PKC axis as a nexus of stemness circuits in TNBC
 
Sima Lev - Virtual Training
Sima Lev - Virtual TrainingSima Lev - Virtual Training
Sima Lev - Virtual Training
 
Sima Lev - Edinburgh Breast Cancer Symposium
Sima Lev - Edinburgh Breast Cancer SymposiumSima Lev - Edinburgh Breast Cancer Symposium
Sima Lev - Edinburgh Breast Cancer Symposium
 
Animal Lectin Galectin-8 by Hadas Shatz-Azoulay, Yaron Vinik, Roi Isaac, Ulri...
Animal Lectin Galectin-8 by Hadas Shatz-Azoulay, Yaron Vinik, Roi Isaac, Ulri...Animal Lectin Galectin-8 by Hadas Shatz-Azoulay, Yaron Vinik, Roi Isaac, Ulri...
Animal Lectin Galectin-8 by Hadas Shatz-Azoulay, Yaron Vinik, Roi Isaac, Ulri...
 
Sima Lev: Non-Vesicular Lipid Transport by Lipid-Transfer Proteins and Beyond
Sima Lev: Non-Vesicular Lipid Transport by Lipid-Transfer Proteins and BeyondSima Lev: Non-Vesicular Lipid Transport by Lipid-Transfer Proteins and Beyond
Sima Lev: Non-Vesicular Lipid Transport by Lipid-Transfer Proteins and Beyond
 
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human CancerSima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer
 
Sima Lev: MEETING OF THE BIOCHEMICAL SOCIETY
Sima Lev: MEETING OF THE BIOCHEMICAL SOCIETYSima Lev: MEETING OF THE BIOCHEMICAL SOCIETY
Sima Lev: MEETING OF THE BIOCHEMICAL SOCIETY
 
Sima Lev | Tethering the SNAREpin in vesicular fusion
Sima Lev | Tethering the SNAREpin in vesicular fusionSima Lev | Tethering the SNAREpin in vesicular fusion
Sima Lev | Tethering the SNAREpin in vesicular fusion
 
Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...
Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...
Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...
 
Prof. Sima Lev - The role of Nir proteins in phosphatidylinositol (PI) transp...
Prof. Sima Lev - The role of Nir proteins in phosphatidylinositol (PI) transp...Prof. Sima Lev - The role of Nir proteins in phosphatidylinositol (PI) transp...
Prof. Sima Lev - The role of Nir proteins in phosphatidylinositol (PI) transp...
 
Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...
Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...
Prof. Sima Lev - Regulatio of Golgi structure and function in interphase and ...
 
Professor Sima Lev - Cell signaling in cancer development and metastasis
Professor Sima Lev - Cell signaling in cancer development and metastasisProfessor Sima Lev - Cell signaling in cancer development and metastasis
Professor Sima Lev - Cell signaling in cancer development and metastasis
 
Prof. Sima Lev: Lipid transfer proteins in cancer biology & metastasis
Prof. Sima Lev: Lipid transfer proteins in cancer biology & metastasisProf. Sima Lev: Lipid transfer proteins in cancer biology & metastasis
Prof. Sima Lev: Lipid transfer proteins in cancer biology & metastasis
 
Professor Sima Lev: Epithelial–Mesenchymal Transition: EMT
Professor Sima Lev: Epithelial–Mesenchymal Transition: EMTProfessor Sima Lev: Epithelial–Mesenchymal Transition: EMT
Professor Sima Lev: Epithelial–Mesenchymal Transition: EMT
 
Prof. Sima Lev - Lipid transfer proteins in cancer biology & metastasis
Prof. Sima Lev - Lipid transfer proteins in cancer biology & metastasisProf. Sima Lev - Lipid transfer proteins in cancer biology & metastasis
Prof. Sima Lev - Lipid transfer proteins in cancer biology & metastasis
 
Sima Lev: VAP-B and its role in Amyotrophic lateral sclerosis
Sima Lev: VAP-B and its role in Amyotrophic lateral sclerosis Sima Lev: VAP-B and its role in Amyotrophic lateral sclerosis
Sima Lev: VAP-B and its role in Amyotrophic lateral sclerosis
 

Recently uploaded

Sonia Journal club presentation (2).pptx
Sonia Journal club presentation (2).pptxSonia Journal club presentation (2).pptx
Sonia Journal club presentation (2).pptxpalsonia139
 
Is Rheumatoid Arthritis a Metabolic Disorder.pptx
Is Rheumatoid Arthritis a Metabolic Disorder.pptxIs Rheumatoid Arthritis a Metabolic Disorder.pptx
Is Rheumatoid Arthritis a Metabolic Disorder.pptxSamar Tharwat
 
Bangalore whatsapp Number Just VIP Brookefield 100% Genuine at your Door Step
Bangalore whatsapp Number Just VIP Brookefield 100% Genuine at your Door StepBangalore whatsapp Number Just VIP Brookefield 100% Genuine at your Door Step
Bangalore whatsapp Number Just VIP Brookefield 100% Genuine at your Door Stepdarmandersingh4580
 
ANAPHYLAXIS BY DR.SOHAN BISWAS,MBBS,DNB(INTERNAL MEDICINE) RESIDENT.pptx
ANAPHYLAXIS BY DR.SOHAN BISWAS,MBBS,DNB(INTERNAL MEDICINE) RESIDENT.pptxANAPHYLAXIS BY DR.SOHAN BISWAS,MBBS,DNB(INTERNAL MEDICINE) RESIDENT.pptx
ANAPHYLAXIS BY DR.SOHAN BISWAS,MBBS,DNB(INTERNAL MEDICINE) RESIDENT.pptxDr. Sohan Biswas
 
ROSE CASE SPINAL SBRT BY DR KANHU CHARAN PATRO
ROSE  CASE SPINAL SBRT BY DR KANHU CHARAN PATROROSE  CASE SPINAL SBRT BY DR KANHU CHARAN PATRO
ROSE CASE SPINAL SBRT BY DR KANHU CHARAN PATROKanhu Charan
 
Mgr university bsc nursing adult health previous question paper with answers
Mgr university  bsc nursing adult health previous question paper with answersMgr university  bsc nursing adult health previous question paper with answers
Mgr university bsc nursing adult health previous question paper with answersShafnaP5
 
VIP Pune 7877925207 WhatsApp: Me All Time Serviℂe Available Day and Night
VIP Pune 7877925207 WhatsApp: Me All Time Serviℂe Available Day and NightVIP Pune 7877925207 WhatsApp: Me All Time Serviℂe Available Day and Night
VIP Pune 7877925207 WhatsApp: Me All Time Serviℂe Available Day and Nightpatanjali9823#S07
 
CAD CAM DENTURES IN PROSTHODONTICS : Dental advancements
CAD CAM DENTURES IN PROSTHODONTICS : Dental advancementsCAD CAM DENTURES IN PROSTHODONTICS : Dental advancements
CAD CAM DENTURES IN PROSTHODONTICS : Dental advancementsNaveen Gokul Dr
 
Renal Replacement Therapy in Acute Kidney Injury -time modality -Dr Ayman Se...
Renal Replacement Therapy in Acute Kidney Injury -time  modality -Dr Ayman Se...Renal Replacement Therapy in Acute Kidney Injury -time  modality -Dr Ayman Se...
Renal Replacement Therapy in Acute Kidney Injury -time modality -Dr Ayman Se...Ayman Seddik
 
Dermatome and myotome test & pathology.pdf
Dermatome and myotome test & pathology.pdfDermatome and myotome test & pathology.pdf
Dermatome and myotome test & pathology.pdfniloofarbarzegari76
 
ESC HF 2024 Spotlights Day-2.pptx heart failure
ESC HF 2024 Spotlights Day-2.pptx heart failureESC HF 2024 Spotlights Day-2.pptx heart failure
ESC HF 2024 Spotlights Day-2.pptx heart failuremahiavy26
 
Failure to thrive in neonates and infants + pediatric case.pptx
Failure to thrive in neonates and infants  + pediatric case.pptxFailure to thrive in neonates and infants  + pediatric case.pptx
Failure to thrive in neonates and infants + pediatric case.pptxclaviclebrown44
 
Signs It’s Time for Physiotherapy Sessions Prioritizing Wellness
Signs It’s Time for Physiotherapy Sessions Prioritizing WellnessSigns It’s Time for Physiotherapy Sessions Prioritizing Wellness
Signs It’s Time for Physiotherapy Sessions Prioritizing WellnessGokuldas Hospital
 
How to buy 5cladba precursor raw 5cl-adb-a raw material
How to buy 5cladba precursor raw 5cl-adb-a raw materialHow to buy 5cladba precursor raw 5cl-adb-a raw material
How to buy 5cladba precursor raw 5cl-adb-a raw materialSherrylee83
 
SEMESTER-V CHILD HEALTH NURSING-UNIT-1-INTRODUCTION.pdf
SEMESTER-V CHILD HEALTH NURSING-UNIT-1-INTRODUCTION.pdfSEMESTER-V CHILD HEALTH NURSING-UNIT-1-INTRODUCTION.pdf
SEMESTER-V CHILD HEALTH NURSING-UNIT-1-INTRODUCTION.pdfSachin Sharma
 
CONGENITAL HYPERTROPHIC PYLORIC STENOSIS by Dr M.KARTHIK EMMANUEL
CONGENITAL HYPERTROPHIC PYLORIC STENOSIS  by Dr M.KARTHIK EMMANUELCONGENITAL HYPERTROPHIC PYLORIC STENOSIS  by Dr M.KARTHIK EMMANUEL
CONGENITAL HYPERTROPHIC PYLORIC STENOSIS by Dr M.KARTHIK EMMANUELMKARTHIKEMMANUEL
 
Hemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.Gawad
Hemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.GawadHemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.Gawad
Hemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.GawadNephroTube - Dr.Gawad
 
Varicose Veins Treatment Aftercare Tips by Gokuldas Hospital
Varicose Veins Treatment Aftercare Tips by Gokuldas HospitalVaricose Veins Treatment Aftercare Tips by Gokuldas Hospital
Varicose Veins Treatment Aftercare Tips by Gokuldas HospitalGokuldas Hospital
 

Recently uploaded (20)

Sonia Journal club presentation (2).pptx
Sonia Journal club presentation (2).pptxSonia Journal club presentation (2).pptx
Sonia Journal club presentation (2).pptx
 
Is Rheumatoid Arthritis a Metabolic Disorder.pptx
Is Rheumatoid Arthritis a Metabolic Disorder.pptxIs Rheumatoid Arthritis a Metabolic Disorder.pptx
Is Rheumatoid Arthritis a Metabolic Disorder.pptx
 
Best medicine 100% Effective&Safe Mifepristion ௵+918133066128௹Abortion pills ...
Best medicine 100% Effective&Safe Mifepristion ௵+918133066128௹Abortion pills ...Best medicine 100% Effective&Safe Mifepristion ௵+918133066128௹Abortion pills ...
Best medicine 100% Effective&Safe Mifepristion ௵+918133066128௹Abortion pills ...
 
Bangalore whatsapp Number Just VIP Brookefield 100% Genuine at your Door Step
Bangalore whatsapp Number Just VIP Brookefield 100% Genuine at your Door StepBangalore whatsapp Number Just VIP Brookefield 100% Genuine at your Door Step
Bangalore whatsapp Number Just VIP Brookefield 100% Genuine at your Door Step
 
ANAPHYLAXIS BY DR.SOHAN BISWAS,MBBS,DNB(INTERNAL MEDICINE) RESIDENT.pptx
ANAPHYLAXIS BY DR.SOHAN BISWAS,MBBS,DNB(INTERNAL MEDICINE) RESIDENT.pptxANAPHYLAXIS BY DR.SOHAN BISWAS,MBBS,DNB(INTERNAL MEDICINE) RESIDENT.pptx
ANAPHYLAXIS BY DR.SOHAN BISWAS,MBBS,DNB(INTERNAL MEDICINE) RESIDENT.pptx
 
ROSE CASE SPINAL SBRT BY DR KANHU CHARAN PATRO
ROSE  CASE SPINAL SBRT BY DR KANHU CHARAN PATROROSE  CASE SPINAL SBRT BY DR KANHU CHARAN PATRO
ROSE CASE SPINAL SBRT BY DR KANHU CHARAN PATRO
 
Mgr university bsc nursing adult health previous question paper with answers
Mgr university  bsc nursing adult health previous question paper with answersMgr university  bsc nursing adult health previous question paper with answers
Mgr university bsc nursing adult health previous question paper with answers
 
VIP Pune 7877925207 WhatsApp: Me All Time Serviℂe Available Day and Night
VIP Pune 7877925207 WhatsApp: Me All Time Serviℂe Available Day and NightVIP Pune 7877925207 WhatsApp: Me All Time Serviℂe Available Day and Night
VIP Pune 7877925207 WhatsApp: Me All Time Serviℂe Available Day and Night
 
CAD CAM DENTURES IN PROSTHODONTICS : Dental advancements
CAD CAM DENTURES IN PROSTHODONTICS : Dental advancementsCAD CAM DENTURES IN PROSTHODONTICS : Dental advancements
CAD CAM DENTURES IN PROSTHODONTICS : Dental advancements
 
Renal Replacement Therapy in Acute Kidney Injury -time modality -Dr Ayman Se...
Renal Replacement Therapy in Acute Kidney Injury -time  modality -Dr Ayman Se...Renal Replacement Therapy in Acute Kidney Injury -time  modality -Dr Ayman Se...
Renal Replacement Therapy in Acute Kidney Injury -time modality -Dr Ayman Se...
 
Dermatome and myotome test & pathology.pdf
Dermatome and myotome test & pathology.pdfDermatome and myotome test & pathology.pdf
Dermatome and myotome test & pathology.pdf
 
ESC HF 2024 Spotlights Day-2.pptx heart failure
ESC HF 2024 Spotlights Day-2.pptx heart failureESC HF 2024 Spotlights Day-2.pptx heart failure
ESC HF 2024 Spotlights Day-2.pptx heart failure
 
Failure to thrive in neonates and infants + pediatric case.pptx
Failure to thrive in neonates and infants  + pediatric case.pptxFailure to thrive in neonates and infants  + pediatric case.pptx
Failure to thrive in neonates and infants + pediatric case.pptx
 
Signs It’s Time for Physiotherapy Sessions Prioritizing Wellness
Signs It’s Time for Physiotherapy Sessions Prioritizing WellnessSigns It’s Time for Physiotherapy Sessions Prioritizing Wellness
Signs It’s Time for Physiotherapy Sessions Prioritizing Wellness
 
How to buy 5cladba precursor raw 5cl-adb-a raw material
How to buy 5cladba precursor raw 5cl-adb-a raw materialHow to buy 5cladba precursor raw 5cl-adb-a raw material
How to buy 5cladba precursor raw 5cl-adb-a raw material
 
SEMESTER-V CHILD HEALTH NURSING-UNIT-1-INTRODUCTION.pdf
SEMESTER-V CHILD HEALTH NURSING-UNIT-1-INTRODUCTION.pdfSEMESTER-V CHILD HEALTH NURSING-UNIT-1-INTRODUCTION.pdf
SEMESTER-V CHILD HEALTH NURSING-UNIT-1-INTRODUCTION.pdf
 
In Kuwait Abortion pills (+918133066128)@Safe abortion pills in Kuwait City
In Kuwait Abortion pills (+918133066128)@Safe abortion pills in Kuwait CityIn Kuwait Abortion pills (+918133066128)@Safe abortion pills in Kuwait City
In Kuwait Abortion pills (+918133066128)@Safe abortion pills in Kuwait City
 
CONGENITAL HYPERTROPHIC PYLORIC STENOSIS by Dr M.KARTHIK EMMANUEL
CONGENITAL HYPERTROPHIC PYLORIC STENOSIS  by Dr M.KARTHIK EMMANUELCONGENITAL HYPERTROPHIC PYLORIC STENOSIS  by Dr M.KARTHIK EMMANUEL
CONGENITAL HYPERTROPHIC PYLORIC STENOSIS by Dr M.KARTHIK EMMANUEL
 
Hemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.Gawad
Hemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.GawadHemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.Gawad
Hemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.Gawad
 
Varicose Veins Treatment Aftercare Tips by Gokuldas Hospital
Varicose Veins Treatment Aftercare Tips by Gokuldas HospitalVaricose Veins Treatment Aftercare Tips by Gokuldas Hospital
Varicose Veins Treatment Aftercare Tips by Gokuldas Hospital
 

Tethering the assembly of SNARE complexes by Sima Lev and WonJin Hong

  • 1. Special Issue: Membrane Trafficking Tethering the assembly of SNARE complexes WanJin Hong1,2 and Sima Lev3 1 School of Pharmaceutical Sciences, Xiamen University, Xiamen, People’s Republic of China 2 Institute of Molecular and Cell Biology, Singapore 3 Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel The fusion of transport vesicles with their target mem- branes is fundamental for intracellular membrane traf- ficking and diverse physiological processes and is driven by the assembly of functional soluble N-ethylmaleimide- sensitive factor attachment protein receptor (SNARE) complexes. Prior to fusion, transport vesicles are physi- cally linked to their target membranes by various teth- ering factors. Recent studies suggest that tethering factors also positively regulate the assembly of function- al SNARE complexes, thereby coupling tethering with fusion events. This coupling is mediated, at least in part, by direct physical interactions between tethering fac- tors, SNAREs, and Sec1/Munc18 (SM) proteins. In this review we summarize recent progress in understanding the roles of tethering factors in the assembly of specific and functional SNARE complexes driving membrane- fusion events. Membrane fusion Membrane fusion along the secretory and endocytic path- ways is mediated by a large group of membrane-bound proteins known as SNAREs. All SNAREs contain a char- acteristic coiled-coil SNARE motif of approximately 70 amino acids comprising heptad repeats and can function on either target membranes (t-SNAREs) or transport vesi- cles (v-SNAREs). The interaction of cognate v- and t- SNAREs leads to the formation of a trans-SNARE complex or SNAREpin, in which four SNARE motifs assemble as a twisted parallel four-helix bundle that brings the opposing membranes together and eventually catalyzes membrane fusion [1,2]. Following fusion, the remaining SNARE com- plex on the fused membranes is referred to as a cis-SNARE complex, which undergoes disassembly catalyzed by ATPase N-ethylmaleimide-sensitive fusion protein (NSF) and its cofactor soluble NSF attachment protein (SNAP) [3] to recycle the SNAREs for a new fusion event [4]. Pairing of distinct SNAREs and subsequent assembly of functional SNAREpins are employed by different mem- brane-trafficking events [5]. In many cases, a single SNARE can be assembled into more than one SNAREpin and thus can regulate several fusion events [6,7]. Hence, the assembly of a functional SNAREpin is remarkably important for both membrane fusion specificity and coor- dinating different membrane-trafficking pathways. Recon- stitution studies suggest that numerous SNARE complexes can be formed in vitro, but only a few can functionally drive membrane fusion [8,9]. Furthermore, the assembly of a functional SNAREpin occurs in a step- wise manner and is attributed to the formation of SNARE complex intermediates, such as t-SNARE subcomplexes on the target membranes that provide templates for v- SNARE binding [7]. These findings raise several critical questions. What are the mechanisms that regulate the assembly of specific SNAREpins? How is the promiscuous assembly of nonfunctional SNARE complexes prevented in intact cells? How are the intermediate steps of the assem- bly process regulated? Numerous studies have shown that membrane-fusion specificity is largely dependent on SNARE proteins, be- cause different SNAREpins can catalyze distinct fusion events [1,6]. Recent studies, however, suggest that both SM proteins and tethering factors play an active role in SNAREpin formation and thus contribute to the specificity of the fusion as well as to its speed and high fidelity [10– 15]. Tethering factors appear to influence SNAREpin as- sembly in several different ways including stabilization of SNARE proteins or the entire SNARE complex, gathering of t-SNAREs on the target membranes, or activating the assembly process by interacting with SM proteins. The direct physical interactions of tethering factors with SNAREs and SM proteins mediate many of these func- tions, as will be discussed in this review. Tethering factors also interact with Rab GTPases and vesicle coats and are believed to bridge the transport vesicles with their target membranes [16,17], thereby functionally coupling tether- ing with fusion events. This review summarizes recent advances in understanding the role of tethering factors in the assembly of functional SNARE complexes. SNARE: general features, classification, and complexes The SNARE hypothesis was first introduced in 1993 [3] and suggested that a v-SNARE on the vesicle pairs with cognate t-SNAREs on the target membrane to form a complex that not only determines the specificity of the Review 0962-8924/$ – see front matter ß 2013 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tcb.2013.09.006 Corresponding author: Lev, S. (Sima.Lev@weizmann.ac.il). Keywords: SNAREs; SM proteins; tethering factors; SNAREpin; membrane fusion. Trends in Cell Biology, January 2014, Vol. 24, No. 1 35
  • 2. fusion but also catalyzes the fusion process. Since then many SNAREs have been identified, of which 38 are known to exist in humans [6,18] (Figure 1). Most SNAREs contain one SNARE motif, whereas four SNAREs (SNAP-23, SNAP- 25, SNAP-29/GS32, and SNAP-47) contain two SNARE motifs [6,18–20] (Figure 1). Initially, SNAREs were classi- fied as v- or t-SNAREs according to their subcellular locali- zation. However, many SNAREs can be found on both vesicles and target membranes; therefore, an alternative classification based on the crystal structure of the synaptic SNARE complex (Syntaxin [Stx]1A, synaptobrevin2, and Synaptosome-associated protein [SNAP]-25B) has been for- mulated [21]. The crystal structure revealed that an ionic layer comprising an arginine (R) (contributed by synapto- brevin) and three glutamine (Q) residues (contributed by Stx1A and SNAP-25) is present in the central position of the four-helix bundle. These findings led to the structural clas- sification of R- and Q-SNAREs [22] and suggested that a functional SNARE complex generally comprises three Q- SNARE motifs and one R-SNARE motif. Most R-SNAREs act as v-SNAREs and most Q-SNAREs act as t-SNAREs. However, there are exceptions: the R-SNAREs Ykt6 and Sec22B act as part of the t-SNARE subcomplexes, whereas GS15, Bet1, and Slt1 are Q-SNAREs that function as v- SNAREs. In addition to the SNARE motif, most SNAREs contain a C-terminal hydrophobic transmembrane domain (TM) (Figure 1). Seven of the 38 SNAREs do not have a TM domain and instead associate with membranes by various lipid modifications. SNAP-23, SNAP-25, SNAP-29, SNAP47, Stx9/19, and Stx11 are palmitoylated at multiple cysteineresidues, whereasYkt6 containsa C-terminalmotif that mediates its prenylation (farnesylation) and subse- quent palmitoylation [23]. Moreover, many SNAREs con- tain N-terminal regulatory regions preceding the SNARE motif (Figure 1). These N-terminal regions regulate impor- tant properties of SNAREs. For example, the short N-ter- minal peptide of Stx1, Stx5, and Stx16 interacts with SM proteins [6]. The Habc region ofStx16folds intoa three-helix bundle and intramolecularily interacts with the SNARE motif, thereby preventing its assembly into a SNARE com- plex [24]. Similarly, the longin domain of Vamp7, an R-SNARE involved in both endocytic and secretory path- ways, interacts with its SNARE motif via hydrophobic inter- actions [25]. The N-terminal extension of VAMP4 contains a dileucine motif and acidic clusters that mediate its recycling fromtheendosometothetrans-Golginetwork(TGN)[26,27]. Because the formation of a functional SNAREpin is the key driver for membrane fusion, identification of functional SNARE complexes in vivo has been an important area of SNARE research. The major SNARE complexes function- ing in the secretory and endocytic pathways are shown in Figure 2 [6]. As can be seen, most SNAREpins contain at least one t-SNARE belonging to the Stx family, and a given SNARE can participate in several distinct SNARE com- plexes. For example, Stx5 is present in at least three different SNAREpins regulating Golgi trafficking and thus can be envisioned as the master SNARE of the Golgi apparatus [28]. Regulation of SNAREpin assembly and function Given that SNAREpins mediate fusion events, it is no surprise that several mechanisms regulate their formation and/or functionality. Below we discuss the role of SM proteins and tethering factors in SNAREpin assembly. SM proteins SM proteins are evolutionarily conserved soluble periph- eral membrane proteins of 60–90 kDa. Four major classes of SM proteins have been identified in mammals: Sly1, VPS45, VPS33 (VPS33A, VPS33B), and Munc18 (Munc18- 1, Munc18-2, and Munc18-3) [6,10]. SM proteins are con- sidered key universal components of the fusion machinery. They function in distinct intracellular transport steps and physically interact with different SNARE proteins (Figure 2) [7,10,29]. Sly1, for example, functions at the endoplasmic reticulum (ER)–Golgi, Vps45 at the endo- some–TGN, Vps33 at the endocytic/lysosomal system, and Muc18 at the plasma membrane (Figure 2). In vitro reconstitution studies convincingly showed that SM pro- teins strongly accelerate the rate of SNARE-mediated fusion and contribute to the specificity of various fusion events [11,30,31]. Indeed, SM proteins are required for all Stx1, 2, 3, 4, 5, 7, 12/13, 16, 17, 18Ha SNARE moƟfHb Hc TMN-Pep VƟ1a, VƟ1b, GS27, GS28, Stx6, 8, 10Ha SNARE moƟfHb Hc TM VAMP7, Sec22b, Sec22a, Sec22cSNARE moƟfLongin TM Slt1/Use1, Sec20 SNARE moƟf TM VAMP1, 2, 3, 5, 8, GS15, Bet1 Ha SNARE moƟfHb HcN-Pep SNARE moƟfLongin VAMP4SNARE moƟfTGN S TM SNARE moƟf TMUnknown SNAP-23, 25, 29, 47 (mulƟple Cys for palmitoylaƟon) Ykt6 (C-terminal prenylaƟon and palmitoylaƟon) Stx9/19, 11 (mulƟple Cys for palmitoylaƟon) SNARE moƟfSNARE MoƟf TRENDS in Cell Biology Figure 1. Domain organization of human soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs). The indicated SNAREs along with their corresponding domains are shown. Abbreviation: TM, transmembrane domain. Review Trends in Cell Biology January 2014, Vol. 24, No. 1 36
  • 3. SNARE-mediated fusion events and recent studies on Vps33 suggest that SM proteins promote the opening of a fusion pore by enhancing the activity of a SNARE com- plex [32]. SM proteins can bind individual SNAREs or SNARE complexes and regulate SNAREpin assembly [10]. Sly1 binds to a short, evolutionarily conserved N-terminal pep- tide of the mammalian t-SNAREs Stx5 and Stx18 and their yeast homologs Sed5p and Ufe1p, respectively [33]. Vps45 similarly interacts with the N-terminal peptide of Stx16 (Tlg2p in yeast) and regulates endosome-to-TGN retro- grade trafficking [34]. Interestingly, Vps45p also interacts with the v-SNARE Snc2p, which participates in SNARE complexes containing Tlg2p in yeast. Hence, Vps45p can bind Tlg2p-containing SNARE complexes independent of its Tlg2p binding [35], suggesting that SM proteins can interact with their cognate SNAREs through distinct mechanisms and at different stages in the SNARE assem- bly/disassembly cycle. Consistent with the multiple modes of binding, yeast Sec1p interacts with a fully assembled exocytic SNARE complex [36]. Mammalian Munc18-1 pro- tein, however, binds the t-SNARE Stx1 in a closed confor- mation [37,38], in which an intramolecular interaction between the N-terminal Habc domain of Stx1 and its SNARE domain preclude its assembly into a SNARE complex. Nevertheless, Munc18-1 stimulates SNARE-me- diated fusion in vitro. It was proposed that Munc18-1 facilitates functional SNAREpin assembly by interacting with the preassembled Stx1–SNAP25 t-SNARE subcom- plex, which serves as a platform for VAMP2 binding [31]. The different modes of interaction between SM proteins and SNAREs demonstrate the important regulatory roles of SM proteins in SNAREpin assembly. Indeed, in vitro binding assays suggested that the interaction of SM pro- teins with SNAREs prevents the formation of non-physio- logical SNARE complexes and stimulates specific SNARE pairing [39]. For additional information on SM proteins and their mechanisms of action, see these recent reviews [40,41]. Tethering factors Tethering factors are a large group of proteins or multiple- protein complexes that link transport vesicles to their cognate target membranes. As such, they regulate funda- mental cellular processes and are implicated in various diseases (Box 1). Tethering factors can be divided into two major groups: homodimeric long coiled-coil proteins and multisubunit tethering complexes (MTCs) [16,42–44]. Coiled-coil tethers are large, hydrophilic, dimeric proteins comprising two globular heads connected by long coiled- coil domains that can interact with vesicles over distances of more than 200 nm [42]. MTCs, which contain three to ten Bet1 Stx5 GS28 Ykt6 GS15 Stx5 GS28 Ykt6 Slt1 Stx18 Sec20 Sec22b Vamp7 Stx7 VƟ1b Stx8 Bet1 Stx5 GS27 Sec22b ER ERGIC PM cis-Golgi Golgi stack Vamp2 Stx1 SNAP-25 Vamp8 Stx4 SNAP-23 Recycling endosome TGN MVB Lysosome Vamp2/3 Stx13 SNAP-25/29 Vamp8 Stx7 VƟ1b Stx8 Early endosome Vamp4 Stx6 Stx16 VƟ1a Munc18 Sly1 Vps45 Vps33 Vps33 Sly1 Sly1 B TRENDS in Cell Biology SynapƟc vesicles Secretory granule Figure 2. Soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes along the secretory and endocytic pathways in mammals. Schematic summary of known mammalian SNARE complexes and their sites of action along the exocytic and endocytic pathways. Potential v-SNAREs are indicated in red; the Sec1/ Munc18 SM proteins for some SNARE complexes are marked in purple. Abbreviations: ER, endoplasmic reticulum; ERGIC, ER–Golgi intermediate compartment; MVBs, multivesicular bodies; PM, plasma membrane; TGN, trans-Golgi network. Review Trends in Cell Biology January 2014, Vol. 24, No. 1 37
  • 4. subunits with an overall molecular weight of approximate- ly 250–800 kDa, can interact with vesicles over much shorter distances (up to 30 nm) [45]. Many long coiled-coil tethers including GM130, Golgin- 45, Golgin-97, Golgin-245, and p115 localize to the Golgi apparatus and belong to the golgin family. The golgins (approximately 20 members) are regulated by small GTPases of the Rab and Arl families and function in membrane–membrane and membrane–cytoskeleton teth- ering at the Golgi apparatus [46]. Other long coiled-coil tethers such as EEA1, Rabaptin-5, Rabenosyn-5, and Rabip4 associate with endosomal compartments [44]. MTCs can be divided into two major classes: complexes associated with tethering containing helical rods (CATCHR) and Class C vacuolar protein-sorting (Vps) complexes. The exocyst, DSL1, Conserved Oligomeric Gol- gi (COG), and Golgi-associated retrograde protein (GARP) (Figure 3A,C,D) belong to the CATCHR family, which exhibits low sequence identity but conserved helical-bun- dle structures [45]. Homotypic fusion and protein sorting (HOPS) (Figure 3B) and class C core vacuole/endosome tethering (CORVET) belong to the Class C Vps complexes and regulate transport from the late Golgi to endosomal/ lysosomal compartments [47]. The multisubunit Transport Protein Particle (TRAPP) complexes (I, II, and III) have also been considered as tethering factors. However, in contrast to other tethering factors that function as small GTPase effectors, TRAPPs act as guanine nucleotide ex- change factors (GEFs) for the Rab small GTPases [48]. TRAPPI, which is required for ER–Golgi transport, inter- acts directly with vesicle coats, thereby coupling Rab acti- vation with coat recognition and vesicular tethering [49,50]. Tethering factors are associated with different intracel- lular membranes via distinct mechanisms, including specif- ic targeting motifs (such as the GRIP domain in several golgins), phospholipid binding, and/or interactions with small GTPases [46,51]. Most tethering factors are Rab or Arl effectors that interact directly with small GTPases in their GTP-bound form [51]. CORVET and EEA1 are Rab5 effectors, whereas HOPS is a Rab7 effector. Rab5 (Vps21p in yeast) functions at the early endosome, whereas Rab7 (Ypt7p in yeast) functions at late endosomes and lysosomes [52]. COG is a Rab1 (Ypt1p) and Rab6 (Ypt6p) effector [53–56], whereas GARP interacts with small GTPases of the Rab and Arl families (Ypt6p and Arl1p, respectively, in Saccharomyces cerevisiae) [57]. The exocyst interacts with the GTP-bound form ofthe Rab small GTPase Sec4p in yeast and with the Rab11, Rab8, and Ral small GTPases in mammals. The exocyst also interacts with Rho and Cdc42 small GTPases [58]. Comprehensive description of these tethers and their structural features and subcellular distri- bution has been published previously [42,45,51,59,60]. Although both MTCs and long coiled-coil tethers have been implicated in vesicular tethering, coiled-coil tethers are presumed to be involved in the initial, highly dynamic stages of tethering, because they can form transient, re- versible, and low-affinity interactions [44]. By contrast, MTCs may regulate later events and be actively involved in SNAREpin assembly. MTCs, which capture vesicles at relatively short distances from their target membranes, can interact simultaneously with different transport com- ponents through their multiple subunits. These multiva- lent interactions could integrate different trafficking components and consequently couple vesicular tethering, docking, and fusion events. The structural and functional differences between long coiled-coil tethers and MTCs suggest that coiled-coil tethers and MTCs could have complementary roles and potentially cooperate during vesicular tethering, docking, and fusion. Consistent with this hypothesis, previous studies have shown direct inter- actions between long coiled-coil tethers such as p115 or Golgin 84 and the COG complex [61,62]. In addition to their established roles in vesicular cap- turing and docking, tethering factors also positively regu- late SNAREpin assembly by direct interaction with components of the fusion machinery – SNAREs and SM proteins [12–15]. These interactions could influence: (i) the stability of certain SNAREs and/or their corresponding complexes; (ii) the assembly of SNARE subcomplex inter- mediates; and (iii) the speed and specificity of SNAREpin assembly. A brief description of these interactions and their physiological roles is given below. Tethering factors interact with SNAREs and SM proteins Both coiled-coil tethers and MTCs interact directly with different SNAREs. The coiled-coil tether p115, for Box 1. Genetic mutations in MTCs, SNAREpin assembly, and inherited diseases In recent years, multiple mutations in genes encoding different subunits of MTCs have been identified as the cause of severe inherited diseases. For example, a single amino acid substitution (L967N) in the carboxyl-terminal region of the Vps54 subunit of the GARP complex was identified as the cause of defective spermiogen- esis and a motor neuron disease in the ‘wobbler’ mouse [92]. Homozygous wobbler mice display unsteady (‘wobbly’) gait with progressive muscle weakness, a phenotype resembling the motor neuron degeneration observed in amyotrophic lateral sclerosis (ALS). This phenotype is accompanied by the formation of ubiquitinated TDP-43 inclusions [93]. It was proposed that the L967N mutation reduces the level of functional GARP and conse- quently its associated autophagic activity, thereby attenuating the clearance of TDP-43 aggregates and facilitating motor neuron death [57]. Likewise, point mutations within the human VPS33B gene, a subunit of the HOPS complex, cause arthrogryposis, renal dysfunc- tion, and cholestasis (ARC) syndrome (Online Mendelian Inheritance in Man [OMIM] 208085). ARC is an autosomal-recessive, multi- system disorder characterized by renal tubular abnormalities, cholestasis, hypotonia, and platelet storage-pool deficiency [94]. It was proposed that the VPS33B mutations cause abnormal assembly of the entire HOPS complex and consequently impair SNAREpin assembly and biogenesis and/or fusion of lysosome-related orga- nelles. Strikingly, a direct influence on the assembly of Golgi SNAREpins has been demonstrated in Cog7- and Cog8-deficient fibroblasts derived from patients with congenital disorders of glycosylation (CDGs) [95]. CDGs are a heterogeneous group of inherited disorders characterized by pleiotropic glycosylation de- fects [96]. In recent years, multiple mutations in the genes encoding different COG subunits have been identified as the cause of CDG [97–101]. These mutations affect the assembly of the COG complex, thereby impairing intra-Golgi retrograde transport and conse- quently the proper localization of Golgi glycosylation enzymes [102]. Overall, these findings demonstrate the importance of tethering factors for normal cell function and their critical physio- logical roles. Review Trends in Cell Biology January 2014, Vol. 24, No. 1 38
  • 5. example, interacts with several Golgi SNAREs including Stx5, GS28, GS15, Ykt6, rSec22, and GS27/membrin [13] and the endosomal EEA1 tether interacts with Stx6 [63]. Different subunits of all MTCs except the TRAPP com- plexes physically interact with various SNAREs both in yeast and mammals (Table 1 in [42]). Mechanistically, tethering factors can interact with multiple structural motifs in SNAREs. For example, the Tip20 and Sec39 subunits of the DSL1 complex, which regulates Golgi-to-ER retrograde transport, interact di- rectly with the N-terminal regulatory domains of the t- SNAREs Use1 and Sec20 at the ER membrane (Figure 3C) [64,65]. The Vps51 subunit of the GARP complex, which regulates endosome-to-TGN retrograde transport, inter- acts with the N-terminal Habc domain of Stx6 (Tlg1p in yeast) [66,67] and the Vps53 and Vps54 subunits of GARP interact with the SNARE motifs of the Golgi SNAREs Stx6, Stx16, and Vamp4 (Figure 3A) [68,69]. Several subunits of the octameric COG complex (Cog1–8), which regulates intra-Golgi and endosome-to-Golgi retrograde transport, also interact with the SNARE motifs of multiple Golgi SNAREs (Figure 3A) [12,70,71]. These interactions with SNARE motifs are mediated by N-terminal coiled-coil domains found in the GARP and COG subunits [43,72]. Subunits of the HOPS complex, which is required for multiple fusion events at the late endosome and homotypic fusion of vacuoles in yeast [73–75], also interact directly with SNAREs. The Vps33 subunit of the HOPS complex binds the SNARE motifs of the t-SNAREs Vam3 and Vam7 as well as the SNARE motif of the v-SNARE Nyv1. The Vps11, Vps16, and Vps18 subunits of HOPS bind the N- terminal Habc domain of Vam3 and the PX domain of Vam7 (Figure 3B) [76–78]. Vps33, which is also an integral subunit of the CORVET complex, interacts directly with endosomal SNAREs, including the yeast syntaxin Pep12p [74]. Remarkably, Vps33 is an SM protein that functions as Vps52 Vps53 Vps54 Vps51 Golgi Stx5 GS28Ykt6 Stx16 VƟ1a Vam p4 Stx6 Sly1 p Exocyst PM Sec9 Sso1/2 Sec4 (Rab) Vesicle Snc1/2 Sec1 Sec15 Sec10Sec6 Sec8 Exo84 Exo70 Sec5 Sec3 DSL1 Sec39 Dsl1 Tip20 ER COP-I Vesicle Sec22 Ufe1 Use1 Sec20 HOPS 16 33 41 18 11 39 Rab/Ypt7 Vam3 Vam7 Vam3 Vam7 Nyv1 Stx6 Stx16 Vamp4 Rab/Ypt7 Vam 3 VƟ1 Vam 7 Nyv1 SNARE abc abc COG 4 1 8 2 3 7 5 6 Vps45 GARP (D) (B)(A) (C) PX t-SNAREs TRENDS in Cell Biology Figure 3. Multisubunit tethering complexes (MTCs) interact with soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) to couple vesicular tethering with fusion. (A) Conserved Oligomeric Golgi (COG) and Golgi-associated retrograde protein (GARP) MTCs interact with multiple SNAREs on the Golgi membranes. The eight subunits of the COG complex are organized in two structurally and functionally distinct lobes (light and dark blue) [103]. The Cog4, Cog6, and Cog7 subunits bind the SNARE domains of the indicated SNAREs. Cog4 also binds the Sec1/Munc18 (SM) proteins Sly1 and Vps45 [12,82]. The Vps52, Vps53, and Vps54 subunits of GARP (purple) can be assembled into a core complex and localize to the Golgi in the absence of Vps51 [67]. The Vps51 subunit (pink) interacts with the N-terminal regulatory Habc domain of Syntaxin (Stx)6 (Tlg1p in yeast) [66,67]. The N-terminal regions of human Vps53 and Vps54 bind to the SNARE motifs of Stx6, Stx16, and Vamp4 [68]. (B) The yeast homotypic fusion and protein-sorting (HOPS) complex via its Vps33 subunit (pink) interacts with the SNARE motifs (pink) of target membrane SNAREs (t- SNAREs) Vam3 (Stx7 in mammals) and Vam7 (Stx8 in mammals) and the transport vesicle SNARE (v-SNARE) Nyv1. The Vps11, Vps16, and Vps18 subunits (green) interact with the regulatory Habc and PX domains of the t-SNAREs Vam3 and Vam7 on the vacuole or lysosome membrane (green). Vps39 and Vps41 subunits (yellow) bind the small Rab GTPase Ypt7 [52]. (C) A schematic presentation of the yeast DSL1 complex according to its crystal structure [64]. DSL1 interacts with the t-SNAREs Sec20 and Use1 via its Tip20 and Sec39 subunits and induces t-SNARE gathering on the endoplasmic reticulum (ER) membrane. The Dsl1p subunit interacts with subunits of the COP-I coat and tethers COP-I vesicles to the ER membranes [90]. (D) The yeast exocyst complex tethers secretory vesicles to the plasma membrane (PM). The Sec3 and Exo70 (pink) subunits interact with the PM via their positively charged residues, whereas the Sec15 subunit interacts with the Rab GTPase Sec4, which localizes on a secretory vesicle. These interactions are required for vesicular tethering [104]. The interactions of the Sec6 subunit with the t-SNARE Sec9 (SNAP23 in mammals) and the SM protein Sec1 and of the Sec3 subunit with Sso1/2 (Stx1 in mammals) regulate the assembly the Sec9–Sso1 t-SNARE complex that binds the v-SNARE Snc1/2 (VAMP3 in mammals) as described in the text. Review Trends in Cell Biology January 2014, Vol. 24, No. 1 39
  • 6. one subunit of the HOPS and CORVET heterohexameric complexes and mediates multiple interactions with SNAREs [52]. The incorporation of an SM protein into MTCs demonstrates the coordinated functions of tethers, SM proteins, and SNAREs in regulating intracellular membrane fusion. Consistent with this mode of action, both the exocyst and the COG complex interact directly with SM proteins and SNAREs. The exocyst, an evolutionarily conserved complex im- plicated in the tethering of secretory vesicles to the plasma membrane, comprises eight subunits (Sec3, Sec5, Sec6, Sec8, Sec10, Sec15, Exo70, and Exo84) [58]. The complex is directly involved in the assembly of the Sec9–Sso1–Snc2 SNAREpin at the plasma membrane in yeast (Figure 3D). Its Sec6 subunit binds the SM protein Sec1 and the t- SNARE Sec9 via overlapping binding sites that prevent the formation of a ternary Sec6–Sec9–Sec1 complex. Fur- thermore, Sec6 interacts with Sec9 in the absence of other exocyst subunits, but interacts with Sec1 in the presence of exocyst subunits [79,80]. When Sec6 is found outside the exocyst complex, its interaction with Sec9 inhibits the interaction between Sec9 and its syntaxin partner Sso1 [79], thereby preventing the formation of a premature t- SNARE complex at the plasma membrane. However, the assembly of the exocyst complex leads to the release of Sec6 from Sec9, opening of the autoinhibitory domain of Sso1, and concomitant recruitment of Sec1 for regulating vesic- ular tethering and fusion events [81]. These observations suggest that the interactions between subunits of MTCs, SNAREs, and SM proteins are crucial for spatial and temporal regulation of SNAREpin assembly. Consistent with these observations, previous studies of the COG complex suggest that the interaction between its Cog4 subunit and the SM protein Sly1 is crucial for SNAR- Epin assembly and membrane fusion at the Golgi mem- branes. However, in this case a ternary complex between Cog4, Sly1, and Stx5 can form due to distinct adjacent binding sites for Sly1 and Stx5 at the N-terminal coiled-coil domain of Cog4 [82]. The interaction of Cog4 with both Sly1 and Stx5 is required for the assembly of Stx5–GS28–Ykt6– GS15 SNAREpin at the Golgi complex and consequently for intra-Golgi retrograde transport [70,82]. Strikingly, Cog4 also interacts with the SM protein Vps45 and Stx16, which regulate endosome-to-Golgi retrograde transport. These interactions are also mediated by the N-terminal coiled-coil domain of Cog4 and compete with the Sly1–Stx5 interac- tions [12]. These observations suggest that the COG com- plex employs similar mechanisms to regulate the assembly of the two different Golgi SNAREpins (Stx5–GS28–Ykt6– GS15 and Stx6–Stx16–Vti1a–VAMP4) and thereby may coordinate their corresponding transport routes. It seems that both COG and exocyst complexes are in- volved in recruiting and/or orienting the SM proteins to a partially assembled SNARE complex, and possibly stabiliz- ing SM–SNARE interactions. These effects might enhance direct interaction between the SM proteins and both the t- and v-SNAREs of SNARE complex intermediates, thereby facilitating the progression of fusion events [11]. The roles of tethering factor–SNARE interactions Functionally, the interactions between tethering factors and SNAREs can influence the assembly of SNARE SNAREpin assembly tSNAREs gathering Vesicle UncoaƟng Vesiclular fusion SNARE complex disassembly Golgi Sly1 GS15 VAMP4 Vps45 COG Vesicle capture UncoaƟng Ykt6 GS28 Stx5 VƟ1a Stx6 Stx16 TRENDS in Cell Biology Figure 4. Roles of the Conserved Oligomeric Golgi (COG) complex in vesicular tethering and SNAREpin assembly at the Golgi apparatus. The COG complex via its different subunits interacts with distinct t-SNAREs and SM proteins on the Golgi membranes and gathers cognate t-SNAREs and SM protein, thereby promoting the assembly of specific ternary t-SNARE complexes. The COG complex also interacts with vesicle coats [56] and consequently promotes vesicular tethering. It is currently unknown at which step COG dissociates from the assembled SNARE complex, but it possibly stabilizes the assembled complex [70]. The COG complex possibly employs similar mechanisms to assemble two distinct fusogenic Golgi SNARE complexes (Stx5–GS28–Ykt6–GS15 and Stx6–Stx16–Vtia–VAMP4), thereby coordinating their associated transport routes. We propose that the assembly of fusogenic SNARE complexes prevents promiscuous assembly of non-fusogenic t-SNARE interactions on the Golgi membranes [12]. Review Trends in Cell Biology January 2014, Vol. 24, No. 1 40
  • 7. complex intermediates, stabilize SNARE complex assem- bly, and spatially and temporally organize fusion events at specific membrane domains. Many MTCs can interact simultaneously through their different subunits with multiple SNAREs and gather spe- cific SNAREs at specific membrane compartments. Sub- units of the GARP, DSL1, and COG complexes, for example, can simultaneously interact with specific t- SNAREs and enhance t-SNARE subcomplex assembly on the Golgi or ER membranes (Figure 3) [12,64,68,69]. The formation of t-SNARE subcomplexes represents an intermediate step in SNAREpin assembly [7]. Hence, SNARE gathering appears to be the major role of MTCs in SNAREpin assembly (Figure 4). This gathering mediat- ed by multivalent interactions not only facilitates the assembly of fusogenic SNARE complexes that functionally drive membrane fusion, but can also prevent the assembly of non-fusogenic SNARE interactions in intact cells. Re- cently it was shown that the Cog4 subunit positively regulates the assembly of fusogenic SNARE complexes at the Golgi complex, thereby preventing non-fusogenic t-SNARE interactions [12]. The interaction of the Sec6 subunit of the exocyst with Sec9 also prevents the forma- tion of the Sec9–Sso1 t-SNARE complex before the assem- bly of the exocyst complex and recruitment of Sec1. This role of MTCs may be a general mechanism that inhibits promiscuous assembly of non-functional or premature SNARE complexes in biological membranes. In vitro stud- ies suggest that SM proteins may also prevent the assem- bly of non-fusogenic SNAREs; the interaction of Sed5p (Stx5 in mammals) with Sly1p, for example, prevents its interactions with non-physiological SNAREs [39]. In addition to SNARE gathering, tethering factors can stabilize SNARE proteins or SNARE complexes and can thereby influence SNAREpin assembly. The Cog6 subunit, for example, interacts directly with and stabilizes Stx6. Depletion of theCog6 subunit leads to abnormal proteasomal degradation of Stx6, thus impairing the assembly of Stx6– Stx16–Vti1a–VAMP4 SNARE complex and consequently endosome-to-Golgiretrograde trafficking [71]. Abnormal pro- teasomal degradation of the Golgi SNAREs GS28 and GS15 wasalsoobservedincellsdepletedofotherCOGsubunits[83] anda directinteractionbetween GS28 andCog4 was recently observed [12]. These direct interactions between MTCs and SNAREs might induce conformational changes in the SNAREs that could protect them from degradation as was previously proposed for SM proteins. Sly1, for example, directly interacts with the ER t-SNARE Ufe1p, a yeast homolog of Stx18, and protects it from ERAD degradation [84],whereasVps45pinteractswithTlg2p,theyeasthomolog of Stx16, and protects it from proteasomal degradation [85]. The HOPS complex employs a different mechanism to stabilize trans-SNARE complexes: it inhibits their disas- sembly by Sec17p/Sec18p (SNAP/NSF) [86]. The HOPS complex has affinity for multiple vacuolar SNAREs and can facilitate the formation of trans-SNARE complexes. Its ability to interact with both t- and v-SNAREs (Figure 3B) possibly inhibits the access of Sec17p and/or Sec18p and consequently SNAREpin disassembly [87]. As discussed, tethers can induce SNARE gathering through multivalent interactions, stabilize SNAREs or SNARE complexes, or activate SNARE complex assembly possibly through interaction with SM proteins [11,40]. However, it appears that SNARE–tether interactions have bidirectional effects and, in some cases, the SNAREs influ- ence the localization of the tethers. SNAREs, for instance, can enhance the binding of HOPS to liposomes and possi- bly stabilize the interaction of HOPS with the vacuole [88]. Likewise, the interaction of the DSL1 complex with ER SNAREs is possibly required for its membrane association, because DSL1 does not interact with the Rab small GTPases that generally mediate membrane recruitment [64,89]. Strikingly, DSL1 also interacts directly with COP- I subunits [90] and HOPS interacts with the Apl5 subunit of the AP-3 coat [91]. These interactions with both coats and SNAREs are possibly required for coupling tethering with fusion events. Concluding remarks Studies over the past few years strongly suggest that tethering factors do not simply bridge two membrane compartments, but rather are actively involved in fusion and influence fusion specificity, efficiency, and speed. A network of protein–protein interactions between tethers, SNAREs, and/or SM proteins is the molecular basis for their critical roles in SNAREpin assembly and conse- quently membrane fusion. The ability of tethers to inter- act with multiple components of the transport machinery, either simultaneously or sequentially, enables them spa- tially to regulate fusion events at specific membrane domains. Multiple tethering factors exist in eukaryotic cells; some regulate similar transport routes and even the assembly of the same SNARE complexes. It remains un- clear how these tethers function coordinately and why there are so many factors with potentially overlapping functions. For example, both GARP and COG regulate the assembly of the Stx6–Stx16–Vti1a–VAMP4 SNARE com- plex and consequently endosome-to-TGN retrograde transport. In light of the described studies, it could be that different tethers regulate the assembly of similar SNARE complexes but concomitantly prevent the assem- bly of distinct SNARE complexes or vice versa. This might depend on the temporal localization of the tethering fac- tors and their ability to interact with other cellular SNAREs. Other aspects including the affinity of tethers to their interacting SNAREs, the avidity of binding, or even post-translational modifications in tethers and/or SNAREs can influence SNAREpin assembly and/or its disassembly. Collectively, these different aspects could dynamically influence SNAREpin assembly and tempo- rally and spatially control membrane-fusion events. Fur- ther in vitro and in vivo studies on tether–SNARE–SM interactions will shed light on the molecular mechanisms that regulate vesicular fusion. Acknowledgments S.L. is the incumbent of the Joyce and Ben B. Eisenberg Chair of Molecular Biology and Cancer Research. This work was supported by the Binational Science Foundation (BSF), Grant No. 2011404. References 1 Chen, Y.A. and Scheller, R.H. (2001) SNARE-mediated membrane fusion. Nat. Rev. Mol. Cell Biol. 2, 98–106 Review Trends in Cell Biology January 2014, Vol. 24, No. 1 41
  • 8. 2 Hanson, P.I. et al. (1997) Neurotransmitter release – four years of SNARE complexes. Curr. Opin. Neurobiol. 7, 310–315 3 Sollner, T. et al. (1993) SNAP receptors implicated in vesicle targeting and fusion. Nature 362, 318–324 4 Jahn, R. et al. (2003) Membrane fusion. Cell 112, 519–533 5 Sollner, T.H. (2003) Regulated exocytosis and SNARE function (review). Mol. Membr. Biol. 20, 209–220 6 Hong, W. (2005) SNAREs and traffic. Biochim. Biophys. Acta 1744, 120–144 7 Malsam, J. et al. (2008) Membrane fusion: SNAREs and regulation. Cell. Mol. Life Sci. 65, 2814–2832 8 Parlati, F. et al. (2002) Distinct SNARE complexes mediating membrane fusion in Golgi transport based on combinatorial specificity. Proc. Natl. Acad. Sci. U.S.A. 99, 5424–5429 9 Parlati, F. et al. (2000) Topological restriction of SNARE-dependent membrane fusion. Nature 407, 194–198 10 Sudhof, T.C. and Rothman, J.E. (2009) Membrane fusion: grappling with SNARE and SM proteins. Science 323, 474–477 11 Shen, J. et al. (2007) Selective activation of cognate SNAREpins by Sec1/Munc18 proteins. Cell 128, 183–195 12 Laufman, O. et al. (2013) The COG complex interacts with multiple Golgi SNAREs and enhances fusogenic SNARE complexes assembly. J. Cell Sci. 126, 1506–1516 13 Shorter, J. et al. (2002) Sequential tethering of Golgins and catalysis of SNAREpin assembly by the vesicle-tethering protein p115. J. Cell Biol. 157, 45–62 14 Kraynack, B.A. et al. (2005) Dsl1p, Tip20p, and the novel Dsl3(Sec39) protein are required for the stability of the Q/t-SNARE complex at the endoplasmic reticulum in yeast. Mol. Biol. Cell 16, 3963–3977 15 Sztul, E. and Lupashin, V. (2006) Role of tethering factors in secretory membrane traffic. Am. J. Physiol. Cell Physiol. 290, C11–C26 16 Lupashin, V. and Sztul, E. (2005) Golgi tethering factors. Biochim. Biophys. Acta 1744, 325–339 17 Cai, H. et al. (2007) Coats, tethers, Rabs, and SNAREs work together to mediate the intracellular destination of a transport vesicle. Dev. Cell 12, 671–682 18 Jahn, R. and Scheller, R.H. (2006) SNAREs – engines for membrane fusion. Nat. Rev. Mol. Cell Biol. 7, 631–643 19 Holt, M. et al. (2006) Identification of SNAP-47, a novel Qbc-SNARE with ubiquitous expression. J. Biol. Chem. 281, 17076–17083 20 Weimbs, T. et al. (1997) A conserved domain is present in different families of vesicular fusion proteins: a new superfamily. Proc. Natl. Acad. Sci. U.S.A. 94, 3046–3051 21 Sutton, R.B. et al. (1998) Crystal structure of a SNARE complex involved in synaptic exocytosis at 2.4 A resolution. Nature 395, 347–353 22 Fasshauer, D. et al. (1998) Conserved structural features of the synaptic fusion complex: SNARE proteins reclassified as Q- and R- SNAREs. Proc. Natl. Acad. Sci. U.S.A. 95, 15781–15786 23 Fukasawa, M. et al. (2004) Localization and activity of the SNARE Ykt6 determined by its regulatory domain and palmitoylation. Proc. Natl. Acad. Sci. U.S.A. 101, 4815–4820 24 Misura, K.M. et al. (2000) Three-dimensional structure of the neuronal-Sec1–syntaxin 1a complex. Nature 404, 355–362 25 Schafer, I.B. et al. (2012) The binding of Varp to VAMP7 traps VAMP7 in a closed, fusogenically inactive conformation. Nat. Struct. Mol. Biol. 19, 1300–1309 26 Zeng, Q. et al. (2003) The cytoplasmic domain of Vamp4 and Vamp5 is responsible for their correct subcellular targeting: the N-terminal extension of VAMP4 contains a dominant autonomous targeting signal for the trans-Golgi network. J. Biol. Chem. 278, 23046–23054 27 Tran, T.H. et al. (2007) VAMP4 cycles from the cell surface to the trans-Golgi network via sorting and recycling endosomes. J. Cell Sci. 120, 1028–1041 28 Malsam, J. and Sollner, T.H. (2013) Organization of SNAREs within the Golgi stack. Cold Spring Harb. Perspect. Biol. 3, a005249 29 Gallwitz, D. and Jahn, R. (2003) The riddle of the Sec1/Munc-18 proteins – new twists added to their interactions with SNAREs. Trends Biochem. Sci. 28, 113–116 30 Scott, B.L. et al. (2004) Sec1p directly stimulates SNARE-mediated membrane fusion in vitro. J. Cell Biol. 167, 75–85 31 Rodkey, T.L. et al. (2008) Munc18a scaffolds SNARE assembly to promote membrane fusion. Mol. Biol. Cell 19, 5422–5434 32 Pieren, M. et al. (2010) The SM protein Vps33 and the t-SNARE H(abc) domain promote fusion pore opening. Nat. Struct. Mol. Biol. 17, 710–717 33 Yamaguchi, T. et al. (2002) Sly1 binds to Golgi and ER syntaxins via a conserved N-terminal peptide motif. Dev. Cell 2, 295–305 34 Dulubova, I. et al. (2002) How Tlg2p/syntaxin 16 ‘snares’ Vps45. EMBO J. 21, 3620–3631 35 Carpp, L.N. et al. (2006) The Sec1p/Munc18 protein Vps45p binds its cognate SNARE proteins via two distinct modes. J. Cell Biol. 173, 927–936 36 Carr, C.M. et al. (1999) Sec1p binds to SNARE complexes and concentrates at sites of secretion. J. Cell Biol. 146, 333–344 37 Yang, B. et al. (2000) nSec1 binds a closed conformation of syntaxin1A. J. Cell Biol. 148, 247–252 38 Dulubova, I. et al. (1999) A conformational switch in syntaxin during exocytosis: role of Munc18. EMBO J. 18, 4372–4382 39 Peng, R. and Gallwitz, D. (2002) Sly1 protein bound to Golgi syntaxin Sed5p allows assembly and contributes to specificity of SNARE fusion complexes. J. Cell Biol. 157, 645–655 40 Rizo, J. and Sudhof, T.C. (2012) The membrane fusion enigma: SNAREs, Sec1/Munc18 proteins, and their accomplices – guilty as charged? Annu. Rev. Cell Dev. Biol. 28, 279–308 41 Jahn, R. and Fasshauer, D. (2012) Molecular machines governing exocytosis of synaptic vesicles. Nature 490, 201–207 42 Brocker, C. et al. (2010) Multisubunit tethering complexes and their role in membrane fusion. Curr. Biol. 20, R943–R952 43 Whyte, J.R. and Munro, S. (2002) Vesicle tethering complexes in membrane traffic. J. Cell Sci. 115, 2627–2637 44 Gillingham, A.K. and Munro, S. (2003) Long coiled-coil proteins and membrane traffic. Biochim. Biophys. Acta 1641, 71–85 45 Yu, I.M. and Hughson, F.M. (2010) Tethering factors as organizers of intracellular vesicular traffic. Annu. Rev. Cell Dev. Biol. 26, 137–156 46 Barr, F.A. and Short, B. (2003) Golgins in the structure and dynamics of the Golgi apparatus. Curr. Opin. Cell Biol. 15, 405–413 47 Peterson, M.R. and Emr, S.D. (2001) The class C Vps complex functions at multiple stages of the vacuolar transport pathway. Traffic 2, 476–486 48 Jones, S. et al. (2000) The TRAPP complex is a nucleotide exchanger for Ypt1 and Ypt31/32. Mol. Biol. Cell 11, 4403–4411 49 Sacher, M. et al. (2001) TRAPP I implicated in the specificity of tethering in ER-to-Golgi transport. Mol. Cell 7, 433–442 50 Cai, H. et al. (2007) TRAPPI tethers COPII vesicles by binding the coat subunit Sec23. Nature 445, 941–944 51 Angers, C.G. and Merz, A.J. (2011) New links between vesicle coats and Rab-mediated vesicle targeting. Semin. Cell Dev. Biol. 22, 18–26 52 Balderhaar, H.J. and Ungermann, C. (2013) CORVET and HOPS tethering complexes – coordinators of endosome and lysosome fusion. J. Cell Sci. 126, 1307–1316 53 VanRheenen, S.M. et al. (1998) Sec35p, a novel peripheral membrane protein, is required for ER to Golgi vesicle docking. J. Cell Biol. 141, 1107–1119 54 VanRheenen, S.M. et al. (1999) Sec34p, a protein required for vesicle tethering to the yeast Golgi apparatus, is in a complex with Sec35p. J. Cell Biol. 147, 729–742 55 Miller, V.J. et al. (2013) Molecular insights into vesicle tethering at the Golgi by the conserved oligomeric Golgi (COG) complex and the golgin TATA element modulatory factor (TMF). J. Biol. Chem. 288, 4229–4240 56 Suvorova, E.S. et al. (2002) The Sec34/Sec35p complex, a Ypt1p effector required for retrograde intra-Golgi trafficking, interacts with Golgi SNAREs and COPI vesicle coat proteins. J. Cell Biol. 157, 631–643 57 Bonifacino, J.S. and Hierro, A. (2011) Transport according to GARP: receiving retrograde cargo at the trans-Golgi network. Trends Cell Biol. 21, 159–167 58 Heider, M.R. and Munson, M. (2012) Exorcising the exocyst complex. Traffic 13, 898–907 59 Oka, T. and Krieger, M. (2005) Multi-component protein complexes and Golgi membrane trafficking. J. Biochem. 137, 109–114 60 Waters, M.G. and Pfeffer, S.R. (1999) Membrane tethering in intracellular transport. Curr. Opin. Cell Biol. 11, 453–459 61 Sohda, M. et al. (2007) The interaction of two tethering factors, p115 and COG complex, is required for Golgi integrity. Traffic 8, 270–284 Review Trends in Cell Biology January 2014, Vol. 24, No. 1 42
  • 9. 62 Sohda, M. et al. (2010) Interaction of Golgin-84 with the COG complex mediates the intra-Golgi retrograde transport. Traffic 11, 1552–1566 63 Simonsen, A. et al. (1999) The Rab5 effector EEA1 interacts directly with Syntaxin-6. J. Biol. Chem. 274, 28857–28860 64 Ren, Y. et al. (2009) A structure-based mechanism for vesicle capture by the multisubunit tethering complex Dsl1. Cell 139, 1119–1129 65 Tripathi, A. et al. (2009) Structural characterization of Tip20p and Dsl1p, subunits of the Dsl1p vesicle tethering complex. Nat. Struct. Mol. Biol. 16, 114–123 66 Conibear, E. et al. (2003) Vps51p mediates the association of the GARP (Vps52/53/54) complex with the late Golgi t-SNARE Tlg1p. Mol. Biol. Cell 14, 1610–1623 67 Siniossoglou, S. and Pelham, H.R. (2002) Vps51p links the VFT complex to the SNARE Tlg1p. J. Biol. Chem. 277, 48318–48324 68 Perez-Victoria, F.J. and Bonifacino, J.S. (2009) Dual roles of the mammalian GARP complex in tethering and SNARE complex assembly at the trans-Golgi network. Mol. Cell. Biol. 29, 5251–5263 69 Perez-Victoria, F.J. et al. (2010) Ang2/fat-free is a conserved subunit of the Golgi-associated retrograde protein complex. Mol. Biol. Cell 21, 3386–3395 70 Shestakova, A. et al. (2007) Interaction of the conserved oligomeric Golgi complex with t-SNARE Syntaxin5a/Sed5 enhances intra-Golgi SNARE complex stability. J. Cell Biol. 179, 1179–1192 71 Laufman, O. et al. (2011) The COG complex interacts directly with Syntaxin 6 and positively regulates endosome-to-TGN retrograde transport. J. Cell Biol. 194, 459–472 72 Whyte, J.R. and Munro, S. (2001) The Sec34/35 Golgi transport complex is related to the exocyst, defining a family of complexes involved in multiple steps of membrane traffic. Dev. Cell 1, 527–537 73 Rieder, S.E. and Emr, S.D. (1997) A novel RING finger protein complex essential for a late step in protein transport to the yeast vacuole. Mol. Biol. Cell 8, 2307–2327 74 Subramanian, S. et al. (2004) The Sec1/Munc18 protein, Vps33p, functions at the endosome and the vacuole of Saccharomyces cerevisiae. Mol. Biol. Cell 15, 2593–2605 75 Mima, J. et al. (2008) Reconstituted membrane fusion requires regulatory lipids, SNAREs and synergistic SNARE chaperones. EMBO J. 27, 2031–2042 76 Stroupe, C. et al. (2006) Purification of active HOPS complex reveals its affinities for phosphoinositides and the SNARE Vam7p. EMBO J. 25, 1579–1589 77 Kramer, L. and Ungermann, C. (2011) HOPS drives vacuole fusion by binding the vacuolar SNARE complex and the Vam7 PX domain via two distinct sites. Mol. Biol. Cell 22, 2601–2611 78 Lobingier, B.T. and Merz, A.J. (2012) Sec1/Munc18 protein Vps33 binds to SNARE domains and the quaternary SNARE complex. Mol. Biol. Cell 23, 4611–4622 79 Sivaram, M.V. et al. (2005) Dimerization of the exocyst protein Sec6p and its interaction with the t-SNARE Sec9p. Biochemistry 44, 6302– 6311 80 Morgera, F. et al. (2012) Regulation of exocytosis by the exocyst subunit Sec6 and the SM protein Sec1. Mol. Biol. Cell 23, 337–346 81 Munson, M. et al. (2000) Interactions within the yeast t-SNARE Sso1p that control SNARE complex assembly. Nat. Struct. Biol. 7, 894–902 82 Laufman, O. et al. (2009) Direct interaction between the COG complex and the SM protein, Sly1, is required for Golgi SNARE pairing. EMBO J. 28, 2006–2017 83 Oka, T. et al. (2004) The COG and COPI complexes interact to control the abundance of GEARs, a subset of Golgi integral membrane proteins. Mol. Biol. Cell 15, 2423–2435 84 Braun, S. and Jentsch, S. (2007) SM-protein-controlled ER-associated degradation discriminates between different SNAREs. EMBO Rep. 8, 1176–1182 85 Bryant, N.J. and James, D.E. (2001) Vps45p stabilizes the syntaxin homologue Tlg2p and positively regulates SNARE complex formation. EMBO J. 20, 3380–3388 86 Xu, H. et al. (2010) HOPS prevents the disassembly of trans-SNARE complexes by Sec17p/Sec18p during membrane fusion. EMBO J. 29, 1948–1960 87 Collins, K.M. et al. (2005) Sec17p and HOPS, in distinct SNARE complexes, mediate SNARE complex disruption or assembly for fusion. EMBO J. 24, 1775–1786 88 Stroupe, C. et al. (2009) Minimal membrane docking requirements revealed by reconstitution of Rab GTPase-dependent membrane fusion from purified components. Proc. Natl. Acad. Sci. U.S.A. 106, 17626–17633 89 Meiringer, C.T. et al. (2011) The Dsl1 protein tethering complex is a resident endoplasmic reticulum complex, which interacts with five soluble NSF (N-ethylmaleimide-sensitive factor) attachment protein receptors (SNAREs): implications for fusion and fusion regulation. J. Biol. Chem. 286, 25039–25046 90 Andag, U. et al. (2001) The coatomer-interacting protein Dsl1p is required for Golgi-to-endoplasmic reticulum retrieval in yeast. J. Biol. Chem. 276, 39150–39160 91 Angers, C.G. and Merz, A.J. (2009) HOPS interacts with Apl5 at the vacuole membrane and is required for consumption of AP-3 transport vesicles. Mol. Biol. Cell 20, 4563–4574 92 Perez-Victoria, F.J. et al. (2010) Structural basis for the wobbler mouse neurodegenerative disorder caused by mutation in the Vps54 subunit of the GARP complex. Proc. Natl. Acad. Sci. U.S.A. 107, 12860–12865 93 Dennis, J.S. and Citron, B.A. (2009) Wobbler mice modeling motor neuron disease display elevated transactive response DNA binding protein. Neuroscience 158, 745–750 94 Gissen, P. et al. (2004) Mutations in VPS33B, encoding a regulator of SNARE-dependent membrane fusion, cause arthrogryposis–renal dysfunction–cholestasis (ARC) syndrome. Nat. Genet. 36, 400–404 95 Laufman, O. et al. (2013) Deficiency of the Cog8 subunit in normal and CDG-derived cells impairs the assembly of the COG and Golgi SNARE complexes. Traffic 14, 1065–1077 96 Jaeken, J. and Matthijs, G. (2007) Congenital disorders of glycosylation: a rapidly expanding disease family. Annu. Rev. Genomics Hum. Genet. 8, 261–278 97 Wu, X. et al. (2004) Mutation of the COG complex subunit gene COG7 causes a lethal congenital disorder. Nat. Med. 10, 518–523 98 Kranz, C. et al. (2007) COG8 deficiency causes new congenital disorder of glycosylation type IIh. Hum. Mol. Genet. 16, 731–741 99 Foulquier, F. et al. (2007) A new inborn error of glycosylation due to a Cog8 deficiency reveals a critical role for the Cog1–Cog8 interaction in COG complex formation. Hum. Mol. Genet. 16, 717–730 100 Foulquier, F. et al. (2006) Conserved oligomeric Golgi complex subunit 1 deficiency reveals a previously uncharacterized congenital disorder of glycosylation type II. Proc. Natl. Acad. Sci. U.S.A. 103, 3764–3769 101 Foulquier, F. (2009) COG defects, birth and rise! Biochim. Biophys. Acta 1792, 896–902 102 Freeze, H.H. and Ng, B.G. (2011) Golgi glycosylation and human inherited diseases. Cold Spring Harb. Perspect. Biol. 3, a005371 103 Ungar, D. et al. (2006) Retrograde transport on the COG railway. Trends Cell Biol. 16, 113–120 104 He, B. and Guo, W. (2009) The exocyst complex in polarized exocytosis. Curr. Opin. Cell Biol. 21, 537–542 Review Trends in Cell Biology January 2014, Vol. 24, No. 1 43