ArticlePDF Available

Two human ARFGAPs associated with COP-I-coated vesicles

Authors:

Abstract and Figures

ADP-ribosylation factors (ARFs) are critical regulators of vesicular trafficking pathways and act at multiple intracellular sites. ADP-ribosylation factor-GTPase-activating proteins (ARFGAPs) are proposed to contribute to site-specific regulation. In yeast, two distinct proteins, Glo3p and Gcs1p, together provide overlapping, essential ARFGAP function required for coat protein (COP)-I-dependent trafficking. In mammalian cells, only the Gcs1p orthologue, named ARFGAP1, has been characterized in detail. However, Glo3p is known to make the stronger contribution to COP I traffic in yeast. Here, based on a conserved signature motif close to the carboxy terminus, we identify ARFGAP2 and ARFGAP3 as the human orthologues of yeast Glo3p. By immunofluorescence (IF), ARFGAP2 and ARFGAP3 are closely colocalized with coatomer subunits in NRK cells in the Golgi complex and peripheral punctate structures. In contrast to ARFGAP1, both ARFGAP2 and ARFGAP3 are associated with COP-I-coated vesicles generated from Golgi membranes in the presence of GTP-gamma-S in vitro. ARFGAP2 lacking its zinc finger domain directly binds to coatomer. Expression of this truncated mutant (DeltaN-ARFGAP2) inhibits COP-I-dependent Golgi-to-endoplasmic reticulum transport of cholera toxin (CTX-K63) in vivo. Silencing of ARFGAP1 or a combination of ARFGAP2 and ARFGAP3 in HeLa cells does not decrease cell viability. However, silencing all three ARFGAPs causes cell death. Our data provide strong evidence that ARFGAP2 and ARFGAP3 function in COP I traffic.
Content may be subject to copyright.
#2007 The Authors
Journal compilation #2007 Blackwell Publishing Ltd
doi: 10.1111/j.1600-0854.2007.00631.x
Traffic 2007; 8: 1644–1655
Blackwell Munksgaard
Two Human ARFGAPs Associated with
COP-I-Coated Vesicles
Gabriella Frigerio
1,2
, Neil Grimsey
1
,
Martin Dale
1
, Irina Majoul
1,3
and
Rainer Duden
1,3,
*
1
Department of Clinical Biochemistry, Cambridge
Institute for Medical Research, University of Cambridge,
Hills Road, Cambridge CB2 2XY, United Kingdom
2
European Bioinformatics Institute, Wellcome Trust
Genome Campus, Hinxton, Cambridge CB10 1SD,
United Kingdom
3
Centre for Biomedical Sciences, School of Biological
Sciences, Royal Holloway University of London,
Egham TW20 0EX, United Kingdom
*Corresponding author: Rainer Duden,
rainer.duden@rhul.ac.uk
ADP-ribosylation factors (ARFs) are critical regulators of
vesicular trafficking pathways and act at multiple intracel-
lular sites. ADP-ribosylation factor-GTPase-activating pro-
teins (ARFGAPs) are proposed to contribute to site-specific
regulation. In yeast, two distinct proteins, Glo3p and
Gcs1p, together provide overlapping, essential ARFGAP
function required for coat protein (COP)-I-dependent traf-
ficking. In mammalian cells, only the Gcs1p orthologue,
named ARFGAP1, has been characterized in detail. How-
ever, Glo3p is known to make the stronger contribution to
COP I traffic in yeast. Here, based on a conserved signature
motif close to the carboxy terminus, we identify ARFGAP2
and ARFGAP3 as the human orthologues of yeast Glo3p.
By immunofluorescence (IF), ARFGAP2 and ARFGAP3 are
closely colocalized with coatomer subunits in NRK cells in
the Golgi complex and peripheral punctate structures. In
contrast to ARFGAP1, both ARFGAP2 and ARFGAP3 are
associated with COP-I-coated vesicles generated from
Golgi membranes in the presence of GTP-g-S in vitro.
ARFGAP2 lacking its zinc finger domain directly binds to
coatomer. Expression of this truncated mutant (DN-ARF-
GAP2) inhibits COP-I-dependent Golgi-to-endoplasmic
reticulum transport of cholera toxin (CTX-K63) in vivo.
Silencing of ARFGAP1 or a combination of ARFGAP2 and
ARFGAP3 in HeLa cells does not decrease cell viability.
However, silencing all three ARFGAPs causes cell death.
Our data provide strong evidence that ARFGAP2 and
ARFGAP3 function in COP I traffic.
Key words: ARF, ARFGAP, coated vesicles, coatomer,
COP I, Golgi
Received 18 June 2007, revised and accepted for publica-
tion 24 July 2007, uncorrected manuscript published
online 27 July 2007, published online 29 August 2007
Cytoplasmic coat proteins govern the transport of proteins
between membrane-bound compartments of the secretory
and endocytic pathways by shaping the membrane of the
donor organelle into vesicular or tubular transport carriers
and selecting appropriate cargo into them (1–3). The well-
characterized minimal machinery to form coat protein (COP)-
I-coated vesicles from Golgi membranes comprises coatomer,
a stable heptameric protein complex comprising a-, b-, b’-,
g-, d-, e- and z-COP, and the small ras-like GTPase ADP-
ribosylation factor (ARF) in its GTP-bound form (4,5). The
GTP/GDP cycle of ARF proteins is regulated by guanine
nucleotide exchange factors and GTPase-activating pro-
teins (GAPs) (6). ARF inactivation by GTP hydrolysis relies
on stimulation of a low intrinsic GTPase activity by ARF-
GTPase-activating proteins (ARFGAPs). ARFGAPs form
a large family of proteins that share a conserved catalytic
domain of approximately 70 residues that includes a zinc
finger motif, but they differ in their non-catalytic domains
(for review see 6,7). In this pathway, ARFGAP activity
triggers uncoating of COP-I-coated vesicles through GTP
hydrolysis on ARF, which renders both ARF and coatomer
cytosolic (4). Interestingly, regulated GTP hydrolysis on
ARF is required not only for uncoating but also for cargo
selection into COP I vesicles because vesicles produced
in the presence of the nonhydrolysable GTP analogue,
GTP-g-S, are depleted of protein cargo (8–10).
There is good evidence that a single ARF species may have
multiple roles and been in different locations in the cell
(6,11). It is generally thought that ARFGAPs with restricted
cellular localizations will contribute to differential regula-
tion of ARF to enable it to function in this way. In yeast,
two ARFGAPs, Gcs1p and Glo3p, provide an overlapp-
ing essential function in the COP-I-mediated Golgi-to-
endoplasmic reticulum (ER) transport (12,13). Several lines
of evidence suggest that Glo3p makes the stronger
contribution to COP-I-mediated trafficking. glo3Dmutants
display a much stronger retrograde Golgi-to-ER transport
defect than gcs1Dmutants (12). Glo3p but not Gcs1p is as-
sociated with yeast COP-I-coated vesicles formed in vitro
(14). A mutant in GLO3, named ret4-1 (13), was isolated
based on a genetic selection for mutants with defects in
dilysine motif-dependent Golgi-to-ER retrieval that had
previously identified several coatomer subunits (15).
Re-use of this article is permitted in accordance with the
Creative Commons Deed, Attribution 2.5, which does not
permit commercial exploitation.
1644 www.traffic.dk
Lastly, Nakano’s laboratory has recently shown that GLO3
but not GCS1 can suppress the temperature-sensitive (ts)-
growth defect of the arf1-16 and arf1-17 mutants, which
display strong defects in retrograde trafficking for several
cargo proteins (16). These data provide compelling evi-
dence that Glo3p in yeast has a critical function in COP-I-
dependent traffic that cannot be complemented by Gcs1p.
We have previously shown that Glo3p but not Gcs1p can
interact with yeast coatomer in vitro (17,18). Using the
two-hybrid system and in vitro binding assays, we dem-
onstrated that this binding occurs via the b0- and g-COP
subunits of coatomer (17,18). All these data combined
functionally tie in Glo3p tightly with the COP I pathway.
Surprisingly, in mammalian cells only ARFGAP1, which by
sequence analysis can be clearly identified as a Gcs1p
orthologue, has been well characterized so far. ARFGAP
was the first ARF-directed GAP to be discovered (19).
ARFGAP1 shuttles between the Golgi complex and cytosol
and is involved in COP-I-dependent trafficking in vivo
(20,21). Assays with Golgi membranes and recombinant
proteins in vitro have suggested that ARFGAP1 is required
for COP I vesicle generation, and under certain in vitro
conditions ARFGAP1 can be observed as a stoichiometric
component of the COP I coat, namely in vesicles produced
in the presence of GTP rather than GTP-g-S (22,23).
Significantly, however, a Drosophila mutant that lacks the
fly orthologue of ARFGAP1 encoded by the Gap69C gene
is viable and fertile (24), consistent with the notion that it is
not an essential, key component to COP I function and that
other, presumably Glo3p-type ARFGAPs can compensate
for the loss of ARFGAP1 function in the fly. In this study,
we aimed to identify and characterize human Glo3p
orthologues.
Sixteen genes encoding ARFGAPs are found in the human
genome. Using a conserved signature motif present in
GLO3 proteins from all species, named the Glo3 motif, we
unambiguously identify ARFGAP2 and ARFGAP3 as the
two human orthologues of yeast Glo3p, and we provide
their initial cell biological characterization. ARFGAP1, ARF-
GAP2 and ARFGAP3 are coexpressed in mammalian cells
(e.g. HeLa and NRK cells). Our data strongly suggest that
ARFGAP1, ARFGAP2 and ARFGAP3 co-operate to perform
essential, overlapping functions in COP-I-mediated traf-
ficking in mammalian cells.
Results
Two human orthologues of yeast Glo3p
We wished to identify a human orthologue for the yeast
ARFGAP Glo3p, which in yeast has been demonstrated to
have a prominent role in coatomer-mediated trafficking.
Among over 16 human proteins with an ARFGAP domain,
we found human Gcs1p, which is named ARFGAP1, and
four novel proteins with a closely related ARFGAP domain.
In order to identify a true orthologue of yeast Glo3p, we
searched for proteins with sequence similarity beyond the
ARFGAP domain. Making use of a Glo3 protein from
Schizosaccharomyces pombe, we were able to identify
a small conserved signature motif, named the Glo3 motif,
found in 19 ARFGAP proteins from 14 different species.
Using this information, we found two distinct human
orthologues of yeast Glo3p, ARFGAP2 and ARFGAP3. In
summary, searching the increasing amount of data avail-
able from systematic sequencing projects, we identified
two distinct human sequence orthologues of the yeast
ARFGAP Glo3p.
The zinc finger domain and the Glo3 motif
Both Glo3p and Gcs1p share high sequence homology in
their amino terminal catalytic zinc finger domain, which is
known to engage ARF. Figure 1A shows an alignment of
Glo3p, Gcs1p and their orthologues from S. pombe,as
well as rat and human ARFGAP1, and the human ARF-
GAP2 and ARFGAP3. Note the presence of the four
conserved cysteine residues characteristic for the zinc
finger domain of ARFGAPs highlighted in yellow.
The conserved Glo3 motif, which is characteristic for Glo3
proteins from a wide variety of eukaryotic organisms
including plants, is about 45 amino acid residues in length
and is situated in the carboxy terminal part of the Glo3
proteins (see Figure 1B). It consists of two repeats of 15
residues separated by a linker of 19–23 residues, depending
on the species. Gaps immediately after some of the repeats
suggest flexibility in the exact length of the sequence in the
region (Figure 1B). Not surprisingly, the overall degree of
conservation is lowest in the parasitic amoeba Plasmodium
yoelii and the fruit fly Drosophila melanogaster.Inthese
organisms, only a single Ile-Ser-Ile tripeptide is present in
the second repeat, whereas only the first repeat is well
conserved in the nematode Caenorhabditis elegans.During
preparation of this manuscript, Nakano’s group also noted
the presence of the Glo3 motif and could demonstrate that
it is important for Glo3p function in yeast (16).
The Glo3 motif, which is always found strictly associated
with an ARFGAP domain, allowed us to identify the true
human orthologues of yeast Glo3p as ARFGAP2 and
ARFGAP3, by sequence comparison. In Figure 2, a protein
sequence alignment of yeast Glo3p, ARFGAP2 and ARF-
GAP3 is shown. ARFGAP2 is a protein of 521 residues
and had not been previously named. However, a zinc
finger protein of unknown function and designated Zfp289
had been previously identified from SCp2 mouse mam-
mary epithelial cells (25). The protein sequence of human
ARFGAP2 and mouse Zfp289 shows complete identity.
ARFGAP2 and ARFGAP3 share 49.6% protein sequence
identity. In an alignment over their entire length, Glo3p
shares only 17.6 and 19.9% identity with ARFGAP2 and
ARFGAP3, respectively. As for ARFGAP3 (516 residues),
both the presence of the ARFGAP domain and its function
as a GTPase-activating protein towards ARF in vitro had
Traffic 2007; 8: 1644–1655 1645
Two Human ARFGAPs Acting in COPI-Dependent Trafficking
been noted (26,27). However, neither ARFGAP2 nor ARF-
GAP3 have previously been recognized as Glo3p rather than
Gcs1p orthologues, and the localization of both ARFGAP2
and ARFGAP3 and their role in COP I transport has notbeen
investigated.
ARFGAP2 and ARFGAP3 are found in the Golgi
complex as well as in pre-Golgi punctate structures
In order to confirm that these two human Glo3p ortho-
logues are involved in COP I transport, we raised mono-
specific polyclonal antibodies against them (see Materials
and Methods) and determined their intracellular localiza-
tion. By IF studies on methanol–acetone-fixed NRK cells,
we find the majority of ARFGAP2 as well as ARFGAP3
associated with a juxtanuclear densely packed structure
typical for the mammalian Golgi complex (Figures 3 and 4).
The remaining protein is associated with small punc-
tate structures scattered throughout the cytoplasm.
Double-labelling experiments reveal that these structures
labelled with antibodies against ARFGAP2 as well as
ARFGAP3 are largely identical to those labelled with anti-
bodies against subunits of the coatomer complex, identi-
fying them as ER-Golgi intermediate compartment
(ERGIC)/vesicular-tubular compartment (VTC) structures.
To extend these results, we performed double-labelling
experiments with a number of well-established marker
proteins. The tethering protein p115 is known to be
associated with the early Golgi complex as well as with
a punctate pre-Golgi compartment (28). Although
because of its exceptional quality, the monoclonal anti-
body against p115 labels a considerably larger number of
peripheral structures, the majority of ARFGAP2- or ARF-
GAP3-positive structures are p115 positive as well.
Within the Golgi compartment, there are areas where
closer inspection of the patterns suggests that the
mammalian ARFGAP2 and ARFGAP3, and p115, respect-
ively, are localized in a closely associated but not
identical pattern. This is in clear contrast to these proteins
Figure 1: ARFGAP domain and
conserved motif of a representa-
tive subset of Glo3 proteins. A)
The N-terminal ARFGAP domains of
Glo3 and Gcs1 proteins from Sac-
charomyces cerevisiae,Schizosac-
charomyces pombe and Homo
sapiens were aligned using the CLUS-
TAL V algorithm in the MEGALIGN pro-
gram (DNASTAR package). Note the
conserved cysteine residues high-
lighted in yellow and the overall high
degree of sequence conservation in
this domain. B). The Glo3 motif
(highlighted in orange) consists of a
repeat of 15 residues separated by
11–17 residues. It is a signature
motif that allows unambiguous
identification of Glo3 orthologues.
Genbank accession numbers: cere-
visiae: 6320969 (S. cerevisiae);
Plasmodium 23478251 (P. yoelii);
worm: 25153991 (Caenorhabditis
elegans); fly: 24668642 (Drosophila
melanogaster); plant A: 7487780,
plant B: 18403775, plant C 15237500
(Arabidopsis thaliana); fish: assem-
bled from several ESTs: 12148139,
12171549, 12158629 and 12265392
(Danio rerio); frog: 27695479 (Xenopus
laevis) and human A: 21263420 and
human B: 31543983 (H. sapiens).
1646 Traffic 2007; 8: 1644–1655
Frigerio et al.
in comparison with subunits of the coatomer complex,
where most features within the Golgi complex are
labelled in a remarkably identical way.
Again, this is not the case for ARFGAP2 and ARFGAP3
when compared with the well-characterized early Golgi
marker GM130 (29) (see Figures 3 and 4). In this case, the
proteins are localized in a closely associated but not
identical fashion in the region of the Golgi complex. As
a control, we analysed the localization of ARFGAP2 and
ARFGAP3 in comparison to g-adaptin, a subunit of the
clathrin adaptor complex AP-1, which is involved in traffic
between the trans Golgi network (TGN) and endosomal
compartments (30). As expected, we found no significant
overlap in the extensive punctate peripheral label as well
as the juxtanuclear label associated with the Golgi complex
(Figures 3 and 4). We conclude that the localization of
ARFGAP2 and ARFGAP3 is consistent with a function in
COP I trafficking.
ARFGAP2 and ARFGAP3 are associated with
COP-I-coated vesicles generated in vitro
We next wished to test whether ARFGAP2 and ARFGAP3
are associated with COP I vesicles produced in vitro. For
this we employed the ‘classic’ budding assay developed by
the Rothman/Wieland labs (31,32), using purified rat liver
Golgi and pig brain cytosol. The budding reaction was
performed in the presence of the nonhydrolysable GTP
analogue, GTP-g-S, which locks ARF on the membrane of
vesicles and thus prevents uncoating. Vesicles and Golgi
donor membranes were separated on a linear sucrose
Figure 2: Sequence alignment of
yeast Glo3p and human ARF-
GAP2 and ARFGAP3. ARFGAP2
and ARFGAP3 share 49.6% protein
sequence identity. ARFGAP2 and
ARFGAP3 share 17.6 and 19.9%
sequence identity with Glo3p,
respectively.
Traffic 2007; 8: 1644–1655 1647
Two Human ARFGAPs Acting in COPI-Dependent Trafficking
gradient by overnight centrifugation (see Materials and
Methods, and 31), and proteins in the fractions obtained
were analysed by immunoblotting and silver staining of
bands after SDS–PAGE (Figure 5). The characteristic set of
coatomer bands (a-, b’-, b-, g- and d-COP) were found
enriched in the expected positions for COP-I-coated
vesicles in the gradient (fractions 8 þ9; corresponding to
40–43% sucrose) where also the blot signals for g-COP
(Figure 5A,B) and b-COP (data not shown) showed a
corresponding major peak. ARFGAP1 was predominantly
detected in the donor Golgi fractions, but only small
amounts were found in the fractions containing COP I
vesicles (Figure 5B). This is consistent with the previously
reported findings from the Hsu lab that ARFGAP1 is
depleted from COP I vesicles formed in the presence of
GTP-g-S (23). On the other hand, ARFGAP2 and ARFGAP3
showed a strong peak in the COP I vesicle fractions
(Figure 5B), indicating that these novel Glo3-type ARFGAPs
can be actively recruited into budding COP I vesicles even in
the presence of GTP-g-S. Clathrin heavy chain and the
g-subunit of the AP-1 adaptor complex used as controls
were absent from the COP I vesicle fractions, as expected.
We find that the dilysine motif-bearing protein ERGIC-53
was also not included into the COP I vesicles. ADP-
ribosylation factor-1, which is involved in many different
transport steps within the Golgi complex, was found
both in the donor Golgi fractions and in the COP I vesicle
fractions, as expected. Our data show that both novel
ARFGAPs, ARFGAP2 and ARFGAP3 are associated with
the COP-I-coated vesicles produced in vitro in the
presence of GTPgS, whereas ARFGAP1 is not or much
less so.
Figure 3: ARFGAP2 colocalizes with coatomer in NRK cells. Methanol–acetone-fixed NRK cells were stained with affinity-purified
ARFGAP2 antibodies and monoclonal antibodies against b-COP, p115, GM130 or g-adaptin. Cy3-coupled anti-rabbit and Alexa488-coupled
anti-mouse secondary antibodies were used. Images were acquired using a Zeiss Axioplan Fluorescent Microscope equipped with a charge-
coupled device camera. The individual channels are shown in monochrome; the composite overlay is shown to the right (green channel,
ARFGAP2; red channel, b-COP, p115, GM130 and gAP, respectively). Bar ¼10 mm.
1648 Traffic 2007; 8: 1644–1655
Frigerio et al.
The catalytic domain on Glo3p or ARFGAP2 is not
required for interaction with coatomer
Yeast Glo3p interacts with g-COP as well as b0-COP in the
two-hybrid system (17). This strong, direct interaction was
confirmed by pull-down experiments using recombinant
His-tagged Glo3p and yeast cytosol (17). Here we show
that the catalytic domain of yeast Glo3p is not required
for this direct interaction with coatomer. Deletion of the
N-terminal 96 amino acids including the Zn finger domain do es
not reduce the interaction of tagged Glo3p with coatomer in
vitro (Figure 6A). In this type of experiment, Gcs1p does not
bind coatomer above background levels defined by recombin-
ant GTP dissociation inhibitor (GDI) as a negative control
(Figure 6A). In order to test whether mammalian ARFGAP2
interacts with coatomer in a way comparable to yeast Glo3p,
glutathione S-transferase (GST)-tagged ARFGAP2 lacking its
ARFGAP domain was used for pull-down experiments from
rat liver cytosol. Indeed, coatomer binding from rat liver
cytosol to GST-tagged ARFGAP2 present on glutathione
beads was readily detectable by silver stain (Figure 6B),
and immunoblots using anti-peptide antibodies against a-
and b-COP demonstrated enrichment of these coatomer
subunits (data not shown). Thus, the catalytic domain of
Glo3-type ARFGAP proteins from yeast and mammals is not
required for in vitro interaction with coatomer.
Expression of DN-ARFGAP2–cyan fluorescent protein
inhibits CTX transport
Full-length ARFGAP2 was tagged at the carboxy terminus
with cyan fluorescent protein (CFP) and expressed at low
Figure 4: ARFGAP3 colocalizes with coatomer. Methanol–acetone fixed NRK cells were labelled with affinity-purified ARFGAP3
antibodies and mAbs against b-COP, p115, GM130 or g-adaptin (see Figure 3 for other details). The individual channels are shown in
monochrome; the composite overlay is shown to the right (green channel, ARFGAP3; red channel, b-COP, p115, GM130 and gAP,
respectively). Bar ¼10 mm.
Traffic 2007; 8: 1644–1655 1649
Two Human ARFGAPs Acting in COPI-Dependent Trafficking
level in Vero cells. This CFP fusion protein localized to the
Golgi complex [as identified using the Golgi enzyme mar-
ker galactosyl transferase tagged with yellow fluorescent
protein (GalT-YFP)] and also localized to punctate structures
scattered through the cytoplasm (Figure 7, upper panel).
This localization pattern is reminiscent of the localization
observed with antibodies against ARFGAP2 (compare with
Figure 3).
To assay COP-I-dependent transport in vivo, we employed
a model cargo protein previously described by us (21,33),
the non-toxic mutant version of cholera toxin fluorescently
labelled with the dye Cy3. After 3 h of internalization, CTX-
K63-Cy3 had been endocytosed from the plasma mem-
brane and was prominently present in the Golgi complex
and in the ER network (Figure 7, upper panel). In cells
expressing higher levels of ARFGAP2–CFP, a strong cyto-
plasmic staining appeared in addition to the Golgi pattern,
perhaps suggesting a limited number of binding sites on
the Golgi. Importantly, however, transport of CTX-K63-Cy3
still proceeded with kinetics very similar to control cells
(data not shown).
To test whether ARFGAP2 may be involved in COP-I-
dependent transport in vivo, we generated a DN-ARFGAP2–
CFP mutant. At low expression level, this protein was
detected at the Golgi complex when expressed in Vero
cells (not shown). At elevated expression levels (12 h after
transfection), a strong cytoplasmic pattern was seen in
addition to the Golgi pattern (Figure 7, middle panel).
Importantly, in such cells, 3 h after addition, CTX-K63-Cy3
was not observed in the ER but remained restricted to the
Golgi complex and punctate structures scattered through
the cytoplasm (Figure 7, middle panel). These data indi-
cate that expression of DN-ARFGAP2–CFP interferes with
COP-I-dependent transport of CTX-K63-Cy3 to the ER.
At even longer time-points after transfection (16 h),
cells expressing DN-ARFGAP2–CFP displayed a strikingly
higher frequency of having two or even three nuclei (indi-
cated by numbers in Figure 8, lower panel). In such cells, as
judged by GalT-YFP, usually only one large Golgi complex
was present to which DN-ARFGAP2–CFP localized (Fig-
ure 7, lower panel). In such cells, transport of CTX-K63-Cy3
was again severely inhibited, with the toxin not arriving at
the ER network even after 3 h. Instead, CTX-K63-Cy3 was
present in the Golgi complex and large cytoplasmic struc-
tures (Figure 7, lower panel). Our data strongly suggest that
ARFGAP2 is involved in the COP-I-dependent trafficking
of cholera toxin from the Golgi to the ER.
Silencing of all three ARFGAPs is lethal
In order to further investigate the role of ARFGAP2 and
ARFGAP3 in COP-I-mediated transport, we analysed NRK
and HeLa cells upon knock-down of individual ARFGAP
transcripts. Protein levels were monitored by immunoblots
of total cell extracts from NRK cells 72 h after transfection
with appropriate small interfering RNA oligonucleotides
(siRNAs). As shown in Figure 8, we indeed found oligos
that allowed knock-down of the individual ARFGAPs in
Figure 5: ARFGAP2 and ARFGAP3 are
associated with COP I vesicles gener-
ated in vitro.Vesicle budding reactions
were performed according to the protocol
developed by (31), using the non-hydro-
lysable GTP analogue, GTP-g-S. Vesicles
produced in the reaction were separated
from the Golgi donor membranes on
sucrose density gradients and peaked at
40–43% sucrose. Fractions were analysed
by silver stain (upper panel) and immuno-
blotting with antibodies as indicated
(lower panel). The positions of coatomer
bands are indicated on the left and also by
asterisks. Note the abundant presence of
ARFGAP2 and ARFGAP3 in the vesicle
fractions defined by the presence of coat-
omer subunits (fractions 8 þ9) but the
absence of ARFGAP1.
1650 Traffic 2007; 8: 1644–1655
Frigerio et al.
transfections with a single RNAi oligo. Antibodies against
g-COP and b-tubulin were used to confirm equal loading of
the samples. In yeast, deletion of either the GLO3 or GCS1
gene results in a relatively mild phenotype. However,
deletion of both genes is lethal. Therefore, we tested the
effect on cell survival of an RNAi-oligo-mediated knock-
down of ARFGAP1, ARFGAP2 and ARFGAP3 either in
pairs or as a combined triple knock-down (see Materials
and Methods for details). No significant cell death was
observed in single knock-downs or in any pairwise knock-
down, compared with cells transfected with a control
oligo. However, a triple knock-down of ARFGAP1, ARF-
GAP2 and ARFGAP3 in HeLa cells resulted in strongly
significant cell death. More than 75% cell death was
routinely observed after three sets of siRNA transfections
at the 72-h time-point after the last transfection. Very
similar results were observed in NRK cells (data not
shown). We conclude that at least one of the three
ARFGAPs is required to maintain cell viability.
Discussion
We wish to understand the role of ARFGAPs in the
regulation of COP-I-dependent trafficking. In yeast, the
two ARFGAPs Glo3p and Gcs1p together provide an
overlapping, essential function in COP-I-dependent mem-
brane traffic. In mammalian cells, only the Gcs1p ortho-
logue, ARFGAP1 has been characterized so far. In this
study, we provide an initial cell biological characterization
of two recently identified human ARFGAPs, ARFGAP2 and
ARFGAP3. We unambiguously demonstrate by sequence
analysis that they are the human orthologues of yeast
Glo3p, through the presence of a conserved ‘signature’
motif at the carboxy terminus, the Glo3 motif. Using novel
antibodies, we demonstrate that they are localized to the
Golgi complex and peripheral punctate structures scat-
tered throughout the cytoplasm in NRK cells. Both ARF-
GAP2 and ARFGAP3 significantly colocalize with COP I
subunits in both locations, strongly suggesting that they
are associated with COP I on both ERGIC/VTC structures
and at the Golgi complex. Here we further show that both
ARFGAP2 and ARFGAP3 are associated with COP I
vesicles produced in vitro in the presence of the non-
hydrolysable GTP analogue, GTP-g-S.
The Golgi and VTC localization of ARFGAP2 and ARFGAP3
appears to be dynamic, as treatment with brefeldin A
(BFA) rapidly renders their localization completely diffuse-
cytoplasmic within 2–5 min of adding the drug, with
kinetics very similar to those of b-COP and ARFGAP1
(unpublished data). Similarly, re-recruitment of ARFGAP2
and ARFGAP3 to Golgi membranes occurs rapidly upon
washout of the drug. Future experiments will need to
address the dynamics of ARFGAP2/3 using live cell
imaging approaches, similar to detailed analysis that has
been reported for ARFGAP1 (20).
The Nakano group has recently reported that Glo3p but not
Gcs1p can suppress arf-Ts mutants with a severe defect in
Golgi-to-ER retrieval in an allele-specific manner (16). They
found that the Glo3 motif is required for suppression of the
growth defect of arf1-16 and arf1-17, suggesting that the
motif is important for Glo3p function (16).
We had previously shown that ARFGAP2, and more
weakly ARFGAP3, can interact with recombinant carboxy
terminal g-COP ‘ear’ domain in pull-down experiments
from rat liver cytosol (18). Furthermore, we could show
this interaction is likely to be physiologically important, as
plasmid-driven overexpression of the g-COP ‘ear’ domain
in NRK cells disrupted the Golgi localization of ARFGAP2,
whereas a point mutant version of the g-COP ‘ear’ domain
(W776S) in which the binding site to ARFGAP2 is abro-
gated had no effect (18). Here we show that the catalytic
domain of ARFGAP2 and ARFGAP3 is not required for
in vitro interactions with mammalian coatomer. The same
phenomenon was observed for their yeast counterpart
Glo3p in binding experiments using yeast cytosol. It has
Figure 6: Direct interaction of yeast and human Glo3 with
coatomer. His6- or GST-tagged proteins were used to pull down
coatomer from yeast or rat liver cytosol, under conditions
described by us previously (17). A) His6-tagged Glo3p lacking its
amino terminus was compared with full-length Glo3p as well as
full-length Gcs1p in its ability to bind coatomer from yeast cytosol.
Protein bound to Ni-NTA agarose beads was resolved by SDS–
PAGE, followed by Western blotting and incubation with anti-
yeast coatomer antibodies detected by ECL. His-tagged Glo3p
lacking its ARFGAP domain (DN96) still binds coatomer from yeast
cytosol in vitro, whereas Gcs1p does not. GTP dissociation
inhibitor is used as a negative control here. B) A GST-fusion
protein harbouring DN96-ARFGAP2 was used for a pull-down from
a centrifugation-cleared TX-100 extract of pig brain crude micro-
somal membranes. Glutathione S-transferase was used as the
negative control. Note absence of binding in the negative control
and the presence of the characteristic coatomer bands in the pull-
down involving the DN96-ARFGAP2 GST fusion as the fishing
hook. Enrichment of a- and b-COP in the bound fraction was
confirmed using antipeptide antibodies (data not shown).
Traffic 2007; 8: 1644–1655 1651
Two Human ARFGAPs Acting in COPI-Dependent Trafficking
recently been shown that ARFGAP1 can also directly
interact, albeit more weakly, with coatomer. However,
this interaction involves two distinct binding sites on
ARFGAP1, one in its catalytic domain and the other in
the non-catalytic region (see 23). Using the two-hybrid
system, we were unable to detect interactions of Gcs1 or
ARFGAP1 with yeast or mammalian coatomer subunits,
respectively, whereas we observed strong interactions of
Glo3p as well as ARFGAP2 and ARFGAP3 with coatomer
subunits (17, unpublished data). We show that in the
presence of GTP-g-S, ARFGAP2 and ARFGAP3 are able
to associate with COP-I-coated vesicles whereas ARF-
GAP1 is barely detectable.
In most mammalian cells there are six ARFs, of which
ARF1 is the most extensively studied. It has been impli-
cated in Golgi-to-ER transport, function of the Golgi, trans-
port from the trans-Golgi network, transport in the
endocytic pathway and recruitment of paxillin to focal
adhesions (7). Becausein mammalian genomes the number
of proteins with an ARFGAP domain is considerably larger
than the number of ARF proteins, it has been suggested
that a number of GAPs might regulate the activities of
ARF1 in a location-dependent manner.
The observed interaction of Glo3p-type ARFGAPs, namely
Glo3p in yeast and ARFGAP2 and ARFGAP3 in mammals,
Figure 7: ARFGAP2 is involved in
COP-I-dependent trafficking in Vero
cells. Vero cells coexpressing full-length
ARFGAP2–CFP and the Golgi marker
GalT-YFP were treated with CTX-K63-
Cy3 (upper panel). After 3 h of internal-
ization, CTX-K63-Cy3, the Cy3-labelled
A-subunit, is prominently present in fine
ER structures (including the nuclear
envelope; arrow) and in the Golgi com-
plex. ARFGAP2–CFP colocalizes well
with GalT-YFP in the Golgi. Additionally,
ARFGAP2–CFP is present in scattered
punctate structures, most likely interme-
diate compartment. In contrast, in Vero
cells expressing DN-ARFGAP2–CFP2
transport of CTX-K63-Cy3 to the ER is
inhibited (middle panel). DN-ARFGAP2–
CFP2 localizes to the Golgi complex.
Vero cells overexpressing DN-ARF-
GAP2–CFP2 for extended periods often
display two to three nuclei and show
severe inhibition of toxin transport
(lower panel). The bottom panel is the
characterization of the Cy3-labelled CTX-
K63 (non-toxic AB5 holotoxin) used for
the experiments (for details see methods).
Mostly the A-subunit of CTX-K63 is
labelled, with traces of B present. Scale
bar: 10 mm.
1652 Traffic 2007; 8: 1644–1655
Frigerio et al.
with coatomer subunits (17,18; this study) may thus
enable regulation of aspects of the COP I vesicle cycle,
such as cargo sorting, coat formation, membrane defor-
mation or uncoating, which may enable regulation of traffic
at different intracellular sites and at specific intracellular
sites. It is perhaps surprising, given the strong direct
interactions of ARFGAP2 and ARFGAP3 with coatomer,
that these proteins were not previously identified in purified
COP I vesicle populations. Apparently, in isolated COP-I-
coated vesicles produced in the presence of GTP-g-S
(31; this study), neither ARFGAP2 nor ARFGAP3 are
stoichiometric coat components.
There are precedents for specific interactions of ARFGAPs
with other coat proteins. For example, it has been shown
that AGAP1 can directly interact with the Golgi/endosome-
localized AP-3 adaptor but not with the closely related AP-2
adaptor involved in endocytosis from the plasma mem-
brane (34,35). The related AGAP2 interacts specifically
with the AP-1 adaptor to regulate trafficking from recycling
endosomes (35) but not with AP-2. The functional signifi-
cance of the direct interactions of ARFGAP2 and ARFGAP3
with coatomer with regard to protein sorting into COP I
vesicles and the regulation of uncoating once the vesicles
are fully assembled needs to be addressed in future
experiments.
In yeast, Glo3p and Gcs1p provide an essential, over-
lapping function for COP-I-mediated transport (12).
However, growing evidence suggests that while Gcs1p
is able to maintain ARF1-regulated membrane traffic in
several transport steps, Age2p and Glo3p are special-
ized for a transport step out of the TGN (36) and COP-I-
mediated transport, respectively (12). Our in vivo data
demonstrate that ARFGAP2 is involved in the COP-I-
dependent Golgi-to-ER transport of a model cargo,
cholera toxin.
We show here that a combined knock-down of ARFGAP1,
ARFGAP2 and ARFGAP3 using siRNAs is lethal in HeLa
and NRK cells. Thus, our data strongly suggest that, similar
to the situation yeast, Gcs1p-type and Glo3p-type ARF-
GAPs together provide an essential function for COP-I-
mediated transport. In the future, it will be important to
analyse in detail the cellular phenotypes resulting from
RNAi-mediated gene silencing of individual ARFGAPs and
use in vivo and in vitro assays to unravel the individual
contributions of ARFGAP1, ARFGAP2 and ARFGAP3 to
COP-I-mediated membrane traffic.
Materials and Methods
DNA cloning, sequencing and computer analysis
Escherichia coli strain DH5awas used for plasmid isolation, and polymerase
chain reaction reactions using a combination of AmpliTaq and TaqExtender
or Vent DNA polymerase, restriction enzyme digests and ligations were
performed by standard methods. All constructs were verified by DNA
sequencing. Database searches were performed using the BLAST and CBLAST
servers at National Institutes of Health (NIH). Multiple alignments were
performed with the program MEGALIGN using the CLUSTAL V algorithm.
Plasmids and IMAGE clones
Full-length clones for both human Glo3 proteins were obtained from the
IMAGE consortium. A hypothetical intron–exon structure established for the
ARFGAP2 locus on chromosome II allowed to explain a number of splice
variants and artefacts found in the expressed sequence tag (EST) database.
Splice variants most closely related to yeast Glo3p were chosen for further
experiments: IMAGE clone 6500690 for ARFGAP3 and a combination of IMAGE
clone 209610498 (ATG to NcoI site) and clone 209860241 (NcoI to TGA) for
ARFGAP2. The coding region of several IMAGE clones with full-length inserts
were sequenced.
Figure 8: Silencing of all three ARFGAPs is lethal in HeLa cells. Silencing of ARFGAP1 or a combination of ARFGAP2 and ARFGAP3
failed to cause lethality in HeLa cells, as well as in NRK cells (data not shown). A) The depletion of ARFGAP1, ARFGAP2 or ARFGAP3 in
single knock-downs in HeLa cells was verified by immunoblotting. Antibodies against tubulin and g-COP were used to verify equal protein
loading. B and C) Cell counting was performed from DAPI-stained HeLa cells grown on coverslips. Note the strong reduction in cell
numbers in the ARFGAP triple knock-down compared with a double knock-down involving ARFGAP2 and ARFGAP3 combined with
a control oligo. For details of the oligos, see Materials and Methods. D) Quantification of the reduction in cell numbers from the above.
More than 75% cell death was routinely observed after three sets of siRNA transfections at the 72-h time-point after the last transfection
(in 10 independent experiments). Bar ¼100 mm.
Traffic 2007; 8: 1644–1655 1653
Two Human ARFGAPs Acting in COPI-Dependent Trafficking
Cloning of ARFGAP2 and ARFGAP3, expression
plasmids and raising antibodies
To obtain His6-tagged recombinant proteins as antigen and for binding
experiments, DNAs encoding full-length ARFGAP2 and ARFGAP3 were
cloned into pET21d (Novagen) and expressed in lDE3 lysogens of strain
BL21 and purified by Ni-NTA chromatography as described (17). Purified
His6-tagged ARFGAP2 and ARFGAP3 were used for raising antibodies in
rabbits. Antibodies were affinity purified over immobilized GST-tagged
ARFGAP2 and ARFGAP3 protein lacking the ARFGAP domain to reduce
the risk of cross-reactivity of these antibodies between the two proteins
and characterized by immunoblotting and IF. Immunoblots on total protein
extracts from NRK and HeLa cells revealed that our antibodies recognized
the human and rat proteins with high affinity, that there is a small difference
between the two proteins in their apparent molecular weight in SDS-
polyacrylamide gels and that both proteins are expressed at readily detect-
able levels (data not shown).
Cell lines and commercial antibodies
HeLa cells, NRK and Vero (green monkey kidney fibroblast) cells were
maintained in DMEM medium þ10% bovine calf serum, 2 mMglutamine
and antibiotics penicillin and streptomycin (100 U/mL and 100 mg/mL,
respectively). In some experiments, BFA was added to the cell culture
medium at a concentration of 5 mg/mL. A stock solution of BFA (2 mg/mL in
ethanol; Epicentre Technologies) was stored at 208C.
For colocalization analysis by IF, we used monoclonal antibodies against
GM130 (clone NN 2C10; Abcam 1299), p115 (clone 5D6; Sigma P3118),
g-adaptin (clone 100/3; Sigma4200) and b-COP (clone mAD; Abcam 6323).
IF and confocal fluorescence microscopy
NRK or HeLa cells grown on glass coverslips were fixed in methanol at
208C for 4 min followed by fixation for 30 seconds with 208C acetone.
Cells were mounted in 50% glycerol on glass slides and epifluorescence
microscopy was performed on a Zeiss Axioplan microscope with a 63,1.4
oil immersion objective, with images taken using a CoolSnap CCD camera.
Pull-down experiments and immunoblot analysis
His6- or GST-tagged proteins were used to pull down coatomer from yeast
or rat liver cytosol, under conditions described by us previously (17). His6-
tagged Glo3p lacking its amino terminus was compared with full-length
Glo3p as well as full-length Gcs1p in its ability to bind coatomer from yeast
cytosol. Protein bound to Ni-NTA agarose beads was resolved by SDS–
PAGE, followed by Western blotting and incubation with anti-yeast coat-
omer antibodies detected by enhanced chemiluminescence (ECL). A
recombinant GST-fusion protein harbouring DN96-ARFGAP2 was used for
pull-downs from rat liver cytosol. Glutathione S-transferase alone or
recombinant GST-tagged GDI (a guanine nucleotide dissociation inhibitor
acting on rab proteins) were used as negative controls. SDS–PAGE and
immunoblotting analysis using ECL were performed as described (17).
In vitro budding reactions
In vitro budding of COP-I Golgi-derived vesicles from purified rat liver Golgi
membranes was performed in the presence of pig brain cytosol as a source
of coatomer, ATP and the nonhydrolysable GTP analogue, GTP-g-S as
described by the Rothman lab (31,32). Pig brain cytosol was prepared by
the method as described by (37). Donor Golgi membranes were prepared
according to a protocol established by the Graham Warren lab (38). Vesicles
were separated from Golgi donor membranes by sedimentation to equilib-
rium in a sucrose density gradient. Following trichloroacetic acid precipitation
and SDS–PAGE on 10% gels, proteins in fractions were analysed by silver
stain and ECL immunoblot using specific antibodies as indicated.
Cell transfections and application of cholera toxin
CTX-K63 and CFP- and YFP-fusion proteins
Vero cells were transfected by electroporation as described by us earlier
(21). Unless otherwise mentioned, cells were tested for expression of
fluorescent fusion proteins and cholera toxin binding 6 h after trans-
fection. For experiments, cells were treated in a pulse-like manner with
CTX-K63, as described before (21,33). CTX-K63 is a nontoxic mutant of
cholera toxin, which has no ADP-ribosylating activity as a result of a Ser
63
/
Lys
63
point mutation in the A-subunit (21,33). Following treatment with
CTX-K63, cells remained in the CO
2
incubator until they were transferred
to the thermostated microscope chamber. Internalization of CTX-K63-
Cy3, labelled as described below, was performed for 3 h at 378C before
imaging.
An expression clone for an YFP-tagged version of the Golgi-resident en-
zyme galactosyl transferase (GalT-YFP) was a kind gift from J. Lippincott-
Schwartz (NIH). We constructed CFP-tagged full-length or DN96-ARFGAP2
expression clones, with the CFP moiety at the carboxy terminus of the
proteins, using standard cloning procedures in the vector pECFP-C1
(ClonTech).
Cy3 labelling of CTX-K63
As described by us (21), CTX-K63 mutant toxin was labelled with Cy3,
resolved by 10% SDS–PAGE and scanned using a Typhoon 8600 Imager
with an excitation wavelength of 532 nm and standard emission filter
560LP. Pixel-by-pixel resolved fluorescence measurement revealed prefer-
ential labelling of the A-subunit in the sample that was used in the
experiments described in Figure 7. The absorption spectrum of the CTX-
K63-Cy3 sample was acquired with a Fluoromax-3 spectrofluorimeter. The
wavelength ratio of 550 nm/280 nm, that is, of dye (550 nm) to protein
(280 nm), was used to measure the concentration of proteins in the sample
and to estimate the labelling ratio, calculated using Beer’s law and the dye
extinction coefficient of Cy3 150 000 M
1
cm
1
. From this presence of
approximately 1.5–2 dye molecules per A-subunit were determined.
siRNA
Sequences were designed following the parameters set out by Dharmacon
by analysing the human genomic sequence for each protein. The following
sequences were selected: ARFGAP1, AAG GUG GUC GCU CUG GCC GAA
G (siACE-RNAi OPTION C DUPLEX); ARFGAP2, AAG CUA UGG GGU GUU
UCU CUG (siACE-RNAi OPTION C DUPLEX); and ARFGAP3, AAC CUA
UGG AGU GUU CCU UUG (siACE-RNA OPTION C DUPLEX). All oligos
were custom synthesized by Dharmacon (http://www.Dharmacon.com/).
A green fluorescent protein duplex was used as a control oligo for all
experiments (Dharmacon, D-001300-01-20).
RNAi-mediated knock-downs of ARFGAP1, 2 and 3
in HeLa cells
HeLa M cells were maintained in DMEM medium supplemented with 10%
foetal bovine serum, 2 mML-glutamine, penicillin (100 U/mL) and strepto-
mycin (100 mg/mL). siRNAs were used at a final concentration of 20 nMin
all transfection experiments. Triple knock-down of all three ARFGAPs and
combinations of these with controls were performed in a staggered 10-day
protocol, using oligofectamine. For double transfections, paired siRNA
oligos were added to 15 nMfinal concentrations. Cells were seeded on
day 0 and transfected on day 1 with either control, single ARFGAP or
ARFGAP2 and 3 oligos. On day 2, cells were split into new wells, allowed to
adhere to the dish for 6 h, then transfected with either control or ARFGAP1
oligo. On day 3, the media was changed to remove transfection reagents.
On day 5, cells were split, allowed to adhere for 6 h and transfected as on
day 1. On day 6, the media were refreshed, and on day 7, cells were
transfected as on day 2. On day 8, cells were split (some cells were
reseeded onto coverslips for 40-60-diamidino-2-phenylindole (DAPI) staining.
On day 9, the cells rested, and on day 10, cells were collected for analysis.
For immunoblot samples, cells washed with PBS, then lysed directly in the
plate by addition of 3Laemmli sample buffer, pipetted out and immedi-
ately boiled at 958C for 5 min. DNA was sheared by passing the samples
through a narrow gauge needle and vortexing at maximum speed. Samples
were then loaded onto SDS–PAGE gels. Cells growing on coverslips for
DAPI staining were washed three times in PBS, then methanol–acetone
fixed and mounted on to glass slides in DAPI-containing mounting medium
1654 Traffic 2007; 8: 1644–1655
Frigerio et al.
(10 mg/mL DAPI, 1 mg/mL p-phenylenediamine in 90% glycerol, 10% PBS,
pH 8.0). A similar protocol was used for NRK cells, yielding almost
identical results.
Acknowledgments
We are grateful to Drs J. Lippincott-Schwartz, M.S. Robinson and Paul
Luzio for generously sharing antibodies and plasmids. Thi s work was
supported by The Wellcome Trust (Senior Research Fellowship; grant
047578 to R. D.).
References
1. Kirchhausen T. Three ways to make a vesicle. Nat Rev Mol Cell Biol
2000;1:187–198.
2. Bonifacino JS, Lippincott-Schwartz J. Coat proteins: shaping mem-
brane transport. Nat Rev Mol Cell Biol 2003;4:409–414.
3. Duden R. ER-to-Golgi transport: COP I & COP II function. Mol Membr
Biol 2003;20:197–207.
4. Rothman JE, Wieland FT. Protein sorting by transport vesicles. Science
1996;272:227–234.
5. Spang A, Matsuoka K, Hamamoto S, Schekman R, Orci L. Coatomer,
Arf1p, and nucleotide are required to bud coat protein complex I-coated
vesicles from large synthetic liposomes. Proc Natl Acad Sci U S A
1998;95:11199–11204.
6. Donaldson JG, Jackson CL. Regulators and effectors of the ARF
GTPases. Curr Opin Cell Biol 2000;12:475–482.
7. Randazzo PA, Hirsch DS. Arf GAPs: multifunctional proteins that
regulate membrane traffic and actin remodelling. Cell Signal 2004;16:
401–413.
8. Lanoix J, Ouwendijk J, Stark A, Szafer E, Cassel D, Dejgaard K,
Weiss M, Nilsson T. Sorting of Golgi resident proteins into different
subpopulations of COPI vesicles: a role for ArfGAP1. J Cell Bi ol 2001;
155:1199–1212.
9. Malsam J, Gommel D, Wieland FT, Nickel W. A role for ADP
ribosylation factor in the control of cargo uptake during COPI-coated
vesicle biogenesis. FEBS Lett 1999;462:267–272.
10. Pepperkok R, Whitney JA, Gomez M, Kreis TE. COPI vesicles
accumulating in the presence of a GTP restricted arf1 mutant are
depleted of anterograde and retrograde cargo. J Cell Sci 2000;113:
135–144.
11. Donaldson JG, Honda A, Weigert R. Multiple activities for Arf1 at the
Golgi complex. Biochim Biophys Acta 2005;1744:364–373.
12. Poon PP, Cassel D, SpangA, Rotman M, Pick E, SingerRA, Johnston GC.
Retrograde transport from the yeast Golgi is mediated by two ARF GAP
proteins with overlapping function. EMBO J 1999;18:555–564.
13. Dogic D, de Chassey B, PickE, Cassel D, Lefkir Y, HenneckeS, Cosson P,
Letourneur F. The ADP-ribosylation factor GTPase-activating protein
Glo3p is involved in ER retrieval. Eur J Cell Biol 1999;78:305–310.
14. Lewis SM, Poon PP, Singer RA, Johnston GC, Spang A. The ArfGAP
Glo3 is required for the generation of COPI vesicles. Mol Biol Cell
2004;15:4064–4072.
15. Letourneur F, Gaynor E, Hennecke S, Demolliere C, Duden R, Emr S,
Riezman H, Cosson P. Coatomer is essential for retrieval of dilysine-
tagged proteins to the ER. Cell 1994;79:1199–1207.
16. Yahara N, Sato K, Nakano A. The Arf1p GTPase-activating protein Glo3p
executes its regulatory function through a conserved repeat motif at
its C-terminus. J Cell Sci 2006;119:2604–2612.
17. Eugster A, Frigerio G, Dale M, Duden R. COP I domains required for
coatomer integrity, and novel interactions with ARF and ARF-GAP.
EMBO J 2000;19:3905–3917.
18. Watson P, Frigerio G, Collins B, Duden R, Owen D. g-COP appendage
domain structure and function. Traffic 2004;5:79–88.
19. Cukierman E, Huber I, Rotman M, Cassel D. The ARF1 GTPase-
activating protein: zinc finger motif and Golgi complex localization.
Science 1995;270:1999–2002.
20. Liu W, Duden R, Phair RD, Lippincott-Schwartz J. ArfGAP1 dynamics
and its role in COPI coat assembly on Golgi membranes of living cells.
J Cell Biol 2005;168:1053–1063.
21. Majoul I, Straub M, Hell SW, Duden R, So
¨ling HD. KDEL-cargo
regulates interactions between proteins involved in COPI vesicle traffic:
measurements in living cells using FRET. Dev Cell 2001;1:139–153.
22. Yang JS, Lee SY, Gao M, Bourgoin S, Randazzo PA, Premont RT,
Hsu VW. ARFGAP1 promotes the formation of COPI vesicles, suggest-
ing function as a component of the coat. J Cell Biol 2002;159:69–78.
23. Lee SY, Yang JS, Hong W, Premont RT, Hsu VW. ARFGAP1 plays
a central role in coupling COPI cargo sorting with vesicle formation.
J Cell Biol 2005;168:281–290.
24. Frolov MV, Alartortsev VE. Molecular analysis of novel Drosophila
gene, Gap69C, encoding a homolog of ADP ribosylation factor
GTPase activating protein. DNA Cell Biol 2001;20:107–113.
25. Singh J, Itahana Y, Parrinello S, Murata K, Desprez PY. Molecular cloning
and characterization of a zinc finger protein involved in Id-1-stimulated
mammary e pithelia l cell growt h. J Biol Chem 2 001;276:11852–11858.
26. Zhang C, Yu Y, Zhang S, Liu M, Xing G, Wei H, Bi J, Liu X, Zhou G,
Dong C, Hu Z, Zhang Y, Luo L, Wu C, Zhao S et al. Characterization,
chromosomal assignment, and tissue expression of a novel human
gene belonging to the ARF GAP family. Genomics 2000;63:400–408.
27. Liu X, Zhang C, Xing G, Chen Q, He F. Functional characterization of
novel human ARFGAP3. FEBS Lett 2001;490:79–83.
28. Sapperstein SK, Walter DM, Grosvenor AR, Heuser JE, Waters MG.
p115 is a general vesicular transport factor related to the yeast
endoplasmic reticulum to Golgi transport factor Uso1p. Proc Natl Acad
Sci U S A 1995;92:522–526.
29. Nakamura N, Rabouille C, Watson R, Nilsson T, Hui N, Slusarewicz P,
Kreis TE, Warren G. Characterization of a cis-Golgi matrix protein,
GM130. J Cell Biol 1995;131:1715–1726.
30. Traub L. Common principles in clathrin-mediated sorting at the Golgi and
the plasma membrane. Biochim Biophys Acta 2005;1744:415–437.
31. Serafini T, Stenbeck G, Brecht A, Lottspeich F, Orci L, Rothman JE,
Wieland FT. A coat subunit of Golgi-derived non-clathrin-coated
vesicles with homology to the clathrin-coated vesicle coat protein
beta-adaptin. Nature 1991;349:215–220.
32. Serafini T, Rothman JE. Purification of Golgi cisternae-derived non-
clathrin-coated vesicles. Methods Enzymol 1992;219:286–299.
33. Majoul I, Sohn K, Wieland FT, Pepperkok R, Pizza M, Hillemann J,
So
¨ling HD. KDEL receptor (Erd2p)-mediated retrograde transport of the
cholera toxin A subunit from the Golgi involves COPI, p23, and the
COOH terminus of Erd2p. J Cell Biol 1998;143:601–612.
34. Nie Z, Boehm M, Boja ES, Vass WC, Bonifacino JS, Fales HM,
Randazzo PA. Specific regulation of the adaptor protein complex
AP-3 by the Arf GAP AGAP1. Dev Cell 2003;5:513–521.
35. Nie Z, Fei J, Premont RT, Randa zzo PA. The Arf GAPs AGAP1 and
AGAP2 distinguish between the adaptor protein complexes AP-1 and
AP-3. J Cell Sci 2005;118:3555–3566.
36. Poon PP, Nothwehr SF, Singer RA, Johnston GC. The Gcs1 and
Age2 ArfGAP proteins provide overlapping essential function for trans-
port from the yeast trans-Golgi network.J Cell Biol 2001;155:1239–1250.
37. Malhotra V, Serafini T, Orci L, Shepherd JC, Rothman JE. Purification
of a novel class of coated vesicles mediating biosynthetic protein
transport through the Golgi stack. Cell 1989;58:329–336.
38. Wang Y, Taguchi T, Warren G. Purification of rat liver Golgi stacks, 3rd
edn. In: Celis J, editor. Cell Biology: A Laboratory Handbook. Elsevier
Science (USA); 2006, pp. 33–39.
Traffic 2007; 8: 1644–1655 1655
Two Human ARFGAPs Acting in COPI-Dependent Trafficking
... solubles ainsi que par les protéines membranaires localisées dans leur partie cytosolique (Jackson et al. 1990;Munro et Pelham 1987). La protéine Arf1 et son régulateur Arfgap1 interviendraient notamment dans ce processus (Lanoix et al. 1999;Frigerio et al. 2007). ...
... En effet, une mutation portée par ce gène a été décrite dans certains cas de gérodermie ostéodysplasique, caractérisée par une peau ridée et une ostéoporose (Hennies et al. 2008). Néanmoins, il serait intéressant de connaître les phénomènes engendrés par une augmentation de l'expression de ce gène, car comme décrit précédemment, la surexpression in vitro de domaines particuliers de certaines golgines peut avoir un effet délétère sur la sécrétion des protéines Luke et al. 2003 (Lanoix et al. 1999;Frigerio et al. 2007). Néanmoins, peu de choses sont connues sur l'impact du vieillissement sur ce transport rétrograde. ...
Thesis
La peau est un organe se trouvant à l’interface de notre organisme et de notre environnement. Ellesubit un vieillissement qui se traduit par des modifications affectant ses différentes couches. Parmi celles-cile derme est particulièrement affecté. Les fibroblastes, présents dans le derme, synthétisent des moléculesde la matrice extracellulaire ainsi que des enzymes de dégradation. Lors du vieillissement, cette sécrétionest modifiée favorisant ainsi la sécrétion d’enzymes et la dégradation du derme. L’un des objectifs de ces travaux de thèse est d’évaluer les modifications ayant lieu chez les fibroblastes lors du vieillissement. Pour cela, trois modèles de vieillissement de fibroblastes primaires dermiques humains ont été développés et caractérisés. Une étude transcriptomique a été réalisée par PCR quantitative en temps réel et a permis de mettre en évidence des différences d’expression de gènes codant pour des composants du derme. Dans le but de développer des actifs cosmétiques à visée « anti-âge », des extraits riches en polysaccharides ont été réalisés à partir de plantes et de microorganismes, puis leur efficacité a été évaluée sur des modèles de peaux humaines. L’appareil de Golgi est un organite jouant un rôle majeur dans la modification post-traductionnelle et la sécrétion. La modification structurale de celui-ci lors du vieillissement a été évaluée sur les modèles de fibroblastes en utilisant des techniques de microcopie optique et électronique. Les résultats montrent une altération de la morphologie du réseau trans-golgien chez les fibroblastes sénescents, l’un des modèles développés au cours de ces travaux. Chez ces cellules, le TGN présente une morphologie particulière qui s’étend dans le cytoplasme. Ainsi, lors de la sénescence, nous avons pu révéler par le biais d’une étude transcriptomique que l’expression de gènes impliqués dans la structure et la fonctionnalité de l’appareil de Golgi étaient modifiée. Les résultats obtenus lors de cette thèse ont permis de mettre en évidence de nouveaux marqueurs biologiques innovants pour le criblage d’actifs dermo-cosmétiques à visée «anti-âge».
... [95][96][97] By contrast, the activity of ArfGAP2 and 3 is not influenced by membrane curvature like ArfGAP1 98 , since no ALPS motif is found in these regulators. 99,100 In summary, the fundamental elements for the formation of COPI vesicles consist of coatomer, Arf1 and its regulators the ArfGEFs and ArfGAPs. 81,101-103 ...
... 143,145,146 On the membrane-associated coat, GTP hydrolysis is speculated to be mediated in a curvature-independent manner at the triad central area by ArfGAP2 close to the γ-COP/Arf1 while a curvature-dependent manner at peripheral triad area by ArfGAP1 close to the β-COP/Arf1. 68,81,109 In eukaryotes, inactivation of one specific sub-type of ArfGAPs (ArfGAP1, or ArfGAP2 and -3) is not lethal 99 , suggesting a certain degree of overlapping function among them. It is not elucidated how the role of one ArfGAP would be replaced by the other. ...
Thesis
Coat protein complex I (COPI) vesicles coated with the heptameric complex coatomer mediate retrograde cargo trafficking from Golgi to endoplasmic reticulum as well as intra-Golgi transport. Whether paralogous subunits of coatomer have different functions is currently unclear. In this thesis, we reveal distinct roles of paralogous coatomer subunits γ1-COP and γ2-COP during the neuronal differentiation of mouse pluripotent cells. Following genome editing experiments, our work shows that γ1-COP specifically facilitates neurite extension and underlines a paralogue-specific function of the COPI pathway and offers evidence for the role of COPI coated vesicles in neuronal polarization. Furthermore, to explore the mechanism of γ1-COP specific functions during pluripotent cells differentiation into neurons, the combination proximity-dependent biotinylation with affinity purification and mass spectrometry (AP-MS) was applied to analyze whether the interactome of γ-COPs reveals paralogue-specific cargos or regulators. In the light of label free quantification (LFQ) analysis, various neurogenesis-related proteins were significantly enriched in the γ1-COP interactome indicating that γ1-COP may preferentially traffic such proteins during the process of pluripotent cells neuronal differentiation. Zusammenfassung Mit dem heptameren Komplex-Coatomer beschichtete Coat Protein Complex I (COPI) -Vesikel vermitteln den retrograden Frachthandel von Golgi zum endoplasmatischen Retikulum sowie den Intra-Golgi-Transport. Ob paraloge Untereinheiten des Coatomers unterschiedliche Funktionen haben, ist derzeit unklar. In dieser Arbeit zeigen wir unterschiedliche Rollen der paralogen Coatomer-Untereinheiten γ1-COP und γ2-COP während der neuronalen Differenzierung pluripotenter Mauszellen. Nach Experimenten zur Bearbeitung des Genoms zeigen unsere Arbeiten, dass γ1-COP die Neuritenverlängerung spezifisch erleichtert, eine paralogspezifische Funktion des COPI-Signalwegs unterstreicht und Hinweise auf die Rolle von COPI-beschichteten Vesikeln bei der neuronalen Polarisation liefert. Um den Mechanismus der γ1-COP-spezifischen Funktionen während der Differenzierung pluripotenter Zellen in Neuronen zu untersuchen, wurde die kombinationsnäherungsabhängige Biotinylierung mit Affinitätsreinigung und Massenspektrometrie (AP-MS) angewendet, um zu analysieren, ob das Interaktom von γ-COPs Paralog zeigt. spezifische Ladungen oder Regulierungsbehörden. Im Lichte der Analyse der markierungsfreien Quantifizierung (LFQ) wurden verschiedene Neurogenese-verwandte Proteine im γ1-COP-Interaktom signifikant angereichert, was darauf hinweist, dass γ1-COP solche Proteine während des Prozesses der neuronalen Differenzierung pluripotenter Zellen bevorzugt transportieren kann.
... (B) Ramos (left) and XL4.1 (right) cells were treated with 50 µM Thiamet-G or DMSO vehicle for 8 h and lysates were subjected to anti-COPγ1IP and analyzed via IB. Vps35 (depleted 3.582-and 2.412-fold from the BFA samples), or are regulators of membrane protein quality control, such as the AAA+ ATPase torsinA (enriched 2.069-and 2.466-fold in the BFA samples) and the ubiquitin E3 ligase NEDD4, which is also a known O-GlcNAc substrate (depleted 3.838and 3.058-fold from the BFA samples) (Figures 4C,D and Supplemental Material) (86)(87)(88)(89)(90)(91)(92)(93)(94)(95)(96)(97)(98)(99)(100)(101). We concluded that our glycoproteomics workflow identified candidate BFA-dependent changes in O-GlcNAc substrates that may impact on protein trafficking. ...
... COPγ1 is a core component of the heteroheptameric COPI complex, which is recruited to Golgi membranes by the small GTPase ADP ribosylation factor 1 (Arf1) to mediate vesicle formation and trafficking within the Golgi or to the ER (49,102,103). COPγ proteins interact with cargo adaptors in the Golgi membrane, with Arf GTPase-activating proteins and with other COPI components in the coat itself (49,88,89,92). COPγ is highly conserved across eukaryotes and is essential for in vitro COPI vesicle formation and for viability in budding yeast (49,104,105). ...
Article
Full-text available
O-linked β-N-acetylglucosamine (O-GlcNAc) is an abundant and essential intracellular form of protein glycosylation in animals and plants. In humans, dysregulation of O-GlcNAcylation occurs in a wide range of diseases, including cancer, diabetes, and neurodegeneration. Since its discovery more than 30 years ago, great strides have been made in understanding central aspects of O-GlcNAc signaling, including identifying thousands of its substrates and characterizing the enzymes that govern it. However, while many O-GlcNAcylated proteins have been reported, only a small subset of these change their glycosylation status in response to a typical stimulus or stress. Identifying the functionally important O-GlcNAcylation changes in any given signaling context remains a significant challenge in the field. To address this need, we leveraged chemical biology and quantitative mass spectrometry methods to create a new glycoproteomics workflow for profiling stimulus-dependent changes in O-GlcNAcylated proteins. In proof-of-principle experiments, we used this new workflow to interrogate changes in O-GlcNAc substrates in mammalian protein trafficking pathways. Interestingly, our results revealed dynamic O-GlcNAcylation of COPγ1, an essential component of the coat protein I (COPI) complex that mediates Golgi protein trafficking. Moreover, we detected 11 O-GlcNAc moieties on COPγ1 and found that this modification is reduced by a model secretory stress that halts COPI trafficking. Our results suggest that O-GlcNAcylation may regulate the mammalian COPI system, analogous to its previously reported roles in other protein trafficking pathways. More broadly, our glycoproteomics workflow is applicable to myriad systems and stimuli, empowering future studies of O-GlcNAc in a host of biological contexts.
... ArfGAP2 and -3, and its S. cerevisiae homologue Glo3 lack an ALPS motif and are unlikely to be regulated by membrane curvature (Yahara et al., 2006). The appendage domain of γ-COP binds to ArfGAP2 and -3 with differential affinities (Frigerio et al., 2007;Watson et al., 2004). The strong binding of ArfGAP2 to coatomer raises the possibility that ArfGAP2/Glo3 is stably associated with cytosolic coatomer and is, hence, co-recruited to membranes (Eugster et al., 2000;Lewis et al., 2004;Watson et al., 2004), where it can be observed in association with γ-Arf . ...
... It is unclear how one ArfGAP would physically supplant the other, deleted subtype with regard to its specific targeting elements and niches within the framework of the coat. Combined deletion or knockdown of both sub-types (ArfGAP1, and ArfGAP2 and -3) critically affects cell viability of yeast and mammalian cells (Frigerio et al., 2007;Kartberg et al., 2010;Poon et al., 1999). Future studies on cargosorting and vesicle uncoating in the absence of either ArfGAP will prove instrumental in elucidating the true nature of their functional roles. ...
Article
Full-text available
The coat protein complex I (COPI) allows the precise sorting of lipids and proteins between Golgi cisternae and retrieval from the Golgi to the ER. This essential role maintains the identity of the early secretory pathway and impinges on key cellular processes, such as protein quality control. In this Cell Science at a Glance and accompanying poster, we illustrate the different stages of COPI-coated vesicle formation and revisit decades of research in the context of recent advances in the elucidation of COPI coat structure. By calling attention to an array of questions that have remained unresolved, this review attempts to refocus the perspectives of the field.
... There is also a similar localization of NBPF15-FLAG in surrounding interphase cells as KLHL9 (regulates AURKB dynamics on mitotic chromosomes and influences mitotic progression and cytokinesis completion (Maerki et al. 2009;Sumara et al. 2007)), ARFGAP2 (involved in protein transport between the Golgi and endoplasmic reticulum (Frigerio et al. 2007)), PAFAH1B3 (involved in neuronal migration and implicated in lissencephaly (Sweeney et al. 2000)), and CEP128 (centriole and spindle pole associated protein that functions in neural development (Mönnich et al. 2018)). Nevertheless, the fewer detected interacting proteins may reflect differences between NBPF-mediated biology in a human versus chimpanzee cellular background due to a greater impact of NBPF dosage in humans, which enhances the proliferative capacity of vRG and oRG hNPCs. ...
Thesis
The human brain evolves under a unique set of adaptive pressures, distinguishing aspects of form and function from the brains of other primate species. An outstanding question in evolutionary biology is the biological tradeoff of human brain adaptations and neurodevelopmental/neuropsychiatric disorders (NND). Two ways to investigate proximate mechanisms of human evolutionary neurobiology are to study human-specific genetic variation and the genetic basis of neuropathology. This dissertation uses an evolutionary medicine approach to investigate three cases of functional divergence of genetic and molecular variation that mediate human-specific features of neural development implicated in NND. Segmental duplications (SD) are a rich source of highly plastic genomic variation important for human-specific adaptive evolution and a major cause of NND. As result of SD, the neuroblastoma breakpoint gene family (NBPF) sequence has undergone significant expansion in humans, yet functions of expressed NBPF proteins remain unknown. Structural rearrangements of NBPF are associated with autism and schizophrenia, and are affected in 1q21.1 deletion/duplication syndrome (1q21DDS, OMIM:612474;612475). In this thesis, we characterize NBPF subcellular localization and expression profiles in human and chimpanzee models of corticogenesis. To investigate dosage effects on cortical development, NBPF-depleted and overexpression in cerebral organoids were generated. These findings support the hypothesis that NBPF dosage impacts proliferative dynamics in human corticogenesis, implicating NBPFs in the etiology of 1q21DDS. Genetic variation in members of the CUB and Sushi Multiple Domains (CSMD) gene family of complement pathway regulators is associated with neuropsychiatric disorders. Unlike young NBPF duplicates, CSMD genes are fixed across species. However, evolved functions in synaptic pruning, specifically of CSMD1, contribute to species-specific synaptic plasticity and neuronal circuitry—features linked to human-specific cognitive adaptations. Expanding on these findings, we identify novel biallelic variants in CSMD1 in individuals with overlapping features of neurodevelopmental disorders, namely microcephaly, intellectual disability, polymicrogyria, and epilepsy. Using CSMD1 knockout human cortical organoids, we identify pathogenic mechanisms of NPC over-proliferation and premature differentiation. Our novel data expand CSMD1-associated functions to a variety of steps in corticogenesis, including neural proliferation, differentiation, neuronal migration, and synaptogenesis. Together, human genetics and functional findings implicate CSMD1 as a novel genetic basis of NND. Proper mRNA maturation and nuclear export regulate eukaryotic gene expression that underlies cellular and species diversity. mRNA processing is coordinated by the multimeric THO subcomplex of the TREX complex. THO component THOC6 is the genetic basis of autosomal recessive THOC6 Intellectual Disability Syndrome (TIDS; OMIM:613680). Although TREX is considered conserved, its structural form is not, facilitated by THOC6 acquisition in metazoans. Consequently, functional divergence of mRNA processing and export coordination by TREX has co-evolved with transcriptomic complexity across species. In this dissertation, we generate mouse and human models of TIDS to investigate THOC6-dependent functions. We identify novel THOC6-associated mRNA splicing functions in neural tissue. We observe dysregulation of key signaling pathways in cortical organoids that dictate the transition from proliferative to neurogenic divisions, resulting in delayed differentiation and increased apoptosis—hallmarks of microcephaly. Lastly, THO is known to preferentially accumulate at repetitive regions and protect against transcription-induced instability. Our findings have further implications for THO processing/splicing of highly repetitive loci (e.g., functional human-specific duplicates) that shape human-specific features of corticogenesis. Together, this thesis extends the contribution of evolutionary medicine research and models of molecular neofunctionalization to our understanding of the intersection of evolutionary and pathogenic mechanisms of human neural development.
... To further investigate the regulation of Arf1 in the coat, we incubated COPI coated vesicles with Arf-GAP1 or ArfGAP2 in the presence of a non-hydrolysable GTP analogue and determined their structures. Previously published biochemical data indicates that almost no ArfGAP1 binds to COPI vesicles generated in vitro in the presence of poorly hydrolysable GTP analogs, while ArfGAP2 is abundant in such vesicle fractions (Frigerio et al., 2007). Note, that in the presence of hydrolysable nucleotide GTP, ArfGAP1, 2 and 3 facilitate efficient vesicle uncoating (Weimer et al., 2008). ...
Article
Full-text available
COPI coated vesicles mediate trafficking within the Golgi apparatus and between the Golgi and the endoplasmic reticulum. Assembly of a COPI coated vesicle is initiated by the small GTPase Arf1 that recruits the coatomer complex to the membrane, triggering polymerization and budding. The vesicle uncoats before fusion with a target membrane. Coat components are structurally conserved between COPI and clathrin/adaptor proteins. Using cryo-electron tomography and subtomogram averaging, we determined the structure of the COPI coat assembled on membranes in vitro at 9 Å resolution. We also obtained a 2.57 Å resolution crystal structure of βδ-COP. By combining these structures we built a molecular model of the coat. We additionally determined the coat structure in the presence of ArfGAP proteins that regulate coat dissociation. We found that Arf1 occupies contrasting molecular environments within the coat, leading us to hypothesize that some Arf1 molecules may regulate vesicle assembly while others regulate coat disassembly.
... Whereas ArfGAP1 appears to reside predominantly on the cis-Golgi membrane and ArfGAP2 seems to co-localize with both cis-Golgi and TGN markers (Pevzner et al., 2012), ArfGAP3 localization remains contentious. Some researchers have reported ArfGAP3 to be on cis-Golgi membranes (Frigerio et al., 2007;Weimer et al., 2008), whereas Randazzo and colleagues found ArfGAP3 exclusively at the TGN (Shiba et al., 2013). ...
Article
Full-text available
We previously proposed a novel mechanism by which the enzyme Golgi-specific BFA resistance factor 1, or GBF1, is recruited to the membranes of thecis-Golgi based onin vivoexperiments. Here, we extend thein vivoanalysis on production of regulatory Arf•GDP and observe that ArfGAP2/3 do not play such a role. We confirm that Arf•GDP localization is critical as a TGN-localized Arf•GDP mutant protein fails to promote GBF1 recruitment. We also report the establishment of anin vitroGBF1 recruitment assay that supports regulation of GBF1 recruitment by Arf•GDP. Thisin vitroassay yielded further evidence for the requirement of a Golgi-localized protein since heat denaturation or protease treatment of Golgi membranes abrogated GBF1 recruitment. Finally, combinedin vivoandin vitromeasurements indicate that the recruitment to Golgi membranes via a putative receptor requires only the HDS1 and HDS2 domains in the C-terminal half of GBF1.
Article
COPI‐coated vesicles mediate transport between Golgi stacks and retrograde transport from the Golgi to the endoplasmic reticulum. The COPI coat exists as a stable heptameric complex in the cytosol termed coatomer and is recruited en bloc to the membrane for vesicle formation. Recruitment of COPI onto membranes is mediated by the Arf family of small GTPases, which, in their GTP‐bound state, bind both membrane and coatomer. Arf GTPases also influence cargo selection, vesicle scission, and vesicle uncoating. Guanine nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs) regulate nucleotide binding by Arf GTPases. To understand the mechanism of COPI‐coated vesicle trafficking it is necessary to characterize the interplay between coatomer and Arf GTPases and their effectors. It is also necessary to understand interactions between coatomer and cargo, cargo adaptors/receptors, and tethers facilitating binding to the target membrane. Here, we summarize current knowledge of COPI coat protein structure, we describe how structural and biochemical studies contributed to this knowledge, we review mechanistic insights into COPI vesicle biogenesis and disassembly, and we discuss the potential to answer open questions in the field. COPI‐coated vesicles mediate transport within the Golgi and retrograde transport from the Golgi to the endoplasmic reticulum. This review discusses the assembly and disassembly of COPI‐coated vesicles with an emphasis on the structure of the COPI coat components and regulatory factors including coatomer, Arf GTPases, and ArfGAPs and ArfGEFs. We highlight recent developments and open questions in the field.
Article
Full-text available
ADP‐ribosylation factors (Arfs) and Arf‐like (Arl) GTPases are key regulators of intracellular vesicle trafficking and Golgi structure. Both Arf and Arl proteins cycle between active GTP‐bound and inactive GDP‐bound forms, where guanine nucleotide exchange factors (GEFs) regulate the exchange of GDP for GTP, whereas GTPase‐activating proteins (GAPs) promote the hydrolysis of bound GTP. Human Arl1 is located at the trans‐Golgi network (TGN) and regulates the function and structure of the Golgi complex. However, neither GEFs nor GAPs for human Arl1 have been identified. Here, we report that ArfGAP1, an Arf1 GAP, can promote GTP hydrolysis of Arl1. We show that ArfGAP1 directly interacts with GTP‐bound Arl1 and exhibits GAP activity toward Arl1 in vitro. Exogenous expression of ArfGAP1, but not ArfGAP2 and ArfGAP3, causes dissociation of endogenous Arl1 from the TGN. In addition, GAP activity‐deficient ArfGAP1 fails to regulate the Golgi localization of Arl1. Using an activity pull‐down assay, we demonstrated that ArfGAP1 regulates the levels of Arl1‐GTP in cells expressing ArfGAP1‐myc or with ArfGAP1 knockdown. Finally, we observed that, similar to expression of putative active Arl1 (Arl1QL), ArfGAP1 knockdown impairs endosome‐to‐TGN retrograde transport of the Shiga toxin B‐subunit. Thus, our findings support the idea that ArfGAP1 acts as an Arl1 GAP to regulate the function of Arl1 in vesicle trafficking at the TGN.
Article
Full-text available
The Arf GTPase controls formation of the COPI vesicle coat. Recent structural models of COPI revealed the positioning of two Arf1 molecules in contrasting molecular environments. Each of these pockets for Arf1 is expected to also accommodate an Arf GTPase-activating protein (ArfGAP). Structural evidence and protein interactions observed between isolated domains indirectly suggest that each niche preferentially recruits one of the two ArfGAPs known to affect COPI, i.e. Gcs1/ArfGAP1 and Glo3/ArfGAP2/3, although only partial structures are available. The functional role of the unique non-catalytic domain of either ArfGAP has not been integrated into the current COPI structural model. Here, we delineate key differences in the consequences of triggering GTP hydrolysis through the activity of one versus the other ArfGAP. We demonstrate that Glo3/ArfGAP2/3 specifically triggers Arf1 GTP hydrolysis impinging on the stability of the COPI coat. We show that the Snf1 kinase complex, the yeast homologue of AMP-activated protein kinase (AMPK), phosphorylates the region of Glo3 that is crucial for this effect and, thereby, regulates its function in the COPI-vesicle cycle. Our results revise the model of ArfGAP function in the molecular context of COPI. This article has an associated First Person interview with the first author of the paper.
Article
Antisera raised to a detergent- and salt-resistant matrix fraction from rat liver Golgi stacks were used to screen an expression library from rat liver cDNA. A full-length clone was obtained encoding a protein of 130 kD (termed GM130), the COOH-terminal domain of which was highly homologous to a Golgi human auto-antigen, golgin-95 (Fritzler et al., 1993). Biochemical data showed that GM130 is a peripheral cytoplasmic protein that is tightly bound to Golgi membranes and part of a larger oligomeric complex. Predictions from the protein sequence suggest that GM130 is an extended rod-like protein with coiled-coil domains. Immunofluorescence microscopy showed partial overlap with medial- and trans-Golgi markers but almost complete overlap with the cis-Golgi network (CGN) marker, syntaxin5. Immunoelectron microscopy confirmed this location showing that most of the GM130 was located in the CGN and in one or two cisternae on the cis-side of the Golgi stack. GM130 was not re-distributed to the ER in the presence of brefeldin A but maintained its overlap with syntaxin5 and a partial overlap with the ER-Golgi intermediate compartment marker, p53. Together these results suggest that GM130 is part of a cis-Golgi matrix and has a role in maintaining cis-Golgi structure.
Article
This chapter describes some simple protocols derived from several earlier methods for obtaining highly purified Golgi stacks from rat liver and for determining their relative purity over the homogenate. Centrifugation is carried out using an L8-70M or Optima LE-80K ultracentrifuge and either a SW-28 rotor containing ultraclear tubes or a SW-41 rotor. It is very important to be as accurate as possible when mixing various components and to check the refractive index of each buffer using a refractometer. Aspirate and discard the lipid at the top and the cytosol and collect Golgi fractions that accumulate at the 0.5/0.86 M interface with a plastic Pasteur pipette. This protocol ensures large amounts of Golgi membranes with minimal contamination by other membranes. The relative purification of the Golgi stacks can be assessed by measuring the increase in specific activity of the Golgi enzyme ?-l,4-galactosyltransferase (GAIT) over that of the whole liver homogenate. The yields of Golgi membranes can be calculated from the ratio between the total GalT activity in the Golgi fractions and that of the homogenate.
Article
We have identified and characterized a novel human ADP-ribosylation factor GTPase-activating protein (ARFGAP1) gene that is related to other members of the ARF GAP family. The full-length cDNA for human ARFGAP1 was cloned following the identification of an EST obtained by large-scale cDNA library sequencing through a Blast search of public databases. Structurally, ARFGAP1 encodes a polypeptide of 516 amino acids, which contained a typical GATA-1-type zinc finger motif (CXXCX16CXXC) with the four cysteine residues that are highly conserved among other members of the ARF GAP family. The conserved ARF GAP domain may emphasize the biological importance of this gene. The ARFGAP1 gene, which contained 16 exons ranging from 0.5 to 9.3 kb, was mapped to human chromosome 22q13.2–q13.3 using radiation hybridization and in silico analyses. ARFGAP1 is strongly expressed in endocrine glands and testis. Interestingly, the expression of ARFGAP1 in testis is about sixfold higher than that in ovary, indicating a possible role of ARFGAP1 in the physiological function of sperm. Expression of ARFGAP1 in four human fetal tissues and seven cancer cell lines was also detected.
Article
This chapter discusses that the availability of antibody reagents specific for the COPs can lead to a more improved vesicle generation protocol. This inhibition, while correlating with COP-coated vesicle accumulation, cannot necessarily reflect the degree of vesicle accumulation or eventual recovery. Parameters now determined are based on the inhibition of acceptor. Function can necessarily be redefined by assaying for vesicle production directly through the use of these reagents. As many low-abundance proteins are present in the vesicles derived from both Chinese hamster ovary (CHO) and rabbit liver membrane sources, improved purification methods are necessary to utilize fully the COP-coated vesicles to unravel the biochemistry of intracellular transport.
Article
Four high-molecular-weight proteins form the main subunits of the coat of Golgi-derived (non-clathrin) coated vesicles. One of these coat proteins, beta-COP, is identical to a Golgi-associated protein of relative mass 110,000 (110K) that shares homology with the adaptin proteins of clathrin-coated vesicles. This connection, and the comparable molecular weights of the coat proteins of Golgi-derived and clathrin-coated vesicles, indicates that they may be structurally related. The identification of beta-COP as the 110K protein explains the blocking of secretion by the drug brefeldin A.
Article
We describe a scheme for the purification of the nonclathrin-coated vesicles that mediate transport of proteins between Golgi cisternae and probably from ER to Golgi. These "Golgi-derived coated vesicles" accumulate when Golgi membranes are incubated with ATP and cytosol in the presence of GTP gamma S, a compound that blocks vesicle fusion. The coated vesicles dissociate from the Golgi cisternae in high salt and can then be purified by employing differential and density gradient centrifugation. Golgi-derived coated vesicles have a putative polypeptide composition that is distinct from both cytosol and Golgi membranes, as well as from that of clathrin-coated vesicles.
Article
A recently discovered vesicular transport factor, termed p115, is required along with N-ethylmaleimide-sensitive fusion protein (NSF) and soluble NSF attachment proteins for in vitro Golgi transport. p115 is a peripheral membrane protein found predominantly on the Golgi. Biochemical and electron microscopic analyses indicate that p115 is an elongated homodimer with two globular "heads" and an extended "tail" reminiscent of myosin II. We have cloned and sequenced cDNAs for bovine and rat p115. The predicted translation products are 90% identical, and each can be divided into three domains. The predicted 108-kDa bovine protein consists of an N-terminal 73-kDa globular domain followed by a 29-kDa coiled-coil dimerization domain, a linker segment of 4 kDa, and a highly acidic domain of 3 kDa. p115 is related to Uso1p, a protein required for endoplasmic reticulum to Golgi vesicular transport in Saccharomyces cerevisiae, which has a similar "head-coil-acid" domain structure. The p115 and Uso1p heads are similar in size, have approximately 25% sequence identity, and possess two highly homologous regions (62% and 60% identity over 34 and 53 residues, respectively). There is a third region of homology (50% identity over 28 residues) between the coiled-coil and acidic domains. Although the acidic nature of the p115 and Uso1p C termini is conserved, the primary sequence is not. We discuss these results in light of the proposed function of p115 in membrane targeting and/or fusion.
Article
Dilysine motifs in cytoplasmic domains of transmembrane proteins are signals for their continuous retrieval from the Golgi back to the endoplasmic reticulum (ER). We describe a system to assess retrieval to the ER in yeast cells making use of a dilysine-tagged Ste2 protein. Whereas retrieval was unaffected in most sec mutants tested (sec7, sec12, sec13, sec16, sec17, sec18, sec19, sec22, and sec23), a defect in retrieval was observed in previously characterized coatomer mutants (sec21-1, sec27-1), as well as in newly isolated retrieval mutants (sec21-2, ret1-1). RET1 was cloned by complementation and found to encode the alpha subunit of coatomer. While temperature-sensitive for growth, the newly isolated coatomer mutants exhibited a very modest defect in secretion at the nonpermissive temperature. Coatomer from beta'-COP (sec27-1) and alpha-COP (ret1-1) mutants, but not from gamma-COP (sec21) mutants, had lost the ability to bind dilysine motifs in vitro. Together, these results suggest that coatomer plays an essential role in retrograde Golgi-to-ER transport and retrieval of dilysine-tagged proteins back to the ER.