ArticlePDF Available

Variable actin dynamics requirement for the exit of different cargo from the 𝘵𝘳𝘢𝘯𝘴-Golgi network

Authors:

Abstract and Figures

Efficient post-Golgi trafficking depends on microtubules, but actin filaments and actin-associated proteins are also postulated. Here we examined, by inverse fluorescence recovery after photobleaching, the role of actin dynamics in the exit from the TGN of fluorescent-tagged apical or basolateral and raft or non-raft-associated cargoes. Either the actin-stabilizing jasplakinolide or the actin-depolymerising latrunculin B variably but significantly inhibited post-Golgi traffic of non-raft associated apical p75NTR and basolateral VSV-G cargoes. The TGN-exit of the apical-destined VSV-G mutant was impaired only by latrunculin B. Strikingly, the raft-associated GPI-anchor protein was not affected by either actin toxin. Results indicate that actin dynamics participates in the TGN egress of both apical- and basolateral-targeted proteins but is not needed for apical raft-associated cargo.
Content may be subject to copyright.
Variable actin dynamics requirement for the exit of different cargo
from the trans-Golgi network
Francisco La
´zaro-Die
´guez
a,b,c,1
, Cecilia Colonna
a,1
, Miguel Cortegano
a
, Marı
´a Calvo
d
,
Susana E. Martı
´nez
a,b
, Gustavo Egea
a,b,c,*
a
Departament de Biologia CelÆlular i Anatomia Patolo
`gica, Facultat de Medicina, Universitat de Barcelona, C/Casanova 143, E-08036 Barcelona, Spain
b
Institut d’Investigacions Biome
`diques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, C/Casanova 143, E-08036 Barcelona, Spain
c
Institut de Nanocie
`ncia i Nanotecnologia (IN
2
UB), Universitat de Barcelona, 08036 Barcelona, Spain
d
Serveis Cientifico-Te
`cnics (SCT-UB), Universitat de Barcelona, 08036 Barcelona, Spain
Received 25 April 2007; revised 6 July 2007; accepted 8 July 2007
Available online 17 July 2007
Edited by Felix Wieland
Abstract Efficient post-Golgi trafficking depends on microtu-
bules, but actin filaments and actin-associated proteins are also
postulated. Here we examined, by inverse fluorescence recovery
after photobleaching, the role of actin dynamics in the exit from
the TGN of fluorescent-tagged apical or basolateral and raft or
non-raft-associated cargoes. Either the actin-stabilizing jasplak-
inolide or the actin-depolymerising latrunculin B variably but
significantly inhibited post-Golgi traffic of non-raft associated
apical p75NTR and basolateral VSV-G cargoes. The TGN-exit
of the apical-destined VSV-G mutant was impaired only by
latrunculin B. Strikingly, the raft-associated GPI-anchor protein
was not affected by either actin toxin. Results indicate that actin
dynamics participates in the TGN egress of both apical- and
basolateral-targeted proteins but is not needed for apical raft-
associated cargo.
2007 Federation of European Biochemical Societies. Published
by Elsevier B.V. All rights reserved.
Keywords: Actin; Cytoskeleton; Golgi apparatus; Raft;
Polarized transport; iFRAP
1. Introduction
The trans-Golgi network (TGN) is defined as the major sort-
ing compartment of the secretory pathway of synthesized pro-
teins upon recognition of specific apical or basolateral sorting
signals [1–3]. Tightly associated with the TGN is the cytoskel-
eton, in particular microtubules [4], whereas actin filaments
have also recently attracted increasing attention [5,6]. Thus,
a variety of actin-regulatory/binding proteins have been impli-
cated in post-Golgi trafficking such as Arp2/3, spir1, myosins
II and VI, cortactin, Cdc42, huntingtin related protein 1 (see
[5] and references herein), and huntingtin [7]. In addition, the
role of actin filaments has also been tested in the transport
from the Golgi to the plasma membrane [8–10] and to early/
late endosomes [11–13] with a variety of results in some cases.
This might be attributable to the use of only one actin-dep-
olymerizing agent (usually cytochalasins) or to utilize different
cargoes or both. In any case, the substantial homology be-
tween the protein-sequestration and sorting machinery acting
at the plasma membrane and the TGN [6,14] points to a signif-
icant role of actin filaments or actin dynamics (actin depoly-
merization/polymerization cycle) at the TGN as occurs at the
plasma membrane, but it remains to be established whether ac-
tin acts at the level of cargo sorting, in membrane budding/
scission, or in the subsequent locomotion of TGN-derived
transport carriers or both [5]. As a first approach to this end,
we here explore how actin dynamics participates in the exit
from the TGN of a variety of cargo proteins using in vivo in-
verse fluorescence recovery after photobleaching (iFRAP)
analysis. This technique allows analysis of the kinetic parame-
ters of the exit of a fluorescently-tagged cargo protein from a
subcellular compartment (the Golgi stack/TGN in our case).
Furthermore, our study is focused on the potential role of
actin dynamics in post-Golgi egress regardless of the intrinsic
molecular mechanisms that determine the sorting and subse-
quent polarized transport of these cargoes. Thus, basolateral
sorting is mediated by peptide sequences containing tyrosine-
and dileucine motifs [15]. In contrast, apical sorting is facili-
tated by one of two mechanisms: either by lipid-lipid and
lipid-protein interactions within the transmembrane or luminal
domains, given by N- and O-glycans [16], or by such interac-
tions within the glycosylphosphatidylinositol (GPI) anchor do-
main, which is invariably associated with lipid rafts [17,18].
Recent lines of evidence indicate that oligomerization of mem-
brane proteins at the TGN is an important sorting determinant
for apical transport of raft-associated proteins [19]. Therefore,
apical sorting and transport can be mediated by raft-depen-
dent and raft-independent mechanisms. It is important to high-
light that both polarized and non-polarized epithelial cells
share molecular mechanisms in the sorting of secretory cargo,
the formation of transport carriers and their subsequent trans-
port to the plasma membrane. This validates the utilization of
non-polarized cell lines, for instance COS-1 or non-polarized
MDCK cells [20,21] to address the aforementioned questions.
In this respect, non-epithelial cells are capable of packaging
and transporting secretory cargo selectively in distinct types
of Golgi-derived carriers like fully-polarized cells do [22].
In this study we examined how actin toxins that depolyme-
rize (latrunculin B/LtB) or stabilize (jasplakinolide/Jpk) actin
filaments impair the TGN-exit of non-raft-associated apical
*
Corresponding author. Address: Departament de Biologia CelÆlular i
Anatomia Patolo
`gica, Facultat de Medicina, Universitat de Barcelona,
C/Casanova 143, E-08036 Barcelona, Spain. Fax: +34 93 4021907.
E-mail address: gegea@ub.edu (G. Egea).
1
These authors equally contributed to this study.
0014-5793/$32.00 2007 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.
doi:10.1016/j.febslet.2007.07.015
FEBS Letters 581 (2007) 3875–3881
(p75NTR)- or basolateral (VSV-G)-targeted proteins. At the
same time, we also examined whether actin dynamics is needed
for the exit of raft-associated GPI from the TGN. Lipid rafts
are membrane platforms enriched in sphingomyelin, choles-
terol and phosphatidylinositol 4,5-biphosphate (PIP
2
). The lat-
ter has a tight relationship with the actin cytoskeleton since it
promotes actin assembly into filaments [23]. We observed that
actin dynamics participates in the exit of both non-raft-associ-
ated apical and basolateral-targeted cargo, but it was not
needed for the TGN exit of raft-anchored GPI. This indicates
that actin does not functionally associate with raft-associated
GPI anchored proteins at the TGN.
2. Materials and methods
2.1. Cell culture and reagents
COS-1 cells were grown at 37 C in complete DMEM culture med-
ium, which consisted of DMEM (Gibco BRL, Eggestein, Germany)
supplemented with penicillin (100 i.u./ml), streptomycin (100 mg/ml),
LL
-glutamine (2 mM), MEM sodium pyruvate (1 mM) and fetal calf
serum (10%). Latrunculin B was purchased from Calbiochem (EMD
Biosiences, Inc., darnstadt, Germany) and Jasplakinolide from Molec-
ular probes (Eugene, OR, USA). Monoclonal antibodies to giantin were
kindly provided by H.P. Hauri (Biozentrum, Basel University) and
those to TGN46 were purchased from Serotec (Oxford, UK). Second-
ary Alexa Fluor 546 donkey anti-sheep IgG (H + L) and Alexa Fluor
647 goat anti-mouse was from Invitrogen (Carlsbad, CA, USA).
2.2. Plasmids
GFP-tagged wild type and apical-targeted GFP-VSV-G mutant
cDNAS, as well as the CFP-tagged VSV-G wild type cDNA were
kindly provided by Kai Simons (Max Planck Institute of Molecular
Cell Biology and Genetics, Dresden, Germany), and GFP-p75NTR
and YFP-GPI cDNAs were kindly supplied by Roman Polishchuk
(CMNS, Chieti, Italy).
2.3. Transient transfection
COS-1 cells were grown on coverslips in a 35 mm Petri dish and
transiently transfected with the corresponding cDNA using Effectene,
in accordance with the manufacture’s instructions (Qiagen) for 16 h at
37 C in complete DMEM.
2.4. Actin toxins treatments and inversal FRAP experiments
To synchronize post-Golgi traffic at the TGN, transfected cells were
incubated at 19.5 C for 2 h in the presence of cycloheximide (100 lm/ml).
Cells were subsequently treated for 20 min at 19.5 C with LtB or Jpk
(500 nM each) (see the experimental scheme in Fig. 1A).
Inverse fluorescence recovery after photobleaching (iFRAP) experi-
ments were carried out using a Leica TCS SL laser scanning confocal
spectral microscope (Leica Microsystems Heidelberg GmbH, Man-
heim, Germany) with Argon and HeNe lasers attached to a Leica
DMIRE2 inverted microscope equipped with an incubation system
with temperature and CO
2
control. All experiments were performed
at 32 C and 5% CO
2
. For visualization of GFP/YFP, images were ac-
quired using a 63·oil immersion objective lens (NA 1.32), 488 nm laser
line, excitation beam splitter RSP 500, emission range detection: 500–
600 nm with the confocal pinhole set at 4.94 Airy units to minimize
changes in fluorescence due to protein-GFP moving away from the
plane of focus. The whole cytoplasm, except the Golgi, of GFP-fusion
protein transfected cell was photobleached using 50–80 scans with the
488 nm laser line at full power. Postbleach images were monitored at
5 s intervals for 15 min. The excitation intensity was attenuated to
approximately 5% of the half laser power to avoid significant photoble-
aching. Longer recording times were also performed to check the arri-
val of cargo at the plasma membrane and the subsequent absence of
fluorescence in the Golgi.
2.5. Inverse FRAP analysis
To evaluate the results of photobleaching experiments, the observed
fluorescence equilibration in the unbleached region (the Golgi com-
plex) was quantified using the Image Processing Leica Confocal Soft-
ware. Background fluorescence was measured in a random field
outside of cells. All experiments were background substracted, cor-
rected and normalized using the equation described below. For each
time point the relative loss of total fluorescent intensity in the
unbleached region of interest was calculated as:
Irel ¼ðItÞ=ðIoÞðTmean =TtÞ
where I
t
is the average intensity of the unbleached region of interest at
time point t,I
o
is the average pre-bleach intensity of the region of inter-
est and T
mean
and T
t
are the total mean cell intensity of the whole post-
bleach period and average total cell intensity at each time of
postbleach, respectively. Fitting of iFRAP curves was performed with
Graphpad Prism Software v.3.0 (Graphpad Software, San Diego, CA)
and modelled assuming two-phase exponential decay iFRAP, whereas
they were equally well modelled with the one-phase exponential decay
equation:
Yðfluorescence decayÞ¼Span expðKXÞþPlateau
where Ystarted at span + plateau and decayed to plateau with a rate
constant K. Half-time was calculated as: 0.69/K. Afterwards, data plot-
ted as fluorescence intensity that remained in the Golgi vs. time.
Mobile fraction (MF) was calculated as:
MF ¼ðFpre FendÞ=Fpre
where F
pre
was the initial fluorescence intensity and F
end
the final
recovered fluorescence intensity. Statistical analysis was performed
by non-parametric ANOVA post-test Bonferroni.
2.6. Immunofluorescence
Indirect immunofluorescence was carried out as previously described
[7] with the following antibody dilutions: anti-giantin, 1:500 and anti-
TGN46, 1:100. Secondary antibodies were used at 1:250.
3. Results and discussion
We analyzed the fluorescence decay curves of a variety of
green or yellow fluorescent protein (GFP/YFP)-tagged cargoes
from the trans-Golgi network (TGN) by iFRAPs in single liv-
ing mammalian cells. This technique allows us to measure the
time of association of the different GFP/YFP-tagged cargoes
within the TGN, and their dissociation is then reflected by
the loss of fluorescence from this region over time monitored
by confocal microscopy.
We performed experiments for three types of GFP/YFP-
tagged proteins: either raft-associated and apical-targeted such
as GPI, or non-raft-associated and basolateral-targeted like
VSV-G protein, or apical-targeted such as p75NTR and
VSV-G mutant. All these proteins have been reported in api-
cal- and basolateral-like transport carriers in non-polarized
[24] and fully-polarized MDCK cells [2,25–28], Vero, BHK
and CHO cell lines [22,29]. Taking this into account, we uti-
lized COS-1 cells (a non-polarized cell line), which are highly
suitable to be transfected, in contrast to MDCK cells which
usually require microinjection. For quantitative analysis, we
chose those cells that express relatively low levels of GFP/
YFP-tagged proteins to overcome potential toxicity of overex-
pression and, furthermore, those with identical Golgi-associ-
ated basal fluorescence to render the data comparable.
We analyzed the kinetics of the TGN-exit of membrane car-
go by plotting Golgi fluorescence intensity over time and by
determining, for each condition, the mobile fraction, which is
defined as the amount of cargo that has left the Golgi after
15 min at 32 C and the halftime point (T
1/2
), which is defined
as the time at which 50% of the mobile fraction has been lost
(Fig. 1A). COS-1 cells were then transfected with the corre-
3876 F. La
´zaro-Die
´guez et al. / FEBS Letters 581 (2007) 3875–3881
sponding plasmids and left overnight at 37 C. Before the
in vivo recording, cargo was allowed to accumulate at the Gol-
gi/TGN by culturing cells at 19.5 C for 2 h in the presence of
cycloheximide. Cells were treated with the corresponding actin
toxin (LtB or Jpk) for the last 20 min at 19.5 C. We confirmed
that LtB and Jpk significantly disrupted the actin cytoskeleton
at this time and temperature (data not shown). Subsequently,
cells were placed in the confocal chamber at 32 C and iFRAP
was carried out (Fig. 1A). When transfected cells were cultured
at 19.5 C, GFP/YFP-tagged proteins invariably accumu-
lated in the Golgi, which was distinguishable because of the
high fluorescence accumulated in a peri-nuclear structure
(Fig. 1B) and by double-labeling experiments using well-estab-
lished Golgi stack and TGN markers such as giantin and
Fig. 1. (A) Scheme of the experimental design used in this study. (B) Representative frames obtained from time-lapse iFRAP confocal microscopy
experiments in transiently transfected COS-1 cells expressing wild-type basolateral GFP-VSV-G. Note that in control cells, the Golgi-associated
fluorescence of GFP-VSV-G decays over time, whereas in cells treated with the actin-stabilizing toxin jasplakinolide (Jpk; 500 mM) the fluorescence
remains virtually unaltered. (C) Colocalization experiments in COS-1 cells of GFP-tagged cargoes used in this study with the Golgi stack marker
giantin and the TGN marker TGN-46 in COS-1 cells incubated at 19.5 C in the presence of cycloheximide. Bar, 10 lm.
F. La
´zaro-Die
´guez et al. / FEBS Letters 581 (2007) 3875–3881 3877
TGN46, respectively (Fig. 1C). Moreover, reticular and punc-
tate cytoplasmic fluorescence pools were also observed, which
corresponded to proteins located in the endoplasmic reticulum
and transport carriers in transit to the Golgi or the plasma
membrane, respectively. A characteristic morphological visual-
ization of an iFRAP experiment for untreated (control) and
Jpk-treated COS-1 cells expressing GFP-VSV-G is shown in
Fig. 1B. In contrast to the control cell, the Golgi-associated
fluorescence corresponding to VSV-G was virtually unaltered
over time as a consequence of Jpk treatment. The kinetics of
Golgi release followed by VSV-G showed a rapid decrease
(Figs. 1B and 2A, red), indicating that protein was released
up to 56% with a T
1/2
of 3.0 min in untreated cells (Fig. 2A,
red; Table 1). In the presence of Jpk, VSV-G release from
the Golgi was robustly inhibited (Fig. 1B), resulting in a fall
of the mobile fraction after 15 min which was only of 10%
(Fig. 2A, blue; Table 1). When cells were treated with the ac-
tin-depolymerizing toxin LtB (Fig. 2A, green), the mobile frac-
tion was also significantly reduced to 27% (T
1/2
of 5.7 min)
but to a lesser extent than Jpk treatment (Fig. 2A; Table 1).
These results indicate that VSV-G exit from the TGN is signif-
icantly reduced by either the stabilization or depolymerization
of actin filaments, which suggests that actin dynamics (for
example, a rapid actin turnover coupled to the formation of
the transport carrier [5]) is necessary for its post-Golgi trans-
port.
In order to test whether this actin dynamics-dependence was
in turn dependent on the intrinsic basolateral destination of
the VSV-G, we used an apical-destined VSV-G mutant in
which the tyrosine-based motif is masked [30]. We reasoned
that since the post-Golgi exit of VSV-G involving actin
dynamics is also signal-mediated, we would expect differences
in the TGN exit kinetic parameters in the apical-targeted mu-
tant in comparison to the basolateral wild-type form. We ob-
served that the apical VSV-G mutant (Fig. 2B, red) showed
a lower mobile fraction than that of the basolateral VSV-G
(36% and 56%, respectively; Table 1) whereas the T
1/2
was practically the same (3.2 min and 3.0 min, respectively;
Table 1). Unexpectedly, the apical VSV-G mutant was only
sensitive to LtB (Fig. 2B, green), giving rise to a significant
reduction of the mobile fraction (25%; Table 1) and a larger
T
1/2
(6.2 min). This indicates that cargo transport was slowed-
down but, as occurred in control cells, the T
1/2
of LtB-treated
cells expressing either wild-type or mutant VSV-G displayed
no significant difference (5.7 min and 6.2 min, respectively;
Table 1). In contrast, Jpk-treated cells showed no changes in
the shape of the decay curve, the mobile fraction, or the T
1/2
in comparison to control cells (Fig. 2B, blue; Table 1).
The delay in the VSVG-GFP egress from the Golgi in LtB-
treated cells implies the involvement of actin in protein export
from the TGN. This result is consistent with previous observa-
tions in which VSV-G exit from the TGN was lower in cells
treated with cytochalasin-B than in untreated cells [8]. These
authors also reported morphological changes in cargo-contain-
ing tubules, which suggested that actin and actin-based cyto-
skeleton components are involved in the detachment of
TGN-derived transport carriers. The arrival of VSV-G at the
basolateral plasma membrane also decreased in LtB-treated
non-polarized MDCK cells [31]. Several groups have found
that interfering with interactions between dynamin II and syn-
dapin II in living cells caused an inhibition of VSV-G exit from
the TGN [32], and the overexpression of a dominant-interfer-
ing mutant of actin nucleator Spir-1 also strongly inhibited
post-Golgi VSV-G transport to the plasma membrane [33].
Not only actin but also actin regulators such as Cdc42 partic-
ipate in the appropriate sorting at the TGN. Thus, VSV-G and
LDL receptor were missorted to the apical membrane in cells
expressing mutants of Cdc42 [31]. Therefore, our results indi-
cate that actin depolymerization/polymerization cycle controls
the exit of basolateral VSV-G protein from the TGN rather
than the presence of filamentous actin alone, whose involve-
ment in apical VSV-G mutant transport seems to be more di-
rect according to the inhibitory effects caused only by LtB.
Next, we tested the effect of both actin toxins on post-Golgi
traffic of membrane protein p75 neurotrophin receptor
(p75NTR) coupled to GFP [9,31]. In stably transfected polar-
ized and non-polarized MDCK cells, p75NTR is a non-raft-
associated protein that is exclusively transported to the apical
plasma membrane via apical sorting signals contained within
the O-glycosylated stalk [9,27]. As shown in Fig. 3A, in control
cells the decay curve shape for GFP-p75NTR was similar to
those observed for both VSV-Gs. However, the mobile frac-
tion (46%) was greater than in the apical VSV-G mutant
Fig. 2. Inverse FRAP decay curves of basolateral GFP-VSV-G (A)
and apical GFP-VSV-G mutant (B) in COS-1 cells. Results are the
means ± S.E.M. from ncells indicated in Table 1.
3878 F. La
´zaro-Die
´guez et al. / FEBS Letters 581 (2007) 3875–3881
(36%) but lower than that obtained for the wild-type basolat-
eral VSV-G (56%) (Table 1). The exit of p75NTR from the
Golgi was significantly inhibited when cells were treated with
Jpk or LtB (Fig. 3A), which resulted in a significant decrease
in their respective protein mobile fractions (28% and
27%, respectively) (Table 1). These results are inconsistent
with those obtained with the apical VSV-G mutant protein,
which is also non-raft-associated, since their respective sensi-
tivity to Jpk appears to be different. In contrast, both are
equally sensitive to LtB, whose use and sensitivity for
p75NTR was previously reported [9]. Note that the decay
curve for p75NTR during the first 3 min at 32 C was the same
in both control and treated cells, and that the differences
caused by actin toxins appeared later (Fig. 3A). This behavior
indicates that actin dynamics is not required for molecular pro-
cesses that lead to the immediate egress of cargo from the
TGN (for instance, the formation of the transport carrier).
In other words, actin toxins do not appear to alter the initial
mobile fraction. In contrast, actin dynamics might be neces-
sary for earlier molecular processes (for instance, cargo sort-
ing) and any alterations would then affect the immobile
fraction. Previous results showed that p75NTR transport to
the membrane is altered by expression of Cdc42 mutants,
but both constitutive active and inactive mutants differentially
altered the exit of apical and basolateral proteins from the
TGN in vitro [31]. The sensitivity to Jpk treatment could be
a particularity of p75NTR, suggesting that actin cytoskeleton
dynamics would mediate different effects depending on the api-
cally-destined cargo examined. p75NTR contains three dileu-
cine motifs that might represent a type of basolateral sorting
signal [34], but that are present on naturally occurring mem-
brane proteins targeted basolaterally because they lack apical
sorting information.
Finally, an unconventional (non-sorting-receptor mediated)
mechanism for apical sorting is the association with lipid rafts
[35]. A role for rafts in protein sorting was first described for
apical sorting of GPI-anchored proteins in polarized epithelial
cells, where these proteins associate with detergent-resistant
microdomains (DRMs) during their passage through the Golgi
apparatus [36]. Therefore, to explore whether the actin dynam-
ics actively participates in post-Golgi traffic of raft-associated
proteins, we examined the raft-associated glycosylphosphat-
idylinositol (GPI)-anchored protein tagged to YFP, which is
sorted to the apical surface [26,29,37]. The release of YFP-
GPI from the Golgi in untreated COS cells showed a similar
decay curve to those observed for other cargoes but, notably,
after the release from the 19.5 C blockage (Fig. 3B) and con-
versely to the other examined apical-targeted proteins, GPI
was totally unaffected by actin toxins (Fig. 3B; Table 1). More-
over, the final YFP-GPI subcellular destination was the plas-
ma membrane and equally occurred both in treated and
untreated cells (not shown). In fact, all this was unexpected
taking into account the attributed role of lipid rafts and actin
cytoskeleton polymerization occurring in the plasma mem-
brane [23]. It could be postulated that this occurs since GPI,
unlike the other examined cargoes, accumulates in a different
perinuclear compartment upon 20 C incubation and, there-
fore, we were monitoring its exit from another subcellular
compartment in which actin dynamics is irrelevant. This possi-
bility was discarded since cells co-expressing YFP-GPI and
Table 1
Mobile fraction (MF; in %) and T
1/2
(in min) values for each GFP/YFP-tagged cargo exiting from the TGN examined by iFRAP in control and actin
toxin-treated cells
Basolateral VSV-G Apical VSV-G p75NTR GPI
MF T
1/2
MF T
1/2
MF T
1/2
MF T
1/2
Control 56 ± 1.9 (n= 6) 3.0 36.6 ± 3.4 (n= 10) 3.2 46.1 ± 4.5 (n= 8) 7.1 44.8 ± 2.9 (n= 11) 4.1
Jpk 9.5 ± 4.2
***
(n= 6) >10 41.2 ± 3.5 (n= 6) 4.6 28.4 ± 6.0
*
(n= 7) 3.5 41.8 ± 4.5 (n= 6) 3.1
LtB 26.8 ± 8.9
**
(n= 6) 5.7 25.2 ± 3.7
*
(n= 7) 6.2 26.9 ± 3.6
**
(n= 9) 2.1 38.7 ± 4.9 (n= 8) 3.0
Results are the means ± S.E.M. Statistical significance:
*
P60.05,
**
P60.01,
***
P60.001.
Fig. 3. Inverse FRAP decay curves of non-raft-associated apical-
targeted GFP-p75NTR (A) and raft-associated apical YFP-GPI-
anchored protein (B). Results are the means ± S.E.M. from ncells
indicated in Table 1.
F. La
´zaro-Die
´guez et al. / FEBS Letters 581 (2007) 3875–3881 3879
CFP-VSV-G showed that both proteins largely colocalized in
the same compartment upon 19.5 C accumulation (Fig. 4).
An alternative and more probable explanation is that Golgi-
associated lipid rafts are not enriched with PIP
2
like those in
the plasma membrane. In this respect, it is known that PIP
2
levels at the Golgi are very low [38]. Recent data have provided
additional clues on how rafts might participate in apical sort-
ing, regardless of the putative participation of actin filaments.
These authors demonstrated that protein oligomerization is a
specific requirement for apical sorting of GPI-tagged apical
proteins in MDCK and Fischer rat thyroid (FRT) cells, and
is not used by transmembrane, non-raft-associated apical pro-
teins [19]. Association with rafts in the Golgi seems to be insuf-
ficient for this process and an additional step that concentrates
the proteins into large complexes is required, as suggested by
the fact that this behavior is not observed for basolateral
GPI-proteins [39].
The results reported here indicate that transport carriers that
contain raft-associated GPI anchored proteins are primarily
sorted or formed at the TGN independently of actin. This is
not the case for those non-raft-associated cargoes destined to
apical or basolateral plasma membrane domains. Moreover,
data suggest that actin dynamics is particularly relevant to car-
go targeted to the basolateral plasma membrane domain.
Acknowledgements: We thank Ine
´s Ferna
´ndez-Ulibarri and Emma
Martı
´nez-Alonso for critical reading of the manuscript, Maite Mun˜oz
and Anna Bosch for technical support and Robin Rycroft for editorial
assistance. This work has been supported by Grants BFU2006-876/
BMC and European Commission RTN2002-00252 to G.E.
References
[1] Griffiths, G. and Simons, K. (1986) The trans-Golgi network:
sorting at the exit site of the Golgi complex. Science 234, 438–443.
[2] Wandinger-Ness, A., Bennett, M.K., Antony, C. and Simons, K.
(1990) Distinct transport vesicles mediate the delivery of plasma
membrane proteins to the apical and basolateral domains of
MDCK cells. J. Cell Biol. 11, 987–1000.
[3] Keller, P., Toomre, D., Diaz, E., White, J. and Simons, K. (2001)
Multicolour imaging of post-Golgi sorting and trafficking in live
cells. Nat. Cell Biol. 3, 40–149.
[4] Musch, A. (2004) Microtubule organization and function in
epithelial cells. Traffic 5, 1–9.
[5] Egea, G., La
´zaro-Die
´guez, F. and Vilella, M. (2006) Actin
dynamics at the Golgi complex in mammalian cells. Curr. Opin.
Cell Biol. 18, 168–178.
[6] McNiven, M.A. and Thompson, H.M. (2006) Vesicle formation
at the plasma membrane and trans-Golgi network: the same but
different. Science 313, 1591–1594.
[7] del Toro, D., Canals, J.M., Gine
´s, S., Kojima, M., Egea, G. and
Alberch, J. (2006) Mutant huntingtin impairs the post-Golgi
trafficking of brain-derived neurotrophic factor but not its
Val66Met polymorphism. J. Neurosci. 26, 12748–12757.
[8] Hirschberg, K., Miller, C.M., Ellenberg, J., Presley, J.F., Siggia,
E.D., Phair, R.D. and Lippincott-Schwartz, J. (1998) Kinetic
analysis of secretory protein traffic and characterization of Golgi
to plasma membrane transport intermediates in living cells. J. Cell
Biol. 143, 1485–1503.
[9] Musch, A., Cohen, D., Kreitzer, G. and Rodriguez-Boulan, E.
(2001) Cdc42 regulates the exit of apical and basolateral proteins
from the TGN. EMBO J. 20, 2171–2179.
[10] Cao, H., Weller, S., Orth, J.D., Chen, J., Huang, B., Chen, J.L.,
Stamnes, M. and McNiven, M.A. (2005) Actin and Arf1-
dependent recruitment of a cortactin-dynamin complex to the
Golgi regulates post-Golgi transport. Nat. Cell Biol. 7, 483–492.
[11] Apodaca, G. (2001) Endocytic traffic in polarized epithelial cells:
role of the actin and microtubule cytoskeleton. Traffic 2, 149–159.
[12] Brown, B.K. and Song, W. (2001) The actin cytoskeleton is
required for the trafficking of the B cell antigen receptor to the late
endosomes. Traffic 2, 414–427.
[13] Mundy, D.I., Machleidt, T., Ying, Y.S., Anderson, R.G. and
Bloom, G.S. (2002) Dual control of caveolar membrane traffic by
microtubules and the actin cytoskeleton. J. Cell Sci. 115, 4327–
4339.
[14] Traub, L.M. (2005) Common principles in clathrin-mediated
sorting at the Golgi and the plasma membrane. Biochim. Biophys.
Acta 1744, 415–437.
[15] Bonifacino, J.S. and Traub, L.M. (2003) Signals for sorting of
transmembrane proteins to endosomes and lysosomes. Annu.
Rev. Biochem. 72, 395–447.
[16] Potter, B.A., Ihrke, G., Bruns, J.R., Weixel, K.M. and Weisz,
O.A. (2004) Specific N-glycans direct apical delivery of trans-
membrane, but not soluble or glycosylphosphatidylinositol-
anchored forms of endolyn in Madin-Darby canine kidney cells.
Mol. Biol. Cell 15, 1407–1416.
[17] Mun˜ iz, M. and Riezman, H. (2000) Intracellular transport of
GPI-anchored proteins. EMBO J. 19, 10–15.
[18] Schuck, S. and Simons, K. (2004) Polarized sorting in epithelial
cells: raft clustering and the biogenesis of the apical membrane. J.
Cell Sci. 117, 5955–5964.
[19] Paladino, S., Sarnataro, D., Tivodar, S. and Zurzolo, C. (2007)
Oligomerization is a specific requirement for apical sorting of
glycosyl-phosphatidylinositol-anchored proteins but not for non-
raft-associated apical proteins. Traffic 8, 251–258.
[20] Keller, P. and Simons, K. (1997) Post-Golgi biosynthetic
trafficking. J. Cell Sci. 110, 3001–3009.
[21] Rodrı
´guez-Boula
´n, E., Kreitzer, G. and Musch, A. (2005)
Organization of vesicular trafficking in epithelia. Nat. Rev. Mol.
Cell. Biol. 6, 233–247.
[22] Rustom, A., Bajohrs, M., Kaether, C., Keller, P., Toomre, D.,
Corbeil, D. and Gerdes, H.H. (2002) Selective delivery of
secretory cargo in Golgi-derived carriers of nonepithelial cells.
Traffic 3, 279–288.
[23] Caroni, P. (2001) New EMBO members’ review: actin cytoskel-
eton regulation through modulation of PI(4,5)P
2
rafts. EMBO J.
20, 4332–4336.
Fig. 4. Colocalization of YFP-GPI and the CFP-VSV-G wild-type form in the Golgi complex of COS-1 cells incubated at 19.5 C in the presence of
cycloheximide. Bar, 10 lm.
3880 F. La
´zaro-Die
´guez et al. / FEBS Letters 581 (2007) 3875–3881
[24] Musch, A., Xu, H., Shields, D. and Rodrı
´guez-Boula
´n, E. (1996)
Transport of vesicular stomatitis virus G protein to the cell
surface is signal mediated in polarized and nonpolarized cells. J.
Cell Biol. 133, 543–558.
[25] Rindler, M.J., Ivanov, I.E., Plesken, H., Rodrı
´guez-Boula
´n, E.
and Sabatini, D.D. (1984) Viral glycoproteins destined for
apical or basolateral plsma membrane domains traverse the
same Golgi apparatus during their intracellular transport in
doubly infected Madin-Darby canine kidney cells. J. Cell Biol. 98,
1304–1319.
[26] Lisanti, M.P., Caras, I.W., Davitz, M.A. and Rodrı
´guez-Boula
´n,
E. (1989) A glycophospholipid membrane anchor acts as an apical
targeting signal in polarized epithelial cells. J. Cell Biol. 109,
2145–2156.
[27] Yeaman, C., Le Gall, A.H., Baldwin, A.N., Monlauzeur, L.,
Le Bivic, A. and Rodriguez-Boulan, E. (1997) The O-glycosylated
stalk domain is required for apical sorting of neurotro-
phin receptors in polarized MDCK cells. J. Cell Biol. 139,
929–940.
[28] Keller, P., Toomre, D., Diaz, E., White, J. and Simons, K. (2001)
Multicolour imaging of post-Golgi sorting and trafficking in live
cells. Nat. Cell Biol. 3, 140–149.
[29] Yoshimori, T., Keller, P., Roth, M.G. and Simons, K. (1996)
Different biosynthetic transport routes to the plasma membrane in
BHK and CHO cells. J. Cell Biol. 133, 247–256.
[30] Schuck, S., Gerl, M.J., Ang, A., Manninen, A., Keller, P.,
Mellman, I. and Simons, K. (2007) Rab10 is involved in
basolateral transport in polarized Madin-Darby Canine Kidney
cells. Traffic 8, 47–60.
[31] Cohen, D., Musch, A. and Rodrı
´guez-Boula
´n, E. (2001) Selective
control of basolateral membrane protein polarity by Cdc42.
Traffic 2, 556–564.
[32] Kessels, M.M., Dong, J., Leibig, W., Westermann, P. and
Qualmann, B. (2006) Complexes of syndapin II with dynamin II
promote vesicle formation at the trans-Golgi network. J. Cell Sci.
119, 1504–1516.
[33] Kerkhoff, E., Simpson, J.C., Leberfinger, C.B., Otto, I.M.,
Doerks, T., Bork, P., Rapp, U.R., Raabe, T. and Pepperkok,
R. (2001) The Spir actin organizers are involved in vesicle
transport processes. Curr. Biol. 11, 1963–1968.
[34] Johnson, D., Lanahan, A., Buck, C.R., Sehgal, A., Morgan, C.,
Mercer, E., Bothwell, M. and Chao, M. (1986) Expression and
structure of the human NGF receptor. Cell 47, 545–554.
[35] Helms, J.B. and Zurzolo, C. (2004) Lipids as targeting signals:
lipid rafts and intracellular trafficking. Traffic 5, 247–254.
[36] Brown, D.A. and Rose, J.K. (1992) Sorting of GPI-anchored
proteins to glycolipid-enriched membrane subdomains during
transport to the apical cell surface. Cell 68, 533–544.
[37] Paladino, S., Pocard, T., Catino, M.A. and Zurzolo, C. (2006)
GPI-anchored proteins are directly targeted to the apical surface
in fully polarized MDCK cells. J. Cell Biol. 172, 1023–1034.
[38] De Matteis, M.A. and Godi, A. (2004) Protein-lipid interactions
in membrane trafficking at the Golgi complex. Biochim. Biophys.
Acta 1666, 264–274.
[39] Sarnataro, D., Paladino, S., Campana, V., Grassi, J., Nitsch, L.
and Zurzolo, C. (2002) PrPC is sorted to the basolateral
membrane of epithelial cells independently of its association with
rafts. Traffic 3, 810–821.
F. La
´zaro-Die
´guez et al. / FEBS Letters 581 (2007) 3875–3881 3881
... In contrast, much less is known about the subcellular localization and regulatory mechanisms of V-ATPase in the Golgi. Our group previously reported significant similarities between events occurring after microfilament disruption (with actin depolymerizing agents) (47)(48)(49) and those seen after the pharmacological inhibition of V-ATPase (with bafilomycin A1 and concanamycin A) (43,44,49). These similarities include: (a) membrane trafficking alterations in the Golgi-to-ER and post-Golgi protein transports; (b) the alkalinisation of the Golgi complex, and (c) strong dilatation of cisternae, observed under the electron microscope. ...
... Inverse fluorescence recovery after photobleaching (iFRAP) experiments were carried out using a Leica TCS SL with Argon and HeNe lasers attached to a Leica DMIRE2 inverted microscope equipped with an incubation system with temperature and CO 2 control as previously reported (48). All experiments were performed at 37°C and 5% CO 2 . ...
... On one hand, we previously reported that actin depolymerisation raises the pH in the Golgi and alters Golgi-to-ER and post-Golgi trafficking (47)(48)(49). These results are similar to those recorded after the pharmacological inhibition of V-ATPase using bafilomycin and concanamycin (44). ...
Article
Full-text available
We previously reported that actin-depolymerizing agents promote the alkalinisation of the Golgi stack and the trans-Golgi network (TGN). The main determinant of acidic pH at the Golgi is the Vacuolar-type H+-translocating ATPase (V-ATPase), whose V1 domain subunits B and C bind actin. We have generated a GFP-tagged subunit B2 construct (GFP-B2) that is incorporated into V1 domain, which in turn is coupled with V0 sector. GFP-B2 subunit is enriched at distal Golgi compartments in HeLa cells. Subcellular fractionation, immunoprecipitation and inversal FRAP experiments show that the actin depolymerisation promotes the dissociation of V1-V0 domains, which entails subunit B2 translocation from Golgi membranes to the cytosol. Moreover molecular interaction between subunits B2 and C1 and actin were detected. In addition, Golgi membrane lipid order disruption by D-ceramide-C6 causes Golgi pH alkalinisation. We conclude that actin regulates the Golgi pH homeostasis maintaining the coupling of V1-V0 domains of V-ATPase through the binding of microfilaments to subunits B and C and preserving the integrity of Detergent Resistant Membranes (DRMs) organization. These results establish the Golgi-associated V-ATPase activity as the molecular link between actin and the Golgi pH.
... While microtubules and dynein clearly play a major role in determining Golgi distribution and organization, recent studies have also proposed roles for actin, actin-associated proteins, and myosin motors in regulating Golgi morphology and function. For example, some or all of these molecules have been implicated in the creation, scission, and transport of vesicles to, within, and away from the Golgi (Campellone, Webb, Znameroski, & Welch 2008;Cao et al., 2005;Carreno, Engqvist-Goldstein, Zhang, McDonald, & Drubin, 2004;Chen, Lacomis, Erdjument-Bromage, Tempst, & Stamnes, 2004;Coudrier & Almeida, 2011;Duran et al., 2003;Guet et al., 2014;Heimann, Percival, Weinberger, Gunning, & Stow, 1999;Jacob, Heine, Alfalah, & Naim, 2003;Kirkbride et al., 2012;Lazaro-Dieguez et al., 2007;Matas, Martinez-Menarguez, & Egea, 2004;Miserey-Lenkei et al., 2010;Stow, Fath, & Burgess, 1998). Most relevant to this study, alterations in Golgi morphology following actin filament disassembly have been reported (Dippold et al., 2009;Egea, Lazaro-Dieguez, & Vilella, 2006;Valderrama et al., 1998), leading to a model wherein actin filaments attached to the sides of Golgi cisternae extend them laterally to create the classic ribbon architecture (Dippold et al., 2009;Egea, Serra-Peinado, Salcedo-Sicilia, & Gutierrez-Martinez, 2013). ...
... That said, arguments supporting such a role will require the identification and targeted disruption of actin networks that specifically associate with the Golgi, as assays involving global actin filament disassembly result in large changes in cell shape that preclude mechanistic insight. Finally, our conclusions do not preclude important roles for actin in Golgi processes like membrane tubulation and vesicle transport for which there is ample scientific support (Campellone et al., 2008;Cao et al., 2005;Carreno et al., 2004;Chen et al., 2004;Coudrier & Almeida, 2011;Duran et al., 2003;Guet et al., 2014;Heimann et al., 1999;Jacob et al., 2003;Kirkbride et al., 2012;Lazaro-Dieguez et al., 2007;Matas et al., 2004;Miserey-Lenkei et al., 2010;Stow et al., 1998). ...
Article
Full-text available
The peri-centrosomal localization and morphology of the Golgi apparatus depends largely on the microtubule cytoskeleton and the microtubule motor protein dynein. Recent studies proposed that myosin 18Aα (M18Aα) also contributes to Golgi morphology by binding the Golgi protein GOLPH3 and walking along adjacent actin filaments to stretch the Golgi into its classic ribbon structure. Biochemical analyses have shown, however, that M18A is not an actin-activated ATPase and lacks motor activity. Our goal, therefore, was to define the precise molecular mechanism by which M18Aα determines Golgi morphology. We show that purified M18Aα remains inactive in the presence of GOLPH3, arguing against the Golgi-specific activation of the myosin. Using M18A-specific antibodies and expression of GFP-tagged M18Aα, we find no evidence that it localizes to the Golgi. Moreover, several cell lines with reduced or eliminated M18Aα expression exhibited normal Golgi morphology. Interestingly, actin filament disassembly resulted in a marked reduction in lateral stretching of the Golgi in both control and M18Aα-deficient cells. Importantly, this reduction was accompanied by an expansion of the Golgi in the vertical direction, vertical movement of the centrosome, and increases in the height of both the nucleus and the cell. Collectively, our data indicate that M18Aα does not localize to the Golgi or play a significant role in determining its morphology, and suggest that global F-actin disassembly alters Golgi morphology indirectly by altering cell shape. This article is protected by copyright. All rights reserved.
... Therefore, AQP1-induced Golgi apparatus extension was ANXA2/F-actin dependent. Actin filaments are involved in the maintenance of Golgi apparatus morphology [35,36] Depolymerization of actin could alter Golgi morphology have been reported in series of studies [36,63]. Therefore, we speculate AQP1 combined with ANXA2 to regulate Golgi apparatus morphology through regulating actin rearrangement. ...
Article
Full-text available
Background Metastasis of breast cancer grows from the local invasion to the distant colonization. Blocking the local invasion step would be promising for breast cancer treatment. Our present study demonstrated AQP1 was a crucial target in breast cancer local invasion. Methods Mass spectrometry combined with bioinformatics analysis was used to identify AQP1 associated proteins ANXA2 and Rab1b. Co-immunoprecipitation, immunofluorescence assays and cell functional experiments were carried out to define the relationship among AQP1, ANXA2 and Rab1b and their re-localization in breast cancer cells. The Cox proportional hazards regression model was performed toward the identification of relevant prognostic factors. Survival curves were plotted by the Kaplan–Meier method and compared by the log-rank test. Results Here, we show that the cytoplasmic water channel protein AQP1, a crucial target in breast cancer local invasion, recruited ANXA2 from the cellular membrane to the Golgi apparatus, promoted Golgi apparatus extension, and induced breast cancer cell migration and invasion. In addition, cytoplasmic AQP1 recruited cytosolic free Rab1b to the Golgi apparatus to form a ternary complex containing AQP1, ANXA2, and Rab1b, which induced cellular secretion of the pro-metastatic proteins ICAM1 and CTSS. Cellular secretion of ICAM1 and CTSS led to the migration and invasion of breast cancer cells. Both in vivo assay and clinical analysis data confirmed above results. Conclusions Our findings suggested a novel mechanism for AQP1-induced breast cancer local invasion. Therefore, targeting AQP1 offers promises in breast cancer treatment.
... The actin cytoskeleton is not only critical for the maintenance of the Golgi structure and its mechanical properties but also provides the structural support favoring carrier biogenesis (15)(16)(17)(18). The Golgi exit of various cargoes is altered in cells treated with drugs either depolymerizing or stabilizing actin filaments (19,20), and the post-Golgi trafficking is affected either by the knockdown of the expression of some actinbinding proteins, which regulate actin dynamics, or by the overexpression of their mutants (12,(21)(22)(23), all together revealing the critical role of actin dynamics for protein trafficking. Only few studies have shown the involvement of actin remodeling proteins in polarized trafficking, mostly in selectively mediating the apical and basolateral trafficking of transmembrane proteins [refs. ...
Article
Significance Our findings represent a fundamental advance in general understanding of the mechanisms of exocytosis in polarized epithelial cells that are crucial for the establishment and maintenance of epithelial cell polarity. Moreover, our data also improve the knowledge on the machinery regulating polarized trafficking of glycosylphosphatidylinositol-anchored proteins, a class of lipid-associated proteins playing diverse vital functions, unraveling an unexpected role of calcium in their apical sorting.
... All these studies clearly indicate that actin cytoskeleton plays a critical role for basolateral and apical transport of transmebrane proteins, but not for GPI-APs. This is also consistent with a pioneering study showing that actin-stabilizing/depolymerizing agents inhibit the exit of apical and basolateral proteins (p75 and VSV-G) without affecting the exit of GPI-APs in Cos-7 cells [129]. Interestingly, FRAP experiments have shown that in unpolarized MDCK cells latrunculin A decreases the apparent diffusion coefficient of transmembrane p75 NTR , but not that of GPI-AP GFP-FR [25]. ...
Article
Glycosylphosphatidylinositol‐anchored proteins (GPI‐APs) are lipid‐anchored proteins attached to the extracellular leaflet of the plasma membrane via a glycolipid anchor. GPI‐APs are commonly associated with cholesterol‐ and sphingolipid‐enriched membrane microdomains. These microdomains help regulating various biological activities, by segregating different proteins and lipids in (nanoscale) membrane compartments. In fibroblasts, GPI‐APs form actin‐ and cholesterol‐dependent nanoclusters directly at the plasma membrane (PM). By contrast, in polarized epithelial cells GPI‐APs cluster in the Golgi apparatus, the major protein‐sorting hub for the secretory pathway. Golgi clustering is required for the selective sorting of GPI‐APs to the apical PM domain, but also regulates their organization and biological activities at the cell surface. In this review, we discuss recent advances in our understanding of the mechanism of GPI‐AP sorting to the apical membrane. We focus on the roles of the protein moiety, lipids, and calcium ions in the regulation of the clustering of GPI‐APs in the Golgi apparatus. This article is protected by copyright. All rights reserved.
... Accordingly, unbiased pulse-chase analysis of total secretory trafficking reveals that knockdown of GOLPH3 or MYO18A arrests overall secretion nearly as completely as treatment with Golgi poisons Brefeldin A or Golgicide A (Ng et al., 2013). Likewise, GOLPH3, MYO18A, and F-actin are required for efficient Golgi-to-PM trafficking of the vesicular stomatitis virus G glycoprotein (VSVG; Dippold et al., 2009;Farber-Katz et al., 2014;Hirschberg et al., 1998;Lá zaro-Dié guez et al., 2007), and exit of Hepatitis C virus from infected cells (Bishé et al., 2012). Overall, the data indicate that the PI4P/GOLPH3/MYO18A/F-actin complex plays a role in vesicle exit from the Golgi for efficient forward trafficking to the PM. ...
Article
Vesicle budding for Golgi-to-plasma membrane trafficking is a key step in secretion. Proteins that induce curvature of the Golgi membrane are predicted to be required, by analogy to vesicle budding from other membranes. Here, we demonstrate that GOLPH3, upon binding to the phosphoinositide PI4P, induces curvature of synthetic membranes in vitro and the Golgi in cells. Moreover, efficient Golgi-to-plasma membrane trafficking critically depends on the ability of GOLPH3 to curve the Golgi membrane. Interestingly, uncoupling of GOLPH3 from its binding partner MYO18A results in extensive curvature of Golgi membranes, producing dramatic tubulation of the Golgi, but does not support forward trafficking. Thus, forward trafficking from the Golgi to the plasma membrane requires the ability of GOLPH3 both to induce Golgi membrane curvature and to recruit MYO18A. These data provide fundamental insight into the mechanism of Golgi trafficking and into the function of the unique Golgi secretory oncoproteins GOLPH3 and MYO18A.
... SNP evoked depolymerization of the actin cytoskeleton in SK-MEL-28 and WM793 cells, regardless of the presence or absence of arginine in the culture medium (Appendix A, Supplementary Fig. 3). Noteworthy, attachment of many membrane-bound proteins to actin filaments determines cell morphology and influences cell adhesion and motility [52]. ...
Article
Anticancer therapy based on recombinant arginine-degrading enzymes has been proposed for the treatment of several types of malignant cells deficient in arginine biosynthesis. One of the predicted side effects of such therapy is restricted bioavailability of nitric oxide as arginine catabolic product. Prolonged NO limitation may lead to unwanted disturbances in NO-dependent vasodilation, cardiovascular and immune systems. This problem can be overcome by co-supplementation with exogenous NO donor. However, NO may potentially counteract anticancer effects of therapy based on arginine deprivation. In this study, we evaluate for the first time the effects of an exogenous NO donor, sodium nitroprusside, on viability and metastatic properties of two human melanoma cell lines SK-MEL-28 and WM793 under arginine-deprived conditions. It was revealed that NO did not rescue melanoma cells from specific effects evoked by arginine deprivation, namely decreased viability and induction of apoptosis, dramatically reduced motility, invasiveness and clonogenic potential. Moreover, sodium nitroprusside co-treatment augmented several of these antineoplastic effects. We report that a combination of NO-donor and arginine deprivation strongly and specifically impaired metastatic behaviour of melanoma cells. Thus, sodium nitroprusside can be considered as an adjuvant for the more efficient treatment of malignant melanoma and possibly other tumors with arginine-degrading enzymes.
... Likewise, GOLPH3 and MYO18A are required for Golgi-to-plasma membrane trafficking as measured by VSVG delivery to the plasma membrane (), total secretory flux by pulse-chase analysis (Ng et al., 2013), secretion of hepatitis C viral particles (Bishé et al., 2012), and for exit of anterograde trafficking vesicles from the Golgi (). Finally, F-actin has been demonstrated to be required for Golgi secretory function (Hirschberg et al., 1998; Lázaro-Diéguez et al., 2007). Taken together, the data indicate that the majority of trafficking from the Golgi to the plasma membrane depends on the PtdIns(4)P/GOLPH3/MYO18A/F- actin pathway. ...
Book
Full-text available
This eBook contains 13 reviews which address the molecular mechanisms of Golgi pathology in Parkinson and Alzheimer disease, amyotrophic lateral sclerosis (ALS) and spinal muscular atrophies, and discuss their potential relevance to the pathological loss of neuronal cell bodies, axons and synapses.
Article
MYO18A is a divergent member of the myosin family characterized by the presence of an amino-terminal PDZ domain. MYO18A has been found in a few different complexes involved in intracellular transport processes. MYO18A is found in a complex with LURAP1 and MRCK that functions in retrograde treadmilling of actin. It also has been found in a complex with PAK2, βPIX, and GIT1, functioning to transport that protein complex from focal adhesions to the leading edge. Finally, a high proportion of MYO18A is found in complex with GOLPH3 at the trans Golgi, where it functions to promote vesicle budding for Golgi-to-plasma membrane trafficking. Interestingly, MYO18A has been implicated as a cancer driver, as have other components of the GOLPH3 pathway. It remains uncertain as to whether or not MYO18A has intrinsic motor activity. While many questions remain, MYO18A is a fascinatingly unique myosin that is essential in higher organisms.
Article
The Golgi complex is the central unit of the secretory pathway, modifying, processing and sorting proteins and lipids to their correct cellular localisation. Changes to proteins at the Golgi complex can have deleterious effects on the function of this organelle, impeding trafficking routes through it, potentially resulting in disease. It is emerging that several Rho GTPase proteins, namely Cdc42, RhoBTB3, RhoA and RhoD are at least in part localised to the Golgi complex, and a number of studies have shown that dysregulation of their levels or activity can be associated with cellular changes which ultimately drive cancer progression. In this mini-review we highlight some of the recent work that explores links between form and function of the Golgi complex, Rho GTPases and cancer.
Article
Full-text available
Live cell, time-lapse microscopy was used to study trafficking of caveolin-1-GFP in stably expressing CHO cells. Multiple cytological and biochemical tests verified that caveolin-1-GFP was a reliable marker for endogenous caveolin-1. At steady state, most caveolin-1-GFP was either at the cell surface associated with invaginated caveolae or near the centrosome in caveosomes. Live cell fluorescence imaging indicated that while much of the caveolin-1-GFP in caveolae at the cell surface was relatively sessile, numerous, highly motile caveolin-1-GFP-positive vesicles were present within the cell interior. These vesicles moved at speeds ranging from 0.3-2 μm/second and movement was abolished when microtubules were depolymerized with nocodazole. In the absence of microtubules, cell surface invaginated caveolae increased more than twofold and they became organized into linear arrays. Complete depolymerization of the actin cytoskeleton with latrunculin A, by contrast, triggered rapid and massive movements of caveolin-positive structures towards the centrosomal region of the cell. The caveolar membrane system of CHO cells therefore appears to be comprised of three caveolin-1-containing compartments. These include caveolae that are confined to the cell surface by cortical actin filaments, the peri-centrosomal caveosomes and caveolar vesicles, which we call `cavicles', that move constitutively and bi-directionally along microtubules between the cell surface and caveosomes. The behavior of cavicles suggests that they function as transport intermediates between caveolae and caveosomes.
Article
Full-text available
Madin-Darby canine kidney (MDCK) cells can sustain double infection with pairs of viruses of opposite budding polarity (simian virus 5 [SV5] and vesicular stomatitis virus [VSV] or influenza and VSV), and we observed that in such cells the envelope glycoproteins of the two viruses are synthesized simultaneously and assembled into virions at their characteristic sites. Influenza and SV5 budded exclusively from the apical plasma membrane of the cells, while VSV emerged only from the basolateral surfaces. Immunoelectron microscopic examination of doubly infected MDCK cells showed that the influenza hemagglutinin (HA) and the VSV G glycoproteins traverse the same Golgi apparatus and even the same Golgi cisternae. This indicates that the pathways of the two proteins towards the plasma membrane do not diverge before passage through the Golgi apparatus and therefore that critical sorting steps must take place during or after passage of the glycoproteins through this organelle. After its passage through the Golgi, the HA accumulated primarily at the apical membrane, where influenza virion assembly occurred. A small fraction of HA did, however, appear on the lateral surface and was incorporated into the envelope of budding VSV virions. Although predominantly found on the basolateral surface, significant amounts of G protein were observed on the apical plasma membrane well before disruption of the tight junctions was detectable. Nevertheless, assembly of VSV virions was restricted to the basolateral domain and in doubly infected cells the G protein was only infrequently incorporated into the envelope of budding influenza virions. These observations indicate that the site of VSV budding is not determined exclusively by the presence of G polypeptides. Therefore, it is likely that, at least for VSV, other cellular or viral components are responsible for the selection of the appropriate budding domain.
Article
Full-text available
The biogenesis and maintenance of asymmetry is crucial to many cellular functions including absorption and secretion, signalling, development and morphogenesis. Here we have directly visualized the segregation and trafficking of apical (glycosyl phosphatidyl inositol-anchored) and basolateral (vesicular stomatitis virus glycoprotein) cargo in living cells using multicolour imaging of green fluorescent protein variants. Apical and basolateral cargo segregate progressively into large domains in Golgi/trans-Golgi network structures, exclude resident proteins, and exit in separate transport containers. These remain distinct and do not merge with endocytic structures suggesting that lateral segregation in the trans-Golgi network is the primary sorting event. Fusion with the plasma membrane was detected by total internal reflection microscopy and reveals differences between apical and basolateral carriers as well as new 'hot spots' for exocytosis.
Article
Full-text available
Quantitative time-lapse imaging data of single cells expressing the transmembrane protein, vesicular stomatitis virus ts045 G protein fused to green fluorescent protein (VSVG–GFP), were used for kinetic modeling of protein traffic through the various compartments of the secretory pathway. A series of first order rate laws was sufficient to accurately describe VSVG–GFP transport, and provided compartment residence times and rate constants for transport into and out of the Golgi complex and delivery to the plasma membrane. For ER to Golgi transport the mean rate constant (i.e., the fraction of VSVG–GFP moved per unit of time) was 2.8% per min, for Golgi to plasma membrane transport it was 3.0% per min, and for transport from the plasma membrane to a degradative site it was 0.25% per min. Because these rate constants did not change as the concentration of VSVG–GFP in different compartments went from high (early in the experiment) to low (late in the experiment), secretory transport machinery was never saturated during the experiments. The processes of budding, translocation, and fusion of post-Golgi transport intermediates carrying VSVG– GFP to the plasma membrane were also analyzed using quantitative imaging techniques. Large pleiomorphic tubular structures, rather than small vesicles, were found to be the primary vehicles for Golgi to plasma membrane transport of VSVG–GFP. These structures budded as entire domains from the Golgi complex and underwent dynamic shape changes as they moved along microtubule tracks to the cell periphery. They carried up to 10,000 VSVG–GFP molecules and had a mean life time in COS cells of 3.8 min. In addition, they fused with the plasma membrane without intersecting other membrane transport pathways in the cell. These properties suggest that the post-Golgi intermediates represent a unique transport organelle for conveying large quantities of protein cargo from the Golgi complex directly to the plasma membrane.
Article
Current model propose that in nonpolarized cells, transport of plasma membrane proteins to the surface occurs by default. In contrast, compelling evidence indicates that in polarized epithelial cells, plasma membrane proteins are sorted in the TGN into at least two vectorial routes to apical and basolateral surface domains. Since both apical and basolateral proteins are also normally expressed by both polarized and nonpolarized cells, we explored here whether recently described basolateral sorting signals in the cytoplasmic domain of basolateral proteins are recognized and used for post TGN transport by nonpolarized cells. To this end, we compared the inhibitory effect of basolateral signal peptides on the cytosol-stimulated release of two basolateral and one apical marker in semi-intact fibroblasts (3T3), pituitary (GH3), and epithelial (MDCK) cells. A basolateral signal peptide (VSVGp) corresponding to the 29-amino acid cytoplasmic tail of vesicular stomatitis virus G protein (VSVG) inhibited with identical potency the vesicular release of VSVG from the TGN of all three cell lines. On the other hand, the VSVG peptide did not inhibit the vesicular release of HA in MDCK cells not of two polypeptide hormones (growth hormone and prolactin) in GH3 cells, whereas in 3T3 cells (influenza) hemagglutinin was inhibited, albeit with a 3x lower potency than VSVG. The results support the existence of a basolateral-like, signal-mediated constitutive pathway from TGN to plasma membrane in all three cell types, and suggest that an apical-like pathway may be present in fibroblast. The data support cargo protein involvement, not bulk flow, in the formation of post-TGN vesicles and predict the involvement of distinct cytosolic factors in the assembly of apical and basolateral transport vesicles.
Article
The question of how membrane proteins are delivered from the TGN to the cell surface in fibroblasts has received little attention. In this paper we have studied how their post-Golgi delivery routes compare with those in epithelia] cells. We have analyzed the transport of the vesicular stomatitis virus G protein, the Semliki Forest virus spike glycoprotein, both basolateral in MDCK cells, and the influenza virus hemagglutinin, apical in MDCK cells. In addition, we also have studied the transport of a hemagglutinin mutant (Cys543Tyr) which is basolateral in MDCK cells. Aluminum fluoride, a general activator of heterotrimeric G proteins, inhibited the transport of the basolateral cognate proteins, as well as of the hemagglutinin mutant, from the TGN to the cell surface in BHK and CHO cells, while having no effect on the surface delivery of the wild-type hemagglutinin. Only wild-type hemagglutinin became insoluble in the detergent CHAPS during transport through the BHK and CHO Golgi complexes, whereas the basolateral marker proteins remained CHAPS-soluble. We also have developed an in vitro assay using streptolysin O-permeabilized BHK cells, similar to the one we have previously used for analyzing polarized transport in MDCK cells (Pimplikar, S.W., E. Ikonen, and K. Simons. 1994. J. Cell Biol. 125:1025-1035). In this assay anti-NSF and rab-GDI inhibited transport of Semliki Forest virus spike glycoproteins from the TGN to the cell surface while having little effect on transport of the hemagglutinin. Altogether these data suggest that fibroblasts have apical and basolateral cognate routes from the TGN to the plasma membrane.
Article
Glycosyl-phosphatidylinositol- (GPI) anchored proteins contain a large extracellular protein domain that is linked to the membrane via a glycosylated form of phosphatidylinositol. We recently reported the polarized apical distribution of all endogenous GPI-anchored proteins in the MDCK cell line (Lisanti, M. P., M. Sargiacomo, L. Graeve, A. R. Saltiel, and E. Rodriguez-Boulan. 1988. Proc. Natl. Acad. Sci. USA. 85:9557-9561). To study the role of this mechanism of membrane anchoring in targeting to the apical cell surface, we use here decay-accelerating factor (DAF) as a model GPI-anchored protein. Endogenous DAF was localized on the apical surface of two human intestinal cell lines (Caco-2 and SK-CO15). Recombinant DAF, expressed in MDCK cells, also assumed a polarized apical distribution. Transfer of the 37-amino acid DAF signal for GPI attachment to the ectodomain of herpes simplex glycoprotein D (a basolateral antigen) and to human growth hormone (a regulated secretory protein) by recombinant DNA methods resulted in delivery of the fusion proteins to the apical surface of transfected MDCK cells. These results are consistent with the notion that the GPI anchoring mechanism may convey apical targeting information.
Article
The B cell antigen receptor (BCR) plays two central roles in B cell activation: to internalize antigens for processing and presentation, and to initiate signal transduction cascades that both promote B cells to enter the cell cycle and facilitate antigen processing by accelerating antigen transport. An early event in B cell activation is the association of BCR with the actin cytoskeleton, and an increase in cellular F-actin. Current evidence indicates that the organization of actin filaments changes in response to BCR-signaling, making actin filaments good candidates for regulation of BCR-antigen targeting. Here, we have analyzed the role of actin filaments in BCR-mediated antigen transport, using actin filament-disrupting reagents, cytochalasin D and latrunculin B, and an actin filament-stabilizing reagent, jasplakinolide. Perturbing actin filaments, either by disrupting or stabilizing them, blocked the movement of BCR from the plasma membrane to late endosomes/lysosomes. Cytochalasin D-treatment dramatically reduced the rate of internalization of BCR, and blocked the movement of the BCR from early endosomes to late endosomes/lysosomes, without affecting BCR-signaling. Thus, BCR-trafficking requires functional actin filaments for both internalization and movement to late endosomes/lysosomes, defining critical control points in BCR-antigen targeting.
Article
The nucleotide sequence for the human nerve growth factor (NGF) receptor has been determined. The 3.8 kb receptor mRNA encodes a 427 amino acid protein containing a 28 amino acid signal peptide, an extracellular domain containing four 40 amino acid repeats with six cysteine residues at conserved positions followed by a serine/threonine-rich region, a single transmembrane domain, and a 155 amino acid cytoplasmic domain. The sequence of the extracellular domain of the NGF receptor predicts a highly ordered structure containing a negatively charged region that may serve as the ligand-binding site. This domain is conserved through evolution. Transfection of a full-length cDNA in mouse fibroblasts results in stable expression of NGF receptors that are recognized by monoclonal antibodies to the human NGF receptor and that bind [125I]NGF.