ArticlePDF Available

Recruitment of the endosomal WASH complex is mediated by the extended ‘tail’ of Fam21 binding to the retromer protein Vps35

Authors:

Abstract and Figures

The retromer complex is a conserved endosomal protein sorting complex that sorts membrane proteins into nascent endosomal tubules. The recognition of membrane proteins is mediated by the cargo-selective retromer complex, a stable trimer of the Vps35 (vacuolar protein sorting 35), Vps29 and Vps26 proteins. We have recently reported that the cargo-selective retromer complex associates with the WASH (Wiskott-Aldrich syndrome homologue) complex, a multimeric protein complex that regulates tubule dynamics at endosomes. In the present study, we show that the retromer-WASH complex interaction occurs through the long unstructured 'tail' domain of the WASH complex-Fam21 protein binding to Vps35, an interaction that is necessary and sufficient to target the WASH complex to endosomes. The Fam21-tail also binds to FKBP15 (FK506-binding protein 15), a protein associated with ulcerative colitis, to mediate the membrane association of FKBP15. Elevated Fam21-tail expression inhibits the association of the WASH complex with retromer, resulting in increased cytoplasmic WASH complex. Additionally, overexpression of the Fam21-tail results in cell-spreading defects, implicating the activity of the WASH complex in regulating the mobilization of membrane into the endosome-to-cell surface pathway.
Content may be subject to copyright.
Biochem. J. (2012) 442, 209–220 (Printed in Great Britain) doi:10.1042/BJ20111761 209
Recruitment of the endosomal WASH complex is mediated by the extended
‘tail’ of Fam21 binding to the retromer protein Vps35
Michael E. HARBOUR1, Sophia Y. BREUSEGEM1and Matthew N. J. SEAMAN2
Cambridge Institute for Medical Research (CIMR)/Department of Clinical Biochemistry, University of Cambridge, Addenbrookes Hospital, Cambridge CB2 0XY, U.K.
The retromer complex is a conserved endosomal protein sorting
complex that sorts membrane proteins into nascent endosomal
tubules. The recognition of membrane proteins is mediated
by the cargo-selective retromer complex, a stable trimer of
the Vps35 (vacuolar protein sorting 35), Vps29 and Vps26
proteins. We have recently reported that the cargo-selective
retromer complex associates with the WASH (Wiskott–Aldrich
syndrome homologue) complex, a multimeric protein complex
that regulates tubule dynamics at endosomes. In the present
study, we show that the retromer–WASH complex interaction
occurs through the long unstructured ‘tail’ domain of the WASH
complex–Fam21 protein binding to Vps35, an interaction that
is necessary and sufficient to target the WASH complex to
endosomes. The Fam21-tail also binds to FKBP15 (FK506-
binding protein 15), a protein associated with ulcerative colitis, to
mediate the membrane association of FKBP15. Elevated Fam21-
tail expression inhibits the association of the WASH complex
with retromer, resulting in increased cytoplasmic WASH complex.
Additionally, overexpression of the Fam21-tail results in cell-
spreading defects, implicating the activity of the WASH complex
in regulating the mobilization of membrane into the endosome-
to-cell surface pathway.
Key words: endosome, FK506-binding protein 15 (FKBP15),
retromer, sorting, tubule, Wiskott–Aldrich syndrome homologue
(WASH) complex.
INTRODUCTION
Endosomes operate as major sorting stations that receive proteins
and lipids from the cell surface via endocytosis as well as
from the biosynthetic pathway [1]. Additionally, membrane flux
through the endocytic system plays a key role in the regulation
of cell size and shape [2–4]. The fidelity of endosomal protein
sorting is therefore of profound physiological importance and
there are numerous examples of pathologies resulting from a
failure of correct protein sorting, including the Niemann–Pick
disease [5], Charcot–Marie–Tooth disease and HSP (hereditary
spastic paraplegia) [6,7]. There is also much evidence to suggest
a causal link between the endosomal protein sorting of amyloid
precursor protein and the mechanisms that govern production of
the neurotoxic amyloid β-peptide that is a key pathological event
in Alzheimer’s disease [8].
Endosomal protein sorting occurs concomitantly with increas-
ing endosomal acidity. The influx of protons into endosomes
is mediated by the V-ATPase (vacuolar ATPase) and regulates
the association of some ligands (e.g. lysosomal hydrolases)
with their respective receptor [9,10]. One of the principal
components of endosomal protein sorting is the retromer complex.
Retromer is an evolutionarily conserved protein complex that
is required for the localization of several membrane proteins
of physiological importance, including the CIMPR (cation-
independent mannose 6-phosphate receptor) [11,12], the Vps10
(Vps is vacuolar protein sorting)-family sorting receptors
sortilin and SorL1 [13,14] and the divalent cation transporter
DMT1 [15].
The retromer complex recognizes cargo proteins through a
trimeric complex comprising Vps35, Vps29 and Vps26 [16,17]
that is recruited to the endosomal membrane by the small GTPase
Rab7 [18,19]. The Vps35 protein is believed to play a key role in
cargo recognition [20], although the structural similarity between
Vps26 and the arrestin family hints that Vps26 may also act
in cargo recognition [21,22]. In addition to the cargo-recognition
complex, retromer-mediated endosome-to-Golgi retrieval also
utilizes the activity of members of the Snx (sorting nexin)-BAR
(Bin/amphiphysin/Rvs) subfamily of Snxs, Snx1 and Snx2, in
complex with Snx5 and Snx6 [23]. The Snx-BAR sorting nexins
mediate the tubulation of endosomal membranes through their
C-terminal BAR domains [24].
The cargo-selective retromer complex associates with a
number of accessory proteins, including the Rab GTPase-
activating protein, TBC1D5 [TBC (Tre-2/Bub2/Cdc16) domain
family, member 5], the WASH (Wiskott–Aldrich syndrome
homologue) complex and FKBP15 (FK506-binding protein 15)
[19,25], a protein implicated in both ulcerative colitis and growth
cone collapse in neurons [26,27]. The WASH complex is a protein
complex containing WASH1, a member of the WASP (Wiskott–
Aldrich syndrome protein)/WAVE (WASP verprolin homologous)
family of actin nucleating promoting factors, KIAA1033 (also
known as SWIP), Strumpellin and Fam21. Loss of function of the
WASH complex results in dysregulation of endosomal tubules,
and since the Strumpellin protein is mutated in HSP, the activity of
the WASH complex has been of interest to researchers studying
the underlying pathology of HSP and related neuropathies
[25,28,29]. Although the precise function of the WASH complex
in endosomal protein sorting is yet to be determined, recent
studies in Dictyostelium discoideum have revealed a role for the
WASH complex in regulating the localization of V-ATPase [30].
Initial reports of the WASH complex association with
endosomes [28,29] contained a number of interesting
discrepancies. For example, the work by Gomez and Billadeau
Abbreviations used: BAR, Bin/amphiphysin/Rvs; CAPZ, capping protein (actin filament) muscle Z-line; CCDC, coiled-coil-domain-containing; CIMPR,
cation-independent mannose 6-phosphate receptor; FKBP15, FK506-binding protein 15; GFP, green fluorescent protein; GST, glutathione transferase; HSP,
hereditary spastic paraplegia; KD, knockdown; siRNA, small interfering RNA; Snx, sorting nexin; V-ATPase, vacuolar ATPase; Vps, vacuolar protein sorting;
WASH, Wiskott–Aldrich syndrome homologue; WASP, Wiskott–Aldrich syndrome protein; WAVE, WASP verprolin homologous; Y2H, yeast two-hybrid.
1These authors contributed equally to this work.
2To whom correspondence should be addressed (email mnjs100@cam.ac.uk).
c
The Authors Journal compilation c
2012 Biochemical Society
210 M. E. Harbour, S. Y. Breusegem and M. N. J. Seaman
[28] did not detect Strumpellin or KIAA1033 in association
with Fam21 and WASH1, whereas the study by Derivery et al.
[29] struggled to detect Fam21 that, when observed, was often
proteolysed and partially degraded. This could indicate that
the WASH complex is not a stable single entity but rather a
looser association of the constituent proteins. Studies conducted
by our laboratory showed that the WASH complex comprising
Strumpellin, KIAA1033, Fam21 and WASH1 bound to retromer
as a single unit. Questions remain, however, as to the nature and
assembly of the endosomal WASH complex [25].
Structural similarities between the endosomal WASH complex
and the analogous plasma membrane-localized WAVE
complex have been recently highlighted [31]. An important
difference between the WASH and WAVE complexes, however,
is the large unstructured ‘tail’ domain of the Fam21 protein. This
clearly distinguishes Fam21 from its WAVE complex counterpart,
Abi. Although Abi is 370 amino acids in size, Fam21 is a
much larger protein of 1300 amino acids containing a predicted
globular ‘head’ domain (200 amino acids) at its N-terminus
with a long (1100 amino acids) unstructured ‘tail’ that contains
a binding site for the actin capping protein [32].
In the present study, we show that Vps35 directly binds
the unstructured tail of Fam21. We show that the interaction
between the Fam21-tail and Vps35 is sufficient to target the
Fam21-tail to endosomes, consistent with the requirement for
the cargo-selective retromer complex to mediate the endosomal
recruitment of the WASH complex. Additionally, we report that
the Fam21-tail can bind to the FKBP15 protein, mediating its
endosomal localization. Overexpression of the Fam21-tail results
in displacement of FKBP15 from the endosome membrane and
competes with the endogenous WASH complex for binding to
retromer, resulting in cell spreading defects, thereby implicating
the activity of the WASH complex in regulating cell shape.
EXPERIMENTAL
Antibodies, general reagents and biochemicals
Unless otherwise stated, chemicals and reagents were purchased
from Sigma. 125I-labelled Protein A used in Western blotting
was obtained from PerkinElmer. Restriction enzymes and other
molecular biology reagents were purchased from New England
Biolabs. siRNA (small interfering RNA) oligonucleotides were
purchased from Dharmacon. Oligofectamine and Optimem used
in siRNA transfections were purchased from Invitrogen.
The anti-Fam21 sera (used for immunofluorescence) and
the anti-Strumpellin sera were both obtained from Santa
Cruz Biotechnology, the anti-FKBP15 serum used for
immunofluorescence was purchased from Abcam and the
anti-FKBP15 antibody used for Western blotting was generated
against a GST (glutathione transferase)–FKBP15 fusion protein
as described previously [25]. Anti-WASH1 antibody was
purchased from Sigma or Millipore. Anti-Fam21 serum used
for Western blotting was produced using a commercial service
(Charles River Laboratories; Margate, Kent, U.K.) using a GST–
Fam21-head domain fusion protein to immunize a rabbit. The
antiserum was affinity purified using GST–Fam21-head domain
coupled with CNBr-activated Sepharose (Amersham Pharmacia).
Monoclonal anti-Snx1 and anti-EEA1 (early endosome antigen
1) were purchased from BD Biosciences. Monoclonal anti-
GFP (green fluorescent protein) antibody was purchased from
Invitrogen, as were fluorescently labelled secondary antibodies
used in immunofluorescence. Polyclonal antisera against GFP,
Vps26, Vps35 and Snx1 have been described previously
[12,19].
Cell culture and production of stably transfected cell lines
HeLa M cells [33] were used throughout the present study
and are referred to simply as HeLa cells. Cells were cultured
in Dulbecco’s modified Eagle’s medium with the addition of
100 units/ml penicillin and 100 μg/ml streptomycin, 10 %(v/v)
fetal calf serum and 0.5 mg/ml G418 as necessary. The cells
stably expressing Vps29–GFP, GFP–Vps35 and Hrs1–GFP have
been described previously [19,25]. Cells stably expressing GFP–
Snx3, GFP–Snx1 and GFP–Fam21 were generated by transfecting
HeLa cells with the appropriate GFP-tagged construct in the
pIRESneo2 vector (Clontech) using Effectene (Qiagen) according
to the manufacturer’s instructions. Transfectants were selected
using G418 (Gibco) at 0.5 mg/ml and colonies that were G418
resistant were screened for expression of the respective construct
by fluorescence or immunofluorescence microscopy.
Production of the GFP-tagged constructs
In the present study, constructs to express GFP–Snx3, GFP–Snx1
and GFP–Fam21-head or -tail were generated. Other GFP-tagged
constructs have been described previously [25]. An IMAGE
consortium [Integrated Molecular Analysis of Genomes and their
Expression consortium (at St. Louis, MO, U.S.A., and at the
Human Genome Mapping Project, Hinxton Hall, Cambridge,
U.K.)] EST (expressed sequence tag) containing full-length
human Snx3 was obtained from GeneService and was used as
a template for a Pfu-mediated PCR reaction to amplify Snx3 and
add BamHI and SalI sites to the 5-and3
-ends respectively. The
PCR product was cloned first using pCRblunt (Invitrogen) and
then sequenced to confirm the identity and fidelity of the PCR
reaction product. The Snx3 ORF (open reading frame) was then
subcloned into pEGFP-C1 at the BglII and SalI sites. The GFP–
Snx3 construct was excised from pEGFP-C1 by digestion with
NheI–BamHI and then cloned into pIRESneo2 that had also been
digested with NheI and BamHI.
The Fam21–GFP construct was described previously [25]. The
Fam21-head and -tail regions were amplified by Pfu-mediated
PCR incorporating BamHI and SalI sites at the 5-and3
-
ends respectively and then cloned into pCRblunt. Following
sequencing, the fragments were subcloned into pEGFP-C1 by
digestion with BglII and SalI and then subsequently subcloned
into pIRESneo2 by excision with NheI and BamHI.
The GFP–Snx1 construct was generated by subcloning the
murine Snx1 coding region (except the start methionine) from
pGEX [12] into pEGFP C1. The GFP–Snx1 construct was then
subcloned into pIRESneo2.
Y2H (yeast two-hybrid) interaction assays
These were performed essentially as described previously [25].
In some instances, constructs were generated by subcloning from
pEGFP-C1 into versions of the pGBT9 (‘bait’) and pGAD424
(‘prey’) vectors that had been modified so that the multi-cloning
sites of the two vectors were the same reading frame as pEGFP-
C1. All constructs used in the Y2H assays were sequenced to
confirm that an in-frame fusion was made.
Automated microscopy and cell spreading assay
Flasks of HeLa cells or HeLa cells expressing GFP-tagged
Vps35 or Fam21-tail were trypsinized and seeded on to 24-well
plates (Greiner) at comparable density, using up to four wells
per cell line. After 75, 150, 250 or 480 min incubation in a
37 C humidified incubator, cells were fixed at room temperature
(20 C) with 4 %(w/v) paraformaldehyde, permeabilized with
c
The Authors Journal compilation c
2012 Biochemical Society
The interactions of the Fam21-tail 211
0.1 %Triton X-100 and stained with Hoechst 33258 and Alexa
Fluor®594-phalloidin. Cell spreading was measured using a
Thermo Fisher Cellomics Arrayscan automated microscope,
using its cell spreading assay protocol and a ×10 objective lens.
For each experiment, 1000 single cells were measured in four
replicate wells at each time point for the cells expressing GFP-
tagged constructs; for the HeLa cells, 800 cells were measured
in duplicate wells. The average single-cell perimeter per well
was obtained from Cellomics vHSC software, whereas further
averaging and normalization was performed using Origin 8.1
(OriginLab).
Microscopy and bafilomycin treatment
Immunofluorescence microscopy was performed essentially
as described previously [25]. For the bafilomycin treatment
experiments described in the present study, a 200×stock solution
of bafilomycin was made by dissolving 10 μgin800 μlofDMSO.
This was divided into aliquots and stored at 20 C. A 1000×
stock solution of chloroquine was made up in water and stored
at 80 C.
HeLa cells were seeded on to coverslips 24 h prior to the
experiment. At 1 h after replacing the medium with fresh medium,
the cells were incubated with 100 nM bafilomycin or 100 μM
chloroquine for 4 h at 37 C. Control cells were treated with
just DMSO. At the end of the incubation period, the cells were
briefly washed with PBS and then fixed for 10 min at room
temperature with 4 %paraformaldehyde. The cells were then
permeabilized with 0.1 %Triton X-100 in PBS and then
blocked with 3 %BSA in PBS prior to incubation with primary
and secondary antibodies. After mounting the coverslips using
ProLong Anti-fade Gold (Invitrogen), the cells were viewed
and imaged using a Zeiss Axioplan epifluorescence microscope
operating with a Hamamatsu C10600 CCD camera (charge-
coupled-device camera). A total of 100 cells were blind scored
for Snx1 tubules in each of three independent experiments.
SDS/PAGE and Western blotting
SDS/PAGE was performed as described previously [12]. Western
blotting employed 125I-labelled Protein A for detection and was
performed as described previously [12]. Signals were obtained by
a phosphoimager (using a GE Typhoon Imager) or by exposure
to X-ray film.
Native immunoprecipitation and MS
Native immunoprecipitations were initially performed (in
Figures 2B and 3A) as described previously [24] using
the PBS +1%Triton X-100 lysis buffer. Subsequently (for
Figures 3B–3D) the buffer was changed to 20 mM Hepes/KOH
(pH 7.0), 50 mM potassium acetate, 1 mM EDTA, 200 mM
sorbitol and 0.1 %Triton X-100, but the methodology was
identical.
MS analysis of gel bands was performed as described
previously [19].
Cell fractionation assay
The assay to separate membrane-associated (pelletable – P)
proteins from cytosolic (soluble – S) proteins was performed as
described previously [19].
RESULTS
The interactions of Vps35 and Fam21
The Vps35, Vps29 and Vps26 proteins assemble to form the
cargo-selective retromer complex that is depicted schematically
in Figure 1(A). To better understand the interactions of Vps35,
we performed a systematic Y2H-based assay for protein–protein
interactions. In Figure 1(B), we show that full-length Vps35
binds to Vps26, Vps29, Fam21 and Snx3. The interactions with
Vps26 and Vps29 act as positive controls in the assay. The
interaction with Fam21 is consistent with our previously published
results [25]. The Vps35–Snx3 interaction is novel, although an
association between Snx3 and retromer has been demonstrated
by native immunoprecipitation [34,35].
Truncation of Vps35 so that the Vps29-binding domain is
separated from the rest of the protein indicated that Snx3 binds to
Vps35 at a site distinct from that of Vps29, but Fam21 interacts
only with full-length Vps35. Additional truncation of Vps35 to
define the Snx3 and Fam21 interaction sites was not possible
due to markedly increased auto-activation of the truncated Vps35
constructs (results not shown). As the Vps35–Snx3 interaction
and also the Vps35–Fam21 interaction were only observed when
Vps35 was expressed from the pGBT9 ‘bait’ plasmid, further
investigation of the interactions of Vps35 using the Y2H system
was not possible.
In addition to interacting with Snx3, Vps35 also binds to the
WASH complex protein Fam21. As stated previously, Fam21
comprises a predicted globular ‘head’ domain of 200 amino
acids and a large (1100 amino acids) unstructured ‘tail’. We
therefore set out to investigate the interactions of the head and tail
of Fam21 and determine whether the Vps35–Fam21 interaction
occurs via one domain or another. Using the Y2H system we tested
the Fam21 head and tail against the retromer proteins and several
retromer-interacting proteins. In Figure 1(C), full-length Fam21
protein demonstrates a robust interaction with KIAA1033/SWIP.
The Fam21–KIAA1033 interaction is mediated exclusively
through the Fam21-head domain, as no interaction was observed
for the Fam21-tail with KIAA1033/SWIP. The Fam21-head
domain is also able to bind to WASH1 and can weakly interact
with both Snx1 and Snx2.
The Fam21-tail interacts with Strumpellin, FKBP15 and the
actin capping protein CAPZa {CAPZ [capping protein (actin
filament) muscle Z-line], α}, interactions that are not observable
for full-length Fam21. By conducting the Y2H assay with the
Fam21 constructs expressed from the pGAD424 ‘prey’ vector,
auto-activation effects could be avoided, and in this configuration
we observe a robust interaction between Fam21 and Vps35 that
appears to be primarily mediated through the tail of Fam21,
whereas the head-domain of Fam21 demonstrates an interaction
with KIAA1033/SWIP (see Figure 1D).
Although we focused primarily on the interactions of Fam21
and Vps35, we also examined the interactions of WASH1,
FKBP15 and Strumpellin. In Supplementary Figure S1 (at
http://www.BiochemJ.org/bj/442/bj4420209add.htm) we show
that WASH1 interacts strongly with KIAA1033/SWIP and also
with Vps35. FKBP15 interacts strongly with itself and also with a
number of other proteins. Conversely, Strumpellin demonstrated
an interaction with KIAA1033/SWIP only.
Localization and
in vivo
interactions of the Fam21-tail
We next investigated whether the Fam21-head or -tail domain
mediate the endosomal localization of the Fam21 protein. GFP-
tagged Fam21-head or -tail constructs were generated and
c
The Authors Journal compilation c
2012 Biochemical Society
212 M. E. Harbour, S. Y. Breusegem and M. N. J. Seaman
Figure 1 Interactions of Vps35 and Fam21
(A) A simple schematic diagram showing the Vps35 protein and the approximate regions where Vps26 and Vps29 bind. The association of Vps26 with Vps35 requires the PRLYL motif at residue
100 in Vps35 [45]. (B) Vps35 in the Y2H ‘bait’ vector (pGBT9) was tested for interactions with a number of retromer and retromer-associated proteins. Interactions were observed for Vps26,
Vps29, Fam21 and Snx3. Truncation of Vps35 abolished the interaction with Fam21 but interactions with Vps26A and Vps29 were retained. (Cand D) Full-length Fam21 or the head or tail domains
were expressed in either the bait vector (pGBT9; C) or prey vector (pGAD424; D) and tested for interactions with retromer and retromer-associated proteins. Expression of the head or tail
domains separately resulted in strong auto-activating activity for the head and weaker auto-activating activity for the tail. In the case of the Fam21-head domain, it was necessary to conduct the
Y2H assays in the presence of 3-amino-1,2,4-triazole (3-AT) to reduce the growth from auto-activation. The Vps35–Fam21 interaction is mediated through the tail of Fam21, whereas the head is
responsible for binding to KIAA1033/SWIP.
transiently transfected into HeLa cells. In Figure 2(A), we show
that the GFP–Fam21-tail construct is sufficient for endosomal
localization and co-localizes with Vps26, similar to the full-length
Fam21–GFP construct. The GFP–Fam21-head domain appears to
be cytoplasmic.
To further investigate the interactions of the Fam21 protein,
we sought to generate a cell line stably expressing Fam21–GFP.
After repeated unsuccessful attempts, a GFP-positive cell line was
obtained in which 50 %of the cells expressed a GFP-tagged
protein. This cell line was used in a native immunoprecipitation
experiment applying methodology we have used recently [25].
In Figure 2(B), the proteins precipitated with anti-GFP from
cells expressing either Fam21–GFP or Vps29–GFP were analysed
by SDS/PAGE, MS and Western blotting. Vps29–GFP co-
immunoprecipitated other retromer proteins and also members
of the WASH complex. The Fam21–GFP protein was observed to
be much smaller than expected (the expected size on SDS/PAGE
is 220 kDa) and relatively few tryptic peptides from Fam21
were identified by MS compared with endogenous Fam21 in the
Vps29–GFP sample (see Figure 2C). The truncated Fam21 did,
however, associate with FKBP15. The interaction between Fam21
and FKBP15 was confirmed by Western blotting of samples
similar to those analysed by SDS/PAGE. In the experiments
presented below, the cells expressing the truncated Fam21–GFP
are designated Fam21–GFP(T).
The total absence of peptides from the N-terminal half of Fam21
in the sample from the Fam21–GFP(T) cells is inconsistent with
some form of post-lysis proteolysis. Also, as several attempts at
generating a cell line stably expressing the Fam21–GFP construct
were required before the Fam21–GFP(T) cell line was obtained,
we surmised that the truncation of the Fam21–GFP construct
occurred stochastically at the time the construct integrated
into the HeLa cell DNA. Attempts to define the N-terminus of
the truncated Fam21–GFP protein by Edman degradation were
unsuccessful due to the N-terminus being blocked. It is, however,
possible to assign the likely start methionine residue with some
confidence as there are relatively few methionine residues in the
Fam21 protein and the only methionine residue that could be
employed as a start methionine residue to produce the truncated
protein detected is at position 801 and is shown in Figure 2(C).
As the use of full-length Fam21–GFP to generate a stably
transfected cell line was problematic, we next used the GFP–
Fam21-tail and GFP–Fam21-head constructs to generate stably
expressing cell lines. We were repeatedly unsuccessful in
producing a GFP–Fam21-head-expressing cell line, but we did
obtain several different cell lines expressing the GFP–Fam21-tail
construct. These cell lines, designated GFP–Fam21-tail(A–D),
were used in native immunoprecipitation/MS experiments along
with positive and negative control cell lines.
Attempts to demonstrate an in vivo interaction between the
GFP–Fam21-tail and retromer were initially unreliable when
we used our standard lysis buffer comprising PBS and 1%
Triton X-100, although the GFP–Fam21-tail construct was able to
reliably co-immunoprecipitate the actin capping proteins CAPZa
and CAPZb (CAPZ, β) (see Figure 3A). We therefore switched
to using a Hepes/potassium acetate buffer with 0.1%Triton
c
The Authors Journal compilation c
2012 Biochemical Society
The interactions of the Fam21-tail 213
Figure 2 The tail of Fam21 is sufficient for targeting to endosomes
(A) HeLa cells were transiently transfected with GFP-tagged constructs of full-length Fam21, or just the head or tail. Both the full-length and the GFP-tail construct targeted to endosomes
and co-localized with Vps26 (indicated with arrowheads). Scale bar, 20 μm. (B) A truncated Fam21–GFP construct co-immunoprecipitates FKBP15. HeLa cells expressing either Vps29–GFP
or Fam21–GFP (a truncated construct) were lysed and treated with anti-GFP. The immunoprecipitations were analysed by SDS/PAGE, MS and Western blotting. FKBP15 was detected in both
immunoprecipitations but more strongly in the immunoprecipitation from Fam21–GFP expressing cells. (C) A schematic diagram showing where the Fam21–GFP construct has been truncated
post-transfection and where peptides detected by MS originate from.
X-100. The results of the large-scale native immunoprecipitation
experiments are shown in Figures 3(A) and 3(B). The GFP–
Fam21-tail cell lines A–C all express the full-length Fam21-tail
at varying levels. The GFP–Fam21-tail(D) cell line, however,
expressed a truncated form that constituted approximately
one-fifth of the Fam21-tail (see Supplementary Figure S2 at
http://www.BiochemJ.org/bj/442/bj4420209add.htm).
Using the Hepes-based lysis buffer, we detected retromer,
FKBP15 and the actin-capping proteins CAPZa and CAPZb in the
immunoprecipitations from cells expressing the full-length GFP–
Fam21-tail. Additionally, two proteins of unknown function,
CCDC22 (coiled-coil-domain-containing 22) and CCDC93, were
detected associated with GFP–Fam21-tail. These proteins were
largely absent in the lane from the GFP–Fam21-tail(D) cell line.
Interactions detected by MS were confirmed by Western blotting
(see Figure 3C).
The increased expression of the Fam21-tail is able to compete
with the endogenous WASH complex for retromer binding.
As shown in Figure 3(D), Vps26 co-immunoprecipitates the
WASH complex from HeLa cells and other cell lines that do not
express the full-length Fam21-tail, but the presence of Strumpellin
and endogenous Fam21 in the immunoprecipitations from the
GFP–Fam21-tail(A–C) cell lines was much reduced, particularly
when compared with the levels of Vps26 and Vps35 in those
immunoprecipitations. Protein levels in the lysates of the samples
shown in Figures 3(C) and 3(D) were examined by Western
blotting and are shown in Figure 3(E).
The reduction in the association of endogenous WASH complex
with retromer in the GFP–Fam21-tail expressing cells is not due
to effects on the WASH complex assembly as incubation of
lysates with anti-WASH sera was able to recover very similar
levels of Strumpellin and Fam21 (Figure 3F). This is consistent
with the Fam21-head domain mediating the interaction with
KIAA1033/SWIP and assembling into the WASH complex,
whereas the tail binds to retromer (through Vps35), FKBP15 and
the actin-capping proteins.
c
The Authors Journal compilation c
2012 Biochemical Society
214 M. E. Harbour, S. Y. Breusegem and M. N. J. Seaman
Figure 3
In vivo
interactions of the Fam21-tail
(A) HeLa cells stably transfected with various GFP constructs were lysed in PBS +1% Triton
X-100 (as we have used previously [25]) and the GFP-tagged proteins were immunoprecipitated
using anti-GFP. After SDS/PAGE, bands were excised and the proteins were identified using
MS. The actin-capping proteins CAPZa and CAPZb are readily detected in the lanes containing
full-length Fam21-tail or the truncated form characterized in Figure 2(B). (B) The same as in
(A), but the lysis buffer was 20 mM Hepes/KOH (pH 7.0), 50 mM potassium acetate, 1mM
EDTA, 200 mM sorbitol and 0.1% Triton X-100. The negative control was untransfected
HeLa cells. Retromer proteins are readily detected in the GFP–Fam21-tail(A-C) lanes (i.e.
full-length Fam21-tail). Two proteins of unknown function, CCDC22 and CCDC93, are also
detected in the Fam21-tail lanes. (C) Similar samples to those shown in (B) were analysed by
Western blotting. (D) Similar lysates to those in (B)and(C) were treated with anti-Vps26 to
immunoprecipitate retromer. The immunoprecipitations were analysed by Western blotting and
showed that increased Fam21-tail inhibits the association of endogenous WASH complex with
The cargo-selective retromer complex mediates the localization
of Fam21
The tail of Fam21 can bind Vps35 and is sufficient to target to
endosomes (see Figures 2 and 3). As shown in Supplementary
Figure S3(A) (at http://www.BiochemJ.org/bj/442/bj4420209add.
htm), in the GFP–Fam21-tail(B) cell line we observe that the
GFP–Fam21-tail is localized to Vps26-positive endosomes. The
GFP–Fam21-tail(A) and (C) cell lines demonstrated a similar
degree of co-localization (results not shown), but the GFP–
Fam21-tail(D) cell line that expresses the truncated Fam21-tail
did not exhibit any co-localization with retromer proteins and
the construct was instead cytoplasmic and also localized to the
nucleus (Supplementary Figure S3B).
Although the tail of Fam21 is sufficient to target to endosomes,
the Fam21-tail retains a requirement for retromer to be membrane-
associated. In Supplementary Figures S3(C) and S3(D), siRNA
KD (knockdown) of Vps26 or Vps35 resulted in no membrane-
associated GFP–Fam21-tail. The siRNA KD of FKBP15,
Strumpellin or endogenous Fam21, on the other hand, did not
cause the GFP–Fam21-tail protein to become cytosolic.
Fam21 mediates the localization of FKBP15
The tail of Fam21 binds the FKBP15 protein. We would therefore
predict that overexpression of the Fam21-tail will displace
FKBP15 from the membrane. In Figure 4(A), cells expressing
GFP–Fam21-tail (cell line B) were mixed with untransfected
HeLa cells and seeded on to coverslips. In cells expressing
GFP–Fam21-tail, the punctate localization of FKBP15 is much
reduced and the protein appears more cytosolic. The localization
of WASH1 was also affected, although to a lesser degree, whereas
the Vps26 localization did not appear to be affected.
Using a simple fractionation assay to separate membranes
from the cytosol, we observe a clear shift of FKBP15 from the
membrane fraction (P) to the cytosolic fraction (S) in the cell
lines expressing GFP–Fam21-tail (see Figures 4B and 4C), but
other cell lines expressing GFP-tagged proteins (e.g. GFP–Snx1)
do not elicit the same effect. Additionally, as the Fam21-tail can
bind to retromer through Vps35, it is able to compete with the
endogenous WASH complex for retromer binding and results in
a partial shift of the WASH complex into the cytosolic fraction as
more Strumpellin was detected in the soluble cytosolic fraction
in the cell lines expressing the full-length GFP–Fam21-tail (see
Figures 4B and 4D).
Although the overexpression of the Fam21-tail causes more
FKBP15 and WASH complex to become cytosolic, the effect
is not as pronounced as observed after siRNA KD of the
Vps26 protein. In Figures 4(E)–4(G), the amount of membrane-
associated FKBP15 and WASH complex (Strumpellin) was
quantitatively determined for the KDs of Vps26, FKBP15
or Fam21. The siRNA KD of either retromer (Vps26) or
Fam21 results in a shift of FKBP15 into the cytosolic fraction.
Additionally, as shown in Supplementary Figure S4 (http://www.
BiochemJ.org/bj/442/bj4420209add.htm), cells treated with
siRNA to KD the expression of Vps26, FKBP15 and Fam21
were fixed and labelled with antibodies against Vps26, FKBP15
retromer. (E) Lysates similar to those for (C)and(D) were analysed by Western blotting. The
levels of some proteins (e.g. FKBP15) vary somewhat between samples but differences in the
levels do not account for the levels detected in (C)and(D). (F) Lysates similar to those in
(A) were treated with antisera against WASH1. The expression of the GFP–Fam21-tail does not
affect the assembly of the WASH complex.
c
The Authors Journal compilation c
2012 Biochemical Society
The interactions of the Fam21-tail 215
Figure 4 The Fam21-tail mediates the membrane association of FKBP15
(A) The GFP–Fam21-tail(B) cells were mixed with untransfected HeLa cells and seeded on to coverslips. After 24 h, the cells were fixed and labelled with antibodies against GFP and either anti-Vps26,
anti-FKBP15 or anti-WASH1. Overexpression of the Fam21-tail does not affect the localization of Vps26 but localization of both FKBP15 and WASH1 is affected, appearing less punctate and more
diffuse in the GFP–Fam21-tail(B) cells. Scale bar, 20 μm. (B) Cells were fractionated into membrane (pelletable – P) and cytosolic (soluble – S) fractions and the samples were subjected to
SDS/PAGE and analysed by Western blotting. FKBP15 and (to a lesser extent) Strumpellin are shifted into the cytosolic fraction in both cell lines expressing the GFP–Fam21-tail construct. (Cand
D) Results from triplicate experiments were quantified and the level of membrane-associated FKBP15 (C) and Strumpellin (D) are shown. (E) Cells treated with various siRNAs were fractionated into
membrane (pelletable – P) and cytosolic (soluble – S) fractions and the samples were subjected to SDS/PAGE and analysed by Western blotting. FKBP15 becomes cytosolic after Vps26 or Fam21
KD. (Fand G) Results from triplicate experiments were quantified and the level of membrane-associated FKBP15 (F) and Strumpellin (G) are shown.
and Fam21 and co-stained with anti-Snx1 antibodies. Labelling of
endogenous FKBP15 and Fam21, although not especially strong,
showed co-localization with Snx1. The loss of Vps26 expression
results in both FKBP15 and Fam21 becoming cytosolic. KD of
FKBP15 does not, however, affect Vps26 or Fam21 localization,
but KD of Fam21 does abolish the localization of FKBP15.
Therefore FKBP15 localization to the endosome requires Fam21,
consistent with the interaction observed between the tail of Fam21
and FKBP15.
Inhibition of the V-ATPase also displaces FKBP15
Recent studies in Dictyostelium have reported that the WASH
complex regulates the luminal pH of endo-/lyso-somes by
mediating the sorting of the V-ATPase from lysosomes [30]. We
therefore asked whether the activity of the V-ATPase can influence
the functioning of the WASH complex using bafilomycin to
inhibit the V-ATPase. Following treatment with bafilomycin, we
observed that cells exhibited markedly less endosomally localized
FKBP15. Comparison of the labelling shown in Figures 5(A)
and 5(B) reveals that cells treated with bafilomycin have little
punctate FKBP15. Treatment with chloroquine, a weak base that
neutralizes endosomal and lysosomal pH, does not produce the
same effect (Figure 5C), indicating that the loss of FKBP15
localization is the result of inhibition of the V-ATPase and not
the result of perturbation of the pH gradient across the endosomal
membrane. We quantified the effect of bafilomycin on the level of
membrane-associated FKBP15 and observed a pronounced shift
(2-fold) of FKBP15 into the cytosolic fraction after treatment
with bafilomycin (see Figure 5D).
Interestingly, we also observed that cells treated with
bafilomycin exhibited almost no Snx1-positive tubules (see
Supplementary Figure S5 at http://www.BiochemJ.org/bj/442/
bj4420209add.htm), although, unlike FKBP15, Snx1 remained
associated with punctate endosomal structures positive for the
retromer protein Vps26.
Dominant-negative effects of the Fam21-tail on cell spreading
Over the course of these studies, we noticed that the cell lines
stably expressing the full-length GFP-tagged Fam21-tail were
slow to spread after trypsinization and seeding on to tissue culture-
treated plastic. Using an automated microscope, we therefore
examined the cell spreading of the four GFP–Fam21-tail cell lines
(A–D) along with cells expressing GFP–Vps35 and untransfected
HeLa cells. As shown in Figure 6, the GFP–Fam21-tail cell lines
c
The Authors Journal compilation c
2012 Biochemical Society
216 M. E. Harbour, S. Y. Breusegem and M. N. J. Seaman
Figure 5 Inhibition of V-ATPase redistributes FKBP15 into the cytoplasm
(AC) HeLa cells were treated with just DMSO (A), 100 nM bafilomycin (B) or 100 μM chloroquine (C) for 4 h. After fixation the cells were labelled with antibodies against Snx1 and either anti-Vps26,
anti-FKBP15 or anti-Fam21. Incubation with bafilomycin results in FKBP15 redistributing to the cytoplasm, but chloroquine treatment does not. Scalebar,20μm. (D) Control or bafilomycin-treated
cells were fractionated into membrane (pelletable – P) and cytosolic (soluble – S) fractions and the samples were subjected to SDS/PAGE and analysed by Western blotting. FKBP15 becomes
cytosolic after incubation with bafilomycin. Results from triplicate blots were quantified and graphed.
A–C all exhibit slower cell spreading when compared with GFP–
Vps35, GFP–Fam21-tail(D) and also untransfected HeLa cells. As
the GFP–Fam21-tail(D) cell line differs from the GFP–Fam21-
tail(A–C) cell lines only by virtue of expressing a truncated form
of the Fam21-tail, the lack of cell spreading defect in this cell line
suggests that the cell spreading defect in the GFP–Fam21-tail(A–
C) cell lines is the result of the full-length constructs titrating a
protein (or proteins) into inactive complexes.
DISCUSSION
In the present study, we have investigated the interactions of the
retromer protein Vps35 and the WASH complex protein Fam21.
The Vps35 protein forms part of the cargo-selective retromer
subcomplex and, as such, associates with Vps26 and Vps29 via
its N- and C-terminal regions respectively [36]. Vps35 has been
directly implicated in cargo selection [20], and it is therefore
important to elucidate how the interactions of Vps35 contribute
to endosomal protein sorting.
Determination of the distinct interactions of the Fam21-head
and -tail domains
In addition to identifying a novel Vps35–Snx3 interaction, we also
show that Vps35 binds to the unstructured ‘tail’ of the Fam21
protein of the WASH complex (see Figure 1B). These novel
interactions were observed using the Y2H system. This proved
a useful tool for examining a large number of binary interactions
of retromer and associated proteins. In some instances, however,
further examination of the interactions using the Y2H system were
curtailed due to the auto-activating effects of particular constructs.
Dissecting Fam21 into its head- and tail-domains revealed
interactions that were not observed for full-length Fam21.
For example, the interactions between the tail of Fam21 and
the FKBP15 and Strumpellin proteins were detected when
no interactions were observed for full-length Fam21 (see
Figures 1C and 1D). Similarly, the Fam21-head domain bound to
WASH1 and the Snx1 and Snx2 proteins when no such interaction
was observed for the full-length Fam21 protein. It is possible that
the head of Fam21 interacts with a region of the tail and
that this interaction prevents full-length Fam21 from interacting
with FKBP15 and Strumpellin in the Y2H experiments. A Fam21-
head–tail interaction could also explain why the head and tail
domains were individually auto-activating but full-length Fam21
did not auto-activate.
Attempts to generate a cell line expressing full-length GFP-
tagged Fam21 were unsuccessful, although a cell line expressing
a truncated version of the Fam21–GFP construct was eventually
obtained that revealed an interaction between Fam21 and FKBP15
(see Figure 2B). This interaction had been predicted from the
Y2H results and therefore provides a useful in vivo confirmation
of the validity of the Y2H results. Although the function of
FKBP15 is yet to be determined, it is a protein of interest
to researchers studying ulcerative colitis and has also been
implicated in neuronal cell function [26,27].
It seems likely that overexpression of the ‘head’ of Fam21
may be toxic, as no cell line stably expressing the GFP–Fam21-
head construct could be obtained. The reason for the apparent
cytotoxicity of the Fam21-head domain is currently unknown and
merits further investigation.
Vps35 is central to the endosomal recruitment of Fam21
When cell lines expressing GFP–Fam21-tail were generated, the
tail of Fam21 was found to interact with a number of proteins
in addition to FKBP15. The actin-capping proteins CAPZa and
c
The Authors Journal compilation c
2012 Biochemical Society
The interactions of the Fam21-tail 217
Figure 6 Overexpression of the Fam21-tail causes cell spreading defects
(A) Average single cell perimeter of HeLa cells and HeLa cells expressing GFP–Vps35 or
GFP–Fam21-tail(A–D) measured at four time points after seeding and normalized to the first time
point (75 min post-seeding). Cells expressing full-length GFP–Fam21-tail(A–C) show markedly
slower cell spreading kinetics compared with control HeLa cells or HeLa cells expressing
GFP–Vps35. This experiment was repeated three times; a representative experiment is shown. (B)
Representative images of the phalloidin (F-actin) stain at three time points post-seeding for HeLa
control cells, HeLa cells expressing GFP–Vps35 and HeLa cells expressing GFP–Fam21-tail(B).
Scale bar, 300 μm. Note the greater proportion of rounder, smaller cells at 8 h for the cells
expressing GFP–Fam21-tail(B). For the cell perimeter analysis in (A), only single cells in the
images are analysed.
CAPZb were readily detected in native immunoprecipitations of
the full-length GFP–Fam21-tail construct, as were the retromer
proteins Vps35, Vps29 and Vps26. Two proteins of unknown
function, CCDC22 and CCDC93, were also detected and, in the
case of CCDC22, confirmed by Western blotting. As CCDC22
was detected only in the GFP–Fam21-tail immunoprecipitations
and not when retromer components were immunoprecipitated,
the CCDC22 protein (and possibly CCDC93) may associate with
Fam21-tail only when the Fam21-tail is not bound to retromer
(see Figures 3A–3C).
The tail of Fam21 is sufficient for recruitment to the endosomal
membrane, a process that requires the retromer cargo-selective
complex (see Supplementary Figure S3). In vivo, an interaction
between Vps35 and the WASH1 protein (revealed in the Y2H ex-
periment) may also contribute to the membrane association of the
WASH complex. Indeed, the inability to co-immunoprecipitate
retromer with the GFP–Fam21-tail under conditions (namely
PBS +1%Triton X-100) in which GFP–Vps35 or Vps29–GFP
are able to co-immunoprecipitate endogenous WASH complex
may be explained by the absence of the contribution of the Vps35–
WASH1 interaction when native immunoprecipitations of GFP–
Fam21-tail constructs were initially performed. The loss of the
Vps35–WASH1 interaction therefore necessitated the use of
the lower stringency Hepes/potassium acetate buffer that was able
to retain the retromer–Fam21-tail interaction. Alternatively, the
Vps35–WASH1 interaction might be involved in regulating
the activity of the WASH1 protein. In either case, the central
role that Vps35 plays in mediating the recruitment of the WASH
complex could explain why loss of Vps35 function in Drosophila
results in extensive actin dysregulation along with defects in
endocytosis [37].
The elevated levels of Fam21-tail in the cell lines expressing
the GFP–Fam21-tail construct disrupted the association of
endogenous WASH complex with retromer, resulting in an
increased cytoplasmic localization of WASH1 and Strumpellin
(see Figures 3D and 4A–4D). This observation significantly
extends our original finding that retromer mediates recruitment
of the WASH complex, which was based on phenotypes observed
after siRNA KD of retromer proteins [25]. Additionally, because
no direct interaction has been observed between WASH1 and the
Fam21-tail domain, these results suggest that the WASH complex
is more likely to be a stable entity in which the Strumpellin,
KIAA1033, WASH1 and Fam21 proteins are recruited to the
endosome as a single unit.
As the overexpression of the Fam21-tail was also able to
induce the displacement of FKBP15 from the membrane, we
suggest that the interaction between Fam21 and FKBP15 may
be the driving force in mediating the membrane association
of FKBP15, a hypothesis supported by the loss of FKBP15
membrane association after Fam21 KD (see Figure 4).
A mechanistic link between endosomal acidification and protein
sorting
Studies in D. discoideum have shown that the WASH complex
is required to mediate the trafficking of the V-ATPase [30].
Results presented in the present paper demonstrate that FKBP15
requires Fam21 for its membrane association (see Figure 4).
Intriguingly, we observed that inhibition of V-ATPase activity
by bafilomycin also markedly reduces the membrane association
of FKBP15, an effect similar to that observed when the Fam21-
tail is overexpressed (see Figures 4 and 5). The coupling of the
V-ATPase activity to the association of FKBP15 with endosomes
(and thereby the WASH complex) could provide a mechanism
to ensure that FKBP15 is only recruited to the membrane when
the V-ATPase is active and the endosome has achieved a measure
of maturity. At this time, however, the precise role of FKBP15
in regulating WASH complex function, or other aspects of
endosomal protein sorting, remains to be determined.
In addition to the displacement of FKBP15 from the membrane
after bafilomycin treatment, we also observed an almost total
loss of Snx1 tubules (see Supplementary Figure S5). We do not
believe, however, that loss of endosomally localized FKBP15
leads to a defect in Snx1-tubule formation as we have not observed
any effect on Snx1-tubules or endosome-to-Golgi retrieval
after FKBP15 KD [25]. A block in Snx1-tubule formation after
inhibition of the V-ATPase is potentially highly significant as it
may provide a regulatory mechanism for ensuring that Snx1-
tubule formation does not occur too early in the endocytic
pathway. This observation may also explain the apparent defect in
endosome-to-Golgi retrieval after pharmacological abolishment
of endosomal acidity [38].
c
The Authors Journal compilation c
2012 Biochemical Society
218 M. E. Harbour, S. Y. Breusegem and M. N. J. Seaman
Figure 7 Schematic diagrams of the WASH complex and its role in endosomal protein sorting
(A) Schematic diagram of the WASH and WAVE complexes showing similar overall architecture (based on Jia et al. [31] and the results of the present study). A key difference is the very long
Fam21-tail domain that is not present in the orthologous WAVE protein, Abi. (B) Schematic diagram summarizing the different Fam21 constructs analysed in the present study and the interactions
detected for the respective constructs. (C) A model for the action of the WASH complex and retromer in regulating endosomal protein sorting. Recruitment of the WASH complex by retromer facilitates
WASH complex-mediated sorting of the V-ATPase and other membrane proteins.
Assembly and physiological role of the WASH complex
The dominant-negative effect(s) resulting from the overexpression
of the Fam21-tail did not extend to assembly of the WASH
complex as there was no apparent defect in the ability of WASH1
to co-immunoprecipitate the Strumpellin or endogenous Fam21
proteins, indicating that the WASH complex was intact and that
the assembly of Fam21 into the WASH complex is most likely to
be mediated by the interaction of the Fam21-head domain with
the KIAA1033/SWIP protein (see Figures 1C, 1D and 3F). The
results presented here are therefore complementary to the in vitro
structural studies of WASH complex assembly [31] and confirm
that the assembly of the WASH complex in vivo is similar to
the assembly of the WAVE complex. A schematic diagram of the
WASH complex and the analogous WAVE complex is shown in
Figure 7(A) and the interactions of the various Fam21 constructs
are summarized in Figure 7(B).
Although we did not observe any profound changes in the
localization of several membrane proteins [e.g. the transferrin
receptor, the CIMPR and LAMP-1 (lysosome-associated mem-
brane protein-1)] in cells expressing the GFP–Fam21-tail (results
not shown), we did observe a significant cell spreading defect
in all three cell lines expressing the full-length GFP–Fam21-tail.
This phenotype was not observed in cells expressing the truncated
GFP–Fam21-tail or GFP–Vps35 and therefore is likely to be due
to dominant-negative effects exerted by the full-length tail (see
Figure 6). Cell spreading following loss of adherence requires
the mobilization of endocytosed membrane to expand the area
of the plasma membrane [2-4]. Interestingly, the genes encoding
Fam21 and WASH1 homologues in the amoeba Naegleria gruberi
(annotated as AM46 and AM5 respectively) have been implicated
in amoeboid locomotion due to their absence in related organisms
incapable of amoeboid motility [39]. Amoeboid motility is
a process that is conceptually similar to the spreading of a
trypsinized HeLa cell that is newly adhered to the substratum.
It is also noteworthy that the two proteins of unknown function,
CCDC22 and CCDC93, that interact with the Fam21-tail, could
be involved in this pathway. The Drosophila melanogaster
c
The Authors Journal compilation c
2012 Biochemical Society
The interactions of the Fam21-tail 219
homologue of CCDC93 has been reported to interact with exo70
[40], a protein of the exocyst complex. CCDC22 has been shown
to bind to Copine 1 and 4, calcium-binding proteins that have
been implicated in membrane trafficking [41,42]. Additionally,
mutations in CCDC22 have recently been identified as causal in
X-linked inherited learning disability [43].
We have previously demonstrated that loss of WASH-complex
function does not profoundly affect endosome-to-Golgi retrieval
of a well-characterized retromer cargo protein, namely the CIMPR
[25]. We therefore suggest that the function of the WASH
complex is more acutely required for membrane trafficking from
endosomes to the cell surface, a pathway that is necessary for
cell spreading [2-4] and recycling of membrane proteins such as
the β-adrenergic receptor [44]. The retromer complex mediates
the endosomal recruitment of the WASH complex through Vps35
binding to the Fam21-tail domain and thereby contributes to the
efficiency of protein sorting into the endosome-to-cell surface
pathway (see Figure 7C).
AUTHOR CONTRIBUTION
Michael Harbour performed the Y2H analysis, generated constructs and cell lines
and performed preliminary native immunoprecipitation experiments. Sophia Breusegem
performed the quantitative cell spreading assay and Snx1-tubule analysis (in the
Supplementary online data). Matthew Seaman performed the immunofluorescence
experiments, cell fractionation assays and native immunoprecipitations and wrote the
manuscript.
ACKNOWLEDGEMENTS
We are grateful to Kamburapola Jayawardena (a.k.a. Jay – ‘the mass-spec-guy’) for MS
identification of gel bands.
FUNDING
This work was funded by the Medical Research Council through a Senior Fellowship
Award [grant number G0701444 (to M.N.J.S.)] and an additional research grant [grant
number G0700750]. Funding for MS in the CIMR was provided by the Wellcome Trust
through a Strategic Award [grant number 079895/Z/06].
REFERENCES
1 Jovic, M., Sharma, M., Rahajeng, J. and Caplan, S. (2010) The early endosome: a busy
sorting station for proteins at the crossroads. Histol. Histopathol. 25, 99–112
2 Balasubramanian, N., Scott, D. W., Castle, J. D., Casanova, J. E. and Schwartz, M. A.
(2007) Arf6 and microtubules in adhesion-dependent trafficking of lipid rafts. Nat. Cell
Biol. 9, 1381–1391
3Jovi
´
c, M., Naslavsky, N., Rapaport, D., Horowitz, M. and Caplan, S. (2007) EHD1
regulates β1 integrin endosomal transport: effects on focal adhesions, cell spreading and
migration. J. Cell Sci. 120, 802–814
4 Veale, K. J., Offenh¨
auser, C., Lei, N., Stanley, A. C., Stow, J. L. and Murray, R. Z. (2011)
VAMP3 regulates podosome organisation in macrophages and together with
Stx4/SNAP23 mediates adhesion, cell spreading and persistent migration. Exp. Cell Res.
317, 1817–1829
5 Rosenbaum, A. I. and Maxfield, F. R. (2011) Niemann–Pick type C disease: molecular
mechanisms and potential therapeutic approaches. J. Neurochem. 116, 789–795
6 Salinas, S., Proukakis, C., Crosby, A. and Warner, T. T. (2008) Hereditary spastic
paraplegia: clinical features and pathogenetic mechanisms. Lancet Neurol. 7, 1127–1138
7 Blackstone, C., O’Kane, C. J. and Reid, E. (2011) Hereditary spastic paraplegias:
membrane traffic and the motor pathway. Nat. Rev. Neurosci. 12, 31–42
8 Burd, C. G. (2011) Physiology and pathology of endosome-to-Golgi retrograde sorting.
Tra ffic 12, 948–955
9 Toei, M., Saum, R. and Forgac, M. (2010) Regulation and isoform function of the
V-ATPases. Biochemistry 49, 4715–4723
10 Marshansky, V. and Futai, M. (2008) The V-type H+-ATPase in vesicular trafficking:
targeting, regulation and function. Curr. Opin. Cell Biol. 20, 415–426
11 Arighi, C. N., Hartnell, L. M., Aguilar, R. C., Haft, C. R. and Bonifacino, J. S. (2004) Role
of the mammalian retromer in sorting of the cation-independent mannose 6-phosphate
receptor. J. Cell Biol. 165, 123–133
12 Seaman, M. N. (2004) Cargo-selective endosomal sorting for retrieval to the Golgi
requires retromer. J. Cell Biol. 165, 111–122
13 Canuel, M., Lefrancois, S., Zeng, J. and Morales, C. R. (2008) AP-1 and retromer play
opposite roles in the trafficking of sortilin between the Golgi apparatus and the
lysosomes. Biochem. Biophys. Res. Commun. 366, 724–730
14 Nielsen, M. S., Gustafsen, C., Madsen, P., Nyengaard, J. R., Hermey, G., Bakke, O., Mari,
M., Schu, P., Pohlmann, R., Dennes, A. and Petersen, C. M. (2007) Sorting by the
cytoplasmic domain of the amyloid precursor protein binding receptor SorLA. Mol. Cell
Biol. 27, 6842–6851
15 Tabuchi, M., Yanatori, I., Kawai, Y. and Kishi, F. (2010) Retromer-mediated direct sorting
is required for proper endosomal recycling of the mammalian iron transporter DMT1. J.
Cell Sci. 123, 756–766
16 Seaman, M. N. (2005) Recycle your receptors with retromer. Trends Cell Biol. 15, 68–75
17 Collins, BM. (2008) The structure and function of the retromer protein complex. Traffic 9,
1811–1822
18 Rojas, R., van Vlijmen, T., Mardones, G. A., Prabhu, Y., Rojas, A. L., Mohammed, S.,
Heck, A. J., Raposo, G., van der Sluijs, P. and Bonifacino, J. S. (2008) Regulation of
retromer recruitment to endosomes by sequential action of Rab5 and Rab7. J. Cell Biol.
183, 513–526
19 Seaman, M. N., Harbour, M. E., Tattersall, D., Read, E. and Bright, N. (2009) Membrane
recruitment of the cargo-selective retromer subcomplex is catalysed by the small GTPase
Rab7 and inhibited by the Rab-GAP TBC1D5. J. Cell Sci. 122, 2371–2382
20 Nothwehr, S. F., Ha, S. A. and Bruinsma, P. (2000) Sorting of yeast membraneproteins
into an endosome-to-Golgi pathway involves direct interaction of their cytosolic domains
with Vps35p. J. Cell Biol. 151, 297–310
21 Shi, H., Rojas, R., Bonifacino, J. S. and Hurley, J. H. (2006) The retromer subunit Vps26
has an arrestin fold and binds Vps35 through its C-terminal domain. Nat. Struct. Mol.
Biol. 13, 540–548
22 Collins, B. M., Norwood, S. J., Kerr, M. C., Mahony, D., Seaman, M. N., Teasdale, R. D.
and Owen, D. J. (2008) Structure of Vps26B and mapping of its interaction with the
retromer protein complex. Traffic 9, 366–379
23 Attar, N. and Cullen, P. J. (2010) The retromer complex. Adv. Enzyme Regul. 50, 216–236
24 Carlton, J., Bujny, M., Peter, B. J., Oorschot, V. M., Rutherford, A., Mellor, H.,
Klumperman, J., McMahon, H. T. and Cullen, P. J. (2004) Sorting nexin-1 mediates
tubular endosome-to-TGN transport through coincidence sensing of high-curvature
membranes and 3-phosphoinositides. Curr. Biol. 14, 1791–1800
25 Harbour, M. E., Breusegem, S. Y., Antrobus, R., Freeman, C., Reid, E. and Seaman, M. N.
(2010) The cargo-selective retromer complex is a recruiting hub for protein complexes
that regulate endosomal tubule dynamics. J. Cell Sci. 123, 3703–3717
26 Viklund, I. M., Aspenstr ¨
om, P., Meas-Yedid, V., Zhang, B., Kopec, J., Agren, D., Schneider,
G., D’Amato, M., Olivo-Marin, J. C., Sansonetti, P., Van Nhieu, G. T. and Pettersson, S.
(2009) WAFL, a new protein involved in regulation of early endocytic transport at the
intersection of actin and microtubule dynamics. Exp. Cell Res. 15, 1040–1052
27 Nakajima, O., Nakamura, F., Yamashita, N., Tomita, Y., Suto, F., Okada, T., Iwamatsu, A.,
Kondo, E., Fujisawa, H., Takei, K. and Goshima, Y. (2006) FKBP133: a novel mouse
FK506-binding protein homolog alters growth cone morphology. Biochem. Biophys. Res.
Commun. 346, 140–149
28 Gomez, T. S. and Billadeau, D. D. (2009) A FAM21-containing WASH complex regulates
retromer-dependent sorting. Dev. Cell 17, 699–711
29 Derivery, E., Sousa, C., Gautier, J. J., Lombard, B., Loew, D. and Gautreau, A. (2009) The
Arp2/3 activator WASH controls the fission of endosomes through a large multiprotein
complex. Dev. Cell 17, 712–723
30 Carnell, M., Zech, T., Calaminus, S. D., Ura, S., Hagedorn, M., Johnston, S. A., May, R.
C., Soldati, T., Machesky, L. M. and Insall, R. H. (2011) Actin polymerization driven by
WASH causes V-ATPase retrieval and vesicle neutralization before exocytosis. J. Cell Biol.
193, 831–839
31 Jia, D., Gomez, T. S., Metlagel, Z., Umetani, J., Otwinowski, Z., Rosen, M. K. and
Billadeau, D. D. (2010) WASH and WAVE actin regulators of the Wiskott–Aldrich
syndrome protein (WASP) family are controlled by analogous structurally related
complexes. Proc. Natl. Acad. Sci. U.S.A. 107, 10442–10447
32 Hernandez-Valladares, M., Kim, T., Kannan, B., Tung, A., Aguda, A. H., Larsson, M.,
Cooper, J. A. and Robinson, R. C. (2010) Structural characterization of a capping protein
interaction motif defines a family of actin filament regulators. Nat. Struct. Mol. Biol. 17,
497–503
33 Tiwari, R. K., Kusari, J. and Sen, G. C. (1987) Functional equivalents of
interferon-mediated signals needed for induction of an mRNA can be generated by
double-stranded RNA and growth factors. EMBO J. 6, 3373–3378
34 Strochlic, T. I., Setty, T. G., Sitaram, A. and Burd, C. G. (2007) Grd19/Snx3p functions as a
cargo-specific adapter for retromer-dependent endocytic recycling. J. Cell Biol. 177,
115–125
35 Harterink, M., Port, F., Lorenowicz, M. J., McGough, I. J., Silhankova, M., Betist, M. C.,
van Weering, J. R., van Heesbeen, R. G., Middelkoop, T. C., Basler, K. et al. (2011) A
SNX3-dependent retromer pathway mediates retrograde transport of the Wnt sorting
receptor Wntless and is required for Wnt secretion. Nat. Cell Biol. 13, 914–923
c
The Authors Journal compilation c
2012 Biochemical Society
220 M. E. Harbour, S. Y. Breusegem and M. N. J. Seaman
36 Hierro, A., Rojas, A. L., Rojas, R., Murthy, N., Effantin, G., Kajava, A. V., Steven, A. C.,
Bonifacino, J. S. and Hurley, J. H. (2007) Functional architecture of the retromer
cargo-recognition complex. Nature 449, 1063–1067
37 Korolchuk, V. I., Sch¨
utz, M. M., G´
omez-Llorente, C., Rocha, J., Lansu, N. R., Collins,
S. M., Wairkar, Y. P., Robinson, I. M. and O’Kane, C. J. (2007)
Drosophila
Vps35 function
is necessary for normal endocytic trafficking and actin cytoskeleton organisation. J. Cell
Sci. 120, 4367–4376
38 Chapman, R. E. and Munro, S. (1994) Retrieval of TGN proteins from the cell surface
requires endosomal acidification. EMBO J. 13, 2305–2312
39 Fritz-Laylin, L. K., Prochnik, S. E., Ginger, M. L., Dacks, J. B., Carpenter, M. L.,
Field, M. C., Kuo, A., Paredez, A., Chapman, J., Pham, J. et al. (2010) The
genome of
Naegleria gruberi
illuminates early eukaryotic versatility. Cell 140,
631–642
40 Formstecher, E., Aresta, S., Collura, V., Hamburger, A., Meil, A., Trehin, A., Reverdy, C.,
Betin, V., Maire, S., Brun, C. et al. (2005) Protein interaction mapping: a
Drosophila
case
study. Genome Res. 15, 376–384
41 Damer, C. K., Bayeva, M., Kim, P. S., Ho, L. K., Eberhardt, E. S., Socec, C. I., Lee, J. S.,
Bruce, E. A., Goldman-Yassen, A. E. and Naliboff, L. C. (2007) Copine A is required for
cytokinesis, contractile vacuole function, and development in
Dictyostelium
. Eukaryot.
Cell 6, 430–442
42 Tomsig, J. L., Snyder, S. L. and Creutz, C. E. (2003) Identification of targets for calcium
signaling through the copine family of proteins. Characterization of a coiled-coil
copine-binding motif. J. Biol. Chem. 278, 10048–10054
43 Voineagu, I., Huang, L., Winden, K., Lazaro, M., Haan, E., Nelson, J., McGaughran, J.,
Nguyen, L. S., Friend, K., Hackett, A. et al. (2011) CCDC22: a novel candidate gene for
syndromic X-linked intellectual disability. Mol. Psychiatry, doi:10.1038/mp.2011.95
44 Temkin, P., Lauffer, B., J¨
ager, S., Cimermancic, P., Krogan, N. J. and von Zastrow, M.
(2011) SNX27 mediates retromer tubule entry and endosome-to-plasma membrane
trafficking of signalling receptors. Nat. Cell Biol. 13, 715–721
45 Gokool, S., Tattersall, D., Reddy, J. V. and Seaman, M. N. (2007) Identification of a
conserved motif required for Vps35p/Vps26p interaction and assembly of the retromer
complex. Biochem. J. 408, 287–295
Received 3 October 2011/9 November 2011; accepted 9 November 2011
Published as BJ Immediate Publication 9 November 2011, doi:10.1042/BJ20111761
c
The Authors Journal compilation c
2012 Biochemical Society
Biochem. J. (2012) 442, 209–220 (Printed in Great Britain) doi:10.1042/BJ20111761
SUPPLEMENTARY ONLINE DATA
Recruitment of the endosomal WASH complex is mediated by the extended
‘tail’ of Fam21 binding to the retromer protein Vps35
Michael E. HARBOUR1, Sophia Y. BREUSEGEM1and Matthew N. J. SEAMAN2
Cambridge Institute for Medical Research (CIMR)/Department of Clinical Biochemistry, University of Cambridge, Addenbrookes Hospital, Cambridge CB2 0XY, U.K.
Figure S1 The interactions of WASH1, FKBP15 and Strumpellin were analysed using the Y2H system
WASH1 demonstrated interactions with KIAA1033 and VPS35. FKBP15 interacted most strongly with itself and also with Fam21. Strumpellin only interacted with KIAA1033.
1These authors contributed equally to this work.
2To whom correspondence should be addressed (email mnjs100@cam.ac.uk).
c
The Authors Journal compilation c
2012 Biochemical Society
M. E. Harbour, S. Y. Breusegem and M. N. J. Seaman
Figure S2 GFP–Fam21-tail constructs
(A) Untransfected HeLa cells or cells expressing GFP-tagged Fam21 constructs were lysed (using PBS +1 % Triton X-100 buffer) and treated with anti-GFP antibody to precipitate (IP) the respective
GFP-tagged protein. The precipitated proteins were analysed by SDS/PAGE and MS. Gel bands marked with a circle were excised and subjected to MALDI–TOF MS (matrix-assisted laser-desorption
ionization–time-of-flight MS) as described previously [1]. The red-filled circles denote bands corresponding to the GFP-tagged Fam21 construct. The protein(s) identified from each band are
shown below the gel. Actin-capping proteins (CAPZa and CAPZb) are readily identifiable in samples that contain C-terminal regions of the Fam21-tail. (B) Peptides identified in gel bands of the
GFP-tagged Fam21 constructs are highlighted (in red boxes) on the Fam21 sequence. The darker blue shaded area is the Fam21-head domain. The light blue shaded area denotes the actin-capping
protein-binding region [2]. The region underlined in the GFP-Fam21(D) data is the region where the construct is likely to be truncated on the basis of the peptides detected in this sample and the
full-length GFP–Fam21-tail(A) sample. The Fam21-tail(D) construct is truncated prior to the actin-capping protein binding region and therefore does not interact with CAPZa or CAPZb.
c
The Authors Journal compilation c
2012 Biochemical Society
The interactions of the Fam21-tail
Figure S3 The tail of Fam21 co-localizes with retromer and requires retromer for its membrane association
(A) The GFP–Fam21-tail(B) cell line was fixed and labelled with anti-GFP and anti-Vps26 antibodies. Scale bar, 20 μm. The regions of the micrograph contained in the boxes is shown as merged
images in the right-hand panels. (B) The GFP–Fam21-tail(D) cell line that expresses a truncated tail does not co-localize with retromer. Scale bar, 20 μm. (C) The GFP–Fam21-tail(B) cell line
was treated with siRNA to KD expression of Vps26 or Vps35. Loss of the cargo-selective retromer complex results in the GFP–Fam21-tail construct becoming cytosolic, but siRNA KD of FKBP15,
Strumpellin or endogenous Fam21 (shown in D) does not affect the localization of the GFP–Fam21-tail(B) construct. Coincident labelling of GFP–Fam21-tail and Snx1 is indicated by arrowheads.
Scale bar, 20 μm. (E) Efficacy of the siRNA KDs. Lysates from HeLa cells treated with siRNA to KD expression of retromer, FKBP15 or WASH complex proteins were subjected to SDS/PAGE and
analysed by Western blotting. The siRNA efficiently silences expression of its respective target, and in some cases, loss of one protein (e.g. Vps26) leads to instability and loss of another (e.g. Vps35).
c
The Authors Journal compilation c
2012 Biochemical Society
M. E. Harbour, S. Y. Breusegem and M. N. J. Seaman
Figure S4 Fam21 is required for the membrane association of FKBP15
(A) HeLa cells were treated with siRNA to KD Vps26, FKBP15 or Fam21. After fixation, the cells were labelled with antibodies against Snx1 and either Vps26, FKBP15 or Fam21. The punctate
localization of FKBP15 and Fam21 was lost after Vps26 KD, and FKBP15 localization was abolished by Fam21 KD, but FKBP15 KD did not markedly affect Fam21 localization. Scale bar, 20 μm.
Figure S5 Absence of Snx1 tubules after bafilomycin treatment
HeLa cells were treated with DMSO, 100 nM bafilomycin or 100 μM chloroquine for 4 h, fixed and labelled with anti-Snx1 and anti-VPS26 antibodies. Scale bar, 20 μm. (B) Cells were treated as in
(A) and 100 cells were blind scored in each of three independent experiments. Results are means +
S.D.
REFERENCES
1 Seaman, M. N., Harbour, M. E., Tattersall, D., Read, E. and Bright, N. (2009) Membrane
recruitment of the cargo-selective retromer subcomplex is catalysed by the small GTPase
Rab7 and inhibited by the Rab-GAP TBC1D5. J. Cell Sci. 122, 2371–2382
2 Hernandez-Valladares, M., Kim, T., Kannan, B., Tung,A., Aguda, A. H., Larsson, M., Cooper,
J. A. and Robinson, R. C. (2010) Structural characterization of a capping protein interaction
motif defines a family of actin filament regulators. Nat. Struct. Mol. Biol. 17, 497–503
Received 3 October 2011/9 November 2011; accepted 9 November 2011
Published as BJ Immediate Publication 9 November 2011, doi:10.1042/BJ20111761
c
The Authors Journal compilation c
2012 Biochemical Society
... The major difference corresponds to FAM21 which is a much larger protein ($1300 amino acids) compared to its counterpart subunit (ABI, $370 amino acids) in the WRC complex. FAM21 is formed by a small ($200 amino acids) "head" domain required for the interaction with other WASH complex subunits and a long ($1100 amino acids) "tail," mostly unstructured, that mediates direct binding to several proteins including the acting-capping proteins CAPZa and CAPZb (Hernandez-Valladares et al., 2010), the DNAJC13 protein, known as receptor-mediated endocytosis-8 (RME-8) (Freeman et al., 2014), the CCDC22 and CCDC93 subunits of the CCC (COMMD/CCDC22/CCDC93) complex (Harbour et al., 2012;Phillips-Krawczak et al., 2015), the FK506-binding protein 15 (FKBP15) (Harbour et al., 2012;Nooh & Bahouth, 2017), the cargo adaptor SNX27 (Lee et al., 2016;Steinberg et al., 2013), and the VPS35 subunit of the retromer complex (Harbour et al., 2012;Helfer et al., 2013;Jia et al., 2012) ( Figure 1a). This network of interactions suggests different mechanisms for controlling actin polymerization on endosomes (Simonetti & Cullen, 2019). ...
... The major difference corresponds to FAM21 which is a much larger protein ($1300 amino acids) compared to its counterpart subunit (ABI, $370 amino acids) in the WRC complex. FAM21 is formed by a small ($200 amino acids) "head" domain required for the interaction with other WASH complex subunits and a long ($1100 amino acids) "tail," mostly unstructured, that mediates direct binding to several proteins including the acting-capping proteins CAPZa and CAPZb (Hernandez-Valladares et al., 2010), the DNAJC13 protein, known as receptor-mediated endocytosis-8 (RME-8) (Freeman et al., 2014), the CCDC22 and CCDC93 subunits of the CCC (COMMD/CCDC22/CCDC93) complex (Harbour et al., 2012;Phillips-Krawczak et al., 2015), the FK506-binding protein 15 (FKBP15) (Harbour et al., 2012;Nooh & Bahouth, 2017), the cargo adaptor SNX27 (Lee et al., 2016;Steinberg et al., 2013), and the VPS35 subunit of the retromer complex (Harbour et al., 2012;Helfer et al., 2013;Jia et al., 2012) ( Figure 1a). This network of interactions suggests different mechanisms for controlling actin polymerization on endosomes (Simonetti & Cullen, 2019). ...
... The major difference corresponds to FAM21 which is a much larger protein ($1300 amino acids) compared to its counterpart subunit (ABI, $370 amino acids) in the WRC complex. FAM21 is formed by a small ($200 amino acids) "head" domain required for the interaction with other WASH complex subunits and a long ($1100 amino acids) "tail," mostly unstructured, that mediates direct binding to several proteins including the acting-capping proteins CAPZa and CAPZb (Hernandez-Valladares et al., 2010), the DNAJC13 protein, known as receptor-mediated endocytosis-8 (RME-8) (Freeman et al., 2014), the CCDC22 and CCDC93 subunits of the CCC (COMMD/CCDC22/CCDC93) complex (Harbour et al., 2012;Phillips-Krawczak et al., 2015), the FK506-binding protein 15 (FKBP15) (Harbour et al., 2012;Nooh & Bahouth, 2017), the cargo adaptor SNX27 (Lee et al., 2016;Steinberg et al., 2013), and the VPS35 subunit of the retromer complex (Harbour et al., 2012;Helfer et al., 2013;Jia et al., 2012) ( Figure 1a). This network of interactions suggests different mechanisms for controlling actin polymerization on endosomes (Simonetti & Cullen, 2019). ...
Article
Full-text available
Endosomal trafficking ensures the proper distribution of lipids and proteins to various cellular compartments, facilitating intracellular communication, nutrient transport, waste disposal, and the maintenance of cell structure. Retromer, a peripheral membrane protein complex, plays an important role in this process by recruiting the associated actin‐polymerizing WASH complex to establish distinct sorting domains. The WASH complex is recruited through the interaction of the VPS35 subunit of retromer with the WASH complex subunit FAM21. Here, we report the identification of two separate fragments of FAM21 that interact with VPS35, along with a third fragment that binds to the VPS29 subunit of retromer. The crystal structure of VPS29 bound to a peptide derived from FAM21 shows a distinctive sharp bend that inserts into a conserved hydrophobic pocket with a binding mode similar to that adopted by other VPS29 effectors. Interestingly, despite the network of interactions between FAM21 and retromer occurring near the Parkinson's disease‐linked mutation (D620N) in VPS35, this mutation does not significantly impair the direct association with FAM21 in vitro.
... Vps26B is a subunit of the retromer complex acting at the budding sites of endosomes, where endosomal tubules grow and fission 40,42,44,[50][51][52][53][54] . Our findings demonstrated that Vps26B interacted with SHIP164, the depletion of which affected Rab14-positive endosomes, suggesting a (13 cells) of Vps26B-RFPnb. ...
Article
Full-text available
Early endosomes (EEs) are crucial in cargo sorting within vesicular trafficking. While cargoes destined for degradation are retained in EEs and eventually transported to lysosomes, recycled cargoes for the plasma membrane (PM) or the Golgi undergo segregation into specialized membrane structures known as EE buds during cargo sorting. Despite this significance, the molecular basis of the membrane expansion during EE bud formation has been poorly understood. In this study, we identify a protein complex comprising SHIP164, an ATPase RhoBTB3, and a retromer subunit Vps26B, which promotes the formation of EE buds at Golgi–EE contacts. Our findings reveal that Vps26B acts as a novel Rab14 effector, and Rab14 activity regulates the association of SHIP164 with EEs. Depletion of SHIP164 leads to enlarged Rab14 ⁺ EEs without buds, a phenotype rescued by wild-type SHIP164 but not the lipid transfer-defective mutants. Suppression of RhoBTB3 or Vps26B mirrors the effects of SHIP164 depletion. Together, we propose a lipid transport-dependent pathway mediated by the RhoBTB3–SHIP164–Vps26B complex at Golgi–EE contacts, which is essential for EE budding.
... Since it is the backbone of Retromer's core complex, a disease-associated mutation in VPS35 is expected to affect Retromer-dependent trafficking. While this might seem obvious, the PD-associated VPS35 mutation, VPS35[D620N] (a mutation that results in an aspartate to an asparagine substitution at residue 620) turns out to have a relatively subtle molecular consequence by specifically impairing its interactions with FAM21 [14], a key member of the Retromer-related WASH complex [15]. More importantly, studies have shown that the PD-associated VPS35 mutation differentially disrupts Retromer's retrograde pathway, compared to Retromer's recycling pathway, ultimately leading to relatively subtle and specific defects in lysosomal function [16,17]. ...
Article
Full-text available
While causative mutations in complex disorders are rare, they can be used to extract a biological pathway whose pathogenicity can generalize to common forms of the disease. Here we begin by relying on the biological consequences of mutations in LRRK2 and VPS35, genetic causes of autosomal-dominant Parkinson's disease, to hypothesize that ‘Retromer-dependent lysosomal stress’ represents a pathway that can generalize to idiopathic Parkinson's disease. Next, we outline a series of studies that can test this hypothesis, including the development of biomarkers of pathway dysfunction. If validated, the hypothesis can suggest a unified mechanism of disease and might inform future diagnostic and therapeutic investigations. This article is part of a discussion meeting issue ‘Understanding the endo-lysosomal network in neurodegeneration’.
... Recruitment of VPS35, a core retromer component, to membranes does not appear impeded by ORF3a expression (Fig EV5B), indicating that tubule formation or scission may be impaired. Endosomal acidification has been proposed to regulate tubule formation, as a homeostatic mechanism to ensure tubulation does not occur too early in the endocytic pathway or from incorrect organelles (Harbour et al, 2012), and so ORF3a-impaired endolysosomal acidification (Ghosh et al, 2020) may prevent proper retrograde tubules from forming on endosomes. ...
Article
Full-text available
The antiviral restriction factor, tetherin, blocks the release of several different families of enveloped viruses, including the Corona-viridae. Tetherin is an interferon-induced protein that forms parallel homodimers between the host cell and viral particles, linking viruses to the surface of infected cells and inhibiting their release. We demonstrate that SARS-CoV-2 infection causes tetherin downregulation and that tetherin depletion from cells enhances SARS-CoV-2 viral titres. We investigate the potential viral proteins involved in abrogating tetherin function and find that SARS-CoV-2 ORF3a reduces tetherin localisation within biosynthetic organelles where Coronaviruses bud, and increases tetherin localisation to late endocytic organelles via reduced retrograde recycling. We also find that expression of Spike protein causes a reduction in cellular tetherin levels. Our results confirm that tetherin acts as a host restriction factor for SARS-CoV-2 and highlight the multiple distinct mechanisms by which SARS-CoV-2 subverts tetherin function.
Article
Full-text available
The identification of multiple genes linked to Parkinson’s Disease invites the question as to how they may cooperate. We have generated isogenic cell lines that inducibly express either wild-type or a mutant form of the retromer component VPS35 (D620N), which has been linked to Parkinson’s Disease. This has enabled us to test proposed effects of this mutation in a setting where the relative expression reflects the physiological occurrence. We confirm that this mutation compromises VPS35 association with the WASH complex, but find no defect in WASH recruitment to endosomes, nor in the distribution of lysosomal receptors, cation-independent mannose-6-phosphate receptor and Sortilin. We show VPS35 (D620N) enhances the activity of the Parkinson’s associated kinase LRRK2 towards RAB12 under basal conditions. Furthermore, VPS35 (D620N) amplifies the LRRK2 response to endolysosomal stress resulting in enhanced phosphorylation of RABs 10 and 12. By comparing different types of endolysosomal stresses such as the ionophore nigericin and the membranolytic agent LLOMe, we are able to dissociate phospho-RAB accumulation from membrane rupture.
Preprint
Full-text available
Endosomal trafficking ensures the proper distribution of lipids and proteins to various cellular compartments, facilitating intracellular communication, nutrient transport, waste disposal, and the maintenance of cell structure. Retromer, a peripheral membrane protein complex, plays an important role in this process by recruiting the associated actin-polymerizing WASH complex to establish distinct sorting domains. The WASH complex is recruited through the interaction of the VPS35 subunit of retromer with the WASH complex subunit FAM21. Here, we report the identification of two separate fragments of Fam21 that interact with VPS35, along with a third fragment that binds to the VPS29 subunit of retromer. The crystal structure of VPS29 bound to a peptide derived from Fam21 shows a distinctive sharp bend that inserts into a conserved hydrophobic pocket with a binding mode similar to that adopted by other VPS29 effectors. Interestingly, despite the network of interactions between Fam21 and retromer occurring near the Parkinson disease-linked mutation (D620N) in VPS35, this mutation does not significantly impair the direct association with Fam21 in vitro.
Article
Transport of macromolecules from the nucleus to the cytoplasm is essential for nearly all cellular and developmental events, and when mis-regulated, is associated with diseases, tumor formation/growth, and cancer progression. Nuclear Envelope (NE)-budding is a newly appreciated nuclear export pathway for large macromolecular machineries, including those assembled to allow co-regulation of functionally related components, that bypasses canonical nuclear export through nuclear pores. In this pathway, large macromolecular complexes are enveloped by the inner nuclear membrane, transverse the perinuclear space, and then exit through the outer nuclear membrane to release its contents into the cytoplasm. NE-budding is a conserved process and shares many features with nuclear egress mechanisms used by herpesviruses. Despite its biological importance and clinical relevance, little is yet known about the regulatory and structural machineries that allow NE-budding to occur in any system. Here we summarize what is currently known or proposed for this intriguing nuclear export process.
Article
Full-text available
Intracellular protein trafficking and sorting are extremely arduous in endocrine and neuroendocrine cells, which synthesize and secrete on-demand substantial quantities of proteins. To ensure that neuroendocrine secretion operates correctly, each step in the secretion pathways is tightly regulated and coordinated both spatially and temporally. At the trans-Golgi network (TGN), intrinsic structural features of proteins and several sorting mechanisms and distinct signals direct newly synthesized proteins into proper membrane vesicles that enter either constitutive or regulated secretion pathways. Furthermore, this anterograde transport is counterbalanced by retrograde transport, which not only maintains membrane homeostasis but also recycles various proteins that function in the sorting of secretory cargo, formation of transport intermediates, or retrieval of resident proteins of secretory organelles. The retromer complex recycles proteins from the endocytic pathway back to the plasma membrane or TGN and was recently identified as a critical player in regulated secretion in the hypothalamus. Furthermore, melanoma antigen protein L2 (MAGEL2) was discovered to act as a tissue-specific regulator of the retromer-dependent endosomal protein recycling pathway and, by doing so, ensures proper secretory granule formation and maturation. MAGEL2 is a mammalian-specific and maternally imprinted gene implicated in Prader-Willi and Schaaf-Yang neurodevelopmental syndromes. In this review, we will briefly discuss the current understanding of the regulated secretion pathway, encompassing anterograde and retrograde traffic. Although our understanding of the retrograde trafficking and sorting in regulated secretion is not yet complete, we will review recent insights into the molecular role of MAGEL2 in hypothalamic neuroendocrine secretion and how its dysregulation contributes to the symptoms of Prader-Willi and Schaaf-Yang patients. Given that the activation of many secreted proteins occurs after they enter secretory granules, modulation of the sorting efficiency in a tissue-specific manner may represent an evolutionary adaptation to environmental cues.
Article
Full-text available
Molecular Psychiatry publishes work aimed at elucidating biological mechanisms underlying psychiatric disorders and their treatment
Article
Full-text available
Wnt proteins are lipid-modified glycoproteins that play a central role in development, adult tissue homeostasis and disease. Secretion of Wnt proteins is mediated by the Wnt-binding protein Wntless (Wls), which transports Wnt from the Golgi network to the cell surface for release. It has recently been shown that recycling of Wls through a retromer-dependent endosome-to-Golgi trafficking pathway is required for efficient Wnt secretion, but the mechanism of this retrograde transport pathway is poorly understood. Here, we report that Wls recycling is mediated through a retromer pathway that is independent of the retromer sorting nexins SNX1-SNX2 and SNX5-SNX6. We have found that the unrelated sorting nexin, SNX3, has an evolutionarily conserved function in Wls recycling and Wnt secretion and show that SNX3 interacts directly with the cargo-selective subcomplex of the retromer to sort Wls into a morphologically distinct retrieval pathway. These results demonstrate that SNX3 is part of an alternative retromer pathway that functionally separates the retrograde transport of Wls from other retromer cargo.
Article
Full-text available
WASP and SCAR homologue (WASH) is a recently identified and evolutionarily conserved regulator of actin polymerization. In this paper, we show that WASH coats mature Dictyostelium discoideum lysosomes and is essential for exocytosis of indigestible material. A related process, the expulsion of the lethal endosomal pathogen Cryptococcus neoformans from mammalian macrophages, also uses WASH-coated vesicles, and cells expressing dominant negative WASH mutants inefficiently expel C. neoformans. D. discoideum WASH causes filamentous actin (F-actin) patches to form on lysosomes, leading to the removal of vacuolar adenosine triphosphatase (V-ATPase) and the neutralization of lysosomes to form postlysosomes. Without WASH, no patches or coats are formed, neutral postlysosomes are not seen, and indigestible material such as dextran is not exocytosed. Similar results occur when actin polymerization is blocked with latrunculin. V-ATPases are known to bind avidly to F-actin. Our data imply a new mechanism, actin-mediated sorting, in which WASH and the Arp2/3 complex polymerize actin on vesicles to drive the separation and recycling of proteins such as the V-ATPase.
Article
Full-text available
Endocytic sorting of signalling receptors between recycling and degradative pathways is a key cellular process controlling the surface complement of receptors and, accordingly, the cell's ability to respond to specific extracellular stimuli. The β2 adrenergic receptor (β2AR) is a prototypical seven-transmembrane signalling receptor that recycles rapidly and efficiently to the plasma membrane after ligand-induced endocytosis. β2AR recycling is dependent on the receptor's carboxy-terminal PDZ ligand and Rab4. This active sorting process is required for functional resensitization of β2AR-mediated signalling. Here we show that sequence-directed sorting occurs at the level of entry into retromer tubules and that retromer tubules are associated with Rab4. Furthermore, we show that sorting nexin 27 (SNX27) serves as an essential adaptor protein linking β2ARs to the retromer tubule. SNX27 does not seem to directly interact with the retromer core complex, but does interact with the retromer-associated Wiskott-Aldrich syndrome protein and SCAR homologue (WASH) complex. The present results identify a role for retromer in endocytic trafficking of signalling receptors, in regulating a receptor-linked signalling pathway, and in mediating direct endosome-to-plasma membrane traffic.
Article
Full-text available
Voluntary movement is a fundamental way in which animals respond to, and interact with, their environment. In mammals, the main CNS pathway controlling voluntary movement is the corticospinal tract, which encompasses connections between the cerebral motor cortex and the spinal cord. Hereditary spastic paraplegias (HSPs) are a group of genetic disorders that lead to a length-dependent, distal axonopathy of fibres of the corticospinal tract, causing lower limb spasticity and weakness. Recent work aimed at elucidating the molecular cell biology underlying the HSPs has revealed the importance of basic cellular processes — especially membrane trafficking and organelle morphogenesis and distribution— in axonal maintenance and degeneration.
Article
Full-text available
The retromer complex is required for the efficient endosome-to-Golgi retrieval of the CIMPR, sortilin, SORL1, wntless and other physiologically important membrane proteins. Retromer comprises two protein complexes that act together in endosome-to-Golgi retrieval; the cargo-selective complex is a trimer of VPS35, VPS29 and VPS26 that sorts cargo into tubules for retrieval to the Golgi. Tubules are produced by the oligomerization of sorting nexin dimers. Here, we report the identification of five endosomally-localised proteins that modulate tubule formation and are recruited to the membrane via interactions with the cargo-selective retromer complex. One of the retromer-interacting proteins, strumpellin, is mutated in hereditary spastic paraplegia, a progressive length-dependent axonopathy. Here, we show that strumpellin regulates endosomal tubules as part of a protein complex with three other proteins that include WASH1, an actin-nucleating promoting factor. Therefore, in addition to a direct role in endosome-to-Golgi retrieval, the cargo-selective retromer complex also acts as a platform for recruiting physiologically important proteins to endosomal membranes that regulate membrane tubule dynamics.
Article
We have previously identified a new gene with sequence homology to the WASP-family of actin regulators denoted WAFL (WASP and FKBP-like). Here we report a possible biological function for WAFL, by demonstrating an association to early endosomes via its central coiled-coil domain. Further we show by functional and structural studies that WAFL is associated with both microtubules and the actin filament system, the two means of transport of early endosomes. In addition, WAFL interacts with WASP-interacting protein (WIP) and actin, thus linking WAFL to actin dynamics. The use of RNAi depletion of WAFL shows that WAFL-deficient cells display delayed transport of endosomal cargo. Our findings are compatible with a model whereby WAFL is involved in the transport of early endosomes at the level of transition between microfilament-based and microtubule-based movement.
Article
The retromer complex is required for the sorting of acid hydrolases to lysosomes, transcytosis of the polymeric immunoglobulin receptor, Wnt gradient formation, iron transporter recycling and processing of the amyloid precursor protein. Human retromer consists of two smaller complexes: the cargo recognition VPS26-VPS29-VPS35 heterotrimer and a membrane-targeting heterodimer or homodimer of SNX1 and/or SNX2 (ref. 13). Here we report the crystal structure of a VPS29-VPS35 subcomplex showing how the metallophosphoesterase-fold subunit VPS29 (refs 14, 15) acts as a scaffold for the carboxy-terminal half of VPS35. VPS35 forms a horseshoe-shaped, right-handed, alpha-helical solenoid, the concave face of which completely covers the metal-binding site of VPS29, whereas the convex face exposes a series of hydrophobic interhelical grooves. Electron microscopy shows that the intact VPS26-VPS29-VPS35 complex is a stick-shaped, flexible structure, approximately 21nm long. A hybrid structural model derived from crystal structures, electron microscopy, interaction studies and bioinformatics shows that the alpha-solenoid fold extends the full length of VPS35, and that VPS26 is bound at the opposite end from VPS29. This extended structure presents multiple binding sites for the SNX complex and receptor cargo, and appears capable of flexing to conform to curved vesicular membranes.
Article
The ability of cells to adhere, spread and migrate is essential to many physiological processes, particularly in the immune system where cells must traffic to sites of inflammation and injury. By altering the levels of individual components of the VAMP3/Stx4/SNAP23 complex we show here that this SNARE complex regulates efficient macrophage adhesion, spreading and migration on fibronectin. During cell spreading this complex mediates the polarised exocytosis of VAMP3-positive recycling endosome membrane into areas of membrane expansion, where VAMP3's surface partner Q-SNARE complex Stx4/SNAP23 was found to accumulate. Lowering the levels of VAMP3 in spreading cells resulted in a more rounded cell morphology and most cells were found to be devoid of the typical ring-like podosome superstructures seen normally in spreading cells. In migrating cells lowering VAMP3 levels disrupted the polarised localisation of podosome clusters. The reduced trafficking of recycling endosome membrane to sites of cell spreading and the disorganised podosome localisation in migrating macrophages greatly reduced their ability to persistently migrate on fibronectin. Thus, this important SNARE complex facilitates macrophage adhesion, spreading, and persistent macrophage migration on fibronectin through the delivery of VAMP3-positive membrane with its cargo to expand the plasma membrane and to participate in organising adhesive podosome structures.
Article
Bidirectional traffic between the Golgi apparatus and the endosomal system sustains the functions of the trans-Golgi network (TGN) in secretion and organelle biogenesis. Export of cargo from the TGN via anterograde trafficking pathways depletes the organelle of sorting receptors, processing proteases, SNARE molecules, and other factors, and these are subsequently retrieved from endosomes via the retrograde pathway. Recent studies indicate that retrograde trafficking is vital to early metazoan development, nutrient homeostasis, and for processes that protect against Alzheimer's and other neurological diseases.