You are on page 1of 10

European Journal of Cell Biology 97 (2018) 523–532

Contents lists available at ScienceDirect

European Journal of Cell Biology


journal homepage: www.elsevier.com/locate/ejcb

Research paper

ATG16 mediates the autophagic degradation of the 19S proteasomal T


subunits PSMD1 and PSMD2
Qiuhong Xionga,b, Sarah Fischera, Malte Karowa, Rolf Müllera, Susanne Meßlinga,
Ludwig Eichingera,

a
Center for Biochemistry, Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 52, 50931 Cologne, Germany.
b
Institute of Biomedical Sciences, Shanxi University, No. 92 Wucheng Road, 030006 Taiyuan, Shanxi, China

ARTICLE INFO ABSTRACT

Keywords: Autophagy and the ubiquitin proteasome system are the two major cellular processes for protein and organelle
Autophagy recycling and clearance in eukaryotic cells. Evidence is accumulating that these two pathways are interrelated
Proteasome through adaptor proteins. Here, we found that PSMD1 and PSMD2, both components of the 19S regulatory
Dictyostelium particle of the proteasome, directly interact with Dictyostelium discoideum autophagy 16 (ATG16), a core au-
UPS
tophagosomal protein. ATG16 is composed of an N-terminal domain, which is responsible for homo-dimerization
ATG16
PSMD1
and binding to ATG5 and a C-terminal β-propeller structure. Deletion analysis of ATG16 showed that the N-
PSMD2 terminal half of ATG16 interacted directly only with PSMD1, while the C-terminal half interacted with both,
PSMD1 and PSMD2. RFP-tagged PSMD1 as well as PSMD2 were enriched in large puncta, reminiscent of au-
tophagosomes, in wild-type cells. These puncta were absent in atg16‾ and atg9‾/16‾ cells and weaker and less
frequent in atg9‾ cells, showing that ATG16 was crucial and the autophagic process important for their for-
mation. Co-expression of ATG16-GFP or GFP-ATG8a(LC3) with RFP-PSMD1 or RFP-PSMD2, respectively, in
atg16‾ or wild-type cells revealed many instances of co-localization, suggesting that RFP-PSMD1 or RFP-PSMD2
positive puncta constitute autophagosomes. LysoTracker® labeling and a proteolytic cleavage assay confirmed
that PSMD1 and PSMD2 were present in lysosomes in wild-type cells. In vivo, ATG16 is required for their en-
richment in ATG8a positive puncta, which mature into autolysosomes. We propose that ATG16 links autophagy
and the ubiquitin proteasome system.

1. Introduction proteasome and, vice versa, proteasomal subunits were found to be


degraded by lysosomes (Cuervo et al., 1995; Gao et al., 2010). More
Cell homeostasis is maintained by a precisely regulated balance recently proteaphagy, i.e. degradation of whole proteasomes by au-
between synthesis and degradation of cellular components. Autophagy tophagy, was reported in HeLa cells, the mouse-ear cress Arabidopsis
and the ubiquitin proteasome system (UPS) are the two major routes for thaliana and in yeast (Cohen-Kaplan et al., 2016; Marshall et al., 2015,
protein and organelle clearance in eukaryotic cells. In general, short- 2016). The autophagy adaptor protein p62/SQSTM1 delivers the ubi-
lived, abnormal or damaged proteins are degraded by the UPS, while quitinated proteasome to the autophagy machinery through binding to
most of the long-lived proteins, protein aggregates as well as cellular LC3 in mammals (Cohen-Kaplan et al., 2016). In A. thaliana the 19S
organelles (mitochondria, peroxisomes, ribosomes, and infectious or- proteasomal subunit RPN10 mediates selective autophagic degradation
ganisms) are cleared by autophagy (Kim et al., 2011; Schreiber and of the 26S proteasome (Marshall et al., 2015) and in yeast the autop-
Peter, 2014). There is increasing evidence that the two degradative hagic turnover of inactive 26S proteasomes is directed by the ubiquitin
pathways, which are of utmost importance for the recycling of cellular receptor Cue5 and Hsp42 (Marshall et al., 2016).
material, are interconnected (Driscoll and Chowdhury, 2012; The core autophagy protein ATG16 is a component of the tetrameric
Korolchuk et al., 2009, 2010). For example, α-synuclein and other ag- ATG12-ATG5/ATG16 complex which consists of two ATG12-ATG5
gregation-prone proteins are substrates of both pathways (Ravikumar conjugates bound to an ATG16 homodimer and is essential for autop-
et al., 2002; Webb et al., 2003). Furthermore, the autophagosomal hagosome formation (Kuma et al., 2002; Mizushima et al., 2003, 1999;
marker protein ATG8 (LC3 in mammals) can be degraded by the 20S Noda and Inagaki, 2015). In the early phase of autophagosome


Corresponding author.
E-mail address: ludwig.eichinger@uni-koeln.de (L. Eichinger).

https://doi.org/10.1016/j.ejcb.2018.09.002
Received 27 February 2018; Received in revised form 6 August 2018; Accepted 10 September 2018
0171-9335/ © 2018 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
Q. Xiong et al. European Journal of Cell Biology 97 (2018) 523–532

generation the ATG12-ATG5/ATG16 complex mainly resides on the


outer side of the phagophore in yeast or the isolation membrane in
higher eukaryotes and catalyzes the covalent linkage of ATG8/LC3 to
the phospholipid phosphatidylethanolamine (PE). In this complex, the
ATG12-ATG5 conjugate holds the catalytic activity while ATG16 is
required for the correct localization of the complex to the isolation
membrane (Fujita et al., 2008; Hanada et al., 2007; Suzuki et al., 2001).
Dictyostelium ATG16 is highly similar to the mammalian orthologue
ATG16L1 and is composed of 3 distinct regions: the N-terminal domain
which is responsible for binding to ATG5, followed by a coiled coil
domain (CCD) which mediates homo-dimerization and seven WD40
repeats in the C-terminal half of the protein, which are predicted to
form a β-propeller structure and are lacking in the yeast protein. The β-
propeller structure is found in a wide range of proteins involved in
distinct biological activities and generally acts as a protein-protein in-
teraction platform (Neer et al., 1994; Smith et al., 1999). In mammalian
ATG16L1 the β-propeller has been identified as an interface for the
transmembrane protein TMEM59 and Nod1 and Nod2, members of the
Nod-like receptor family, which mediate the autophagic response to
bacterial pathogens by recruiting ATG16L1 to the bacterial entry site
(Boada-Romero et al., 2013; Travassos et al., 2010). Furthermore, a
large subset of WD40 repeats was defined as a new class of ubiquitin-
binding domains that interact with ubiquitin in a similar fashion
(Pashkova et al., 2010). Along this line it was found that ubiquitin is
recognized by the autophagic machinery during infection of HeLa cells
with Salmonella typhimurium through adaptor proteins, including a di-
rect interaction between ubiquitin and Atg16L1 (Fujita et al., 2013).
ATG16 also appears to play a role in the link between autophagy
and the UPS. It is indirectly involved in Cullin-3-mediated ubiquitina-
tion of p62 followed by proteasomal degradation because in its absence
the E3 ubiquitin ligase Cullin-3 could not be neddylated which resulted
in the accumulation of p62 (Lee et al., 2012). In addition, the half-life
and cellular concentration of ATG16 itself is possibly regulated by the
UPS, since treatment of MEFs with proteasome inhibitors led to an in-
crease of ATG16 (Fujita et al., 2009). On the other hand, ATG16 is also
crucial for UPS function because the Dictyostelium atg16 knock-out
mutant displayed strongly reduced proteasomal activity (Xiong et al.,
2015).
The social amoeba D. discoideum is a well-established model or-
ganism for the investigation of the autophagic process (Mesquita et al.,
2017). In this organism novel conserved autophagy genes have been
discovered and the analysis of single or double knock-outs of core au-
tophagy genes revealed informative phenotypes (Calvo-Garrido and
Escalante, 2010; Calvo-Garrido et al., 2014; Messling et al., 2017;
Munoz-Braceras et al., 2015; Otto et al., 2004; Tung et al., 2010; Xiong
et al., 2015). Here, we identified PSMD1 and PSMD2, both components
of the 19S regulatory particle of the proteasome, as ATG16-interacting
proteins. We show that PSMD1 and PSMD2 are degraded in lysosomes
and that ATG16 has an essential function in this process. Our results
reveal a novel role for ATG16 in the crosstalk between autophagy and
the UPS.

2. Results
(caption on next page)
2.1. PSMD1 and PSMD2 interact with ATG16

To identify novel ATG16 binding proteins we incubated cell lysates


from Dictyostelium AX2 wild-type cells with recombinant GST-ATG16
followed by GST pull-down. Mass spectrometric analysis revealed a
number of proteins that specifically interacted with GST-ATG16, among
them were PSMD1 with 108 kDa and PSMD2 with 90 kDa (Fig. 1A).
PSMD1 and PSMD2 are two subunits of the regulatory 19S particle of
the proteasome. Pairwise alignments of the protein sequences of PSMD1
and PSMD2 with the corresponding orthologues of human and yeast
showed significantly higher sequence identity of Dictyostelium PSMD1
and PSMD2 with the human orthologues (Table 1).

524
Q. Xiong et al. European Journal of Cell Biology 97 (2018) 523–532

Fig. 1. Identification and verification of PSMD1 and PSMD2 as new binding GST-PSMD2, while ATG161−309-GFP was only pulled down by GST-
partners of ATG16. (A) GST-ATG16 pulled down PSMD1 and PSMD2. PSMD1 (Fig. S1). Taken together, these results show that the C-terminal
Recombinant GST-ATG16 was incubated with total cell lysates from AX2 cells half of ATG16, which harbors the seven WD repeats, can bind directly
followed by GST pull-down assays. Precipitated proteins were separated by to both proteasomal subunits, PSMD1 and PSMD2, while the N-terminal
SDS-PAGE and stained with Coomassie Brilliant Blue (CBB). Specific protein
half of ATG16 only interacts directly with PSMD1.
bands were cut out and the identity of the proteins determined by mass spec-
trometry. The positions of PSMD1, PSMD2 and GST-ATG16 are marked by
black arrowheads. (B) GST-PSMD1 and GST-PSMD2 pulled down ATG16-GFP. 2.3. PSMD1 and PSMD2 puncta co-localize with ATG16 and ATG8a
Recombinant GST-PSMD1 or GST-PSMD2 was incubated with total cell lysates
from atg16‾/atg16-gfp cells followed by GST pull-down assays. ATG16-GFP was To investigate possible co-localizations of PSMD1 or PSMD2 with
detected with a GFP specific antibody. Upper part, Western Blot; lower part ATG16 and ATG8a (LC3) in vivo, we expressed either RFP-PSMD1 or
PonceauS stain. (C) ATG16 interacted with PSMD1 and PSMD2 in vivo. Total RFP-PSMD2, respectively, in atg16‾/atg16-gfp and AX2/gfp-atg8a cells
cell lysates of atg16‾/atg16-gfp cells expressing RFP, RFP-PSMD1 or RFP- (Matthias et al., 2016; Xiong et al., 2015). Immunofluorescence analysis
PSMD2 were incubated with GFP-Trap®_A beads. Precipitated proteins were was performed after treatment of the cells with NH4Cl for 4 h to slow
separated by SDS-PAGE, transferred by Western blotting and ATG16-GFP, RFP- down the autophagic flux. We found that RFP-PSMD1 and RFP-PSMD2
PSMD1 and RFP-PSMD2 were detected by GFP or RFP specific antibodies. (*):
were enriched in punctate structures in atg16‾/atg16-gfp cells and
Degradation product of RFP-PSMD1.
partially co-localized with ATG16-GFP (Fig. 3A, B). RFP-PSMD1 and
RFP-PSMD2 positive punctate structures were only observed in
Table 1 NH4Cl–treated but not in untreated AX2 cells. Since ATG16 belongs to
Protein sequence identities of human, yeast and Dictyostelium PSMD1 and 2. the core autophagy genes and associates with the isolation membrane
in the early phase of autophagosome biogenesis, we considered the
Dictyostelium Yeast
possibility that the PSMD1 and PSMD2 positive puncta might be au-
PSMD1 Human 46 41 tophagosomes. To address this question, we analyzed the co-localiza-
Dictyostelium 38 tion of the autophagosomal marker GFP-ATG8a with RFP-PSMD1 and
PSMD2 Human 51 42 RFP-PSMD2 in AX2 wild-type cells in vivo. Immunofluorescence ana-
Dictyostelium 35
lysis revealed many instances of partial co-localization of GFP-ATG8a
Sequence identity was determined by aligning the corresponding protein se-
with RFP-PSMD1 or RFP-PSMD2. In addition, we found that the
quences using the alignment program (bl2seq) at the NCBI (http://www.ncbi. punctate structures were frequently encircled to various degrees by
nlm.nih.gov/blast/Blast.cgi). Percentage values of sequence identities are GFP-ATG8a (Fig. 3C, D). Furthermore, the GFP-ATG8a signal changed
given. position in tact with the RFP-PSMD1 puncta suggesting that the co-
localization is real (supplementary movie 1). Quantitative analysis
We verified the interaction between ATG16 and the two protea- showed that 40.5 and 31.4% of RFP-PSMD1 or RFP-PSMD2 positive
somal subunits by utilizing recombinant GST-PSMD1 and GST-PSMD2 puncta co-localized with ATG16-GFP, respectively. The corresponding
in pull-down assays with cell lysates from atg16‾/atg16-gfp cells. In values for the co-localization with GFP-ATG8a were 12.1 and 23.9%,
these assays both proteins, GST-PSMD1 and GST-PSMD2, could pull respectively (Table 2). Taken together, these results show that a sub-
down ATG16-GFP (Fig. 1B). We next ectopically expressed either RFP- fraction of RFP-PSMD1 and RFP-PSMD2 co-localises with autophago-
PSMD1 or RFP-PSMD2 in atg16‾/atg16-gfp cells, respectively. Im- somes.
munoprecipitation (IP) experiments with GFP-Trap®A beads showed
that RFP-PSMD1 as well as RFP-PSMD2 could be immune-precipitated 2.4. The autophagosomal localization of PSMD1 and PSMD2 is dependent
by ATG16-GFP (Fig. 1C). These results suggest that PSMD1 and PSMD2 on ATG16
are interacting with ATG16 in vivo.
Next we expressed RFP-PSMD1 also in AX2, atg16‾, atg9‾, atg9‾/
16‾ and atg9‾/16‾/atg16-gfp cells and RFP-PSMD2 also in AX2 and
2.2. The interaction of ATG16 with PSMD1 and PSMD2 is direct atg16‾ cells (Figure S2 A). Immunofluorescence analysis showed that
tagged PSMD1 and PSMD2 were enriched in punctate structures upon
We next wanted to determine, if ATG16 directly interacts with treatment with NH4Cl in AX2 wild-type cells but not in atg16‾ cells
PSMD1 and PSMD2 and, if this is the case, which region of ATG16 is (Fig. 4A). Similar results were obtained after treatment of the cells with
responsible for the interaction. For this purpose we generated and Bafilomycin A1, which inhibits lysosomal acidification (Fig. S2B).
purified a series of GST tagged ATG16 fragments. These were, based on Quantitative analysis revealed that 98% of AX2 cells contained RFP-
the ATG16 domain structure, ATG161−309, the N-terminal half, PSMD1 and 97% RFP-PSMD2 positive punctate structures, while such
ATG16310–612, the C-terminal half with the seven WD-repeats, RFP-PSMD1 or RFP-PSMD2 positive punctae were only seen in less than
ATG161–198, which contains the ATG5-interacting motif (AFIM) and the 1% of atg16‾ cells (Fig. 4B). After expression of ATG16-GFP in ATG16‾
coiled coil domain (CCD) and ATG16160–312, which contains the CCD cells we observed the re-appearance of RFP-PSMD1 and RFP-PSMD2
and the region between the CCD and the WD-repeats (Fig. 2A). In pull- positive puncta in 86 and 94% of the cells, respectively (Fig. 4A, B).
down assays we found that the N- and C-terminal halves of ATG16, Next, we performed immunofluorescence analysis of atg9‾ and atg9‾/
ATG161−309 and ATG16310–612, respectively, could both pull down 16‾ cells expressing RFP-PSMD1 and found that RFP-PSMD1 positive
PSMD1 but only ATG16310-612 was able to also pull down PSMD2 puncta were only formed in 23% of the atg9‾ cells and were absent in
(Fig. 2B). Further truncation analysis of the interaction of the N-term- atg9‾/16‾ cells. Expression of ATG16-GFP in atg9‾/16‾ cells partially
inal half of ATG16 with PSMD1 revealed that ATG161-198 but not rescued puncta formation to similar levels as in atg9‾ cells. However,
ATG16160-312 was able to pull down PSMD1 (Fig. 2C). We next gener- the punctate structures were significantly less prominent in atg9‾ and
ated constructs for the expression of ATG161−309-GFP, ATG16310-612- atg9‾/16‾/atg16-gfp cells in comparison to AX2 (Fig. S2C, D). In pre-
GFP and ATG16156–612-GFP in Dictyostelium atg16‾ cells (Fig. 2A). Un- vious studies it was shown that ATG16 binds to the isolation membrane
fortunately, we were not able to obtain an ATG16310-612-GFP expressing in an ATG5-dependent manner and more recently an AFIM was iden-
atg16‾ strain, therefore, we could only analyze the interaction of tified in ATG16L1 (Kim et al., 2015; Romanov et al., 2012). Sequence
ATG161−309-GFP and ATG16156–612-GFP with PSMD1 and PSMD2, re- alignments with ATG16 protein sequences showed that the AFIM is
spectively. Consistent with the in vitro interaction studies we found that nicely conserved in other ATG16 proteins as for example yeast, Danio
ATG16156–612-GFP was pulled down by both proteins, GST-PSMD1 and rerio, Mus musculus, Homo sapiens and Dictyostelium (Fig. 4C). To test

525
Q. Xiong et al. European Journal of Cell Biology 97 (2018) 523–532

Fig. 2. The interaction of ATG16 with PSMD1


and PSMD2 is direct. (A) Domain structure of
ATG16 and schematic representation of in-
vestigated ATG16 fragments. Amino acid po-
sitions are given above full length ATG16 and
each ATG16 fragment. The N-terminal domain
is depicted in green and the C-terminal β-pro-
peller domain in blue. CC, coiled coil domain;
WD1-7 depicts the seven WD repeats. (B) The N-
terminal half of ATG16 interacted directly with
PSMD1 and the C-terminal half with PSMD1
and PSMD2. GST-tagged ATG161−309 and
ATG16310-612 were purified from E. coli, bound
to Glutathione Sepharose 4B beads and in-
cubated with purified recombinant PSMD1 or
PSMD2. Proteins were separated by 12% SDS-
PAGE, transferred to nitrocellulose membrane
and stained with PonceauS. PSMD1 and
PSMD2 were detected by specific polyclonal
antibodies. Upper part, Western Blot (WB) with
anti PSMD1 (left) and anti PSMD2 (right) an-
tibodies; (*), unspecific binding of the PSMD2
antibody; lower part, WB with anti GST anti-
body. (C) ATG161-198 but not ATG16160-312
interacted directly with PSMD1. Experimental
procedure was as described for (B) (For inter-
pretation of the references to colour in this
figure legend, the reader is referred to the web
version of this article.).

whether the N-terminal region of ATG16 containing the AFIM is crucial presumably takes place in autolysosomes. To investigate the putative
for mediating the formation of RFP-PSMD1 and RFP-PSMD2 punctate localization of PSMD1 and PSMD2 in autolysosomes we expressed RFP-
structures we expressed ATG161−309-GFP or ATG16156-612-GFP, lacking GFP-PSMD1 or RFP-GFP-PSMD2 in AX2 cells (Fig. S3). The fluorescence
the AFIM, with RFP-PSMD1 or RFP-PSMD2, respectively in atg16‾ cells. of GFP is very sensitive to the acidic environment of the lysosome, thus,
Immunofluorescence analysis showed that in atg16‾ cells expressing the presence of red puncta lacking the green fluorescence is an in-
ATG161−309-GFP, but not in those expressing ATG16156-612-GFP, RFP- dication of the acidification of the autophagosome through fusion with
PSMD1 and RFP-PSMD2 were again enriched in punctate structures the lysosome (Calvo-Garrido et al., 2011). We found that treatment
(Fig. 4D, E). These results show that the enrichment of PSMD1 and with NH4Cl or Bafilomycin A1, which both slow down autophagosomal
PSMD2 in punctate structures, which are presumably autophagosomes, degradation, resulted in the appearance of red puncta in these cells. The
is dependent on the N-terminal 309 amino acids of ATG16, which disappearance of the green fluorescence indicates the acidification of
contain the AFIM. The results are consistent with the assumption that PSMD1 and PSMD2 positive puncta through fusion with the lysosome
ATG16 lacking the AFIM cannot be recruited to the autophagosome. We (Fig. 5B). Finally, to determine whether PSMD1 and PSMD2 are indeed
conclude that the N-terminal 155 amino acids of ATG16, which contain present in lysosomes, we stained live AX2 and atg16‾ cells expressing
the AFIM motif, are required and the C-terminal half of ATG16, which RFP-PSMD1 or RFP-PSMD2 with LysoTracker. In AX2 cells we found
harbors the seven WD repeats, is dispensable for the formation of that many of the PSMD1 and PSMD2 positive puncta co-localized with
PSMD1 and PSMD2 puncta in atg16‾ cells. LysoTracker, while no such puncta were formed and hence no co-lo-
calization was seen in atg16‾ cells (Fig. 5C). Quantification of the co-
localization showed that approximately 50% of the RFP-PSMD1 or RFP-
2.5. PSMD1 and PSMD2 are degraded by autophagy PSMD2 positive puncta were also positive for LysoTracker. Interest-
ingly, the ratio of co-localization increased to nearly 90% for puncta
Our results so far suggest that a subfraction of PSMD1 and PSMD2 with a diameter larger than 1 μm (Table 3). We conclude that PSMD1
positive puncta are autophagosomes, because these structures partially and PSMD2 are degraded by ATG16-mediated autophagy.
co-localized with ATG8a, could only be detected when autophagosomal
degradation was slowed down and their formation was dependent on
ATG16. Autophagic degradation of substrates can be monitored with 3. Discussion
the GFP cleavage assay (Calvo-Garrido et al., 2011). This assay is based
on the observation that the GFP moiety is often cleaved as a whole from The core autophagy protein ATG16 is crucial for the expansion of
GFP-tagged autophagic substrates inside the autolysosome and accu- the isolation membrane by acting in concert with the ATG12-5 con-
mulates because of its resistance to further digestion. We expressed jugate as an E3 ligase in the ATG8(LC3) conjugation system (Noda and
GFP-PSMD1 and GFP-PSMD2 in AX2 and atg16‾ cells, respectively, and Inagaki, 2015). The protein is composed of an N-terminal half with a
found that upon treatment with NH4Cl the released free GFP was clearly CCD for homo-dimerization and binding sites for ATG5, clathrin,
detected in AX2 but not in atg16‾ cells (Fig. 5A). We conclude that the FIP200, and Rab33B (Gammoh et al., 2013; Itoh et al., 2008; Kuma
degradation of PSMD1 and PSMD2 is mediated by ATG16 and et al., 2002; Mizushima and Levine, 2010; Nishimura et al., 2013;

526
Q. Xiong et al. European Journal of Cell Biology 97 (2018) 523–532

Fig. 3. ATG16 and ATG8a (LC3) co-localize with RFP-PSMD1 and RFP-PSMD2 positive puncta. (A, B) Immunofluorescence microscopy of live atg16‾/atg16-gfp/rfp-
psmd1 and atg16‾/atg16-gfp/rfp-psmd2 cells. Arrows indicate partial co-localization of ATG16-GFP and RFP-PSMD1 and of ATG16-GFP and RFP-PSMD2. Scale bar:
2 μm. (C, D) Immunofluorescence microscopy of live AX2/gfp-atg8a/rfp-psmd1 and AX2/gfp-atg8a/rfp-psmd2 cells. RFP-PSMD1 or RFP-PSMD2, respectively, were
ectopically expressed in AX2 cells together with GFP-ATG8a. RFP-PSMD1 and RFP-PSMD2 puncta partially co-localized with the autophagosomal marker ATG8a
(LC3) (row 1). We often observed that the GFP-ATG8a signal expanded along the puncta to different degrees and sometimes completely encircled the puncta (row
2–4). Scale Bar: 2 μm.

Table 2 particle of the proteasome, as novel ATG16 interacting proteins and


Co-localization of RFP-PSMD1 and RFP-PSMD2 with ATG16-GFP and GFP- found that their interaction with ATG16 is direct (Figs. 1, 2 and S1). We
ATG8a, respectively. furthermore narrowed down the binding sites and could show that the
RFP-PSMD1 RFP-PSMD2 N-terminal 198 amino acids of ATG16 are sufficient for the binding to
PSMD1, while the C-terminal half (aa 310–612) directly interacts with
RFP puncta Co-localization RFP puncta Co-localization both proteins, PSMD1 and PSMD2 (Figs. 2B, C, S1). At present it is
with GFP with GFP
unclear how the interaction between ATG16 and PSMD1/2 is regulated.
# # % # # %
Possible elicitors could be e.g. environmental stress, non-stoichiometric
presence of PSMD1/2 with respect to other proteasome subunits or
ATG16-GFP 425 172 40.5 341 107 31.4 folding or activity problems.
GFP-ATG8a 1284 156 12.1 1013 242 23.9 The direct interaction raised the question whether ATG16 acts as an
autophagy receptor or adaptor for the degradation of PSMD1/2. Since
For ATG16-GFP the puncta from 5 and for GFP-ATG8a from 6 independent
experiments were counted. For each combination of tagged proteins at least 300 autophagy is a fast process we made use of the lysosomotropic com-
cells were analyzed. pound NH4Cl and of Bafilomycin A1, which both slow down the au-
tophagic flux by increasing the lysosomal pH, albeit by different me-
Ravikumar et al., 2010) and a C-terminal β-propeller structure, that chanisms (Aubry et al., 1993; Calvo-Garrido et al., 2011; Klionsky et al.,
binds NOD 1 and 2, TMEM59 and ubiquitin and appears to be crucial in 2016; Mesquita et al., 2013; Yoshimori et al., 1991). Treatment with
xenophagy (Boada-Romero et al., 2013; Fujita et al., 2013; Travassos these compounds led to the frequent detection of RFP-PSMD1 and RFP-
et al., 2010; Ver Heul et al., 2013). The numerous so far identified in- PSMD2 positive puncta in AX2 wild-type cells, however, we only very
teractors imply that ATG16 acts as a major hub in autophagy and we set rarely could detect such puncta in atg16‾ cells (Figs. 4A, B, S2B).
out to identify novel binding proteins in the Dictyostelium system. Using PSMD1 and PSMD2 puncta formation was rescued in atg16‾ cells ex-
GST-ATG16 as bait in pull down assays with total cell lysates, we pressing ATG16-GFP or ATG161−309-GFP but not ATG16156–612-GFP
identified PSMD1 and PSMD2, two subunits of the 19S regulatory (Fig. 4A, D). It was previously shown that the N-terminal half of ATG16

527
Q. Xiong et al. European Journal of Cell Biology 97 (2018) 523–532

respectively. RFP-PSMD1 or RFP-PSMD2 positive puncta were formed in atg16‾


cells after expression of ATG161−309-GFP (left images) but not after expression
of ATG16156-312-GFP, which lacks the AFIM (right images). Scale Bar: 2 μm.
Expression of tagged constructs was confirmed by Western blotting; 1, atg16‾/
rfp-psmd1/atg161-309-gfp; 2, atg16‾/rfp-psmd1/atg16156-312-gfp; 3, atg16‾/rfp-
psmd2/atg161-309-gfp; 4, atg16‾/rfp-psmd2/atg16156-312-gfp. PSMD1 and PSMD2
were detected with specific polyclonal antibodies, ATG161−309-GFP and
ATG16156-312-GFP were detected with a GFP specific monoclonal antibody.
Actin served as loading control and was detected with the Act1-7 anti actin
antibody (For interpretation of the references to colour in this figure legend, the
reader is referred to the web version of this article.).

is required for the recruitment of the ATG12-5/16 complex to the au-


tophagosomal membrane (Romanov et al., 2012). In this region of
ATG16 the AFIM (Kim et al., 2015) is located between amino acids 43
and 60 (Fig. 4B). The ATG16156–612-GFP fusion protein lacks the AFIM
and hence is not able to rescue puncta formation in atg16‾ cells. Al-
though ATG161−309 was not able to interact with PSMD2, expression of
ATG161−309-GFP in atg16‾ rescued also the formation of RFP-PSMD2
puncta (Figs. 2C, 4 D, S1A). It was reported that PSMD1 and PSMD2
directly interact in S. cerevisiae (Rosenzweig et al., 2008). In in vitro
pull-down assays we could, however, not find a direct interaction be-
tween Dictyostelium PSMD1 and PSMD2 (Fig. S4A), nevertheless,
binding of PSMD1 to ATG161−309-GFP could mediate an indirect in-
teraction between ATG16 and PSMD2 in vivo.
We next wanted to determine the nature and function of the puncta.
They could constitute protein aggregates, which could have been caused
by the overexpression of RFP-PSMD1 or RFP-PSMD2 in AX2 and the
different transgenic strains. However, the expression level of RFP-PSMD1
in atg16‾ and atg9‾/16‾ cells, which did not form RFP-PSMD1 positive
puncta, was as high as in AX2, atg16‾/atg16-gfp and atg9‾/16‾/atg16-gfp
cells (Fig. S2A). Furthermore, we frequently observed ubiquitinated
protein aggregates in atg16‾ cells (Xiong et al., 2015) but hardly any
RFP-PSMD1 or RFP-PSMD2 positive puncta (Figs. 4, S2B, C). Taken to-
gether, these results strongly argue against the hypothesis that the puncta
are protein aggregates. We then considered the possibility that the
puncta are autophagosomes and/or autolysosomes since their formation
was dependent on ATG16 (Fig. 4A, B). We found that around 40% of the
RFP-PSMD1 and 30% of the RFP-PSMD2 positive puncta co-localized
with ATG16, and 12 and 24%, respectively, with the autophagosomal
marker ATG8a (Fig. 3 and Table 2), suggesting that a considerable
fraction of the puncta are indeed autophagosomes. At present it is un-
clear why RFP-PSMD2 co-localization is lower with ATG16 and higher
with ATG8a as compared to RFP-PSMD1 puncta. To further investigate
whether a subfraction or all of the remaining puncta could be auto-
lysosomes, we applied three approaches: i) in a proteolytic cleavage
assay (Calvo-Garrido et al., 2011) we detected the released free GFP from
the GFP-PSMD1 and GFP-PSMD2 fusion proteins in AX2 wild-type cells
but not in atg16‾ cells (Fig. 5A). ii) We were able to monitor the lyso-
somal localization of PSMD1 and PSMD2 through the expression of RFP-
GFP-PSMD1 and RFP-GFP-PSMD2. The acidic pH of the lysosome
Fig. 4. The formation of PSMD1 and PSMD2 positive puncta is dependent on
quenches the GFP fluorescence, thus the emission of only red fluores-
ATG16. (A) Immunofluorescence analysis of live Dictyostelium cells expressing
RFP-PSMD1 or RFP-PSMD2, respectively. After treatment with NH4Cl, RFP- cence indicates acidification of the autophagosome through fusion with a
PSMD1 (top) or RFP-PSMD2 (bottom) were enriched in punctate structures lysosome (Fig. 5B). This strategy was originally described for RFP-GFP-
(white arrows) in AX2 and atg16‾/atg16-gfp cells but not in atg16‾ cells. Scale ATG8 which is often used to monitor autophagic flux (Mesquita et al.,
Bar: 2 μm. (B) Quantification of the percentage of AX2, atg16‾ and atg16‾/ 2013). iii) Finally, we investigated the co-localization of RFP-PSMD1 or
atg16-gfp cells containing RFP-PSMD1 or RFP-PSMD2 positive puncta. For each RFP-PSMD2 with LysoTracker and found that approximately 50% of the
experiment and strain more than 200 cells were analyzed. Error bars represent puncta co-localized with LysoTracker (Fig. 5C, Table 3). These results
standard deviations of three independent experiments. (C) Sequence alignment suggest that, dependent on ATG16, PSMD1 and PSMD2 get enriched in
of the ATG5-interacting motif (AFIM) of ATG16 from different organisms. Sc, autophagosomes, followed by degradation in autolysosomes. At present
Saccharomyces cerevisiae; Dr, Danio rerio; Hs, Homo sapiens; Mm, Mus musculus;
it is not clear if only excess PSMD1 and PSMD2, tagged as well as en-
Dd, Dictyostelium discoideum. Numbers denote the position of the first and the
dogenous, or the whole 19S regulatory particle or even the 26S protea-
last amino acid of the AFIM in ATG16 of the different organisms. Red letters
highlight key residues for ATG5 binding. (D) Immunofluorescence analysis of
some get degraded in this way in AX2 wild-type cells. In the latter case,
live Dictyostelium atg16‾ cells expressing either ATG161−309-GFP (left) or we would expect an increase of the protein levels of the proteasomal
ATG16156-312-GFP (right) and RFP-PSMD1 (top) or RFP-PSMD2 (bottom), subunits and a concomitant increase in proteasomal activity in atg16‾
and other autophagy-compromised cells. However, based on protein

528
Q. Xiong et al. European Journal of Cell Biology 97 (2018) 523–532

Fig. 5. PSMD1 and PSMD2 are degraded in autolysosomes. (A) Proteolytic


cleavage assay to monitor autophagy. GFP-PSMD1 and GFP-PSMD2 were ex-
pressed in AX2 and atg16‾ cells, respectively, cells were either left untreated (-)
or treated (+) two times for two hours with 100 mM NH4Cl, lysed and total cell
lysates subjected to Western blotting. The cleaved free GFP was detected with a
GFP-specific monoclonal antibody in AX2 cells but not in atg16‾ cells. The
positions of GFP-PSMD1, GFP-PSMD2 and of GFP are indicated. (B)
Fluorescence analysis of AX2 cells expressing RFP-GFP-PSMD1 or RFP-GFP-
PSMD2. Treatment of these cells with NH4Cl or Bafilomycin A1 followed by
confocal microscopy revealed the formation of red punctate structures for both,
RFP-GFP-PSMD1 and RFP-GFP-PSMD2 (white arrows). Since the GFP fluores-
cence is rapidly lost in the acidic environment of the lysosome these puncta are
by definition autolysosomes. Scale bar: 2 μm. (C) Fluorescence analysis of
LysoTracker labelled AX2 and atg16‾ cells expressing RFP-PSMD1 or RFP-
PSMD2. RFP-PSMD1 and RFP-PSMD2 positive puncta co-localize in many cases
with LysoTracker in AX2 cells (for quantitative analysis see Table 3). No such
RFP-PSMD1 and RFP-PSMD2 positive puncta are present in atg16‾ cells and
hence there is no co-localization with LysoTracker. Scale Bar: 2 μm (For inter-
pretation of the references to colour in this figure legend, the reader is referred
to the web version of this article.).

levels of subunits 4 and 5 (SU4 and SU5) of the 20S proteasome (Xiong
et al., 2015) and of PSMD1 and PSMD2 of the 19S regulatory particle
(Fig. S2A) there was no increase in the number of proteasomes in these
strains. Moreover, we previously found that proteasomal activity was
strongly reduced in different autophagy-compromised strains (Arhzaouy
et al., 2012; Messling et al., 2017; Xiong et al., 2015). We therefore
speculate that in atg16‾ and other autophagy-compromised cells pro-
teasomes with reduced activity, which are normally cleared by autop-
hagy, are not at all or only insufficiently removed. This then causes the
observed decrease in proteasomal activity (Arhzaouy et al., 2012;
Messling et al., 2017; Xiong et al., 2015).
For a long time autophagy and the UPS were thought to act in-
dependently but evidence is accumulating that these two major in-
tracellular pathways for protein and organelle clearance in eukaryotic
cells are interrelated (Bartel, 2015; Driscoll and Chowdhury, 2012;
Korolchuk et al., 2009, 2010; Kraft et al., 2010). An early finding was
that proteasomes accumulated in lysosomes upon nutrient starvation or
treatment with the lysosomal inhibitor leupeptin (Cuervo et al., 1995).
In colon cancer cells pharmacological inhibition of autophagy and also
downregulation of autophagy genes by RNAi resulted in an increase of
proteasomal subunits and proteasomal activity (Wang et al., 2013). In
contrast, inhibition of lysosomal activities by chloroquine in neuro-
blastoma cells led to an accumulation of ubiquitinated proteins and
reduced proteasomal activities (Qiao and Zhang, 2009). Furthermore,
inhibition of autophagy with Bafilomycin A1 or siRNA knock-down of
atg7 or atg12 resulted in an impaired clearance of UPS clients in HeLa
cells, consistent with reduced proteasomal activity (Korolchuk et al.,
2009). A quantitative proteomic analysis of autophagosomes showed,
that proteasomal proteins were abundant among the stimulus-in-
dependent common autophagosome-associated proteins (Dengjel et al.,
2012). Recently, it was reported that in Arabidopsis the degradation of
inactive 26S proteasomes is mediated by the proteasomal subunit
RPN10 (PSMD4) via ubiquitin and ATG8, and in S. cerevisiae via the
ubiquitin receptor Cue5 and the Hsp42 chaperone, a process the au-
thors termed proteaphagy (Marshall et al., 2015, 2016). Furthermore, it
was found that the autophagic clearance of proteasomes in yeast re-
quires the sorting nexin 4 and that upon starvation the proteaphagy
pathways for the core and regulatory particles of the proteasome are
different (Nemec et al., 2017; Waite et al., 2016). In mammals, stress-
induced proteaphagy was dependent on p62/SQSTM1 and ubiquitin
(Cohen-Kaplan et al., 2016). It appears therefore that proteaphagy
might be universal in eukaryotes, however, the mechanism and the
autophagy receptors appear to vary from organism to organism. In
Dictyostelium, impaired disposal of inactive proteasomes in autophagy-
compromised strains could cause the observed dramatic decrease in
proteasomal activity (Arhzaouy et al., 2012; Messling et al., 2017;

529
Q. Xiong et al. European Journal of Cell Biology 97 (2018) 523–532

Table 3
Co-localization of RFP-PSMD1 or RFP-PSMD2 with LysoTracker.
# of green # of red # of red and % of red puncta co- # of red puncta # of red and green % of red puncta > 1 μm co-localizing
puncta puncta green puncta localizing with green puncta > 1 μm puncta > 1 μm with green puncta

RFP-PSMD1 AX2 1318 968 483 49.9 69 60 87.0


atg16‾ 432 0 0 N/A 0 0 N/A
RFP-PSMD2 AX2 1403 1191 578 48.5 128 108 84.4
atg16‾ 447 0 0 N/A 0 0 N/A

Data from two independent experiments were quantified. Green (LysoTracker) and red (RFP-PSMD1 or RFP-PSMD2) puncta larger than 0.25 μm of 566 AX2/rfp-
psmd1, 322 atg16¯/rfp-psmd1, 607 AX2/rfp-psmd2 and 333 atg16¯/rfp-psmd2 cells were counted and the percentage of RFP-PSMD1 or RFP-PSMD2, respectively, co-
localizing with LysoTracker was calculated.

Xiong et al., 2015). These results and our identification of the 19S selected in the presence of proper antibiotics. Dictyostelium cells expres-
regulatory particle subunits PSMD1 and PSMD2 as direct binding sing RFP- and/or GFP-tagged proteins were identified by visual inspec-
partners of the core autophagy protein ATG16, suggest that ATG16 may tion under a fluorescence microscope and expression of fusion proteins
not only mediate the degradation of PSMD1 and PSMD2 but of the 19S was verified by Western blotting. All primers used in PCR reactions in
regulatory particle or even the complete 26S proteasome. Further work this study are listed in table S2.
is needed to solve the question whether ATG16 indeed acts as an au-
tophagy receptor or adaptor for proteaphagy in D. discoideum. 4.3. Protein quantification, antibody generation, SDS PAGE and western
blotting
4. Material and methods
For generation of PSMD1 and PSMD2 specific polyclonal antibodies
4.1. Dictyostelium strains (pAbs), the cDNA sequences encoding a hydrophilic fragment (aa
263–312 for PSMD1 and aa 13–79 for PSMD2) were amplified by PCR
D. discoideum AX2 was used as wild-type strain. The generation and and cloned into the pGEX-6P-1 GST expression vector. The fusion
phenotypes of atg9‾, atg16‾, atg16‾/atg16-gfp, atg9‾/16‾ and atg9‾/ protein was expressed in E. coli XL1 blue, purified using GST-Sepharose
16‾/atg16-gfp strains are described elsewhere (Tung et al., 2010; Xiong beads, released through cleavage with PreScission protease (GE
et al., 2015). Expression plasmids for RFP-PSMD1, RFP-PSMD2, GFP- Healthcare Life Science) and used for the immunization of rabbits
PSMD1, GFP-PSMD2, RFP-GFP-PSMD1, RFP-GFP-PSMD2, ATG161−309- (BioGenes GmbH, Germany). SDS-PAGE and Western blotting were
GFP and ATG16156-612-GFP were used for transformation of different essentially performed as described (Laemmli, 1970; Towbin et al.,
strains by electroporation. AX2 and mutant cells were grown at 21 °C in 1979). Dictyostelium total cell lysates were prepared and the proteins of
AX2 medium (for 1 L: 14.3 g bacteriological peptone, 7.15 g yeast ex- 2 × 105 cells were separated per lane by SDS gel electrophoresis. The
tract, 18 g maltose, 0.62 g Na2HPO4 x 2H2O, 0.49 g KH2PO4, pH 6.7) generated PSMD1 pAbs were affinity purified and used for Western
with shaking at 160 rpm in Erlenmeyer flasks or on SM agar plates with blotting at a 1:2000 dilution. PSMD2 pAbs were used at a 1:2000 di-
Klebsiella aerogenes (Brink et al., 1990; Watts and Ashworth, 1970; lution. GFP was detected with monoclonal antibody K3-184-2 at a 1:20
Williams and Newell, 1976). The different transgenic strains used in dilution (Noegel et al., 2004). RFP was detected with monoclonal an-
this study are listed in table S1. tibody K64-372-1 at a 1:20 dilution (Fischer et al., 2004). Actin was
detected with monoclonal antibody Act1-7 at a 1:15 dilution (Simpson
4.2. Vector construction and transformation et al., 1984). Secondary antibodies used were anti-mouse and anti-
rabbit lgG conjugated with peroxidase (POD) (Sigma, Germany) at a
cDNAs encoding either full-length or parts of the ATG16 (gene ID: 1:10,000 dilution followed by chemiluminescence detection. Images
DDB_G0275323, http://www.dictybase.org), of PSMD1 were recorded and analyzed using the Fluorchem SP imaging system
(DDB_G0287953) and PSMD2 (DDB_G0293752) were amplified by PCR (Alpha Innotech, USA). Relative protein amounts were determined
and cloned into the pGEX-6P-1 or pGEX-4T-1 GST expression vectors. For densitometrically using the Spot Denso tool of the AlphaEaseFC soft-
ectopic expression of PSMD1 and PSMD2 fused C-terminally to RFP in ware (Alpha Innotech, USA).
different Dictyostelium strains the p389-2-mRFPMars vector was used
(Fischer et al., 2004). The PSMD1 and PSMD2 coding sequences were 4.4. GFP cleavage assay
amplified by PCR. The PSMD1 cDNA was cloned into the p389-2-
mRFPMars vector via the BamHI and EcoRI restriction sites and the The GFP cleavage assay was performed essentially as described
PSMD2 cDNA via the EcoRI and ClaI restriction sites. For ectopic ex- (Calvo-Garrido et al., 2011). Briefly, 1 mL AX2 or atg16‾ cells expressing
pression of PSMD1 and PSMD2 fused C-terminally to GFP the pBsr-N-GFP GFP-PSMD1 or GFP-PSMD2 in AX2 medium at a concentration of
vector was used (Blau-Wasser et al., 2009). The PSMD1 and PSMD2 1 × 106C/ml were deposited in the wells of a 6-well plate. The cells were
coding sequences were cloned into the pBsr-N-GFP vector via the PstI and allowed to settle down for 15 min, washed twice with Soerensen buffer
XmaI restriction sites, respectively. For ectopic expression of (2.0 mM Na2PO4, 14.6 mM KH2PO4, pH 6.0), then treated with 100 mM
ATG161−309–GFP and ATG16156-612–GFP the pBsr-C1-GFP vector was NH4Cl in Soerensen buffer for 2 h. The treatment with 100 mM NH4Cl in
used (Blau-Wasser et al., 2009). Both coding sequences were amplified Soerensen buffer was repeated for another 2 h. Proteins of total cell ly-
by PCR and cloned into pBsr-C1-GFP via the PstI and XmaI restriction sates were separated in 12% SDS-PAGE gels, transferred by Western
sites. RFP-GFP-PSMD1 and RFP-GFP-PSMD2 expressing constructs were blotting and GFP was detected with a specific monoclonal antibody.
generated by insertion of the GFP coding sequence, amplified by PCR
from the pBsr-N2-GFP vector, into the p389-2-mRFPMars/RFP-PSMD1 4.5. Pulldown and co-immunoprecipitation (IP) assays
and p389-2-mRFPMars/RFP-PSMD2 plasmids (see above) via BamHI and
EcoRI restriction sites, respectively. All final expression constructs were To identify new binding partners of ATG16, we used recombinant
verified by sequencing and were introduced into bacterial cells by heat GST-ATG16 fusion protein bound to Glutathione Sepharose 4B beads as
shock and into Dictyostelium cells by electroporation. Transformants were “bait” with a lysate from AX2 cells. Negative controls were GST-ATG16

530
Q. Xiong et al. European Journal of Cell Biology 97 (2018) 523–532

beads without AX2 Lysate and AX2 lysate with GST beads only. AX2 Loflo medium. Cells expressing RFP fusion proteins were analyzed
cells were lysed in 25 mM Tris/HCl pH 7.5, 5 mM EDTA, 150 mM NaCl, without pre-incubation in Loflo medium. GFP and/or RFP fusion pro-
10% Glycerol, 0.1% Triton X-100, 1 mM DTT and 1x Protease Inhibitor teins expressing cells were washed once with Soerensen buffer followed
Cocktail (Roche). Cell lysates were centrifuged at 20,000 xg for 10 min by an incubation with 100 mM NH4Cl or 5 μM Bafilomycin A1 (Enzo
at 4 °C and the supernatant containing the soluble proteins was in- life sciences) in Soerensen buffer for 2 h. Treatment with NH4Cl or
cubated with either GST or GST-ATG16 bound to Glutathione Bafilomycin A1 increases the lysosomal pH and thus slows autophagic
Sepharose beads for 3 h at 4 °C on a rotating wheel. The beads were degradation of proteins (Aubry et al., 1993; Mesquita et al., 2013). The
collected by centrifugation at 500 x g for 15 s and then washed three cells were treated in the same way with 100 mM NH4Cl and with or
times with the same buffer. The bound proteins were eluted from the without 0.1 μM LysoTracker Deep Red (Molecular Probes) to visualize
beads by boiling in 1 x SDS buffer at 95 °C for 5 min, separated in 10% lysosomes or with 5 μM Bafilomycin A1 for another 2 h. Delivery of
and 15% SDS-PAGE gels and stained with the Imperial Protein Stain. 24 RFP-GFP-PSMD1 or RFP-GFP-PSMD2 from the autophagosome to the
distinct bands in the size range between 400 and 15 kDa that were autophagolysosome can be monitored in this way because the acidic
missing in the negative controls were excised and proteins were in-gel environment of the lysosome quenches the more sensitive fluorescence
digested after reduction and alkylation with trypsin. Tandem mass of GFP, while the fluorescence of RFP is preserved. Thus, the presence
spectrometry (LCeMS/MS) was performed at the Center for Molecular of red puncta lacking the green fluorescence indicates the presence of
Medicine Cologne (CMMC) as described (Feldkirchner et al., 2012). RFP-GFP-tagged fusion proteins in autolysosomes (Mesquita et al.,
LCeMS data were acquired on a HCT ETD II ion-trap mass spectrometer 2013). Confocal images of cells were taken with a TCS SP5 laser
equipped with a nano ESI source (Bruker Daltonics). Data-dependent scanning microscope (Leica) with a 63x HCX PL APO lambda blue 1.40
acquisition of MS and MS/MS spectra was controlled by the Compass oil UV immersion objective. Excitation of GFP was at 488 nm, emission
3.0 software using previously published parameters (Albert et al., 500–550 nm; of RFP at 561 nm, emission 570-620 nm; and of Lyso-
2010). Proteins were identified by searching the NCBInr release Tracker Deep Red at 647 nm, emission 646-727 nm. Images were pro-
20,080,210 (Dictyostelium) using a local installation of MASCOT 2.2 cessed using the Leica Application Suite (LAS AF) software.
(Matrix Science Ltd). Searches were submitted via Proteinscape 2.0
(Bruker Daltonics) with the following parameter settings: enzyme Author’s contributions
“trypsin”, species “Dictyostelium”, fixed modifications “carbamido-
methyl (Cys)”, optional modifications “Methionine oxidation” and QX carried out the molecular lab work, performed data analysis and
missed cleavages “1″. The mass tolerance was set to 0.3 Da for peptide drafted the manuscript; SF, MK, RM and SM contributed to the mole-
and fragment spectra. The identified proteins are listed in table S3. cular lab work; LE conceived of the study, designed and coordinated it
Co-immunoprecipitation assays were performed with axenically and finalised the manuscript. All authors gave final approval for pub-
grown atg16‾/atg16-gfp/rfp-psmd1 or atg16‾/atg16-gfp/rfp-psmd2 cells lication.
using GFP-Trap® (ChromoTek). Log phase cells (2–4 × 106C/ml) were
harvested, centrifuged at 500 xg for 4 min and washed twice with Acknowledgments
Soerensen buffer. Cell pellets were frozen at −20 °C overnight and
lysed according to the manufacturer’s instructions (ChromoTek). We thank Dr. Angelika A. Noegel and Dr. Christoph Clemen for
Recombinant PSMD1, PSMD2, full-length ATG16 and fragments of helpful discussions, Dr. Ricardo Escalante for providing plasmids, Dr.
ATG16 were expressed in E. coli XL1 blue or DH5α or ArcticExpress RIL Salvatore Bozzaro for advice with LysoTracker and Maria Stumpf for help
cells (Agilent Technologies) and purified using Glutathione Sepharose with immunofluorescence studies. LE acknowledges support of this work
beads. For pull-down assays of PSMD1 and PSMD2 with full-length by the German Research Foundation (Deutsche Forschungsgemeinschaft:
ATG16 and fragments of ATG16, PSMD1 and PSMD2 were released CRC670, TP01) and by Köln Fortune. This work was supported by the
from the beads through cleavage with PreScission protease, diluted China scholarship council to QX.
with RIPA buffer (50 mM Tris/HCl pH 7.5, 150 mM NaCl, 1% NP-40,
0.5% Na-deoxycholate) and then incubated with ATG16 and fragments Appendix A. Supplementary data
of ATG16 bound to Glutathione Sepharose beads for 3 h at 4 °C. Beads
were washed with RIPA buffer, bound proteins were eluted by boiling Supplementary material related to this article can be found, in the
the beads in SDS sample buffer, proteins were separated by SDS-PAGE online version, at doi:https://doi.org/10.1016/j.ejcb.2018.09.002.
and then analyzed by western blotting. To test binding of ATG16 from
total Dictyostelium cell lysates to recombinant PSMD1 and PSMD2, cells References
were lysed in 25 mM Tris/HCl pH 7.5, 5 mM EDTA, 150 mM NaCl, 10%
Glycerol, 0.1% Triton X-100, 1 mM DTT and 1x Protease Inhibitor Albert, T.K., Laubinger, W., Muller, S., Hanisch, F.G., Kalinski, T., Meyer, F., Hoffmann,
W., 2010. Human intestinal TFF3 forms disulfide-linked heteromers with the mucus-
Cocktail (Roche). Cell lysates were centrifuged at 20,000 xg for 10 min
associated FCGBP protein and is released by hydrogen sulfide. J. Proteome Res. 9,
at 4 °C, the supernatant containing the soluble proteins was incubated 3108–3117.
with GST-PSMD1 or GST-PSMD2 bound to Glutathione Sepharose beads Arhzaouy, K., Strucksberg, K.H., Tung, S.M., Tangavelou, K., Stumpf, M., Faix, J.,
Schroder, R., Clemen, C.S., Eichinger, L., 2012. Heteromeric p97/p97R155C com-
for 3 h at 4 °C. Beads were washed twice with the same buffer and plexes induce dominant negative changes in wild-type and autophagy 9-deficient
bound proteins were analyzed by western blotting. Dictyostelium strains. PLoS One 7, e46879.
Aubry, L., Klein, G., Martiel, J.L., Satre, M., 1993. Kinetics of endosomal pH evolution in
Dictyostelium discoideum amoebae. Study by fluorescence spectroscopy. J. Cell. Sci.
4.6. Fluorescence microscopy 105 (Pt 3), 861–866.
Bartel, B., 2015. Proteaphagy-selective autophagy of inactive proteasomes. Mol. Cell 58,
970–971.
For live cell imaging, Dictyostelium cells ectopically expressing RFP Blau-Wasser, R., Euteneuer, U., Xiong, H., Gassen, B., Schleicher, M., Noegel, A.A., 2009.
or GFP fusion proteins, RFP-GFP-PSMD1 or RFP-GFP-PSMD2, were CP250, a novel acidic coiled-coil protein of the Dictyostelium centrosome, affects
detached from the plastic surface of a cell culture dish (Ø 100 mm) and growth, chemotaxis, and the nuclear envelope. Mol. Biol. Cell 20, 4348–4361.
Bloomfield, G., Traynor, D., Sander, S.P., Veltman, D.M., Pachebat, J.A., Kay, R.R., 2015.
transferred to a μ-Dish (Ø 35 mm, Ibidi). Cells were allowed to attach Neurofibromin controls macropinocytosis and phagocytosis in Dictyostelium. Elife 4,
for 15 min. Dictyostelium cells that expressed GFP fusion proteins were e04940.
washed once with Loflo (low fluorescence) medium (Formedium) and Boada-Romero, E., Letek, M., Fleischer, A., Pallauf, K., Ramon-Barros, C., Pimentel-
Muinos, F.X., 2013. TMEM59 defines a novel ATG16L1-binding motif that promotes
pre-incubated overnight in Loflo medium to decrease the cytosolic local activation of LC3. EMBO J. 32, 566–582.
background fluorescence (Bloomfield et al., 2015). Approximately two Brink, M., Gerisch, G., Isenberg, G., Noegel, A.A., Segall, J.E., Wallraff, E., Schleicher, M.,
1990. A Dictyostelium mutant lacking an F-actin cross-linking protein, the 120-kD
hours before treatment of the cells, the medium was replaced with fresh

531
Q. Xiong et al. European Journal of Cell Biology 97 (2018) 523–532

gelation factor. J. Cell Biol. 111, 1477–1489. Dictyostelium discoideum autophagy 8 proteins have distinct autophagic functions.
Calvo-Garrido, J., Escalante, R., 2010. Autophagy dysfunction and ubiquitin-positive Eur. J. Cell Biol. 96, 312–324.
protein aggregates in Dictyostelium cells lacking Vmp1. Autophagy 6, 100–109. Mizushima, N., Levine, B., 2010. Autophagy in mammalian development and differ-
Calvo-Garrido, J., Carilla-Latorre, S., Mesquita, A., Escalante, R., 2011. A proteolytic entiation. Nat. Cell Biol. 12, 823–830.
cleavage assay to monitor autophagy in Dictyostelium discoideum. Autophagy 7, Mizushima, N., Noda, T., Ohsumi, Y., 1999. Apg16p is required for the function of the
1063–1068. Apg12p-Apg5p conjugate in the yeast autophagy pathway. EMBO J. 18, 3888–3896.
Calvo-Garrido, J., King, J.S., Munoz-Braceras, S., Escalante, R., 2014. Vmp1 regulates Mizushima, N., Kuma, A., Kobayashi, Y., Yamamoto, A., Matsubae, M., Takao, T.,
PtdIns3P signaling during autophagosome formation in Dictyostelium discoideum. Natsume, T., Ohsumi, Y., Yoshimori, T., 2003. Mouse Apg16L, a novel WD-repeat
Traffic 15, 1235–1246. protein, targets to the autophagic isolation membrane with the Apg12-Apg5 con-
Cohen-Kaplan, V., Livneh, I., Avni, N., Fabre, B., Ziv, T., Kwon, Y.T., Ciechanover, A., jugate. J. Cell. Sci. 116, 1679–1688.
2016. p62- and ubiquitin-dependent stress-induced autophagy of the mammalian 26S Munoz-Braceras, S., Calvo, R., Escalante, R., 2015. TipC and the chorea-acanthocytosis
proteasome. Proc. Natl. Acad. Sci. U. S. A. 113, E7490–E7499. protein VPS13A regulate autophagy in Dictyostelium and human HeLa cells.
Cuervo, A.M., Palmer, A., Rivett, A.J., Knecht, E., 1995. Degradation of proteasomes by Autophagy 11, 918–927.
lysosomes in rat liver. Eur. J. Biochem. 227, 792–800. Neer, E.J., Schmidt, C.J., Nambudripad, R., Smith, T.F., 1994. The ancient regulatory-
Dengjel, J., Hoyer-Hansen, M., Nielsen, M.O., Eisenberg, T., Harder, L.M., Schandorff, S., protein family of WD-repeat proteins. Nature 371, 297–300.
Farkas, T., Kirkegaard, T., Becker, A.C., et al., 2012. Identification of autophagosome- Nemec, A.A., Howell, L.A., Peterson, A.K., Murray, M.A., Tomko Jr, R.J., 2017.
associated proteins and regulators by quantitative proteomic analysis and genetic Autophagic clearance of proteasomes in yeast requires the conserved sorting nexin
screens. Mol. Cell Proteomics 11 (M111), 014035. Snx4. J. Biol. Chem. 292, 21466–21480.
Driscoll, J.J., Chowdhury, R.D., 2012. Molecular crosstalk between the proteasome, ag- Nishimura, T., Kaizuka, T., Cadwell, K., Sahani, M.H., Saitoh, T., Akira, S., Virgin, H.W.,
gresomes and autophagy: translational potential and clinical implications. Cancer Mizushima, N., 2013. FIP200 regulates targeting of Atg16L1 to the isolation mem-
Lett. 325, 147–154. brane. EMBO Rep. 14, 284–291.
Feldkirchner, S., Schessl, J., Muller, S., Schoser, B., Hanisch, F.G., 2012. Patient-specific Noda, N.N., Inagaki, F., 2015. Mechanisms of autophagy. Annu. Rev. Biophys. 44,
protein aggregates in myofibrillar myopathies: laser microdissection and differential 101–122.
proteomics for identification of plaque components. Proteomics 12, 3598–3609. Noegel, A.A., Blau-Wasser, R., Sultana, H., Muller, R., Israel, L., Schleicher, M., Patel, H.,
Fischer, M., Haase, I., Simmeth, E., Gerisch, G., Muller-Taubenberger, A., 2004. A brilliant Weijer, C.J., 2004. The cyclase-associated protein CAP as regulator of cell polarity
monomeric red fluorescent protein to visualize cytoskeleton dynamics in and cAMP signaling in Dictyostelium. Mol. Biol. Cell 15, 934–945.
Dictyostelium. FEBS Lett. 577, 227–232. Otto, G.P., Wu, M.Y., Kazgan, N., Anderson, O.R., Kessin, R.H., 2004. Dictyostelium
Fujita, N., Itoh, T., Omori, H., Fukuda, M., Noda, T., Yoshimori, T., 2008. The Atg16L macroautophagy mutants vary in the severity of their developmental defects. J. Biol.
complex specifies the site of LC3 lipidation for membrane biogenesis in autophagy. Chem. 279, 15621–15629.
Mol. Biol. Cell 19, 2092–2100. Pashkova, N., Gakhar, L., Winistorfer, S.C., Yu, L., Ramaswamy, S., Piper, R.C., 2010.
Fujita, N., Saitoh, T., Kageyama, S., Akira, S., Noda, T., Yoshimori, T., 2009. Differential WD40 repeat propellers define a ubiquitin-binding domain that regulates turnover of
involvement of Atg16L1 in Crohn disease and canonical autophagy: analysis of the F box proteins. Mol. Cell 40, 433–443.
organization of the Atg16L1 complex in fibroblasts. J. Biol. Chem. 284, Qiao, L., Zhang, J., 2009. Inhibition of lysosomal functions reduces proteasomal activity.
32602–32609. Neurosci. Lett. 456, 15–19.
Fujita, N., Morita, E., Itoh, T., Tanaka, A., Nakaoka, M., Osada, Y., Umemoto, T., Saitoh, Ravikumar, B., Duden, R., Rubinsztein, D.C., 2002. Aggregate-prone proteins with poly-
T., Nakatogawa, H., et al., 2013. Recruitment of the autophagic machinery to en- glutamine and polyalanine expansions are degraded by autophagy. Hum. Mol. Genet.
dosomes during infection is mediated by ubiquitin. J. Cell Biol. 203, 115–128. 11, 1107–1117.
Gammoh, N., Florey, O., Overholtzer, M., Jiang, X., 2013. Interaction between FIP200 Ravikumar, B., Moreau, K., Jahreiss, L., Puri, C., Rubinsztein, D.C., 2010. Plasma mem-
and ATG16L1 distinguishes ULK1 complex-dependent and -independent autophagy. brane contributes to the formation of pre-autophagosomal structures. Nat. Cell Biol.
Nat. Struct. Mol. Biol. 20, 144–149. 12, 747–757.
Gao, Z., Gammoh, N., Wong, P.M., Erdjument-Bromage, H., Tempst, P., Jiang, X., 2010. Romanov, J., Walczak, M., Ibiricu, I., Schuchner, S., Ogris, E., Kraft, C., Martens, S., 2012.
Processing of autophagic protein LC3 by the 20S proteasome. Autophagy 6, 126–137. Mechanism and functions of membrane binding by the Atg5-Atg12/Atg16 complex
Hanada, T., Noda, N.N., Satomi, Y., Ichimura, Y., Fujioka, Y., Takao, T., Inagaki, F., during autophagosome formation. EMBO J. 31, 4304–4317.
Ohsumi, Y., 2007. The Atg12-Atg5 conjugate has a novel E3-like activity for protein Rosenzweig, R., Osmulski, P.A., Gaczynska, M., Glickman, M.H., 2008. The central unit
lipidation in autophagy. J. Biol. Chem. 282, 37298–37302. within the 19S regulatory particle of the proteasome. Nat. Struct. Mol. Biol. 15,
Itoh, T., Fujita, N., Kanno, E., Yamamoto, A., Yoshimori, T., Fukuda, M., 2008. Golgi- 573–580.
resident small GTPase Rab33B interacts with Atg16L and modulates autophagosome Schreiber, A., Peter, M., 2014. Substrate recognition in selective autophagy and the
formation. Mol. Biol. Cell 19, 2916–2925. ubiquitin-proteasome system. Biochim. Biophys. Acta 1843, 163–181.
Kim, H.M., Yu, Y., Cheng, Y., 2011. Structure characterization of the 26S proteasome. Simpson, P.A., Spudich, J.A., Parham, P., 1984. Monoclonal antibodies prepared against
Biochim. Biophys. Acta 1809, 67–79. Dictyostelium actin: characterization and interactions with actin. J. Cell Biol. 99,
Kim, J.H., Hong, S.B., Lee, J.K., Han, S., Roh, K.H., Lee, K.E., Kim, Y.K., Choi, E.J., Song, 287–295.
H.K., 2015. Insights into autophagosome maturation revealed by the structures of Smith, T.F., Gaitatzes, C., Saxena, K., Neer, E.J., 1999. The WD repeat: a common ar-
ATG5 with its interacting partners. Autophagy 11, 75–87. chitecture for diverse functions. Trends Biochem. Sci. 24, 181–185.
Klionsky, D.J., Abdelmohsen, K., Abe, A., Abedin, M.J., Abeliovich, H., Acevedo Arozena, Suzuki, K., Kirisako, T., Kamada, Y., Mizushima, N., Noda, T., Ohsumi, Y., 2001. The pre-
A., Adachi, H., Adams, C.M., Adams, P.D., et al., 2016. Guidelines for the use and autophagosomal structure organized by concerted functions of APG genes is essential
interpretation of assays for monitoring autophagy, 3rd edition. Autophagy, vol. 12. for autophagosome formation. EMBO J. 20, 5971–5981.
pp. 1–222. Towbin, H., Staehelin, T., Gordon, J., 1979. Electrophoretic transfer of proteins from
Korolchuk, V.I., Mansilla, A., Menzies, F.M., Rubinsztein, D.C., 2009. Autophagy inhibi- polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc.
tion compromises degradation of ubiquitin-proteasome pathway substrates. Mol. Cell Natl. Acad. Sci. U. S. A. 76, 4350–4354.
33, 517–527. Travassos, L.H., Carneiro, L.A., Ramjeet, M., Hussey, S., Kim, Y.G., Magalhaes, J.G., Yuan,
Korolchuk, V.I., Menzies, F.M., Rubinsztein, D.C., 2010. Mechanisms of cross-talk be- L., Soares, F., Chea, E., et al., 2010. Nod1 and Nod2 direct autophagy by recruiting
tween the ubiquitin-proteasome and autophagy-lysosome systems. FEBS Lett. 584, ATG16L1 to the plasma membrane at the site of bacterial entry. Nat. Immunol. 11,
1393–1398. 55–62.
Kraft, C., Peter, M., Hofmann, K., 2010. Selective autophagy: ubiquitin-mediated re- Tung, S.M., Unal, C., Ley, A., Pena, C., Tunggal, B., Noegel, A.A., Krut, O., Steinert, M.,
cognition and beyond. Nat. Cell Biol. 12, 836–841. Eichinger, L., 2010. Loss of Dictyostelium ATG9 results in a pleiotropic phenotype
Kuma, A., Mizushima, N., Ishihara, N., Ohsumi, Y., 2002. Formation of the approximately affecting growth, development, phagocytosis and clearance and replication of
350-kDa Apg12-Apg5.Apg16 multimeric complex, mediated by Apg16 oligomeriza- Legionella pneumophila. Cell. Microbiol. 12, 765–780.
tion, is essential for autophagy in yeast. J. Biol. Chem. 277, 18619–18625. Ver Heul, A.M., Fowler, C.A., Ramaswamy, S., Piper, R.C., 2013. Ubiquitin regulates
Laemmli, U.K., 1970. Cleavage of structural proteins during the assembly of the head of caspase recruitment domain-mediated signaling by nucleotide-binding oligomeriza-
bacteriophage T4. Nature 227, 680–685. tion domain-containing proteins NOD1 and NOD2. J. Biol. Chem. 288, 6890–6902.
Lee, J., Kim, H.R., Quinley, C., Kim, J., Gonzalez-Navajas, J., Xavier, R., Raz, E., 2012. Waite, K.A., De-La Mota-Peynado, A., Vontz, G., Roelofs, J., 2016. Starvation induces
Autophagy suppresses interleukin-1beta (IL-1beta) signaling by activation of p62 proteasome autophagy with different pathways for core and regulatory particles. J.
degradation via lysosomal and proteasomal pathways. J. Biol. Chem. 287, Biol. Chem. 291, 3239–3253.
4033–4040. Wang, X.J., Yu, J., Wong, S.H., Cheng, A.S., Chan, F.K., Ng, S.S., Cho, C.H., Sung, J.J., Wu,
Marshall, R.S., Li, F., Gemperline, D.C., Book, A.J., Vierstra, R.D., 2015. Autophagic de- W.K., 2013. A novel crosstalk between two major protein degradation systems: reg-
gradation of the 26S proteasome is mediated by the dual ATG8/Ubiquitin receptor ulation of proteasomal activity by autophagy. Autophagy 9, 1500–1508.
RPN10 in Arabidopsis. Mol. Cell 58, 1053–1066. Watts, D.J., Ashworth, J.M., 1970. Growth of myxameobae of the cellular slime mould
Marshall, R.S., McLoughlin, F., Vierstra, R.D., 2016. Autophagic turnover of inactive 26S Dictyostelium discoideum in axenic culture. Biochem. J. 119, 171–174.
proteasomes in yeast is directed by the ubiquitin receptor Cue5 and the Hsp42 cha- Webb, J.L., Ravikumar, B., Atkins, J., Skepper, J.N., Rubinsztein, D.C., 2003. Alpha-sy-
perone. Cell Rep. 16, 1717–1732. nuclein is degraded by both autophagy and the proteasome. J. Biol. Chem. 278,
Matthias, J., Messling, S., Eichinger, L., 2016. The two Dictyostelium autophagy eight 25009–25013.
proteins, ATG8a and ATG8b, associate with the autophagosome in succession. Eur. J. Williams, K.L., Newell, P.C., 1976. A genetic study of aggregation in the cellular slime
Cell Biol. 95, 15–25. mould Dictyostelium discoideum using complementation analysis. Genetics 82,
Mesquita, A., Calvo-Garrido, J., Carilla-Latorre, S., Escalante, R., 2013. Monitoring au- 287–307.
tophagy in Dictyostelium. Methods Mol. Biol. 983, 461–470. Xiong, Q., Unal, C., Matthias, J., Steinert, M., Eichinger, L., 2015. The phenotypes of
Mesquita, A., Cardenal-Munoz, E., Dominguez, E., Munoz-Braceras, S., Nunez-Corcuera, ATG9, ATG16 and ATG9/16 knock-out mutants imply autophagy-dependent and
B., Phillips, B.A., Tabara, L.C., Xiong, Q., Coria, R., et al., 2017. Autophagy in -independent functions. Open Biol. 5, 150008.
Dictyostelium: mechanisms, regulation and disease in a simple biomedical model. Yoshimori, T., Yamamoto, A., Moriyama, Y., Futai, M., Tashiro, Y., 1991. Bafilomycin A1,
Autophagy 13, 24–40. a specific inhibitor of vacuolar-type H(+)-ATPase, inhibits acidification and protein
Messling, S., Matthias, J., Xiong, Q., Fischer, S., Eichinger, L., 2017. The two degradation in lysosomes of cultured cells. J. Biol. Chem. 266, 17707–17712.

532

You might also like