Download The LIR motif – crucial for selective autophagy

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Cell membrane wikipedia , lookup

Phosphorylation wikipedia , lookup

SR protein wikipedia , lookup

Hedgehog signaling pathway wikipedia , lookup

Magnesium transporter wikipedia , lookup

Cytokinesis wikipedia , lookup

Proteasome wikipedia , lookup

Protein moonlighting wikipedia , lookup

Protein wikipedia , lookup

Endomembrane system wikipedia , lookup

Protein phosphorylation wikipedia , lookup

Intrinsically disordered proteins wikipedia , lookup

Apoptosome wikipedia , lookup

Programmed cell death wikipedia , lookup

G protein–coupled receptor wikipedia , lookup

JADE1 wikipedia , lookup

List of types of proteins wikipedia , lookup

Proteolysis wikipedia , lookup

Signal transduction wikipedia , lookup

Paracrine signalling wikipedia , lookup

Autophagy wikipedia , lookup

Transcript
Commentary
3237
The LIR motif – crucial for selective autophagy
Åsa Birna Birgisdottir, Trond Lamark and Terje Johansen*
Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsø, 9037 Tromsø, Norway
*Author for correspondence ([email protected])
Journal of Cell Science 126, 3237–3247
ß 2013. Published by The Company of Biologists Ltd
doi: 10.1242/jcs.126128
Journal of Cell Science
Summary
(Macro)autophagy is a fundamental degradation process for macromolecules and organelles of vital importance for cell and tissue
homeostasis. Autophagy research has gained a strong momentum in recent years because of its relevance to cancer, neurodegenerative
diseases, muscular dystrophy, lipid storage disorders, development, ageing and innate immunity. Autophagy has traditionally been
thought of as a bulk degradation process that is mobilized upon nutritional starvation to replenish the cell with building blocks and keep
up with the energy demand. This view has recently changed dramatically following an array of papers describing various forms of
selective autophagy. A main driving force has been the discovery of specific autophagy receptors that sequester cargo into forming
autophagosomes (phagophores). At the heart of this selectivity lies the LC3-interacting region (LIR) motif, which ensures the targeting
of autophagy receptors to LC3 (or other ATG8 family proteins) anchored in the phagophore membrane. LIR-containing proteins include
cargo receptors, members of the basal autophagy apparatus, proteins associated with vesicles and of their transport, Rab GTPaseactivating proteins (GAPs) and specific signaling proteins that are degraded by selective autophagy. Here, we comment on these new
insights and focus on the interactions of LIR-containing proteins with members of the ATG8 protein family.
Key words: ATG8, LC3, GABARAP, LIR, p62, Selective autophagy
Introduction
Macroautophagy (hereafter referred to as autophagy) is an
intracellular degradation process, in which a double membrane
structure called the phagophore expands and closes upon itself to
sequester part of the cytoplasm to form an autophagosome
(varying in diameter from 0.5 to 1.5 mm). First, the
autophagosome fuses either with a late endosome forming an
amphisome or directly with a lysosome forming an autolysosome.
Amphisomes also fuse with lysosomes to form autolysosomes
(Mizushima et al., 2011) (Fig. 1). This process can degrade all
kinds of molecules and supramolecular structures in the cytoplasm,
including organelles such as peroxisomes and mitochondria
(Johansen and Lamark, 2011). Because of its fundamental
importance in cellular homeostasis and cellular signaling,
autophagy is highly relevant for a number of diseases, including
cancer, neurodegenerative diseases, muscular dystrophy, lipidstorage disorders and processes such as development, ageing and
innate immunity (Levine and Kroemer, 2008; Levine et al., 2011;
Mizushima and Komatsu, 2011; Deretic, 2012). In addition to
(macro)autophagy, microautophagy and chaperone-mediated
autophagy represent distinct autophagy pathways (Arias and
Cuervo, 2011; Mijaljica et al., 2011).
Pioneering genetic studies in yeast have revealed a number of
AuTophaGy (ATG) genes (Nakatogawa et al., 2009; Mizushima
et al., 2011). Currently, 38 Atg proteins are known in yeast. Of
these, 17 (Atg1 to Atg10, Atg12 to Atg16, Atg18 and Atg22) are
part of the core autophagy machinery used by all the different
autophagy pathways. The components of the core autophagy
machinery are well conserved from yeast to mammals and appear
to act in a similar hierarchical manner. In mammals the core
machinery consists of (i) the complex of uncoordinated 51-like
kinase 1 and 2 (ULK1–ULK2 ); (ii) a class III phosphatidylinositol
3-kinase (PI 3-kinase) complex; (iii) ATG2A and ATG2B, and the
mammalian Atg18 homologs WD-repeat protein interacting with
phosphoinositides 1, 2, 3 and 4 (WIPI1, WIPI2, WIPI3 and WIPI4,
respectively); (iv) ATG9; (v) a complex of the ATG12–ATG5
conjugate and Atg16L1 and; (vi) ATG8 or the microtubuleassociated proteins 1A/1B light chain 3 (MAP1LC3 or LC3)
proteins (Fig. 1).
Members of the ATG8 family are the only known ubiquitinlike (Ubl) proteins that are conjugated to a lipid, namely
phosphatidylethanolamine (PE). ATG8-PE is present both on the
outer and inner membranes of the phagophore. During
autophagosome maturation, ATG8 is deconjugated from the
outer membrane by ATG4. This is necessary for autophagosome
biogenesis (Mizushima et al., 2011). In mammals, two
subfamilies of at least seven ATG8 proteins exist: the LC3
proteins LC3A, LC3B and LC3C, with two N-terminal splice
variants of LC3A, and GABARAP (c-amino butyric acid
receptor-associated protein), GABARAPL1 and GABARAPL2.
In mammals, LC3B is the most prevalent and well-established
autophagosome marker. Yeast has only one Atg8 homolog,
Caenorhabditis elegans and Drosophila melanogaster have two,
whereas Arabidopsis thaliana has nine ATG8 homologs
(reviewed by Shpilka et al., 2011).
Autophagy was traditionally regarded as a non-selective, bulk
degradation process mainly induced to replenish energy stores
upon starvation. A distinction is usually made between basal,
housekeeping autophagy that is important for quality control of
proteins and organelles, and starvation- or stress-induced
autophagy. During the last decade evidence has accumulated
that autophagy can be highly selective (Kirkin et al., 2009a; Kraft
et al., 2010; Johansen and Lamark, 2011). Selective autophagy
refers to the selective degradation of, for instance, organelles
3238
Journal of Cell Science 126 (15)
Induction
Nucleation
Expansion
Cargo recruitment
Closure
Maturation
Cargo degradation
Endosome
Lysosome
PI 3-kinase
complex
ULK1/2
?
ATG9
vesicle
Autophagosome
Phagophore
ATG5
ATG12
ATG7
ATG10
Journal of Cell Science
ATG5 ATG12
proLC3z
ATG4B
LC3-I
ATG7
ATG3
ATG5 ATG12
ATG16L1
Key
LC3-II
PE
PtdIns(3)P
DFCP1
Autolysosome
Protein misfolded/aggregate
Ubiquitin
WIPI proteins
Peroxisome
ATG9
Bacteria
ATG12–ATG5–ATG16L1
Mitophagy receptor
LC3-PE
Mitochondria
Autophagy receptor (SLRs)
ATG8-LIR interaction
Fig. 1. Overview of selective autophagy in mammalian cells. Activation of the complex between uncoordinated 51-like kinases 1 and 2 (ULK1–ULK2) and the
scaffold proteins ATG13, FIP200 and ATG101 is essential for the induction of autophagy. At the nucleation step, proteins and lipids are recruited to the
phagophore. ATG9, a multi-spanning transmembrane protein, is located on vesicles that dynamically traffic to and from the phagophore. The class III
phosphatidylinositol 3-kinase (PI 3-kinase) complex, with the catalytic subunit Vps34, the Ser/Thr kinase Vps15 and the regulatory subunits beclin-1 and
ATG14L, generates PtdIns(3)P at the phagophore. PtdIns(3)P is required for the recruitment of WD-repeat proteins that interact with phosphoinositides (WIPIs)
and double-FYVE-containing protein 1 (DFCP1). WIPIs, in turn, recruit ATG2A and ATG2B into a complex, which can communicate with ATG9. Expansion of
the phagophore depends on two ubiquitin-like (Ubl) conjugation systems (boxed). Conjugation of ATG5 to ATG12, which requires the E1 enzyme ATG7 and the
E2 enzyme ATG10, generates an oligomeric complex between the ATG12–ATG5 conjugate and ATG16L1. ATG8/LC3 proteins are subsequently conjugated to
phosphatidylethanolamine (PE) following cleavage by the cysteine protease ATG4 acting on nascent ATG8s (proLC3) to expose a C-terminal glycine residue
required for covalent attachment to PE. The exposed glycine of ATG8 (LC3-I) is activated by ATG7 (E1), activated ATG8 is transferred to ATG3 (E2-like
enzyme) forming an ATG8,ATG3 thioester intermediate, before ATG8 is conjugated to PE by the E3-like ATG12–ATG5–ATG16 complex. The cargo for
selective autophagy is recruited to the inner, concave, surface of the growing phagophore by autophagy receptors that are associated both with the cargo and with
lipidated ATG8/LC3 (LC3 II). The phagophore expands and encloses its cargo to form the double-membrane autophagosome. Fusion of autophagosomes with late
endosomes or lysosomes (maturation) forms autolysosomes where the enclosed cargo is degraded.
(mitophagy and pexophagy), bacteria (xenophagy), ribosomes,
macromolecular structures, specific proteins and protein
aggregates (aggrephagy) by autophagy. The cytoplasm-tovacuole targeting (Cvt) pathway, which selectively directs
aggregated precursors of aminopeptidase 1 and a-mannosidase
to the vacuole, is the only biosynthetic pathway that uses the
autophagy core machinery (Lynch-Day and Klionsky, 2010).
Together with the discovery of bona fide selective autophagy
receptors in mammalian cells, studies of the Cvt pathway have
helped elucidate some of the molecular basis for selective
autophagy.
Emerging selectivity – discovery of selective
autophagy receptors and the LIR motif
A selective autophagy receptor needs to be able to bind
specifically to cargo and to dock onto the forming phagophore
enabling autophagic sequestration and degradation of the cargo.
The first selective autophagy receptor to be identified was p62
[also known as sequestosome-1 (SQSTM1)] (Bjørkøy et al.,
2005; Komatsu et al., 2007; Pankiv et al., 2007). p62 was well
known to act as a scaffold protein in signaling pathways
involving NF-kB (Moscat et al., 2007), but to also accumulate
in ubiquitin-containing protein inclusions in many proteinaggregation diseases including Alzheimer disease, Pick disease,
dementia with Lewy bodies, Parkinson disease and multiple
system atrophy (Kuusisto et al., 2001; Zatloukal et al., 2002). We
found that p62 is both a selective autophagy substrate and a cargo
receptor for autophagic degradation of ubiquitylated protein
aggregates (Bjørkøy et al., 2005; Pankiv et al., 2007).
Consistently, knockout of autophagy in the liver of mice
demonstrated that p62, which binds both ubiquitin and LC3,
regulates the formation of protein aggregates and is removed by
autophagy (Komatsu et al., 2007). The authors showed that
blocking of autophagy resulted in a failure to degrade p62 and
lead to extensive accumulation of protein aggregates, severe
hepatomegaly and liver dysfunction (Komatsu et al., 2007).
The LIR motif in selective autophagy
3239
cargo receptor Atg19 (Pankiv et al., 2007; Ichimura et al., 2008b;
Noda et al., 2008). The motif has also been called Atg8-family
interacting motif (AIM) (Noda et al., 2010). The structures of p62
and Atg19 peptides bound to LC3B and Atg8, respectively,
revealed a common W-x-x-L motif (x5any amino acid)
(Ichimura et al., 2008b; Noda et al., 2008) (Fig. 3) (see also
p62 consists of 440 amino acids and contains an N-terminal
PB1 domain, followed by a ZZ-type zinc-finger domain and a
C-terminally located ubiquitin-binding UBA domain (Fig. 2).
Detailed deletion mapping and point mutation analyses, together
with X-ray crystallography and NMR lead to the elucidation of
the LC3-interacting region (LIR) motifs of p62 and of the Cvt
Sequestosome-1-like receptors (SLRs)
LIR KIR
p62
ZZ
PB1
Ub
UBA
NLS2
440
NLS1 NES
LIR2
NBR1
CC1
ZZ
ZZ
PB1
FW
LIR
NDP52
ZF
OPTN
CC
Ub
CC
Coiled-Coil
CC
CC
LIR
Journal of Cell Science
966
446
LIR
SKICH
Ub
UBA
UBA
Ub
CC
SKICH
TAX1BP1
LIR1
CC2
ZF ZF
789
Ub
CC
CC
UBAN
ZF
577
Mitophagy receptors
LIR
FUNDC1
TM TM
TM
155
LIR
BH3
BNIP3
TM
194
LIR
BH3
NIX
TM
219
LIR
Atg32
TM
529
Specialized receptors
4H
Cbl
EF
SH2
LIR
UBA
Ring
906
LIR
Stbd1
CBM20
TM
358
Cvt cargo receptors
LIR
Atg19
CC
415
ABD
LIR
Atg34
ABD
412
Fig. 2. Domain architecture of selective autophagy cargo receptors known to date. The sequestosome-1-like receptors (SLRs) constitute of p62, NBR1,
NDP52, TAX1BP and OPTN (optineurin) in mammals. The known mitophagy receptors FUNDC1, BNIP3, NIX (BNIP3L) in mammals, and Atg32 in yeast, are
shown. The specialized receptors Cbl and Stbd1, characterized in mammals, are involved in selective autophagy of Src kinase and glycogen, respectively. The Cvt
cargo receptors, Atg19 and Atg34 in yeast, are essential for the Cvt pathway. PB1, Phox and Bem1 domain (dark pink); ZZ, ZZ-type zink finger domain (blue);
CC, coiled-coil domain (light pink); NLS1 and NLS2, nuclear localization signals 1 and 2 (dark gray); NES, nuclear export signal (dark gray); LIR, LC3interacting region (dark red); KIR, Keap interacting region (green); UBA, ubiquitin-associated domain (yellow); FW, four tryptophan domain (dark yellow);
SKICH, SKIP carboxyl homology domain (light green); ZF, Zinc-finger domain (yellow); UBAN, ubiquitin binding in ABIN and NEMO domain (yellow); TM,
transmembrane domain (light blue); BH3, Bcl-2 homology (BH) domain 3 (light purple); 4H, four-helix bundle domain (light gray); EF, EF-hand-fold domain
(light gray); SH2, Src-homology 2 domain (light gray); Ring, really-interesting-new-gene-finger domain (blue); CBM20, family 20 carbohydrate-binding module
domain (light gray); ABD, Ams1-binding domain (orange). The size of the receptors (in numbers of amino acids) is indicated.
3240
Journal of Cell Science 126 (15)
A
p62
Atg19
R28
HP1
W340
L343
E414
D338
HP2
R11
L415
W412
D337
E413
R67
R10
NBR1
LC3B
Atg8
NDP52
K24
K20
V135
E730
K16
Y732
I735
L134
V136
D731
K48
LC3C
GABARAPL1
C
B
4
Bits
2
R11
D338
W340
0
6
D337
Bits
R10
4
2
L343
0
4
Ub-like domain
Bits
Journal of Cell Science
N-terminal arm
2
0
X–3 X–2 X–1W0 X1 X2 L3 X4 X5
Fig. 3. LIR motif consensus and structural determinants of LIR–ATG8 interactions. (A) Surface representation of LC3B bound to the p62-LIR peptide (top
left), yeast Atg8 bound to the Atg19-LIR peptide (top right), GABARAP-L1 bound to the NBR1-LIR peptide (bottom left) and LC3C bound to the NDP52-LIR
peptide (non-canonical LIR-motif) (bottom right). The hydrophobic pockets (HP1 and HP2) of LC3B, Atg8 and GABARAP-L1 as well as the hydrophobic
patch of LC3C are indicated in bright yellow. The amino acids (yellow) of the different LIR peptides that bind in the pockets are shown as well as the amino acids
(red) that interact with basic residues of the ATG8 proteins (blue). (B) Ribbon diagram of LC3B with the N-terminal arm (blue) and the Ubl domain (gray). The
bound p62-LIR peptide is depicted in red. Amino acids D337 and D338 in the p62-LIR peptide interact with the basic residues R10 and R11 in the N-terminal arm
of LC3B. Amino acids W340 and L343 in the p62-LIR peptide binding to hydrophobic pockets in LC3B are also indicated. (C) Sequence logos that are a
graphical representation of amino acid residues as stacks at each position in multiple sequence alignments of LIR motifs. The overall height of the stack indicates
the sequence conservation at that position, whereas the height of symbols within the stack indicates the relative frequency of each amino at that position. The
sequence logos were created on the basis of 42 verified LIR motifs (upper panel) and were split into 22 W-type LIRs (middle panel) and 15 F-type LIRs (lower
panel). The analysis of these 42 LIRs (33 of which are published, see supplementary material Table S1) confirms the core consensus sequence [W/F/Y]xx[L/I/V],
in which alternative letters are placed in square brackets with a solidus between them. Only five LIRs have Tyr (Y) at the aromatic position binding to the HP1
pocket. W-type LIRs prefer Leu (L) in HP2 (13 out of 22). Such a preference is not seen among the 15 F-type LIRs, in which I, L and V are similarly distributed.
F-type LIRs have a significantly higher average number of acidic residues than W-type LIRs. The average number of E, D, S, or T in the three positions Nterminal to the core hydrophobic residue (positions X21 to X23) is 1.7 and 2.5 for W- and F-type LIRs, respectively. The Seq2Logo-1.0 server (http://www.cbs.
dtu.dk/biotools/Seq2Logo-1.0/) was used with Kullback-Leibler logo type and Hobohm1 clustering (threshold 0.63 and 0 weight on prior pseudo counts)
(Thomsen and Nielsen, 2012).
The LIR motif in selective autophagy
Journal of Cell Science
Box 1. Specificities of the LIR-ATG8 protein
interaction
A compilation of verified LIR motifs reveals a core consensus
sequence [W/F/Y]xx[L/I/V] (see Fig. 3C). Most LIR motifs have a
W or an F at the aromatic position binding to the HP1 pocket, but a
few have Y at this position. Structural data show that the Y-type
LIR1 of NBR1 binds in a manner similar to W-type LIRs, but
mutation of the core Y residue into W or F demonstrated that W
results in higher binding affinity than F or Y (Rozenknop et al.,
2011). In addition to the core motif, the importance of an acidic
charge (E, D, S or T), either N- or C-terminal to the conserved
aromatic residue, is evident. The prevalent use of S and T flanking
the core motif indicates a regulation by phosphorylation.
Electrostatic interactions may determine the substrate specificity
since they often involve residues found only in a subset of the
ATG8 homologs. For F-type LIRs, a higher number of electrostatic
interactions appear to compensate for a lower affinity between F
and HP1. The choice of amino acid at position X1 is also more
important for F-type LIRs than for W-type LIRs. The F-type LIRs of
ULK1 and ATG13 have a preference for the GABARAP subfamily.
Mutagenesis of these LIRs showed V (Val), C (Cys), I (Ile), E (Glu)
and F as the only amino acids acceptable in position X1 (Alemu
et al., 2012).
The LC3C-specific LIR of NDP52 represents a more-specialized
variant, because it has lost the aromatic residue and the binding to
the HP1 pocket (see Fig. 3A). Lacking this aromatic residue, it is
unable to bind to most ATG8 proteins but does interact with LC3C
because a rotation of the b-strand of the LIR improves shape
complementarity and creates additional interstrand hydrogen
bonds with the binding cleft in LC3C (von Muhlinen et al., 2012).
LIR-independent interactions involving ATG8 proteins also exist.
For example, C. elegans autophagy receptors do not contain LIR
motifs (Lin et al., 2013), and several of the proteins identified by
Behrends and colleagues interact with ATG8 in a manner that is
not affected by mutations in the LIR docking site (e.g. ATG16L,
ATG7 and ATG5) (Behrends et al., 2010).
Box 1), and the importance of the acidic residues N-terminal to
the core of the DDDWTHL LIR motif of p62 was verified by
alanine substitutions (Pankiv et al., 2007; Ichimura et al., 2008a;
Noda et al., 2008). The LIR motif of p62 presents as an extended
b-strand that forms an intermolecular parallel b-sheet with the b2
strand of LC3B. The ATG8 family proteins have a C-terminal,
‘core’ Ubl domain that contains the conserved ‘ubiquitin fold’
and an additional N-terminal arm with two a-helices that are
closed onto the core Ubl domain (Fig. 3B). The LIR-containing
peptide is located in the interface of the N-terminal arm and the
Ubl domain. In this LIR docking site, two hydrophobic pockets
HP1 and HP2 in the Ubl domain of LC3 accommodate the side
chains of the W and L residues (Ichimura et al., 2008b; Noda
et al., 2008; Noda et al., 2010) (Fig. 3A). The two pockets are
located on the opposite side of the hydrophobic patch (L8-I44V70) of ubiquitin. Electrostatic interactions, which involve two
of the three aspartic acid residues of the LIR motif and basic
residues in the N-terminal arm and Ubl domain of LC3 (R10,
R11, K49 and K50), are also important for the interaction
between p62 and LC3 (Fig. 3A,B). The importance of the basic
residues in the N-terminal arm of LC3B for binding and
autophagic degradation of p62 has been demonstrated by
domain swap experiments (Shvets et al., 2008; Shvets et al.,
2011).
3241
Different strategies have been used to identify proteins that
interact with ATG8 proteins through LIR motifs, including
candidate approaches (Pankiv et al., 2007; Noda et al., 2008;
Sancho et al., 2012), bioinformatics searches (Kraft et al., 2012),
proteomics (Behrends et al., 2010; Pankiv et al., 2010), phage
display (Mohrlüder et al., 2007b) and yeast two-hybrid assays
(Kirkin et al., 2009b; Novak et al., 2010; Wild et al., 2011;
Popovic et al., 2012). LIR motifs have been identified by using
deletion mapping and protein–protein interaction assays, and by
testing deletion and point-mutated constructs. We have found that
peptide array analysis is a specific and efficient method for
identification of LIR motifs (Alemu et al., 2012).
Cargo receptors in selective autophagy
Following discovery of p62 as a selective autophagy receptor, the
related neighbor of BRCA1 gene 1 (NBR1) was found to act as
an aggrephagy receptor (Kirkin et al., 2009b). Subsequently,
nuclear dot protein 52 kDa (NDP52) was found to be an
important xenophagy receptor (Thurston et al., 2009) together
with optineurin (Wild et al., 2011). These and the other
autophagy receptors discussed below use LIR-motif-dependent
interactions to target their cargos for autophagic degradation.
Sequestosome-1-like receptors
In addition to the role of p62, NDP52 and optineurin in selective
autophagy, these proteins have also recently been shown to
regulate innate immunity signaling pathways and, thus, were
suggested to represent a new class of pattern recognition
receptors, the sequestosome-1-like receptors (SLRs) (Deretic,
2012). The SLRs currently consists of p62, NBR1, NDP52,
optineurin and Tax1-binding protein 1 (TAX1BP1) (Fig. 2).
They all contain a dimerization or multimerization domain, a LIR
domain (an atypical LIR motif in the case of NDP52 and
TAX1BP1) and an ubiquitin-binding domain. These three
features of SLRs are required for the efficient execution of
their role as autophagic cargo receptors (Pankiv et al., 2007;
Ichimura et al., 2008b; Itakura and Mizushima, 2011; Deosaran
et al., 2013). Studies of selective autophagy in mammalian cells
and of the Cvt pathway in yeast revealed that the cargo must
either be aggregated or represent a reasonably large structure that
enables the binding of many receptor molecules; alternatively,
the autophagy receptors themselves need to be able to
multimerize the cargo (Lynch-Day and Klionsky, 2010;
Johansen and Lamark, 2011).
The dual nature of SLRs as autophagy receptors and
scaffolding proteins that act in signaling pathways is intriguing.
Since the levels of SLRs are regulated by autophagy, the rate of
autophagy obviously impacts on signaling that involves SLRs. To
discuss these signaling pathways is beyond the scope of this
Commentary, but it is worth noting that accumulation of SLRs
occurs during cellular stresses, including infection and
inflammation, oxidative stress, ER-stress and metabolic stress
(Johansen and Lamark, 2011; Deretic, 2012). This accumulation
may have dramatic effects on stress-related signaling pathways,
but the exact role of autophagy in the control of signal
transduction – beyond its effect on receptor levels – is poorly
understood. One exception is the regulation of the KEAP1–NRF2
oxidative-stress-response pathway, in which p62 binds to and
sequesters KEAP1, leading to its autophagic degradation and the
concomitant induction of NRF2 (Komatsu et al., 2010; Jain et al.,
2010; Taguchi et al., 2012). A positive feedback loop is
Journal of Cell Science
3242
Journal of Cell Science 126 (15)
established in that increased p62 levels activate NRF2, which, in
turn, further increases p62 levels (Jain et al., 2010). Recently, it
has been found that the liver toxicity of accumulated p62 is due
to constitutive upregulation of the NRF2 oxidative-stressresponse pathway (Inami et al., 2011).
One of the striking features of SLRs is their ability to mediate
the selective autophagy of substrates that apparently have no
structural similarities. Substrates targeted by p62 include
ubiquitylated protein aggregates and membrane-embedded
structures, such as intracellular bacteria and peroxisomes. The
single feature that unites the various structures appears to be that
they become ubiquitylated before they are degraded. The LIR
motif is absolutely required for targeting of SLRs and bound
cargoes into the lumen of autophagosomes (Johansen and Lamark,
2011). Selective autophagy depends on a direct interaction
between the LIR motif and ATG8 homologs that are conjugated
to the inner, concave membrane of the phagophore. However, the
LIR motif by itself does not bring a protein to the inner surface of a
phagophore, and the majority of LIR-motif-containing proteins are
not substrates for selective autophagy. Selective autophagy of p62
depends on its PB1-domain-driven polymerization, but for the
delivery of p62-associated cargos, ubiquitin binding and
interactions with other proteins are also important. For the
selective autophagy of protein aggregates, p62 collaborates with
autophagy-linked FYVE protein (ALFY), a nuclear scaffolding
protein that is recruited to cytosolic protein aggregates in a p62dependent manner. ALFY interacts directly with ATG5 and
phosphatidylinositol (3)-phosphate [PtdIns(3)P], and may act as a
scaffold protein that induces the assembly of an autophagycompatible structure (Clausen et al., 2010; Filimonenko et al.,
2010). In flies that lack the ALFY ortholog Blue cheese (Bchs),
accumulation of the p62 ortholog Ref(2)P in ubiquitin-positive
protein aggregates has been observed (Clausen et al., 2010)
suggesting a conserved role for ALFY.
Redundancy and/or collaboration clearly exist between different
SLRs, although this is not very well studied. NBR1 and p62 bind
directly to each other through their PB1 domains, and collaborate
in selective autophagy of misfolded proteins and probably also
midbody rings (Kirkin et al., 2009b; Pohl and Jentsch, 2009; Kuo
et al., 2011). These two proteins also collaborate in pexophagy.
Here, binding and clustering of peroxisomes is mediated by NBR1
in a process that depends on the coincident membrane binding of
its amphipathic J domain and the adjacent UBA domain (Deosaran
et al., 2013). Specialized intracellular pathogens have often
developed strategies to avoid or use autophagy for their own
purposes, but other pathogens are efficiently degraded by selective
autophagy if they are released into the cytoplasm or upon
membrane rupture (Mostowy and Cossart, 2012). Microbes that
are released into the cytosol are ubiquitylated and then recognized
by SLRs (Dupont et al., 2009; Thurston et al., 2009; Zheng et al.,
2009). Membrane remnants associated with exposed microbes can
also be polyubiquitylated and targeted for autophagic degradation
by p62 (Dupont et al., 2009). Furthermore, p62 promotes
autophagic killing of intracellular microbes. Cytoplasmic
precursors of antimicrobial peptides (ubiquitin or ribosomal
precursor proteins) are transported by p62 into autolysosomes or
microbe-containing autolysosomes. Here, the precursors are
converted into peptides that have been shown to kill
Mycobacterium tuberculosis (Ponpuak et al., 2010); and these
peptides might also be potent against other microbes. Viruses can
also act as substrates and p62 has been implicated in xenophagic
elimination of Sindbis virus (Orvedahl et al., 2010). Efficient
xenophagy of Salmonella enterica serotype Typhimurium (S.
typhimurium) is mediated by p62, NDP52 and optineurin, with the
p62-containing microdomains on ubiquitin-coated bacteria
appearing to be physically separated from areas that are
occupied by NDP52 and optineurin (Cemma et al., 2011;
Mostowy et al., 2011; Wild et al., 2011). However, although
NDP52 is recruited to ubiquitin-coated bacteria through its Cterminal Zinc-finger (ZF) domain (Fig. 2), it is initially targeted to
damaged Salmonella-containing vacuoles that are marked by
galectin-8, which binds exposed b-galactoside-containing glycans.
In this way, cytosolic galectin-8 functions as an ubiquitinindependent ‘danger’ receptor and ‘eat-me’ signal (Thurston
et al., 2012). Galectin 8 also detects non-bacteria induced damage
to endosomes or lysosomes, suggesting that membrane rupture is
the initial common event detected during invasion by microbes
(Thurston et al., 2012). The atypical LIR motif in NDP52, termed
CLIR, comprises the tripeptide Leu-Val-Val and binds specifically
to LC3C (Fig. 3A). Efficient recruitment of the other ATG8 family
members to bacteria-degrading autophagosomes depends on both
NDP52 and LC3C (von Muhlinen et al., 2012). TAX1BP1 (T6BP)
is a cargo receptor with homology to NDP52. TAX1BP1 binds
ubiquitin and contains the same atypical LIR motif as NDP52
(Newman et al., 2012) but its role in xenophagy is unknown.
NDP52 is required for degradation of the micro RNA
(miRNA)-processing enzyme DICER, and the main miRNA
effector AGO2 by selective autophagy (Gibbings et al., 2012).
An ubiquitin-independent role of optineurin in aggrephagy has
recently been reported (Korac et al., 2013). It should also be
noted that p62 has a role in mitophagy (Johansen and Lamark,
2011), although this process is primarily mediated by specific
mitochondrial membrane receptors, as discussed below.
Mitophagy receptors
Both yeast and mammalian cells can selectively eliminate
damaged or superfluous mitochondria by mitophagy (reviewed
by Ashrafi and Schwarz, 2013). In yeast, mitophagy is
orchestrated by Atg32, an integral protein of the outer
mitochondrial membrane (OMM) with a N-terminus that faces
the cytosol and C-terminus located in the intermembrane space
(Kanki et al., 2009; Okamoto et al., 2009). Atg32 can interact
with Atg8 indirectly through Atg11 and directly through its LIR
motif in the N-terminal cytosolic domain. Atg32 recruits Atg8
and Atg11 to the mitochondria surface to form an initiator
complex essential for mitophagy (Kondo-Okamoto et al., 2012).
In mammalian cells, three integral OMM proteins that all have a
LIR motif in their cytosolic N-terminal domain are implicated in
mitophagy (Fig. 2). Two homologous BCL2 homology 3 (BH3)only proteins, Bnip3 and Nix (also known as Bnip3L), are able to
induce mitophagy and can also activate cell death (reviewed by
Zhang and Ney, 2009). Bnip3 induces the removal of both
mitochondria and endoplasmic reticulum (Hanna et al., 2012).
Homodimerization of Bnip3 through the transmembrane domain
facilitates the interaction between the LIR motif of Bnip3 and
LC3B. Nix also facilitates LIR-dependent mitophagy (Novak
et al., 2010) (supplementary material Table S1). During erythroid
cell maturation, Nix mediates the complete removal of
mitochondria (Schweers et al., 2007; Sandoval et al., 2008).
Additionally, Nix is involved in depolarization-induced
mitophagy. Nix has a core LIR motif identical to Bnip3.
However, in contrast to Bnip3, Nix does not interact with
The LIR motif in selective autophagy
LC3B but with GABARAP-L1 during mitochondrial stress
(Schwarten et al., 2009; Novak et al., 2010). Hence, residues
flanking the core LIR motif might be involved in determining
specificity. Bnip3 and Nix are both involved in hypoxia-induced
mitophagy (Zhang et al., 2008; Bellot et al., 2009). The third
mitophagy receptor in the OMM is FUNDC1; it acts in hypoxiainduced mitophagy, but with a different mechanism that involves
the dephosphorylation of its LIR motif, which enhances its
binding to LC3B (see below) (Liu et al., 2012).
Journal of Cell Science
Specialized autophagy receptors
So far few autophagy receptors are known to only interact with
one substrate under certain circumstances. Starch-bindingdomain-containing protein 1 (Stbd1) and the E3-ubiquitin
ligase Cbl represent such specialized receptors (Fig. 2). Stbd1
binds glycogen in vitro and is associated with glycogen in cells; it
binds more tightly to abnormal glycogen that is poorly branched
(Jiang et al., 2011). Stbd1 binds to GABARAP-L1 through a LIR
motif and has been proposed to act as an autophagy receptor for
glycogen in a process termed glycophagy (Jiang et al., 2011).
Kinase activity can also be regulated by selective autophagy
that involves interaction with the LIR motif. For instance, Cbl has
been identified as an autophagy receptor for the active, nonreceptor, membrane-associated tyrosine kinase Src (Sandilands
et al., 2012). Increased Src activity promotes tumorigenesis but
excessive Src signaling can be cytotoxic (Yeatman, 2004). When
integrin signaling through the focal adhesion kinase (FAK)–Src
pathway is disrupted in cancer this can lead to excessive and
cytotoxic Src activity. By using its LIR motif to bind LC3B, Cbl
is able to switch the targeting of Src from the proteasome to
autophagic degradation, thereby promoting cancer cell survival
(Sandilands et al., 2012).
Many LIR-containing proteins do not act as
cargo receptors
The presence of functional LIR motifs in components of the core
autophagy machinery demonstrates that LIR-motif-mediated
Cargo recruitment
interactions do not only help targeting cargo receptors to
autophagosomes but are also involved in regulating autophagosome
formation and maturation. In addition to the core autophagy
machinery, several other LIR-motif-containing proteins are
involved in autophagosome formation, transport and maturation
(fusion to lysosomes) (see Fig. 4) (supplementary material Table S1).
LIR-motif-containing proteins in the core autophagy
machinery
The ATG proteins of the core autophagy machinery are involved in
all steps of autophagosome formation (Fig. 1) (Mizushima et al.,
2011). The yeast serine/threonine kinase Atg1 (ULK1 in
mammals) forms a large complex with Atg13 and the Atg17–
Atg31–Atg29 ternary complex. Recently, two independent studies
reported a LIR-motif-dependent interaction between Atg1 and
Atg8 (Kraft et al., 2012; Nakatogawa et al., 2012). Atg1 is present
on autophagosomes in an Atg8-dependent manner before it is
transported to the vacuole for its degradation. Kraft et al. (Kraft
et al., 2012) also showed that Atg13, in complex with Atg1, is
degraded by autophagy. Similarly, Atg1 and Atg13 are degraded
by autophagy during nutrient starvation in Arabidopsis
(Suttangkakul et al., 2011), but the role of the interaction
between their LIR motifs and ATG8 is currently unknown.
Mutations in the LIR motif of Atg1 result in reduced autophagy but
do not influence its functions during initiation of autophagosome
formation (Nakatogawa et al., 2012). This indicates that Atg1 is
also involved in late events of autophagy. Kraft et al. showed that
the Atg1–Atg8 interaction is conserved and maintained in
mammals, by demonstrating that ULK1 associates with
autophagosomes in a LIR-motif-dependent manner (Kraft et al.,
2012). We identified that the same LIR motif in ULK1 is required
for its starvation-induced association with autophagosomes
(Alemu et al., 2012). In contrast to that in Atg1, the LIR motif
of ULK1 does not significantly mediate its degradation. We also
mapped LIR motifs in ULK2, and the ULK complex proteins
ATG13 and FIP200, and demonstrated their binding to ATG8
proteins with a preference for the GABARAP-subfamily (Alemu
Maturation
+
Phagophore
Autolysosome
Autophagosome
Transport
Microtubule
–
Key
ULK1–ULK2 complex
SLRs (p62, NBR1, NDP52, OPTN)
ATG4
LC3-PE (LC3-II)
Ubiquitylated misfolded protein
TBC1D5, TBC1D25
Kinesin
DOR and TP53INP1
Mitophagy receptor
Mitochondria
3243
ATG8–LIR-motif interaction
FYCO1
Fig. 4. Involvement of LIR-ATG8 interaction in
selective autophagy. Selective recruitment of cargo
to the inner membrane of the phagophore is
mediated by interaction between a LIR-motif
containing autophagy receptor and lipidated ATG8
(shown here LC3-PE). Transport of
autophagosomes towards plus ends of microtubules
involves the interaction of the LIR motif of FYCO1
with LC3-PE on the outer autophagosomal
membrane. Maturation of the autophagosome is
dependent on interaction between the LIR motif of
factors involved in the autophagy machinery (e.g.
the ULK1–ULK2 complex or ATG4, or regulatory
factors, such as TBC1D5, TBC1D25, DOR and
TP53INP1) with ATG8 proteins, which then recruit
effector proteins to the outer membrane.
Journal of Cell Science
3244
Journal of Cell Science 126 (15)
et al., 2012). It is possible that LIR-ATG8 interactions of ULK
complex proteins facilitate and/or stabilize tethering of the ULK
complex to the phagophore.
Other members of the core autophagy apparatus, Atg3 in yeast
and ATG4B in mammals, undergo LIR-motif-dependent
interactions with ATG8 that potentially serve a regulatory role
(Satoo et al., 2009; Yamaguchi et al., 2010). Among the four
ATG4 homologs (ATG4A, ATG4B, ATG4C, ATG4D), ATG4B
is the main human ATG4 homolog that efficiently processes
ATG8 precursors and ATG8-PE (Li et al., 2011). The crystal
structure of the human ATG4B–LC3B complex indicates
conformational changes in ATG4B upon binding of the LC3
substrate that facilitate access of LC3 to the catalytic site of
ATG4B (Satoo et al., 2009). Interestingly, in the crystal structure,
the N-terminal LIR motif of ATG4B interacts with a LIR-binding
site on an adjacent (non-substrate) LC3. This interaction with the
LIR motif stabilizes an open conformation of the N-terminal tail
of ATG4B, which presumably favors membrane targeting. Since
ATG4 also mediates deconjugation of ATG8 proteins, a process
that requires membrane targeting, the conformation of the Nterminal tail might, therefore, regulate the deconjugation activity
of ATG4 (Satoo et al., 2009). The yeast E2-like enzyme Atg3
contains a canonical LIR motif (WEDL) that is essential for the
efficient transfer of Atg8 from Atg3 to PE. The Atg3 LIR motif is
required for the Cvt pathway but not for starvation-induced
autophagy. The interaction between the LIR motif of Atg3 and
Atg8 liberates Atg8 from being bound by the LIR motif of Atg19,
thus allowing Atg8–PE conjugation (Yamaguchi et al., 2010).
LIR-containing proteins associated with autophagosomes
and other vesicles
Although some steps of autophagosome formation are well
understood, the membrane origins of autophagosomes are still
debated. Multiple membrane sources were found to be involved,
such as endoplasmic reticulum (ER), mitochondria, ERmitochondria contact sites and plasma membrane (Hamasaki
et al., 2013; Weidberg et al., 2011). The plasma membrane can
contribute directly to the formation of ATG16L1-positive
autophagosome precursors that depend on interactions between
ATG16L1 and the clathrin heavy chain (Ravikumar et al., 2010).
Hence, clathrin-mediated endocytosis might be involved in
regulating the initial stages of autophagosome formation
(Ravikumar et al., 2010). Interestingly, the clathrin heavy chain
also interacts with GABARAP through a LIR motif on a surfaceexposed a-helix in the flexible linker region (Mohrlüder et al.,
2007a). Structural studies show that LIR motifs adopt a bconformation when bound to ATG8-proteins and form an
intermolecular parallel b-sheet (Fig. 3B) (Noda et al., 2010). It
will, therefore, be interesting to learn which conformation the
clathrin LIR has upon binding to GABARAP. Clathrin and
GABARAP are both involved in trafficking of the GABAA
receptor, suggesting that the LIR-mediated interaction has a
physiological relevance. However, it has not been studied whether
this interaction impacts on autophagosome formation. Calreticulin,
which competes with clathrin for binding to GABARAP
(Mohrlüder et al., 2007a), also has a LIR motif very similar to
that of clathrin (supplementary material Table S1) (Mohrlüder
et al., 2007b). Calreticulin is a luminal Ca2+-dependent chaperone
of the ER, but is also involved in variety of cytosolic functions as a
regulator of intracellular Ca2+ homeostasis (Wang et al., 2012). It
is presently not known whether these LIR interactions are relevant
for both autophagosome formation and trafficking of the GABAA
receptor, or for only the latter.
The autophagosome precursor that is generated by clathrindependent endocytosis might represent phagophore precursors.
During and after phagophore formation, proteins are recruited to
the forming autophagosome in a ‘retrieve–recycle’ manner. The
tumor protein 53-induced nuclear protein 2 (TP53INP2; also
known as and, hereafter, referred to as DOR) exits the nucleus in
response to cellular stress or the activation of autophagy (Nowak
et al., 2009; Mauvezin et al., 2010). Cytoplasmic DOR then
localizes to autophagosomes where it interacts with the
transmembrane protein VMP1 (Nowak et al., 2009). On
autophagosomes, DOR interacts with LC3B through its LIR
motif (Sancho et al., 2012), but it does not colocalize with the
autolysosome-associated protein LAMP1, indicating that DOR
localizes only to early autophagosomes (Mauvezin et al., 2010).
Through its interaction with VMP1, DOR presumably acts as a
scaffold protein that recruits ATG8 proteins to the
autophagosome (Nowak et al., 2009). The LIR motif in DOR
overlaps with its nuclear export signal (Sancho et al., 2012).
Hence, mutation of the core LIR residues of DOR blocks its
nuclear exit in response to autophagy activation. Interestingly,
DOR and its homolog TP53INP1 share two highly conserved
regions, including the LIR motif (Sancho et al., 2012). LIRmediated localization of TP53INP1 to autophagosomes induces
autophagy- and caspase-dependent cell death, and it has been
suggested that TP53INP1 displaces p62 from LC3B, which then
promotes cell death (Seillier et al., 2012).
The mechanisms that regulate membrane trafficking in
autophagy are poorly understood. The Rab GTPases (a large
family of monomeric, small GTPases) in their active form are
spatially organized into distinct membrane regions, where they
recruit effectors to regulate intracellular vesicle trafficking events
(Stenmark, 2009). Rab GTPase-activating proteins (GAPs)
negatively regulate the activity of Rab GTPases. The Rab GAP
TBC1D25 is recruited to phagophores and autophagosomes
through direct interactions between its LIR motif and ATG8
homologs, and its GAP activity regulates the fusion between
autophagosomes and lysosomes (Itoh et al., 2011). TBC1D25
inhibits Rab33B, a Golgi-resident Rab (Itoh et al., 2011). Active
Rab33B binds to ATG16L1 and is involved in recruitment of the
ATG12-ATG5-ATG16L complex to preautophagosomal structures
(Itoh et al., 2008). The authors suggest a model whereby TBC1D25
uses ATG8 proteins as scaffolds to regulate autophagosomal
maturation (Itoh et al., 2011).
Recently, 14 of 36 human TBC (Tre2, Bub2, Cdc16)-domaincontaining Rab GAPs were shown to interact with ATG8 proteins
in yeast two-hybrid screens (Popovic et al., 2012). One of these,
TBC1D5 contains two LIR motifs, which are both required for
ATG8 binding and its co-localization with ATG8 to
autophagosomes upon starvation-induced autophagy (Popovic
et al., 2012). TBC1D5 is involved in retrograde traffic from
endosomes to the Golgi. Interestingly, the N-terminal LIR of
TBC1D5 interacts with the Vps29 subunit of the retromer
complex, a recycling endosome sorting complex responsible for
vesicle delivery from early endosomes to the Golgi. The binding of
TBC1D5 to Vps29 can be titrated out by LC3 (Popovic et al.,
2012), indicating that TBC1D5 acts as a molecular switch between
endosomes and autophagy. Furthermore, the C-terminal LIR of
TBC1D5 can tether the endosome and autophagosome, thereby
mediating autophagosome maturation (Popovic et al., 2012).
Journal of Cell Science
The LIR motif in selective autophagy
The examples above demonstrate crosstalk between
endocytosis and autophagy, which then converge in lysosomal
degradation. Rab7 is involved in maturation of both
autophagosomes and endosomes, as well as in the transport of
autophagosomes and endosomes towards lysosomes for
degradation. The Rab7 effector FYVE and coiled-coil-domaincontaining protein 1 (FYCO1) is localized on phagophores,
autophagosomes and late endosomes and, in addition to Rab7,
interacts with LC3 and PtdIns(3)P (Pankiv et al., 2010). FYCO1
binds to LC3 through a LIR motif in the middle of the connecting
loop between its FYVE and GOLD domains. This flexible loop is
predicted to be folded in a way that blocks the interaction
between the FYVE domain and PtdIns(3)P. Binding to LC3B on
autophagic structures releases this inhibition and targets FYCO1
exclusively to PtdIns(3)P-containing membranes that contain
LC3B (Pankiv et al., 2010). FYCO1 couples autophagosomes and
other Rab7-positive vesicles to molecular motors. Depending on
the direction of vesicle movement, FYCO1 is coupled to kinesin
molecular motors. This is further supported by the identification
of a potential kinesin-binding site in FYCO1 (Pankiv et al.,
2010).
Mitogen-activated protein kinase 15 (MAPK15) is another
protein that localizes to autophagosomes through an interaction
between its LIR motif and ATG8 proteins (supplementary material
Table S1) (Colecchia et al., 2012). The kinase activity of MAPK15
is known to affect the rate of both basal and starvation-induced
autophagy (Colecchia et al., 2012) but the substrates of MAPK15
that are involved in autophagy are unknown.
LIR-containing signaling proteins that act as substrates for
selective autophagy
One signaling pathway regulated by autophagy is Wnt signaling;
autophagy enhances the degradation of Dishevelled2 (Dvl2), a
transducer of the Wnt pathway and, thus, negatively affects Wnt
signaling (Gao et al., 2010). The C-terminal DEP domain of Dvl2
contains a LIR motif that binds to ATG8 proteins (Gao et al., 2010;
Zhang et al., 2011). The N-terminal DIX domain mediates the selfoligomerization of Dvl2 that is necessary for its ubiquitylation, and
facilitates its binding to LC3B and GABARAP (Gao et al., 2010).
Ubiquitylation of Dvl2 is enhanced during starvation, and is
essential for its interaction with p62 and subsequent targeting to
autophagosomes. Thus, p62 mediates the indirect association of
Dvl2 with LC3B and GABARAP (Gao et al., 2010). This is also
likely to be the case for the interaction of Dvl2 with GABARAPL1 (Zhang et al., 2011). Consequently, Dvl2 is degraded by
autophagy through its LIR-dependent interaction with ATG8
proteins and by means of the autophagy receptor p62. Very
recently, it was shown that b-catenin is selectively degraded by
autophagy during nutrient deprivation via the formation of a bcatenin-LC3 complex depending on a LIR motif in b-catenin. A
regulatory feedback mechanism is at work, in which active Wnt/bcatenin signalling represses autophagy and p62 expression, while
b-catenin is itself targeted for autophagic clearance in
autolysosomes upon autophagy induction (Petherick et al., 2013).
Regulation of the interaction between LIR and
ATG8 through phosphorylation
Since 25% of the known LIR motifs harbour an S or T residue as the
‘any amino acid residue’ at position –1 immediately N-terminal to
the aromatic residue of the LIR motif (supplementary material
Table S1), it is conceivable that binding affinity of LIR motifs is
3245
regulated through phosphorylation. Indeed, NDP52 recruits TANKbinding kinase 1 (TBK1) to the bacterial surface (Thurston et al.,
2009). Optineurin also recruits TBK1 to ubiquitylated Salmonella,
resulting in the subsequent phosphorylation of optineurin at S177
located at the ‘any amino acid residue’ at position –1, which
strongly enhances the binding to LC3B (Wild et al., 2011).
Optineurin and NDP52 occupy the same microdomains on the
bacteria. Thus, NDP52-bound TBK1 can also phosphorylate the
LIR of optineurin, thereby enhancing the response.
The phosphorylation state of the LIR motif of Bnip3 has been
shown to determine whether it executes pro-survival mitophagy
or apoptosis. Phosphorylation of S17 and S24 that flank the LIR
increases binding of Bnip3 to LC3B and GABARAP-L2, and
induces mitophagy. When its LIR motif is unphosphorylated,
Bnip3 functions as a BH3-only protein and promotes apoptosis
(Zhu et al., 2013). Furthermore, the interaction of FUNDC1 with
LC3B is enhanced during hypoxia through the dephosphorylation
of the tyrosine residue (Y18 binding to HP1) in its LIR, which
facilitates mitophagy (Liu et al., 2012).
Not only the LIR motifs but also the binding surface of ATG8
proteins may be phosphorylated to regulate binding of LIR-motifcontaining proteins. For instance, phosphorylation of S12 in the Nterminal arm of rat LC3B by protein kinase A (PKA) negatively
affects autophagy (Cherra et al., 2010). This residue is adjacent to
the two Arg residues (R10 and R11) that bind to two aspartic acid
residues (bold) in the DDDWTHL LIR motif of p62, but it has not
been studied whether phosphorylation of S12 affects the docking
of p62. Further studies are required to thoroughly address the
question to which extent LIR motifs and LIR docking sites are
regulated by posttranslational modifications.
Concluding remarks
The interaction between LIR motifs and ATG8 proteins is crucial
for the recruitment of cargo to the inner surface of the
phagophore, and for the recruitment of effector proteins to the
outer autophagosomal membrane where these effectors mediate
transport and maturation of autophagosomes (Fig. 4). The
characterization of LIR-motif-containing proteins and the
elucidation of their roles in autophagy are still at an early
stage. Additional examples on how LIR–ATG8 interactions are
regulated through phosphorylation or other post-translational
modifications are clearly anticipated in future studies. It will be
interesting to see whether an interaction of LIR motifs with
ATG8 proteins is also involved in processes other than
autophagy. The binding of TBC1D5 to Vps29 through its Nterminal LIR motif suggests that there are binding partners for
LIR-motifs other than ATG8 proteins. Finally, it will also be
interesting to investigate whether these LIR–ATG8 interactions
can be explored as druggable targets.
Acknowledgements
We thank members of our group for critical reading of the
manuscript, and Steingrim Svenning for help with Fig. 3.
Funding
This work was funded in part by grants from the FUGE and FRIBIO
programs of the Norwegian Research Council, the Norwegian
Cancer Society and the Blix foundation to T.J.
Supplementary material available online at
http://jcs.biologists.org/lookup/suppl/doi:10.1242/jcs.126128/-/DC1
3246
Journal of Cell Science 126 (15)
Journal of Cell Science
References
Alemu, E. A., Lamark, T., Torgersen, K. M., Birgisdottir, A. B., Larsen, K. B., Jain,
A., Olsvik, H., Øvervatn, A., Kirkin, V. and Johansen, T. (2012). ATG8 family
proteins act as scaffolds for assembly of the ULK complex: sequence requirements for
LC3-interacting region (LIR) motifs. J. Biol. Chem. 287, 39275-39290.
Arias, E. and Cuervo, A. M. (2011). Chaperone-mediated autophagy in protein quality
control. Curr. Opin. Cell Biol. 23, 184-189.
Ashrafi, G. and Schwarz, T. L. (2013). The pathways of mitophagy for quality control
and clearance of mitochondria. Cell Death Differ. 20, 31-42.
Behrends, C., Sowa, M. E., Gygi, S. P. and Harper, J. W. (2010). Network
organization of the human autophagy system. Nature 466, 68-76.
Bellot, G., Garcia-Medina, R., Gounon, P., Chiche, J., Roux, D., Pouysségur, J. and
Mazure, N. M. (2009). Hypoxia-induced autophagy is mediated through hypoxiainducible factor induction of BNIP3 and BNIP3L via their BH3 domains. Mol. Cell.
Biol. 29, 2570-2581.
Bjørkøy, G., Lamark, T., Brech, A., Outzen, H., Perander, M., Øvervatn, A.,
Stenmark, H. and Johansen, T. (2005). p62/SQSTM1 forms protein aggregates
degraded by autophagy and has a protective effect on huntingtin-induced cell death.
J. Cell Biol. 171, 603-614.
Cemma, M., Kim, P. K. and Brumell, J. H. (2011). The ubiquitin-binding adaptor
proteins p62/SQSTM1 and NDP52 are recruited independently to bacteria-associated
microdomains to target Salmonella to the autophagy pathway. Autophagy 7, 341-345.
Cherra, S. J., 3rd, Kulich, S. M., Uechi, G., Balasubramani, M., Mountzouris, J.,
Day, B. W. and Chu, C. T. (2010). Regulation of the autophagy protein LC3 by
phosphorylation. J. Cell Biol. 190, 533-539.
Clausen, T. H., Lamark, T., Isakson, P., Finley, K., Larsen, K. B., Brech, A.,
Øvervatn, A., Stenmark, H., Bjørkøy, G., Simonsen, A. et al. (2010). p62/
SQSTM1 and ALFY interact to facilitate the formation of p62 bodies/ALIS and their
degradation by autophagy. Autophagy 6, 330-344.
Colecchia, D., Strambi, A., Sanzone, S., Iavarone, C., Rossi, M., Dall’Armi, C.,
Piccioni, F., Verrotti di Pianella, A. and Chiariello, M. (2012). MAPK15/ERK8
stimulates autophagy by interacting with LC3 and GABARAP proteins. Autophagy 8,
1724-1740.
Deosaran, E., Larsen, K. B., Hua, R., Sargent, G., Wang, Y., Kim, S., Lamark, T.,
Jauregui, M., Law, K., Lippincott-Schwartz, J. et al. (2013). NBR1 acts as an
autophagy receptor for peroxisomes. J. Cell Sci. 126, 939-952.
Deretic, V. (2012). Autophagy as an innate immunity paradigm: expanding the scope
and repertoire of pattern recognition receptors. Curr. Opin. Immunol. 24, 21-31.
Dupont, N., Lacas-Gervais, S., Bertout, J., Paz, I., Freche, B., Van Nhieu, G. T., van
der Goot, F. G., Sansonetti, P. J. and Lafont, F. (2009). Shigella phagocytic
vacuolar membrane remnants participate in the cellular response to pathogen invasion
and are regulated by autophagy. Cell Host Microbe 6, 137-149.
Filimonenko, M., Isakson, P., Finley, K. D., Anderson, M., Jeong, H., Melia, T. J.,
Bartlett, B. J., Myers, K. M., Birkeland, H. C., Lamark, T. et al. (2010). The
selective macroautophagic degradation of aggregated proteins requires the PI3Pbinding protein Alfy. Mol. Cell 38, 265-279.
Gao, C., Cao, W., Bao, L., Zuo, W., Xie, G., Cai, T., Fu, W., Zhang, J., Wu, W.,
Zhang, X. et al. (2010). Autophagy negatively regulates Wnt signalling by promoting
Dishevelled degradation. Nat. Cell Biol. 12, 781-790.
Gibbings, D., Mostowy, S., Jay, F., Schwab, Y., Cossart, P. and Voinnet, O. (2012).
Selective autophagy degrades DICER and AGO2 and regulates miRNA activity. Nat.
Cell Biol. 14, 1314-1321.
Hain, A. U., Weltzer, R. R., Hammond, H., Jayabalasingham, B., Dinglasan, R. R.,
Graham, D. R., Colquhoun, D. R., Coppens, I. and Bosch, J. (2012). Structural
characterization and inhibition of the Plasmodium Atg8-Atg3 interaction. J. Struct.
Biol. 180, 551-562.
Hamasaki, M., Shibutani, S. T. and Yoshimori, T. (2013). Up-to-date membrane
biogenesis in the autophagosome formation. Curr. Opin Cell Biol. 25, doi: 10.1016/
j.ceb.2013.03.004.
Hanna, R. A., Quinsay, M. N., Orogo, A. M., Giang, K., Rikka, S. and Gustafsson,
A. B. (2012). Microtubule-associated protein 1 light chain 3 (LC3) interacts with
Bnip3 protein to selectively remove endoplasmic reticulum and mitochondria via
autophagy. J. Biol. Chem. 287, 19094-19104.
Ichimura, Y., Kominami, E., Tanaka, K. and Komatsu, M. (2008a). Selective
turnover of p62/A170/SQSTM1 by autophagy. Autophagy 4, 1063-1066.
Ichimura, Y., Kumanomidou, T., Sou, Y. S., Mizushima, T., Ezaki, J., Ueno, T.,
Kominami, E., Yamane, T., Tanaka, K. and Komatsu, M. (2008b). Structural basis
for sorting mechanism of p62 in selective autophagy. J. Biol. Chem. 283, 2284722857.
Inami, Y., Waguri, S., Sakamoto, A., Kouno, T., Nakada, K., Hino, O., Watanabe,
S., Ando, J., Iwadate, M., Yamamoto, M. et al. (2011). Persistent activation of Nrf2
through p62 in hepatocellular carcinoma cells. J. Cell Biol. 193, 275-284.
Itakura, E. and Mizushima, N. (2011). p62 Targeting to the autophagosome formation
site requires self-oligomerization but not LC3 binding. J. Cell Biol. 192, 17-27.
Itoh, T., Fujita, N., Kanno, E., Yamamoto, A., Yoshimori, T. and Fukuda,
M. (2008). Golgi-resident small GTPase Rab33B interacts with Atg16L and
modulates autophagosome formation. Mol. Biol. Cell 19, 2916-2925.
Itoh, T., Kanno, E., Uemura, T., Waguri, S. and Fukuda, M. (2011). OATL1, a novel
autophagosome-resident Rab33B-GAP, regulates autophagosomal maturation. J. Cell
Biol. 192, 839-853.
Jain, A., Lamark, T., Sjøttem, E., Larsen, K. B., Awuh, J. A., Øvervatn, A.,
McMahon, M., Hayes, J. D. and Johansen, T. (2010). p62/SQSTM1 is a target gene
for transcription factor NRF2 and creates a positive feedback loop by inducing
antioxidant response element-driven gene transcription. J. Biol. Chem. 285, 2257622591.
Jiang, S., Wells, C. D. and Roach, P. J. (2011). Starch-binding domain-containing
protein 1 (Stbd1) and glycogen metabolism: Identification of the Atg8 family
interacting motif (AIM) in Stbd1 required for interaction with GABARAPL1.
Biochem. Biophys. Res. Commun. 413, 420-425.
Johansen, T. and Lamark, T. (2011). Selective autophagy mediated by autophagic
adapter proteins. Autophagy 7, 279-296.
Kanki, T., Wang, K., Cao, Y., Baba, M. and Klionsky, D. J. (2009). Atg32 is a
mitochondrial protein that confers selectivity during mitophagy. Dev. Cell 17, 98-109.
Kirkin, V., McEwan, D. G., Novak, I. and Dikic, I. (2009a). A role for ubiquitin in
selective autophagy. Mol. Cell 34, 259-269.
Kirkin, V., Lamark, T., Sou, Y. S., Bjørkøy, G., Nunn, J. L., Bruun, J. A., Shvets, E.,
McEwan, D. G., Clausen, T. H., Wild, P. et al. (2009b). A role for NBR1 in
autophagosomal degradation of ubiquitinated substrates. Mol. Cell 33, 505-516.
Komatsu, M., Waguri, S., Koike, M., Sou, Y. S., Ueno, T., Hara, T., Mizushima, N.,
Iwata, J., Ezaki, J., Murata, S. et al. (2007). Homeostatic levels of p62 control
cytoplasmic inclusion body formation in autophagy-deficient mice. Cell 131, 11491163.
Komatsu, M., Kurokawa, H., Waguri, S., Taguchi, K., Kobayashi, A., Ichimura, Y.,
Sou, Y. S., Ueno, I., Sakamoto, A., Tong, K. I. et al. (2010). The selective
autophagy substrate p62 activates the stress responsive transcription factor Nrf2
through inactivation of Keap1. Nat. Cell Biol. 12, 213-223.
Kondo-Okamoto, N., Noda, N. N., Suzuki, S. W., Nakatogawa, H., Takahashi, I.,
Matsunami, M., Hashimoto, A., Inagaki, F., Ohsumi, Y. and Okamoto, K. (2012).
Autophagy-related protein 32 acts as autophagic degron and directly initiates
mitophagy. J. Biol. Chem. 287, 10631-10638.
Korac, J., Schaeffer, V., Kovacevic, I., Clement, A. M., Jungblut, B., Behl, C.,
Terzic, J. and Dikic, I. (2013). Ubiquitin-independent function of optineurin in
autophagic clearance of protein aggregates. J. Cell Sci. 126, 580-592.
Kraft, C., Peter, M. and Hofmann, K. (2010). Selective autophagy: ubiquitin-mediated
recognition and beyond. Nat. Cell Biol. 12, 836-841.
Kraft, C., Kijanska, M., Kalie, E., Siergiejuk, E., Lee, S. S., Semplicio, G., Stoffel, I.,
Brezovich, A., Verma, M., Hansmann, I. et al. (2012). Binding of the Atg1/ULK1
kinase to the ubiquitin-like protein Atg8 regulates autophagy. EMBO J. 31, 36913703.
Kuo, T. C., Chen, C. T., Baron, D., Onder, T. T., Loewer, S., Almeida, S.,
Weismann, C. M., Xu, P., Houghton, J. M., Gao, F. B. et al. (2011). Midbody
accumulation through evasion of autophagy contributes to cellular reprogramming
and tumorigenicity. Nat. Cell Biol. 13, 1214-1223.
Kuusisto, E., Salminen, A. and Alafuzoff, I. (2001). Ubiquitin-binding protein p62 is
present in neuronal and glial inclusions in human tauopathies and synucleinopathies.
Neuroreport 12, 2085-2090.
Levine, B. and Kroemer, G. (2008). Autophagy in the pathogenesis of disease.
Cell 132, 27-42.
Levine, B., Mizushima, N. and Virgin, H. W. (2011). Autophagy in immunity and
inflammation. Nature 469, 323-335.
Li, M., Hou, Y., Wang, J., Chen, X., Shao, Z. M. and Yin, X. M. (2011). Kinetics
comparisons of mammalian Atg4 homologues indicate selective preferences toward
diverse Atg8 substrates. J. Biol. Chem. 286, 7327-7338.
Lin, L., Yang, P., Huang, X., Zhang, H., Lu, Q. and Zhang, H. (2013). The scaffold
protein EPG-7 links cargo-receptor complexes with the autophagic assembly
machinery. J. Cell Biol. 201, 113-129.
Liu, L., Feng, D., Chen, G., Chen, M., Zheng, Q., Song, P., Ma, Q., Zhu, C., Wang,
R., Qi, W. et al. (2012). Mitochondrial outer-membrane protein FUNDC1 mediates
hypoxia-induced mitophagy in mammalian cells. Nat. Cell Biol. 14, 177-185.
Lynch-Day, M. A. and Klionsky, D. J. (2010). The Cvt pathway as a model for
selective autophagy. FEBS Lett. 584, 1359-1366.
Mauvezin, C., Orpinell, M., Francis, V. A., Mansilla, F., Duran, J., Ribas, V.,
Palacı́n, M., Boya, P., Teleman, A. A. and Zorzano, A. (2010). The nuclear cofactor
DOR regulates autophagy in mammalian and Drosophila cells. EMBO Rep. 11, 37-44.
Mijaljica, D., Prescott, M. and Devenish, R. J. (2011). Microautophagy in mammalian
cells: revisiting a 40-year-old conundrum. Autophagy 7, 673-682.
Mizushima, N. and Komatsu, M. (2011). Autophagy: renovation of cells and tissues.
Cell 147, 728-741.
Mizushima, N., Yoshimori, T. and Ohsumi, Y. (2011). The role of Atg proteins in
autophagosome formation. Annu. Rev. Cell Dev. Biol. 27, 107-132.
Mohrlüder, J., Hoffmann, Y., Stangler, T., Hänel, K. and Willbold, D. (2007a).
Identification of clathrin heavy chain as a direct interaction partner for the gammaaminobutyric acid type A receptor associated protein. Biochemistry 46, 14537-14543.
Mohrlüder, J., Stangler, T., Hoffmann, Y., Wiesehan, K., Mataruga, A. and
Willbold, D. (2007b). Identification of calreticulin as a ligand of GABARAP by
phage display screening of a peptide library. FEBS J. 274, 5543-5555.
Moscat, J., Diaz-Meco, M. T. and Wooten, M. W. (2007). Signal integration and
diversification through the p62 scaffold protein. Trends Biochem. Sci. 32, 95-100.
Mostowy, S. and Cossart, P. (2012). Bacterial autophagy: restriction or promotion of
bacterial replication? Trends Cell Biol. 22, 283-291.
Mostowy, S., Sancho-Shimizu, V., Hamon, M. A., Simeone, R., Brosch, R.,
Johansen, T. and Cossart, P. (2011). p62 and NDP52 proteins target intracytosolic
Shigella and Listeria to different autophagy pathways. J. Biol. Chem. 286, 2698726995.
Journal of Cell Science
The LIR motif in selective autophagy
Nakatogawa, H., Suzuki, K., Kamada, Y. and Ohsumi, Y. (2009). Dynamics and
diversity in autophagy mechanisms: lessons from yeast. Nat. Rev. Mol. Cell Biol. 10,
458-467.
Nakatogawa, H., Ohbayashi, S., Sakoh-Nakatogawa, M., Kakuta, S., Suzuki, S. W.,
Kirisako, H., Kondo-Kakuta, C., Noda, N. N., Yamamoto, H. and Ohsumi,
Y. (2012). The autophagy-related protein kinase Atg1 interacts with the ubiquitin-like
protein Atg8 via the Atg8 family interacting motif to facilitate autophagosome
formation. J. Biol. Chem. 287, 28503-28507.
Newman, A. C., Scholefield, C. L., Kemp, A. J., Newman, M., McIver, E. G., Kamal,
A. and Wilkinson, S. (2012). TBK1 kinase addiction in lung cancer cells is mediated
via autophagy of Tax1bp1/Ndp52 and non-canonical NF-kB signalling. PLoS ONE 7,
e50672.
Noda, N. N., Kumeta, H., Nakatogawa, H., Satoo, K., Adachi, W., Ishii, J., Fujioka,
Y., Ohsumi, Y. and Inagaki, F. (2008). Structural basis of target recognition by
Atg8/LC3 during selective autophagy. Genes Cells 13, 1211-1218.
Noda, N. N., Ohsumi, Y. and Inagaki, F. (2010). Atg8-family interacting motif crucial
for selective autophagy. FEBS Lett. 584, 1379-1385.
Novak, I., Kirkin, V., McEwan, D. G., Zhang, J., Wild, P., Rozenknop, A., Rogov,
V., Löhr, F., Popovic, D., Occhipinti, A. et al. (2010). Nix is a selective autophagy
receptor for mitochondrial clearance. EMBO Rep. 11, 45-51.
Nowak, J., Archange, C., Tardivel-Lacombe, J., Pontarotti, P., Pébusque, M. J.,
Vaccaro, M. I., Velasco, G., Dagorn, J. C. and Iovanna, J. L. (2009). The
TP53INP2 protein is required for autophagy in mammalian cells. Mol. Biol. Cell 20,
870-881.
Okamoto, K., Kondo-Okamoto, N. and Ohsumi, Y. (2009). Mitochondria-anchored
receptor Atg32 mediates degradation of mitochondria via selective autophagy. Dev.
Cell 17, 87-97.
Orvedahl, A., MacPherson, S., Sumpter, R., Jr, Tallóczy, Z., Zou, Z. and Levine,
B. (2010). Autophagy protects against Sindbis virus infection of the central nervous
system. Cell Host Microbe 7, 115-127.
Pankiv, S., Clausen, T. H., Lamark, T., Brech, A., Bruun, J. A., Outzen, H.,
Øvervatn, A., Bjørkøy, G. and Johansen, T. (2007). p62/SQSTM1 binds directly to
Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy.
J. Biol. Chem. 282, 24131-24145.
Pankiv, S., Alemu, E. A., Brech, A., Bruun, J. A., Lamark, T., Overvatn, A.,
Bjørkøy, G. and Johansen, T. (2010). FYCO1 is a Rab7 effector that binds to LC3
and PI3P to mediate microtubule plus end-directed vesicle transport. J. Cell Biol. 188,
253-269.
Petherick, K. J., Williams, A. C., Lane, J. D., Ordonez-Moran, P., Huelsken, J.,
Collard, T. J., Smart, H. J., Batson, J., Malik, K., Paraskeva, C., et al, (2013).
Autolysosomal beta-catenin degradation regulates Wnt-autophagy-p62 crosstalk.
EMBO J.
Pohl, C. and Jentsch, S. (2009). Midbody ring disposal by autophagy is a postabscission event of cytokinesis. Nat. Cell Biol. 11, 65-70.
Ponpuak, M., Davis, A. S., Roberts, E. A., Delgado, M. A., Dinkins, C., Zhao, Z.,
Virgin, H. W., 4th, Kyei, G. B., Johansen, T., Vergne, I. et al. (2010). Delivery of
cytosolic components by autophagic adaptor protein p62 endows autophagosomes
with unique antimicrobial properties. Immunity 32, 329-341.
Popovic, D., Akutsu, M., Novak, I., Harper, J. W., Behrends, C. and Dikic, I. (2012).
Rab GTPase-activating proteins in autophagy: regulation of endocytic and autophagy
pathways by direct binding to human ATG8 modifiers. Mol. Cell. Biol. 32, 17331744.
Ravikumar, B., Moreau, K., Jahreiss, L., Puri, C. and Rubinsztein, D. C. (2010).
Plasma membrane contributes to the formation of pre-autophagosomal structures.
Nat. Cell Biol. 12, 747-757.
Rozenknop, A., Rogov, V. V., Rogova, N. Y., Löhr, F., Güntert, P., Dikic, I. and
Dötsch, V. (2011). Characterization of the interaction of GABARAPL-1 with the LIR
motif of NBR1. J. Mol. Biol. 410, 477-487.
Sancho, A., Duran, J., Garcı́a-España, A., Mauvezin, C., Alemu, E. A., Lamark, T.,
Macias, M. J., DeSalle, R., Royo, M., Sala, D. et al. (2012). DOR/Tp53inp2 and
Tp53inp1 constitute a metazoan gene family encoding dual regulators of autophagy
and transcription. PLoS ONE 7, e34034.
Sandilands, E., Serrels, B., McEwan, D. G., Morton, J. P., Macagno, J. P., McLeod,
K., Stevens, C., Brunton, V. G., Langdon, W. Y., Vidal, M. et al. (2012).
Autophagic targeting of Src promotes cancer cell survival following reduced FAK
signalling. Nat. Cell Biol. 14, 51-60.
Sandoval, H., Thiagarajan, P., Dasgupta, S. K., Schumacher, A., Prchal, J. T.,
Chen, M. and Wang, J. (2008). Essential role for Nix in autophagic maturation of
erythroid cells. Nature 454, 232-235.
Satoo, K., Noda, N. N., Kumeta, H., Fujioka, Y., Mizushima, N., Ohsumi, Y. and
Inagaki, F. (2009). The structure of Atg4B-LC3 complex reveals the mechanism of
LC3 processing and delipidation during autophagy. EMBO J. 28, 1341-1350.
Schwarten, M., Mohrlüder, J., Ma, P., Stoldt, M., Thielmann, Y., Stangler, T.,
Hersch, N., Hoffmann, B., Merkel, R. and Willbold, D. (2009). Nix directly binds
to GABARAP: a possible crosstalk between apoptosis and autophagy. Autophagy 5,
690-698.
3247
Schweers, R. L., Zhang, J., Randall, M. S., Loyd, M. R., Li, W., Dorsey, F. C.,
Kundu, M., Opferman, J. T., Cleveland, J. L., Miller, J. L. et al. (2007). NIX is
required for programmed mitochondrial clearance during reticulocyte maturation.
Proc. Natl. Acad. Sci. USA 104, 19500-19505.
Seillier, M., Peuget, S., Gayet, O., Gauthier, C., N’Guessan, P., Monte, M., Carrier,
A., Iovanna, J. L. and Dusetti, N. J. (2012). TP53INP1, a tumor suppressor, interacts
with LC3 and ATG8-family proteins through the LC3-interacting region (LIR) and
promotes autophagy-dependent cell death. Cell Death Differ. 19, 1525-1535.
Shpilka, T., Weidberg, H., Pietrokovski, S. and Elazar, Z. (2011). Atg8: an
autophagy-related ubiquitin-like protein family. Genome Biol. 12, 226.
Shvets, E., Fass, E., Scherz-Shouval, R. and Elazar, Z. (2008). The N-terminus and
Phe52 residue of LC3 recruit p62/SQSTM1 into autophagosomes. J. Cell Sci. 121,
2685-2695.
Shvets, E., Abada, A., Weidberg, H. and Elazar, Z. (2011). Dissecting the
involvement of LC3B and GATE-16 in p62 recruitment into autophagosomes.
Autophagy 7, 683-688.
Stenmark, H. (2009). Rab GTPases as coordinators of vesicle traffic. Nat. Rev. Mol.
Cell Biol. 10, 513-525.
Suttangkakul, A., Li, F., Chung, T. and Vierstra, R. D. (2011). The ATG1/ATG13
protein kinase complex is both a regulator and a target of autophagic recycling in
Arabidopsis. Plant Cell 23, 3761-3779.
Suzuki, K., Kondo, C., Morimoto, M. and Ohsumi, Y. (2010). Selective transport of
alpha-mannosidase by autophagic pathways: identification of a novel receptor,
Atg34p. J. Biol. Chem. 285, 30019-30025.
Svenning, S., Lamark, T., Krause, K. and Johansen, T. (2011). Plant NBR1 is a
selective autophagy substrate and a functional hybrid of the mammalian autophagic
adapters NBR1 and p62/SQSTM1. Autophagy 7, 993-1010.
Taguchi, K., Fujikawa, N., Komatsu, M., Ishii, T., Unno, M., Akaike, T., Motohashi,
H. and Yamamoto, M. (2012). Keap1 degradation by autophagy for the maintenance
of redox homeostasis. Proc. Natl. Acad. Sci. USA 109, 13561-13566.
Thomsen, M. C. and Nielsen, M. (2012). Seq2Logo: a method for construction and
visualization of amino acid binding motifs and sequence profiles including sequence
weighting, pseudo counts and two-sided representation of amino acid enrichment and
depletion. Nucleic Acids Res. 40, W281-W287.
Thurston, T. L., Ryzhakov, G., Bloor, S., von Muhlinen, N. and Randow, F. (2009).
The TBK1 adaptor and autophagy receptor NDP52 restricts the proliferation of
ubiquitin-coated bacteria. Nat. Immunol. 10, 1215-1221.
Thurston, T. L., Wandel, M. P., von Muhlinen, N., Foeglein, A. and Randow,
F. (2012). Galectin 8 targets damaged vesicles for autophagy to defend cells against
bacterial invasion. Nature 482, 414-418.
von Muhlinen, N., Akutsu, M., Ravenhill, B. J., Foeglein, A., Bloor, S., Rutherford,
T. J., Freund, S. M., Komander, D. and Randow, F. (2012). LC3C, bound
selectively by a noncanonical LIR motif in NDP52, is required for antibacterial
autophagy. Mol. Cell 48, 329-342.
Wang, W. A., Groenendyk, J. and Michalak, M. (2012). Calreticulin signaling in
health and disease. Int. J. Biochem. Cell Biol. 44, 842-846.
Weidberg, H., Shvets, E. and Elazar, Z. (2011). Biogenesis and cargo selectivity of
autophagosomes. Annu. Rev. Biochem. 80, 125-156.
Wild, P., Farhan, H., McEwan, D. G., Wagner, S., Rogov, V. V., Brady, N. R.,
Richter, B., Korac, J., Waidmann, O., Choudhary, C. et al. (2011).
Phosphorylation of the autophagy receptor optineurin restricts Salmonella growth.
Science 333, 228-233.
Yamaguchi, M., Noda, N. N., Nakatogawa, H., Kumeta, H., Ohsumi, Y. and Inagaki,
F. (2010). Autophagy-related protein 8 (Atg8) family interacting motif in Atg3
mediates the Atg3-Atg8 interaction and is crucial for the cytoplasm-to-vacuole
targeting pathway. J. Biol. Chem. 285, 29599-29607.
Yeatman, T. J. (2004). A renaissance for SRC. Nat. Rev. Cancer 4, 470-480.
Zatloukal, K., Stumptner, C., Fuchsbichler, A., Heid, H., Schnoelzer, M., Kenner,
L., Kleinert, R., Prinz, M., Aguzzi, A. and Denk, H. (2002). p62 Is a common
component of cytoplasmic inclusions in protein aggregation diseases. Am. J. Pathol.
160, 255-263.
Zhang, J. and Ney, P. A. (2009). Role of BNIP3 and NIX in cell death, autophagy, and
mitophagy. Cell Death Differ. 16, 939-946.
Zhang, H., Bosch-Marce, M., Shimoda, L. A., Tan, Y. S., Baek, J. H., Wesley, J. B.,
Gonzalez, F. J. and Semenza, G. L. (2008). Mitochondrial autophagy is an HIF-1dependent adaptive metabolic response to hypoxia. J. Biol. Chem. 283, 10892-10903.
Zhang, Y., Wang, F., Han, L., Wu, Y., Li, S., Yang, X., Wang, Y., Ren, F., Zhai, Y.,
Wang, D. et al. (2011). GABARAPL1 negatively regulates Wnt/b-catenin signaling
by mediating Dvl2 degradation through the autophagy pathway. Cell Physiol
Biochem. 27, 503-512.
Zheng, Y. T., Shahnazari, S., Brech, A., Lamark, T., Johansen, T. and Brumell,
J. H. (2009). The adaptor protein p62/SQSTM1 targets invading bacteria to the
autophagy pathway. J. Immunol. 183, 5909-5916.
Zhu, Y., Massen, S., Terenzio, M., Lang, V., Chen-Lindner, S., Eils, R., Novak, I.,
Dikic, I., Hamacher-Brady, A. and Brady, N. R. (2013). Modulation of serines 17
and 24 in the LC3-interacting region of Bnip3 determines pro-survival mitophagy
versus apoptosis. J. Biol. Chem. 288, 1099-1113.
Table 1. Known LIR-containing proteins and details of their LIR motifs and interaction partners
LIR protein
LIR motif
LIRpos
ATG8 ortholog interaction
Role in autophagy
References
Sequestosome-1-like receptors (SLRs)
p62
SGGDDDWTHLSS
338
Binds to LC3A, LC3B, LC3C,
GABARAP, GABARAP-L1 and
GABARAP-L2.
Autophagy receptor for degradation of ubiquitylated
cargo: protein aggregates, soluble proteins, midbody
rings, damaged mitochondria, peroxisomes and
phagocytic membrane remnants. Also involved in
xenophagy.
(Pankiv et al., 2007;
Ichimura et al.,
2008b)
SASSEDYIIILP
732
Binds to LC3A, LC3B, LC3C,
GABARAP, GAPARAP-L1 and
GAPARAP-L2.
Autophagy receptor; targets ubiquitylated cargo. Involved
in aggrephagy. Acts as an autophagy receptor for
peroxisomes. Also involved in xenophagy.
(Kirkin et al.,
2009b; Behrends et
al., 2010)
LCGVSEWDPILE
661
Binds to six of the eight AtATG8
family members that were tested.
Plant (Arabidopsis thaliana) NBR1 that is a functional
hybrid of mammalian p62 and NBR1.
(Svenning et al.,
2011)
ENEEDILVVTT
134
Strong preference for LC3C. Note
the non-canonical LIR motif.
Autophagy receptor; targets invasive bacteria for
degradation (xenophagy)
(Thurston et al.,
2009; von Muhlinen
et al., 2012)
EGNSDMLVVTT
141
Binds to LC3C and GABARAP.
Very weak binding to LC3B.
*Note non-canonical LIR motif.
Recently identified cargo receptor where the N- terminal
part shows homology with NDP52. Role in xenophagy
has not been studied.
(Newman et al.,
2012)
GSSEDSFVEIRM
178
Binds to LC3A, LC3B,
GABARAP, GABARAP-L1 and
GABARAP-L2. (LC3C not
tested).
Autophagy receptor; targets ubiquitin-coated bacteria
(xenophagy). Also targets various protein aggregates in an
ubiquitin-independent manner.
(Wild et al., 2011)
(Q13501)
NBR1
(Q14596)
AtNBR1
(Q9SB64)
NDP52
(Q13137)
Tax1bp1/T6BP
(Q86VP1)
Optineurin
(Q96CV9)
Mitophagy receptors
Nix/Bnip3L
(O60238)
AGLNSSWVELPM
36
Binds to LC3A, LC3C,
GABARAP, GABARAP-L1 and
–L2. Relatively weak interaction
A mitophagy receptor required for selective mitochondrial
clearance after mitochondrial damage and during
(Schwarten et al.,
2009;
LIR protein
Bnip3
LIR motif
LIRpos
References
with LC3B.
erythroid differentiation.
Novak et al., 2010)
18
Binds to LC3B and GABARAPL2.
A mitophagy receptor that induces removal of both
mitochondria and ER.
(Hanna et al., 2012;
Zhu et al., 2013)
ESDDDSYEVLDL
18
Binds to LC3A, LC3B,
GABARAP and GABARAP-L2.*
A mitophagy receptor; mediates hypoxia-induced
mitophagy.
(Liu et al., 2012)
DSISGSWQAIQP
86
Binds to ScAtg8.
A mitophagy receptor; regulates selective degradation of
mitochondria in yeast
(Okamoto et al.,
2009; KondoOkomoto et al.,
2012)
(Q8IVP5)
ScAtg32
Role in autophagy
ESLQGSWVELHF
(Q12983)
FUNDC1
ATG8 ortholog interaction
(P40458)
Specialized receptors
Stbd1
RVDHEEWEMVPR
203
Binds to LC3B, LC3C,
GABARAP, GABARAP-L1 and L2. (LC3A not tested).
Specialized autophagy receptor; involved in degradation
of glycogen particles
(Behrends et al.,
2010; Jiang et al.,
2011)
ASSSFGWLSLDG
802
Binds to LC3B*.
Specialized autophagy receptor. An E3 ubiquitin ligase
that targets active tyrosine kinase Src for autophagy
(Sandilands et al.,
2012)
NEKALTWEEL
412
Binds to ScAtg8.
A cargo receptor for aminopeptidase I (Ape1) and αmannosidase in the Cvt pathway in yeast.
(Noda et al., 2008)
LSRPFTWEEI
409
Binds to ScAtg8.
A cargo receptor (Atg19 homolog) that acts cooperatively
with Atg19 in the Cvt pathway in yeast; mediates the
delivery of α-mannosidase to the vacuole but not of
aminopeptidase I (Ape1).
(Suzuki et al., 2010)
(O95210)
c-Cbl
(P22681)
Cvt cargo receptors
ScAtg19
(P35193)
ScAtg34
(Q12292)
Components of the core autophagy machinery
ULK1
SCDTDDFVMVPA
357
Preference for GABARAP and
GABARAP-L1. Also binds to
Member of the core autophagy machinery. Required for
(Alemu et al., 2012;
LIR protein
LIR motif
LIRpos
(O75385)
ULK2
ATG8 ortholog interaction
autophagosome biogenesis.
Kraft et al., 2012)
SCDTDDFVLVPH
353
Same as ULK1.
Member of the core autophagy machinery. Strongly
related to ULK1. Not needed for autophagy in cells
expressing ULK1.
(Alemu et al., 2012)
HEDSDDFVLVPK
391
Binds to DmATG8A.
Member of the core autophagy machinery in D.
melanogaster. Important for autophagosome biogenesis.
(Alemu et al., 2012)
RSFEREYVVVEK
429
Binds to ScAtg8.
Member of the core autophagy machinery in yeast.
Required for autophagosome biogenesis. Atg8 binding
triggers vacuolar degradation of the Atg1–Atg13 complex
in yeast.
(Nakatogawa et al.,
2012; Kraft et al.,
2012)
GNTHDDFVMIDF
444
Preference for GABARAP and
GABARAP-L1. Also binds to
GABARAP-L2, LC3A and
LC3C. Very weak binding to
LC3B.
Member of the core autophagy machinery; A component
of the ULK1–ULK2 complex.
(Alemu et al., 2012)
DAHTFDFETIPH
702
Binds to GABARAP and
GABARAP-L1.
Member of the core autophagy machinery; A component
of the ULK1–ULK2 complex.
(Alemu et al., 2012)
DAATLTYDTLRF
8
Binds to LC3B and LC3C.
Discrepancy in references
regarding binding to GABARAP
and GABARAP-L2.
Member of the core autophagy machinery. A cysteine
protease used for processing ATG8 family proteins prior
to conjugation of ATG8/LC3 to
phosphatidylethanolamine (PE).
(Satoo et al., 2009;
Behrends et al.,
2010; Li et al.,
2011)
LDGVGDWEDLQD
270
Binds to ScAtg8.
Member of the core autophagy machinery in yeast. An
E2-like enzyme involved in conjugation of Atg8 to
phosphatidylethanolamine.
(Yamaguchi et al.,
2010)
KIVDNDWLLPSY
105
Binds to PfAtg8.
A LIR-like motif in Plasmodium falciparum (Pf) Atg3 has
been described that interacts with PfAtg8. The importance
of the interaction for PfAtg 8 lipidation and autophagy has
(Hain et al., 2012)
(Q8MQJ7)
ScAtg1
(P53104)
ATG13
(O75143)
FIP200
(Q8TDY2)
ATG4B
(Q9Y4P1)
ScAtg3
(P40344)
PfAtg3
(C0H519)
References
GABARAP-L2, LC3A and
LC3C. Weak binding to LC3B.
(Q8IYT8)
DmATG1B
Role in autophagy
LIR protein
LIR motif
LIRpos
ATG8 ortholog interaction
Role in autophagy
References
not been investigated.
Proteins associated with autophagosomes and other vesicles
Clathrin HC
VGYTPDWIFLLR
514
Binds to GABARAP.*
It has been proposed that clathrin mediated endocytosis
contributes to the formation of early autophagic
precursors. The role of the LIR interaction in
autophagosome formation has not been addressed.
(Mohrlüder et al.,
2007a)
GSLEDDWDFLPP
200
Binds to GABARAP.*
Calreticulin competes with clathrin for binding to
GABARAP. Calreticulin might have an important
regulatory role for the GABARAP interaction with
clathrin. Direct role in autophagy is not known.
(Mohrlüder et al.,
2007a and 2007b)
EDEVDGWLIIDL
35
Binds to LC3A, LC3B, LC3C,
GABARAP, GABARAP-L1 and
GABARAP-L2.
Required for autophagosome development.
(Nowak et al.,
2009; Sancho et al.,
2012)
(Q00610)
Calreticulin
(P27797)
TP53INP2/DOR
(Q8IXH6)
TP53INP1
EKEDDEWILVDF
31
Binds to LC3A, LC3B, LC3C
GABARAP, GABARAP-L1 and
GABARAP-L2.
Localizes to autophagosomes mediated by LIR binding
and induces autophagy-dependent cell death.
(Seillier et al.,
2012; Sancho et al.,
2012)
SPLLEDWDIISP
136
Binds to LC3B, GABARAP and
GABARAP-L2.*
GTPase-activating protein for Rab33B. Regulates the
interaction of autophagosomes with lysosomes by
inactivating Rab33B.
(Itoh et al., 2011)
NSYRKEWEELFV
58
Binds to LC3A, LC3B and
GABARAP-L1.*
A Rab GTPase-activating protein recruited from
endosomes to autophagosomes during starvation-induced
autophagy. LIR 1 mediates binding to Vps29 in the
retromer complex and this binding can be titrated out by
LC3.
(Popovic et al.,
2012)
SSKDSGFTIVSP
788
Binds to LC3A, LC3B and
GABARAP-L1.*
TBC1D5 LIR2 can bridge endosomes and
autophagosomes. TBC1D5 appears to mediate
autophagosome maturation
(Popovic et al.,
2012)
(Q96A56)
TBC1D25
(Q3MII6)
TBC1D5 LIR1
(Q92609)
TBC1D5 LIR2
(Q92609)
Interacts with VMP1 and presumably acts as a scaffold
protein recruiting ATG8 proteins to the autophagosome
membrane.
LIR protein
FYCO1
LIR motif
LIRpos
ATG8 ortholog interaction
PPDDAVFDIITD
1280
Preference for LC3A and LC3B.
Weak binding to LC3C,
GABARAP, GABARAP-L1 and L2.
Involved in movement of autophagosomes to lysosomes
using microtubuli plus end-directed transport.
(Pankiv et al., 2010;
Alemu et al., 2012)
EYRSRVYQMILE
340
Binds to LC3B, GABARAP and
GABARAPL1.*
A MAP kinase that stimulates autophagy.
(Colecchia et al.,
2012)
(Q9BQS8)
MAP15K
(Q8TD08)
Role in autophagy
References
Signaling proteins that acts as a substrates for selective autophagy
Dvl2
EVRDRMWLKITI
444
Binds to LC3B and GABARAP.
Does not bind GABARAP-L2. *
Important adaptor protein in the Wnt signaling pathway.
Autophagy negatively regulates Wnt signaling by
accelerating Dvl degradation under stress conditions.
Functional role in autophagy is unknown.
(Gao et al., 2010)
LHPPSHWPLIKA
504
Binds to LC3B. Other ATG8
proteins were not tested.
Wnt signalling negatively regulates autophagy and p62
expression while β-catenin, like Dvl2 is degraded by
autophagy upon nutrient deprivation revealing a
regulatory feedback mechanism.
(Petherick et al.,
2013)
(O14641)
β-catenin
(P35222)
*Only these were tested for interaction.
Bold amino acids indicate the core consensus sequence.