Você está na página 1de 12

REVIEW

Autophagy and autophagy-related proteins


in the immune system
Shusaku T Shibutani1,2,5, Tatsuya Saitoh3, Heike Nowag4, Christian Mnz4 & Tamotsu Yoshimori1,2

Autophagy is an intracellular bulk degradation system that is highly conserved in eukaryotes. The discovery of autophagy-related
('ATG') proteins in the 1990s greatly advanced the mechanistic understanding of autophagy and clarified the fact that autophagy
serves important roles in various biological processes. In addition, studies have revealed other roles for the autophagic machinery
2015 Nature America, Inc. All rights reserved.

beyond autophagy. In this Review, we introduce advances in the knowledge of the roles of autophagy and its components in
immunity, including innate immunity, inflammatory responses and adaptive immunity.

In macroautophagy (called simply autophagy here), a double-mem- related genes in the 1990s (refs. 24). 40 genes encoding autophagy-
brane organelle called the autophagosome has a central role. The related (ATG) proteins have been identified so far, and the 15 core
formation of autophagosomes is maintained at relatively low levels genes encoding ATG proteins in yeast (ATG1ATG10, ATG12ATG14,
under non-stress conditions but is induced substantially by various ATG16 and ATG18) are conserved in mammals, which indicates that
cellular stresses (for example, amino acid starvation). The forma- autophagy is an evolutionarily conserved process. As in yeast, mamma-
tion of autophagosomes starts with the emergence of a membrane lian core ATG proteins can be classified into several functional units:
cup called the isolation membrane (or phagophore) (Fig. 1). The the ULK complex, ATG9L, the class III PI(3)K (phosphatidylinositol-
isolation membrane elongates and closes, which results in the forma- 3-OH kinase) complex, the ATG2-WIPI complex, the ATG12 conjuga-
tion of a double-membranous autophagosome that engulfs a portion tion system, and the LC3 conjugation system (Fig. 1).
of the cytoplasm. Then, subsequent autophagosome-lysosome fusion The ULK complex (a serine-threonine kinase complex composed
leads to the digestion of its contents and inner membrane by lysosomal of ULK1, ULK2, ATG13, ATG101 and FIP200) and ATG9L (ATG9L1
hydrolases (Fig. 1). and ATG9L2) are thought to be the most upstream components in
A growing body of evidence suggests that autophagy has important the autophagy pathway5. The activity of the ULK complex is regu-
roles in various biological processes at cellular and physiological levels lated negatively by the kinase complex mTORC1 and is regulated
and is generally beneficial to cells and whole organisms1. Furthermore, positively by the kinase AMPK. Both the ULK complex and ATG9L1
npg

it has also become clear that proteins involved in autophagy have func- are required for recruitment of the autophagy-specific class III PI(3)
tions other than autophagy as well. In this Review, we will discuss K complex5,6.
advances in the understanding of autophagy and its machinery as it PI(3)Ks are lipid kinases that phosphorylate phosphatidylinositol
is related to innate immunity, inflammatory responses and adaptive at the hydroxyl group at position 3 of the inositol ring, which results
immunity. in the generation of phosphatidylinositol 3-phosphate. The autoph-
agy-specific PI(3)K complex (called the ATG14L complex here) is
Autophagy proteins and their function composed of ATG14L, VPS34 (the catalytic subunit), beclin-1 and
Although autophagic structures were reported in electron microscopy VPS15. ATG14L localizes on the endoplasmic reticulum (ER), and
studies as early as the 1950s, the molecular mechanism of autophagy this localization is required for its autophagy-inducing ability7. The
remained mostly unknown until the discovery of yeast autophagy- ATG14L complex accumulates at ER-mitochondria contact sites dur-
ing starvation; this accumulation requires syntaxin-17, a SNARE (sol-
1Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier uble N-ethylmaleimide-sensitive factorattachment protein receptor)
Biosciences, Osaka University, Osaka, Japan. 2Department of Genetics, Graduate protein that resides on the ER and mitochondria8. It is likely that the
School of Medicine, Osaka University, Osaka, Japan. 3Division of Molecular ATG14L complex generates phosphatidylinositol 3-phosphate on the
Genetics, Institute for Enzyme Research, Tokushima University, Tokushima, ER at the ER-mitochondria contact site to generate a platform for the
Japan. 4Viral Immunobiology, Institute of Experimental Immunology, University of formation of an isolation membrane. The generation of phosphati-
Zrich, Zrich, Switzerland. 5Present address: Laboratory of Veterinary Hygiene, dylinositol 3-phosphate provides the binding site for its binding pro-
Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi Japan. teins, including members of the WIPI family (WIPI1WIPI4), the
Correspondence should be addressed to T.Y. (tamyoshi@fbs.osaka-u.ac.jp). mammalian orthologs of yeast ATG18. These binding proteins form
a complex with ATG2 (ATG2A and ATG2B). A study has shown that
Received 11 June; accepted 17 August; published online 18 September 2015; WIPI2 directly binds to ATG16L1 and recruits the ATG16L1 complex
doi:10.1038/ni.3273. to the autophagosome-formation site9.

1014 VOLUME 16 NUMBER 10 OCTOBER 2015 NATURE IMMUNOLOGY


REVIEW

Figure 1 The autophagy pathway. Autophagy-


Amino acid starvation
inducing signals (for example, amino acid Pathogen infection, etc.
starvation and pathogen infection) initiate Cell membrane
formation of the isolation membrane (or
phagophore). Closure of the isolation
membrane results in formation of the
autophagosome. The subsequent lysosome- ULK FIP200
autophagosome fusion leads to degradation of ATG13 ATG101 Isolation membrane Autophagosome Autolysosome
the contents of the autophagosome by lysosomal ULK1 complex
hydrolases in the autolysosome. Labels in green
boxes indicate core ATG proteins required for

Kim Caesar/Nature Publishing Group


autophagosome formation.
ATG9L

The ATG16L1 complex (composed of


ATG16L1, ATG5 and ATG12) specifically local- ATG14L VPS34
izes to the isolation membrane and dissociates Beclin-1 VPS15 LC3 Fusion
from it upon the completion of autophagosome ATG14L complex WIPI ATG16L1 ATG5 ATG12 LC3
Hydrolases
formation10. The ubiquitin-like protein ATG12 ATG2 ATG16L1 complex
Lysosome
is covalently conjugated to ATG5 by ATG7
(an E1-like enzyme) and ATG10 (a ubiquitin
E2-like enzyme)11. ATG16L1 and ATG12-ATG5 form a 2:2:2 complex12. including ligands for pattern-recognition receptors (PRRs) (for
This ATG16L1 complex functions as the E3-like enzyme for the other example, Toll-like receptors (TLRs) and Nod-like receptors (NLRs))
ubiquitin-like system: LC3-PE11. There are seven mammalian orthologs and cytokines (Box 2). In addition, the stimulation of phagocytes by
2015 Nature America, Inc. All rights reserved.

of ATG8 (LC3A, LC3B, LC3C, GABARAP, GABARAPL1, GABARAPL2 particles coated with TLR ligands also induces LC3-PE conjugation on
and GABARAPL3; these are represented by LC3 in this Review). LC3 is phagosomal membranes. This phenomenon is called LC3-associated
conjugated to the membrane lipid molecule phosphatidylethanolamine phagocytosis (LAP) (Fig. 2). We note that phagosomes (which have a
(PE) by ATG7 (an E1-like enzyme) and ATG3 (an E2-like enzyme)11. single membrane) involved in LAP are different from autophagosomes
LC3-PE conjugation takes place at the isolation membrane, where the (which have a double membrane) and that the detection of LC3 by
ATG16L1 complex (an E3-like enzyme) accumulates13. LC3 is widely fluorescence microscopy should be interpreted carefully, because this
used as a marker for the microscopic detection of isolation membranes technique cannot distinguish these two structures. Although LAP was
and autophagosomes. In addition, PE-conjugated LC3 (LC3-II) and initially shown to facilitate phagosome-lysosome fusion and accelerate
unconjugated LC3 (LC3-I) can be detected separately by immunoblot pathogen killing17, further investigation has revealed various functions
analysis, and the amount of LC3-II is also widely used for the quantifica- for LAP beyond the acceleration of pathogen killing, including antigen
tion of autophagic activity14. It has been suggested that LC3 functions in presentation (discussed below in the section on adaptive immunity).
the closure of the isolation membrane15,16.
Xenophagy
Autophagy and pathogen infection Although the role of autophagy and ATG proteins in the protection
Autophagy has been studied in relation to infection by bacteria, from pathogen infection in vivo is probably complex, the most direct
viruses, and parasites. Various in vivo studies have shown that autoph- method of autophagy-dependent elimination of pathogens is engulf-
agy serves a protective function against pathogens (Box 1). Other stud- ment of the pathogen by the autophagosome and subsequent killing
ies have suggested that autophagy suppresses excessive inflammatory of the pathogen by lysosome-autophagosome fusion.
npg

responses to pathogen infection. The relationship between autophagy Autophagy was initially thought to be a nonspecific degradation
and inflammation will be discussed later in this Review. process. However, it is now apparent that autophagy has an ability
to selectively target large structures, such as intracellular pathogens,
Regulation of the autophagy machinery by immunological signals damaged organelles and protein aggregates. This type of autophagy is
Autophagy is regulated by a plethora of immunological signals, called selective autophagy and is thought to maintain intracellular

Box 1 In vivo roles of autophagy in pathogen infection


In mice, knockout of genes encoding autophagy proteins must be conditional, since systemic knockout of these genes results in
death shortly after birth1. For example, knockout of Atg5 in myeloid cells resulted in diminished resistance to L. monocytogenes and
Toxoplasma gondii50 and M. tuberculosis80,91. Knockout of Atg5 or Atg16l1 in intestinal epithelial cells also results in diminished
resistance to Salmonella ser. Typhimurium149,150. The survival of neonatal mice after infection with Sindbis virus is diminished by
neuron-specific knockout of Atg5 (ref. 42). Knockout of Atg5 in DCs diminishes the survival of mice after infection with herpes simplex
virus type 2 (ref. 108). In Drosophila, the knockdown of ATG proteins diminishes host survival after infection with L. monocytogenes151.
Autophagy suppresses the replication of vesicular stomatitis virus in Drosophila152. Caenorhabditis elegans treated by RNA-mediated
interference targeting genes encoding ATG proteins, as well as Dictyostelium expressing mutant ATG proteins, showed reduced survival
after infection with Salmonella ser. Typhimurium153.
In contrast, however, mice with systemic hypomorphic Atg16l1, which show abnormality in the packaging and secretion of
antimicrobial granules by Paneth cells64,154, exhibit greater resistance to urinary tract infection with uropathogenic E. coli and
intestinal infection with Citrobacter rodentium, with enhanced inflammatory cytokine responses and more recruitment of monocytes
and neutrophils to infected sites70,155. The greater clearance of bacteria in these mice might be partly explained by the heightened
inflammatory responses seen in autophagy-deficient macrophages 60.

NATURE IMMUNOLOGY VOLUME 16 NUMBER 10 OCTOBER 2015 1015


REVIEW

Box 2 Immunological signals that regulate autophagy


The stimulation of TLR4 by lipopolysaccharide induces autophagosomes in macrophages156. Other TLR ligands also induce green
fluorescent proteintagged LC3 dots and autolysosomes in macrophages157. Nod1 and Nod2, which are cytosolic receptors for bacterial
peptidoglycan, interact with ATG16L1 and mediate the recruitment of ATG16L1 to the bacterial entry site in non-phagocytic cells,
macrophages and lymphocytes158. Stimulation of Nod2 also induces autophagosomes in DCs159. NLRX1 is required for vesicular
stomatitis virusinduced autophagy by interacting with the ATG16L1 complex through the mitochondrial protein TUFM 87. In contrast,
other NLRs, including NLRC4, NLRP3, NLRP4 and NLRP10, suppress autophagy by sequestering beclin-1 (ref. 160). The alarmin
HMGB1, which is released into the extracellular space and activates certain TLRs, is required for autophagy and the suppression of
endotoxin-induced death of macrophages161. HMGB1 has also been shown to bind to beclin-1 and induces autophagy by releasing
beclin-1 from the antiapoptotic protein Bcl-2 (ref. 162). HIV-1 envelope glycoproteins expressed at the cell surface activate autophagy and
cause apoptosis in bystander CD4+ T cells through the chemokine receptor CXCR4 (ref. 163). IFN-g has been shown to induce autophagy
in macrophages in a manner dependent on the GTPase family member Irgm1 (refs. 21,164). TNF induces autophagy in myoblasts in a
manner independent of the transcription factor NF-kB165 and enhances the recruitment of NDP52 to Shigella species in HeLa human
cervical cancer cells166. IL-1b also induces autophagy in macrophages in a manner dependent on the adaptor TBK-1 (ref. 167). In
contrast, the type 2 helper T cell cytokines IL-4 and IL-13 suppress autophagy168. The transcription factor STAT3, which is activated by
various cytokines, has been shown to suppress autophagy through inhibition of protein kinase R 169.

homeostasis by eliminating unwanted structures. In particular, selec- E3 ubiquitin ligases recognize substrates and therefore determine
tive autophagy directed against pathogens is called xenophagy. substrate specificity. Ubiquitination has been well characterized as a
Antibacterial xenophagy in both non-phagocytic cells and phago- hallmark event in the ubiquitin-proteasome pathway, which degrades
2015 Nature America, Inc. All rights reserved.

cytic cells has been investigated extensively. For phagocytes such as individual ubiquitinated proteins by the proteasome26. However, stud-
macrophages, dendritic cells (DCs) and neutrophils, bacteria are rec- ies have also shown its involvement in selective autophagy. Notably,
ognized by PRRs on the phagocyte surface, which leads to the activa- the fusion of ubiquitin to monomeric red fluorescent protein or
tion of intracellular signaling that induces immune responses. At the peroxisomes leads to autophagic degradation of the monomeric red
same time, bacteria are internalized by phagocytosis and are effectively fluorescent protein or peroxisomes, respectively27. Moreover, invasive
killed by phagosome-lysosome fusion as well as by exposure to reactive bacteria often localize together with ubiquitin28. Two E3 ubiquitin
oxygen species (ROS) and antimicrobial peptides18. However, inva- ligases have been related to antibacterial xenophagy thus far: LRSAM1
sive bacteria, such as Salmonella, Listeria and Shigella species, invade (refs. 29,30) and parkin31. Parkin is also known as the main E3 ubiq-
non-phagocytic cells (for example, cells of the intestinal epithelium). uitin ligase responsible for mitophagy (the selective autophagy of
They modify endosomes into a niche suitable for their survival and mitochondria)32.
proliferation and often break into the nutrient-rich cytosol from the One possible mechanism underlying the bacteria-ubiquitin colo-
vacuole19. calization noted above might be direct ubiquitination of bacteria, and
Xenophagy directed against invasive bacteria in non-phagocytic an alternative possibility is the ubiquitination of bacteria-containing
cells was first demonstrated in a study showing that the bacterium endosomes. Several studies support the latter model. The endosomal
group A Streptococcus (GAS) is efficiently targeted by an LC3+ membrane is ubiquitinated after S. flexneri escapes into the cytosol
autophagosome-like double-membraned structure and is later sub- from the endosome33. Salmonella ser. Typhimurium inside endosomes
jected to lysosomal degradation20. Moreover, this study showed that has been found to be targeted by autophagosomes6,23. Strikingly, even
genetic deletion of Atg5 in mice results in the absence of the LC3+ latex beads ingested by endocytosis can induce autophagosome forma-
npg

structure and in improved survival of GAS. This study clearly showed tion after the bead-containing endosomal membrane is ruptured34,35,
that autophagy functions as an intracellular innate immune system which suggests that no bacterial factors are required for the selective
directed against bacterial invasion in non-phagocytic cells. Around formation of autophagosomes. Moreover, rupture of the lysosomal
the same time as that study, another study showed that the survival of membrane induces selective ubiquitination and autophagy of the
Mycobacterium bovis bacillus Calmette-Gurin and Mycobacterium ruptured lysosome (a process called lysophagy)36,37. An important
tuberculosis in macrophages is suppressed by the induction of autoph- insight from these results is that cells might be simply detecting the
agy21. Follow-up studies have shown that other invasive bacteria, rupture in pathogen-containing endosomes as an autophagy-inducing
including Shigella flexneri22, Salmonella enterica serovar Typhimurium signal rather than preparing different autophagy-inducing mecha-
(Salmonella ser. Typhimurium)23 and Listeria monocytogenes24, are nisms for various kinds of pathogens.
also targeted by autophagy.
However, there are bacterial species that have evolved mechanisms Recruitment of LC3 through autophagy adaptors
to prevent autophagy in a strain-specific manner and therefore have How can ubiquitination function as the signal for selective autophagy?
become more vulnerable to antibacterial xenophagy when these self- Autophagy adaptors (receptors) such as the ubiquitin sensor SQSTM1
defense mechanisms are impaired. These bacteria include S. flexneri, (p62), NDP52 and optineurin have been suggested to serve an impor-
L. monocytogenes and Legionella pneumophila, as reviewed in detail tant role in xenophagy directed against intracellular bacteria25. These
elsewhere25. adaptors contain ubiquitin-binding domains and LC3-interacting
regions, and therefore it is proposed that these adaptors recruit the LC3
Mechanisms of xenophagy: ubiquitination of the substrate machinery to ubiquitinated substrates (Fig. 2). In addition, NDP52
The key event that triggers antibacterial xenophagy and other selective binds galectin-8, a b-galactose-binding lectin. Since b-galactose chains
autophagy is ubiquitination (the covalent conjugation of ubiquitin to exist in the lumen of endosomes, galectin-8 and its binding partner
a substrate). Ubiquitination is mediated by E1 (ubiquitin-activating NDP52 are recruited from the cytoplasm to the endosomal membrane
enzyme), E2 (ubiquitin-conjugating enzyme) and E3 (ubiquitin ligase). when the endosome is ruptured38.

1016 VOLUME 16 NUMBER 10 OCTOBER 2015 NATURE IMMUNOLOGY


REVIEW

Figure 2 Xenophagy and LAP. In ubiquitin-


dependent xenophagy (left), the recruitment
of ATG proteins occurs after the pathogen- Ubiquitin-dependent
containing endosome is ruptured. Endosomal xenophagy LAP in phagocytes
membrane proteins are ubiquitinated (Ub) Pathogen
immediately after the membrane rupture. In (bacteria, etc.)
addition, adaptors such as SQSTM1 (p62), Cell membrane
NDP52 and optineurin mediate recruitment
of LC3 to the ubiquitinated substrate. NDP52
can be recruited to the ruptured endosomal
membrane through binding to galectin-8, which
recognizes b-galactose chains in the lumen of
endosomes (not shown here). Formation of the
isolation membrane takes place in the proximity
of the pathogen-containing endosome, and
finally, the autophagosome engulfs the pathogen-
containing endosome. The subsequent lysosome-
autophagosome fusion eliminates the pathogen.
In contrast to the process used for xenophagy,
the double-membraned autophagosome is not WIPI or
formed in LAP (right). Instead, the phagosomal ATG2

membrane is decorated directly by LC3. LAP Ub ULK


complex
facilitates lysosome-phagosome fusion or ATG14L1
complex
prolonged antigen processing for presentation by
2015 Nature America, Inc. All rights reserved.

Ub ATG9L LC3
MHC class II in a cell typespecific manner.
ATG16L1
Ub complex LC3

Recruitment of ATG proteins


Ub Adaptor LC3
However, even in the absence of the recruit- Fusion
ment of LC3 to intracellular Salmonella
species (in cells deficient in Atg3, Atg5 or
Atg7), formation of an isolation membrane Lysosome or
can initiate around Salmonella-containing endosome

Kim Caesar/Nature Publishing Group


endosomes6. In the absence of LC3 recruit-
ment, other ATG proteins such as the ULK1
complex and ATG9L1 are recruited to intra-
Fusion
cellular Salmonella, which suggests that the
recruitment of these early ATG proteins is
sufficient to begin the formation of an isola- Lysosome
tion membrane independently of the adap-
tor-mediated recruitment of LC36, although
these isolation membranes cannot mature genes, SMURF1, has been shown to be required for cell survival after
to complete autophagosome formation without LC3-PE. A follow-up infection with Sindbis virus or herpes simplex virus type 1 (ref. 43).
npg

study has revealed that recruitment of the ULK1 complex, ATG9L1 The human immunodeficiency virus type 1 (HIV-1) virulence fac-
and the ATG16L1 complex (and the downstream ATG proteins tor Nef has been shown to suppress autophagy by binding to beclin-1
ATG14L, WIPI1 and LC3) to ruptured endosomes is all dependent (ref. 44). The Nef-binding sequence of beclin-1 (amino acids 267284)
on ubiquitination35. Recruitment of the ATG16L1 complex involves induces autophagy when modified as a cell-permeable peptide (Tat
direct ATG16L1-ubiquitin binding through the carboxy-terminal WD beclin-1). Tatbeclin-1 suppresses Sindbis virus, Chikungunya virus,
domain of ATG16L1 and direct ATG16L1-FIP200 binding (FIP200 West Nile virus and HIV-1 in vitro and diminishes the lethality of
is a subunit of the ULK1 complex)35. The exact mechanisms of the infection with Chikungunya virus or West Nile virus in vivo45.
ubiquitination-dependent recruitment of the ULK1 complex and
ATG9L1 remain unknown. Autophagy-independent functions of ATG proteins
The role of ATG proteins in immunity is not restricted to xenophagy.
Xenophagy directed against viruses They can also function in autophagy-independent ways. One striking
The role of autophagy in viral infection is complex and depends on the example is LAP. Unlike the process of canonical autophagy, single-
virus. Although several viruses have autophagy-inhibitory functions membrane phagosomes are modified by LC3 during LAP in a way
and even exploit autophagy for their replication39,40, the virulence of dependent on ATG5, ATG7 and beclin-1 but independent of ATG14L
several other viruses is suppressed by autophagy. For example, capsid or the ULK1 complex17,4648. In another example, Brucella abortus
proteins of Sindbis virus are degraded by autophagy in an SQSTM1- exploits autophagic machinery by forming autophagic Brucella-
dependent manner, and mice with neuron-specific Atg5 deficiency containing vacuoles. Although these vacuoles have double-mem-
exhibit greater susceptibility to infection with Sindbis virus than do braned structures reminiscent of autophagosomes and require ULK1,
their wild-type counterparts41,42. Genome-wide screening with small beclin-1 and ATG14L, they lack the autophagosome marker LC3 and
interfering RNA has identified 141 genes whose knockdown results do not require ATG5 or ATG16L1 (ref. 49). Other examples of autoph-
in a change in the colocalization of Sindbis virus capsid protein with agy-independent functions of ATG proteins include ATG5-dependent
green fluorescent proteintagged LC3. The product of one of those membrane damage in Toxoplasma-containing vesicles50, ATG16L1

NATURE IMMUNOLOGY VOLUME 16 NUMBER 10 OCTOBER 2015 1017


REVIEW

complexdependent inhibition of the formation of membranous rep- However, ATG16L1 in intestinal epithelial cells also controls inflam-
lication complex in mouse norovirus infection51, and LC3-associated mation. Hypomorphic mutation of Atg16l1 in mice results in Paneth
ejection of intracellular pathogens such as poliovirus52, coxsackie cell abnormality due to infection with norovirus and induces elevated
virus53 and uropathogenic Escherichia coli54 toward the extracellular production of tumor-necrosis factor and interferon-g (IFN-g), which
space. Other examples are also discussed below. Such examples indi- results in enhanced susceptibility to dextran sodium sulfateinduced
cate that the observation of one feature associated with autophagy (for colitis64. Hence, autophagy has a key role in the control of inflamma-
example, the recruitment of LC3, the formation of double-membrane tory cytokine production.
structures, etc.) does not necessarily guarantee that the phenomenon NLRP3 is a well-characterized PRR of the NLR family that induces
is canonical autophagy. Therefore, such phenomena should be inter- the production of IL-1b and IL-18 (ref. 57). Various inducers of the
preted carefully, and the involvement of autophagy should be assessed NLRP3 inflammasome, such as ATP, palmitic acid, monosodium urate
by multiple, functionally different experiments. crystals, silica nanoparticles, cholesterol crystals and influenza A virus,
cause mitochondrial damage, which leads to the release of ROS. NLRP3
Autophagy in innate immunity forms an inflammasome complex with ASC and caspase-1 and induces
The innate immune system induces inflammation to protect the host the production of IL-1b and IL-18 in response to mitochondrial ROS.
from microbial infection55. PRRs sense microbial components and Because autophagy maintains cellular homeostasis by promoting the
induce activation of signal-transduction pathways, which leads to the turnover of organelles, a deficiency in autophagy results in the accu-
expression of inflammatory mediators such as cytokines and type I mulation of old and dysfunctional mitochondria. Accumulated old and
interferons. Although inflammation is essential for host defense, aber- dysfunctional mitochondria produce an excess of ROS upon stimu-
rant induction of inflammation causes the development of inflam- lation, which potently activates the NLRP3 inflammasome65,66 (Fig.
matory diseases, such as septic shock, autoimmune diseases and 3). In addition, lysophagy can also be important for suppressing the
metabolic diseases. Thus, the innate immune system is strictly regu- activation of inflammasomes, since extracellular irritant particles such
2015 Nature America, Inc. All rights reserved.

lated to prevent either insufficient inflammatory responses or excessive as monosodium urate and silica are internalized into cells via the endo-
inflammatory responses. Accumulating evidence has shown that the cytic pathway and break lysosomes, which might damage mitochon-
intracellular degradation system has an important role in regulating dria. Lysophagy sequesters lysosomes damaged by monosodium urate
inflammatory responses. In particular, studies have shown the involve- or silica in vitro and in vivo37. Upon infection with influenza A virus,
ment of autophagy in inflammatory responses56. the cytosolic PRR Nod2 and its downstream regulator RIPK2 induce
mitophagy by inducing phosphorylation of ULK1 to prevent excessive
Autophagy in inflammasome activation activation of the NLRP3 inflammasome67. A deficiency in autophagy
The inflammasome is a cytosolic protein complex consisting of NLRs worsens the symptoms of various inflammatory conditions, such as
or of the cytosolic receptor AIM2 together with the adaptor ASC colitis, sepsis, pneumonia, diabetes, atherosclerosis and urinary tract
and the protease caspase-1 (ref. 57). Activation of inflammasomes infection, by enhancing NLRP3 inflammasomedependent production
induces the proteolytic maturation of interleukin 1b (IL-1b) and the of IL-1b and IL-18 (refs. 6670). In contrast, inducers of autophagy
related cytokine IL-18 by caspase-1, which in turn induces produc- can suppress the inflammasome-dependent production of IL-1b and
tion of these cytokines. Inflammasomes drive cytokine-dependent IL-18 and ameliorate symptoms of inflammation-associated tissue
host defense against microbes. However, aberrant activation of damage7173. Hence, autophagy might be a useful therapeutic target
inflammasomes by an excess of microbial components, host-derived for the treatment of inflammasome-related inflammatory diseases.
stimulatory factors and environmental irritants often causes massive Autophagy inhibits not only the NLRP3 inflammasome but also
inflammation, which results in severe tissue damage. Hence, inflam- other inflammasomes, such as the AIM2 inflammasome. Lys63
masomes are closely associated with the development of infectious (K63)-linked ubiquitination of ASC, an essential adaptor of inflam-
npg

and inflammatory diseases. masomes, is induced upon activation of the NLRP3 and AIM2 inflam-
Genome-wide association studies have identified ATG16L1 as a gene masomes74. Autophagy recognizes ubiquitinated ASC by the ubiquitin
encoding a factor responsible for susceptibility to Crohns disease, a sensor SQSTM1 and induces selective degradation of inflammasomes
chronic inflammatory disease of the intestine58,59. The importance and thereby suppresses the production of IL-1b and IL-18 (Fig. 3).
of ATG16L1 in the maintenance of intestinal homeostasis has been Furthermore, the autophagy system delivers IL-1b into lysosomal com-
confirmed in mouse models. Mice with hematopoietic cellspecific partments to induce its degradation75 (Fig. 3). Hence, autophagy can
deficiency in ATG16L1 and mice that express the Crohns disease target multiple steps of inflammasome activation and thus can potently
associated ATG16L1 variant ATG16L1 T300A are susceptible to colitis inhibit the production of IL-1b and IL-18.
induced by dextran sodium sulfate and colitis induced by pathogenic Autophagy limits the activation of inflammasomes under normal
bacteria, respectively6062. ATG16L1-deficient macrophages produce and nutrient-rich conditions. However, a study has shown that autoph-
large amounts of IL-1b and IL-18 in response to lipopolysaccharide, agy often mediates unconventional release of IL-1b under conditions
ATP or monosodium urate crystals (Fig. 3). Expression of the Crohns of nutrient starvation76. GORASP and RAB8A, which are membrane
disease-associated variant ATG16L1 T300A in peripheral blood mono- traffic regulators, are involved in the autophagy-dependent uncon-
nuclear cells from patients with Crohns disease and in macrophages ventional release of IL-1b. However, other studies have shown that
from genetically engineered mice results in elevated production of autophagy suppresses IL-1b production under conditions of nutri-
IL-1b6163. Caspase-3 and caspase-7, which are apoptotic proteases, ent starvation70,74. Hence, the role of autophagy in IL-1b production
enhance the production of IL-1b by inducing proteolytic cleavage of under conditions of nutrient starvation is still unclear and needs to
the Crohns diseaseassociated variant ATG16L1 T300A61,62. Loss be investigated further.
of autophagy in macrophages greatly augments the production of Other studies have shown that the non-canonical inflammasome
IL-1b, as macrophages deficient in ATG7 or treated with an inhibi- consisting of caspase-11 in mice and caspase-4 and caspase-5 in
tor of the ATG14L complex catalytic subunit VPS34 produce large humans induces the production of IL-1a and/or IL-1b and IL-18 in
amounts of IL-1b in response to various inflammasome inducers60. response to cytosolic lipopolysaccharide77. During infection with

1018 VOLUME 16 NUMBER 10 OCTOBER 2015 NATURE IMMUNOLOGY


REVIEW

DNA virus Bacteria Uric acid crystals Influenza Palmitic LPS ATP
Cholesterol crystals virus acid

TLR4 P2X7 Cell membrane

Phagosome
damage

Phagosome Autophagy
damage induction

Autophagosomes

dsDNA LPS
leakage leakage
Non- Mitochondria
canonical damage
pathway ROS Engulfment
generation of substrates
AIM2 NLRP3 Ub
PRR activation Ub Ub
2015 Nature America, Inc. All rights reserved.

ASC

Ub Lysosome
ASC oligomerization ASC Ub Ub
fusion

Cytokine production IL-1

Kim Caesar/Nature Publishing Group


Autophagic
degradation
Inflammation

Autolysosomes

Figure 3 Autophagy suppresses the inflammasome-mediated production of IL-1b. NLRP3 forms an inflammasome with its adaptor ASC, and its excessive
activation causes inflammatory diseases such as gout. Activators of the NLRP3 inflammasome cause mitochondrial damage, which in turn causes the
production of ROS; this results in activation of the NLRP3 inflammasome. Autophagy induces the elimination of old and dysfunctional mitochondria to
suppress ROS generation and subsequent activation of the NLRP3 inflammasome. Autophagy also induces the elimination of damaged phagosomes,
npg

ubiquitinated ASC and IL-1b to suppress IL-1b production mediated by other inflammasomes. LPS, lipopolysaccharide.

Gram-negative bacteria, a cluster of guanylate-binding proteins, T cells, which results in severe lung tissue damage in a mouse model
which are small interferon-inducible GTPases, moves to the surface of tuberculosis80. Autophagy-deficient macrophages produce large
of the bacteria-containing vacuoles (BCVs) and induces membrane amounts of IL-1a in response to M. tuberculosis due to an elevation
rupture78. The destabilization of BCVs releases bacterial components in mitochondrial ROS. Hence, mitochondrial ROS have a critical role
into the cytosol and thereby facilitates recognition of their lipopolysac- in activation of the inflammasome and calpain in autophagy-deficient
charide by the non-canonical inflammasome. A deficiency in autoph- macrophages.
agy causes accumulation of the destabilized BCVs, which results in
enhanced activation of the non-canonical inflammasome (Fig. 3). Autophagy in type I interferon production
Autophagy targets damaged BCVs via ubiquitin and galectin-8 and The innate immune system senses viral nucleic acids through PRRs
induces their autophagic degradation as described above. Hence, such as TLRs, RLRs and cGAS and induces production of the type
autophagy-mediated clearance of BCVs prevents excessive induction I interferons IFN-a and IFN-b8183. Type I interferons induce the
of the inflammatory response mediated by the non-canonical inflam- expression of a series of antiviral factors to establish an antiviral state
masome. and have a crucial role in host defense against viruses.
TLR7 and TLR9 sense viral single-stranded RNA and unmethylated
Autophagy in inflammasome-independent IL-1 production CpG DNA, respectively, in endolysosomes and mediate robust produc-
The maturation and production of IL-1a involve caspase 1-depen- tion of type I interferons by plasmacytoid DCs (pDCs)81. Loss of ATG5
dent and caspase 1-independent processes79. In caspase-1-indepen- severely impairs TLR7-dependent production of type I interferons by
dent process, the maturation of IL-1a by calpain is triggered by ROS pDCs infected with vesicular stomatitis virus or Sendai virus84. Loss
released from damaged mitochondria. A deficiency in autophagy in of ATG5 also severely impairs TLR9-dependent production of type I
phagocytes causes enhanced responses of the TH17 subset of helper interferons by pDCs stimulated with A- or D-type unmethylated CpG

NATURE IMMUNOLOGY VOLUME 16 NUMBER 10 OCTOBER 2015 1019


REVIEW

oligonucleotides (ligands for TLR9). Furthermore, LAP deficiency gens to the autophagy machinery results in the depletion of specific T
severely impairs pDC production of type I interferon induced by com- cells during negative selection, which eliminates autoreactive T cells103.
plexes of DNA and immunoglobulins and dependent on TLR9 (ref. Therefore, the processing of endogenous MHC class II antigens via
48). Hence, autophagy and LAP are required for the sensing of nucleic autophagy contributes to the induction of T cell tolerance and pro-
acids and TLR-mediated production of type I interferons by pDCs. motes the presentation of intracellular antigens to CD4+ T cells.
The RLRs RIG-I and Mda5 are caspase-recruitment domaincon-
taining RNA helicases and sense viral double-stranded RNA (dsRNA) Exogenous antigen processing for presentation by MHC class II
in the cytosol82. After sensing viral dsRNA, RLRs induce the produc- In addition to the function noted above of classical autophagy in anti-
tion of type I interferons through activation of the mitochondrial gen processing, the LC3-PEconjugation machinery also regulates
caspase-recruitment domaincontaining adaptor IPS-1 (also known the processing of exogenous antigens via phagocytosis. During this
as MAVS, VISA and CARDIF). ATG5 deficiency enhances the produc- LAP, LC3 is coupled to a subset of phagosomal membranes that sur-
tion of type I interferons by RNA viruses such as vesicular stomatitis round TLR2, TIM4, Fc receptors or dectin-1-binding cargo17,46,104106.
virus and inhibits viral replication. ATG5 deficiency results in the The production of ROS seems to be needed to maintain the conjuga-
accumulation of old and dysfunctional mitochondria, which in turn tion of LC3 to phagosomes involved in LAP47,105. The fate of these
induces elevated expression of IPS-1 and the generation of mitochon- phagosomes depends on the cellular background in which they form.
drial ROS; this results in enhanced production of type I interferons by Mouse macrophages rapidly degrade the contents of LAP phago-
dsRNA85. ATG12-ATG5 conjugates and their binding partner TUFM somes in lysosomes, possibly by LC3B-dependent recruitment of
suppress dsRNA-induced production of type I interferons by disrupt- the vesicular transport protein FYCO1, which might accelerate the
ing signals from RLRs and IPS-1 (refs. 86,87). Hence, autophagy and microtubule transport of LAP phagosomes17,46,107. In contrast, pDCs
a unique function of autophagy-related proteins are involved in nega- and human macrophages, as well as monocyte-derived DCs, seem to
tive regulation of the RLR-mediated production of type I interferons. stabilize these vesicles for fusion with TLR-containing endosomes and
2015 Nature America, Inc. All rights reserved.

Studies have shown that cGAS senses microbial dsDNA in the cyto- prolonged processing of antigen for presentation by MHC class II,
sol83. After recognizing dsDNA, cGAS generates cGAMP to activate respectively48,105. Cargo that engages dectin-1 or TLR2 seems to be
STING, a downstream regulator, which in turn induces the production efficiently processed via LAP for presentation by MHC class II mol-
of type I interferons; this results in the establishment of an antiviral ecules105,106 (Fig. 4). Similarly, the LC3-PEconjugation machinery
state. Loss of ATG9L1 causes hyper-aggregation of STING on Golgi- seems to be required for efficient presentation by MHC class II in
derived compartments and enhances STING-dependent production vivo108. Moreover, hydrolases that facilitate antigen processing can be
of type I interferons88. ATG9L1 controls the subcellular localiza- transported into lysosomes via autophagy, as shown for peptidylargi-
tion of STING via unknown mechanisms. Inhibition of ULK1 also nine deiminases that generate citrullinated peptides, which constitute
enhances the STING-dependent production of type I interferons89. epitopes that are recognized by CD4+ T cells in rheumatoid arthri-
ULK1 induces the phosphorylation of STING to prevent sustained tis109. However, LAP does not seem to affect all phagocytosed cargo
production of type I interferons. Inhibition of beclin-1 enhances similarly. Although the autophagy machinery has been reported to
cGAS-dependent production of type I interferons. Beclin-1 binds to assist the transport of antigens recognized by B cell antigen receptors
cGAS to inhibit cGAMP synthesis90. In contrast, the cGAS-STING to TLR-containing endosomes110, this does not seem to be required
signaling axis is required for the targeting of autophagosomes to and for the establishment of the presentation of antigens to helper T cells
their elimination of M. tuberculosis9193. Hence, both autophagy and during germinal center reactions111. Thus, the autophagy machinery
unique functions of autophagy-related proteins regulate the cGAS- that mediates LC3-PEconjugation can modify the fate and content
mediated anti-microbial response. of phagosomes and facilitate the presentation of exogenous antigens
by MHC class II molecules.
npg

Endogenous antigen processing by autophagy


In contrast to sensing by the innate immune system via PRRs, the adap- Antigen processing for presentation by MHC class I
tive immune system relies on the recognition of peptides processed The classical function of the LC3-PEconjugation machinery (deliv-
from pathogen- or tumor-associated proteins and their presentation on ering substrates for lysosomal degradation) might limit the access of
major histocompatibility complex (MHC) molecules. For this purpose, antigens to proteasomal turnover112. In addition, proteasomes can be
MHC class I molecules are loaded mainly with proteasomal products targeted by the autophagy machinery113,114, but this might affect mainly
for their recognition by CD8+ T cells, while MHC class II molecules dysfunctional proteasomes115 and contribute little to further constrain
receive fragments of lysosomal degradation for their recognition by proteasomal processing of antigens for presentation by MHC class
CD4+ T cells94. Autophagy delivers cytoplasmic constituents for lyso- I molecules112. However, viral infection can block this proteasomal
somal hydrolysis and thereby contributes to the processing of endog- degradation and the import of antigenic peptides into the ER, medi-
enous antigens for presentation by MHC class II molecules95 (Fig. 4). ated by the transporter TAP, for loading onto MHC class I molecules.
This was first noted for a prominent antigen of Epstein-Barr virus Under these conditions, autophagy and possibly even LAP might con-
recognized by CD4+ T cells96,97. Further studies have demonstrated tribute to the loading of antigen onto MHC class I molecules (Fig. 4).
that rapamycin-induced autophagy or targeting of model antigens to Indeed, involvement of autophagy in the presentation of antigens by
autophagosomes by fusion to LC3 augments the presentation of intra- MHC class I molecules during herpesvirus infection in vitro has been
cellular antigens on MHC class II molecules98100. However, the best- shown for herpes simplex virus type 1 and human cytomegalovirus,
characterized physiological process that requires intracellular antigen which both encode TAP-blocking proteins116,117. Notably, autophagy
loading is T cell education in the thymus101. Thymic epithelial cells is reported to assist in the upregulation of MHC class I mediated by
present self antigens on MHC molecules for the induction of central T IFN-g118. Therefore, autophagy might contribute to the delivery of
cell tolerance. Indeed, thymic epithelial cells have high constitutively intracellular antigens to vesicular MHC class Iloading compart-
active levels of autophagy101,102; absence of this autophagy results in ments and to the processing of extracellular antigens for presenta-
the compromised education of CD4+ T cells101. The targeting of anti- tion by MHC class I, called cross-presentation. Cross-presentation of

1020 VOLUME 16 NUMBER 10 OCTOBER 2015 NATURE IMMUNOLOGY


REVIEW

Figure 4 ATG proteins in MHC-restricted


antigen presentation. Autophagy-related ATG8 and LC3 Cell
proteins contribute, by at least three pathways, MHCII membrane
Antigen-
to the loading of vesicular antigens onto MHC MHCI donor cell
molecules. Autophagosomes can fuse with Antigen
endosomal MHC loading compartments and
deliver cytoplasmic constituents for loading onto
MHC class II (MHCII). Loading of MHC class
I (MHCI) might also occur in these vesicular Exocytosis of
Endosomal MHC
compartments, especially under conditions in loading compartment
LC3-decorated
which the classical processing of classical MHC vesicles
class I antigens, including import into the ER by LAP
TAP, is inhibited. LAP can deliver extracellular
antigens to this loading compartment. LC3
might need to be deconjugated from the outer
autophagosomal and the LAP phagosome
membrane by ATG4 for efficient fusion with the ATG4

Kim Caesar/Nature Publishing Group


endosomal MHC loading compartment. Finally,
signal peptideindependent but autophagy-
dependent exocytosis seems to efficiently
transfer antigens from donor cells to antigen- ATG4
presenting cells, mainly for cross-presentation by
MHC class I molecules.
Antigen or
2015 Nature America, Inc. All rights reserved.

Macroautophagy presenting cell

antigens from respiratory syncytial virus, HIV, Chlamydia bacterial Autophagy also has a crucial role in the function of mature T cells.
species and Aspergillus mold species are reported to benefit from the Proliferating T cells upregulate autophagy134, and the survival of T cell
LC3-PEconjugation machinery119122, presumably via the fusion of during proliferation depends on this134,138. A lack of autophagy leads
LAP phagosomes with vesicular MHC class I loading compartments. to terminally differentiated senescent CD8+ T cells with dysfunctional
Hence, vesicular loading of MHC class I might benefit from autophagy mitochondria, increased ROS production and elevated amounts of
and LAP, especially when TAP is blocked. p38 (a mitogen-activated protein kinase)139. Indeed, p38 suppresses
autophagy in these cells, and its inhibition restores proliferative func-
Antigen packaging for cross-presentation tion. Furthermore, the altered cell organelle homeostasis with enlarged
In addition to the catabolic functions of the LC3 lipidation machinery, ER structures compromises Ca2+ mobilization after T cell stimula-
alterations that affect completion of the autophagosome-maturation tion140. This seems to affect not only effector T cells but also regulatory
process might promote exocytosis. This has been demonstrated in T cells141. Moreover, CD4+ T cells defective in autophagy are suscep-
yeast for signal peptideindependent secretion123,124 and has been sug- tible to apoptosis after they are activated142. In addition, a deficiency
gested for the release of, among others, the alarmin HMGB1, IL-1b, in autophagy causes impaired survival of memory CD8+ T cells during
ATP and secretory lysosomes by mammalian cells76,125127. Moreover, infection with lymphocytic choriomeningitis virus, influenza virus
some viruses, such as HIV, polio and Epstein-Barr virus, might uti- or mouse cytomegalovirus143,144. T cell memory responses are par-
npg

lize exocytosis involving autophagic membranes to exit infected tially restored by the autophagy stimulator spermidine143. Therefore,
cells44,128,129. This autophagic exocytosis might also benefit antigen autophagy might be required for the population expansion and mem-
processing and has been shown to facilitate the cross-presentation of ory maintenance of lymphocytes.
tumor antigens and antigens from influenza A virus130,131 (Fig. 4).
Accordingly, vesicular fractions generated from antigen-donor cells Autophagy in the development and function of B cells
under autophagy-stimulating and lysosome-inhibiting conditions, The loss of autophagy in developing B cells results in a marginally
some of which are called defective ribosomal products in blebs (or decreased total number of cells135,145. However, the number of mouse
dribbles), are now being explored as antigen formulations for better B-1a cells is substantially compromised when genes encoding essential
cross-presentation132. Thus, autophagic membranes might contribute ATG proteins are deficient111,145. This decrease seems to originate
to antigen release for efficient cross-presentation. from the autophagy-dependent transition from pre-B cell to pro-B
cell. Therefore, a subset of B cells is dependent on autophagy during
Autophagy in T cells their differentiation.
Beyond its role in antigen processing, autophagy shapes adaptive immu- In line with the dependence of B cell development on functional
nity through its contribution to the development and effector function autophagy, effector functions of the humoral immune response are
of lymphocytes. While myeloid cells seem able to differentiate in the also affected by a deficiency in autophagy. The terminal effector func-
absence of autophagy and can even cause severe myeloproliferation, tion resides with plasma cells that continuously generate antibodies.
a deficiency in autophagy in hematopoietic progenitor cells in mice In addition, memory B cells can be reactivated for antibody produc-
severely compromises the lymphoid lineage133. Accordingly, thymo- tion upon encountering cognate antigen. Both T celldependent
cyte development upregulates and is dependent on functional autoph- antibody responses and T cellindependent antibody responses that
agy134,135. The number of naive T cells is much lower in the absence lead to plasma cell differentiation require autophagy111,146. Moreover,
of autophagic clearance of damaged mitochondria (mitophagy)136,137. the long-term survival of plasma cells in the bone marrow of mice is
Thus, mitophagy seems to be a prerequisite for T cell development. diminished by a deficiency in autophagy111. The absence of autophagy

NATURE IMMUNOLOGY VOLUME 16 NUMBER 10 OCTOBER 2015 1021


REVIEW

in plasma cells results in massive enlargement of the ER, which trig- 13. Fujita, N. et al. The Atg16L complex specifies the site of LC3 lipidation for membrane
biogenesis in autophagy. Mol. Biol. Cell 19, 20922100 (2008).
gers unfolded protein responses during the production of immuno- 14. Kabeya, Y. et al. LC3, a mammalian homologue of yeast Apg8p, is localized in
globulins. This uncontrolled stress response is thought to induce cell autophagosome membranes after processing. EMBO J. 19, 57205728 (2000).
death111. However, a direct effect of autophagy on antibody production 15. Fujita, N. et al. An Atg4B mutant hampers the lipidation of LC3 paralogues and
causes defects in autophagosome closure. Mol. Biol. Cell 19, 46514659 (2008).
by plasma cells and multiple myeloma cells has not been shown111,147. 16. Sou, Y. et al. The Atg8 conjugation system is indispensable for proper develop-
In addition to the maintenance of plasma cells, autophagy is also ment of autophagic isolation membranes in mice. Mol. Biol. Cell 19, 47624775
(2008).
required for the survival of memory B cells. Mice with autophagy- 17. Sanjuan, M.A. et al. Toll-like receptor signalling in macrophages links the autophagy
deficient B cells have severely impaired specific secondary antibody pathway to phagocytosis. Nature 450, 12531257 (2007).
responses to influenza A virus due to compromised maintenance of Identification of LAP in this study.
18. Flannagan, R.S., Coso, G. & Grinstein, S. Antimicrobial mechanisms of phagocytes
memory B cells148. Consequently, humoral immune responses, par- and bacterial evasion strategies. Nat. Rev. Microbiol. 7, 355366 (2009).
ticularly the protection due to sustained antibody production and 19. Cossart, P. & Sansonetti, P.J. Bacterial invasion: the paradigms of enteroinvasive
pathogens. Science 304, 242248 (2004).
secondary responses, necessitate autophagy. 20. Nakagawa, I. et al. Autophagy defends cells against invading group A Streptococcus.
Science 306, 10371040 (2004).
Concluding remarks Study clarifying, through the use of and LC3 and cells deficient in genes encoding
ATG proteins, the fact that autophagy can target intracellular bacteria.
Since the discovery of the ATG proteins, the molecular mechanism of 21. Gutierrez, M.G. et al. Autophagy is a defense mechanism inhibiting BCG and
autophagy and its relationship with immunity has been investigated Mycobacterium tuberculosis survival in infected macrophages. Cell 119, 753766
extensively. As noted in this Review, it is now indisputable that autoph- (2004).
22. Ogawa, M. et al. Escape of intracellular Shigella from autophagy. Science 307,
agy and its machinery have critical roles in immunity, including the 727731 (2005).
elimination of pathogens, the regulation of inflammatory responses, 23. Birmingham, C.L., Smith, A.C., Bakowski, M.A., Yoshimori, T. & Brumell, J.H.
Autophagy controls Salmonella infection in response to damage to the Salmonella-
and antigen processing and presentation, as well as the development containing vacuole. J. Biol. Chem. 281, 1137411383 (2006).
of cells of the immune system. However, the exact functions of ATG
2015 Nature America, Inc. All rights reserved.

24. Py, B.F., Lipinski, M.M. & Yuan, J. Autophagy limits Listeria monocytogenes intracel-
proteins and mechanism of autophagosome formation are still in being lular growth in the early phase of primary infection. Autophagy 3, 117125 (2007).
25. Huang, J. & Brumell, J.H. Bacteria-autophagy interplay: a battle for survival. Nat.
elucidated. Future investigations should clarify the fundamental mech- Rev. Microbiol. 12, 101114 (2014).
anism of autophagy in more detail and will probably reveal unexpected 26. Welchman, R.L., Gordon, C. & Mayer, R.J. Ubiquitin and ubiquitin-like proteins as
roles for autophagy in immunity. multifunctional signals. Nat. Rev. Mol. Cell Biol. 6, 599609 (2005).
27. Kim, P.K., Hailey, D.W., Mullen, R.T. & Lippincott-Schwartz, J. Ubiquitin signals
autophagic degradation of cytosolic proteins and peroxisomes. Proc. Natl. Acad. Sci.
ACKNOWLEDGMENTS USA 105, 2056720574 (2008).
Supported by the Ministry of Education, Culture, Sports, Science and Technology 28. Fujita, N. & Yoshimori, T. Ubiquitination-mediated autophagy against invading bac-
of Japan (S.T.S., T.S. and Y.T.), the Japan Agency for Medical Research and teria. Curr. Opin. Cell Biol. 23, 492497 (2011).
Development CREST (S.T.S., T.S. and Y.T.), Cancer Research Switzerland (KFS- 29. Ng, A.C.Y. et al. Human leucine-rich repeat proteins: a genome-wide bioinformatic
3234-08-2013 for H.N. and C.M.), Worldwide Cancer Research (14-1033 for categorization and functional analysis in innate immunity. Proc. Natl. Acad. Sci. USA
H.N. and C.M.), the clinical research programs KFSPMS and KFSPHHLD of the 108, 46314638 (2011).
University of Zurich (H.N. and C.M.), the Baugarten Foundation (H.N. and C.M.), 30. Huett, A. et al. The LRR and RING domain protein LRSAM1 is an E3 ligase crucial
for ubiquitin-dependent autophagy of intracellular Salmonella Typhimurium. Cell
the Sobek Foundation (H.N. and C.M.), Fondation Acteria (H.N. and C.M.), the
Host Microbe 12, 778790 (2012).
Swiss Vaccine Research Institute (H.N. and C.M.) and the Swiss National Science 31. Manzanillo, P.S. et al. The ubiquitin ligase parkin mediates resistance to intracellular
Foundation (310030_143979 and CRSII3_136241 for H.N. and C.M.). pathogens. Nature 501, 512516 (2013).
32. Narendra, D., Tanaka, A., Suen, D.-F. & Youle, R.J. Parkin is recruited selectively to
COMPETING FINANCIAL INTERESTS impaired mitochondria and promotes their autophagy. J. Cell Biol. 183, 795803
The authors declare no competing financial interests. (2008).
33. Dupont, N. et al. Shigella phagocytic vacuolar membrane remnants participate in
Reprints and permissions information is available online at http://www.nature.com/ the cellular response to pathogen invasion and are regulated by autophagy. Cell Host
reprints/index.html. Microbe 6, 137149 (2009).
npg

34. Kobayashi, S. et al. Artificial induction of autophagy around polystyrene beads in


nonphagocytic cells. Autophagy 6, 3645 (2010).
1. Mizushima, N. & Komatsu, M. Autophagy: renovation of cells and tissues. Cell 147, 35. Fujita, N. et al. Recruitment of the autophagic machinery to endosomes during
728741 (2011). infection is mediated by ubiquitin. J. Cell Biol. 203, 115128 (2013).
2. Clark, S.L. Jr. Cellular differentiation in the kidneys of newborn mice studies with Study demonstrating that ubiquitination is required for the recruitment of autophagy-
the electron microscope. J. Biophys. Biochem. Cytol. 3, 349362 (1957). related proteins to ruptured endosomes, and clarifying the mechanism for the recruit-
3. Novikoff, A.B. The proximal tubule cell in experimental hydronephrosis. J. Biophys. ment of ATG16L1 to ubiquitinated endosomes.
Biochem. Cytol. 6, 136138 (1959). 36. Hung, Y.-H., Chen, L.M.-W., Yang, J.-Y. & Yuan Yang, W. Spatiotemporally con-
4. Ohsumi, Y. Historical landmarks of autophagy research. Cell Res. 24, 923 (2014). trolled induction of autophagy-mediated lysosome turnover. Nat. Commun. 4, 2111
5. Itakura, E. & Mizushima, N. Characterization of autophagosome formation site by a (2013).
hierarchical analysis of mammalian Atg proteins. Autophagy 6, 764776 (2010). 37. Maejima, I. et al. Autophagy sequesters damaged lysosomes to control lysosomal
6. Kageyama, S. et al. The LC3 recruitment mechanism is separate from Atg9L1- biogenesis and kidney injury. EMBO J. 32, 23362347 (2013).
dependent membrane formation in the autophagic response against Salmonella. 38. Thurston, T.L.M., Wandel, M.P., von Muhlinen, N., Foeglein, A. & Randow, F. Galectin
Mol. Biol. Cell 22, 22902300 (2011). 8 targets damaged vesicles for autophagy to defend cells against bacterial invasion.
Study demonstrating that early ATG proteins are recruited to intracellular Salmonella. Nature 482, 414418 (2012).
7. Matsunaga, K. et al. Autophagy requires endoplasmic reticulum targeting of the Study identifying galectin-8 as an ubiquitin-independent mediator of the recruitment
PI3-kinase complex via Atg14L. J. Cell Biol. 190, 511521 (2010). of NDP52 to ruptured endosomes.
8. Hamasaki, M. et al. Autophagosomes form at ER-mitochondria contact sites. Nature 39. Dreux, M. & Chisari, F.V. Viruses and the autophagy machinery. Cell Cycle 9, 1295
495, 389393 (2013). 1307 (2010).
Study identifying ER-mitochondria contact sites as the initiation sites for autophago- 40. Yordy, B., Tal, M.C., Hayashi, K., Arojo, O. & Iwasaki, A. Autophagy and selective
some formation. deployment of Atg proteins in antiviral defense. Int. Immunol. 25, 110 (2013).
9. Dooley, H.C. et al. WIPI2 links LC3 conjugation with PI3P, autophagosome forma- 41. Liang, X.H. et al. Protection against fatal Sindbis virus encephalitis by beclin, a novel
tion, and pathogen clearance by recruiting Atg125-16L1. Mol. Cell 55, 238252 Bcl-2-interacting protein. J. Virol. 72, 85868596 (1998).
(2014). 42. Orvedahl, A. et al. Autophagy protects against Sindbis virus infection of the central
10. Mizushima, N. et al. Dissection of autophagosome formation using Apg5-deficient nervous system. Cell Host Microbe 7, 115127 (2010).
mouse embryonic stem cells. J. Cell Biol. 152, 657668 (2001). 43. Orvedahl, A. et al. Image-based genome-wide siRNA screen identifies selective
11. Nakatogawa, H., Suzuki, K., Kamada, Y. & Ohsumi, Y. Dynamics and diversity in autoph- autophagy factors. Nature 480, 113117 (2011).
agy mechanisms: lessons from yeast. Nat. Rev. Mol. Cell Biol. 10, 458467 (2009). 44. Kyei, G.B. et al. Autophagy pathway intersects with HIV-1 biosynthesis and regulates
12. Fujita, N. et al. Differential involvement of Atg16L1 in Crohn disease and canoni- viral yields in macrophages. J. Cell Biol. 186, 255268 (2009).
cal autophagy: analysis of the organization of the Atg16L1 complex in fibroblasts. 45. Shoji-Kawata, S. et al. Identification of a candidate therapeutic autophagy-inducing
J. Biol. Chem. 284, 3260232609 (2009). peptide. Nature 494, 201206 (2013).

1022 VOLUME 16 NUMBER 10 OCTOBER 2015 NATURE IMMUNOLOGY


REVIEW

46. Martinez, J. et al. Microtubule-associated protein 1 light chain 3 a (LC3)-associated 79. Netea, M.G., van de Veerdonk, F.L., van der Meer, J.W.M., Dinarello, C.A. & Joosten,
phagocytosis is required for the efficient clearance of dead cells. Proc. Natl. Acad. L.A.B. Inflammasome-independent regulation of IL-1-family cytokines. Annu. Rev.
Sci. USA 108, 1739617401 (2011). Immunol. 33, 4977 (2014).
47. Martinez, J. et al. Molecular characterization of LC3-associated phagocytosis reveals 80. Castillo, E.F. et al. Autophagy protects against active tuberculosis by suppressing
distinct roles for Rubicon, NOX2 and autophagy proteins. Nat. Cell Biol. 17, 893906 bacterial burden and inflammation. Proc. Natl. Acad. Sci. USA 109, E3168E3176
(2015). (2012).
48. Henault, J. et al. Noncanonical autophagy is required for type I interferon secretion 81. Kawai, T. & Akira, S. The role of pattern-recognition receptors in innate immunity:
in response to DNA-immune complexes. Immunity 37, 986997 (2012). update on Toll-like receptors. Nat. Immunol. 11, 373384 (2010).
49. Starr, T. et al. Selective subversion of autophagy complexes facilitates completion of 82. Yoneyama, M., Onomoto, K., Jogi, M., Akaboshi, T. & Fujita, T. Viral RNA detection
the Brucella intracellular cycle. Cell Host Microbe 11, 3345 (2012). by RIG-I-like receptors. Curr. Opin. Immunol. 32, 4853 (2015).
50. Zhao, Z. et al. Autophagosome-independent essential function for the autophagy 83. Cai, X., Chiu, Y.-H. & Chen, Z.J. The cGAS-cGAMP-STING pathway of cytosolic DNA
protein Atg5 in cellular immunity to intracellular pathogens. Cell Host Microbe 4, sensing and signaling. Mol. Cell 54, 289296 (2014).
458469 (2008). 84. Lee, H.K., Lund, J.M., Ramanathan, B., Mizushima, N. & Iwasaki, A. Autophagy-
51. Hwang, S. et al. Nondegradative role of Atg5-Atg12/ Atg16L1 autophagy protein dependent viral recognition by plasmacytoid dendritic cells. Science 315, 1398
complex in antiviral activity of interferon g. Cell Host Microbe 11, 397409 (2012). 1401 (2007).
52. Taylor, M.P., Burgon, T.B., Kirkegaard, K. & Jackson, W.T. Role of microtubules in First study to report involvement of ATG proteins in PRR-mediated innate immune
extracellular release of poliovirus. J. Virol. 83, 65996609 (2009). response.
53. Robinson, S.M. et al. Coxsackievirus B exits the host cell in shed microvesicles 85. Tal, M.C. et al. Absence of autophagy results in reactive oxygen species-dependent
displaying autophagosomal markers. PLoS Pathog. 10, e1004045 (2014). amplification of RLR signaling. Proc. Natl. Acad. Sci. USA 106, 27702775
54. Miao, Y., Li, G., Zhang, X., Xu, H. & Abraham, S.N. A TRP channel senses lyso- (2009).
some neutralization by pathogens to trigger their expulsion. Cell 161, 13061319 86. Jounai, N. et al. The Atg5 Atg12 conjugate associates with innate antiviral immune
(2015). responses. Proc. Natl. Acad. Sci. USA 104, 1405014055 (2007).
55. Kawai, T. & Akira, S. The roles of TLRs, RLRs and NLRs in pathogen recognition. 87. Lei, Y. et al. The mitochondrial proteins NLRX1 and TUFM form a complex that
Int. Immunol. 21, 317337 (2009). regulates type I interferon and autophagy. Immunity 36, 933946 (2012).
56. Deretic, V., Saitoh, T. & Akira, S. Autophagy in infection, inflammation and immunity. 88. Saitoh, T. et al. Atg9a controls dsDNA-driven dynamic translocation of STING and the
Nat. Rev. Immunol. 13, 722737 (2013). innate immune response. Proc. Natl. Acad. Sci. USA 106, 2084220846 (2009).
57. Latz, E., Xiao, T.S. & Stutz, A. Activation and regulation of the inflammasomes. Nat. First study to report involvement of ATG proteins in cytosolic dsDNAinduced innate
Rev. Immunol. 13, 397411 (2013). immune response.
2015 Nature America, Inc. All rights reserved.

58. Hampe, J. et al. A genome-wide association scan of nonsynonymous SNPs identifies 89. Konno, H., Konno, K. & Barber, G.N. Cyclic dinucleotides trigger ULK1 (ATG1)
a susceptibility variant for Crohn disease in ATG16L1. Nat. Genet. 39, 207211 phosphorylation of STING to prevent sustained innate immune signaling. Cell 155,
(2007). 688698 (2013).
59. Rioux, J.D. et al. Genome-wide association study identifies new susceptibility loci 90. Liang, Q. et al. Crosstalk between the cGAS DNA sensor and Beclin-1 autophagy pro-
for Crohn disease and implicates autophagy in disease pathogenesis. Nat. Genet. tein shapes innate antimicrobial immune responses. Cell Host Microbe 15, 228238
39, 596604 (2007). (2014).
60. Saitoh, T. et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced 91. Watson, R.O., Manzanillo, P.S. & Cox, J.S. Extracellular M. tuberculosis DNA tar-
IL-1b production. Nature 456, 264268 (2008). gets bacteria for autophagy by activating the host DNA-sensing pathway. Cell 150,
First study to report that autophagy-deficiency enhances inflammasome activation. 803815 (2012).
61. Murthy, A. et al. A Crohns disease variant in Atg16l1 enhances its degradation by 92. Dey, B. et al. A bacterial cyclic dinucleotide activates the cytosolic surveillance
caspase 3. Nature 506, 456462 (2014). pathway and mediates innate resistance to tuberculosis. Nat. Med. 21, 401406
Study providing evidence that the Crohns diseaseassociated variant of ATG16L1 (2015).
enhances inflammation in vivo. 93. Collins, A.C. et al. Cyclic GMP-AMP synthase is an innate immune DNA sensor for
62. Lassen, K.G. et al. Atg16L1 T300A variant decreases selective autophagy resulting Mycobacterium tuberculosis. Cell Host Microbe 17, 820828 (2015).
in altered cytokine signaling and decreased antibacterial defense. Proc. Natl. Acad. 94. Blum, J.S., Wearsch, P.A. & Cresswell, P. Pathways of antigen processing. Annu. Rev.
Sci. USA 111, 77417746 (2014). Immunol. 31, 443473 (2013).
63. Plantinga, T.S. et al. Crohns disease-associated ATG16L1 polymorphism modulates 95. Romao, S., Gannage, M. & Mnz, C. Checking the garbage bin for problems in the
pro-inflammatory cytokine responses selectively upon activation of NOD2. Gut 60, house, or how autophagy assists in antigen presentation to the immune system.
12291235 (2011). Semin. Cancer Biol. 23, 391396 (2013).
64. Cadwell, K. et al. Virus-plus-susceptibility gene interaction determines Crohns dis- 96. Paludan, C. et al. Endogenous MHC class II processing of a viral nuclear antigen
ease gene Atg16L1 phenotypes in intestine. Cell 141, 11351145 (2010). after autophagy. Science 307, 593596 (2005).
65. Zhou, R., Yazdi, A.S., Menu, P. & Tschopp, J. A role for mitochondria in NLRP3 97. Leung, C.S., Haigh, T.A., Mackay, L.K., Rickinson, A.B. & Taylor, G.S. Nuclear
inflammasome activation. Nature 469, 221225 (2011). location of an endogenously expressed antigen, EBNA1, restricts access to macro
First study to report that autophagy targets old or dysfunctional mitochondria to limit autophagy and the range of CD4 epitope display. Proc. Natl. Acad. Sci. USA 107,
inflammasome activation. 21652170 (2010).
npg

66. Nakahira, K. et al. Autophagy proteins regulate innate immune responses by inhibit- 98. Jagannath, C. et al. Autophagy enhances the efficacy of BCG vaccine by increasing
ing the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat. peptide presentation in mouse dendritic cells. Nat. Med. 15, 267276 (2009).
Immunol. 12, 222230 (2011). 99. Schmid, D., Pypaert, M. & Mnz, C. Antigen-loading compartments for major histo-
67. Lupfer, C. et al. Receptor interacting protein kinase 2-mediated mitophagy regu- compatibility complex class II molecules continuously receive input from autopha-
lates inflammasome activation during virus infection. Nat. Immunol. 14, 480488 gosomes. Immunity 26, 7992 (2007).
(2013). Study demonstrating that autophagosomes deliver cytosolic antigen to MHC class
68. Wen, H. et al. Fatty acid-induced NLRP3-ASC inflammasome activation interferes II loading compartments.
with insulin signaling. Nat. Immunol. 12, 408415 (2011). 100. Jin, Y. et al. Regulation of SIV antigen-specific CD4+ T cellular immunity via autopha-
69. Razani, B. et al. Autophagy links inflammasomes to atherosclerotic progression. Cell gosome-mediated MHC II molecule-targeting antigen presentation in mice. PLoS
Metab. 15, 534544 (2012). ONE 9, e93143 (2014).
70. Wang, C. et al. Atg16L1 deficiency confers protection from uropathogenic Escherichia 101. Nedjic, J., Aichinger, M., Emmerich, J., Mizushima, N. & Klein, L. Autophagy in
coli infection in vivo. Proc. Natl. Acad. Sci. USA 109, 1100811013 (2012). thymic epithelium shapes the T-cell repertoire and is essential for tolerance. Nature
71. Shaw, S.Y. et al. Selective modulation of autophagy, innate immunity, and adaptive 455, 396400 (2008).
immunity by small molecules. ACS Chem. Biol. 8, 27242733 (2013). 102. Kasai, M. et al. Autophagic compartments gain access to the MHC class II compart-
72. Guo, W. et al. Small molecule-driven mitophagy-mediated NLRP3 inflammasome ments in thymic epithelium. J. Immunol. 183, 72787285 (2009).
inhibition is responsible for the prevention of colitis-associated cancer. Autophagy 103. Aichinger, M., Wu, C., Nedjic, J. & Klein, L. Macroautophagy substrates are loaded
10, 972985 (2014). onto MHC class II of medullary thymic epithelial cells for central tolerance. J. Exp.
73. Abderrazak, A. et al. Anti-inflammatory and anti-atherogenic effects of the inflam- Med. 210, 287300 (2013).
masome NLRP3 inhibitor, arglabin, in ApoE2.Ki mice fed a high fat diet. Circulation Study demonstrating that autophagic cargo is presented on MHC class II molecules
131, 10611070 (2015). for negative selection of CD4+ T cells in the thymus.
74. Shi, C.-S. et al. Activation of autophagy by inflammatory signals limits IL-1b produc- 104. Florey, O., Kim, S.E., Sandoval, C.P., Haynes, C.M. & Overholtzer, M. Autophagy
tion by targeting ubiquitinated inflammasomes for destruction. Nat. Immunol. 13, machinery mediates macroendocytic processing and entotic cell death by targeting
255263 (2012). single membranes. Nat. Cell Biol. 13, 13351343 (2011).
75. Harris, J. et al. Autophagy controls IL-1b secretion by targeting pro-IL-1b for degrada- 105. Romao, S. et al. Autophagy proteins stabilize pathogen-containing phagosomes for
tion. J. Biol. Chem. 286, 95879597 (2011). prolonged MHC II antigen processing. J. Cell Biol. 203, 757766 (2013).
76. Dupont, N. et al. Autophagy-based unconventional secretory pathway for extracellular Study demonstrating that LAP retains endocytosed antigen for prolonged presentation
delivery of IL-1b. EMBO J. 30, 47014711 (2011). by MHC class II to CD4+ T cells.
77. Lamkanfi, M. & Dixit, V.M. Mechanisms and functions of inflammasomes. Cell 157, 106. Ma, J., Becker, C., Lowell, C.A. & Underhill, D.M. Dectin-1-triggered recruitment
10131022 (2014). of light chain 3 protein to phagosomes facilitates major histocompatibility complex
78. Meunier, E. et al. Caspase-11 activation requires lysis of pathogen-containing vacu- class II presentation of fungal-derived antigens. J. Biol. Chem. 287, 3414934156
oles by IFN-induced GTPases. Nature 509, 366370 (2014). (2012).

NATURE IMMUNOLOGY VOLUME 16 NUMBER 10 OCTOBER 2015 1023


REVIEW

107. Ma, J., Becker, C., Reyes, C. & Underhill, D.M. Cutting edge: FYCO1 recruitment to 137. Farfariello, V., Amantini, C. & Santoni, G. Transient receptor potential vanilloid 1
dectin-1 phagosomes is accelerated by light chain 3 protein and regulates phagosome activation induces autophagy in thymocytes through ROS-regulated AMPK and Atg4C
maturation and reactive oxygen production. J. Immunol. 192, 13561360 (2014). pathways. J. Leukoc. Biol. 92, 421431 (2012).
108. Lee, H.K. et al. In vivo requirement for Atg5 in antigen presentation by dendritic 138. Willinger, T. & Flavell, R.A. Canonical autophagy dependent on the class III phos-
cells. Immunity 32, 227239 (2010). phoinositide-3 kinase Vps34 is required for naive T-cell homeostasis. Proc. Natl.
109. Ireland, J.M. & Unanue, E.R. Autophagy in antigen-presenting cells results in pre- Acad. Sci. USA 109, 86708675 (2012).
sentation of citrullinated peptides to CD4 T cells. J. Exp. Med. 208, 26252632 139. Henson, S.M. et al. p38 signaling inhibits mTORC1-independent autophagy in senes-
(2011). cent human CD8+ T cells. J. Clin. Invest. 124, 40044016 (2014).
110. Chaturvedi, A., Dorward, D. & Pierce, S.K. The B cell receptor governs the subcellular 140. Jia, W., Pua, H.H., Li, Q.-J. & He, Y.-W. Autophagy regulates endoplasmic reticu-
location of Toll-like receptor 9 leading to hyperresponses to DNA-containing antigens. lum homeostasis and calcium mobilization in T lymphocytes. J. Immunol. 186,
Immunity 28, 799809 (2008). 15641574 (2011).
111. Pengo, N. et al. Plasma cells require autophagy for sustainable immunoglobulin 141. Parekh, V.V. et al. Impaired autophagy, defective T cell homeostasis, and a wast-
production. Nat. Immunol. 14, 298305 (2013). ing syndrome in mice with a T cell-specific deletion of Vps34. J. Immunol. 190,
Study demonstrating that plasma cells require autophagy for their antibody-producing 50865101 (2013).
function. 142. Kovacs, J.R. et al. Autophagy promotes T-cell survival through degradation of proteins
112. Wenger, T. et al. Autophagy inhibition promotes defective neosynthesized proteins of the cell death machinery. Cell Death Differ. 19, 144152 (2012).
storage in ALIS, and induces redirection toward proteasome processing and MHCI- 143. Puleston, D. J. et al. Autophagy is a critical regulator of memory CD8+ T cell forma-
restricted presentation. Autophagy 8, 350363 (2012). tion. eLife 3, e03706 (2014).
113. Kristensen, A.R. et al. Ordered organelle degradation during starvation-induced 144. Xu, X. et al. Autophagy is essential for effector CD8+ T cell survival and memory
autophagy. Mol. Cell. Proteomics 7, 24192428 (2008). formation. Nat. Immunol. 15, 11521161 (2014).
114. Cuervo, A.M., Palmer, A., Rivett, A.J. & Knecht, E. Degradation of proteasomes by 145. Miller, B.C. et al. The autophagy gene ATG5 plays an essential role in B lymphocyte
lysosomes in rat liver. Eur. J. Biochem. 227, 792800 (1995). development. Autophagy 4, 309314 (2008).
115. Marshall, R.S., Li, F., Gemperline, D.C., Book, A.J. & Vierstra, R.D. Autophagic 146. Conway, K.L. et al. ATG5 regulates plasma cell differentiation. Autophagy 9, 528537
degradation of the 26S proteasome is mediated by the dual ATG8/ubiquitin receptor (2013).
RPN10 in Arabidopsis. Mol. Cell 58, 10531066 (2015). 147. Milan, E. et al. A plastic SQSTM1/p62-dependent autophagic reserve maintains
116. English, L. et al. Autophagy enhances the presentation of endogenous viral antigens proteostasis and determines proteasome inhibitor susceptibility in multiple myeloma
on MHC class I molecules during HSV-1 infection. Nat. Immunol. 10, 480487 cells. Autophagy 11, 11611178 (2015).
(2009). 148. Chen, M. et al. Essential role for autophagy in the maintenance of immunological
2015 Nature America, Inc. All rights reserved.

117. Tey, S.-K. & Khanna, R. Autophagy mediates transporter associated with antigen memory against influenza infection. Nat. Med. 20, 503510 (2014).
processing-independent presentation of viral epitopes through MHC class I pathway. Study demonstrating that the maintenance of memory B cells requires autophagy.
Blood 120, 9941004 (2012). 149. Benjamin, J.L., Sumpter, R. Jr., Levine, B. & Hooper, L.V. Intestinal epithelial
Study demonstrating that autophagy can contribute to the loading of antigen onto autophagy is essential for host defense against invasive bacteria. Cell Host Microbe
MHC class I molecules under TAP-blocking conditions. 13, 723734 (2013).
118. Li, B. et al. Autophagy facilitates major histocompatibility complex class I expres- 150. Conway, K.L. et al. Atg16l1 is required for autophagy in intestinal epithelial cells and
sion induced by IFN-g in B16 melanoma cells. Cancer Immunol. Immunother. 59, protection of mice from Salmonella infection. Gastroenterology 145, 13471357
313321 (2010). (2013).
119. Johnstone, C. et al. Exogenous, TAP-independent lysosomal presentation of a respira- 151. Yano, T. et al. Autophagic control of listeria through intracellular innate immune
tory syncytial virus CTL epitope. Immunol. Cell Biol. 90, 978982 (2012). recognition in Drosophila. Nat. Immunol. 9, 908916 (2008).
120. Blanchet, F.P. et al. Human immunodeficiency Virus-1 inhibition of immunoam- 152. Shelly, S., Lukinova, N., Bambina, S., Berman, A. & Cherry, S. Autophagy is an essen-
phisomes in dendritic cells impairs early innate and adaptive immune responses. tial component of Drosophila immunity against vesicular stomatitis virus. Immunity
Immunity 32, 654669 (2010). 30, 588598 (2009).
121. Fiegl, D. et al. Amphisomal route of MHC class I cross-presentation in bacteria- 153. Jia, K. et al. Autophagy genes protect against Salmonella typhimurium infection and
infected dendritic cells. J. Immunol. 190, 27912806 (2013). mediate insulin signaling-regulated pathogen resistance. Proc. Natl. Acad. Sci. USA
122. De Luca, A. et al. CD4+ T cell vaccination overcomes defective cross-presentation of 106, 1456414569 (2009).
fungal antigens in a mouse model of chronic granulomatous disease. J. Clin. Invest. 154. Cadwell, K. et al. A key role for autophagy and the autophagy gene Atg16l1 in mouse
122, 18161831 (2012). and human intestinal Paneth cells. Nature 456, 259263 (2008).
123. Duran, J.M., Anjard, C., Stefan, C., Loomis, W.F. & Malhotra, V. Unconventional secre- Study demonstrating that ATG16L1 and ATG5 are required for maintenance of the
tion of Acb1 is mediated by autophagosomes. J. Cell Biol. 188, 527536 (2010). granule-secretion pathway in Paneth cells in the intestine.
124. Manjithaya, R., Anjard, C., Loomis, W.F. & Subramani, S. Unconventional secretion 155. Marchiando, A.M. et al. A deficiency in the autophagy gene Atg16L1 enhances
of Pichia pastoris Acb1 is dependent on GRASP protein, peroxisomal functions, and resistance to enteric bacterial infection. Cell Host Microbe 14, 216224 (2013).
autophagosome formation. J. Cell Biol. 188, 537546 (2010). 156. Xu, Y. et al. Toll-like receptor 4 is a sensor for autophagy associated with innate
Two studies (refs. 123,124) establishing the fact that the autophagic machinery immunity. Immunity 27, 135144 (2007).
contributes to signal peptideindependent secretion. 157. Delgado, M.A., Elmaoued, R.A., Davis, A.S., Kyei, G. & Deretic, V. Toll-like receptors
npg

125. Thorburn, J. et al. Autophagy regulates selective HMGB1 release in tumor cells that control autophagy. EMBO J. 27, 11101121 (2008).
are destined to die. Cell Death Differ. 16, 175183 (2009). 158. Travassos, L.H. et al. Nod1 and Nod2 direct autophagy by recruiting ATG16L1
126. Martins, I. et al. Molecular mechanisms of ATP secretion during immunogenic cell to the plasma membrane at the site of bacterial entry. Nat. Immunol. 11, 5562
death. Cell Death Differ. 21, 7991 (2014). (2010).
127. DeSelm, C.J. et al. Autophagy proteins regulate the secretory component of osteo- 159. Cooney, R. et al. NOD2 stimulation induces autophagy in dendritic cells influencing
clastic bone resorption. Dev. Cell 21, 966974 (2011). bacterial handling and antigen presentation. Nat. Med. 16, 9097 (2010).
128. Jackson, W.T. et al. Subversion of cellular autophagosomal machinery by RNA viruses. 160. Jounai, N. et al. NLRP4 negatively regulates autophagic processes through an asso-
PLoS Biol. 3, e156 (2005). ciation with Beclin1. J. Immunol. 186, 16461655 (2011).
129. Nowag, H. et al. Macroautophagy proteins assist Epstein Barr virus production and 161. Yanai, H. et al. Conditional ablation of HMGB1 in mice reveals its protective func-
Get incorporated into the virus particles. EBioMedicine 1, 116125 (2014). tion against endotoxemia and bacterial infection. Proc. Natl. Acad. Sci. USA 110,
130. Uhl, M. et al. Autophagy within the antigen donor cell facilitates efficient antigen 2069920704 (2013).
cross-priming of virus-specific CD8+ T cells. Cell Death Differ. 16, 9911005 (2009). 162. Tang, D. et al. Endogenous HMGB1 regulates autophagy. J. Cell Biol. 190, 881892
131. Li, Y. et al. Efficient cross-presentation depends on autophagy in tumor cells. Cancer (2010).
Res. 68, 68896895 (2008). 163. Espert, L. et al. Autophagy is involved in T cell death after binding of HIV-1 envelope
132. Ye, W. et al. Cross-presentation of viral antigens in dribbles leads to efficient activa- proteins to CXCR4. J. Clin. Invest. 116, 21612172 (2006).
tion of virus-specific human memory T cells. J. Transl. Med. 12, 100 (2014). 164. Singh, S.B., Davis, A.S., Taylor, G.A. & Deretic, V. Human IRGM induces autophagy
133. Mortensen, M. et al. The autophagy protein Atg7 is essential for hematopoietic stem to eliminate intracellular mycobacteria. Science 313, 14381441 (2006).
cell maintenance. J. Exp. Med. 208, 455467 (2011). 165. Keller, C.W. et al. TNF-a induces macroautophagy and regulates MHC class II expres-
Study demonstrating which hematopoietic lineages requires autophagy for their sion in human skeletal muscle cells. J. Biol. Chem. 286, 39703980 (2011).
development. 166. Mostowy, S. et al. p62 and NDP52 proteins target intracytosolic Shigella and Listeria
134. Pua, H.H., Dzhagalov, I., Chuck, M., Mizushima, N. & He, Y.-W. A critical role for the to different autophagy pathways. J. Biol. Chem. 286, 2698726995 (2011).
autophagy gene Atg5 in T cell survival and proliferation. J. Exp. Med. 204, 2531 167. Pilli, M. et al. TBK-1 promotes autophagy-mediated antimicrobial defense by control-
(2007). ling autophagosome maturation. Immunity 37, 223234 (2012).
135. Arsov, I. et al. A role for autophagic protein beclin 1 early in lymphocyte development. 168. Harris, J. et al. T helper 2 cytokines inhibit autophagic control of intracellular
J. Immunol. 186, 22012209 (2011). Mycobacterium tuberculosis. Immunity 27, 505517 (2007).
136. Pua, H.H., Guo, J., Komatsu, M. & He, Y.-W. Autophagy is essential for mitochondrial 169. Shen, S. et al. Cytoplasmic STAT3 represses autophagy by inhibiting PKR activity.
clearance in mature T lymphocytes. J. Immunol. 182, 40464055 (2009). Mol. Cell 48, 667680 (2012).

1024 VOLUME 16 NUMBER 10 OCTOBER 2015 NATURE IMMUNOLOGY


Reproduced with permission of the copyright owner. Further reproduction prohibited without
permission.

Você também pode gostar