Sunteți pe pagina 1din 16

REVIEWS

Autophagy in infection, inflammation


and immunity
Vojo Deretic1, Tatsuya Saitoh2 and Shizuo Akira2
Abstract | Autophagy is a fundamental eukaryotic pathway that has multiple
effects on immunity. Autophagy is induced by pattern recognition receptors and,
through autophagic adaptors, it provides a mechanism for the elimination of
intracellular microorganisms. Autophagy controls inflammation through regulatory
interactions with innate immune signalling pathways, by removing endogenous
inflammasome agonists and through effects on the secretion of immune mediators.
Moreover, autophagy contributes to antigen presentation and to T cell homeostasis,
and it affects T cell repertoires and polarization. Thus, as we discuss in this Review,
autophagy has multitiered immunological functions that influence infection,
inflammation and immunity.

The term autophagy (from the Greek words auto The autophagy pathway
Macroautophagy
The autophagy-related protein meaning ‘self ’ and phagein meaning ‘to eat’) refers ATG-dependent autophagy. The principal morphologi-
(ATG)-dependent pathway, to a collection of diverse processes — including cal feature of autophagy is the formation of endomem-
sequestering cargo into macroautophagy, microautophagy, chaperone-mediated branous organelles, which are known as autophagosomes
autophagosomes that fuse autophagy1 and non-canonical autophagy2 — that enable (BOX 1). An ensemble of ATG factors drives the formation
with lysosomes, in which
the cargo is degraded.
cells to digest their cytoplasmic contents in lysosomes. in the cytoplasm of the autophagic isolation membrane
Proteins, organelles (such as Macroautophagy, which is autophagy in its strictest (also known as the phagophore)1 (BOX 1). Beclin 1 (the
mitochondria) and invading form and which is the focus of this Review, depends mammalian orthologue of the yeast protein ATG-6), the
microorganisms are selectively on specialized autophagy-related proteins (ATGs)1 serine/threonine protein kinase ULK1 (the mammalian
degraded by macroautophagy.
(BOX 1) and is distinct from other cytoplasmic diges- paralogue of the yeast protein ATG-1), autophagy-
Bulk cytoplasmic autophagy
occurs as a starvation tive processes, including proteasomal degradation, as related LC3 proteins and γ-aminobutyric acid recep-
response. a result of its ability to capture and to eliminate large tor-associated proteins (GABARAPs; the mammalian
targets such as toxic protein aggregates, defunct or dis- paralogues of the yeast ATG-8 protein) are key regula-
used organelles and invading microorganisms. Several tors of phagophore formation1 (BOX 1). A phagophore,
autophagy-related factors have also been recognized to which is often depicted in models as an isolated crescent
have autophagy-independent immune functions3, but in the cytoplasm, is transiently connected to and derived
1
Department of Molecular these are not discussed in this Review. from phosphatidylinositol-3‑phosphate (PtdIns3P)-
Genetics and Microbiology, Autophagy may be the primordial form of eukary- positive domains of the endoplasmic reticulum (ER),
University of New Mexico otic innate immunity against invading microorganisms. which are known as omegasomes 1. Other cellular
Health Sciences Center, 915
Camino de Salud, North East,
Our understanding of the immunological functions compartments, such as the Golgi apparatus, the mito-
Albuquerque, New Mexico of autophagy has dramatically increased in recent chondria and the plasma membrane-derived endocytic
87131, USA. years4, and it is now appreciated that, in mammals, organelles, also contribute to phagophore formation1.
2
Laboratory of Host Defense, these primordial functions of autophagy have evolved Recent studies suggest that the ER‑derived autophago-
World Premier International
and have been incorporated into multiple innate and somes form at ER–mitochondria contact sites5. A pha-
Immunology Frontier
Research Center, Osaka adaptive immune pathways. In this Review, we dis- gophore sequesters the captured cytoplasmic cargo
University, 3–1 Yamada-oka, cuss the four main roles of autophagy in immunity: that is destined for autophagic disposal and, following
Suita, Osaka 565–0871, the direct elimination of microorganisms, the control elongation and closure, an autophagosome is formed.
Japan. of inflammation, the control of adaptive immunity At this stage, the corresponding endomembranes are
Correspondence to V.D.
e‑mail:
through the regulation of antigen presentation and commonly visualized as double membrane structures1.
vderetic@salud.unm.edu lymphocyte homeostasis, and the secretion of immune The degradation of the captured cargo begins when the
doi:10.1038/nri3532 mediators (FIG. 1). double membrane autophagosome matures into a single

722 | O CTOBER 2013 | VOLUME 13 www.nature.com/reviews/immunol

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 1 | The autophagy pathway


Autophagy is controlled by the regulatory serine/threonine protein kinases ULK1 and ULK2 (which are the mammalian homologues of the yeast
autophagy-related protein 1 (ATG‑1)) and by the lipid kinase activity of the class III phosphatidylinositol-3‑phosphate kinase (PtdIns3K) VPS34 (VPS‑34 in
yeast), which forms a complex with beclin 1 (which is the mammalian orthologue of yeast ATG‑6) and ATG14‑like protein (ATG14L). ULK1, ULK2 and the
VSP34–beclin 1–ATG14L complex integrate upstream signals. These factors, which are engaged in the activation of autophagy during nutritional and
immune responses, induce the downstream ATG conjugation cascade1. This involves the association of the ATG5–ATG12 conjugate with ATG16L1. The
ATG5–ATG12–ATG16L1 complex facilitates the addition of a phosphatidylethanolamine group to the carboxyl terminus of the mammalian paralogues
of ATG-8: LC3A, LC3B (which has been used as a marker for the identification
of autophagosomes in mammalian cells), LC3C, γ-aminobutyric acid Donor membrane
receptor-associated protein (GABARAP), GABARAP-like 1 (GABARAPL1) • ER omegasome
and GABARAPL2 (REF. 1). Lipidated mammalian paralogues of ATG-8 function • Golgi apparatus
together with other factors to assemble, elongate and lead to the closure • ER–mitochondria contact sites
• Endosomes
of nascent autophagic organelles1 (see the figure). The lipid kinase VPS34, • Plasma membrane
through its production of PtdIns3 phosphate (PtdIns3P), has a dual function in
autophagy1. During initiation, VPS34‑derived PtdIns P is recognized by the Initiation
3
PtdIns3P-binding factors WD repeat domain phosphoinositide-interacting Isolation
ULK1 ATG16L1
protein 1 (WIPI1)–WIPI4 (which are the mammalian paralogues of yeast membrane
ATG‑18), and these factors cooperate with ATG2 and ATG9 to form the Beclin 1 VPS34 ATG5 ATG12
phagophore1. During the final maturation of autophagosomes into LC3
ATG14L
autolysosomes, the attachment of a tail-anchored SNARE, syntaxin 17 LC3A GABARAP
(REF. 6), to the autophagosomal membrane enables fusion with lysosomes.
WIPI ATG2 Elongation LC3B GABARAPL1
At this stage, the VPS34–beclin 1 complex contains ultraviolet radiation
ATG9 and closure
resistance‑associated gene protein (UVRAG) instead of ATG14L1. LC3C GABARAPL2
Autophagy requires the rapid remodelling of endomembranes in the
cytoplasm, which involves the formation of transient PtdIns3P-positive
structures (known as omegasomes) on the endoplasmic reticulum (ER), Autophagosome
the ER–mitochondria contact sites, the Golgi apparatus and the plasma Lysosome
membrane-derived endosomal organelles. This happens independently of
transcriptional changes. However, transcriptional adjustments are necessary Syntaxin 17
for sustained autophagy, for instance, to replenish ‘consumables’ such as LC3 Fusion
and autophagic adaptors including sequestosome 1 (also known as p62), which Maturation
are degraded along with the captured cargo in autolysosomes. Transcriptional
adjustments are accomplished by transcription-positive regulators (for Beclin 1 VPS34
example, transcription factor EB (TFEB), forkhead box protein O3A (FOXO3A) UVRAG
Autolysosome
and NFE2‑related factor 2 (NRF2)) and transcription-negative regulators (for HOPS
example, zinc finger protein with KRAB and SCAN domains 3 (ZKSCAN3; also
known as ZNF306), which coordinate autophagy with lysosomal, proteasomal,
lipolytic and oxidative stress response systems138,139.
Degradation
HOPS, homotypic fusion and vacuole protein sorting.
Nature Reviews | Immunology

membrane-delimited autolysosome. Autophagosomal The ancient defence mechanism of autophagy has


maturation is induced by the translocation of the SNARE been well integrated with other immune-sensing sys-
protein syntaxin 17 (REF. 6) to the outer membrane of tems. Pattern recognition receptor (PRR) signalling
the completed autophagosome. This facilitates fusion that is induced following the recognition of pathogen-
of the autophagosome with lysosomes5,6 and results associated molecular patterns (PAMPs) and damage-
in the acidification of the autophagosome lumen, the associated molecular patterns (DAMPs) can activate
acquisition of lysosomal hydrolases and the degradation autophagy (BOX 2); for example, Toll-like receptor 4
Microautophagy of the cargo as well as the inner of the two membranes1. (TLR4) signalling leads to ubiquitylation of the ATG
Lysosomal import and beclin 1 by the E3 ligase TNF receptor-associated fac-
degradation of small Immune signalling and autophagy. A deficiency of tor 6 (TRAF6)9. Ubiquitylated beclin 1 is released from
portions of the cytoplasm
that is independent of
intracellular nutrients because of competition from its inhibitor B cell lymphoma 2 (BCL‑2) and becomes
autophagy-related proteins invading microorganisms is likely to have been one active9. TRAF6 also activates ULK1 through ubiquityla-
(ATGs), and that is often of the primordial danger signals that was available tion10 and thereby controls both key pathways that lead
manifested as invaginations of to eukaryotic cells to detect microbial invasion and to to autophagy (BOX 1). TRAF6, ubiquitylated ULK1 and
the lysososomal membrane
eliminate pathogens through autophagy. Consistent the IκB kinase (IKK) regulators TAK1‑binding protein 2
into the lysosomal lumen.
with this idea, metabolic signalling downstream of (TAB2) and TAB3 also have roles in starvation-induced
Chaperone-mediated amino acid starvation has been associated with anti- autophagy (BOX 2), which supports the idea that a simi-
autophagy microbial autophagy in response to bacterial7 and viral lar circuitry, with some variations, becomes induced
The import and degradation of infection8 (FIG. 1a). Metabolic regulation by mammalian by nutritional and innate immune signals. In BOX 2,
soluble cytoplasmic proteins
target of rapamycin (mTOR), which inhibits autophagy, we propose a model by which immune and nutritional
by chaperone-dependent
direct translocation across and AMP-activated protein kinase (AMPK), which acti- triggers converge in common signalling pathways to
the lysosomal membrane. vates autophagy, has been covered in detail elsewhere1. activate autophagy.

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | O CTOBER 2013 | 723

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

a Elimination of microorganisms b Control of pro-inflammatory signalling


Bacteria Virus Virus

Ubiquitin DAMPs ATG12


K+ efflux RIG-I
IPS1 ATG5
Amino acid TLR
starvation
Galectin Damaged
SLR
mitochondria

mTOR TRAF6 NOX2 NOD2 Viral


ssRNA
ROS ATG16L1 Leak of
lysosomal
protease ROS mtDNA
LC3 SLR

NLRP3 AIM2
inflammasome inflammasome
Endosome
Auto-
phago- Delivery of
somes PAMPs to PRRs IL-1β
IL-18
Autophagy LAP Xenophagy TLR7 or SLR
TLR8 Failing
autophagy

TRAF6 Lysosome
Lysosome

Pro-inflammatory
mediators Autolysosome
Autolysosome

c Adaptive immunity d Secretion of immune mediators


Regulated Constitutive Unconventional
Particulate
secretion secretion secretion
antigen
Cathepsin K
B cell effects (in osteoclasts) and DAMPs (e.g. IL-1β,
Conventional • HSC self renewal lysozyme (in Paneth cells) IL-6 and IL-8 IL-18 and HMGB1)
MHC class I • B1 cell development
presentation • Plasma cell survival
and IgG secretion

Cytosolic T cell effects


proteins • Thymic selection of
T cell repertoire ATGs
DICER • T cell maturation
AGO (clearance of
Citrullinated mitochondria and ER) ?
antigen • T cell survival after TCR
activation
miRNA
• Immunological synapse
destabilization
Autophagy NOD2 LAP mRNA
Secretory
Innate immune cell lysosomes
effects
Proteins • Control of activation ATGs
and release of IL-1α and
IL-1β (which influences
T cell polarization to
TH17 cell phenotype)
Proteasome Golgi apparatus
MHC
class II ER

ER
Unconventional
MHC MHC class I
class I presentation

Nature Reviews | Immunology


724 | O CTOBER 2013 | VOLUME 13 www.nature.com/reviews/immunol

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Non-canonical autophagy DAMPs, such as DNA complexes11, ATP12 and high- probably through the downstream recruitment of
Macroautophagy that mobility group box 1 protein (HMGB1)13, also activate TRAF6 and the subsequent TRAF6‑dependent ubiqui-
has been reported to occur autophagy. HMGB1 derepresses beclin 1 by displacing tylation of beclin 1 (REF. 9). T helper 1 (TH1) cell-derived
independently of one or its negative regulator BCL‑2 (BOX 2), and can also extra- cytokines, such as interferon‑γ (IFNγ), also induce
more components of the
autophagy-related protein
cellularly activate autophagy by interacting with its cell autophagy in effector cells; for example, these cytokines
(ATG) system. It should not be surface receptor RAGE (receptor for advanced glycation induce autophagy in macrophages to enable them to
confused with non-canonical end products). resist mycobacteria infection15–17. IFNγ might activate
functions of ATGs, which Inflammatory cytokines are also involved in the autophagy both through the function of immunity-
refers to the participation
activation of autophagy (BOX  2). Indeed, the induc- related GTPases15–17 and through the phosphorylation
of individual ATG factors
in processes other than
tion of autophagy by the pro-inflammatory cytokine of beclin 1 on Thr119 of its BH3 domain by death-
autophagy. interleukin‑1β (IL‑1β) is crucial for the control of associated protein kinase 1 (DAPK1). This results in
Mycobacterium tuberculosis in infected macrophages14. the dissociation of beclin 1 from its inhibitor BCL‑2
IL‑1β signals through the IL‑1 receptor (IL‑1R)14 and (REF.18) and, together with other processes (BOX 2), leads
to beclin 1 activation. In human macrophages, but not
in mouse macrophages, IFNγ cooperates with 1,25‑dihy-
droxyvitamin D3 (also known as calcitriol) to induce
◀ Figure 1 | Four principal roles of autophagy in immunity.  a | The role of autophagy
in the elimination of microorganisms is shown. An incoming microorganism can induce antimycobacterial autophagic activities19. Similarly,
autophagy by competing for nutrients or by stimulating innate immune receptors, tumour necrosis factor (TNF) has been shown to stimu-
such as Toll-like receptors (TLRs). When the microorganism is taken up by phagocytosis late autophagy to restrict intracellular bacteria such as
and remains in an intact vacuole, an autophagic process termed LC3‑associated Shigella spp. and Listeria spp20.
phagocytosis (LAP) can promote the maturation of autophagosomes into autolysosomes. By contrast, TH2 cell-associated cytokines, such as
Xenophagy of pathogens that enter the cytosol can be initiated by sequestosome 1‑like IL‑4 and IL‑13, inhibit autophagy 16. The signalling path-
receptors (SLRs) or other mechanisms, including nucleotide-binding oligomerization ways that lead to the inhibition of autophagy by IL‑4
domain-containing protein 2 (NOD2)–autophagy-related protein 16‑like 1 (ATG16L1) and IL‑13 are context dependent: they occur through
interactions. b | Several examples of the role of autophagy in the control of AKT signalling when autophagy is induced by starva-
pro-inflammatory signalling (see also FIG. 3) are shown. Failure to remove SLRs by
tion, and are AKT independent and signal transducer
autophagy can increase the levels of these receptors and the levels of pro-inflammatory
signalling. Autophagy can deliver cytoplasmic pathogen-associated molecular patterns and activator of transcription 6 (STAT6) dependent
(PAMPs) to endocytic TLRs and can stimulate their activity. NOD-like receptors (NLRs; when autophagy is induced by IFNγ16. IL‑10 can also
such as NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3)) and RIG‑I‑like inhibit autophagy through AKT signalling 21. Moreover,
receptors (RLRs; such as absent in melanoma 2 (AIM2)) show complex positive and STAT3, which transduces signals downstream of vari-
negative co‑regulation with autophagy: the ATG5–ATG12 complex inhibits retinoic ous signals including IL‑6, can inhibit autophagy 22. This
acid-inducible gene I (RIG‑I) signalling, and autophagy limits inflammasome activation inhibitory pathway does not involve transcriptional
by removing damaged mitochondria, which then release the inflammasome activators signalling, but instead involves the binding of cyto­
reactive oxygen species (ROS) and mitochondrial DNA. c | The role of autophagy in plasmic STAT3 to protein kinase R (PKR; also known
adaptive immunity is shown. Autophagy can increase the MHC class II presentation as IFN-induced double-stranded RNA-activated protein
of cytoplasmic antigens, including self or viral antigens, as well as promoting the
kinase)22. STAT3 interacts through its SH2 domain with
citrullination of antigens. LAP can enhance the processing of particulate antigens
for MHC class II presentation. NOD2 enhances autophagic antigen presentation. PKR and inhibits it. This prevents PKR from facilitating
Autophagy may directly or indirectly affect MHC class I presentation by competing with autophagy through the hyperphosphorylation of eukary-
the proteasome for substrates, by influencing the peptidome pools through the control otic translation initiation factor 2α (EIF2α; also known
of levels of components of microRNA (miRNA) machinery (for example, argonaute (AGO) as EIF2S1) and the subsequent inhibition of cellular and
and DICER), or by supporting unconventional MHC class I presentation. In addition, viral protein synthesis22.
autophagy affects the self-renewal of haematopoietic stem cells (HSCs), B1 cell Reactive oxygen species (ROS) are classical anti­
development, plasma cell survival and IgG secretion. Autophagy affects T cell survival microbial effectors, which have an important role in
following T cell receptor (TCR) activation, and it destabilizes the immunological synapse. immune signalling. ROS, and the oxidases that generate
It also controls innate immune cell (such as macrophage) signalling through the release them, affect autophagy 23–27 (BOX 2; FIG. 1a). NADPH oxi-
of interleukin‑1α (IL‑1α) and IL‑1β, which influence the polarization of T cells into
dase and the autophagic machinery are connected via an
T helper 17 (TH17) cells. Autophagy also affects naive T cell repertoire selection in the
thymus and the survival and function of maturing T cells by removing the mitochondria autophagy regulatory protein known as RUBICON (run
and endoplasmic reticulum (ER), thus ensuring calcium homeostasis. d | The role of domain beclin 1‑interacting and cysteine-rich-containing
autophagy in the secretion of immune mediators is shown. Autophagy affects the quality protein), which interacts with beclin 1 and the PtdIns3
of regulated secretion from pre-stored granules. Autophagy affects the quality and the kinase catalytic subunit VPS34. By physically disassociat-
quantity of the output of the constitutive secretory pathway (which is the conventional ing from the autophagy inhibitory complex and associating
pathway of protein secretion via the ER, the Golgi apparatus and the plasma membrane). with the NADPH oxidase activating complex, RUBICON
Autophagy supports a form of unconventional secretion that captures cytoplasmic activates two bactericidal mechanisms (that is, autophagy
proteins for extracellular release. Note that secretory protein cargo in the regulated and ROS production), although it is not known whether
and constitutive secretory pathways contains conventional leader peptides for the activation of these two mechanisms occurs simulta-
co‑translational import into the ER lumen, whereas protein cargo that enters the
neously 26. Nitric oxide inhibits autophagy by inactivating
unconventional secretory pathway lacks leader peptides and does not enter the ER.
Dashed arrow indicates that this pathway remains to be defined. DAMPs, damage- Jun N‑terminal kinase 1 (JNK1) and IKKβ through direct
associated molecular patterns; HMGB1, high-mobility group box 1 protein; IPS1, IFNβ S‑nitrosylation27. This prevents JNK1‑dependent BCL‑2
promoter stimulator protein 1; mtDNA, mitochondrial DNA; mTOR, mammalian target phosphorylation27 and BCL‑2 dissociation from beclin
of rapamycin; NOX2, NADPH oxidase 2; PRRs, pattern recognition receptors; 128,29 and inhibits IKKβ-associated AMPK-dependent
ssRNA, single-stranded RNA; TRAF6, TNF receptor-associated factor 6. autophagy initiation27,30.

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | O CTOBER 2013 | 725

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 2 | Parallels between metabolic and immune signalling in autophagy activation


The model (see the figure) proposes that immune and nutritional signals converge to activate a similar cascade. Nutritional
triggers, such as starvation, are transduced via mammalian target of rapamycin (mTOR) and AMP-activated protein kinase
(AMPK). Starvation inhibits mTOR and activates AMPK, which in turn activates the serine/threonine protein kinase
ULK1 to phosphorylate beclin 1 (REF. 140), activating molecule in BECN1‑regulated autophagy protein 1 (AMBRA1)10
and TAK1‑binding protein 2 (TAB2)141. In the resting state, TAB2 and TAB3 bind beclin 1 and repress its activity30,140.
TAB2 activates TGFβ-activated kinase 1 (TAK1)30 to further enhance AMPK activity. AMBRA1 (REF. 10), along with
beclin 1 (REF. 107), recruits TNF receptor-associated factor 6 (TRAF6). TRAF6 functions as an E3 ubiquitin ligase, generating
polyubiquitin chains that in turn stabilize and activate ULK1 (REF. 10) and beclin 1 (REF. 107), and that probably activate
the TAB2 and TAB3–TAK1 complexes. Once fully amplified, the concomitant activation of ULK1 and beclin 1 leads to
autophagy through both branches
SNARE of autophagy regulatory kinases —
(Soluble NSF attachment that is, the protein kinase ULK1 (via AMPK P TAK1
Starvation
protein receptor). A member AMPK) and the lipid kinase VPS34
of a class of proteins that (via beclin 1) (not shown)107.
catalyse membrane fusion P
Immune signals activate
and thus regulate organelle RAPTOR
identity and vesicular
autophagy and engage at least
trafficking. The membrane some components of the mTOR
fusion occurs through the pathways, including ULK1
P
formation of a cognate Qa-, (REF. 108), beclin 1 (REF. 142) and
Qb-, Qc- and R‑SNARE TRAF6 (REF. 107). Downstream of NOD2 RIPK2 ULK1 P TAB2 and
TAB3
four-helix bundle, which the TAB2 and TAB3–TAK1 Ub
consists of SNAREs on donor IFNγ DAPK1 Ub
complexes, IκB kinases (IKKs)
and acceptor membranes. affect autophagy independently of Ub
HMGB1 JNK1
nuclear factor-κB143. TANK-binding Ub
Mammalian target of P Ub
rapamycin
kinase 1 (TBK1) also controls
NLRP3,
(mTOR). A serine/threonine autophagy14,77 and promotes the NLRP4 and Beclin 1
protein kinase that regulates elimination of intracellular NLRP10 Ub
cell growth and metabolism. microorganisms independently of Ub
mTOR is stimulated by growth its role in type I interferon (IFN) Cardiolipin IRGM P AMBRA1
P
factor receptor and phosphati- activation (not shown)144. TBK1
dylinositol‑3,4,5‑phosphate- controls both the capture of TRAF6 BCL-2
RUBICON
dependent signalling. It autophagic cargo (for example,
responds to the availability of NOX2 ROS
microorganisms) via specialized
nutrients (for example, amino
acids). Active mTOR inhibits
adaptor proteins14,35,36,45 and the
TLR4
autophagy via the serine/ maturation of autophagosomal
threonine protein kinase ULK1. organelles into degradative
compartments14,77. TBK1 may also IL-1β
AMP-activated protein contribute to autophagosome
kinase formation at stages earlier than CD40 Autophagy
(AMPK). A sensor of cellular autophagosome maturation77.
energetic state. It is activated
by increased AMP levels which Nature
BCL‑2, B cell lymphoma 2; DAPK1, death-associated protein kinase 1; HMGB1, high-mobility group box 1 Reviews
protein; IL‑1β,| Immunology
indicate decreased energy interleukin‑1β; IRGM, immunity-related GTPase family M protein; JNK1, Jun N-terminal kinase 1; NLRP, NOD-, LRR- and pyrin
status following hypoxia or domain-containing protein; NOD2, nucleotide-binding oligomerization domain-containing protein 2; NOX2, NADPH oxidase 2;
nutrient deprivation. P, phosphorylation; RAPTOR, regulatory-associated protein of mTOR; RIPK2, receptor-interacting serine/threonine-protein kinase 2;
ROS, reactive oxygen species; RUBICON, run domain beclin 1-interacting and cysteine-rich-containing protein; TLR4, Toll-like
Xenophagy receptor 4; Ub, ubiquitylation.
The selective degradation
of intracellular pathogens
(such as bacteria or viruses)
through macroautophagy. The connections between autophagy and immune confined in the nascent and presumably intact phago-
signalling seem to be surprisingly complex. From an some24,31,32 (BOX 3). Finally, a group of autophagic adap-
LC3‑associated evolutionary perspective, these connections reflect the tors33, known as sequestosome 1‑like receptors (SLRs), are
phagocytosis
(LAP). A shared pathway that
integration of autophagy with several immune regulatory involved in eliminating microorganisms from the cyto-
involves conventional systems. plasm. SLRs recognize molecular tags (such as ubiqui-
phagocytosis and autophagy tin, galectin and membrane phospholipid modifications)
at the maturation stage that is Direct elimination of microorganisms present on invading microorganisms or on damaged
mediated by the recruitment
Autophagy intercepts pathogen invasion. The antimi- host membranes that are associated with the pathogen
of the autophagy protein LC3
(which is the mammalian crobial functions of autophagy provide a series of barri- and that physically recruit the autophagic machinery 34–38
homologue of yeast ATG‑8). ers against invading microorganisms (FIG. 1a). The first (FIG. 2). The importance of autophagy in protecting the
LAP results in a more robust antimicrobial function is xenophagy, which is the uptake cytoplasm from microbial invasion is highlighted by the
phagolysosome, which can of intracellular microorganisms into double-membrane microbial countermeasures and adaptations that have
also function as a specialized
signalling compartment or
autophagosomes4, and the second is LC3‑associated evolved to inhibit, to block specific stages or to com-
an antigen-presentation phagocytosis (LAP), which involves the engagement mandeer autophagy 39 (see BOX 4 for recently discovered
compartment. of the autophagic machinery while the bacterium is examples).

726 | O CTOBER 2013 | VOLUME 13 www.nature.com/reviews/immunol

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 3 | LAP: a shared maturation step for autophagy and phagocytosis


NLRs interact with ATGs to localize autophagy. The
cooperation between NLRs and autophagy in antimi-
Autophagy is often morphologically defined by the formation of double-membrane crobial defence is conserved from flies64 to humans41,42.
autophagosomes in the cytoplasm. This is the geometrical consequence of the NOD1 (nucleotide-binding oligomerization domain-
formation of a phagosome that is derived from a pre-existing membranous intracellular containing protein 1) and NOD2 detect muramyl pep-
compartment, such as the endoplasmic reticulum (ER), rather than the consequence of
tides in the cytoplasm and they direct the autophagic
a functional requirement for a double membrane. An exception to this rule is the
formation of standard phagolysosomes by the autophagy-related protein (ATG) LC3,
machinery by recruiting ATG16‑like 1 (ATG16L1) to
through a process known as LC3‑associated phagocytosis (LAP)24,31,32,53. LAP occurs the plasma membrane at the site of bacterial entry 41
following the uptake of various extracellular targets: particles coated with Toll-like (FIG. 1a). The NOD2‑assisted localization of ATG16L1
receptor (TLR) agonists, phagocytosed dead cells, live epithelial cells that are at the plasma membrane is consistent with the role of
engulfed via entosis by neighbouring cells, and the Fcγ receptor-dependent uptake ATG16L1 in the formation of a portion of autophagic
of immune complexes. When a microorganism or a TLR ligand is taken up by precursors from the plasma membrane65 (BOX 1). Of
conventional phagocytosis31,53, the autophagic machinery enhances the maturation note, polymorphisms at the ATG16L1 and NOD2
of the conventional phagosomes through the same maturation pathway that is used loci have been associated with an increased risk of
in internally formed autophagosomes. Thus, LAP uses beclin 1–VPS34 complexes and Crohn’s disease66. Cells from donors who are homozy-
LC3 conjugation systems. LAP is independent of the serine/threonine protein kinase
gous for the Crohn’s disease risk allele ATG16L1*300T
ULK1 (REF. 32) — ULK1 is only required to generate autophagosomes from internal
ER membranes during starvation.
have a decreased capacity for autophagy induction in
response to the NOD2 agonist muramyl dipeptide.
The truncation of NOD2 that occurs in patients with
To initiate autophagy, mammalian cells detect the Crohn’s disease renders it cytoplasmic, which results in
presence, location and extent of the cytoplasmic inva- the retention of ATG16L1 in the cytoplasm. This pre-
sion by a pathogen. Conventional PRRs can elicit cludes ATG16L1 recruitment to bacterial entry sites
autophagic responses at different stages of the host– and thereby prevents timely and site-specific control of
pathogen encounter 40. First, TLRs and NOD-like recep- autophagy.
tors (NLRs) detect released microbial products (that is, Other NLRs also interact with the autophagic machin-
PAMPs) very early following infection and this stimu- ery. NLRX1 and its interacting partner mitochondrial
lates autophagy. Second, autophagy can be initiated Tu elongation factor (TUFM), which associates with
during adhesion- and pathogen-induced uptake of bac- ATG5–ATG12 complexes and with ATG16L1, promote
teria by the host cell, or during active phagocytosis of autophagy 67 (FIG. 3b). NLRC4 (NOD-, LRR- and CARD-
bacteria by macro­phages31,41,42. Third, at stages after bac- containing protein 4), NLRP3 (NOD-, LRR- and pyrin
terial uptake, autophagy is induced following pathogen- domain-containing protein 3), NLRP4 and NLRP10
inflicted damage to the newly formed parasitophorous interact with beclin 1 (REF. 68) (BOX 2). The recruitment
vacuoles35,36,38,43–45 and on the escape of bacteria into the of NLPR4 to the plasma membrane during the phago-
cytoplasm34,46,47. Similarly, the initiation of autophagy cytosis of group A streptococci leads to its transient dis-
following virus infection occurs at various stages of the sociation from beclin 1. This enables the initiation of
virus life cycle25,48–57 (FIG. 1a). beclin 1‑mediated autophagic responses68. Collectively,
NLRs may gather autophagy factors in the vicinity of the
TLRs and autophagy cooperate in the response to incoming microorganism (or resident mitochondria),
PAMPs. The early induction of autophagy downstream of which leads to the activation of autophagy.
TLR stimulation ensures the prompt upregulation
of antimicrobial activities, including the upregulation of Nucleic acid sensors and autophagy. RIG‑I‑like receptors
autophagy systems in advance of microbial invasion58,59 (RLRs), including retinoic acid-inducible gene I (RIG‑I),
Sequestosome 1‑like (FIG.  1a) . This and other triggers, such as IFNγ and melanoma differentiation-associated protein 5 (MDA5;
receptors 1,25‑dihydroxyvitamin D3, may promote the expres- also known as IFIH1) and LGP2 (also known as DHX58),
(SLRs). These are autophagic
sion and the early delivery of antimicrobial peptides recognize viral RNA to induce the production of type I
adaptors that recognize
microbial targets and that link to the parasitophorous vacuoles19,60–62, or they may IFNs and other pro-inflammatory cytokines. Unlike other
them to autophagy machinery direct the autophagic response to the points of micro- PRRs, RLRs seem to negatively regulate autophagy67,69.
by binding to mammalian bial entry, as occurs during LAP31 (FIG. 1a). In addition, However, downstream molecules that are engaged by
autophagy-related protein 8 autophagic membranes deliver cytoplasmic PAMPs, RLRs may have an RLR-independent (and possibly
(ATG-8) proteins (for example,
autophagy related LC3
such as single-stranded viral RNA, to the endosomal competing) function in the induction of autophagy; for
proteins and γ-aminobutyric lumen where they can make contact with the luminal example, downstream of nucleic acid sensing by RLRs,
acid receptor-associated portion of endosomal TLRs, such as TLR7, to stimulate the cytoplasmic adaptor protein stimulator of IFN genes
proteins (GABARAPs)). other responses, including the type I IFN response 50 protein (STING) activates TANK-binding kinase 1
(FIG. 1b). Autophagy that is stimulated by TLRs, in con- (TBK1) signalling and type I IFN production, but can
Crohn’s disease
A form of chronic inflammatory junction with LAP53,63, enhances antigen presentation also induce autophagy, possibly by the direct recogni-
bowel disease that can affect by dendritic cells (DCs) (FIG. 1c). LAP also contributes tion of DNA or second messengers that are associated
the entire gastrointestinal to the trafficking of TLR9 into specialized IFN signal- with the presence of cytoplasmic DNA. Indeed, infec-
tract, but is most common ling compartments in plasmacytoid DCs (pDCs) (FIG. 3). tion with double-stranded DNA viruses, such as herpes
in the terminal ileum. It is
characterized by transmural
Thus, TLRs and autophagy influence each other, which simplex virus 1 (HSV1) or human cytomegalovirus,
inflammation, strictures and amplifies the outputs of both systems in response to induces autophagy that is dependent on STING70,71.
granuloma formation. microbial invasion. Similarly, bacterial DNA that is released during infection

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | O CTOBER 2013 | 727

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

can induce autophagy via STING45 (FIG. 3e). Such bacte- host cell systems generate a second messenger, cyclic
rial DNA can escape from M. tuberculosis-containing GMP–AMP (cGAMP)72 (FIG. 3e). This cyclic dinucleotide
phagosomes through pores that are introduced by the is generated by host cGAMP synthase and activates the
bacterial secretion system ESX1 (REF. 45). It has recently type I IFN pathway via STING72. It remains to be shown
become evident that, in response to cytoplasmic DNA, whether cGAMP production in response to bacterial and

a Sequestosome 1-like receptors b


NLS NES UBA Bacteria
Sequestosome 1 PB1 ZZ Parasitophorous
PAMP vacuole
TRAF6-interacting region LIR KIR
332 342 β-galactoside Galectin

X/(D,E,S,T)-X/(D,E,S,T)-X/(D,E,S,T)-W/F/Y-X/(D,E,S,T)-X-L/I/V
LC3C NDP52
NBR1 PB1 ZZ CC FW CC
Isolation
LIR UBA
membrane
NDP52 (human) SKICH CC GIR UBZ LRSAM1
(E3 ligase)
CLIR TBK1
NDP52 (mouse) SKICH RAB8B
Optineurin

TAX1BP1 SKICH CC CC CC UBZ UBZ

Optineurin CC CC CC UBAN ZnF


LIR
Figure 2 | Autophagy-mediated clearance of intracellular pathogens.  a | Protein domains
of sequestosome 1‑like receptors (SLRs) are shown. The LC3‑interacting region (LIR) motif
of an SLR binds to autophagy-related LC3 proteins through its consensus sequence at
amino acids 332–342 in sequestosome 1 (also known as p62). The conserved residues are Ub
shown. X/(D,E,S,T) indicates that any amino acid (X) is allowed but that acidic (D,E) or Ub
Ub
phosphorylatable amino acids (S,T) are often present (usually at least one or more within Ub Ub
Ub
the entire consensus sequence). The core LIR motif residues are aromatic pocket-filling W
(or F or Y) residues and aliphatic pocket-filling L (or I or V). They form an intermolecular Ubiquitylation
parallel β-sheet with LC3 proteins or γ-aminobutyric acid receptor-associated proteins
(GABARAPs). The CLIR motif, which is a LIR motif that is specific for LC3C, lacks the aromatic
residue found in the LIR motif and, instead, uses hydrophobic contacts provided by
additional aliphatic residues located between the W and L position anchors to stabilize
interactions with LC3C. All human SLRs also contain a ubiquitin-binding domain (UBD):
UBA (as found in sequestosome 1 and NBR1) is a three-helix bundle UBD that has affinity TBK1
for monoubiquitin and K63 ubiquitin linkages; UBAN (as found in optineurin) is a parallel RAB8B
coiled-coiled dimer UBD that has specificity for linear ubiquitin chains; and UBZ
(as found in nuclear dot protein 52 (NDP52)) is a zinc finger ββα-fold UBD that binds to
monoubiquitin and polyubiquitin. b | A model of cooperative action between different
SLRs and E3 ligases in bacterial targeting for xenophagy is shown. The schematic shows a
parasitophorous vacuole with glycosylated molecules (in this case β‑galactosides) facing
the lumen of the vacuole that contains a bacterium and that is experiencing membrane
damage. This membrane tear exposes β‑galactosides to galectins (for example, galectin 8)
which in turn bind to the galectin-interacting region (GIR) motif of NDP52. NDP52 also LC3 or
directly interacts with the E3 ligase LRSAM1 and indirectly with the serine/threonine protein GABARAP
P TBK1
kinase TANK-binding kinase 1 (TBK1), which interacts with optineurin. The CLIR motif of Ub
NDP52 binds to LC3C, which is a proposed initiator in the LC3 and GABARAP cascade Ub
Sequestosome 1 Ub
during bacterial xenophagy. LRSAM1 or other E3 ubiquitin ligases polymerize ubiquitin at multimer Ub
molecular targets that are yet to be identified. The hypothetical model includes the putative Ub
Ub
recognition of bacterial pathogen-associated molecular patterns (PAMPs) by LRSAM1
through its leucine-rich repeat domain. Ubiquitin tags are recognized by UBDs of NDP52, Ub
optineurin and sequestosome 1. The LIR motif of optineurin is phosphorylated by TBK1 and Ub
Ub
this improves LC3 and GABARAP binding. The UBA of sequestosome 1 is also phosphorylated Ub
by TBK1 and this improves ubiquitin chain binding. As a consequence, the autophagic Ub Ub
P
isolation membrane initiates at the appropriate location and grows to capture the bacterium Ub
and to eliminate it through autophagy. CC, coiled-coil domain; FW, four W domain (also Ub
Ub
known as the NBR1 domain); KIR, KEAP1 interacting region; NES, nuclear export signal; NLS, Ub
nuclear localization signal; P, phosphorylation; PB1, protein-binding domain 1; SKICH, skeletal
muscle and kidney enriched inositol phosphatase carboxyl homology domain; TAX1BP1,
TAX1-binding protein 1; Ub, ubiquitylation; ZnF, zinc finger domain; ZZ, ZZ‑type ZnF domain.

728 | O CTOBER 2013 | VOLUME 13 www.nature.com/reviews/immunol

© 2013 Macmillan Publishers Limited. All rights reserved Nature Reviews | Immunology
REVIEWS

host-derived cytoplasmic DNA can induce autophagy. SLRs contain one or more cargo recognition domains
Analogous cyclic dinucleotides that are secreted by (CRDs) that recognize ubiquitin-tagged 20,34–36,45,46,73
bacteria (for example, cyclic di‑GMP or di‑AMP) bind or galectin-tagged38,79 microbial or microorganism-
to STING and can activate autophagy when they are associated targets. Ubiquitin-binding domains (UBDs)
introduced into the host cell cytoplasm45 (FIG. 3e). that are specific for different ubiquitin chains (UBA,
In conclusion, conventional PRRs recognize microbial UBZ or UBAN)80 are present in all known SLRs. SLR
products at different stages of an infection and initiate CRDs that recognize tags other than ubiquitin include
an autophagic response that contributes to the efficient a hook-like CRD (which is the galectin-interacting
elimination of the invading microorganism. However, region (GIR) motif) that enables NDP52 to interact
in some instances, autophagy can suppress type I IFN with galectin 8 (REF. 79) (FIG. 2a). Galectin 8 links NDP52
responses, notably by inhibiting RLR signalling 67,69. This to cytoplasmically exposed β‑galactoside glycans on
relationship paradoxically promotes viral replication, and pathogen-damaged host membranes38 (FIG. 2b).
may explain why autophagy can have a pro-infective role. Moreover, all SLRs have an LC3‑interacting region
motif (LIR motif), which is X/(D,E,S,T)-X/(D,E,S,T)-X/
SLRs clear microorganisms from the cytoplasm. If a path- (D,E,S,T)-W/F/Y-X/(D,E,S,T)-X-L/I/V, where X/
ogen escapes the autophagy barriers that are controlled (D,E,S,T) indicates that any amino acid (X) is allowed
by conventional PRRs, it can still be captured in the cyto- but that acidic (D,E) or phosphorylatable amino acids
plasm or even in the cytosol. The autophagy-mediated (S,T) are often present (at least one or more within the
elimination of cytoplasmic pathogens depends on special- entire motif)33. A modified LIR, known as a CLIR motif
ized adaptors known as SLRs20,34–36,46,73,74 (FIG. 2a). SLRs, (L-V-V) has been identified in NDP52 (REF. 81) and pos-
Ubiquitin-binding domains which are named after the archetypical protein seques- sibly in TAX1BP1 (REF. 77) (FIG. 2a). Phosphorylation of
(UBDs). Domains that have tosome 1 (also known as p62) include sequestosome 1 the LIR motif and of the CRDs of SLRs modulates their
different specificities for
(REF. 75), NBR1 (REF. 76), optineurin36, nuclear dot protein autophagic activities14,36,82 (FIG. 2b). The number of SLRs
ubiquitin chains: UBA, which
is found in sequestosome 1, 52 (NDP52; also known as CALCOCO2; it is full size and the type of unique or repetitive structures that are
favours K63 polyubiquitin in humans but truncated in mice)35 and an NDP52‑like recognized as tags may increase as more research is
chains; UBZ, which is found receptor TAX1-binding protein 1 (TAX1BP1; also known carried out.
in nuclear dot protein 52
as CALCOCO3)77 (FIG. 2a). SLRs have been shown to The process of autophagic clearance of microorgan-
(NDP52), binds to
monoubiquitin and affect inflammation in several ways34,74,78 and, as they can isms from the cytoplasm involves the sensing of DAMPs
polyubiquitin; and UBAN, be consumed during autophagy 33, it is possible that SLR and PAMPs, and often the function of multiple SLRs. An
which is found in optineurin, accumulation (following autophagy inhibition) or SLR example of autophagy-inducing DAMPs are the glycans
has specificity for linear depletion (following autophagy activation) may modulate that become exposed following damage to host mem-
ubiquitin chains.
inflammatory processes (FIG. 1b). branes, as in the case of infection with Salmonella spp.38,79.
LC3‑interacting region motif
(LIR motif). Canonical LIR
motif, which is X/(D,E,S,T)-X/ Box 4 | Microbial countermeasures against autophagy
(D,E,S,T)-X/(D,E,S,T)-W/F/Y-X/
Microorganisms use a wide range of mechanisms to prevent, to counteract or to commandeer autophagy39.
(D,E,S,T)-X-L/I/V, where X/
(D,E,S,T) indicates that any
These inhibitory mechanisms involve the targeting of beclin 1 (REFS 49,145), the inhibition of autophagosomal
amino acid (X) is allowed maturation51,57, the perforation of autophagosomal membranes to prevent acidification44, the proteolytic cleavage
but that acidic (D,E) or of autophagy-related protein 8 (ATG8) to irreversibly remove carboxy-terminal lipid modifications146, and the
phosphorylatable amino masking of epitopes or tags that are recognized by sequestosome 1‑like receptors (SLRs)46,147. Autophagy may
acids (S,T) are often present. even be activated to generate nutrients for invading microorganisms148.
Aromatic residues (W/F/Y) With regard to bacteria, Listeria spp. proteins AktA46 and InlK147 interfere with recognition via host ubiquitin tags,
occupy the aromatic pocket and Shigella spp. protein IcsB masks bacterial epitopes149, which are recognized by the autophagic machinery, either
and aliphatic side chains (L/I/V) directly or by recruiting cytoplasmic proteins. The Salmonella spp. deubiquitinase SseL removes ubiquitin tags88.
occupy an aliphatic pocket
The Legionella spp. effector protein RavZ is injected into the host cytoplasm through the bacterial type IV secretion
present in all autophagy-
related protein 8 (ATG-8)
system and inhibits autophagy through irreversible deconjugation of mammalian homologues of ATG-8. This involves
homologues. The LIR motif the proteolysis of the C‑terminal glycine in ATG-8 homologues, which prevents future phosphatidylethanolamine
forms an intermolecular modification146. Listeria spp. block autophagosome acidification through the pore-forming toxin listeriolysin O44.
β‑sheet with ATG-8 homologues, Bacterial actin-based intracytoplasmic motility may be another factor in evading capture by autophagosomes.
not discriminating between This is supported by the role of septin scaffolds in enabling autophagy of cytoplasmic Shigella spp. as they start
mammalian paralogues of to polymerize actin20.
yeast ATG-8. Viruses also interfere with autophagy; for example, herpes simplex virus 1 infected cell protein 34.5 (ICP34.5)49,
influenza virus M2 protein52 and HIV protein Nef51,57, all target beclin 1 to either completely block autophagy or
CLIR motif
to inhibit autophagosomal maturation. Nef binds to the evolutionarily conserved domain of beclin 1 at the
A variant of the LC3‑interacting
region (LIR) motif (L-V-V),
same region as the endogenous inhibitor GAPR1 (Golgi-associated plant pathogenesis-related protein 1)57.
which is found in nuclear Mouse herpesvirus 68‑encoded protein M11 (which is the viral homologue of B cell lymphoma 2) inhibits beclin 1
dot protein 52 (NDP52). through its BH3 domain145, whereas Kaposi sarcoma-associated virus (KSHV) FLICE-like inhibitory protein (FLIP)
The CLIR motif binds to the blocks ATG3 in the LC3 lipidation cascade127. HIV Nef, hepatitis C virus NS3 and measles virus Mev3 interact with
LIR-interacting region in LC3C. autophagy factor immunity-related GTPase family M protein (IRGM), which has consequences that are yet to be
The CLIR motif lacks the fully understood150.
signature aromatic residue of Finally, several microorganisms can harness autophagy for their own benefit, including using autophagy for
the LIR motif. Instead, it makes replication39; for example, the obligatory intracellular pathogen Anaplasma phagocytophilum uses its type IV
compensatory hydrophobic
secretion effector Ats1 to activate autophagy to supply nutrients148.
contacts with LC3C.

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | O CTOBER 2013 | 729

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

a DNA-containing b c
antigen
RIG-I Dectin RIG-I
B cell pDC
BCR ATG12
↑ Autophagy RUBICON
ATG5 BCL-10
Beclin 1
IPS1 MALT1 CARD9 ↓ Autophagy
LAP VPS34
TUFM
TLR9 NF-κB
NLRX1
Type I
IFN BCL-10
MALT1
CARD9 RUBICON
B cell activation Type I IFN
Degradation
BCL-10 Ub of BCL-10 by
autophagy
Autoimmune plasma cells Sequestosome 1

d Autophagy e
Virus DNA cGAMP
synthase

ROS ↑ RLR signalling Mitochondria cGAMP

GMP AMP
Depolarized Bacteria
mitochondria Inflammasome Calpain Type I IFNs
Cyclic di-AMP STING ATG9 ↑ Autophagy
Cyclic di-GMP
IL-1β IL-1α

Autophagy TBK1

Type I IFN
Figure 3 | Autophagy controls inflammatory processes.  a | Autophagy promotes Toll-like receptor 9 (TLR9)
Nature Reviews
signalling in B cells and type I interferon (IFN) production by plasmacytoid dendritic cells (pDCs). | Immunology
b | The autophagy
protein complex autophagy-related protein 5 (ATG5)–ATG12 inhibits RIG‑I‑like receptor (RLR) signalling by binding
to the caspase recruitment domains of retinoic acid-inducible gene I (RIG‑I) and IFNβ promoter stimulator protein 1
(IPS1), which is the mitochondrial adaptor of RIG‑I signalling. The NOD-like receptor X1 (NLRX1)-interacting partner
mitochondrial Tu elongation factor (TUFM) associates with the ATG5–ATG12 complex to promote autophagy while
inhibiting RLR-dependent type I IFN activation. c | Autophagy factors negatively regulate the caspase recruitment
domain-containing protein 9 (CARD9)–B cell lymphoma 10 (BCL‑10)– mucosa-associated lymphoid tissue lymphoma
translocation protein 1 (MALT1) complex. RUBICON (run domain beclin 1-interacting and cysteine-rich-containing
protein), which is a binding partner and a negative regulator of beclin 1, inhibits CARD9, whereas sequestosome 1
leads to the degradation of BCL‑10. d | Excessive production of reactive oxygen species (ROS) by depolarized
mitochondria that are not cleared by autophagy enhance RLR signalling. e | Viral, mitochondrial or bacterial DNA lead
to the activation of stimulator of IFN genes protein (STING), probably through cGAMP synthase and cyclic GMP–AMP
(cGAMP) production, which increases the type I IFN response. Autophagy removes sources of agonists that stimulate
STING, whereas autophagic factors (for example, ATG9) inhibit the activation of STING by affecting its cytoplasmic
translocation. Bacterial cyclic dinucleotides (di‑AMP and di‑GMP) can activate autophagy, thereby functioning as a
regulatory loop that amplifies the removal of infectious or endogenous irritants. BCR, B cell receptor; IL, interleukin;
LAP, LC3‑associated phagocytosis; NK-κB, nuclear factor-κB; TBK1, TANK-binding kinase 1; Ub, ubiquitylation.

Moreover, microbial polymers, such as DNA, that are There is further cooperation (FIG. 2b) between the
present in the cytoplasm might function as autophagy- recognition of the initial tears in microorganism-
inducing PAMPs, as occurs during M. tuberculosis infec- harbouring vacuoles and ubiquitylation, as exemplified
Leucine-rich repeat tion45. In the cytoplasm, microorganisms may become in the case of Salmonella spp. infection. In this exam-
(LRR). A domain that is often coated with ubiquitin20,34–36,45,46,73,which may involve ple, LRSAM1 directly interacts with NDP52 (REF. 83). In
found in pattern recognition
specialized E3 ubiquitin ligases, such as LRSAM1, which human cells, NDP52 seems to function as an important
receptors and that is involved
in pathogen-associated recognize Salmonella spp. through their leucine-rich hub, as it can recognize LRSAM1 (REF. 83), galectin 8
molecular pattern recognition. repeat (LRR) domains83. Following auto-ubiquitylation, bound to β‑galactoside glycans38,79, ubiquitin tags35 and
LRRs are repeats of L-X-X-L-X- LRSAM1 ubiquitylates microbial targets that are yet to be LC3C (also known as MAP1LC3C), which is required
L-X-X-N-X-L or L-X-X-X-L-X-L-X- defined and possibly host molecules that are associated for the acquisition of other LC3 proteins to mediate
X-C-X-X-L motifs (where X
represents any amino acid),
with microorganisms. Any of these ubiquitin tags alone autophagy 81. The action of NDP52 is non-redundantly
which form a horseshoe or or in combination may lead to the recruitment of SLRs reinforced by other SLRs that may be recruited to
solenoid tertiary structure. through their UBDs (FIG. 2b). Salmonella spp., such as optineurin (which localizes with

730 | O CTOBER 2013 | VOLUME 13 www.nature.com/reviews/immunol

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

NDP52 in microdomains surrounding Salmonella spp.) phospholipid-binding domain C2 is required. In keep-


and sequestosome 1 (REF. 36). These SLRs bind to ubiqui- ing with the idea that membrane phospholipids or their
tin through their UBDs that have varied specificities for derivatives are potential tags or signalling intermedi-
ubiquitin chains80, and, through their LIR motifs, they ates, diacylglycerol has also been implicated in bacterial
recruit the remaining LC3 proteins and GABARAPs to autophagy 37. Thus, the presence of an E3 ligase domain
mediate autophagosome formation. The affinity of SLRs in an autophagy-targeting factor does not necessarily
for LC3 and ubiquitin is further enhanced by phospho- indicate that it ubiquitylates the cargo as a requirement
rylation of the LIR motif in optineurin and the UBD for selective autophagy.
in sequestosome 1 by TBK1 (FIG. 2b). TBK1 is recruited In summary, we now understand in greater detail
to these sites via protein complexes containing NDP52 how autophagy is activated in response to microbial
(REF. 35), optineurin36 and RAB8B14, which is a small presence. Autophagy is controlled by nearly all classes
GTPase that regulates membrane trafficking. The cargo of PRRs and is also regulated by cytokines and recep-
capture and autophagy is coordinated with maturation tors that modulate innate and adaptive immunity.
into autolysosomes by RAB8B14. Notably, the NDP52 Integration of the immune triggers is an obvious area
product that is encoded by the corresponding mouse for continuing study, and it is already clear that the
gene Calcoco2 is carboxy‑terminally truncated (and lacks antimicrobial functions of autophagy can be promoted
motifs for ubiquitin and galectin binding) (FIG. 2a) and (for example, by TH1 cell-associated cytokines and
may be primarily expressed in embryonic tissues (see the IL‑1β) or inhibited (for example, by TH2 cell-associated
expressed sequence tags NCBI database). This suggests cytokines) by immune mediators. At the intracellular
that, in mice, the function of CALCOCO2 may be pri- level, an idea has emerged of a graded, multitiered sys-
marily conferred through the recruitment of LRSAM1, tem of autophagic responses that are commensurate
TBK1 and LC3, as the binding sites for these proteins are with the extent of microbial penetration. An interesting
retained in the truncated mouse protein. Alternatively, recent development was the discovery of the hierarchi-
NDP52 activities in mice may be compensated for by cal cooperation of several proteins in controlling anti-
the NDP52‑like receptor CALCOCO3. microbial autophagy. However, gaps in our knowledge
remain, especially with regard to alternative, that is, not
Other pathways to target cytoplasmic microorgan- ubiquitin-based, modes of target recognition. At pre-
isms for autophagy. Autophagy-associated factors have sent, recognition of ubiquitin tags seems to be crucial
been shown to directly target microbial proteins 47; for autophagy, and this is further supported by evidence
for example, ATG5 binds directly to a Shigella spp. of bacterial countermeasures that involve deubiquity-
surface protein, VirG. The ATG5–VirG interaction lation88. Moreover, ubiquitin tags might only activate
only occurs when the VirG recognition epitope is not autophagy after more specialized mechanisms of recog-
masked by another Shigella spp. protein, IcsB (BOX 4), nition have failed. It remains to be determined whether
and requires the ATG5‑binding partner TECPR1 (tec- overly strong activation of antimicrobial autophagy
tonin β-propeller repeat-containing protein 1) which comes at a price, as exemplified by its interference with
interacts with WIPI2 (WD repeat domain phospho- type I IFN signalling. Finally, although autophagy is a
inositide-interacting protein 2)47. WIPI2 is one of the mode of self defence that is available, in principle, to
four mammalian PtdIns3P-binding ATG18 paralogues any eukaryotic cell, the magnitude of its contribution
and has a role in phagophore formation (BOX 1). Of note, in complex organisms is cell type dependent, as shown
the control of icsB-mutant Shigella spp. also requires by the fact that it has an important role in protecting
NOD1 activity to induce autophagy and to limit intra- neurons against HSV1 infection whereas it is absent
cellular growth41. Moreover, these mechanisms against in keratinocytes of the vaginal mucosa56. Thus, at the
Shigella spp. are further complemented by ubiquitylation organismal level, tissue- and cell-specific engagement
and SLR activity 20,34. Taken together, these findings sup- of antimicrobial autophagy may be a key factor.
port the idea that autophagic defences are multilayered
and cooperative in nature. Control of inflammation
Other antibacterial pathways include the autophagy Genetic links with immune disorders. The role of
factors beclin 1 (REF. 84) and immunity-related GTPase autophagy in inflammatory diseases was initially
family M protein (IRGM) which directly bind to cardio­ established through genome-wide association stud-
lipin, a lipid that is only present in bacteria and mito- ies (GWASs)66. Polymorphisms in autophagy-asso-
chondria17. IRGM is necessary for the optimal induction ciated genes, such as ATG16L1 and IRGM, are linked
of autophagy 17, is partially localized in mitochondria to Crohn’s disease66. In addition to single nucleotide
(hence its affinity for cardiolipin)17 and is a genetic polymorphisms, IRGM is an example of a gene dosage
predisposition factor for Crohn’s disease66,85,86. The (through copy number variants) correlation with a pre-
E3 ubiquitin-protein ligase SMURF1 is required for anti- disposition to Crohn’s disease in human populations85.
Mitophagy viral responses to Sindbis virus and to HSV1 (REF. 87). One of the common IRGM polymorphisms in Crohn’s
A special form of autophagy SMURF1 is involved in mitophagy87, which in some disease leads to an escape from the negative regula-
by which mitochondria (in a respects resembles bacterial xenophagy. Surprisingly, the tion of IRGM expression by a microRNA (miRNA)86.
damaged or depolarized
state) are engulfed by
HECT (homologous to the E6‑AP C terminus) domain A recent GWAS analysis of 75,000 individuals indicates
autophagosomes and of SMURF1 (and hence the region that has its E3 ligase an overlap between susceptibility loci for inflammatory
degraded. activity) is dispensable for mitophagy and, instead, its bowel disease and mycobacterial infections89. Moreover,

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | O CTOBER 2013 | 731

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

a link has been reported between Crohn’s disease and are a source of ROS that also enhance RLR effects25
the autophagy-targeting factor SMURF1 (REF.  89) . (FIG. 3d). NLRX1 promotes autophagy and inhibits the
Furthermore, polymorphisms in ULK1 have also been RLR-dependent induction of type I IFN signalling and
linked with Crohn’s disease90. inflammation (FIG. 3b); indeed, the two processes are
In addition, polymorphisms in autophagy-associated reciprocally affected by NLRX1 (REF. 67). Finally, ATG9
genes have been associated with autoimmune dis- negatively controls the trafficking of STING and sup-
orders. IRGM polymorphisms may be a risk factor presses the activation of TBK1 in type I IFN signalling
in systemic lupus erythematosus (SLE)91. Moreover, in response to double-stranded DNA100 (FIG. 3e).
GWASs have linked ATG5 variants with SLE92–94 and Collectively, these phenomena may represent feed-
asthma95. Rheumatoid arthritis has been associated back loops by which autophagy downregulates type I
with variations in the PR domain-containing gene 1 IFN responses following a period of productive induc-
(PRDM1)–ATG5 intergenic region96. A new human tion or to increase the threshold for activation of type I
autophagy locus, which was first functionally identified IFN signalling. Alternatively, autophagic interference
in Caenorhabditis elegans screens as epg5 and which was with RLR signalling could reflect competition for limited
shown in mice to be required for autophagosomal matu- resources, such as TBK1, that are shared between type I
ration97, has been linked to the complex Vici syndrome IFN signalling and autophagy pathways14,36,45,101. These
that includes immunodeficiency 98. Thus, autophagy phenomena, in addition to other factors that are not dis-
shows clinical relevance as a result of genetic links with cussed here, could be an explanation for why autophagy
immunological and inflammatory disorders. paradoxically enhances the replication of certain viruses.

Autophagy affects PRR-mediated type I IFN signalling. Autophagy suppresses inflammasome activation. The
The autophagic machinery can amplify TLR signalling; recognition that autophagy has an anti-inflammatory
for example, autophagy enhances the delivery of cyto- function stems from the observation that the production
plasmic PAMPs to endosomal TLR7, which enables the of IL‑1β and IL‑18 is increased in the absence of func-
recognition of cytoplasmic viral replication intermedi- tional ATG16L1 in a mouse model of Crohn’s disease102.
ates and IFNα production by pDCs50. However, the role Inflammasomes are cytoplasmic complexes that respond
of autophagy in PAMP–PRR amplification loops can to PAMPs and DAMPs by inducing the proteolytic pro-
lead to aberrations and auto­immunity. In the case of cessing and secretion of IL‑1β and IL‑18 (REF. 103). An
TLR9, which normally responds to microbial unmethyl- inflammasome consists of pro-caspase 1, the adaptor
ated CpG DNA, the autophagic machinery can enhance protein ASC and a sensor protein from the NLR family
aberrant self DNA reactivity. This occurs via intracellular (such as NLRP1, NLRP3, NLRC4, NLRP6 or NLRP12)
trafficking events following B cell receptor (BCR) stim- or from the PYHIN (pyrin and HIN domain-containing
ulation by self DNA-containing antigens11. Autophagy protein) family (which includes absent in melanoma 2
promotes the fusion of DNA-containing antigen-loaded (AIM2) and IFNγ-inducible protein 16 (IFI16)) 103.
BCR compartments and TLR9‑containing endosomes Several convergent reports show that autophagy has a
and leads to B  cell hyper­responsiveness 11 (FIG.  3a) . negative role in inflammasome activation104–108. Under
Similarly, in pDCs the autophagic machinery enhances sterile conditions, autophagy clears the cytoplasm of
the trafficking of particulate self DNA-containing debris, protein aggregates and defective organelles that
immune complexes and of TLR9 to signalling- can function as endogenous inflammasome agonists.
competent compartments, which results in increased Studies suggest that basal levels of autophagy control the
IFNα production32 (FIG. 3a). It is possible that these aber- set point for inflammasome activation104,105. If autophagy
rant autophagic activities in B cells and pDCs could is blocked, this leads to an accumulation of depolarized
cooperate and lead to the generation of autoimmune mitochondria that leak endogenous inflammasome ago-
plasma cells (FIG. 3a). nists, such as mitochondrial DNA (which is detected,
By contrast, there are examples of autophagy-related at least partly, by AIM2) and ROS (which activate the
factors that directly inhibit the formation or that sup- NLRP3 inflammasome)104,105 (FIG. 1b).
press the activation of pro-inflammatory protein com- During infection, the removal of damaged mitochon-
plexes. The ATG5–ATG12 complex negatively regulates dria may not be a passive process; for example, during
RLR signalling by directly binding to the caspase recruit- infection with influenza A virus, a NOD2–receptor-
ment domains (CARDs) of RIG‑I and IFNβ promoter interacting serine/threonine protein kinase 2 (RIPK2)
stimulator protein 1 (IPS1; also known as MAVS, VISA pathway activates the autophagy factor ULK1 to main-
or CARDIF; it is a mitochondrial adaptor of RIG‑I sig- tain or to increase mitophagy and to thereby reduce
nalling)69 (FIG. 3b). RUBICON inhibits CARD9–BCL‑10– inflammasome activation108. This RIPK2‑dependent
MALT1 (mucosa-associated lymphoid tissue lymphoma regulation of autophagy may complement the
translocation protein 1) signalling complexes by binding RIPK2‑independent action of NOD2 following the
to CARD9, which thereby terminates pro-inflammatory recruitment of ATG16L1 to microorganisms41 and both
signalling downstream of RIG‑I or dectin 1 (REF. 99) processes may contribute to inflammatory syndromes
(FIG. 3c). The absence of autophagy amplifies RLR sig- such as Crohn’s disease. Importantly, autophagy may
nalling through increased IPS1 levels as a result of an downregulate prolonged inflammasome activity by
accumulation of mitochondria, and increased pools of removing aggregated inflammasome components107.
depolarized mitochondria in the absence of mitophagy This depends on the recruitment of sequestosome 1 to

732 | O CTOBER 2013 | VOLUME 13 www.nature.com/reviews/immunol

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

K63‑ubiquitylated ASC107. A role for exocyst has been targeting of other inflammatory signalling pathways is
implicated in the overall process107, but the process could also becoming apparent. This is consistent with the idea
be self-starting, as sequestosome 1 associates with the that autophagy eliminates microorganisms (and micro-
E3 ligase TRAF6 (REF. 78) that ubiquitylates beclin 1 to bial products or corpses — an area that deserves more
initiate autophagy 9; however, this has not been inves- investigation) and endogenous irritants to prevent exces-
tigated. In summary, basal autophagy protects cells sive inflammation. When autophagic clearance fails,
from inadvertent inflammasome activation initiating inflammation ensues as the body’s response to persistent
damaging sterile inflammation, whereas a deficiency in danger. This may be an explanation for the selection of
autophagy causes increased IL‑1β levels (FIG. 1b). genetic alleles in autophagy-related factors that promote
inflammation at certain anatomical sites. This has been
Autophagy suppresses calpain-dependent IL‑1α activa- recently shown in the case of the Crohn’s disease risk
tion. Similarly to IL‑1β, IL‑1α is synthesized as a cyto- factor ATG16L1, whereby a defect in autophagy may
plasmic pro-form, and is processed by calpain or other paradoxically confer protection against uropathogenic
proteases before being actively secreted from cells or Escherichia coli (UPEC) by increasing the production
passively released following cell death109. The activation of pro-inflammatory cytokines and by promoting the
and the secretion of IL‑1α involve caspase 1‑dependent recruitment of innate immune cells to UPEC-infected
and caspase 1‑independent processes109. Autophagy- bladders in ATG16L1‑hypomorphic mice113.
defective (Atg5fl/fl LysM–Cre+) macrophages secrete
high levels of IL‑1α through a calpain-dependent but Autophagy in adaptive immunity
inflammasome-independent pathway 109. This pheno- Autophagy in antigen presentation and T cell responses.
type has been observed both in vitro and in vivo 45,109. Autophagy functions as a bulk ‘topological inverter’ by
The processing of pro‑IL‑1α by calpain is induced by transporting proteins from the cytoplasm into the lumen
ROS that have been released from accumulated depo- of antigen-processing compartments54,114,115 (FIG. 1c). This
larized mitochondria in autophagy-deficient cells109. property can be artificially exploited by fusing antigens
Thus, ROS in autophagy-defective cells activate both the (such as influenza virus matrix protein 1) to LC3 to
inflammasome and the calpain pathways that lead to the enhance MHC class II‑mediated antigen presentation
excess production of IL‑1β and IL‑1α, respectively 104,109. to CD4+ T cells116. The topological inversion principle
IL‑1α that has been released from ATG5‑defective mac- is more generally applicable as it supplies cytoplasmic
rophages has important consequences in vivo, as it leads ligands to endosomal receptors; for example, autophagy
to enhanced and prolonged TH17 cell responses in the transfers viral RNA replication intermediates to lumenal
context of M. tuberculosis infection, which contributes to TLR7, as discussed above.
lung tissue damage in a mouse model of tuberculosis109. The contributions of autophagy to MHC class II
presentation are not restricted to cytoplasmic pro-
Autophagy and degradation of pro-inflamma- teins and extend through LAP to exogenous antigens.
tory signalling factors. Autophagy factors inhibit Autophagy augments the MHC class II‑dependent pro-
BCL‑10‑containing complexes99, and autophagy degrades cessing and presentation of phagocytosed extracellular
BCL‑10 to reduce nuclear factor‑κB (NF‑κB) activation, particulate antigens, as shown using PRR-stimulated
as shown in antigen-activated T cells110. This occurs via LAP of ovalbumin-coated latex beads in DCs53. PRR-
sequestosome 1 association with K63‑polyubiquitylated mediated enhancement of MHC class II presenta-
BCL‑10 (FIG. 3c). As mentioned above, BCL‑10‑containing tion also occurs through the induction of autophagy
complexes are also inhibited by the binding of RUBICON following NOD2 stimulation with bacterial muramyl
to CARD9 (REF. 99). As RUBICON is a negative regulator dipeptides42 (FIG. 1c).
of autophagy, its translocation to BCL‑10 complexes leads Autophagy-enhanced MHC class II presentation
to the direct inhibition of CARD9–BCL‑10–MALT1 possibly has a role in the selection of naive T cell rep-
signalling and may also enhance the autophagic deg- ertoires in the thymus. In autophagy-deficient mice,
radation of these complexes (FIG. 3c). NF‑κB signalling both positive and negative selection of CD4 + T cells
may also be downregulated by autophagy via NSFL1C but not of CD8+ T cells were affected, and this had
cofactor p47, which is a protein that has a ubiquitin- consequences for the selection of appropriate T cell
binding UBA domain and its orthologue in yeast binds receptor (TCR) repertoires and for the elimination
to ATG‑8. This potential adaptor functions as a negative of autoreactive CD4 + T  cells 115. Transplantation of
regulator of IKK through the lysosomal (and presum- ATG5‑deficient thymi into wild-type mice caused an
ably autophagic) degradation of polyubiquitylated NF‑κB infiltration of autoreactive CD4+ T cells into multiple
essential modulator (NEMO)111. The targeting of NEMO organs and induced autoimmune colitis that was similar
Exocyst
for autophagic degradation is specifically induced by the to Crohn’s disease-related phenotypes115. These effects
An evolutionarily conserved
protein complex that consists murine cytomegalo­virus protein M45; this suppresses have been attributed to the defective presentation of
of eight subunits and that is antiviral processes during infection112. cytoplasmic antigens by autophagy-deficient thymic
best known for targeting In summary, genetic and functional studies indicate epithelial cells115. Other studies investigating DC func-
exocytic vesicles to sites of that autophagy is an anti-inflammatory mechanism tion indicate that autophagy-dependent antigen pres-
docking and fusion at the
plasma membrane. It also
that affects numerous pathways. The most prominent entation is defective in DCs from patients with Crohn’s
functions as a protein complex examples are the dampening of inflammasome acti- disease that express the ATG16L1 or NOD2 disease risk
assembly platform. vation and type I IFN signalling; however, broader variants42. The contributions of autophagy-dependent

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | O CTOBER 2013 | 733

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

MHC class II antigen presentation in antimicrobial Following T cell activation, autophagy can influence
defence are experimentally detectable in the context TH cell polarization, partly by controlling the inflamma-
of adaptive immunity against viral54,114 and bacterial63 tory function of innate immune cells; for example, the
infections. As a result of the enrichment of peptidy- excessive secretion of IL‑1α and IL‑1β from autophagy-
larginine deaminase in autophagosomes117, autophagy deficient macrophages, functions together with IL‑6
promotes the presentation of citrullinated antigens that and transforming growth factor‑β (TGFβ), to promote
may be relevant to autoreactivity (FIG. 1c). This process TH17 cell responses109 (FIG. 1c). Indeed, the levels of IL‑17
is constitutive in DCs and macrophages, but in B cells it are increased in the lungs of mice with ATG5‑deficient
only occurs following BCR stimulation117. myeloid cells during M.  tuberculosis infection 109.
In contrast to this augmentation of MHC class II CD4+ T cells show increased IL‑17 expression when
antigen processing by autophagy, autophagic degrada- lung-infiltrating leukocytes from mice with an ATG5
tion promotes the disassembly of immunological syn- deletion in the myeloid lineage are restimulated with
apses118. Inhibition of ATG16L1 and IRGM expression mycobacterial antigens ex vivo109. The increased TH17
in DCs leads to hyperstable interactions with T cells and cell response might also result from the increased dura-
increases T cell activation118. This activation might be bility of immunological synapses between T cells and
further enhanced in the T cells by the lack of autophagic autophagy-defective DCs109,118.
degradation of BCL‑10 (REF. 110) (FIG. 3c). Therefore,
although autophagy initially promotes MHC class II Autophagy in plasma cells and humoral immunity.
antigen processing, at later stages it may help to down- Although autophagy seems not to be important for the
regulate the response. Consistent with this idea, during survival of the majority of mature B cells130, lymphoid pre-
cancer cell invasion the epithelial-to‑mesenchymal transi- cursor stages are affected by the absence of autophagy 131.
tion is associated with an attenuation of immunological Autophagy is needed for the survival of B1 cells, which
synapses between target cells and cytotoxic lympho- are a subset of self-renewing B cells that cannot be
cytes in a process that is dependent on autophagy and replenished from the adult bone marrow 130. Moreover,
beclin 1 (REF. 119). autophagy has a role in ER maintenance in plasma cells
Autophagy affects conventional MHC class I presen- under conditions of high secretory demands. Somewhat
tation by competing with the proteasome for the deg- paradoxically, absence of autophagy leads to excessive
radation of newly synthesized cytoplasmic proteins120 immunoglobulin secretion132. Autophagy is important
(FIG.  1c) . Autophagy may also contribute to uncon- not only for the function of plasma cells but also for their
ventional MHC class I presentation pathways in ways survival and homeostasis; it is especially important for
that are yet to be fully understood121,122. In principle, the preservation of the bone marrow plasma cell pool
Citrullinated antigens
autophagy could broadly affect MHC class I peptide rep- and, thus, is relevant for the maintenance of long-lived
Citrullin is enzymatically ertoires; for example, NDP52‑dependent autophagy tar- humoral immunity 132 (FIG. 1c).
generated from arginine gets DICER and argonaute (AGO)123, which are involved In summary, autophagy influences adaptive immune
residues by peptidylarginine in miRNA processing and function. This might correlate responses through its effects on antigen presentation,
deaminase, which is an
with the abundance of MHC class I‑associated peptides naive T cell repertoire selection, T cell homeostasis and
enzyme that may be enriched
in autophagosomes. The that seem to be derived from transcripts containing TH cell polarization. Remaining questions include whether
presence of autoantibodies in miRNA response elements (FIG. 1c). autophagy affects T cell polarization beyond TH17 cell
patients with rheumatoid responses — for example, whether it affects regulatory
arthritis correlates with the Autophagy in T cell homeostasis and TH17 cell polari- T cells and other T cell subsets — and how autophagy
levels of citrullinated antigens
(such as vimentin).
zation. In addition to its role in antigen presentation, in one cell type affects other cells and immune networks
autophagy affects both the homeostasis and the func- in specific anatomical sites. Answers to these questions
Epithelial-to‑mesenchymal tion of T cells (FIG. 1c). Autophagy and clearance of should help to explain the complex phenotypes that are
transition mitochondria are needed for normal haematopoietic associated with risk mutations in autophagy genes.
A reversal of the mesenchymal-
stem cell (HSC) maintenance and function, which
to‑epithelial transition that
occurs during development. are essential for the production of both myeloid and Secretion of immune mediators
Epithelial-to‑mesenchymal lymphoid progenitor cells124. After exiting the thymus, In addition to its intracellular actions, autophagy influ-
transition or the naive T cells depend on autophagy and mitochon- ences the extracellular release of immune mediators133–135
dedifferentiation of epithelial drial content reduction for their maturation125. T cells (FIG. 1d), some of which are stored as pre-made secretary
cells can have normal
physiological roles (such as
require maintenance of an ER that is capable of calcium granules. The defective secretion of lysozyme from
in wound healing) or can be homeostasis126. Calcium ions are sequestered in ER‑like Paneth cells in ATG16L1‑hypomorphic mice results in an
associated with fibrotic structures that accumulate in ATG7‑deficient T cells. intestinal phenotype that corresponds to that of patients
pathologies and cancer. This curtails calcium fluxes in response to TCR engage- with Crohn’s disease133. Autophagy modulates the secre-
ment 126. In naive T cells, autophagy is suppressed by tion of low-molecular-mass immune mediators such as
miRNA response elements
Sequences on RNA cellular FLICE-like inhibitory protein (CFLIP; also ATP, which function extracellularly — either directly or
transcripts that have partial known as CFLAR)127,128, but TCR signalling and CD28 following ectonucleotidase action — on purinergic P2Y
complementarity to co‑stimulation induce autophagy in activated T cells129. and P2X receptors to promote immune cell chemotaxis
microRNAs (miRNAs). miRNA Autophagy is a pro-survival process in activated T cells and inflammasome activation135. Autophagic processes
binding to the response
elements typically leads to
and counteracts the pro-apoptotic function of CD95 in senescent cells affect the secretion of IL‑6 and IL‑8
repression of target gene (also known as FAS) and CD95L (also known as FASL), through the constitutive ER‑to‑Golgi apparatus secre-
expression. which are upregulated by TCR stimulation128. tory pathway 136. Autophagy reduces immunoglobulin

734 | O CTOBER 2013 | VOLUME 13 www.nature.com/reviews/immunol

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

secretion from plasma cells and, thus, seems to be a set of adaptors, the SLRs, to eliminate invading micro­
mechanism that prevents excessive antibody produc- organisms and, in turn, pathogens have evolved strategies
tion132. The effects of autophagy on secretory func- to evade autophagic capture. It seems that as evolution
tions are not limited to immune effectors: for example, has progressed, nearly all components of the innate
autophagy factors and LC3 have a role in the exocytosis immune system, such as conventional PRRs and inflam-
of secretory lysosomes, as has been observed for a form masomes, have become integrated with autophagy. In the
of LAP during osteoclastic bone resorption137. chordate lineage, this has extended to adaptive immunity,
Autophagy may contribute to the unconventional as is best shown in mammalian systems. As a result, in
secretion of cytosolic proteins that have extracellular humans, a failure in parts of the autophagic apparatus can
immune functions. The exact mechanism by which this lead to inflammatory, autoimmune or general immune
occurs is not fully understood. IL‑1β and IL‑18 do not disorders.
have signal peptides that facilitate entry to the ER and The current knowledge of immunological autophagy
the conventional secretory pathway (ER to Golgi appa- is still in its infancy and important questions remain. As
ratus to plasma membrane). Instead, they are delivered IKKs and TBK1 regulate autophagy, why is autophagy
following inflammasome activation to the extracellular often at cross-purposes with type I IFN-mediated inflam-
environment via unconventional secretion. Although mation? Is the ubiquitylation of microbial components the
autophagy suppresses inflammasome activation under main process that leads to pathogen recognition by SLRs,
normal nutrient-rich conditions, autophagy can contrib- or does it result from a failure to detect microorganisms
ute to the unconventional secretion of IL‑1β under stress by other means? How are the recognition of microbial
conditions, for example, following nutrient starvation106. entry and the subsequent targeting of microorganisms
It is possible that autophagy functions as a double-switch for autophagy integrated through the functions of host
mechanism to repress the inflammasome under basal con- membrane damage sensing, E3 ubiquitin ligases, and pro-
ditions but to temporarily increase its physiological out- tein and lipid kinases? Are mitochondria and mitophagy
put in response to DAMPs or PAMPs during infection. the present-day manifestation of an evolutionary battle
The pro-inflammatory role of autophagy also extends to between autophagy and the bacterium endosymbiont
the unconventional secretion of IL‑18 and the DAMP precursor to mitochondria? The increased understanding
HMGB1 (REF. 106). This role depends on ATG factors of the roles of autophagy in secretion during inflamma-
and on a specialized unconventional secretion regulator tion and tissue remodelling is an important development.
Golgi reassembly-stacking protein (GRASP; for example In this context, the relationship between autophagy and
GRASP of 55 kDa (GRASP55)). GRASP is important not unconventional secretion, including the secretion of
only in unconventional secretion but also affects canonical DAMPs and of inflammasome-dependent substrates
starvation-induced autophagy in mammalian cells106. such as IL‑1β, needs further work. Finally, although early
Our understanding of the molecular and cellular progress has been made in understanding the roles of
mechanisms that are involved in the secretory role of autophagy in adaptive immunity, this area warrants
autophagy is still in its infancy. It nevertheless extends more study. Some of the questions that have recently
the idea of the importance of autophagy in the intracel- been asked in this area refer to the role of autophagy in the
lular space to the extracellular milieu, where it affects polarization of TH17 cells and possibly other T cell subsets,
tissue organization, remodelling and the delivery of along with the role of autophagy in general lymphocyte
extracellular mediators of immunity and inflammation. homeostasis and differentiation.
In summary, autophagy and immunity are fully
Conclusions integrated and our continuing study of the interface
Autophagy influences many aspects of innate and adap- between these processes will be a fruitful area of sci-
tive immunity. Indeed, it is possible that autophagy entific inquiry for many years to come. In translational
evolved as one of the first antimicrobial defences in terms, autophagy is undoubtedly an attractive target for
eukaryotic cells, and that it was shaped early on in evolu- developing new treatments in inflammatory disorders
tion from what may initially have arisen as a metabolic and autoimmunity, and may eventually be harnessed as
and quality control pathway. Autophagy uses a distinct an anti-infective mechanism.

1. Mizushima, N., Yoshimori, T. & Ohsumi, Y. The role of to autophagosomes for fusion with endosomes/ 11. Chaturvedi, A., Dorward, D. & Pierce, S. K. The B cell
atg proteins in autophagosome formation. Annu. lysosomes. Cell 151, 1256–1269 (2012). receptor governs the subcellular location of Toll-like
Rev. Cell Dev. Biol. 27, 107–132 (2011). 7. Tattoli, I. et al. Amino acid starvation induced by receptor 9 leading to hyperresponses to DNA-
2. Codogno, P., Mehrpour, M. & Proikas-Cezanne, T. invasive bacterial pathogens triggers an innate host containing antigens. Immunity 28, 799–809 (2008).
Canonical and non-canonical autophagy: variations on defense program. Cell Host Microbe 11, 563–575 12. Biswas, D. et al. ATP-induced autophagy is associated
a common theme of self-eating? Nature Rev. Mol. Cell (2012). with rapid killing of intracellular mycobacteria within
Biol. 13, 7–12 (2012). 8. Shelly, S., Lukinova, N., Bambina, S., Berman, A. & human monocytes/macrophages. BMC Immunol. 9, 35
3. Virgin, H. W. & Levine, B. Autophagy genes in Cherry, S. Autophagy is an essential component of (2008).
immunity. Nature Immunol. 10, 461–470 (2009). Drosophila immunity against vesicular stomatitis 13. Tang, D. et al. Endogenous HMGB1 regulates
4. Levine, B., Mizushima, N. & Virgin, H. W. Autophagy in virus. Immunity 30, 588–598 (2009). autophagy. J. Cell Biol. 190, 881–892 (2010).
immunity and inflammation. Nature 469, 323–335 9. Shi, C. S. & Kehrl, J. H. TRAF6 and A20 regulate lysine 14. Pilli, M. et al. TBK‑1 promotes autophagy-mediated
(2011). 63‑linked ubiquitination of Beclin‑1 to control antimicrobial defense by controlling autophagosome
5. Hamasaki, M. et al. Autophagosomes form at TLR4‑induced autophagy. Sci. Signal. 3, ra42 (2010). maturation. Immunity 37, 223–234 (2012).
ER‑mitochondria contact sites. Nature 495, 389–393 10. Nazio, F. et al. mTOR inhibits autophagy by controlling 15. Gutierrez, M. G. et al. Autophagy is a defense
(2013). ULK1 ubiquitylation, self-association and function mechanism inhibiting BCG and Mycobacterium
6. Itakura, E., Kishi-Itakura, C. & Mizushima, N. The through AMBRA1 and TRAF6. Nature Cell Biol. 15, tuberculosis survival in infected macrophages.
hairpin-type tail-anchored SNARE syntaxin 17 targets 406–416 (2013). Cell 119, 753–766 (2004).

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | O CTOBER 2013 | 735

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

16. Harris, J. et al. T helper 2 cytokines inhibit autophagic 40. Saitoh, T. & Akira, S. Regulation of innate immune 63. Jagannath, C. et al. Autophagy enhances the efficacy
control of intracellular Mycobacterium tuberculosis. responses by autophagy-related proteins. J. Cell Biol. of BCG vaccine by increasing peptide presentation in
Immunity 27, 505–517 (2007). 189, 925–935 (2010). mouse dendritic cells. Nature Med. 15, 267–276
17. Singh, S. B. et al. Human IRGM regulates autophagy and 41. Travassos, L. H. et al. Nod1 and Nod2 direct (2009).
cell-autonomous immunity functions through autophagy by recruiting ATG16L1 to the plasma 64. Yano, T. et al. Autophagic control of listeria through
mitochondria. Nature Cell Biol. 12, 1154–1165 membrane at the site of bacterial entry. Nature intracellular innate immune recognition in drosophila.
(2010). Immunol. 11, 55–62 (2010). Nature Immunol. 9, 908–916 (2008).
18. Zalckvar, E. et al. DAP-kinase-mediated phosphorylation 42. Cooney, R. et al. NOD2 stimulation induces autophagy 65. Moreau, K., Ravikumar, B., Renna, M., Puri, C. &
on the BH3 domain of beclin 1 promotes dissociation in dendritic cells influencing bacterial handling and Rubinsztein, D. C. Autophagosome precursor
of beclin 1 from Bcl‑XL and induction of autophagy. antigen presentation. Nature Med. 16, 90–97 maturation requires homotypic fusion. Cell 146,
EMBO Rep. 10, 285–292 (2009). (2010). 303–317 (2011).
19. Fabri, M. et al. Vitamin D is required for IFNγ- 43. Birmingham, C. L., Smith, A. C., Bakowski, M. A., 66. Wellcome Trust Case Control Consortium. Genome-
mediated antimicrobial activity of human Yoshimori, T. & Brumell, J. H. Autophagy controls wide association study of 14,000 cases of seven
macrophages. Sci. Transl. Med. 3, 104ra102 (2011). Salmonella infection in response to damage to the common diseases and 3,000 shared controls. Nature
This is a translationally important study showing Salmonella-containing vacuole. J. Biol. Chem. 281, 447, 661–678 (2007).
that vitamin D has a key cooperative role with IFNγ 11374–11383 (2006). 67. Lei, Y. et al. The mitochondrial proteins NLRX1 and
in inducing antimicrobial autophagy in human cells, 44. Birmingham, C. L. et al. Listeriolysin O allows Listeria TUFM form a complex that regulates type I interferon
whereas vitamin D metabolites are diminished in monocytogenes replication in macrophage vacuoles. and autophagy. Immunity 36, 933–946 (2012).
tuberculosis and HIV. Nature 451, 350–354 (2008). 68. Jounai, N. et al. NLRP4 negatively regulates
20. Mostowy, S. et al. p62 and NDP52 proteins target 45. Watson, R. O., Manzanillo, P. S. & Cox, J. S. autophagic processes through an association with
intracytosolic Shigella and Listeria to different Extracellular M. tuberculosis DNA targets bacteria beclin1. J. Immunol. 186, 1646–1655 (2011).
autophagy pathways. J. Biol. Chem. 286, for autophagy by activating the host DNA-sensing 69. Jounai, N. et al. The Atg5 Atg12 conjugate associates
26987–26995 (2011). pathway. Cell 150, 803–815 (2012). with innate antiviral immune responses. Proc. Natl
21. Van Grol, J. et al. HIV‑1 inhibits autophagy in This study shows that microbial DNA that is Acad. Sci. USA 104, 14050–14055 (2007).
bystander macrophage/monocytic cells through Src- released into the host cell cytosol can induce This is a key study that led to the appreciation of
Akt and STAT3. PLoS ONE 5, e11733 (2010). autophagy via STING, which forms an important the interference between autophagy and type I IFN
22. Shen, S. et al. Cytoplasmic STAT3 represses intersection with type I IFN signalling. It also shows signalling.
autophagy by inhibiting PKR activity. Mol. Cell 48, the in vivo role of autophagy in protection against 70. McFarlane, S. et al. Early induction of autophagy
667–680 (2012). M. tuberculosis. in human fibroblasts after infection with human
23. Scherz-Shouval, R. et al. Reactive oxygen species are 46. Yoshikawa, Y. et al. Listeria monocytogenes ActA- cytomegalovirus or herpes simplex virus 1.
essential for autophagy and specifically regulate the mediated escape from autophagic recognition. J. Virol. 85, 4212–4221 (2011).
activity of Atg4. EMBO J. 26, 1749–1760 (2007). Nature Cell Biol. 11, 1233–1240 (2009). 71. Rasmussen, S. B. et al. Activation of autophagy by
24. Huang, J. et al. Activation of antibacterial autophagy 47. Ogawa, M. et al. A Tecpr1‑dependent selective α-herpesviruses in myeloid cells is mediated by
by NADPH oxidases. Proc. Natl Acad. Sci. USA 106, autophagy pathway targets bacterial pathogens. cytoplasmic viral DNA through a mechanism
6226–6231 (2009). Cell Host Microbe 9, 376–389 (2011).
48. dependent on stimulator of IFN genes. J. Immunol.
25. Tal, M. C. et al. Absence of autophagy results in Talloczy, Z., Virgin, H. W.t. & Levine, B. PKR-dependent 187, 5268–5276 (2011).
reactive oxygen species-dependent amplification autophagic degradation of herpes simplex virus 72. Sun, L., Wu, J., Du, F., Chen, X. & Chen, Z. J.
of RLR signaling. Proc. Natl Acad. Sci. USA 106, type 1. Autophagy 2, 24–29 (2006). Cyclic GMP-AMP synthase is a cytosolic DNA sensor
2770–2775 (2009). 49. Orvedahl, A. et al. HSV‑1 ICP34.5 confers that activates the type I interferon pathway.
26. Yang, C. S. et al. Autophagy protein Rubicon mediates neurovirulence by targeting the beclin 1 autophagy Science 339, 786–791 (2013).
phagocytic NADPH oxidase activation in response protein. Cell Host Microbe 1, 23–35 (2007). 73. Zheng, Y. T. et al. The adaptor protein p62/SQSTM1
to microbial infection or TLR stimulation. 50. Lee, H. K., Lund, J. M., Ramanathan, B., targets invading bacteria to the autophagy pathway.
Cell Host Microbe 11, 264–276 (2012). Mizushima, N. & Iwasaki, A. Autophagy-dependent J. Immunol. 183, 5909–5916 (2009).
27. Sarkar, S. et al. Complex inhibitory effects of nitric viral recognition by plasmacytoid dendritic cells. 74. Yang, J. Q., Liu, H., Diaz-Meco, M. T. & Moscat, J.
oxide on autophagy. Mol. Cell 43, 19–32 (2011). Science 315, 1398–1401 (2007). NBR1 is a new PB1 signalling adapter in Th2
28. Maiuri, M. C. et al. Functional and physical interaction 51. Kyei, G. B. et al. Autophagy pathway intersects with differentiation and allergic airway inflammation
between Bcl‑XL and a BH3‑like domain in Beclin‑1. HIV‑1 biosynthesis and regulates viral yields in in vivo. EMBO J. 29, 3421–3433 (2010).
EMBO J. 26, 2527–2539 (2007). macrophages. J. Cell Biol. 186, 255–268 (2009). 75. Bjorkoy, G. et al. p62/SQSTM1 forms protein
29. Wei, Y., Pattingre, S., Sinha, S., Bassik, M. & Levine, B. 52. Gannage, M. et al. Matrix protein 2 of influenza A aggregates degraded by autophagy and has a
JNK1‑mediated phosphorylation of Bcl‑2 regulates virus blocks autophagosome fusion with lysosomes. protective effect on huntingtin-induced cell death.
starvation-induced autophagy. Mol. Cell 30, 678–688 Cell Host Microbe 6, 367–380 (2009). J. Cell Biol. 171, 603–614 (2005).
(2008). 53. Lee, H. K. et al. In vivo requirement for Atg5 in This is a seminal study that led to the identification
30. Egan, D. F. et al. Phosphorylation of ULK1 (hATG1) antigen presentation by dendritic cells. Immunity 32, of autophagic receptors and mechanisms of
by AMP-activated protein kinase connects energy 227–239 (2010). selective autophagy in mammalian cells.
sensing to mitophagy. Science 331, 456–461 54. Blanchet, F. P. et al. Human immunodeficiency virus‑1 76. Kirkin, V. et al. A role for NBR1 in autophagosomal
(2011). inhibition of immunoamphisomes in dendritic cells degradation of ubiquitinated substrates. Mol. Cell 33,
31. Sanjuan, M. A. et al. Toll-like receptor signalling in impairs early innate and adaptive immune responses. 505–516 (2009).
macrophages links the autophagy pathway to Immunity 32, 654–669 (2010). 77. Newman, A. C. et al. TBK1 kinase addiction in lung
phagocytosis. Nature 450, 1253–1257 (2007). 55. Orvedahl, A. et al. Autophagy protects against cancer cells is mediated via autophagy of Tax1bp1/
32. Henault, J. et al. Noncanonical autophagy is required sindbis virus infection of the central nervous system. Ndp52 and non-canonical NF‑κB signalling.
for type I interferon secretion in response to DNA- Cell Host Microbe 7, 115–127 (2010). PLoS ONE 7, e50672 (2012).
immune complexes. Immunity 37, 986–997 (2012). 56. Yordy, B., Iijima, N., Huttner, A., Leib, D. & Iwasaki, A. 78. Moscat, J. & Diaz-Meco, M. T. p62 at the crossroads
This is an important study that differentiates LAP A neuron-specific role for autophagy in antiviral of autophagy, apoptosis, and cancer. Cell 137,
from conventional autophagy by defining it at the defense against herpes simplex virus. Cell Host 1001–1004 (2009).
molecular level as a process that is independent of Microbe 12, 334–345 (2012). 79. Li, S. et al. Sterical hindrance promotes selectivity of
ULK1 but that is dependent on beclin 1. 57. Shoji-Kawata, S. et al. Identification of a candidate the autophagy cargo receptor NDP52 for the danger
33. Johansen, T. & Lamark, T. Selective autophagy therapeutic autophagy-inducing peptide. Nature 494, receptor galectin‑8 in antibacterial autophagy.
mediated by autophagic adapter proteins. 201–206 (2013). Science signaling 6, ra9 (2013).
Autophagy 7, 279–296 (2011). This study is a key step towards customizing the 80. Husnjak, K. & Dikic, I. Ubiquitin-binding proteins:
34. Dupont, N. et al. Shigella phagocytic vacuolar induction of autophagy without using mTOR decoders of ubiquitin-mediated cellular functions.
membrane remnants participate in the cellular inhibitors. The peptide that was designed on the Annu. Rev. Biochem. 81, 291–322 (2012).
response to pathogen invasion and are regulated by basis of the Nef-binding site on beclin 1 showed 81. von Muhlinen, N. et al. LC3C, bound selectively by
autophagy. Cell Host Microbe 6, 137–149 (2009). efficacy in various infections and in other in vitro a noncanonical LIR motif in NDP52, is required for
35. Thurston, T. L., Ryzhakov, G., Bloor, S., von and in vivo models. antibacterial autophagy. Mol. Cell 48, 329–342
Muhlinen, N. & Randow, F. The TBK1 adaptor and 58. Xu, Y. et al. Toll-like receptor 4 is a sensor for (2012).
autophagy receptor NDP52 restricts the proliferation autophagy associated with innate immunity. This was an important step in defining the roles of
of ubiquitin-coated bacteria. Nature Immunol. 10, Immunity 27, 135–144 (2007). different mammalian ATG-8 paralogues in
1215–1221 (2009). 59. Delgado, M. A., Elmaoued, R. A., Davis, A. S., Kyei, G. autophagic antimicrobial defence.
36. Wild, P. et al. Phosphorylation of the autophagy & Deretic, V. Toll-like receptors control autophagy. 82. Matsumoto, G., Wada, K., Okuno, M., Kurosawa, M.
receptor optineurin restricts Salmonella growth. EMBO J. 27, 1110–1121 (2008). & Nukina, N. Serine 403 phosphorylation of p62/
Science 333, 228–233 (2011). 60. Alonso, S., Pethe, K., Russell, D. G. & Purdy, G. E. SQSTM1 regulates selective autophagic clearance of
37. Shahnazari, S. et al. A diacylglycerol-dependent Lysosomal killing of Mycobacterium mediated by ubiquitinated proteins. Mol. Cell 44, 279–289
signaling pathway contributes to regulation of ubiquitin-derived peptides is enhanced by (2011).
antibacterial autophagy. Cell Host Microbe 8, autophagy. Proc. Natl Acad. Sci. USA 104, 83. Huett, A. et al. The LRR and RING domain protein
137–146 (2010). 6031–6036 (2007). LRSAM1 is an E3 ligase crucial for ubiquitin-
38. Thurston, T. L., Wandel, M. P., von Muhlinen, N., 61. Ponpuak, M. et al. Delivery of cytosolic components dependent autophagy of intracellular Salmonella
Foeglein, A. & Randow, F. Galectin 8 targets damaged by autophagic adaptor protein p62 endows Typhimurium. Cell Host Microbe 12, 778–790 (2012).
vesicles for autophagy to defend cells against bacterial autophagosomes with unique antimicrobial This paper identifies an E3 ubiquitin ligase that
invasion. Nature 482, 414–418 (2012). properties. Immunity 32, 329–341 (2010). recognizes cytoplasmic bacteria, that ubiquitylates
39. Deretic, V. & Levine, B. Autophagy, immunity, and 62. Kim, B. H. et al. A family of IFNγ-inducible 65‑kD autophagic targets and that cooperates with other
microbial adaptations. Cell Host Microbe 5, 527–549 GTPases protects against bacterial infection. Science receptors to guide autophagic capture and
(2009). 332, 717–721 (2011). elimination of the invading microorganisms.

736 | O CTOBER 2013 | VOLUME 13 www.nature.com/reviews/immunol

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

84. Huang, W. et al. Crystal structure and biochemical 108. Lupfer, C. et al. Receptor interacting protein kinase 132. Pengo, N. et al. Plasma cells require autophagy
analyses reveal Beclin 1 as a novel membrane binding 2‑mediated mitophagy regulates inflammasome for sustainable immunoglobulin production.
protein. Cell Res. 22, 473–489 (2012). activation during virus infection. Nature Immunol. 14, Nature Immunol. 14, 298–305 (2013).
85. Craddock, N. et al. Genome-wide association study 480–488 (2013). 133. Cadwell, K. et al. A key role for autophagy and the
of CNVs in 16,000 cases of eight common diseases 109. Castillo, E. F. et al. Autophagy protects against autophagy gene Atg16l1 in mouse and human
and 3,000 shared controls. Nature 464, 713–720 active tuberculosis by suppressing bacterial burden intestinal Paneth cells. Nature 456, 259–263 (2008).
(2010). and inflammation. Proc. Natl Acad. Sci. USA 109, This is a key study that shows the general secretory
86. Brest, P. et al. A synonymous variant in IRGM alters a E3168–E3176 (2012). defects in autophagy-deficient cells that are of
binding site for miR‑196 and causes deregulation of This paper is an in vivo demonstration of the role consequence for inflammatory bowel diseases such
IRGM-dependent xenophagy in Crohn’s disease. of autophagy in controlling both inflammation and as Crohn’s disease.
Nature Genet. 43, 242–245 (2011). bacteria during infection. 134. Ushio, H. et al. Crucial role for autophagy in
87. Orvedahl, A. et al. Image-based genome-wide siRNA 110. Paul, S., Kashyap, A. K., Jia, W., He, Y. W. & degranulation of mast cells. J. Allergy Clin. Immunol.
screen identifies selective autophagy factors. Nature Schaefer, B. C. Selective autophagy of the adaptor 127, 1267–1276 e6 (2011).
480, 113–117 (2011). protein Bcl10 modulates T cell receptor activation 135. Michaud, M. et al. Autophagy-dependent anticancer
88. Mesquita, F. S. et al. The Salmonella deubiquitinase of NF‑κB. Immunity 36, 947–958 (2012). immune responses induced by chemotherapeutic
SseL inhibits selective autophagy of cytosolic 111. Shibata, Y. et al. p47 negatively regulates IKK agents in mice. Science 334, 1573–1577 (2011).
aggregates. PLoS Pathog. 8, e1002743 (2012). activation by inducing the lysosomal degradation of 136. Narita, M. et al. Spatial coupling of mTOR and
89. Jostins, L. et al. Host-microbe interactions have polyubiquitinated NEMO. Nature Commun. 3, 1061 autophagy augments secretory phenotypes.
shaped the genetic architecture of inflammatory (2012). Science 332, 966–970 (2011).
bowel disease. Nature 491, 119–124 (2012). 112. Fliss, P. M. et al. Viral mediated redirection of NEMO/ 137. DeSelm, C. J. et al. Autophagy proteins regulate the
90. Henckaerts, L. et al. Genetic variation in the IKKγ to autophagosomes curtails the inflammatory secretory component of osteoclastic bone resorption.
autophagy gene ULK1 and risk of Crohn’s disease. cascade. PLoS Pathog. 8, e1002517 (2012). Dev. Cell 21, 966–974 (2011).
Inflamm. Bowel Dis. 17, 1392–1397 (2011). 113. Wang, C. et al. Atg16L1 deficiency confers protection 138. Chauhan, S. & Boyd, D. ZKSCAN3 is a master
91. Ramos, P. S. et al. A comprehensive analysis of from uropathogenic Escherichia coli infection in vivo. transcriptional repressor of autophagy.
shared loci between systemic lupus erythematosus Proc. Natl Acad. Sci. USA 109, 11008–11013 Mol. Cell 50, 16–28 (2013).
(SLE) and sixteen autoimmune diseases reveals (2012). 139. Settembre, C. et al. TFEB controls cellular lipid
limited genetic overlap. PLoS Genet. 7, e1002406 114. Paludan, C. et al. Endogenous MHC class II processing metabolism through a starvation-induced
(2011). of a viral nuclear antigen after autophagy. Science autoregulatory loop. Nature Cell Biol. 15, 647–658
92. Harley, J. B. et al. Genome-wide association scan in 307, 593–596 (2005). (2013).
women with systemic lupus erythematosus identifies 115. Nedjic, J., Aichinger, M., Emmerich, J., Mizushima, N. 140. Russell, R. C. et al. ULK1 induces autophagy by
susceptibility variants in ITGAM, PXK, KIAA1542 and & Klein, L. Autophagy in thymic epithelium shapes the phosphorylating Beclin‑1 and activating VPS34 lipid
other loci. Nature Genet. 40, 204–210 (2008). T‑cell repertoire and is essential for tolerance. Nature kinase. Nature Cell Biol. 15, 741–750 (2013).
93. Han, J. W. et al. Genome-wide association study in a 455, 396–400 (2008). This is a study that functionally links two key
Chinese Han population identifies nine new 116. Schmid, D., Pypaert, M. & Munz, C. Antigen-loading branches of autophagy regulators, the ULK1
susceptibility loci for systemic lupus erythematosus. compartments for major histocompatibility complex branch and the beclin 1 branch.
Nature Genet. 41, 1234–1237 (2009). class II molecules continuously receive input from 141. Takaesu, G., Kobayashi, T. & Yoshimura, A.
94. Yang, W. et al. Meta-analysis followed by replication autophagosomes. Immunity 26, 79–92 (2007). TGFβ-activated kinase 1 (TAK1)-binding proteins
identifies loci in or near CDKN1B, TET3, CD80, 117. Ireland, J. M. & Unanue, E. R. Autophagy in antigen- (TAB) 2 and 3 negatively regulate autophagy.
DRAM1, and ARID5B as associated with systemic presenting cells results in presentation of J. Biochem. 151, 157–166 (2012).
lupus erythematosus in Asians. Am. J. Hum. Genet. citrullinated peptides to CD4 T cells. J. Exp. Med. 142. Zalckvar, E., Berissi, H., Eisenstein, M. & Kimchi, A.
92, 41–51 (2013). 208, 2625–2632 (2011). Phosphorylation of Beclin 1 by DAP-kinase promotes
95. Martin, L. J. et al. Functional variant in the autophagy- 118. Wildenberg, M. E. et al. Autophagy attenuates autophagy by weakening its interactions with Bcl‑2
related 5 gene promotor is associated with childhood the adaptive immune response by destabilizing the and Bcl‑XL. Autophagy 5, 720–722 (2009).
asthma. PLoS ONE 7, e33454 (2012). immunologic synapse. Gastroenterology 142, 143. Criollo, A. et al. Inhibition of autophagy by TAB2
96. Raychaudhuri, S. et al. Genetic variants at CD28, 1493–1503 e6 (2012). and TAB3. EMBO J. 30, 4908–4920 (2011).
PRDM1 and CD2/CD58 are associated with 119. Akalay, I. et al. Epithelial-to‑mesenchymal transition 144. Radtke, A. L., Delbridge, L. M., Balachandran, S.,
rheumatoid arthritis risk. Nature Genet. 41, and autophagy induction in breast carcinoma promote Barber, G. N. & O’Riordan, M. X. TBK1 protects
1313–1318 (2009). escape from T‑cell-mediated lysis. Cancer Res. 73, vacuolar integrity during intracellular bacterial
97. Zhao, H. et al. Mice deficient in Epg5 exhibit selective 2418–2427 (2013). infection. PLoS Pathog. 3, e29 (2007).
neuronal vulnerability to degeneration. J. Cell Biol. 120. Wenger, T. et al. Autophagy inhibition promotes 145. Ku, B. et al. Structural and biochemical bases for the
200, 731–741 (2013). defective neosynthesized proteins storage in ALIS, and inhibition of autophagy and apoptosis by viral BCL‑2
98. Cullup, T. et al. Recessive mutations in EPG5 cause induces redirection toward proteasome processing of murine γ-herpesvirus 68. PLoS Pathog. 4, e25
Vici syndrome, a multisystem disorder with defective and MHCI-restricted presentation. Autophagy 8, (2008).
autophagy. Nature Genet. 45, 83–87 (2013). 350–363 (2012). 146. Choy, A. et al. The Legionella effector RavZ inhibits
99. Yang, C. S. et al. The autophagy regulator Rubicon is 121. Fiegl, D. et al. Amphisomal route of MHC class I host autophagy through irreversible Atg8
a feedback inhibitor of CARD9‑mediated host innate cross-presentation in bacteria-infected dendritic cells. deconjugation. Science 338, 1072–1076 (2012).
immunity. Cell Host Microbe 11, 277–289 (2012). J. Immunol. 190, 2791–2806 (2013). This is an elegant recent example of how bacteria
100. Saitoh, T. et al. Atg9a controls dsDNA-driven 122. Tey, S. K. & Khanna, R. Autophagy mediates defend themselves against autophagy by
dynamic translocation of STING and the innate transporter associated with antigen processing- preventing LC3 lipidation, which thus stalls the
immune response. Proc. Natl Acad. Sci. USA 106, independent presentation of viral epitopes through autophagic response.
20842–20846 (2009). MHC class I pathway. Blood 120, 994–1004 147. Dortet, L. et al. Recruitment of the major vault protein
101. Li, S., Wang, L., Berman, M., Kong, Y. Y. & Dorf, M. E. (2012). by InlK: a Listeria monocytogenes strategy to avoid
Mapping a dynamic innate immunity protein 123. Gibbings, D. et al. Selective autophagy degrades autophagy. PLoS Pathog. 7, e1002168 (2011).
interaction network regulating type I interferon DICER and AGO2 and regulates miRNA activity. 148. Niu, H., Xiong, Q., Yamamoto, A., Hayashi-Nishino, M.
production. Immunity 35, 426–440 (2011). Nature Cell Biol. 14, 1314–1321 (2012). & Rikihisa, Y. Autophagosomes induced by a bacterial
102. Saitoh, T. et al. Loss of the autophagy protein 124. Mortensen, M. et al. The autophagy protein Atg7 is Beclin 1 binding protein facilitate obligatory
Atg16L1 enhances endotoxin-induced IL‑1β essential for hematopoietic stem cell maintenance. intracellular infection. Proc. Natl Acad. Sci. USA 109,
production. Nature 456, 264–268 (2008). J. Exp. Med. 208, 455–467 (2011). 20800–20807 (2012).
This is a key study that recognizes the 125. Pua, H. H., Guo, J., Komatsu, M. & He, Y. W. 149. Ogawa, M. et al. Escape of intracellular Shigella
anti-inflammatory role of autophagy. Autophagy is essential for mitochondrial clearance in from autophagy. Science 307, 727–731 (2005).
103. Rathinam, V. A., Vanaja, S. K. & Fitzgerald, K. A. mature T lymphocytes. J. Immunol. 182, 4046–4055 150. Gregoire, I. P. et al. IRGM is a common target of
Regulation of inflammasome signaling. Nature (2009). RNA viruses that subvert the autophagy network.
Immunol. 13, 333–332 (2012). 126. Jia, W., Pua, H. H., Li, Q. J. & He, Y. W. Autophagy PLoS Pathog. 7, e1002422 (2011).
104. Zhou, R., Yazdi, A. S., Menu, P. & Tschopp, J. A role regulates endoplasmic reticulum homeostasis and
for mitochondria in NLRP3 inflammasome activation. calcium mobilization in T lymphocytes. J. Immunol. Acknowledgements
Nature 469, 221–225 (2011). 186, 1564–1574 (2011). We apologize to our colleagues whose work or specific studies
This is a conceptually important study that shows 127. Lee, J. S. et al. FLIP-mediated autophagy regulation in have not been cited. This is not a reflection of lack of interest
that housekeeping functions, such as the autophagic cell death control. Nature Cell Biol. 11, 1355–1362 on the authors’ part or on the significance for the field but
clearance of depolarized mitochondria when they are (2009). was dictated by the article scope and reference number limi-
failing, can lead to endogenous sterile inflammation. 128. He, M. X. & He, Y. W. A role for c‑FLIPL in the regulation tations. V.D. is supported by US National Institutes of Health
105. Nakahira, K. et al. Autophagy proteins regulate of apoptosis, autophagy, and necroptosis in grant AI042999 and by a grant from the Bill and Melinda
innate immune responses by inhibiting the release T lymphocytes. Cell Death Differ. 20, 188–197 Gates Foundation.
of mitochondrial DNA mediated by the NALP3 (2013).
inflammasome. Nature Immunol. 12, 222–230 129. Hubbard, V. M. et al. Macroautophagy regulates Competing interests statement
(2011). energy metabolism during effector T cell activation. The authors declare no competing financial interests.
106. Dupont, N. et al. Autophagy-based unconventional J. Immunol. 185, 7349–7357 (2010).
secretory pathway for extracellular delivery of IL‑1β. 130. Miller, B. C. et al. The autophagy gene ATG5 plays an
EMBO J. 30, 4701–4711 (2011). essential role in B lymphocyte development. DATABASES
107. Shi, C. S. et al. Activation of autophagy by Autophagy 4, 309–314 (2008). Expressed sequence tags NCBI database:
inflammatory signals limits IL‑1β production by 131. Arsov, I. et al. A role for autophagic protein beclin 1 http://www.ncbi.nlm.nih.gov/nucest
targeting ubiquitinated inflammasomes for early in lymphocyte development. J. Immunol. 186, ALL LINKS ARE ACTIVE IN THE ONLINE PDF
destruction. Nature Immunol. 13, 255–263 (2012). 2201–2209 (2011).

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | O CTOBER 2013 | 737

© 2013 Macmillan Publishers Limited. All rights reserved

S-ar putea să vă placă și