Available online at www.sciencedirect.com

ScienceDirect Emerging themes in bacterial autophagy Matthew T Sorbara1 and Stephen E Girardin2 The role of autophagy in the control of intracellular bacterial pathogens, also known as xenophagy, is well documented. Here, we highlight recent advances in the field of xenophagy. We review the importance of bacterial targeting by ubiquitination, diacylglycerol (DAG) or proteins such as Nod1, Nod2, NDP52, p62, NBR1, optineurin, LRSAM1 and parkin in the process of xenophagy. The importance of metabolic sensors, such as mTOR and AMPK, in xenophagy induction is also discussed. We also review the in vitro and in vivo evidence that demonstrate a global role for xenophagy in the control of bacterial growth. Finally, the mechanisms evolved by bacteria to escape xenophagy are presented. Addresses 1 Department of Immunology, University of Toronto, Toronto M5S 1A8, Canada 2 Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada Corresponding author: Girardin, Stephen E ([email protected])

Current Opinion in Microbiology 2015, 23:163–170 This review comes from a themed issue on Host-microbe interactions: bacteria Edited by David Holden and Dana Philpott

http://dx.doi.org/10.1016/j.mib.2014.11.020 1369-5274/# 2014 Elsevier Ltd. All rights reserved.

the cytosolic side of late endosomes and lysosomes, known as the mTOR complex 1 (mTORC1) [2]. mTORC1 is assembled and functional only when cellular nutrients or cofactors (including amino acids, growth factors, oxygen and ATP) are not limiting, and active mTORC1 in turns phosphorylates several cellular proteins involved in cell proliferation, growth, ribosomal biogenesis and mRNA translation [2]; in addition, active mTORC1 potently inhibits autophagy by promoting the phosphorylation of the ULK1 complex, including ULK1 itself and ATG13 [3]. The kinase AMPK, which is another sensor of cellular energy (and in particular of the AMP/ATP ratio), also regulates autophagy through the phosphorylation of the ULK1 complex, although the global effect of AMPK on autophagy might depend on the trigger (glucose versus amino acid starvation) [3]. In addition to its role in cellular metabolism, numerous lines of evidence indicate that autophagy also plays a key role in innate immunity, by promoting the entrapment and lysosome-mediated degradation of bacterial pathogens, including bacteria that escape into the host cytosol (such as Shigella or Listeria) or those that reside in a modified intracellular vacuole (such as Salmonella and Mycobacterium). While several comprehensive reviews have detailed the role of autophagy in host defense also known as ‘xenophagy’ [4–6], we will highlight here the most recent advances in this rapidly expanding field of research, and present our views on the most important questions that remain to be solved in the coming years.

Bacterial targeting in xenophagy

Introduction Autophagy is an essential cellular process through which cellular materials, such as large macromolecular complexes or defective organelles, are progressively engulfed by a double membrane, or phagophore, resulting in the formation of an autophagosome [1]. After fusion with lysosomes, the constituents are degraded and metabolites recycled. While a constitutive level of autophagy is necessary for normal cellular housekeeping functions, autophagy can be strongly upregulated in conditions of metabolic starvation in order to provide a spare source of metabolites for a limited time before cell death ensues. Several signaling pathways contribute to the regulation of autophagy by metabolic checkpoints [1]. One of the most conserved of those is dependent on the kinase Tor (mTOR in mammals), which forms a large complex on www.sciencedirect.com

An active area of research in the field of xenophagy relates to the question of how infected cells can specifically direct the phagophore to bacteria that are either free in the cytosol or confined into a vacuole, while avoiding to target other cellular constituents, such as organelles. Four main molecular mechanisms are currently known to confer bacterial targeting during xenophagy, which involve: Nod proteins, ubiquitination, galectin-dependent pathway and diacylglycerol (DAG). Nod1 and Nod2: the Nod-like receptor (NLR) proteins Nod1 and Nod2 are cytosolic proteins that detect specific fragments derived from bacterial peptidoglycan and initiate pro-inflammatory signaling (NF-kB and MAP kinase pathways) following the recruitment of the adaptor protein Rip2 [7]. In the initial phase of bacterial invasion, Nod1/2 also directly interacts with the autophagy protein ATG16L1 in a Rip2-independent manner, thereby initiating the recruitment of the autophagic machinery at the site of bacterial entry [8]. However, it remains unclear if Current Opinion in Microbiology 2015, 23:163–170

164 Host-microbe interactions: bacteria

Nod1/2 can target bacteria (such as Shigella or Listeria) once they are free in the cytosol. Ubiquitination: it was observed that cytosolic and vacuoleconfined bacteria are rapidly decorated by ubiquitination [9], and different types of ubiquitin chains constitute the ubiquitin coat surrounding bacteria [10,11,12]. Several adaptor proteins, including NDP52, p62, NBR1 and optineurin, have been shown to serve as a bridge between ubiquitin groups and the autophagy protein LC3 [13]. At least some of these adaptor proteins likely serve nonredundant functions, as evidenced by the reported effect of targeting NDP52 [14], p62 [15] or optineurin [16] on bacterial restriction. The nature of the proteins targeted by ubiquitination, and even if those are from the host or the bacteria, remains unclear. However, recent progress was made with the identification of E3 ligases implicated in this ubiquitination process. LRSAM1 was identified as an important E3 ligase in the case of infection with Salmonella [17]. More recently, the ubiquitin ligase parkin was shown to be required for the formation of the ubiquitin coat surrounding Mycobacterium tuberculosis, and for the restriction of M. tuberculosis growth in macrophages [11]. Interestingly, the ubiquitin ligase activity of parkin has a well-established role in mitophagy, and mutations in the human gene encoding parkin lead to increased susceptibility to Parkinson’s disease, but also to infections by Mycobacterium Leprae and Salmonella enterica serovar Typhi, suggesting links between xenophagy and mitophagy, and between susceptibility to neurodegenerative disorders and to infectious diseases [13]. In addition, a role for the intracellular DNA sensor STING in the formation of the ubiquitin coat surrounding M. tuberculosis was identified [18], suggesting that STING and parkin might operate in the same pathway. Finally, S-guanylation of cysteine residues on the surface of intracellular Group A streptococcus by 8-nitro-cGMP was shown to drive polyubiquitination and autophagy [19]. Exposure of galectin proteins: the membrane remnants of vacuolar rupture in Shigella-infected cells were found to accumulate ubiquitinated proteins and autophagy markers [20], suggesting that the event of membrane damage could trigger xenophagy. In agreement, Thurston et al. demonstrated that galectin proteins, such as galectin-8/3/9, which are glycan-binding lectin proteins that can be found in the cytosol, accumulated at sites of membrane damage induced by either Salmonella or by glycyl-L-phenylalanine 2-naphthylamide (GPN), a molecule inducing sterile lysosomal damage [14]. Moreover, galectin-8 recruited NDP52 to the Salmonella-containing vacuole (SCV) through a mechanism distinct and more rapid than the ubiquitinmediated recruitment of NDP52 to the SCV [14]. Thus, galectin proteins might serve as important sensors of danger signals initiated at the damaged membrane by the exposure of glycans to the cytosol, while those glycans are normally only exposed to the luminal side of vesicles. Current Opinion in Microbiology 2015, 23:163–170

DAG: in Salmonella-infected cells, Shahnazari et al. observed early accumulation of DAG around the SCV, and inhibition of DAG formation using pharmacological or genetic approaches resulted in inhibition of bacterial autophagy [21]. While the authors proposed that DAGdependent and ubiquitin-dependent pathways of autophagy targeting to the SCV were independent, the role of DAG in Nod1/2-dependent or galectin/NDP52-dependent xenophagy remains to be addressed.

Mechanisms of xenophagy induction While much attention was given to the mechanisms responsible for bacterial targeting by autophagy (see above), little is known about how autophagy pathways are turned on during infection. Until recently, it was assumed that the constitutive levels of housekeeping autophagy were likely sufficient to drive xenophagy in infected cells. However, it seems counter-intuitive that a critical innate immune mechanism of bacterial restriction would be limited by nutrient sufficiency and the metabolic status of the infected cells; indeed, metabolic sensors, such as mTOR, play key roles in limiting autophagy. Studies in epithelial cell lines demonstrated that Shigella, Listeria and Salmonella all triggered a rapid inhibition of mTOR signaling, which is associated with a burst of autophagy in infected cells, thus showing that xenophagy induction can occur irrespective of the nutrient status of the infected cells [22–24]. Mechanistically, the inhibition of mTOR was driven by amino acid starvation (AAS) caused by membrane damage in the infected cells, resulting in mTOR dissociation from late endosome/lysosome membranes [23]. While Shigella caused a sustained inhibition of mTOR and induction of AAS pathways, infection with Salmonella or Listeria resulted in transient AAS stress and mTOR inhibition, a mechanism that likely accounts, at least in part, for the capacity of these bacteria to escape xenophagy-mediated bacterial restriction [23]. Although the mechanism by which bacteriainduced membrane damage causes AAS is unknown, it is interesting to note that bacterial pore-forming toxins, which induce severe membrane damage, have also been shown to inhibit mTOR and to turn on AAS pathways [25,26]. Moreover, recent evidence showed that the tyrosine kinase focal adhesion kinase (FAK) was recruited to the surface of the SCV in macrophages, resulting in increased activity of the mTORC1 through an unknown mechanism dependent on the Salmonella Pathogenicity Island 2 (SPI-2) [27]. This subversion mechanism thereby amplified autophagy inhibition through mTOR induction.mTOR is not the only critical regulator of nutrient status that influences the autophagy pathway. As mentioned above, AMPK also emerges as a potential key player in these pathways, but the role of this energy sensor in the context of host responses to bacterial pathogens remains poorly characterized. A recent study demonstrated that, in M. tuberculosis infected macrophages, www.sciencedirect.com

Emerging themes in bacterial autophagy Sorbara and Girardin 165

AMPK induction by the drug AICAR was sufficient to promote antibacterial autophagy, in part through the transcriptional upregulation of autophagy-associated genes [28]. However, it remains unknown if bacterial pathogens can directly activate AMPK in infected cells, thereby affecting xenophagy.

Role of xenophagy in bacterial restriction Substantial work in cell lines and primary cell cultures has identified and supported a bactericidal role for autophagy (see Table 1). Interestingly, several groups have found that wild type strains of bacteria that have evolved to replicate in the cytoplasm, including S. flexneri, Francisella tularensis, and L. monocytogenes are not restricted by autophagy. Strikingly, only attenuated strains, lacking specific virulence factors, are efficiently targeted and degraded in autolysosomes. In contrast, bacteria species that reside and replicate within damaged-vacuoles, including M. tuberculosis or S. typhimurium, generally appear to be more efficiently restricted by autophagy (Table 1). In in vitro infection models where loss or stimulation of autophagy does impact on bacterial replication, the loss of autophagy generally increases bacterial replication by less than an order of magnitude with typical values of 2–5 the replication observed in autophagy competent cells. Furthermore, pathogens adapted to an intracellular lifestyle are still able to replicate in autophagy competent cells, and, in the case of bacteria such as Shigella and Listeria, spread from cell to cell. Therefore, a major question in the field has been whether the slight delay or decrease in intracellular replication within a cell translates to a substantial effect at the whole organism level. Several studies in non-mammalian model organisms have identified a critical role for autophagy in the control of bacterial infection. Seminal work from Yano et al. in Drosophila demonstrated that inhibition of autophagy through loss of either ATG5 or the peptidoglycan recognition protein, PGRL-LE, results in a dramatic increase in susceptibility to L. monocytogenes infection [29]. More recently, a critical role for the ubiquitin ligase parkin has been identified in clearance of S. typhimurium, M. marinum and L. monocytogenes in Drosophila [11]. Mostowy et al. recently proposed a zebrafish larvae model for studying the role of autophagy in Shigella infection, and found that loss of the autophagy adaptor p62 results in increased susceptibility to S. flexneri infection [30]. Finally, loss of core autophagy components in either Caenorhabditis elegans or the single cell Dictyostelium triggered increased susceptibility to S. typhimurium infection [31]. Therefore, in non-mammalian systems both the core components of the autophagy machinery (ATG proteins) and the components specific to xenophagy or subsets of autophagy (PGRP-LE, parkin, p62) have been implicated in host defense. However, it must be noted that these infection models in which xenophagy was shown to confer control of intracellular bacteria relied on the interaction between www.sciencedirect.com

bacterial pathogens and host cells that are not the natural targets of these bacteria, raising the possibility that xenophagy could mainly restrict pathogens that are not adapted to their host environment. Recent experiments using murine models of infection have supported a role for autophagy in bacterial pathogen clearance. Mice lacking expression of ATG5 or ATG16L1 in intestinal epithelial cells had increased Salmonella translocation to systemic sites and increased bacterial load [32,33]. Furthermore, mice lacking macrophage expression of ATG5 were more susceptible to M. tuberculosis or L. monocytogenes infection [18,34]. One caveat of these studies is that they do not exclude contributions from the autophagy machinery beyond direct bactericidal activity, such as maintenance of host cell fitness by controlling the turnover of cellular organelles and macromolecular complexes. Interestingly, during infection of mice with the extracellular pathogen Citrobacter rodentium, decreased expression of ATG16L1 triggered a protective hyper-inflammatory response, and a similar phenotype was observed following UPEC infection (see Table 1). In agreement, ATG16L1 was found to negatively regulate Nod1/2-dependent induction of proinflammatory pathways in an autophagy-independent manner [35]. Therefore, further work is needed to clarify the relative contributions of direct bactericidal activity and ancillary functions in the immune response.

Bacterial escape mechanisms from xenophagy One of the most compelling pieces of evidence that xenophagy is a critical innate immune mechanism of bacterial restriction comes from the identification of bacterial strategies that were evolved to escape host autophagy. Schematically, two main escape mechanisms seem to have been designed by bacterial pathogens: autophagy disarming and camouflage. Autophagy disarming: several bacteria have evolved means to inhibit autophagy to favor their intracellular replication. This is the case of M. tuberculosis, which is targeted by autophagy only after addition of rapamycin [36], suggesting that mTOR activity in infected cells is sufficient to turn down xenophagy. In addition, the ESX-1 secretion system was shown to specifically inhibit autophagosome-lysosome fusion, a key step in xenophagy [37]. Salmonella and Listeria also escape autophagy at least in part through the rapid reactivation of mTOR after an initial phase of inhibition caused by membrane damage (see above) [22,23]. A sophisticated example of autophagy disarming was provided in the case of Legionella pneumophila with the discovery that the effector RavZ could cleave the C-terminal end of lipid-conjugated LC3, thus irreversibly preventing the reconjugation of the protein to autophagosomes and thereby inhibiting autophagy [37]. Finally, Listeria phospholipases C were shown to stall Current Opinion in Microbiology 2015, 23:163–170

166 Host-microbe interactions: bacteria

Table 1 Autophagy-dependent restriction of bacterial replication. Evidence of a role for autophagy in restricting the proliferation of different model pathogens is indicated with tissue culture models of infection shaded in green, and in vivo infection models shaded in gray Species Group A Streptococcus (GAS)

Primary observation Restriction in xenophagy competent cells

Increased restriction through autophagy stimulation Increased mortality and bacterial load with increased autophagy Mycobacterium species

Restriction in xenophagy competent cells

Restriction with artificial autophagy induction Decreased survival, increased pathology and bacterial load Decreased survival Increased bacterial load, inflammation

Shigella flexneri

Restriction of DIcsB strain No restriction of wild type Shigella No restriction of wild type with autophagy induction Restriction of wild type Shigella, increased difference with DIcsB strain Increased mortality

Salmonella typhimurium

Restriction in xenophagy competent cells

Protection from GAS infection in RNF5 / [46]

mice

Knockdown in macrophage/monocyte lines: IRGM [51], Rubicon [52], parkin [11], NDP52 [18] Knockout cells: PARK2 / BMDMs [11], STING / , ATG5 / , TBK1 / BMDMs [18] RAW264.7 and primary macrophages [36], ATG5 fl/ fl and ATG5 fl/fl x Lyz-Cre BMDM [34] M. tuberculosis infection of ATG5 fl/fl LyzM-Cre mice [18] M. marinum infection of parkin-deficient Drosophila [11] M. marinum infection of p62, or DRAM1 deficient (morpholino treated) zebrafish embryos. Decreased bacterial load with DRAM1 overexpression [53] Wild type and ATG5 / MEFs [38], Tecpr1 siRNA in HeLa, Tecpr1 / MEFs [44] LC3C siRNA, NDP52 siRNA in HeLa [54] Rapamycin treatment HeLa [23] Control compared to ATG16L1 T300A MEFs, primary intestinal epithelial cells (24821797) p62 deficient (morpholino treated) zebrafish embryos [30]

Increased bacterial translocation, and intestinal inflammation Increased intestinal inflammation with ATG16L1 variant expression

MEFs: wild type compared to ATG5 / [55], PKCd / [21], ATG7 / [56], ATG16L1-deficient [57] HeLa knockdown studies: control compared to NDP52, Gal8 [14], WIPI2 [58], LC3C, NDP52 [54], LRSAM1, ATG16L1 [17], OPTN [16], NDP52, TBK1 [59], p62 [15], PKCd [21], FNBP1L [60] silenced cells U2OS knockdown studies: control compared to Barkor siRNA [56] Henle-407 knockdown studies: control compared to ATG12 or P22 knockdowns [61] LC3, p62, ATG5 siRNAs in HeLa [62] Rapamycin treatment and ATG16L1 shRNA in HeLa [23], MDP treatment and Nod2, Rip2, ATG16L1, or ULK1 knockdown in HCT116 [63,64] ATG7, BEC-1, or IGG-1 RNAi-fed C. elegans. Atg1 , Atg6 , or Atg7 Dictyostelium [31] Parkin-deficient Drosophila [11] Oral Salmonella infection of WT and FAK fl/fl LyzMCre mice [27] Oral Salmonella infection of ATG5fl/fl x villin Cre mice [32] Salmonella streptomycin-pretreatment infection model in ATG16L1fl/fl x villin Cre mice [33] Salmonella streptomycin-pretreatment infection model in ATG16L1 T300A mice [65]

Restriction of O-antigen mutants

ATG5 fl/fl Lyz-Cre macrophages [66]

Enhances replication Increased restriction through autophagy stimulation Decreased survival, increased bacterial load

Decreased bacterial load and pathology with increased autophagy Increased bacterial translocation

F. tularensis

Model system Knockout cells: wild type and ATG5 / MEFs [45]. Wild type and RNF5 / BMDMs [46] Knockdown in HeLa: Rab9a, Rab23 [47], Rab7 [48], ATG5, CD46 (cyt-1 isoform), GOPC [49], VAMP8 VTI1b [50] 8-Nitro-cGMP treatment in RAW264.7 cells [19]

Current Opinion in Microbiology 2015, 23:163–170

www.sciencedirect.com

Emerging themes in bacterial autophagy Sorbara and Girardin 167

Table 1 (Continued ) Species L. monocytogenes

Primary observation

Model system

Restriction of wild type Listeria in xenophagy competent cells

PTPN2 knockdown in T84 and primary human colon lamina propria fibroblasts with PTPN2 variant [67], wild type and ATG5 / MEFs [68], Rubicon knockdown in THP-1, BMDM, and RAW264.7 [52], NOD2 / or TLR2 / BMDM [69] DPrfA infection of wild type and ATG5 / MEFs [70]. DActA infection of wild type and ATG5 / MEFs or p62 / and p62 / + p62 MEFs [39] Dependent on host MVP expression (siRNA in RAW264.7) [40] Wildtype and autophagy deficient ATG16L1 DCCD MEFs [22] Parkin-deficient [11], PGRP-LE deficient, or ATG5 siRNA [29] Drosophila Intraperitoneal challenge of ATG5 fl/fl and ATG5 fl/fl x Lyz-Cre mice [34]

No restriction of wild type Listeria but restriction of mutant Listeria in xenophagy competent cells. Decreased autophagy with InlK expression in DActA strain Wild type Listeria protects co-infecting DPlcA/B bacteria in-trans Increased bacterial load, decreased survival Increased bacterial load, decreased survival C. rodentium Yersinia enterocolitica

Decreased replication in autophagy deficient Restriction in xenophagy competent cells

ATG16L1 hypomorphic mice [71] ATG16L1 T300A human macrophages, or T316A murine macrophages [72]

E. coli

Restriction in xenophagy competent cells

AIEC infection in wild type and ATG5 / MEFs, ATG16L1 knockdown HeLa cells [73], ATG16L1 siRNA in THP-1 macrophages, wild type or NOD2 / murine primary macrophages [74]. miR-30c or miR-130a upregulation following AIEC infection in T84 [75] ATG16L1 hypomorphic mice infected with UPEC [76]

Increased replication with autophagy downregulation Decreased replication in autophagy deficient

early phagophore structures by inhibiting autophagic flux, resulting in bacterial escape from autophagy [22]. Camouflage: one of the first examples of xenophagy escape by camouflage came with the observation that Shigella protein IcsB efficiently prevented the entrapment of Shigella into autophagosomes [38]. The proposed mechanism suggested that IcsB interacted with VirG, another Shigella protein, thereby competitively inhibiting the interaction of the autophagy protein ATG5 to VirG [38]. In a similar fashion, Listeria protein ActA was shown to favor the recruitment of the Arp2/3 complex and VASP around bacteria, thereby masking bacterial surface and restricting access to autophagy adaptor proteins such as p62, resulting in decreased autophagy targeting [39]. The Listeria protein InlK also contributes to autophagy escape by recruiting the host protein Major Vault Protein at the bacterial surface [40]. Finally, the serotype M1T1 of Group A Streptococcus expresses a protease that cleaves p62, NDP52 and NBR1, thus limiting targeting by the autophagic machinery [41].

Conclusions and outstanding questions It is now clear that targeting of intracellular bacteria by the autophagic system, or xenophagy, plays an essential role in innate immunity. Despite considerable recent progress in the field of xenophagy, many unanswered questions remain: Why are there several bacterial targeting mechanisms for xenophagy (Nods/ubiquitin/galectins/DAG) and are they www.sciencedirect.com

inter-connected to increase sensitivity or specificity? Similarly, what are the relative contributions of the adaptor proteins that display domains of interaction with ubiquitinated proteins and LC3, such as p62, NDP52, NBR1 and optineurin? Finally, since ATG5-12-16L1 is required to target LC3 to the bacterial autophagosome, how critical is the capacity of p62, NDP52, NBR1 and optineurin to recruit LC3? Besides Nod1 and Nod2, are there any factors that are truly specific for xenophagy, as opposed to other processes of selective autophagy? Indeed, NBR1, p62 and ubiquitination are critical for the clearance of aggregated proteins [42,43], NDP52 and galectin-8 are recruited to damaged membranes even in the absence of infection [14], optineurin associates with LC3-positive vesicles upon nutrient deprivation [16], and Tecpr1 is involved in selective autophagy targeting pathogens, aggresomes and mitochondria [44]. Therefore, is the role of Nod proteins to direct towards bacteria an autophagic machinery that would be triggered by non-microbial signals, such as membrane damage and accumulation of ubiquitinated molecules? In any given cell, autophagy typically only targets a small fraction of the intracellular bacteria. If the same cause produces the same effect, why would not all the bacteria be targeted? Is there an unknown feature that would make a specific bacterium more prone to targeting by autophagy than others? Current Opinion in Microbiology 2015, 23:163–170

168 Host-microbe interactions: bacteria

How important xenophagy is for bacterial growth restriction as compared to other innate immune defense mechanisms, such as for instance phagocytosis? This is a difficult question since cellular models of defective xenophagy usually either target general or selective autophagy proteins, which both result in altered cellular fitness, potentially leading to indirect effects on the cell’s capacity to control bacterial pathogens. Answering this question will require the identification of factors involved in xenophagy, but not in general or other selective autophagy pathways.

Acknowledgements Research in the lab of SEG is supported by grants from Canadian Institutes of Health Research (CIHR), Natural Sciences and Engineering Research Council (NSERC), Canadian Cancer Society Research Institutes and Crohn’s and Colitis Canada. MTS. was supported by fellowships from the NSERC (CGS-M), the Ontario Graduate Scholarship (OGS), CIHR (CGSD) and by CIHR Strategic Training Fellowship STP-53877.

References and recommended reading Papers of particular interest, published within the period of review, have been highlighted as:  of special interest  of outstanding interest 1.

Yang Z, Klionsky DJ: Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol 2010, 22:124-131.

2.

Laplante M, Sabatini DM: mTOR signaling in growth control and disease. Cell 2012, 149:274-293.

3.

Wong PM, Puente C, Ganley IG, Jiang X: The ULK1 complex: sensing nutrient signals for autophagy activation. Autophagy 2013, 9:124-137.

4.

Huang J, Brumell JH: Bacteria-autophagy interplay: a battle for survival. Nat Rev Microbiol 2014, 12:101-114.

5.

Gomes LC, Dikic I: Autophagy in antimicrobial immunity. Mol Cell 2014, 54:224-233.

6.

Deretic V, Saitoh T, Akira S: Autophagy in infection, inflammation and immunity. Nat Rev Immunol 2013, 13:722-737.

7.

Philpott DJ, Girardin SE: Nod-like receptors: sentinels at host membranes. Curr Opin Immunol 2010, 22:428-434.

8.

Travassos LH, Carneiro LA, Ramjeet M, Hussey S, Kim YG, Magalhaes JG, Yuan L, Soares F, Chea E, Le Bourhis L et al.: Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry. Nat Immunol 2010, 11:55-62.

9.

Perrin AJ, Jiang X, Birmingham CL, So NS, Brumell JH: Recognition of bacteria in the cytosol of mammalian cells by the ubiquitin system. Curr Biol 2004, 14:806-811.

10. van Wijk SJ, Fiskin E, Putyrski M, Pampaloni F, Hou J, Wild P, Kensche T, Grecco HE, Bastiaens P, Dikic I: Fluorescence-based sensors to monitor localization and functions of linear and K63-linked ubiquitin chains in cells. Mol Cell 2012, 47:797-809. 11. Manzanillo PS, Ayres JS, Watson RO, Collins AC, Souza G,  Rae CS, Schneider DS, Nakamura K, Shiloh MU, Cox JS: The ubiquitin ligase parkin mediates resistance to intracellular pathogens. Nature 2013, 501:512-516. Demonstrates that Parkin deficiency results in increased susceptibility to infection with intracellular bacteria in mice and flies and the importance of K63-linked ubiquitin targeting for autophagic restriction. 12. Collins CA, De Maziere A, van Dijk S, Carlsson F, Klumperman J, Brown EJ: Atg5-independent sequestration of ubiquitinated mycobacteria. PLoS Pathog 2009, 5:e1000430. Current Opinion in Microbiology 2015, 23:163–170

13. Randow F, Youle RJ: Self and nonself: how autophagy targets mitochondria and bacteria. Cell Host Microbe 2014, 15:403-411. 14. Thurston TL, Wandel MP, von Muhlinen N, Foeglein A, Randow F: Galectin 8 targets damaged vesicles for autophagy to defend cells against bacterial invasion. Nature 2012, 482:414-418. 15. Zheng YT, Shahnazari S, Brech A, Lamark T, Johansen T, Brumell JH: The adaptor protein p62/SQSTM1 targets invading bacteria to the autophagy pathway. J Immunol 2009, 183:59095916. 16. Wild P, Farhan H, McEwan DG, Wagner S, Rogov VV, Brady NR, Richter B, Korac J, Waidmann O, Choudhary C et al.: Phosphorylation of the autophagy receptor optineurin restricts Salmonella growth. Science 2011, 333:228-233. 17. Huett A, Heath RJ, Begun J, Sassi SO, Baxt LA, Vyas JM, Goldberg MB, Xavier RJ: The LRR and RING domain protein LRSAM1 is an E3 ligase crucial for ubiquitin-dependent autophagy of intracellular Salmonella Typhimurium. Cell Host Microbe 2012, 12:778-790. 18. Watson RO, Manzanillo PS, Cox JS: Extracellular M. tuberculosis DNA targets bacteria for autophagy by activating the host DNA-sensing pathway. Cell 2012, 150:803-815. 19. Ito C, Saito Y, Nozawa T, Fujii S, Sawa T, Inoue H, Matsunaga T, Khan S, Akashi S, Hashimoto R et al.: Endogenous nitrated  nucleotide is a key mediator of autophagy and innate defense against bacteria. Mol Cell 2013, 52:794-804. This paper identifies S-guanylation by 8-nitro-cGMP as a new mechanism for the specific targeting of intracellular bacteria, in association with ubiquitination. 20. Dupont N, Lacas-Gervais S, Bertout J, Paz I, Freche B, Van Nhieu GT, van der Goot FG, Sansonetti PJ, Lafont F: Shigella phagocytic vacuolar membrane remnants participate in the cellular response to pathogen invasion and are regulated by autophagy. Cell Host Microbe 2009, 6:137-149. 21. Shahnazari S, Yen WL, Birmingham CL, Shiu J, Namolovan A, Zheng YT, Nakayama K, Klionsky DJ, Brumell JH: A diacylglycerol-dependent signaling pathway contributes to regulation of antibacterial autophagy. Cell Host Microbe 2010, 8:137-146. 22. Tattoli I, Sorbara MT, Yang C, Tooze SA, Philpott DJ, Girardin SE: Listeria phospholipases subvert host autophagic defenses by stalling pre-autophagosomal structures. EMBO J 2013, 32:3066-3078. 23. Tattoli I, Sorbara MT, Vuckovic D, Ling A, Soares F, Carneiro LA, Yang C, Emili A, Philpott DJ, Girardin SE: Amino acid starvation induced by invasive bacterial pathogens triggers an innate host defense program. Cell Host Microbe 2012, 11:563-575. 24. Tattoli I, Philpott DJ, Girardin SE: The bacterial and cellular determinants controlling the recruitment of mTOR to the Salmonella-containing vacuole. Biol Open 2012, 1:1215-1225. 25. von Hoven G, Kloft N, Neukirch C, Ebinger S, Bobkiewicz W, Weis S, Boller K, Janda KD, Husmann M: Modulation of translation and induction of autophagy by bacterial exoproducts. Med Microbiol Immunol 2012, 201:409-418. 26. Kloft N, Neukirch C, Bobkiewicz W, Veerachato G, Busch T, von Hoven G, Boller K, Husmann M: Pro-autophagic signal induction by bacterial pore-forming toxins. Med Microbiol Immunol 2010, 199:299-309. 27. Owen KA, Meyer CB, Bouton AH, Casanova JE: Activation of focal adhesion kinase by Salmonella suppresses autophagy via an Akt/mTOR signaling pathway and promotes bacterial survival in macrophages. PLoS Pathog 2014, 10:e1004159. 28. Yang CS, Kim JJ, Lee HM, Jin HS, Lee SH, Park JH, Kim SJ, Kim JM, Han YM, Lee MS et al.: The AMPK–PPARGC1A pathway is required for antimicrobial host defense through activation of autophagy. Autophagy 2014, 10:785-802. 29. Yano T, Mita S, Ohmori H, Oshima Y, Fujimoto Y, Ueda R, Takada H, Goldman WE, Fukase K, Silverman N et al.: Autophagic control of Listeria through intracellular innate immune recognition in Drosophila. Nat Immunol 2008, 9:908-916. www.sciencedirect.com

Emerging themes in bacterial autophagy Sorbara and Girardin 169

30. Mostowy S, Boucontet L, Mazon Moya MJ, Sirianni A, Boudinot P,  Hollinshead M, Cossart P, Herbomel P, Levraud JP, ColucciGuyon E: The zebrafish as a new model for the in vivo study of Shigella flexneri interaction with phagocytes and bacterial autophagy. PLoS Pathog 2013, 9:e1003588. Demonstrates the potential of using zebrafish as a new model to study Shigella infection and the involvement of cellular processes such as phagocytosis or autophagy. 31. Jia K, Thomas C, Akbar M, Sun Q, Adams-Huet B, Gilpin C, Levine B: Autophagy genes protect against Salmonella typhimurium infection and mediate insulin signalingregulated pathogen resistance. Proc Natl Acad Sci U S A 2009, 106:14564-14569. 32. Benjamin JL, Sumpter R Jr, Levine B, Hooper LV: Intestinal  epithelial autophagy is essential for host defense against invasive bacteria. Cell Host Microbe 2013, 13:723-734. This study provides the first in vivo evidence to suggest that autophagy in enterocytes is critical for the control of intestinal bacterial pathogens. 33. Conway KL, Kuballa P, Song JH, Patel KK, Castoreno AB,  Yilmaz OH, Jijon HB, Zhang M, Aldrich LN, Villablanca EJ et al.: Atg16l1 is required for autophagy in intestinal epithelial cells and protection of mice from Salmonella infection. Gastroenterology 2013, 145:1347-1357. Demonstrates that loss of ATG16L1 in intestinal epithelial cells results in increased Salmonella replication and pathology. 34. Zhao Z, Fux B, Goodwin M, Dunay IR, Strong D, Miller BC, Cadwell K, Delgado MA, Ponpuak M, Green KG et al.: Autophagosome-independent essential function for the autophagy protein Atg5 in cellular immunity to intracellular pathogens. Cell Host Microbe 2008, 4:458-469. 35. Sorbara MT, Ellison LK, Ramjeet M, Travassos LH, Jones NL, Girardin SE, Philpott DJ: The protein ATG16L1 suppresses inflammatory cytokines induced by the intracellular sensors Nod1 and Nod2 in an autophagy-independent manner. Immunity 2013, 39:858-873.

44. Ogawa M, Yoshikawa Y, Kobayashi T, Mimuro H, Fukumatsu M, Kiga K, Piao Z, Ashida H, Yoshida M, Kakuta S et al.: A Tecpr1dependent selective autophagy pathway targets bacterial pathogens. Cell Host Microbe 2011, 9:376-389. 45. Nakagawa I, Amano A, Mizushima N, Yamamoto A, Yamaguchi H, Kamimoto T, Nara A, Funao J, Nakata M, Tsuda K et al.: Autophagy defends cells against invading Group A Streptococcus. Science 2004, 306:1037-1040. 46. Kuang E, Okumura CY, Sheffy-Levin S, Varsano T, Shu VC, Qi J, Niesman IR, Yang HJ, Lopez-Otin C, Yang WY et al.: Regulation of ATG4B stability by RNF5 limits basal levels of autophagy and influences susceptibility to bacterial infection. PLoS Genet 2012, 8:e1003007. 47. Nozawa T, Aikawa C, Goda A, Maruyama F, Hamada S, Nakagawa I: The small GTPases Rab9A and Rab23 function at distinct steps in autophagy during Group A Streptococcus infection. Cell Microbiol 2012, 14:1149-1165. 48. Yamaguchi H, Nakagawa I, Yamamoto A, Amano A, Noda T, Yoshimori T: An initial step of GAS-containing autophagosome-like vacuoles formation requires Rab7. PLoS Pathog 2009, 5:e1000670. 49. Joubert PE, Meiffren G, Gregoire IP, Pontini G, Richetta C, Flacher M, Azocar O, Vidalain PO, Vidal M, Lotteau V et al.: Autophagy induction by the pathogen receptor CD46. Cell Host Microbe 2009, 6:354-366. 50. Furuta N, Fujita N, Noda T, Yoshimori T, Amano A: Combinational soluble N-ethylmaleimide-sensitive factor attachment protein receptor proteins VAMP8 and Vti1b mediate fusion of antimicrobial and canonical autophagosomes with lysosomes. Mol Biol Cell 2010, 21:1001-1010. 51. Singh SB, Davis AS, Taylor GA, Deretic V: Human IRGM induces autophagy to eliminate intracellular mycobacteria. Science 2006, 313:1438-1441.

36. Gutierrez MG, Master SS, Singh SB, Taylor GA, Colombo MI, Deretic V: Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell 2004, 119:753-766.

52. Yang CS, Lee JS, Rodgers M, Min CK, Lee JY, Kim HJ, Lee KH, Kim CJ, Oh B, Zandi E et al.: Autophagy protein Rubicon mediates phagocytic NADPH oxidase activation in response to microbial infection or TLR stimulation. Cell Host Microbe 2012, 11:264-276.

37. Romagnoli A, Etna MP, Giacomini E, Pardini M, Remoli ME, Corazzari M, Falasca L, Goletti D, Gafa V, Simeone R et al.: ESX-1 dependent impairment of autophagic flux by Mycobacterium tuberculosis in human dendritic cells. Autophagy 2012, 8: 1357-1370.

53. van der Vaart M, Korbee CJ, Lamers GE, Tengeler AC, Hosseini R, Haks MC, Ottenhoff TH, Spaink HP, Meijer AH: The DNA damageregulated autophagy modulator DRAM1 links mycobacterial recognition via TLP-MYD88 to authophagic defense. Cell Host Microbe 2014, 15:753-767.

38. Ogawa M, Yoshimori T, Suzuki T, Sagara H, Mizushima N, Sasakawa C: Escape of intracellular Shigella from autophagy. Science 2005, 307:727-731.

54. von Muhlinen N, Akutsu M, Ravenhill BJ, Foeglein A, Bloor S, Rutherford TJ, Freund SM, Komander D, Randow F: LC3C, bound selectively by a noncanonical LIR motif in NDP52, is required for antibacterial autophagy. Mol Cell 2012, 48:329-342.

39. Yoshikawa Y, Ogawa M, Hain T, Yoshida M, Fukumatsu M, Kim M, Mimuro H, Nakagawa I, Yanagawa T, Ishii T et al.: Listeria monocytogenes ActA-mediated escape from autophagic recognition. Nat Cell Biol 2009, 11:1233-1240. 40. Dortet L, Mostowy S, Samba-Louaka A, Gouin E, Nahori MA, Wiemer EA, Dussurget O, Cossart P: Recruitment of the major vault protein by InlK: a Listeria monocytogenes strategy to avoid autophagy. PLoS Pathog 2011, 7:e1002168. 41. Barnett TC, Liebl D, Seymour LM, Gillen CM, Lim JY, Larock CN,  Davies MR, Schulz BL, Nizet V, Teasdale RD et al.: The globally disseminated M1T1 clone of Group A Streptococcus evades autophagy for intracellular replication. Cell Host Microbe 2013, 14:675-682. While it was previously believed that autophagy efficiently clears Group A Streptococcus, the authors of this study demonstrate that a serotype frequently associated with disease in humans expresses a protease that degrades autophagy proteins. 42. Kim PK, Hailey DW, Mullen RT, Lippincott-Schwartz J: Ubiquitin signals autophagic degradation of cytosolic proteins and peroxisomes. Proc Natl Acad Sci U S A 2008, 105:20567-20574. 43. Kirkin V, Lamark T, Sou YS, Bjorkoy G, Nunn JL, Bruun JA, Shvets E, McEwan DG, Clausen TH, Wild P et al.: A role for NBR1 in autophagosomal degradation of ubiquitinated substrates. Mol Cell 2009, 33:505-516. www.sciencedirect.com

55. Birmingham CL, Smith AC, Bakowski MA, Yoshimori T, Brumell JH: Autophagy controls Salmonella infection in response to damage to the Salmonella-containing vacuole. J Biol Chem 2006, 281:11374-11383. 56. Sun Q, Fan W, Chen K, Ding X, Chen S, Zhong Q: Identification of Barkor as a mammalian autophagy-specific factor for Beclin 1 and class III phosphatidylinositol 3-kinase. Proc Natl Acad Sci U S A 2008, 105:19211-19216. 57. Fujita N, Saitoh T, Kageyama S, Akira S, Noda T, Yoshimori T: Differential involvement of Atg16L1 in Crohn disease and canonical autophagy: analysis of the organization of the Atg16L1 complex in fibroblasts. J Biol Chem 2009, 284: 32602-32609. 58. Dooley HC, Razi M, Polson HE, Girardin SE, Wilson MI, Tooze SA: WIPI2 links LC3 conjugation with PI3P, autophagosome formation, and pathogen clearance by recruiting Atg12-516L1. Mol Cell 2014, 55:238-252. 59. Thurston TL, Ryzhakov G, Bloor S, von Muhlinen N, Randow F: The TBK1 adaptor and autophagy receptor NDP52 restricts the proliferation of ubiquitin-coated bacteria. Nat Immunol 2009, 10:1215-1221. 60. Huett A, Ng A, Cao Z, Kuballa P, Komatsu M, Daly MJ, Podolsky DK, Xavier RJ: A novel hybrid yeast–human network Current Opinion in Microbiology 2015, 23:163–170

170 Host-microbe interactions: bacteria

analysis reveals an essential role for FNBP1L in antibacterial autophagy. J Immunol 2009, 182:4917-4930. 61. Huang J, Canadien V, Lam GY, Steinberg BE, Dinauer MC, Magalhaes MA, Glogauer M, Grinstein S, Brumell JH: Activation of antibacterial autophagy by NADPH oxidases. Proc Natl Acad Sci U S A 2009, 106:6226-6231. 62. Yu HB, Croxen MA, Marchiando AM, Ferreira RB, Cadwell K, Foster LJ, Finlay BB: Autophagy facilitates Salmonella replication in HeLa cells. MBio 2014, 5 e00865–14. 63. Homer CR, Kabi A, Marina-Garcia N, Sreekumar A, Nesvizhskii AI, Nickerson KP, Chinnaiyan AM, Nunez G, McDonald C: A dual role for receptor-interacting protein kinase 2 (RIP2) kinase activity in nucleotide-binding oligomerization domain 2 (NOD2)dependent autophagy. J Biol Chem 2012, 287:25565-25576. 64. Homer CR, Richmond AL, Rebert NA, Achkar JP, McDonald C: ATG16L1 and NOD2 interact in an autophagy-dependent antibacterial pathway implicated in Crohn’s disease pathogenesis. Gastroenterology 2010, 139:1630-1641 e1631–1632. 65. Lassen KG, Kuballa P, Conway KL, Patel KK, Becker CE,  Peloquin JM, Villablanca EJ, Norman JM, Liu TC, Heath RJ et al.: Atg16L1 T300A variant decreases selective autophagy resulting in altered cytokine signaling and decreased antibacterial defense. Proc Natl Acad Sci U S A 2014, 111:7741-7746. Identifies a Caspase-3 cleavage site in Atg16L1, which is differentially used depending on the adjacent Crohn’s disease-associated polymorphism (T300A). Using a knock-in mouse model, effects on bacterial autophagy were identified. 66. Case ED, Chong A, Wehrly TD, Hansen B, Child R, Hwang S, Virgin HW, Celli J: The Francisella O-antigen mediates survival in the macrophage cytosol via autophagy avoidance. Cell Microbiol 2014, 16:862-877. 67. Scharl M, Wojtal KA, Becker HM, Fischbeck A, Frei P, Arikkat J, Pesch T, Kellermeier S, Boone DL, Weber A et al.: Protein tyrosine phosphatase nonreceptor type 2 regulates autophagosome formation in human intestinal cells. Inflamm Bowel Dis 2012, 18:1287-1302. 68. Py BF, Lipinski MM, Yuan J: Autophagy limits Listeria monocytogenes intracellular growth in the early phase of primary infection. Autophagy 2007, 3:117-125.

Current Opinion in Microbiology 2015, 23:163–170

69. Anand PK, Tait SW, Lamkanfi M, Amer AO, Nunez G, Pages G, Pouyssegur J, McGargill MA, Green DR, Kanneganti TD: TLR2 and RIP2 pathways mediate autophagy of Listeria monocytogenes via extracellular signal-regulated kinase (ERK) activation. J Biol Chem 2011, 286:42981-42991. 70. Birmingham CL, Canadien V, Gouin E, Troy EB, Yoshimori T, Cossart P, Higgins DE, Brumell JH: Listeria monocytogenes evades killing by autophagy during colonization of host cells. Autophagy 2007, 3:442-451. 71. Marchiando AM, Ramanan D, Ding Y, Gomez LE, Hubbard Lucey VM, Maurer K, Wang C, Ziel JW, van Rooijen N, Nunez G et al.: A deficiency in the autophagy gene Atg16L1 enhances resistance to enteric bacterial infection. Cell Host Microbe 2013, 14:216-224. Identification of a role for Atg16L1 in suppressing inflammation during infection with a non-invasive bacteria. 72. Murthy A, Li Y, Peng I, Reichelt M, Katakam AK, Noubade R,  Roose-Girma M, DeVoss J, Diehl L, Graham RR et al.: A Crohn’s disease variant in Atg16l1 enhances its degradation by caspase 3. Nature 2014, 506:456-462. Identifies a Caspase-3 cleavage site in Atg16L1, and increased susceptibility to Yersinia infection in mice expressing Atg16L1 risk variant. 73. Lapaquette P, Glasser AL, Huett A, Xavier RJ, DarfeuilleMichaud A: Crohn’s disease-associated adherent-invasive E. coli are selectively favoured by impaired autophagy to replicate intracellularly. Cell Microbiol 2010, 12:99-113. 74. Lapaquette P, Bringer MA, Darfeuille-Michaud A: Defects in autophagy favour adherent-invasive Escherichia coli persistence within macrophages leading to increased proinflammatory response. Cell Microbiol 2012, 14:791-807. 75. Nguyen HT, Dalmasso G, Muller S, Carriere J, Seibold F, Darfeuille-Michaud A: Crohn’s disease-associated adherent invasive Escherichia coli modulate levels of microRNAs in intestinal epithelial cells to reduce autophagy. Gastroenterology 2014, 146:508-519. 76. Wang C, Mendonsa GR, Symington JW, Zhang Q, Cadwell K, Virgin HW, Mysorekar IU: Atg16L1 deficiency confers protection from uropathogenic Escherichia coli infection in vivo. Proc Natl Acad Sci U S A 2012, 109:11008-11013.

www.sciencedirect.com

Emerging themes in bacterial autophagy.

The role of autophagy in the control of intracellular bacterial pathogens, also known as xenophagy, is well documented. Here, we highlight recent adva...
379KB Sizes 0 Downloads 14 Views