Microbes and Infection 16 (2014) 985e990 www.elsevier.com/locate/micinf

Recognition of Legionella pneumophila nucleic acids by innate immune receptors Larissa D. Cunha, Dario S. Zamboni* Department of Cell Biology, Ribeir~ao Preto Medical School, University of S~ao Paulo (FMRP/USP), Ribeir~ao Preto, SP 14049-900, Brazil Received 10 July 2014; accepted 18 August 2014 Available online 27 August 2014

Abstract Innate immune receptors evolved to sense conserved molecules that are present in microbes or are released during non-physiological conditions. Activation of these receptors is essential for early restriction of microbial infections and generation of adaptive immunity. Among the conserved molecules sensed by innate immune receptors are the nucleic acids, which are abundantly contained in all infectious organisms including virus, bacteria, fungi and parasites. In this review we focus in the innate immune proteins that function to sense nucleic acids from the intracellular bacterial pathogen Legionella pneumophila and the importance of these processes to the outcome of the infection. © 2014 Institut Pasteur. Published by Elsevier Masson SAS. All rights reserved.

Keywords: Legionella; Nucleic acid sensing; Innate immune receptors

1. Introduction Legionella pneumophila is the etiological agent of Legionnaire's disease, an acute pneumonia that affects mostly immunosuppressed patients, with an elevated mortality risk [1]. L. pneumophila is a facultative intracellular Gramnegative bacterium that parasite free-living protozoa of fresh water reservoirs, thus human contamination occurs by inhalation of aerosols containing with infected protozoan host [2]. In humans, L. pneumophila thrives in endosomal-like vacuoles (LCV, Legionella-containing vacuole), which are formed in alveolar macrophages and monocytes, eventually invading non-professional phagocytes such as epithelial cells [3,4]. In order to establish and maintain the LCV, the bacteria modulate host cell vesicular traffic and endosomal maturation pathway, inducing recruitment of vesicles derived from endoplasmic * Corresponding author. Department of Cell Biology, Ribeir~ao Preto Medical School, University of S~ao Paulo (FMRP/USP), Av. Bandeirantes 3900, Ribeir~ao Preto, SP 14049-900, Brazil. Tel.: þ55 (16) 3602 3265; fax: þ55 (16) 3633 1786. E-mail address: [email protected] (D.S. Zamboni).

reticulum while avoiding LCV fusion with lysosomes [5]. Legionella express a unique type IVB secretion system denominated Dot/Icm, through which more than 300 bacterial effector proteins are secreted into host cytosol to subvert host functions Contributing to LCV maintenance [6,7]. Consequently, L. pneumophila mutants that express defective Dot/ Icm machinery fail to establish a functional LCV and are rapidly controlled by the host cells [8,9]. Murine models of infection with L. pneumophila have effectively contributed to unravel the mechanisms that lead to infection control by a competent host. Effective innate immune responses are essential to control L. pneumophila infection [10]. Susceptibility to infection by L. pneumophila in A/J mice has been mapped to a gene called Naip5 or Birc1e, encoded within the chromosome 13 locus Lgn1 [11e14]. Birc1e (NAIP5) is a cytosolic receptor that belongs to the Nod-like family. Recognition of bacterial flagellin that escapes into macrophages cytosol through the Dot/Icm system interacts with NAIP5, forming a complex with CARDcontaining NLRC4 (also a member of Nod-like receptor family), and triggering caspase-1 recruitment and activation in a molecular platform generally denominated inflammasome

http://dx.doi.org/10.1016/j.micinf.2014.08.008 1286-4579/© 2014 Institut Pasteur. Published by Elsevier Masson SAS. All rights reserved.

986

L.D. Cunha, D.S. Zamboni / Microbes and Infection 16 (2014) 985e990

[15e18]. The NAIP5/NLRC4 pathway of inflammasome activation in response to flagellin is considered the main mechanism involved with generation of responses that culminate in infection control [19e21]. In accordance, restrictive C57BL/6 mice are susceptible to early pulmonary bacterial multiplication in the absence of Naip5, Nlrc4 or caspase-1, as well as by infection with flagellin deficient L. pneumophila ( flaA) [15,22]. Neutrophil recruitment to the lungs of infected mice is required for successful eradication of L. pneumophila. Deficiency on Myd88, an adaptor of Toll-like (TLR) receptors, IL1 and IL-18 receptors, is well known to impact neutrophil recruitment and infection control and different players have already been shown to collaborate to this process [23e25]. In addition, the Nod-like receptor Nod2 and the adaptor molecule Rip2, which are known to trigger NF-kB signaling pathway, also contribute to recruitment of neutrophils to the lungs of infected mice [26]. Type I interferon (IFN) production, induced by pathways that trigger activation of transcription factor IRF3, is another important player in the immune response against L. pneumophila. Mice lacking type I interferon (IFN) signaling are defective in bacterial clearance by mechanisms not completely understood [27e29]. Finally, other components of the innate immune response, such as the Nod-like receptor NLRP3, also participate in the recognition of L. pneumophila [30,31]. In this review, we discuss the role of nucleic acid recognition in immune responses to L. pneumophila. So far, different signaling pathways dependent on nucleic acid sensing have already been shown to recognize L. pneumophila and contribute to infection control. Among them are the recognition of bacterial DNA by TLR9 and induction of type I interferon mediated by IRF3 activation. Moreover, modulation by L. pneumophila Dot/Icm effectors of host cell responses driven by acid nucleic sensing is also discussed. 2. TLRs activation and DNA-mediated TLR9 activation in response to L. pneumophila As previously mentioned, innate immune responses mediated by the adaptor protein Myd88 are pivotal to clearance of L. pneumophila [23e25]. Recognition of L. pneumophila peptidoglycans and lipopeptides by TLR2 contributes to bacterial clearance in the lungs of infected mice [24,25,32,33]. Moreover, alveolar macrophages deficient in TLR5 produce less TNF-a as compared to cells from wild-type animals [34]. Although Tlr5/  mice restrict L. pneumophila infection normally, they show lung pathology consistent with development of pneumonia in the late stages of infection [34]. IL-18 signaling, mediated by Myd88, was also shown to be important for IFN-g induction by NK cells [35]. In agreement, IFN-g deficiency moderately impairs L. pneumophila clearance [23,35]. Recently, it was shown that the IL-1a production in the lungs of infected mice was dependent on the expression of a functional Dot/Icm dependent system that mediates neutrophil recruitment to the infection site [36]. Consequently, IL-1a deficient mice failed to control infection by virulent L. pneumophila [36].

The initial studies supporting a role of TLR9 for L. pneumophila recognition demonstrated that infection with L. pneumophila or stimulation with heat-killed bacteria induced IL-12p40 secretion in macrophages [37]. Of note, the cytokine production was reduced by treatment of cells with chloroquine, an inhibitor of endosome acidification, which is essential for TLR9 activation [37]. Further studies performed with macrophages derived from Tlr2//Tlr9/ indicated that these cells produced similar levels of IL-12 compared Tlr2/ littermates controls, suggesting that in macrophages TLR9 is not essential for cytokine production in response to L. pneumophila infection [35]. Of note, these studies were performed using cells encoding the Naip5 mutant allele derived from the A/J mice background. Therefore, the TLR9-mediated restriction of L. pneumophila replication is not affected by Naip5mediated flagellin recognition. In the case of dendritic cells (DC), inhibition of Tlr9 activation by either chloroquine or by an inhibitory ligand of TLR9 (ODN2088) reduced IL-12p40 secretion in cells derived from both susceptible A/J and restrictive BALB/c mice (that express a functional Naip5) [37]. Importantly, Tlr9/ mice in the restrictive BALB/c genetic background are more susceptible to pulmonary infection with L. pneumophila, with increased mortality and bacterial burden in comparison to wild-type mice in an intratracheal route of infection [38]. Susceptibility of mice deficient for Tlr9 could be associated with reduced levels of myeloid DC and CD4þ lymphocytes in the lung of infected mice [38]. This impairment observed in Tlr9/ mice was associated with isolation from infected mice of alveolar macrophages displaying alternative polarization, with reduced phagocytosis of pulmonary bacteria and expression of nitric oxide, which is important to bacterial clearance. As Tlr9/ macrophages infected in vitro does not display impaired bacterial phagocytosis, it is suggestive that the cytokine milieu in the lungs is important for macrophage response to Legionella. Indeed, Tlr9/ mice presented reduced production of chemokines and type I cytokines, such as MCP-1, TNF-a, IL-12 and IFN-g. IL-12, for instance, stimulates IFN-g production by NK cells, which is important to host resistance to L. pneumophila infection. Importantly, Tlr9-dependent chemokine and cytokine profile in the lungs and bacterial clearance could be restored by adoptive transfer of wild-type dendritic cells directly in the lungs of Tlr9/ mice. These data unequivocally supported a role for TLR9-mediated DNA sensing for host immunity against L. pneumophila. Therefore, recognition of L. pneumophila by TLR9 is important for bacterial sensing by dendritic cells, collaborating to the development of a chemokine and cytokine microenvironment that sustain effective activation of macrophages in the lungs. Finally, is important to emphasize that so far, mice deficient to each component that signal through Myd88, as well as doubleknockouts such as Tlr2//Tlr5/ and Tlr2//Tlr9/, fail to recapitulate the severe defect in L. pneumophila clearance present by Myd88/ mice [35]. Therefore, despite the important role of TLR2, -5 and -9 for innate immune recognition of L. pneumophila, the severe deficiency in MyD88 suggest the

L.D. Cunha, D.S. Zamboni / Microbes and Infection 16 (2014) 985e990

existence of a redundant function of TLRs and/or additional receptors operate in the MyD88-mediated innate immunity to L. pneumophila. 3. Induction of type I IFN in response to L. pneumophila Induction of antimicrobial genes through type I interferon (e.g., IFN-a and IFN-b) signaling is a classic antiviral response. During viral infections, type I IFN transcription is mediated by activation of transcription factors IRF3 and IRF7, upon sensing of viral dsRNA by cytosolic helicases retinoic acid-inducible gene I (RIG-I) or melanoma differentiationassociated gene 5 (MDA5), both requiring the common adaptor protein mitochondrial antiviral signaling protein (MAVS/IPS-1) [39]. However, it is now well accepted that type I interferon is also relevant in immune responses to intracellular bacterial pathogens such as Listeria monocytogenes, Mycobacterium tuberculosis, Franciella tularensis and Brucella abortus [40e43]. Production of IFN-b in response to bacteria has been suggested to occur by recognition of cytosolic dsDNA, supported by the fact that purified bacterial DNA can induce type I IFN production by BMM [44]. Recently, different sensors leading to IRF3 activation and type I IFN expression by recognition of cytosolic DNA have emerged, such as RNA polymerase III (PolIII) [45,46], DNAdependent activator of IFN- regulatory factors (DAI)/Z-DNAbinding protein (ZBP)1 [47], STING [48] and cGAS [49]. The Dot/Icm machinery diverged from ancestral bacterial DNA conjugation system, while retaining the capacity to translocate plasmidial DNA [50]. Therefore, recognition of cytosolic bacterial DNA during the infection with L. pneumophila is likely to occur. In fact, infection of bone marrowderived macrophages and epithelial-like A549 human cells revealed that the bacteria trigger IRF3-dependent IFN-b expression dependently on a functional Dot/Icm secretion system but independently of bacterial flagellin [44,51]. In macrophages, IFN induction is independent of TLR receptors or de novo protein synthesis by the host cell [44]. Accordingly, epithelial cells silenced for IRF3 and macrophages Irf3/ are more susceptible to infection with L. pneumophila [27,51]. These phenotypes could be reverted by treatment with recombinant IFN-b, thus suggesting that type I IFNs contributes to the restriction of L. pneumophila replication in macrophages. Interestingly, induction of Irgm1, an immunity-related GTPase induced by type I IFN response, is partially required for resistance to L. pneumophila in macrophages [27]. Chiu et al. (2009) demonstrated that AT-rich cytosolic DNA triggers IFN production through a mechanism involving Polymerase III (Pol III)-dependent synthesis of poly (A-U) and recognition of newly synthetized RNA by RIG-I. Pol III inhibition was found to significantly reduce IFN-b production in response to L. pneumophila, also impairing infection control. Macrophages treated with Pol III inhibitor ML60218 were susceptible to bacterial infection, and resistance was restored by treatment with IFN-b. Results by other authors argued that, although MDA5 and RIG1 partially contribute to induction of IFN-b in response to L. pneumophila in BMM, this response

987

occurs by recognition of bacterial RNA, not DNA [52]. This latter work also argued that inhibition of Pol III activity by ML60218 does not impair resistance to L. pneumophila. Although the adaptor IPS-1 is required downstream for IFN-b, both in BMM and in vivo, Ips-1/ mice do not fail to control bacterial multiplication [45,52]. Interestingly, in human epithelial cells, the use of RNAi was used to demonstrate that MDA5, RIG1 and other CARD-containing receptors (such as ASC and Nod2) are not necessary for IFN induction, suggesting that in humans other receptors may contribute to IFN production in response to bacteria [51]. Finally, differently than L. pneumophila, mice deficient in IFN-b signaling are more resistant to infections with Listeria monocytogenes and with Brucella abortus [53,54]. This contrasting role of type I IFN in host resistance suggests that type I IFN might be a pathway subverted by certain bacterial pathogens thus influencing infection outcome in a pathogen specific fashion. Most recently, induction of a unique eukaryotic cyclic dinucleotide, denominated cyclic guanosine monophosphateadenosine monophosphate (cyclic GMP-AMP, or cGAMP), has been recognized as a major pathway of type IFN expression in response to cytosolic DNA in innate immune cells [55]. Cyclic dinucleotides are conserved secondary messengers in bacterial signaling cascades, and had been previously suggested as immunomodulatory molecules in bacterial infection of murine cells [56]. Nonetheless cGAMP is produced in response to DNA recognition by a cGAMP synthase (cGAS), belonging to the nucleotidyltransferase family [49]. Of note, cGAMP intermediates activation of STING, also important for IRF3-dependent induction of type I IFN [55,57,58]. The axis cGAS/cGAMP/STING contributes to IFN-b production by human macrophages in response to Listeria monocytogenes [59]. Interestingly, silencing of STING by RNAi reduces IFNb in BMM infected with L. pneumophila [27]. In this context, it will be important to verify whether cGAS and cGAMP synthesis are also key players in the generation of type I IFN in response to L. pneumophila. 4. Inflammasome activation in response to recognition of L. pneumophila nucleic acid Induction of inflammasome and caspase-1 activation by the Nod-like receptors NAIP5/NLRC4 is pivotal for recognition of L. pneumophila by murine macrophages and infection control [18e21]. However, additional cytosolic sensors that trigger inflammasome activation also participate in a flagellinindependent pathway for caspase-1 activation in response to L. pneumophila. Of note, the central adaptor protein of inflammasome receptors ASC, is required for cleavage of caspase-1 dependent and independent of expression of flagellin by L. pneumophila [60]. In addition, non-canonical inflammasome activation mediated by caspase-11, most likely in response to cytosolic L. pneumophila LPS, triggers flagellin-independent pore formation in the macrophages membranes and regulates cleavage of caspase-1 by NLRP3 [30]. Similarly to ASC, the NLRP3 receptor is essential to caspase-1 cleavage in response to flaA

988

L.D. Cunha, D.S. Zamboni / Microbes and Infection 16 (2014) 985e990

L. pneumophila, as caspase-1 cleavage and IL-1b is abrogated in Nlrp3/ macrophages [30]. In this context, it is important to emphasize that L. pneumophila RNA has been shown to activate NLRP3-dependent inflammasome activation [31]. The role of NLRP3 in the recognition of RNA:DNA hybrid complexes has been recently demonstrated, corroborating the importance of this sensor for recognition and signaling against the presence of foreign nucleic acid inside the cell [61]. In this context, it was reported that NLRP3 participate in the recognition of prokaryotic mRNA thus favoring the recognition of microbial viability, a feature that would account for differential discrimination of live versus dead microbes. It is still unclear if this pathway operates for recognition of L. pneumophila. The observations that live L. pneumophila that is deficient in functional Dot/Icm fail to trigger NLRP3 activation do not favor this hypothesis. An important DNA-sensing inflammasome was revealed in 2009, this complex is involved in recognition of cytosolic DNA and is composed by the cytosolic helicase AIM2 and uses the adaptor protein ASC to trigger caspase-1 activation, the so-called AIM2 inflammasome [62e65]. AIM2 directly interacts with DNA, and have already been shown to participate in macrophage response to DNA virus, such as vaccinia virus and mouse cytomegalovirus, and bacterial pathogens, such as Francisella tularensis and L. monocytogenes [66e70]. It is unclear if AIM2 participate in L. pneumophila recognition. Certainly the L. pneumophila DNA trigger AIM2 activation, but it has been reported that a Dot/Icm effector SdhA, which was known to inhibit host cell death and stabilize the vacuole containing bacteria, prevent the access of bacterial DNA to the cytosol, thus inhibiting the AIM2 recognition of L. pneumophila by human macrophage-like cells and murine macrophages [71e73]. Thus, determination of the role of AIM2 in bacterial recognition and host resistance during L. pneumophila infections may be a subject of additional investigation. 5. Conclusion The resistance to infectious organisms, including L. pneumophila, relies on the innate immune recognition of invading organisms. Understanding of this process is essential to unravel the mechanisms underlying host resistance. Although the recognition of L. pneumophila flagellin by cytosolic receptors NAIP5 and NLRC4 represent a pivotal mechanism for host resistance, recent studies identified additional mechanisms that operate independent on the axis flagellin/NAIP5/NLRC4. In this context, the recognition of L. pneumophila DNA and RNA may effectively contribute to early innate immune detection of the pathogen. Different innate immune receptors have already been shown to be involved in the recognition of the L. pneumophila nucleic acids, including TLR9, Pol III, NLRP3 and AIM2 (Fig. 1). These receptors trigger different signaling pathways, such as NF-kB pathway, signaling through type I interferon and cleavage of caspase-1, that result in expression, activation and secretion of diverse chemokines and cytokines. Activation of these pathways may favor the

Fig. 1. Overview of innate immune responses pathways triggered by sensing of L. pneumophila DNA. LCV indicate the Legionella-containing vacuole. DNA releases from death (dotted square in blue) bacteria trigger TLR9 activation in the LCV membrane leading to NF-kB activation. Nucleic acid from live L. pneumophila (square in blue) translocate into the host cell cytoplasm in a process dependent of the bacterial Dot/Icm. This process enables activation of RIG1/MDA5, STING and AIM2 pathways, which culminated in caspase-1 activation (via AIM2 and ASC) and IFN-b production. The Dot/Icm effector protein SdhA was shown to stabilize the vacuole containing bacteria preventing the access of bacterial DNA to the cytosol, thus reducing AIM2 activation in response to L. pneumophila infection.

boosting an effective innate immune response for host resistance during infections with Legionella. We envisage that in the near future additional pathways for DNA and RNA recognition will be identified as relevant for host response during L. pneumophila infections. Moreover, further investigation using L. pneumophila as a model of infection may facilitate identification of novel nucleic acid recognition pathways that operate in response to infections with pathogenic microbes. Conflict of interest There is no conflict of interest. Acknowledgments We apologize to our colleagues whose papers we were unable to cite due to space limitations. Work in our laboratory is supported by grants from the Fundaç~ao de Amparo a Pesquisa do Estado de S~ao Paulo (FAPESP, Grants 2012/09363-6, 2013/08216-2, and 2014/04684-4) and Instituto Nacional de Ci^encia e Tecnologia em Vacinas (INCTV/CNPq). D.S.Z. is a research fellow from CNPq. References [1] Phin N, Parry-Ford F, Harrison T, Stagg HR, Zhang N, Kumar K, et al. Epidemiology and clinical management of Legionnaires' disease. Lancet Infect Dis 2014;14:70713e3.

L.D. Cunha, D.S. Zamboni / Microbes and Infection 16 (2014) 985e990 [2] Cateau E, Delafont V, Hechard Y, Rodier MH. Free-living amoebae: what part do they play in healthcare-associated infections? J Hosp Infect 2014;87:131e40. [3] Horwitz MA, Silverstein SC. Legionnaires' disease bacterium (Legionella pneumophila) multiples intracellularly in human monocytes. J Clin Invest 1980;66:441e50. [4] Nash TW, Libby DM, Horwitz MA. Interaction between the legionnaires' disease bacterium (Legionella pneumophila) and human alveolar macrophages. Influence of antibody, lymphokines, and hydrocortisone. J Clin Invest 1984;74:771e82. [5] Escoll P, Rolando M, Gomez-Valero L, Buchrieser C. From amoeba to macrophages: exploring the molecular mechanisms of Legionella pneumophila infection in both hosts. Curr Top Microbiol Immunol 2013;376:1e34. [6] Hubber A, Roy CR. Modulation of host cell function by Legionella pneumophila type IV effectors. Annu Rev Cell Dev Biol 2010;26:261e83. [7] Ge J, Shao F. Manipulation of host vesicular trafficking and innate immune defence by Legionella Dot/Icm effectors. Cell Microbiol 2011;13:1870e80. [8] Marra A, Blander SJ, Horwitz MA, Shuman HA. Identification of a Legionella pneumophila locus required for intracellular multiplication in human macrophages. Proc Natl Acad Sci U S A 1992;89:9607e11. [9] Berger KH, Isberg RR. Two distinct defects in intracellular growth complemented by a single genetic locus in Legionella pneumophila. Mol Microbiol 1993;7:7e19. [10] Massis LM, Zamboni DS. Innate immunity to legionella pneumophila. Front Microbiol 2011;2:109. [11] Dietrich WF, Damron DM, Isberg RR, Lander ES, Swanson MS. Lgn1, a gene that determines susceptibility to Legionella pneumophila, maps to mouse chromosome 13. Genomics 1995;26:443e50. [12] Beckers MC, Yoshida S, Morgan K, Skamene E, Gros P. Natural resistance to infection with Legionella pneumophila: chromosomal localization of the Lgn1 susceptibility gene. Mammalian Genome: Off J Int Mammalian Genome Soc 1995;6:540e5. [13] Diez E, Lee SH, Gauthier S, Yaraghi Z, Tremblay M, Vidal S, et al. Birc1e is the gene within the Lgn1 locus associated with resistance to Legionella pneumophila. Nat Genet 2003;33:55e60. [14] Wright EK, Goodart SA, Growney JD, Hadinoto V, Endrizzi MG, Long EM, et al. Naip5 affects host susceptibility to the intracellular pathogen Legionella pneumophila. Curr Biol: CB 2003;13:27e36. [15] Lightfield KL, Persson J, Brubaker SW, Witte CE, von Moltke J, Dunipace EA, et al. Critical function for Naip5 in inflammasome activation by a conserved carboxy-terminal domain of flagellin. Nat Immunol 2008;9:1171e8. [16] Kofoed EM, Vance RE. Innate immune recognition of bacterial ligands by NAIPs determines inflammasome specificity. Nature 2011;477:592e5. [17] Zhao Y, Yang J, Shi J, Gong YN, Lu Q, Xu H, et al. The NLRC4 inflammasome receptors for bacterial flagellin and type III secretion apparatus. Nature 2011;477:596e600. [18] Zamboni DS, Kobayashi KS, Kohlsdorf T, Ogura Y, Long EM, Vance RE, et al. The Birc1e cytosolic pattern-recognition receptor contributes to the detection and control of Legionella pneumophila infection. Nat Immunol 2006;7:318e25. [19] Ren T, Zamboni DS, Roy CR, Dietrich WF, Vance RE. Flagellin-deficient Legionella mutants evade caspase-1- and Naip5-mediated macrophage immunity. PLoS Pathog 2006;2:e18. [20] Molofsky AB, Byrne BG, Whitfield NN, Madigan CA, Fuse ET, Tateda K, et al. Cytosolic recognition of flagellin by mouse macrophages restricts Legionella pneumophila infection. J Exp Med 2006;203:1093e104. [21] Amer A, Franchi L, Kanneganti TD, Body-Malapel M, Ozoren N, Brady G, et al. Regulation of Legionella phagosome maturation and infection through flagellin and host Ipaf. J Biol Chem 2006;281:35217e23. [22] Pereira MS, Morgantetti GF, Massis LM, Horta CV, Hori JI, Zamboni DS. Activation of NLRC4 by flagellated bacteria triggers

[23]

[24]

[25]

[26]

[27]

[28]

[29]

[30]

[31]

[32]

[33]

[34]

[35]

[36]

[37]

[38]

[39]

[40]

989

caspase-1-dependent and -independent responses to restrict Legionella pneumophila replication in macrophages and in vivo. J Immunol 2011;187:6447e55. Sporri R, Joller N, Albers U, Hilbi H, Oxenius A. MyD88-dependent IFN-gamma production by NK cells is key for control of Legionella pneumophila infection. J Immunol 2006;176:6162e71. Archer KA, Roy CR. MyD88-dependent responses involving toll-like receptor 2 are important for protection and clearance of Legionella pneumophila in a mouse model of Legionnaires' disease. Infect Immun 2006;74:3325e33. Hawn TR, Smith KD, Aderem A, Skerrett SJ. Myeloid differentiation primary response gene (88)- and toll-like receptor 2-deficient mice are susceptible to infection with aerosolized Legionella pneumophila. J Infect Dis 2006;193:1693e702. Frutuoso MS, Hori JI, Pereira MS, Junior DS, Sonego F, Kobayashi KS, et al. The pattern recognition receptors Nod1 and Nod2 account for neutrophil recruitment to the lungs of mice infected with Legionella pneumophila. Microbes infection/Institut Pasteur 2010;12:819e27. Lippmann J, Muller HC, Naujoks J, Tabeling C, Shin S, Witzenrath M, et al. Dissection of a type I interferon pathway in controlling bacterial intracellular infection in mice. Cell Microbiol 2011;13:1668e82. Schiavoni G, Mauri C, Carlei D, Belardelli F, Pastoris MC, Proietti E. Type I IFN protects permissive macrophages from Legionella pneumophila infection through an IFN-gamma-independent pathway. J Immunol 2004;173:1266e75. Coers J, Vance RE, Fontana MF, Dietrich WF. Restriction of Legionella pneumophila growth in macrophages requires the concerted action of cytokine and Naip5/Ipaf signalling pathways. Cell Microbiol 2007;9:2344e57. Case CL, Kohler LJ, Lima JB, Strowig T, de Zoete MR, Flavell RA, et al. Caspase-11 stimulates rapid flagellin-independent pyroptosis in response to Legionella pneumophila. Proc Natl Acad Sci U S A 2013;110:1851e6. Kanneganti TD, Ozoren N, Body-Malapel M, Amer A, Park JH, Franchi L, et al. Bacterial RNA and small antiviral compounds activate caspase-1 through cryopyrin/Nalp3. Nature 2006;440:233e6. Girard R, Pedron T, Uematsu S, Balloy V, Chignard M, Akira S, et al. Lipopolysaccharides from Legionella and Rhizobium stimulate mouse bone marrow granulocytes via Toll-like receptor 2. J Cell Sci 2003;116:293e302. Akamine M, Higa F, Arakaki N, Kawakami K, Takeda K, Akira S, et al. Differential roles of Toll-like receptors 2 and 4 in in vitro responses of macrophages to Legionella pneumophila. Infect Immun 2005;73:352e61. Hawn TR, Berrington WR, Smith IA, Uematsu S, Akira S, Aderem A, et al. Altered inflammatory responses in TLR5-deficient mice infected with Legionella pneumophila. J Immunol 2007;179:6981e7. Archer KA, Alexopoulou L, Flavell RA, Roy CR. Multiple MyD88dependent responses contribute to pulmonary clearance of Legionella pneumophila. Cell Microbiol 2009;11:21e36. Barry KC, Fontana MF, Portman JL, Dugan AS, Vance RE. IL-1alpha signaling initiates the inflammatory response to virulent Legionella pneumophila in vivo. J Immunol 2013;190:6329e39. Newton CA, Perkins I, Widen RH, Friedman H, Klein TW. Role of Tolllike receptor 9 in Legionella pneumophila-induced interleukin-12 p40 production in bone marrow-derived dendritic cells and macrophages from permissive and nonpermissive mice. Infect Immun 2007;75:146e51. Bhan U, Trujillo G, Lyn-Kew K, Newstead MW, Zeng X, Hogaboam CM, et al. Toll-like receptor 9 regulates the lung macrophage phenotype and host immunity in murine pneumonia caused by Legionella pneumophila. Infect Immun 2008;76:2895e904. Yoneyama M, Kikuchi M, Natsukawa T, Shinobu N, Imaizumi T, Miyagishi M, et al. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat Immunol 2004;5:730e7. Charrel-Dennis M, Latz E, Halmen KA, Trieu-Cuot P, Fitzgerald KA, Kasper DL, et al. TLR-independent type I interferon induction in response to an extracellular bacterial pathogen via intracellular recognition of its DNA. Cell Host Microbe 2008;4:543e54.

990

L.D. Cunha, D.S. Zamboni / Microbes and Infection 16 (2014) 985e990

[41] Henry T, Brotcke A, Weiss DS, Thompson LJ, Monack DM. Type I interferon signaling is required for activation of the inflammasome during Francisella infection. J Exp Med 2007;204:987e94. [42] Roux CM, Rolan HG, Santos RL, Beremand PD, Thomas TL, Adams LG, et al. Brucella requires a functional Type IV secretion system to elicit innate immune responses in mice. Cell Microbiol 2007;9:1851e69. [43] Stanley SA, Johndrow JE, Manzanillo P, Cox JS. The Type I IFN response to infection with Mycobacterium tuberculosis requires ESX-1mediated secretion and contributes to pathogenesis. J Immunol 2007;178:3143e52. [44] Stetson DB, Medzhitov R. Recognition of cytosolic DNA activates an IRF3-dependent innate immune response. Immunity 2006;24:93e103. [45] Chiu YH, Macmillan JB, Chen ZJ. RNA polymerase III detects cytosolic DNA and induces type I interferons through the RIG-I pathway. Cell 2009;138:576e91. [46] Ablasser A, Bauernfeind F, Hartmann G, Latz E, Fitzgerald KA, Hornung V. RIG-I-dependent sensing of poly(dA:dT) through the induction of an RNA polymerase III-transcribed RNA intermediate. Nat Immunol 2009;10:1065e72. [47] Takaoka A, Wang Z, Choi MK, Yanai H, Negishi H, Ban T, et al. DAI (DLM-1/ZBP1) is a cytosolic DNA sensor and an activator of innate immune response. Nature 2007;448:501e5. [48] Ishikawa H, Ma Z, Barber GN. STING regulates intracellular DNAmediated, type I interferon-dependent innate immunity. Nature 2009;461:788e92. [49] Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 2013;339:786e91. [50] Vogel JP, Andrews HL, Wong SK, Isberg RR. Conjugative transfer by the virulence system of Legionella pneumophila. Science 1998;279:873e6. [51] Opitz B, Vinzing M, van Laak V, Schmeck B, Heine G, Gunther S, et al. Legionella pneumophila induces IFNbeta in lung epithelial cells via IPS1 and IRF3, which also control bacterial replication. J Biol Chem 2006;281:36173e9. [52] Monroe KM, McWhirter SM, Vance RE. Identification of host cytosolic sensors and bacterial factors regulating the type I interferon response to Legionella pneumophila. PLoS Pathog 2009;5:e1000665. [53] de Almeida LA, Carvalho NB, Oliveira FS, Lacerda TL, Vasconcelos AC, Nogueira L, et al. MyD88 and STING signaling pathways are required for IRF3-mediated IFN-beta induction in response to Brucella abortus infection. PloS one 2011;6:e23135. [54] O'Connell RM, Saha SK, Vaidya SA, Bruhn KW, Miranda GA, Zarnegar B, et al. Type I interferon production enhances susceptibility to Listeria monocytogenes infection. J Exp Med 2004;200:437e45. [55] Wu J, Sun L, Chen X, Du F, Shi H, Chen C, et al. Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA. Science 2013;339:826e30. [56] Burdette DL, Monroe KM, Sotelo-Troha K, Iwig JS, Eckert B, Hyodo M, et al. STING is a direct innate immune sensor of cyclic di-GMP. Nature 2011;478:515e8. [57] Diner EJ, Burdette DL, Wilson SC, Monroe KM, Kellenberger CA, Hyodo M, et al. The innate immune DNA sensor cGAS produces a

[58]

[59]

[60]

[61]

[62]

[63]

[64]

[65]

[66]

[67]

[68]

[69]

[70]

[71]

[72]

[73]

noncanonical cyclic dinucleotide that activates human STING. Cell Reports 2013;3:1355e61. Ablasser A, Goldeck M, Cavlar T, Deimling T, Witte G, Rohl I, et al. cGAS produces a 2'-5'-linked cyclic dinucleotide second messenger that activates STING. Nature 2013;498:380e4. Hansen K, Prabakaran T, Laustsen A, Jorgensen SE, Rahbaek SH, Jensen SB, et al. Listeria monocytogenes induces IFNbeta expression through an IFI16-, cGAS- and STING-dependent pathway. EMBO J 2014;33(15):1654e66. Case CL, Shin S, Roy CR. Asc and Ipaf inflammasomes direct distinct pathways for caspase-1 activation in response to Legionella pneumophila. Infect Immun 2009;77:1981e91. Kailasan Vanaja S, Rathinam VA, Atianand MK, Kalantari P, Skehan B, Fitzgerald KA, et al. Bacterial RNA: DNA hybrids are activators of the NLRP3 inflammasome. Proc Natl Acad Sci U S A 2014;111:7765e70. Burckstummer T, Baumann C, Bluml S, Dixit E, Durnberger G, Jahn H, et al. An orthogonal proteomic-genomic screen identifies AIM2 as a cytoplasmic DNA sensor for the inflammasome. Nat Immunol 2009;10:266e72. Fernandes-Alnemri T, Yu JW, Datta P, Wu J, Alnemri ES. AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature 2009;458:509e13. Hornung V, Ablasser A, Charrel-Dennis M, Bauernfeind F, Horvath G, Caffrey DR, et al. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature 2009;458:514e8. Roberts TL, Idris A, Dunn JA, Kelly GM, Burnton CM, Hodgson S, et al. HIN-200 proteins regulate caspase activation in response to foreign cytoplasmic DNA. Science 2009;323:1057e60. Belhocine K, Monack DM. Francisella infection triggers activation of the AIM2 inflammasome in murine dendritic cells. Cell Microbiol 2012;14:71e80. Fernandes-Alnemri T, Yu JW, Juliana C, Solorzano L, Kang S, Wu J, et al. The AIM2 inflammasome is critical for innate immunity to Francisella tularensis. Nat Immunol 2010;11:385e93. Jones JW, Kayagaki N, Broz P, Henry T, Newton K, O'Rourke K, et al. Absent in melanoma 2 is required for innate immune recognition of Francisella tularensis. Proc Natl Acad Sci U S A 2010;107:9771e6. Rathinam VA, Jiang Z, Waggoner SN, Sharma S, Cole LE, Waggoner L, et al. The AIM2 inflammasome is essential for host defense against cytosolic bacteria and DNA viruses. Nat Immunol 2010;11:395e402. Wu J, Fernandes-Alnemri T, Alnemri ES. Involvement of the AIM2, NLRC4, and NLRP3 inflammasomes in caspase-1 activation by Listeria monocytogenes. J Clin Immunol 2010;30:693e702. Creasey EA, Isberg RR. The protein SdhA maintains the integrity of the Legionella-containing vacuole. Proc Natl Acad Sci U S A 2012;109:3481e6. Ge J, Gong YN, Xu Y, Shao F. Preventing bacterial DNA release and absent in melanoma 2 inflammasome activation by a Legionella effector functioning in membrane trafficking. Proc Natl Acad Sci U S A 2012;109:6193e8. Laguna RK, Creasey EA, Li Z, Valtz N, Isberg RR. A Legionella pneumophila-translocated substrate that is required for growth within macrophages and protection from host cell death. Proc Natl Acad Sci U S A 2006;103:18745e50.

Recognition of Legionella pneumophila nucleic acids by innate immune receptors.

Innate immune receptors evolved to sense conserved molecules that are present in microbes or are released during non-physiological conditions. Activat...
411KB Sizes 2 Downloads 9 Views