Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Dec 1.
Published in final edited form as: Curr Opin Virol. 2011 Oct 28;1(6):447–454. doi: 10.1016/j.coviro.2011.10.006

Toll-like Receptors: Key Players in Antiviral Immunity

Nicholas Arpaia 1, Gregory M Barton 1
PMCID: PMC3311989  NIHMSID: NIHMS331647  PMID: 22440908

Abstract

TLRs are a family of innate receptors whose specificities are predetermined in the germline. Therefore, TLRs have evolved to recognize conserved features of microbes. Viruses typically lack the conserved features common to other pathogen classes, so the innate immune system has evolved to recognize viral nucleic acid as a hallmark of viral infection. In this review we discuss examples of TLR-mediated viral recognition and the functional consequences of this recognition for antiviral immunity.

Introduction

Overview

Toll-like receptors (TLRs) are an essential arm of the innate immune response to bacteria, viruses and fungi and link recognition of distinct features of these microbes to the induction of pro-inflammatory signaling pathways [1,2]. These receptors are able to respond to broad classes of pathogens because each TLR recognizes specific conserved microbial features. Multiple microbial products can serve as ligands, including LPS (TLR4), lipopetides (TLR2/1 and TLR2/6), flagellin (TLR5), unmethylated CpG motifs in DNA (TLR9), and RNA (TLRs 3, 7 and 8). It has been proposed that targeting nucleic acids is a necessary strategy for viral recognition by the innate immune system because viruses lack any other shared, conserved features suitable for innate detection [3].

TLRs can be subdivided into two groups based on their localization within the cell and type of ligand they recognize. While TLR1, TLR2, TLR4, TLR5 and TLR6 are localized at the cell surface, the nucleic acid-sensing TLRs, TLR3, TLR7, TLR8 and TLR9 are localized intracellularly within endolysosomes [4]. This intracellular localization facilitates recognition of nucleic acids released from microbes degraded within endolysosomes. It is also likely that localization helps to limit recognition of self nucleic acids, although we will not discuss this aspect of TLR function as the topic has been recently reviewed elsewhere[3].

TLR signaling pathways

TLRs consist of an extracellular ligand-binding domain, a transmembrane domain, and a cytosolic signaling domain called the Toll-interleukin 1 receptor homology domain (TIR) [1]. Signaling occurs via homotypic binding of TIR domains on the receptor with those on signaling adaptors. All TLRs, with the exception of TLR3, utilize the common signaling adaptor myeloid differentiation factor 88 (MyD88). MyD88 also contains a death domain that recruits IL-1R-associated kinase (IRAK) family members upon recruitment to the TLR TIR. IRAK proteins then dissociate from the MyD88/TLR complex and interact with tumor necrosis factor receptor-associated factor 6 (TRAF6) to mediate downstream signaling. Both TLR3 and TLR4 are able to recruit a second adaptor, TIR-domain-containing adapter-inducing interferon-β (TRIF). TRIF activation initiates a TRAF3-dependent signaling cascade that results in the dimerization and activation of TANK (TRAF family member–associated NF-κB activator)-binding kinase 1 (TBK1) and inhibitor of NF-κB kinase (IKKi). This TBK1/IKKi complex leads to phosphorylation of the transcription factor IRF3, allowing it to translocate into the nucleus.

Type I IFN production by TLRs

One of the most important consequences of TLR activation for antiviral immunity is the induction of type I IFN, and all TLRs implicated in viral recognition can induce expression of this family of cytokines[5]. As mentioned above, TLR3 and TLR4 are able to activate the signaling adaptor TRIF, inducing IRF3 phosphorylation, translocation and the transcription of type I IFN genes. A second pathway for the induction of type I IFN involves the transcription factor IRF7 and takes place downstream of TLR7 and TLR9. Interestingly, this pathway functions primarily in a specialized subset of dendritic cells know as plasmacytoid dendritic cells (pDC) [3]. The ability for these cells to uniquely respond to TLR7 and TLR9 ligands by making IFN has been attributed to their high expression of IRF7. However, by delivering TLR7 and TLR9 ligands to early endosomal compartments via lipid complexes, researchers have been able to activate type I IFNs in macrophages as well [6]. These observations suggest that ligand trafficking may be the unique feature that allows IFN production within pDCs. Whether macrophages are able to make IFN via specialized ligand delivery in vivo is still unknown. However, the importance of this pathway in the recognition and control of viral replication has been repeatedly shown and will be discussed further, below.

TLR Recognition of Viruses

As discussed above, nucleic-acid sensing plays an important role in recognizing viral genomes. This requirement for sensing nucleic acid is due, in part, to limited viral ‘foreignness.’ Viruses utilize host machinery and nucleotides to assemble into infectious particles, and the innate immune system relies on ligand delivery of viral nucleic acid to intracellular compartments to distinguish viral ligands from self. Although there are examples of recognition of viral envelope proteins by TLRs, we will first discuss nucleic acid sensing and touch upon these other forms of recognition later.

Recognition of DNA viruses by TLR9

Unmethylated CpG motifs within bacterial nucleic acid were the first ligands identified to activate TLR9 [7]. Since then, many DNA viruses have also been shown to activate this TLR, including those from the herpesvirus[8-17], adenovirus[18], poxvirus[19] and torquetenovirus (TTV) families[20]. Both α- and β-herpesviruses have genomes that are rich in CpG motifs. Herpes simplex viruses (HSV) 1 and 2 have been shown to activate TLR9 in spleen and bone marrow cultures to produce pro-inflammatory cytokines and type I IFN [16,17], and similar results have also been observed with mouse cytomegalovirus (MCMV) [10,21,22]. Although the contribution of TLR9 recognition of these viruses has clearly been reported in vitro, the phenotype of TLR9-deficient mice in vivo does not completely phenocopy that of mice deficient in the signaling adaptor MyD88. These results suggested that other members of the TLR/IL-1R family might play a redundant role in recognition of these viruses, and it was later shown that the remaining MyD88-dependent recognition may be mediated by detection of viral envelope proteins by TLR2 and possibly RNA intermediates produced during viral replication by TLR3 and TLR7 (discussed later) [9,10,23].

It is noteworthy that very few examples exist of viruses suppressing CpG motifs within their genomes as a strategy to evade nucleic acid detection. This lack of evidence can perhaps be explained by the constraint on mutability of compact viral genomes with few non-coding sequences. One example of CpG suppression has recently been reported for a member of the γ-herpesvirus family, murine herpesvirus 68 (MHV68) [24]. In this study it was reported that CpG suppression, most likely mediated by cytosine to thymine conversion, within the genome of MHV68, allows the virus to evade TLR9 detection and largely avoid detection in vivo. Further, during latent stages of infection, CpG motifs within the viral genome were found to be methylated, and therefore unable to stimulate TLR9. These alterations in genome composition lead to reduced type I IFN and pro-inflammatory cytokine production. Nevertheless, it is difficult to prove that TLR9 recognition was the selective pressure leading to suppression of these motifs.

Recognition of ssRNA by TLR7/8

The first ligands identified to stimulate TLRs 7 and 8 were members of the imidazoquinolones family which share structural similarity with ribonucleosides [25]. The imidazoquinolones were long known to stimulate a potent antiviral response, and it wasn’t until the discovery of these TLRs that the receptors mediating this response were appreciated. The precise structural motifs or ssRNA sequences (analogous to CpG motifs for TLR9 stimulation) that are capable of stimulating TLR7 are not precisely known. G-U rich and even PolyU RNA are capable of stimulating TLR7 [26]. Single stranded RNA is extremely labile, and host-derived RNA rarely comes into contact with intracellular TLRs because it is rapidly broken down by extracellular RNAses. In contrast, viral particles protect viral RNA from degradation; destruction of viral particles within phagosomes releases viral RNA for recognition by TLRs 7 and 8.

Similarly to TLR9, TLRs 7 and 8 are capable of inducing type I IFNs in pDCs via the signaling adaptor MyD88. HIV, influenza and vesicular stomatitis virus (VSV) have all been reported to stimulate TLR7[26-28]. The genomic RNA of influenza virus is capable of inducing IFNalpha from mouse dendritic cells in a TLR7 dependent manner, and similar results were obtained when comparing stimulations of wildtype and TLR7-deficient cells with the U5 region of HIV-1 RNA.

Recognition of dsRNA by TLR3

TLR3 recognizes dsRNA as well as the synthetic analog, polyI:C [29-32]. This receptor signals through TRIF and leads to production of IFN in all cell types in which it is expressed. Despite the discovery that TLR3 is a potent inducer of IFN in response to dsRNA, in vivo evidence that TLR3 deficiency results in exacerbated disease in response to viral infection was lacking for many years —leading some to believe that TLR3 may not actually be a receptor of viral dsRNA directly [33].

It has since been appreciated, however, that TLR3 may play a role in recognition of dsRNA intermediates within engulfed apoptotic cells that have been infected with ssRNA viruses [34]. The viruses used in this study were encephalomyocarditis virus and Semliki Forest virus, both of which are ssRNA viruses, lending to the possibility that dsRNA, produced during the replication of these viruses, is detected by TLR3 within the apoptotic cells after phagocytosis. Intersetingly, several reports have also shown that in models of West Nile virus and Influenza A virus (both ssRNA viruses), pro-inflammatory cytokines produced in a TLR3-dependent manner can actually lead to increased viral pathology [35-39]. These reports further confirm the notion that dsRNA intermediates are likely generated and detected in vivo. Adding further complexity to how TLR3 may participate in viral recognition, TLR3-deficient mice are more susceptible to the DNA virus MCMV [40]. Also, human patients with deficiencies in TLR3 or signaling molecules downstream of this receptor show predisposition to HSV-induced encephalitis [41-45]. This suggests that dsRNA may be produced by the secondary structures of viral transcripts derived from the viral genome of DNA viruses.

Non-nucleic acid recognition of Viral ligands by TLRs

As mentioned earlier, the recognition of viral nucleic acid by TLRs may be the most reliable method of detecting viruses because of ligand delivery, the constraints of mutating viral genomic contents, and the fact that the viral particle is assembled using host machinery. Despite these conceptual considerations, there are a handful of examples of viral proteins serving as TLR ligands. In principle, the sequence or structure of these detected proteins must be constrained for this strategy to work. Mutating the regions that are detected by TLRs should make the viral particle non-functional; otherwise, the virus would easily be able to mutate these proteins to avoid detection. The clear exception to this idea is when TLR-mediated inflammation is important for viral pathology, as is supported in some infection models discussed below.

HSV-1, HSV-2, HCMV, MCMV, VV, respiratory syncytial virus (RSV) and the hemagglutinin protein of measles virus have all been shown to activate TLR2 [23,46-51]. In response to VV infection, TLR2 on inflammatory monocytes is capable of inducing type I IFN, and depletion of these cells leads to elevated levels of Vaccinia virus in ovaries of mice[51]. TLR2 signaling has also been shown to be important in CD8 T cells where it is critical for clonal expansion and memory formation following VV infection[52]. These examples suggest that TLR2-mediated detection of VV is important for the host to be able to generate a protective response against the virus.

In a very different example, TLR2 activation by certain isolates of HSV-1 seems to induce excessive inflammation. In this model, TLR2-deficient animals are somewhat protected from HSV-induced encephalitis [50]. The responding cells in this HSV example may be generating a pro-inflammatory response without the production of protective IFN, aberrantly leading to increased encephalitis pathology.

Mouse mammary tumor virus (MMTV) and RSV have both been implicated to activate TLR4 [53-57]. In the case of MMTV, TLR4 leads to the production of IL-10, an anti-inflammatory cytokine, and allows for the virus to enter a stage of persistence by dampening the immune response against it [57]. On the other hand, the role of TLR4 detection of RSV seems to be protective. TLR4-deficient mice have reduced IL-6 production in response to RSV and infants with polymorphisms in TLR4 are at higher risk for severe RSV infection [55,56].

Detecting the contribution of non-nucleic acid-sensing TLRs to the overall innate immune response may require that mice deficient in TLR2 or TLR4 are crossed to those lacking nucleic acid-sensing TLRs that can mask the contribution of viral protein detection. Infection of these mice will allow us to gain insight into the potentially wider contribution of TLR detection of viral proteins as a strategy for host protection or a viral strategy for exploiting these signals.

Functional Consequences of TLR activation

Viral evasion strategies

As discussed above, there are many examples of TLRs detecting viral nucleic acid or viral envelope proteins. Concomitant with the evolution of these innate detection strategies, viruses have acquired genes whose products inhibit TLR signaling pathways, thus evading detection by TLRs or specific downstream consequences of TLR activation. While not the main focus of this review, we have outlined examples of viral evasion of TLR signaling pathways in Table 1. Notably, there are relatively few examples of viral gene products that specifically inhibit TLR signaling, especially when compared to the many examples of viral inhibition of cytosolic innate immune sensors. Furthermore, several of the downstream TLR signaling components targeted by viruses are common to both TLR and RLR signaling. Even TRIF has recently been implicated as a downstream adaptor for the cytosolic dsRNA-sensing helicases DDX1, DDX21, and DHX36 [58].

Target Viruses References
MyD88, IRAK1/2/4 Herpes simplex virus,
Poxviruses, Hepatitis C virus
[59-61]
RIP1, TRAF3/6 West Nile virus, Respiratory syncytial
virus
[62-64]
TRIF Influenza, Reovirus, Coxsackievirus,
Karposi’s sacrcoma-associated herpes
virus, African swine fever virus,
Hepatitis A/C viruses
[58,65-69]
TBK1, IKKi, IRF3/7 Ebola virus, Hepatitis C virus, Vaccinia
virus, Varicella-Zoster virus, Rabies
virus, Foot-and-mouth disease virus,
Epstein-Barr virus, Measles virus
[70-78]

An important consideration when evaluating the selective pressure responsible for these evasion strategies is that TLRs detect viral particles that fail to infect cells. In fact, the ability of innate immune cells to recognize viruses without being infected is likely an important mechanism for the host to avoid viral immune evasion strategies. Even viruses within infected cells that undergo apoptosis can be be detected by TLRs expressed by phagocytic cells. Once viruses have infected a cell, their genomes, replication intermediates, and products of gene expression can be sensed by innate cytosolic sensors. This potential recognition coupled with the ability to express evasion genes in infected cells may explain why so many viruses have evolved strategies to block or circumvent cytosolic innate sensing.

Activation of TLRs on antigen-presenting cells, such as macrophages and dendritic cells (DCs), leads to upregulation of costimulatory molecules, the production of inflammatory cytokines, and the generation of a functional adaptive immune response. We focus on these functional consequences of TLR signaling in the next section as a means of understanding precisely how TLR-detection of viruses contributes to induction of antiviral immunity.

Phagosome-autonomous responses

A specific feature of TLR responses that does not apply to cytosolic sensors is the detection of defective viral particles that do not cause active infection but activate TLRs upon degradation in the phagosome. This also applies to the engulfment of apoptotic virally infected cells. The detection of these viral products highlights the phagosome-autonomous functions of TLR signaling, outlined in Figure 1. As mentioned, TLR3, TLR7 and TLR9 reside within the phagosome and TLR4 has been shown to be internalizaed with ligand upon activation [79]. Once activated, these TLRs induce more rapid acidification of the phagosome, presumably through the recruitment of the V1 sector subunits A and E (of the Vacuolar-ATPase) to the phagosomal membrane[80-83]. The release of reactive oxygen and nitrogen species by recruitment of NADPH oxidase or mitochondria to the phagosome is another hallmark of phagosome-autonomous TLR-mediated maturation [84-86]. Along with proteases that are active at the low pH of the mature phagosome, viral proteins are degraded into peptides, loaded into MHC class II and shuttled to the surface of the cell. Further, antigens present within this mature phagosome can be loaded into MHC class I for activation of cytotoxic CD8 T cells via a process known as cross presentation [87-89]. The precise details of the cross presentation pathway are still elusive, however, it is widely accepted that engulfed, virally-infected dead or dying cells activate TLRs and that their viral antigens are capable of being presented in class I MHC after engulfment by CD8α+ dendritic cells [34].

Figure 1. Functional Consequences of TLR Signaling.

Figure 1

Engulfed viruses are degraded in endosomes and/or phagosomes. Released viral nucleic acid stimulates intracellular nucleic acid-sensing TLRs (red) leading to phagosome autonomous responses including recruitment of V-ATPase (green) and NADPH oxidase (purple). Viral antigens generated upon degradation of the virus are loaded into MHC class II and shuttled to the surface (pink). In certain cell types, antigens are loaded into MHC class I via cross presentation (dashed line, green). TLR activation also leads to the upregulation of costimulatory molecules, CD40 (brown) and CD86 (lime), as well as a signaling cascade that leads to the transcription of cytokine genes.

Upregulation of Cell Surface Molecules

TLR signaling also leads to the upregulation of costimulatory molecules on the surface of the signaling cell. These include CD80, CD86 and CD40 (Figure 1). These molecules are required along with antigen presentation for the activation of T lymphocytes. Class II MHC molecules presenting viral antigens are also upregulated on the cells surface. These molecules, upregulated via TLR signaling, serve to activate T cells for protective immunity.

Cytokine production

As discussed previously, the intracellular nucleic acid sensing TLRs are capable of inducing type I IFN in pDCs and TLR2 and TLR4 both recognize viral products and lead to the induction of IFN in certain cell types. The overall importance of this cytokine is exemplified by the extreme susceptibility of type I IFN receptor mice to viral infections [5]. Signaling via the type I IFN receptor leads to the upregulation of many genes that serve the function of shutting off host translation and inducing apoptosis, thus rendering the cell inhospitable for viral replication. This cytokine is also critically important for other immune responses. For example, IFN can lead to the differentiation and activation of natural killer (NK) cells. These cells serve the function of killing virally infected cells that try to evade antigen presentation via MHC down-regulation [22,90,91]. Type I IFN has also been shown to induce cross presentation by dendritic cells and leads to the upregualtion of MHC Class I, further increasing the activation of CD8 T cells.

Other pro-inflammatory cytokines that are produced downstream of TLRs include IL-6, IL-12 and TNF-α. These cytokines serve numerous functions including promoting the survival and proliferation of B and T cells, activating NK cells, and inducing local inflammation to recruit other cells to sites of infection. In one circumstance, as discussed above, the production of IL-10 downstream of TLR4 seems to be exploited by MMTV because IL-10 is a potent suppressant of immune cell function.

Conclusion

Our conceptual understanding of how TLRs recognize viruses has expanded significantly over the past few years. Recent discoveries demonstrate a role for TLR3 in recognizing virally infected apoptotic cells and for the detection of HSV in the central nervous system. Further, the role of TRIF as an adaptor for cytosolic RNA sensors and the surprising finding that TLR2 is capable of making IFN in certain cell types have been reported. A common recurring theme is that viral recognition by TLRs always seems to lead to the production of type I IFN. The role of this cytokine in the generation of an effective antiviral response has been repeatedly shown, and inhibiting these signaling pathways seems to be a common viral evasion mechanism. Detection of non-nucleic acid viral ligands by TLRs has also been more recently appreciated. The implications of these detection strategies may be that viruses have begun to suppress nucleic acid motifs that can be detected by TLRs, although there remain very few examples of this evasion mechanism. Further understanding of the interactions between virally infected cells and TLR signaling pathways may provide important advances in the study of protective adaptive immunity and vaccine development.

Highlights.

  • - TLRs detect viral nucleic acid and envelope proteins

  • - Defective viral particles that do not infect cells can also activate TLRs

  • - TLRs can detect viral nucleic acid by engulfing infected apoptotic cells

  • - TLR signaling upregulates costimulatory molecules for T cell activation

  • - Selection of antigens for presentation is mediated by phagosomal TLR activation

Acknowledgements

We apologize to any colleagues whose work was not cited because of space constraints. We thank members of the Barton lab for helpful discussions. Research in our laboratory related to this review is funded by NIH/NIAID R01AI072429.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: Update on toll-like receptors. Nat Immunol. 2010;11(5):373–384. doi: 10.1038/ni.1863. [DOI] [PubMed] [Google Scholar]
  • 2.Iwasaki A, Medzhitov R. Regulation of adaptive immunity by the innate immune system. Science. 2010;327(5963):291–295. doi: 10.1126/science.1183021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Barbalat R, Ewald SE, Mouchess ML, Barton GM. Nucleic acid recognition by the innate immune system. Annual review of immunology. 2011;29:185–214. doi: 10.1146/annurev-immunol-031210-101340. [DOI] [PubMed] [Google Scholar]
  • 4.Barton GM, Kagan JC. A cell biological view of toll-like receptor function: Regulation through compartmentalization. Nat Rev Immunol. 2009;9(8):535–542. doi: 10.1038/nri2587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Stetson DB, Medzhitov R. Type i interferons in host defense. Immunity. 2006;25(3):373–381. doi: 10.1016/j.immuni.2006.08.007. [DOI] [PubMed] [Google Scholar]
  • 6.Honda K, Ohba Y, Yanai H, Negishi H, Mizutani T, Takaoka A, Taya C, Taniguchi T. Spatiotemporal regulation of myd88-irf-7 signalling for robust type-i interferon induction. Nature. 2005;434(7036):1035–1040. doi: 10.1038/nature03547. [DOI] [PubMed] [Google Scholar]
  • 7.Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, Matsumoto M, Hoshino K, Wagner H, Takeda K, Akira S. A toll-like receptor recognizes bacterial DNA. Nature. 2000;408(6813):740–745. doi: 10.1038/35047123. [DOI] [PubMed] [Google Scholar]
  • 8.Sato A, Iwasaki A. Induction of antiviral immunity requires toll-like receptor signaling in both stromal and dendritic cell compartments. Proc Natl Acad Sci USA. 2004;101(46):16274–16279. doi: 10.1073/pnas.0406268101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Fiola S, Gosselin D, Takada K, Gosselin J. Tlr9 contributes to the recognition of ebv by primary monocytes and plasmacytoid dendritic cells. J Immunol. 2010;185(6):3620–3631. doi: 10.4049/jimmunol.0903736. [DOI] [PubMed] [Google Scholar]
  • 10.Zucchini N, Bessou G, Traub S, Robbins SH, Uematsu S, Akira S, Alexopoulou L, Dalod M. Cutting edge: Overlapping functions of tlr7 and tlr9 for innate defense against a herpesvirus infection. J Immunol. 2008;180(9):5799–5803. doi: 10.4049/jimmunol.180.9.5799. [DOI] [PubMed] [Google Scholar]
  • 11.Lund JM, Linehan MM, Iijima N, Iwasaki A. Cutting edge: Plasmacytoid dendritic cells provide innate immune protection against mucosal viral infection in situ. J Immunol. 2006;177(11):7510–7514. doi: 10.4049/jimmunol.177.11.7510. [DOI] [PubMed] [Google Scholar]
  • 12.Feldman SB, Ferraro M, Zheng HM, Patel N, Gould-Fogerite S, Fitzgerald-Bocarsly P. Viral induction of low frequency interferon-alpha producing cells. Virology. 1994;204(1):1–7. doi: 10.1006/viro.1994.1504. [DOI] [PubMed] [Google Scholar]
  • 13.Dalod M, Hamilton T, Salomon R, Salazar-Mather TP, Henry SC, Hamilton JD, Biron CA. Dendritic cell responses to early murine cytomegalovirus infection: Subset functional specialization and differential regulation by interferon alpha/beta. J Exp Med. 2003;197(7):885–898. doi: 10.1084/jem.20021522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Rölle A, Olweus J. Dendritic cells in cytomegalovirus infection: Viral evasion and host countermeasures. APMIS. 2009;117(5-6):413–426. doi: 10.1111/j.1600-0463.2009.02449.x. [DOI] [PubMed] [Google Scholar]
  • 15.Asselin-Paturel C, Boonstra A, Dalod M, Durand I, Yessaad N, Dezutter-Dambuyant C, Vicari A, O’Garra A, Biron C, Brière F, Trinchieri G. Mouse type i ifn-producing cells are immature apcs with plasmacytoid morphology. Nat Immunol. 2001;2(12):1144–1150. doi: 10.1038/ni736. [DOI] [PubMed] [Google Scholar]
  • 16.Lund J, Sato A, Akira S, Medzhitov R, Iwasaki A. Toll-like receptor 9-mediated recognition of herpes simplex virus-2 by plasmacytoid dendritic cells. J Exp Med. 2003;198(3):513–520. doi: 10.1084/jem.20030162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Krug A, Luker GD, Barchet W, Leib DA, Akira S, Colonna M. Herpes simplex virus type 1 activates murine natural interferon-producing cells through toll-like receptor 9. Blood. 2004;103(4):1433–1437. doi: 10.1182/blood-2003-08-2674. [DOI] [PubMed] [Google Scholar]
  • 18.Zhu J, Huang X, Yang Y. Innate immune response to adenoviral vectors is mediated by both toll-like receptor-dependent and -independent pathways. J Virol. 2007;81(7):3170–3180. doi: 10.1128/JVI.02192-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Samuelsson C, Hausmann J, Lauterbach H, Schmidt M, Akira S, Wagner H, Chaplin P, Suter M, O’Keeffe M, Hochrein H. Survival of lethal poxvirus infection in mice depends on tlr9, and therapeutic vaccination provides protection. J Clin Invest. 2008;118(5):1776–1784. doi: 10.1172/JCI33940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Rocchi J, Ricci V, Albani M, Lanini L, Andreoli E, Macera L, Pistello M, Ceccherini-Nelli L, Bendinelli M, Maggi F. Torquetenovirus DNA drives proinflammatory cytokines production and secretion by immune cells via toll-like receptor 9. Virology. 2009;394(2):235–242. doi: 10.1016/j.virol.2009.08.036. [DOI] [PubMed] [Google Scholar]
  • 21.Krug A, French AR, Barchet W, Fischer JAA, Dzionek A, Pingel JT, Orihuela MM, Akira S, Yokoyama WM, Colonna M. Tlr9-dependent recognition of mcmv by ipc and dc generates coordinated cytokine responses that activate antiviral nk cell function. Immunity. 2004;21(1):107–119. doi: 10.1016/j.immuni.2004.06.007. [DOI] [PubMed] [Google Scholar]
  • 22.Delale T, Paquin A, Asselin-Paturel C, Dalod M, Brizard G, Bates EEM, Kastner P, Chan S, Akira S, Vicari A, Biron CA, et al. Myd88-dependent and -independent murine cytomegalovirus sensing for ifn-alpha release and initiation of immune responses in vivo. J Immunol. 2005;175(10):6723–6732. doi: 10.4049/jimmunol.175.10.6723. [DOI] [PubMed] [Google Scholar]
  • 23.Sato A, Linehan MM, Iwasaki A. Dual recognition of herpes simplex viruses by tlr2 and tlr9 in dendritic cells. Proc Natl Acad Sci USA. 2006;103(46):17343–17348. doi: 10.1073/pnas.0605102103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **24.Pezda AC, Penn A, Barton GM, Coscoy L. Suppression of tlr9 immunostimulatory motifs in the genome of a gammaherpesvirus. J Immunol. 2011;187(2):887–896. doi: 10.4049/jimmunol.1003737. CpG suppresion within the genome of MHV68 is suggested to be a method for inhibiting TLR9 recognition when compared to MCMV. During latent MHV68 infection CpG motifs are methylated.
  • 25.Hemmi H, Kaisho T, Takeuchi O, Sato S, Sanjo H, Hoshino K, Horiuchi T, Tomizawa H, Takeda K, Akira S. Small anti-viral compounds activate immune cells via the tlr7 myd88-dependent signaling pathway. Nat Immunol. 2002;3(2):196–200. doi: 10.1038/ni758. [DOI] [PubMed] [Google Scholar]
  • 26.Diebold SS, Kaisho T, Hemmi H, Akira S, Reis e Sousa C. Innate antiviral responses by means of tlr7-mediated recognition of single-stranded rna. Science. 2004;303(5663):1529–1531. doi: 10.1126/science.1093616. [DOI] [PubMed] [Google Scholar]
  • 27.Lund JM, Alexopoulou L, Sato A, Karow M, Adams NC, Gale NW, Iwasaki A, Flavell RA. Recognition of single-stranded rna viruses by toll-like receptor 7. Proc Natl Acad Sci USA. 2004;101(15):5598–5603. doi: 10.1073/pnas.0400937101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C, Akira S, Lipford G, Wagner H, Bauer S. Species-specific recognition of single-stranded rna via toll-like receptor 7 and 8. Science. 2004;303(5663):1526–1529. doi: 10.1126/science.1093620. [DOI] [PubMed] [Google Scholar]
  • 29.Choe J, Kelker MS, Wilson IA. Crystal structure of human toll-like receptor 3 (tlr3) ectodomain. Science. 2005;309(5734):581–585. doi: 10.1126/science.1115253. [DOI] [PubMed] [Google Scholar]
  • 30.Bell JK, Botos I, Hall PR, Askins J, Shiloach J, Davies DR, Segal DM. The molecular structure of the tlr3 extracellular domain. J Endotoxin Res. 2006;12(6):375–378. doi: 10.1179/096805106X118780. [DOI] [PubMed] [Google Scholar]
  • 31.Bell JK, Botos I, Hall PR, Askins J, Shiloach J, Segal DM, Davies DR. The molecular structure of the toll-like receptor 3 ligand-binding domain. Proc Natl Acad Sci USA. 2005;102(31):10976–10980. doi: 10.1073/pnas.0505077102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Alexopoulou L, Holt AC, Medzhitov R, Flavell RA. Recognition of double-stranded rna and activation of nf-kappab by toll-like receptor 3. Nature. 2001;413(6857):732–738. doi: 10.1038/35099560. [DOI] [PubMed] [Google Scholar]
  • 33.Edelmann KH, Richardson-Burns S, Alexopoulou L, Tyler KL, Flavell RA, Oldstone MBA. Does toll-like receptor 3 play a biological role in virus infections? Virology. 2004;322(2):231–238. doi: 10.1016/j.virol.2004.01.033. [DOI] [PubMed] [Google Scholar]
  • 34.Schulz O, Diebold SS, Chen M, Näslund TI, Nolte MA, Alexopoulou L, Azuma Y-T, Flavell RA, Liljeström P, Reis e Sousa C. Toll-like receptor 3 promotes cross-priming to virus-infected cells. Nature. 2005;433(7028):887–892. doi: 10.1038/nature03326. [DOI] [PubMed] [Google Scholar]
  • 35.Le Goffic R, Pothlichet J, Vitour D, Fujita T, Meurs E, Chignard M, Si-Tahar M. Cutting edge: Influenza a virus activates tlr3-dependent inflammatory and rig-i-dependent antiviral responses in human lung epithelial cells. J Immunol. 2007;178(6):3368–3372. doi: 10.4049/jimmunol.178.6.3368. [DOI] [PubMed] [Google Scholar]
  • 36.Le Goffic R, Balloy V, Lagranderie M, Alexopoulou L, Escriou N, Flavell R, Chignard M, Si-Tahar M. Detrimental contribution of the toll-like receptor (tlr)3 to influenza a virus-induced acute pneumonia. PLoS Pathog. 2006;2(6):e53. doi: 10.1371/journal.ppat.0020053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hidaka F, Matsuo S, Muta T, Takeshige K, Mizukami T, Nunoi H. A missense mutation of the toll-like receptor 3 gene in a patient with influenza-associated encephalopathy. Clin Immunol. 2006;119(2):188–194. doi: 10.1016/j.clim.2006.01.005. [DOI] [PubMed] [Google Scholar]
  • *38.Daffis S, Samuel MA, Suthar MS, Gale M, Diamond MS. Toll-like receptor 3 has a protective role against west nile virus infection. J Virol. 2008;82(21):10349–10358. doi: 10.1128/JVI.00935-08. Demonstrate a protecrive role for TLR3 in restricting West Nile virus replication in neurons.
  • 39.Wang T, Town T, Alexopoulou L, Anderson JF, Fikrig E, Flavell RA. Toll-like receptor 3 mediates west nile virus entry into the brain causing lethal encephalitis. Nat Med. 2004;10(12):1366–1373. doi: 10.1038/nm1140. [DOI] [PubMed] [Google Scholar]
  • 40.Tabeta K, Georgel P, Janssen E, Du X, Hoebe K, Crozat K, Mudd S, Shamel L, Sovath S, Goode J, Alexopoulou L, et al. Toll-like receptors 9 and 3 as essential components of innate immune defense against mouse cytomegalovirus infection. Proc Natl Acad Sci USA. 2004;101(10):3516–3521. doi: 10.1073/pnas.0400525101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Audry M, Ciancanelli M, Yang K, Cobat A, Chang H-H, Sancho-Shimizu V, Lorenzo L, Niehues T, Reichenbach J, Li X-X, Israel A, et al. Nemo is a key component of nf-κb- and irf-3-dependent tlr3-mediated immunity to herpes simplex virus. J Allergy Clin Immunol. 2011;128(3):610–617. e614. doi: 10.1016/j.jaci.2011.04.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **42.de Diego R Pérez, Sancho-Shimizu V, Lorenzo L, Puel A, Plancoulaine S, Picard C, Herman M, Cardon A, Durandy A, Bustamante J, Vallabhapurapu S, et al. Human traf3 adaptor molecule deficiency leads to impaired toll-like receptor 3 response and susceptibility to herpes simplex encephalitis. Immunity. 2010;33(3):400–411. doi: 10.1016/j.immuni.2010.08.014. Show a link between TLR3 signaling via TRAF3 for the induction of protective IFNs and immunity against HSV-1 in the central nervous system.
  • 43.Zhang S-Y, Jouanguy E, Ugolini S, Smahi A, Elain G, Romero P, Segal D, Sancho-Shimizu V, Lorenzo L, Puel A, Picard C, et al. Tlr3 deficiency in patients with herpes simplex encephalitis. Science. 2007;317(5844):1522–1527. doi: 10.1126/science.1139522. [DOI] [PubMed] [Google Scholar]
  • 44.Casrouge A, Zhang S-Y, Eidenschenk C, Jouanguy E, Puel A, Yang K, Alcais A, Picard C, Mahfoufi N, Nicolas N, Lorenzo L, et al. Herpes simplex virus encephalitis in human unc-93b deficiency. Science. 2006;314(5797):308–312. doi: 10.1126/science.1128346. [DOI] [PubMed] [Google Scholar]
  • **45.Guo Y, Audry M, Ciancanelli M, Alsina L, Azevedo J, Herman M, Anguiano E, Sancho-Shimizu V, Lorenzo L, Pauwels E, Philippe PB, et al. Herpes simplex virus encephalitis in a patient with complete tlr3 deficiency: Tlr3 is otherwise redundant in protective immunity. J Exp Med. 2011 doi: 10.1084/jem.20101568. A patient with TLR3 deficiency highlights the importance of this receptor in the central nervous system for immuity against HSV-1. PBMCs showed no defect in response to HSV-1 suggesting a redundant role of TLR3 outside of the nervous system.
  • 46.Murawski MR, Bowen GN, Cerny AM, Anderson LJ, Haynes LM, Tripp RA, Kurt-Jones EA, Finberg RW. Respiratory syncytial virus activates innate immunity through toll-like receptor 2. J Virol. 2009;83(3):1492–1500. doi: 10.1128/JVI.00671-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Bieback K, Lien E, Klagge IM, Avota E, Schneider-Schaulies J, Duprex WP, Wagner H, Kirschning CJ, Ter Meulen V, Schneider-Schaulies S. Hemagglutinin protein of wild-type measles virus activates toll-like receptor 2 signaling. J Virol. 2002;76(17):8729–8736. doi: 10.1128/JVI.76.17.8729-8736.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Szomolanyi-Tsuda E, Liang X, Welsh RM, Kurt-Jones EA, Finberg RW. Role for tlr2 in nk cell-mediated control of murine cytomegalovirus in vivo. J Virol. 2006;80(9):4286–4291. doi: 10.1128/JVI.80.9.4286-4291.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Compton T, Kurt-Jones EA, Boehme KW, Belko J, Latz E, Golenbock DT, Finberg RW. Human cytomegalovirus activates inflammatory cytokine responses via cd14 and toll-like receptor 2. J Virol. 2003;77(8):4588–4596. doi: 10.1128/JVI.77.8.4588-4596.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kurt-Jones EA, Chan M, Zhou S, Wang J, Reed G, Bronson R, Arnold MM, Knipe DM, Finberg RW. Herpes simplex virus 1 interaction with toll-like receptor 2 contributes to lethal encephalitis. Proc Natl Acad Sci USA. 2004;101(5):1315–1320. doi: 10.1073/pnas.0308057100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *51.Barbalat R, Lau L, Locksley RM, Barton GM. Toll-like receptor 2 on inflammatory monocytes induces type i interferon in response to viral but not bacterial ligands. Nat Immunol. 2009;10(11):1200–1207. doi: 10.1038/ni.1792. The first account that TLR2 is able to induce type I IFN in response to Vaccinia and other viruses, but only in inflammatory monocytes. Depletion of these cells resulted in higher Vaccinia titres in vivo.
  • **52.Quigley M, Martinez J, Huang X, Yang Y. A critical role for direct tlr2- myd88 signaling in cd8 t-cell clonal expansion and memory formation following vaccinia viral infection. Blood. 2009;113(10):2256–2264. doi: 10.1182/blood-2008-03-148809. This paper demonstrates a role for TLR2 recognition of vaccinia virus in CD8 T cells. TLR2 signaling induces clonal expansion and memory T cell formation and is critical for immunity to infection.
  • 53.Burzyn D, Rassa JC, Kim D, Nepomnaschy I, Ross SR, Piazzon I. Toll-like receptor 4-dependent activation of dendritic cells by a retrovirus. J Virol. 2004;78(2):576–584. doi: 10.1128/JVI.78.2.576-584.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Rassa JC, Meyers JL, Zhang Y, Kudaravalli R, Ross SR. Murine retroviruses activate b cells via interaction with toll-like receptor 4. Proc Natl Acad Sci USA. 2002;99(4):2281–2286. doi: 10.1073/pnas.042355399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Tal G, Mandelberg A, Dalal I, Cesar K, Somekh E, Tal A, Oron A, Itskovich S, Ballin A, Houri S, Beigelman A, et al. Association between common toll-like receptor 4 mutations and severe respiratory syncytial virus disease. J Infect Dis. 2004;189(11):2057–2063. doi: 10.1086/420830. [DOI] [PubMed] [Google Scholar]
  • 56.Kurt-Jones EA, Popova L, Kwinn L, Haynes LM, Jones LP, Tripp RA, Walsh EE, Freeman MW, Golenbock DT, Anderson LJ, Finberg RW. Pattern recognition receptors tlr4 and cd14 mediate response to respiratory syncytial virus. Nat Immunol. 2000;1(5):398–401. doi: 10.1038/80833. [DOI] [PubMed] [Google Scholar]
  • 57.Jude BA, Pobezinskaya Y, Bishop J, Parke S, Medzhitov RM, Chervonsky AV, Golovkina TV. Subversion of the innate immune system by a retrovirus. Nat Immunol. 2003;4(6):573–578. doi: 10.1038/ni926. [DOI] [PubMed] [Google Scholar]
  • **58.Zhang Z, Kim T, Bao M, Facchinetti V, Jung SY, Ghaffari AA, Qin J, Cheng G, Liu Y-J. Ddx1, ddx21, and dhx36 helicases form a complex with the adaptor molecule trif to sense dsrna in dendritic cells. Immunity. 2011;34(6):866–878. doi: 10.1016/j.immuni.2011.03.027. Demonstrate a role for TRIF as the signaling adaptor that complexes with dsRNA sensor helicases in myeloid dendritic cells.
  • 59.van Lint AL, Murawski MR, Goodbody RE, Severa M, Fitzgerald KA, Finberg RW, Knipe DM, Kurt-Jones EA. Herpes simplex virus immediate-early icp0 protein inhibits toll-like receptor 2-dependent inflammatory responses and nf-kappab signaling. J Virol. 2010;84(20):10802–10811. doi: 10.1128/JVI.00063-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Keating SE, Maloney GM, Moran EM, Bowie AG. Irak-2 participates in multiple toll-like receptor signaling pathways to nfkappab via activation of traf6 ubiquitination. J Biol Chem. 2007;282(46):33435–33443. doi: 10.1074/jbc.M705266200. [DOI] [PubMed] [Google Scholar]
  • 61.Abe T, Kaname Y, Hamamoto I, Tsuda Y, Wen X, Taguwa S, Moriishi K, Takeuchi O, Kawai T, Kanto T, Hayashi N, et al. Hepatitis c virus nonstructural protein 5a modulates the toll-like receptor-myd88-dependent signaling pathway in macrophage cell lines. J Virol. 2007;81(17):8953–8966. doi: 10.1128/JVI.00649-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Swedan S, Andrews J, Majumdar T, Musiyenko A, Barik S. Multiple functional domains and complexes of the two nonstructural proteins of human respiratory syncytial virus contribute to interferon suppression and cellular location. J Virol. 2011;85(19):10090–10100. doi: 10.1128/JVI.00413-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Swedan S, Musiyenko A, Barik S. Respiratory syncytial virus nonstructural proteins decrease levels of multiple members of the cellular interferon pathways. J Virol. 2009;83(19):9682–9693. doi: 10.1128/JVI.00715-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Arjona A, Ledizet M, Anthony K, Bonafé N, Modis Y, Town T, Fikrig E. West nile virus envelope protein inhibits dsrna-induced innate immune responses. J Immunol. 2007;179(12):8403–8409. doi: 10.4049/jimmunol.179.12.8403. [DOI] [PubMed] [Google Scholar]
  • *65.Mukherjee A, Morosky SA, Delorme-Axford E, Dybdahl-Sissoko N, Oberste MS, Wang T, Coyne CB. The coxsackievirus b 3c protease cleaves mavs and trif to attenuate host type i interferon and apoptotic signaling. PLoS Pathog. 2011;7(3):e1001311. doi: 10.1371/journal.ppat.1001311. A cysteine protease encoded by coxsackievirus cleaves TRIF and inhibits the production of type I IFN and apoptosis. Cleavage sites within the N- and C- terminus of TRIF are determined and the intracellular colocalization between TRIF and the protease are shown.
  • 66.Ahmad H, Gubbels R, Ehlers E, Meyer F, Waterbury T, Lin R, Zhang L. Kaposi sarcoma-associated herpesvirus degrades cellular toll-interleukin-1 receptor domain-containing adaptor-inducing beta-interferon (trif) J Biol Chem. 2011;286(10):7865–7872. doi: 10.1074/jbc.M110.191452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.de Oliveira VL, Almeida SCP, Soares HR, Crespo A, Marshall-Clarke S, Parkhouse RME. A novel tlr3 inhibitor encoded by african swine fever virus (asfv) Arch Virol. 2011;156(4):597–609. doi: 10.1007/s00705-010-0894-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **68.Qu L, Feng Z, Yamane D, Liang Y, Lanford RE, Li K, Lemon SM. Disruption of tlr3 signaling due to cleavage of trif by the hepatitis a virus protease-polymerase processing intermediate, 3cd. PLoS Pathog. 2011;7(9):e1002169. doi: 10.1371/journal.ppat.1002169. This paper reports on a protease within Hepatitis A virus derived from 3Cpro that cleaves TRIF. 3Cpro is processed into two proteases, 3ABC and 3CD that have specificity for MAVS and TRIF cleavage, respectively.
  • 69.Li K, Foy E, Ferreon JC, Nakamura M, Ferreon ACM, Ikeda M, Ray SC, Gale M, Lemon SM. Immune evasion by hepatitis c virus ns3/4a protease-mediated cleavage of the toll-like receptor 3 adaptor protein trif. Proc Natl Acad Sci USA. 2005;102(8):2992–2997. doi: 10.1073/pnas.0408824102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wang D, Fang L, Li P, Sun L, Fan J, Zhang Q, Luo R, Liu X, Li K, Chen H, Chen Z, et al. The leader proteinase of foot-and-mouth disease virus negatively regulates the type i interferon pathway by acting as a viral deubiquitinase. J Virol. 2011;85(8):3758–3766. doi: 10.1128/JVI.02589-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Rieder M, Brzózka K, Pfaller CK, Cox JH, Stitz L, Conzelmann K-K. Genetic dissection of interferon-antagonistic functions of rabies virus phosphoprotein: Inhibition of interferon regulatory factor 3 activation is important for pathogenicity. J Virol. 2011;85(2):842–852. doi: 10.1128/JVI.01427-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Sen N, Sommer M, Che X, White K, Ruyechan WT, Arvin AM. Varicella-zoster virus immediate-early protein 62 blocks interferon regulatory factor 3 (irf3) phosphorylation at key serine residues: A novel mechanism of irf3 inhibition among herpesviruses. J Virol. 2010;84(18):9240–9253. doi: 10.1128/JVI.01147-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Pfaller CK, Conzelmann K-K. Measles virus v protein is a decoy substrate for ikappab kinase alpha and prevents toll-like receptor 7/9-mediated interferon induction. J Virol. 2008;82(24):12365–12373. doi: 10.1128/JVI.01321-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *74.Ning S, Pagano JS. The a20 deubiquitinase activity negatively regulates lmp1 activation of irf7. J Virol. 2010;84(12):6130–6138. doi: 10.1128/JVI.00364-10. A link between A20 upregulation and inhibition of IRF7 activity is made. The authors suggest that lmp1 upregulates A20 during viral latency and may directly deubiquitinate IRF7 to inhibit its activity and prevent antiviral responses.
  • 75.Prins KC, Cárdenas WB, Basler CF. Ebola virus protein vp35 impairs the function of interferon regulatory factor-activating kinases ikkepsilon and tbk-1. J Virol. 2009;83(7):3069–3077. doi: 10.1128/JVI.01875-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Otsuka M, Kato N, Moriyama M, Taniguchi H, Wang Y, Dharel N, Kawabe T, Omata M. Interaction between the hcv ns3 protein and the host tbk1 protein leads to inhibition of cellular antiviral responses. Hepatology. 2005;41(5):1004–1012. doi: 10.1002/hep.20666. [DOI] [PubMed] [Google Scholar]
  • 77.Foy E, Li K, Sumpter R, Loo Y-M, Johnson CL, Wang C, Fish PM, Yoneyama M, Fujita T, Lemon SM, Gale M. Control of antiviral defenses through hepatitis c virus disruption of retinoic acid-inducible gene-i signaling. Proc Natl Acad Sci USA. 2005;102(8):2986–2991. doi: 10.1073/pnas.0408707102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **78.Unterholzner L, Sumner RP, Baran M, Ren H, Mansur DS, Bourke NM, Randow F, Smith GL, Bowie AG. Vaccinia virus protein c6 is a virulence factor that binds tbk-1 adaptor proteins and inhibits activation of irf3 and irf7. PLoS Pathog. 2011;7(9):e1002247. doi: 10.1371/journal.ppat.1002247. The C6 protein of Vaccinia virus is shown to interact with members of the TBK1 signaling scaffold and is suggested to inhibit IRF3 and IRF7 activation at this level. Mutant viruses lacking the C6L gene were attenuated in vivo.
  • 79.Kagan JC, Su T, Horng T, Chow A, Akira S, Medzhitov R. Tram couples endocytosis of toll-like receptor 4 to the induction of interferon-beta. Nat Immunol. 2008;9(4):361–368. doi: 10.1038/ni1569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Blander JM, Medzhitov R. Regulation of phagosome maturation by signals from toll-like receptors. Science. 2004;304(5673):1014–1018. doi: 10.1126/science.1096158. [DOI] [PubMed] [Google Scholar]
  • 81.Arpaia N, Godec J, Lau L, Sivick KE, McLaughlin LM, Jones MB, Dracheva T, Peterson SN, Monack DM, Barton GM. Tlr signaling is required for salmonella typhimurium virulence. Cell. 2011;144(5):675–688. doi: 10.1016/j.cell.2011.01.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Blander JM, Medzhitov R. Toll-dependent selection of microbial antigens for presentation by dendritic cells. Nature. 2006;440(7085):808–812. doi: 10.1038/nature04596. [DOI] [PubMed] [Google Scholar]
  • 83.Trombetta ES, Ebersold M, Garrett W, Pypaert M, Mellman I. Activation of lysosomal function during dendritic cell maturation. Science. 2003;299(5611):1400–1403. doi: 10.1126/science.1080106. [DOI] [PubMed] [Google Scholar]
  • 84.Underhill DM, Ozinsky A. Phagocytosis of microbes: Complexity in action. Annu Rev Immunol. 2002;20:825–852. doi: 10.1146/annurev.immunol.20.103001.114744. [DOI] [PubMed] [Google Scholar]
  • 85.Lambeth JD. Nox enzymes and the biology of reactive oxygen. Nat Rev Immunol. 2004;4(3):181–189. doi: 10.1038/nri1312. [DOI] [PubMed] [Google Scholar]
  • 86.West AP, Brodsky IE, Rahner C, Woo DK, Erdjument-Bromage H, Tempst P, Walsh MC, Choi Y, Shadel GS, Ghosh S. Tlr signalling augments macrophage bactericidal activity through mitochondrial ros. Nature. 2011;472(7344):476–480. doi: 10.1038/nature09973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Katunuma N, Matsunaga Y, Saibara T. Mechanism and regulation of antigen processing by cathepsin b. Adv Enzyme Regul. 1994;34:145–158. doi: 10.1016/0065-2571(94)90014-0. [DOI] [PubMed] [Google Scholar]
  • 88.Fonteneau JF, Kavanagh DG, Lirvall M, Sanders C, Cover TL, Bhardwaj N, Larsson M. Characterization of the mhc class i cross-presentation pathway for cell-associated antigens by human dendritic cells. Blood. 2003;102(13):4448–4455. doi: 10.1182/blood-2003-06-1801. [DOI] [PubMed] [Google Scholar]
  • 89.Shen L, Sigal LJ, Boes M, Rock KL. Important role of cathepsin s in generating peptides for tap-independent mhc class i crosspresentation in vivo. Immunity. 2004;21(2):155–165. doi: 10.1016/j.immuni.2004.07.004. [DOI] [PubMed] [Google Scholar]
  • 90.Biron CA, Byron KS, Sullivan JL. Severe herpesvirus infections in an adolescent without natural killer cells. N Engl J Med. 1989;320(26):1731–1735. doi: 10.1056/NEJM198906293202605. [DOI] [PubMed] [Google Scholar]
  • 91.Biron CA, Brossay L. Nk cells and nkt cells in innate defense against viral infections. Curr Opin Immunol. 2001;13(4):458–464. doi: 10.1016/s0952-7915(00)00241-7. [DOI] [PubMed] [Google Scholar]

RESOURCES