Skip to main content
Journal of Orthopaedic Translation logoLink to Journal of Orthopaedic Translation
. 2017 Jun 7;10:68–85. doi: 10.1016/j.jot.2017.05.004

Danger of frustrated sensors: Role of Toll-like receptors and NOD-like receptors in aseptic and septic inflammations around total hip replacements

Michiaki Takagi a,, Yuya Takakubo a, Jukka Pajarinen b, Yasushi Naganuma a, Hiroharu Oki a, Masahiro Maruyama a,b, Stuart B Goodman b
PMCID: PMC5676564  NIHMSID: NIHMS884057  PMID: 29130033

Summary

The innate immune sensors, Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), can recognize not only exogenous pathogen-associated molecular patterns (PAMPs), but also endogenous molecules created upon tissue injury, sterile inflammation, and degeneration. Endogenous ligands are called damage-associated molecular patterns (DAMPs), and include endogenous molecules released from activated and necrotic cells as well as damaged extracellular matrix. TLRs and NLRs can interact with various ligands derived from PAMPs and DAMPs, leading to activation and/or modulation of intracellular signalling pathways. Intensive research on the innate immune sensors, TLRs and NLRs, has brought new insights into the pathogenesis of not only various infectious and rheumatic diseases, but also aseptic foreign body granuloma and septic inflammation of failed total hip replacements (THRs). In this review, recent knowledge is summarized on the innate immune system, including TLRs and NLRs and their danger signals, with special reference to their possible role in the adverse local host response to THRs.

Translational potential of this article: A clear understanding of the roles of Toll-like receptors and NOD-like receptors in aseptic and septic loosening of joint replacements will facilitate potential strategies to mitigate these events, thereby extending the longevity of implants in humans.

Keywords: innate immune sensors, NLR, TLR, total hip replacement

Introduction

Aseptic loosening and deep infection are two major reasons for reoperation after primary total hip replacement (THR) [1]. Both “aseptic” and “septic” loosening are conditions in which a loose THR implant is accompanied by unfavourable inflammatory immune reactions in periprosthetic connective tissue. Aseptic loosening is considered to be a local foreign body reaction to implant wear debris, in which monocytes/macrophages recognize and phagocytose debris derived from the implant. The septic condition around THRs is attributed to interplay of the host and microbial attacks, and is referred to as a periprosthetic joint infection, in which monocytes/macrophages and neutrophils both play an important role in combating the bacterial infection. Monocytes/macrophages and neutrophils are the major cell types responsible for the innate host defence. These two immune responses drive and define the pathologic state. Various mediators have been identified and linked to the physiologic and pathologic roles of aseptic and/or septic inflammation around THR implants. However, the precise mechanisms of aseptic loosening and the septic condition have not been elucidated yet. In addition, despite “aseptic” and “septic” inflammatory conditions being defined in THRs, the mechanisms, similarities, and differences of these two conditions are still poorly understood. Continued research on the molecular and cellular mechanisms underlying these complex inflammatory processes will undoubtedly contribute to better longevity of patients with THRs.

The human immune system consists of both innate and adaptive immunity. Innate immunity is found in both animals and plants, and serves to protect the host from foreign intruders. The adaptive immune system is found only in vertebrates, and developed at a later evolutionary stage than innate immunity. In vertebrates, the innate and adaptive immunities work together in the host defence against pathogens. The innate immune response is induced not only by microbial attack, but also by sterile tissue injury and degeneration with no invading pathogen present. This paradox was first explained by Matzinger [2] in 1994 (the Matzinger hypothesis). She proposed that the immune system was activated by both external and internal molecules signalling dangers, rather than by recognition of self versus nonself. Sensing receptors, termed “pattern recognition receptors” (PRRs), have since been identified as important components of this theory. Mammalian cells can sense danger signals from both pathogens and damaged tissues via PRRs of the innate immune system, which evolved earlier than the highly diverse receptors of the adaptive immune system in vertebrates. Subsequent analyses of innate immune receptors have led to the identification and classification of PRRs into five families: Toll-like receptors (TLRs), retinoid acid inducible gene-I like receptors (RLRs), nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), C-type lectins (CTLs) and absent-in-melanoma-like receptors. These receptor families can respond to exogenous molecules and endogenous danger signals [3], [4]. (Abbreviations of the molecules and related structures used in this review are summarized alphabetically in Appendix 1.)

TLRs are located on the cell-surface and on endosomes within the cell. They can sense exogenous “pathogen-associated molecular patterns” (PAMPs) [5] and endogenous “damage-associated molecular patterns” (DAMPs), also known as alarmins [6], [7]. RLRs are cytosolic sensors for nucleic acids and are largely responsible for the recognition of cytosolic, virus-derived RNA, subsequently inducing the production of interferons. RLRs can sense RNA species. CTLs, located on the cell membrane, recognize carbohydrate-based ligands and are important receptors in the detection and response to different infections, e.g., fungi and tubercle bacillus. CTLs can function, like TLRs located on the cell membrane, to scan the extracellular milieu for PAMPs. Human immunodeficiency virus exploits CTLs for cell entry. NLRs are cytoplasmic sensors and can promote the inflammatory response via signal transduction and/or inflammasome formation. NLRs also function in the process of autophagy [4], [8], [9], [10], [11], [12], [13], [14]. Moreover, possible mechanisms of collaboration and/or cross-talk among these different innate immune sensors have been suggested [15].

Recent research on TLRs and NLRs has brought new insights into not only the pathogenesis of infectious and rheumatic diseases, but also the adverse local host response around THRs, especially aseptic and septic conditions. TLRs and NLRs are particularly known to be involved in the local host response around THRs [16], [17], [18], [19]. This review summarizes recent knowledge and examines the innate immune response of TLRs and NLRs, with special reference to aseptic loosening due to foreign body granuloma and septic condition by microbial attack in THRs.

Toll-like receptors

Toll receptor was originally identified in Drosophila melanogaster as a receptor essential for the establishment of the dorso-ventral pattern in developing embryos [20]. In 1996, Lemaitre et al. [21] demonstrated that Toll-mutant flies were highly susceptible to fungal infections. A mammalian toll homologue was first reported in 1997 and named TLR [22]. TLRs belong to the PRR family, and are the main innate immune sensors, together with RLRs and NLRs. Thus far, 13 distinct mammalian TLRs have been identified, 10 of which are functional in humans (TLR1–10), and 12 in mice (TLR1–9 and TLR11–13) [23], [24], [25]. TLR1, TLR2, TLR5, TLR6, TLR10, and TLR11 are located on the cell surface. TLR3, TLR7, TLR8, and TLR9 are located in the endosome. TLR4 is located both on the cell surface and in the endosome. The high levels and broadest spectra of TLR expression have been observed in a variety of cell types in the immune system, including monocytes/macrophages [16], [26], [27], [28], [29], [30] and neutrophils [17], [31].

TLRs can recognize a wide variety of PAMPs derived from bacteria, fungi, parasites, and viruses. They act as homo- or heterodimers of type I transmembrane glycoproteins, each comprising a ligand-binding ectodomain containing 19–25 tandem leucine-rich repeat (LRR) motifs, a single transmembrane helix, and a cytoplasmic Toll/interleukin (IL)-1 receptor (TIR) domain, which is required for downstream signalling. Combinations of TLRs and corresponding PAMP ligands include the TLR1–2 heterodimer with triacyl lipopeptides, the TLR2–6 heterodimer with diacyl lipopeptides, the TLR3 homodimer and dsRNA, the TLR4 homodimer and lipopolysaccharide (LPS), the TLR5 homodimer and flagellin, the TLR7 and TLR8 homodimers and ssRNA, the TLR9 homodimer and CpGDNA and the TLR11 homodimer and protozoan profilin-like protein. These receptor/ligand pairs are summarized in Table 1, Table 2 and Figure 1 [23], [24], [32], [33], [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], [60], [61], [62], [63], [64], [65]. TLR10 has an inhibitory effect and is predominantly involved in the modulation of the TLR2-driven activation of immune cells [66].

Table 1.

Exogenous TLR ligands derived from organisms.

Location/receptor Major ligands Origin of the ligand Refs
Plasma membrane
 TLR 1 triacyl lipopeptide bacteria & mycobacteria [32]
soluble factors Neisseria meningiditis [33]
 TLR 2 lipoprotein/lipopeptides various pathogens [34]
peptidoglycan gram-positive bacteria [35]
[36]
lipoteichoic acid gram-positive bacteria [36]
lipoarabinomannan mycobacteria [37]
phenol-soluble modulin Staphylococcus epidermis [38]
glycoinositolphospholipid Trypanosoma cruzi [39]
glycolipid Treponema maltophilum [40]
porins Neisseria [41]
atypical LPS Leptospira interrogans [42]
atypical LPS Porphyromonas gingivalis [43]
zymosan Fungi [44]
 TLR 5 flagellin bacteria [52]
 TLR 6 diacylpolypeptide mycoplasma [53]
lipoteichoic acid gram-positive bacteria [46]
 TLR 10 Profilin-like molecule ND [57], [66]
Plasma membrane/endosomal membrane
 TLR 4 LPS gram-negative bacteria [46]
Taxol plants [47]
fusion protein respiratory syncytial virus [48]
envelope protein mouse mammary-tumour virus [49]
Hsp70 (exogenous) Chlamydia pneumoniae [50], [51]
Endosomal membrane
 TLR 3 dsRNA viruses [45]
 TLR 7 ssRNA viruses [54], [55]
 TLR 8 ssRNA viruses [54]
 TLR 9 CpG DNA bacteria & viruses [56]
 TLR 11 ND uropathogenic bacteria [58]
Profilin like molecule Toxoplasma Gondi [59], [63]
 TLR 12 Profilin like molecule ND [63]
 TLR 13 bacteria 23S rRNA ND [63]

ND = not determined. see Appendix 1 for list of all other abbreviations. Note. Adapted and modified with permission from copyright holder [3], [63].

Table 2.

Endogenous TLR ligands of host origin and synthetic analogues.

Location TLRs Ligands
Signal adaptor
Major DAMPs Synthetic analogue
Plasma membrane
TLR1–2 ND triacyl lipopeptides MAL/MyD88
TLR2–6 heat-shock proteins diacyl lipopetides HMGB1, versican, hyaluronic acid MAL/MyD88
TLR5 ND discontinuous 13-amino acid peptide CBLC502 MyD88
TLR10 ND ND MyD88
Plasma membrane/Endosomal membrane
TLR4 heat-shock proteins, HMGB1, β-defensin-3, extra domain A of a fibronectin, hyaluronic acid, heparin sulfate, fibrinogen, surfactant-protein A, oxidized phospholipids lipid A mimetics (monophosphoryl lipid A minoalkyl glucosamine 4-phosphate), E620, E5531, E5564 MAL/
MyD88
TRAM/
TRIF
Endosomal membrane
TLR3 mRNA poly(I:C). poly(I; C12U) TRIF
TLR7 ssRNA (immune complex) oligonucleotides, imidazoquinoline, guanosine nucleotides, bropirimine MyD88
TLR8 ssRNA (immune complex) imidazoquinolines (resiquimod) MyD88
TLR9 chromatin IgG complex CpG oligo-deoxynucleotide MyD88
TLR11 ND ND MyD88
TLR12 ND ND MyD88
TLR13 ND ND MyD88

ND = not determined; see Appendix 1 for list of all other abbreviations.

Note. Adapted with permission from copyright holder [3], [65].

Figure 1.

Figure 1

Representative signalling pathway of human TLRs in the cell membrane (TLR1/2, TLR2/6, TLR4, TLR5, and TLR10) and endosome (TLR3, TLR4, TLR7, and TLR9). TLRs signal through two major adaptors, MyD88 and TRIF, which bind directly or indirectly to TLRs through TIR-domain interactions. MyD88 and Mal promote the recruitment of TRAF6 (ubiquitin ligase) and several kinase complexes, such as IRAKs, TAB/TAK, IKKαβ, MAKKs, and/or PI3K/Akt. Subsequent phosphorylation and ubiquitination promote the translocation of the transcription factors CREB, AP-1, and NF-κB, which can induce transcription of certain messenger RNAs, such as TNFs, pro-IL-1β, and other inflammatory molecules. By contrast, TRIF interacts with TRAF3 and TBK1/IKKε in endosomal TLR4 signalling, and with TRAF3 in TLR3 signaling, followed by IRF3 activation. In TLR7 and TLR9 signaling, TRIF interacts with IRAKs and TRAF3, followed by IRF7 activation. Activated and phosphorylated IRF3 and IRF7 in the cytoplasm are translocated into the nucleus, which induces subsequent induction of type I IFNs [63], [64], [65]. See Appendix 1 for list of abbreviations.

Adapted and modified with permission from copyright holder [63].

TLRs can sense not only exogenous PAMPs, but also DAMPs of endogenous origin. Intracellular molecules become DAMPs as they are released into the extracellular milieu by activated and necrotic cells. Similarly, molecules released from the damaged extracellular matrix due to tissue injury, sterile inflammation, or degradation can function as DAMPs. Definite, probable, or possible endogenous TLR DAMP ligands have been reported and can be categorized as proteins/peptides, fatty acids/lipoproteins, proteoglycans/glycosaminoglycans, and nucleic acids/protein-nucleic acids complex [3], [10] (Table 2). Proteins and peptides that activate the TLRs include: β-defensin-3 for TLR1; HSP60, HSP70, gp96, HMGB1, HMGB1-nucleosome complex, β-defensin-3, surfactant proteins, eosinophil-derived neurotoxin, and antiphospholipid antibodies for TLR2; HMGB1, fibronectin EDA, fibrinogen, tenascin C, surfactant proteins, β-defensin-3, HSP60, HSP70, HSP72, HSP22(B8), gp96, S100A8 (Mrp8), S100A9 (Mrp14), neutrophil elastase, antiphospholipid antibodies, and lactoferrin for TLR4 and antiphospholipid antibodies for TLR7 and TLR8. Fatty acids and lipoproteins that activate the TLRs include: serum amyloid A for TLR2, and serum amyloid A, oxidized LDL and saturated fatty acid for TLR4. Proteoglycans and glycosaminoglycans that activate the TLRs include: biglycan, versican and hyaluronic acid fragments for TLR2, and biglycan, heparan sulfate fragments, and hyaluronic acid fragments for TLR4. Nucleic acid and protein-nucleic acid ligands of the TLRs include dsRNA for TLR3, ssRNA for TLR7 and TLR8, and IgG-chromatin complexes for TLR9. Thus, TLRs can interact with both PAMPs and DAMPs, and represent a key molecular link among microbial infection, tissue injury, sterile inflammation, and degeneration. Crystal structure analyses proposed possible diverse modes of exogenous ligand recognition by TLRs, involving TLR homodimerization and heterodimerization, as well as direct TLR-ligand interactions or interactions with coreceptors and accessory molecules [65], [66], [67], [68], [69], [70], [71], [72] (Table 2).

Coreceptors and accessory molecules are known to assist PAMP and/or DAMP recognition by some TLRs [69], [71], e.g., MD-2, CD14, Mrp8, RP105, and PAR-2 for the TLR4 homodimer, and CD14, CD36, and dectin for the TLR1–TLR2 and TLR2–TLR6 [72], [73], [74]. MD-2 and CD14 can work to assist DAMP recognition, either independently or together [10]. In addition, P2X4/P2X7 have been shown to activate TLR2 and TLR4 by biglycan ligand stimulation in macrophages [75]. HMGB1 mediates activation of plasmacytoid dendritic cells and B cells through TLR9 by DNA-containing immune complexes with immunoglobulin superfamily member receptor for advanced glycation end products (RAGE) [76]. CD44, together with MD-2, can assist in the recognition of hyaluronic acid by TLR4 [77].

The initial role of TLRs is to function as sensors for danger signals of PAMPs, and to bind DAMPs to initiate innate host responses [21], [22]. The binding of a TLR ligand to the receptor triggers conformational rearrangement of the cytoplasmic TIR domains and recruitment of specific adaptors via homotype TIR-TIR interactions. The main adaptor molecules include MyD88, TRIF (also known as TICAM-1), TIRAP (also known as Mal), and TRAM (also known as TICAM-2) [78], [79]. MyD88 and TRIF are the main adaptors of TLRs. Most TLRs use MyD88. TLR4 uses both MyD88 and TRIF, and TLR3 uses only TRIF. In spite of direct interaction of the intracellular portion of TLRs with MyD88 or TRIF, TLR1, TLR2, TLR4, and TLR6, which are located in the plasma membrane, require additional adaptors for downstream signalling, e.g., TIRAP for MyD88 and TRAM for TRIF. The interaction of adaptor molecules is followed by the recruitment of kinases and ubiquitin ligases, leading to activation and nuclear translocation of several transcription factors, such as NF-κB, AP1, CREB, and IRF3/7, which differ between cell types in the innate immune system [8], [69], [80] (Figure 1).

Activation of these transcription factors via TLR signalling leads to upregulation of proinflammatory cytokines, chemokines, and chemokine receptors, such as TNF-α [81], IL-1β [82], IL-6 [81], [82], [83], IFN-α/β/γ [81], [83], [84], [85], CC (or CCβ) chemokines [83], [85], [86], and CXC (or CXCα) chemokines [83], [85]. TLR-ligand interactions also upregulate costimulatory molecules, such as intercellular adhesion molecule-1 [82] and lymphocyte function-associated antigen-1 and -3 [84]. Costimulatory molecules are essential for the induction of pathogen-specific adaptive immune responses [87]. Thus, TLRs link innate host responses to adaptive immunity [88], [89]. Synthetic analogues can also stimulate TLR signalling (Table 2).

TLRs are important not only for the identification of infectious agents, but also for various other types of diseases. In inflammatory rheumatoid synovium, increased expression of TLR2, TLR3–5, TLR7, and TLR9 was demonstrated [90], [91]. In systemic lupus erythematosus, the possible involvement of TLRs at onset was proposed [92], [93]. Pathologic involvement of TLR signalling has been suggested in asthma [94], psoriasis [95], inflammatory bowel disease [96], Alzheimer's disease [97], multiple sclerosis [98], and atherosclerosis [99]. Thus, TLRs have emerged as attractive therapeutic targets, and several biologic and synthetic antagonists to modulate their function have been developed, e.g., for TLR2, OPN-305 (humanized antibody) for delayed graft function; for TLR4, SPA4 (peptide) for lung inflammation, E5564 (small molecules) for sepsis, TAK-242 (small molecule) for septic shock and NI-0101 (antibody) for rheumatoid arthritis; for TLR7-9, IMO-3100 (DNA-based small molecule) for psoriasis; and for TLR7-9: ODN antagonist (oligonucleotides) for inflammation, IMO-880 (DNA-based small molecules) for dermatomyositis and psoriasis and CpG-ODN c41 (oligonucleotide) for inflammation [65], [100], [101], [102].

NOD-like receptors

The concept of pathogen recognition and the host defence system in plants was intensively studied in the early 2000s [103], [104]. In plants, resistance to pathogens is mediated by a group of disease resistance genes that elicit the hypersensitivity response. This response includes physical isolation of infected lesions through remodelling of the cell wall structure, production of reactive oxygen species (ROS), cell death, and production of antipathogen molecules at the site of infection. Some resistance gene products are transmembrane proteins with ectodomains containing LRRs, such as mammalian TLRs [104]. Another major class of resistance gene products is cytosolic NOD proteins, which typically contain amino-terminal α-helix-rich or TIR domains, a central NOD, and carboxyl-terminal LRRs [104]. These receptors were also identified in mammalian cells, and their role in host-microbial interactions and inflammatory diseases have been comprehensively summarized [105], [106].

NLRs are cytoplasmic receptors and can recognize PAMPs and DAMPs. Evolutionary and domain structure analyses of the NLRs have clarified four subfamilies: NLRA, NLRB, NLRC, and NLRP [12], [107]. Recent findings regarding each subfamily of gene, function and activator is summarized in Figure 2 and Table 3 [108], [109], [110], [111], [112], [113], [114], [115], [116], [117], [118], [119], [120], [121], [122], [123], [124], [125], [126], [127], [128], [129], [130], [131], [132], [133], [134], [135], [136], [137], [138], [139], [140], [141], [142], [143], [144]. NLRs share a common domain, a central NOD domain, and carboxyl-terminal LRRs. The NOD domain in NLRs is also known as the NACHT domain, which has been named after the NAIP, CIITA, HET-E, and TP1 proteins. The NACHT domain consists of seven distinct conserved motifs, including the ATP/GTPase specific loop, the Mg2+-binding site, and five more specific motifs, including a transactivation domain (TAD), caspase recruitment and activation domain (CARD), baculovirus IAP repeat (BIR), pyrin domain (PYD) and an undefined region (Figure 2) [4], [107], [161], [162].

Figure 2.

Figure 2

The NLR subfamily. There are four NLR: NLRA, NLRB, NLRC, and NLRP. In their protein structures, the NACHT, CARD, LRR, BIR, TAD, PYD, and undefined regions are major components [4], [12], [107]. See Appendix 1 for list of abbreviations.

Adapted and modified with permission of copyright holder [4].

Table 3.

Gene, function and the activators of NLRs.

Subfamily Gene Function Activator Refs
NLRA CIITA MHC class II expression IFNγ [108]
NLRB Naip control of legionella pneumophila infection flagellin [109]
NLRC NOD1 NF-κB DAP [110], [111], [112]
NOD2 NF-κB, autophagy, type 1 IFN production MDP [113], [114]
NLRC3 negative regulator of T cell activation, response to LPS & TRAF6 unknown [115], [116]
NLRC5 inflammatory response & MHC class I upregulation IFNγ, IFNβ [117], [118], [119]
NLRC4 caspase 1 activation, cell death, phagosome maturation rod proteins, flagellin [120], [121], [122], [123]
NLRX1 ROS production & antiviral response unknown [124], [125], [126], [127]
NLRP NLRP1 response to anthrax bacteria lethal toxin, MDP [128], [129]
NLRP10 dendritic cell migration unknown [130], [131]
NLRPs 2–9, 11–14
NLRP2 embryonic development, caspase 1 activation, TRAF6 suppression unknown [132], [133], [134]
NLRP3 caspase 1 activation ATP, K+ efflux, alum, asbestos, silica, ROS, fungi, MSU, HA, amyloid, nanoparticle [135], [136], [137], [138], [139], [140], [142], [143], [144], [145], [146]
NLRP4 embryogenesis, suppression of type 1 IFN, autophagy, & TRAF6 unknown [147], [148], [149], [150]
NLRP5 embryogenesis unknown [151]
NLRP6 NF-κB inhibition & caspase 1 activation unknown [152], [153]
NLRP7 embryonic development, caspase 1 activation lipopetides [154], [155], [156]
NLRP12 inhibition of noncanonical NF-κB, ERK, & AKT pathway, activation caspase 1 Yersinia [157], [158], [159]
NLRP14 spermatogenesis [160]

See Appendix 1 for list of abbreviations.

Motta et al. [4] put forward a functional concept of the NLR family, in which the roles of NLRs were divided into “Signalsome” (signal transduction), “Enhansosome” (transcriptional activation), “Autophagosome” (autophagy), and “Inflammasome” (inflammasome assembly). For example, in signal transduction, NOD1 and NOD2 are known to activate the NF-κB pathway via Rip2 [163]. Nuclear translocation of NF-κB induces cytokine production. The peptidoglycans diaminopimelic acid and muramyl dipeptide (MDP) are activators for NOD1 [111], [112] and NOD2 [113], respectively. This pathway is different from TLR signalling mediated by MyD88 and TRAF6 [164]. To the contrary, the NALP subfamily members NLRC3 (NLR family CARD domain containing 3) [116], NLRP2 (NLR family pyrin domain containing 2) [134], and NLRP4 [147] negatively regulate this pathway by the suppression of TRAF6 (Figure 3). In response to IFN-γ stimulation, nuclear translocation of CIITA (class II, major histocompatibility complex, transactivator) [108] and NLRC5 [117], [118], [165] induces MHC class II and MHC class I upregulations, respectively. Nuclear translocation of NLRC5 is followed by dimer formation. IFN-β stimulation can upregulate the inflammatory response via NLRC5. Autophagy, a bulk cytoplasmic degradation system, aids in xenography, which plays an important role of bacterial clearance. NOD2 can recognize bacteria at the cell entry site and recruit autophagy-related protein 16L1 (ATG 16L1) to the plasma membrane. Autophagosome formation around intracellular bacteria is initiated and followed by the exclusion of bacteria [114], [166], [167], [168]. Inflammasomes can be assembled by NLRP3, NLRP1, and NLRC4 and are triggered by various cell stressors: NLPP3 by ATP, K+ efflux, aluminium, asbestos, silica, ROS, potassium efflux, fungus, uric acid, hyaluronic acid, amyloid, and nanoparticles; NLRP1 by anthrax lethal toxin and MDP and NLRC4 by flagellin and rod proteins (T3SS and T4SS; injectisome, found in several gram negative bacteria). These are summarized in Figure 2, Figure 3. Upon activation, NLRs form multimeric protein complexes including ASC and procaspase. Procaspase is maturated into caspase-1, which induces activation of pro-IL-1β into IL-1β and of proIL-18 into IL-18 (Figure 3). Thus, inflammasome assembly leads to an inflammatory response, and abnormalities of the genes involved are related to specific diseases [169], [170]. Mutation in CIITA is responsible for bare lymphocyte syndrome, in which the immune system is severely compromised and cannot effectively fight infection [171]. Gene deletion in Naip was found in spinal muscular atrophy [172]. Asthma [173] and inflammatory bowel disease [174] have been related to NOD1 mutations, and Crohn's disease [175] and Blau syndrome [176] to NOD2 mutations. NALP1 mutations have been associated with vitiligo [128], Addison's disease [177] and type-1 diabetes mellitus [177]. NLRP12 mutations were also found in one type of hereditary periodic fever syndrome [178]. In addition, many genetic mutations encoding NLRPs have been reported, including Beckwith-Wiedemann Syndrome [179], neonatal-onset multisystem inflammatory disease (also called CINCA syndrome) [180], [181], familial cold autoinflammatory syndrome [182], and Muckle-Wells syndrome [182]. Genetic mutations in NLRP3 and inflammasome-related molecules have also been found in human autoinflammatory diseases, hereditary periodic fever syndromes, familial Mediterranean fever, hyperimmunoglobulinaemia D and periodic fever syndrome and necrosis factor receptor superfamily 1A-associated periodic syndrome [183], [184]. Gene mutations in the NLRs and their related molecules can affect the onset of diseases and syndromes more distinctly compared to the TLRs. In addition, NLRs are involved in the process of ontogeny in mammals [185], [186]. The NLR family of intracellular sensors has emerged not only as a crucial component of the innate immune response, but also a key regulator of life phenomena.

Figure 3.

Figure 3

NLRs play four major roles as intracellular sensors: signal transduction via NF-κB, enhancement of immune and inflammatory cytokine induction, autophagosome-related antigen presentation and/or bacterial clearance and inflammasome formation leading to inflammatory reactions induced by activated IL-1β and IL-18, mediated by caspase-1 [4]. See Appendix 1 for list of all other abbreviations.

Adapted and modified with permission from copyright holder [4].

As previously described for TLRs, the molecular mechanisms responsible for innate immunity have become possible therapeutic targets in various diseases. However, when compared to clinical trials examining the therapeutic use of several TLR-related molecules [65], [100], [101], [102], those related to NLRs have not yet progressed at the clinical trial level. This is probably due to more complicated interactions of various ligands with NLRs, as well as at least four different roles of the intracellular receptors (signal transduction, transcriptional activation, autophagy, and inflammasome assembly) which potentially interact with each other in a complicated fashion. In addition, TLR and NLR interaction has become a great concern [4], [15], [187]. TLRs can promote the induction of autophagy and promote the delivery of infecting pathogens to the lysosome. Induction of autophagy and interplay between autophagy and NOD1/NOD2, which are associated with ATG 16L1, has been demonstrated [168]. Several NLRs, including NLRP3 and NLRC4, have been shown to mediate caspase-1-dependent cell death of infected cells, which is termed pyroptosis. TLRs and NLRs coordinately influence the outcome of infections by controlling the fate of host cells [187]. However, the precise mechanism of the interactions between the two innate sensors remains unclear.

Frustrated TLRs and NLRs in the local host response to THR implants

Expression of TLRs in aseptic periprosthetic connective tissue was first reported in 2007. CD68-positive monocytes/macrophages in the synovial membrane showed marked immunoreactivity of TLR4 and TLR9, which was more intense than that found in osteoarthritic synovial tissue [16]. Enhanced immunoreactivity of TLR1, TLR2, TLR5, and TLR6 in CD68-positive monocytes/macrophages was also observed [17], [18], [19]. Upregulation of various TLRs, including TLR1, TLR2, TLR6, and TLR9, in aseptic interface tissue was also demonstrated using quantitative reverse transcription polymerization reaction [188]. Subsequently, the expression of TLR4 was found to correlate with several proinflammatory and chemotactic mediators in peri-implant tissue, suggesting a role of TLR4 in the peri-implant inflammatory process [189]. Similarly, increased expression of TLRs was observed in an animal model of wear particle disease [190]. In the septic synovial tissue of THR patients, enhanced immunoreactivity of TLR1, TLR2, TLR4, TLR5, and TLR9, was seen in CD15-positive polymorphonuclear neutrophils and CD68-positive monocytes/macrophages [17], [191] (Figure 4). These findings indicate that both aseptic and septic periprosthetic tissues around THRs are well equipped with TLRs, which can respond promptly to microbial stimuli (PAMPs) and DAMPs induced in the process of tissue damage, degradation, and remodelling. The cell types responsible for TLR signalling are mainly monocytes/macrophages in aseptic periprosthetic tissue and neutrophils and monocytes/macrophages in septic inflammation. Increased expression of NLRs and related molecules was also observed both in aseptic and septic periprosthetic tissues around THRs. NLRP3, caspase-1, ASC, and IL-1β, were found both in aseptic and septic inflammatory periprosthetic tissues [19], [191] (Figure 4). Prior to immunohistochemical analysis of the retrieved tissue samples, secretion of IL-1β from macrophages after titanium particle exposure was demonstrated. IL-1β secretion was dependent on inflammasome activation, namely, NALP3, ASC, and caspase-1 [192]. In addition, involvement of NLRP3 in polymethylmethacrylate particle-induced inflammation and osteolysis was demonstrated in both in vitro macrophage culture and an in vivo mouse calvarial model [193]. This suggests the potential role of the inflammasome in the pathogenesis of periprosthetic osteolysis.

Figure 4.

Figure 4

Immunohistochemical localization of (A) TLR1, TLR2, and TLR6 and (B) NLRP3, Caspase-1, and ASC in aseptic foreign body granuloma and septic synovial membrane in failed THRs. The main immune-reactive cells were monocytes/macrophages in aseptic tissues, and monocytes/macrophages and neutrophils in septic tissues. Retrieved tissue samples were embedded in OCT compound, followed by thin-cut and fixation with formaldehyde. Avidin-biotin peroxidase complex method was applied as previously described [19]. See Appendix 1 for list of abbreviations.

Taken together, these studies indicate that the innate immune sensors, TLRs and NLRP3, are possibly involved in the pathogenesis of “aseptic” and “septic” inflammation around THR implants. In the “septic” condition, monocytes/macrophages and neutrophils utilize TLRs, and at least NLRP3, to combat microbes. Microbial components, PAMPs, can stimulate these sensors; thus, the inflammatory reaction is evoked and enhanced. In the process of inflammation, these sensors can also possibly react with DAMPs from damaged cellular components and the various mediators produced and/or released in the response to pathogen-induced inflammation. For example, proteins/peptides, fatty acids/lipoproteins, proteoglycans/glycosaminoglycans, and nucleic acids/protein-nucleic acids complexes found in infectious sites are candidates for DAMPs as TLR-ligands in septic inflammation around THRs, as described previously [3], [17]. Potential ligands that activate NLRP3 include ATP, ROS, and uric acid released from the damaged cells.

The question remains as to how the sensors behave in the “aseptic” condition. Immunohistochemical analyses confirm that monocytes/macrophages involved in foreign body granulomas are equipped with TLRs, NLRP3, and associated molecules. Monocytes/macrophages work as scavengers of the innate immune system to eliminate unknown foreign wear particles. However, except for subclinical infections, there does not appear to be a PAMP ligand to stimulate the sensors in loco. One possible explanation for the function of the sensors abundantly found in aseptic foreign body granulomas and osteolysis is the induction and/or enhancement of inflammatory reactions by direct contact and/or phagocytosis of wear debris from implants. Maitra et al. [194] demonstrated that polymeric alkaline structures, with side chain oxidation, directly bind and activate the TLR1–TLR2 signalling pathway and initiate phagocytosis of the particle, inducing enlargement, fusion and disruption of endosomal components. This leads to NALP3 inflammasome activation and ROS release by cathepsins [194], [195]. Hydroxyapatite and cobalt-alloy particles and/or cobalt ions also enhance proinflammatory cytokine production via TLR4. Several previous studies have suggested that direct contact and/or phagocytosis of the debris itself evokes innate immune responses via the sensors [196], [197], [198]. Indeed, it has recently been demonstrated that, in contrast to ions, cobalt particles induce inflammation and osteolysis mainly by activating NLRP3 rather than TLR4 [199]. Similarly, endotoxin-free titanium particles were reported to be insufficient to induce and enhance proinflammatory cytokines via TLR signalling pathways. Removal of LPS (a ligand for TLR4) contamination from titanium particles almost completely inhibited the production of TNF-α, IL-1β, and IL-6 at the protein level, and did not affect osteoclastic differentiation in vitro. In addition, the removal of endotoxin from the particles reduced wear-driven osteolysis by 50–70% in vivo [200]. LPS-free and lipoteichoic acid (LTA) (ligand for TLR2)-free titanium particles were also shown not to elevate TNF-α, IL-1β, and IL-6 mRNA levels in macrophages stimulated by these particles in vitro [19], [30]. Additional coating of particles by LPS and LTA in vitro reversed this phenomenon. Thus, these in vitro studies may support the hypothesis that PAMPs, either present on the implant surface [201] or accumulated in the effective joint space via circulation [202], can adhere to wear particles and stimulate the innate immune sensors, and induce and/or enhance the osteolytic inflammatory reaction [203]. This hypothesis is further supported by the observations from genetic association studies that polymorphisms in innate immune sensors and relevant signalling molecules can alter the risk for periprosthetic osteolysis [204].

However, the discrepancy in the findings between titanium wear particles and those made of other materials, including polymers, cobalt-alloy, and hydroxyapatite, seems unusual. Rigorous treatment of wear particles to remove residual endotoxins by strong acid (nitric acid) and strong alkali (sodium hydroxide) may alter the surface chemistry of the wear particles, including their hydrophobicity, oxidation, ionization, and/or electric charge, and result in an impaired cellular response. This might account for the fact that removal of endotoxins reduced particle-induced in vivo osteolysis by 50–70%, but not completely [200]. The surfaces of the endotoxin-free particles in vivo are exposed to cells, extracellular matrix, and fluid after particle implantation. This can allow the adherence of various molecules of host-origin to the particles. These substances may include DAMPs derived from cells, extracellular matrices, and fluid content from exudate of serum and/or extracellular space, which are induced as a result of the stimulus and invasive particle implantation. Adherence of DAMPs to particles potentially evokes TLR-related inflammatory responses, although the reaction is probably weaker compared to PAMP adherence. This phenomenon could be explained an increase in TNF-α and IL-1β mRNA, even in the absence of LPS and LTA coatings [30], [190]. Cellular debris, containing DAMPs, produced during cell culture can potentially adhere to particles. The particles and/or cellular DAMPs themselves can stimulate innate immune sensors in vitro. Alternatively, particle implantation in vivo can evoke foreign body reactions apart from their effects on TLRs or other innate immune sensors. These effects might also be due to misrecognition relating to phagocytosis, such as the interaction between the Fc receptor of immune cells and nonspecific immunoglobulin attached on the particle surfaces, or between scavenger receptors and denatured cholesterol and/or allied substances of serum origin after invasive processes of particle implantation and the ensuing tissue response. Serum amyloid A, complement components, hyaluronic acid, fibronectin, and other endogenous molecules in the serum might act as additional ligands covering the particles.

Foreign body granuloma formation and periprosthetic osteolysis occur gradually over several years, and often over more than 10 years in aseptic loosening of THRs. Pathologic extracellular connective tissue remodelling by proteinases and cell cycling (recruit, proliferation, apoptosis, and/or necrosis) continues, but proceeds slowly in response to the phagocytosis of wear particles. This process can also provide DAMPs in loco, which stimulate TLRs and NLRs, but is probably regulated by a self-protection mechanism, as the addition of particles to monocytes/macrophages in vitro precipitated downregulation of TLR mRNA [16], [30], and, in spite of the upregulation of IL-1β mRNA, the release at protein levels was not high [19]. Eventual downregulation of TLRs after particle stimulation was also observed in a mouse model [205]. This phenomenon seems compatible with the in loco status around the aseptic loosening of THRs, where inflammatory markers, e.g., C-reactive protein, are within normal limits, along with the absence of redness and local heat driven by inflammasomes and active IL-1β. Down- and/or upregulation of the TLR- and NLR-mediated response to wear particles probably reflects the proactive behaviour of effector cells in vitro, but appears to be more complex in vivo. Further analyses on the effect of DAMPs from tissue debris, as well as PAMPs in vitro and in vivo, are required. We have to keep in mind that in vitro experiments and in vivo animal models only provide short time points when compared to the extended timeline of the aseptic loosening of THRs in humans.

The boundary between “aseptic” and “septic” conditions still seems unclear. For example, clinically unapparent microbial attack, which does not show the expected tissue response with active neutrophil invasion, and the presence of biofilms, probably makes the situation more complex to decipher. Multifactorial interactions among the genetic variations, including deletions and polymorphism in immune cells, types of foreign substances, including PAMPs and biomaterial related-wear, and DAMPs produced in the process of tissue response, need to be precisely taken into account to explain the individually different local tissue and systemic reactions, and require more study [206]. In addition, the problem of adverse local host response to metals and various types of tissue reaction, often overlapping perivascular lymphocytic infiltration and/or lymphocyte--dominant reaction, foreign body granuloma, tissue necrosis, fibrosis, or infection, is now raising additional questions on the role of innate immune sensors [207], [208].

In the basic research field of TLRs and NLRs, intensive and broad analyses are ongoing. Although the precise mechanism of TLRs, NLRs, and their interaction is not yet completely elucidated, current knowledge can certainly provide hints to clarify the pathology of unfavourable host reactions around THRs. In addition, the clinical approach of modifying TLR and NLR signalling using synthetic agents and/or by identification of individual differences at genetic and epigenetic levels probably improves the pathologic status of aseptic and septic conditions around THRs, leading to upgrading of therapeutic and prophylactic levels. Further intensive research not only on the frustrated immune sensors, TLRs and NLRs, but also on the global interaction of the innate and adaptive immune system in the local host response are required. These studies provide relevant information on more accurate patient-specific implant selection, and therapeutic and/or prophylactic approaches for unfavourable host responses around THRs in the future.

ND = not determined. see Appendix 1 for list of all other abbreviations. Note. From “Toll-like receptor--A potent driving force behind rheumatoid arthritis,” by M. Takagi, 2011, J Clin Exp Hematop, 51, p. 77–92. Copyright 201X, Name of Copyright Holder. From “The history of Toll-like receptors – redefining innate immunity,” by L.A. O'Neill, D. Golenbock, A.G. Bowie, Nat Rev Immunol, 2013, 13, p. 453–60. Copyright 201X, Name of Copyright Holder. Modified with permission.

Conflict of interest

There is no conflict as for the review article.

Funding/Support

We acknowledge support from the Grant-in-Aid for Scientific Research, the Ministry of Science and Culture (Basic Research C-24592252), Yamagata Health Support Society for the Promotion of Science 2015–2017 (H26-10, H27-13, H28-8, H29-4), Japan, and National Institutes of Health (2R01AR055650 and 1R01AR063717).

Acknowledgments

We greatly thank Istuho Tomuro for skilful illustration and Eiko Saito for pathologic analyses.

Appendix 1. List of abbreviations for molecules and related structures.

Akt protein kinase B
AP-1 activating protein-1
ATG16L1 autophagy-related protein 16
BIR baculovirus inhibitor of apoptosis protein(IAP) repeat
CARD caspase activation and recruitment domain
CC C-C motif
CD cluster of differentiation
CIITA MHC class II transcription activator
CpG DNA DNA containing unmethylated CpG motifs
CTL C-type lectin
CREB cyclic-AMP response element-binding protein
CXC CXC-motif
DAI DNA-dependant activator of IFN-regulatory factor
DAMP damage-associated molecular pattern
ds double-stranded
EDA extradomain A
ERK extracellular signal-regulated kinase
FcγR fragment crystallisable gamma receptor
gp96 glycoprotein 96
HET-E heterocaryon incompatibility locus E protein from Podspora anserina
HMGB high mobility group box
Hsp heat shock protein
IAP inhibitor of apoptosis protein
IFN interferon
IL interleukin
IL-1ra IL-1 receptor antagonist
IRAK IL-1 receptor-associated kinase
IRF interferon regulatory factor
IKK inhibitor κB kinase
IκB inhibitor kappa B
JNK c-Jun N-terminal kinase
LDL low-density lipoprotein cholesterol
LPS lipopolysaccharide
LR laminin receptor
LRR leucine-rich repeat
MAD5 melanoma differentiation-associated gene 5
Mal MyD88-adapter-like
MAPK mitogen-activated protein kinase
MD-1, 2 myeloid differentiation factor-1, 2
MDP muramyldipeptide
MHC major histocompatibility complex
MKK MAPK kinase
MSK mitogen- and stress-activated kinase
MSU monosodium urate
MyD88 myeloid differentiation primary response protein 88
NACHT NAIP, CIITA, HET-E and TP1
NAIP neuronal apoptosis inhibitor protein
NALP NACHT, leucine-rich repeat, and pyrin-domain-containing protein
NF-IL6 nuclear factor interleukin-6
NF-κB nuclear factor-kappa B
NLR NOD-like receptor
NOD nucleotide-binding oligomerization domain
NLRC3-5 NLR family CARD domain-containing 3-5
NLRP NLR with PYD-containing protein
PAMP Pathogen-associated molecular pattern
PAR-2 proteinase-activated G-protein-coupled receptor -2
PI3K phosphatidylinositol 3-kinase
PRR pattern recognition receptor
PYD pyrin domain
RAGE receptor for advanced glycation end-products
RIP receptor-interacting protein
RLR retinoic acid-inducible gene 1-like receptor
RP105 radioprotective 105
ss single-stranded
T3SS/T4SS type III/IV secretion system
TAD transactivation domain
TANK TRAF family associated NF-kB activator
TBK TANK-binding kinase
TIR Toll/IL-1 receptor
TICAM-1,2 TIR-containing adaptor molecule-1, 2
TIRAP Toll/interleukin receptor domain-containing adaptor protein
TLR Toll-like receptor
TMED transmembrane emp24 domain-containing protein
TNF tumour necrosis factor
TP1 telomerase-associated protein 1
TRAF TNF receptor-associated factor
TRAM TRIF-related adaptor molecule
TRIF TIR-domain-containing adapter-inducing IFN

References

  • 1.Garellick G., Kärrholm J., Lindahl H., Malchau H., Rogmark C., Lolfson O. Reoperation. In: Garellick G., Kärrholm J., Lindahl H., Malchau H., Rogmark C., Rolson O., editors. The Swedish hip arthroplasty register, annual report. Göran Garellick; Göteborg: 2014. pp. 51–56. [Google Scholar]
  • 2.Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol. 1994;12:991–1045. doi: 10.1146/annurev.iy.12.040194.005015. [DOI] [PubMed] [Google Scholar]
  • 3.Takagi M. Toll-like receptor–A potent driving force behind rheumatoid arthritis. J Clin Exp Hematop. 2011;51:77–92. doi: 10.3960/jslrt.51.77. [DOI] [PubMed] [Google Scholar]
  • 4.Motta V., Soares F., Sun T., Philpott D.J. NOD-like receptors: Versatile cytosolic sentinels. Physiol Rev. 2015;95:149–178. doi: 10.1152/physrev.00009.2014. [DOI] [PubMed] [Google Scholar]
  • 5.Akira S., Hemmi H. Recognition of pathogen-associated molecular patterns by TLR family. Immunol Lett. 2003;85:85–95. doi: 10.1016/s0165-2478(02)00228-6. [DOI] [PubMed] [Google Scholar]
  • 6.Rifkin I.R., Leadbetter E.A., Busconi L., Viglianti G., Marshak-Rothstein A. Toll-like receptors, endogenous ligands, and systemic autoimmune disease. Immunol Rev. 2005;204:27–42. doi: 10.1111/j.0105-2896.2005.00239.x. [DOI] [PubMed] [Google Scholar]
  • 7.Bianchi M.E. DAMPs, PAMPs and alarmins: All we need to know about danger. J Leukoc Biol. 2007;81:1–5. doi: 10.1189/jlb.0306164. [DOI] [PubMed] [Google Scholar]
  • 8.Baccala R., Gonzalez-Quintial R., Lawson B.R., Stern M.E., Kono D.H., Buetler B. Sensors of the innate immune system: Their mode of action. Nat Rev Rheumatol. 2009;5:448–456. doi: 10.1038/nrrheum.2009.136. [DOI] [PubMed] [Google Scholar]
  • 9.Theofilopoulos A.N., Gonzalez-Quintal R., Lawson B.R., Koh Y.T., Stem M.E., Kono D.H. Sensors of the innate immune system; their link to rheumatic diseases. Nature Rev Rheumatol. 2010;6:146–156. doi: 10.1038/nrrheum.2009.278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Piccinini A.M., Midwood K.S. DAMPening inflammation by modulating TLR signaling. Mediators Inflamm. 2010;2010:672395. doi: 10.1155/2010/672395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kastner D.L., Aksentijevich I., Goldbach-Mansky R. Autoinflammatory disease reloaded: A clinical perspective. Cell. 2010;140:784–790. doi: 10.1016/j.cell.2010.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Schroder K., Tschopp J. The inflammasomes. Cell. 2010;140:821–832. doi: 10.1016/j.cell.2010.01.040. [DOI] [PubMed] [Google Scholar]
  • 13.Vanaja S., Rathinam V.K., Fitzerald K.A. Mechanisms of inflammasome activation: Recent advances and novel insight. Trends Cell Biol. 2015;25:308–315. doi: 10.1016/j.tcb.2014.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Hansen J.D., Vojtech L.N., Laing K.J. Sensing disease and danger: A survey of vertebrate PRRs and their origins. Dev Comp Immunol. 2011;35:886–897. doi: 10.1016/j.dci.2011.01.008. [DOI] [PubMed] [Google Scholar]
  • 15.Joosten L.A., Netea M.G., Mylona E., Koenders M.I., Malireddi R.K., Oosting M. Engagement of fatty acids with Toll-like receptor 2 drives interleukin 1β production via the ASC/caspase 1 pathway in monosodium urate monohydrate crystal-induced gouty arthritis. Arthritis Rheum. 2010;62:3237–3248. doi: 10.1002/art.27667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Takagi M., Tamaki Y., Hasegawa H., Takakubo Y., Konttinen L., Tiainen V.M. Toll-like receptors in the interface membrane around loosening total hip replacement implants. J Biomed Mater Res A. 2007;81:1017–1026. doi: 10.1002/jbm.a.31235. [DOI] [PubMed] [Google Scholar]
  • 17.Tamaki Y., Takakubo Y., Goto K., Hirayama T., Sasaki K., Konttinen Y.T. Increased expression of toll-like receptors in aseptic loose periprosthetic tissues and septic synovial membranes around total hip implants. J Rheumatol. 2009;36:598–608. doi: 10.3899/jrheum.080390. [DOI] [PubMed] [Google Scholar]
  • 18.Pajarinen J., Cenni E., Savarino L., Gomez-Barrena E., Tamaki Y., Takagi M. Profile of toll-like receptor-positive cells in septic and aseptic loosening of total hip arthroplasty implants. J Biomed Mater Res A. 2010;94:84–92. doi: 10.1002/jbm.a.32674. [DOI] [PubMed] [Google Scholar]
  • 19.Naganuma Y., Takakubo Y., Hirayama T., Tamaki Y., Pajarinen J., Sasaki K. Lipoteichoic acid modulates inflammatory response in macrophages after phagocytosis of titanium particles through Toll-like receptor 2 cascade and inflammasomes. J Biomed Mater Res A. 2016;104:435–444. doi: 10.1002/jbm.a.35581. [DOI] [PubMed] [Google Scholar]
  • 20.Hashimoto C., Hudson K.L., Anderson K.V. The Toll gene of Drosophila, required for dorsal-ventral embryonic polarity, appears to encode a transmembrane protein. Cell. 1988;52:269–279. doi: 10.1016/0092-8674(88)90516-8. [DOI] [PubMed] [Google Scholar]
  • 21.Lemaitre B., Nicolas E., Michaut L., Reichhart J.M., Hoffman J.A. The dorsoventral regulatory gene cassette spatzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell. 1996;86:973–983. doi: 10.1016/s0092-8674(00)80172-5. [DOI] [PubMed] [Google Scholar]
  • 22.Medzhitov R., Preston-Hurlburt P., Janeway C.A., Jr. A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature. 1997;388:394–397. doi: 10.1038/41131. [DOI] [PubMed] [Google Scholar]
  • 23.Uematsu S., Akira S. Toll-like receptors and innate immunity. J Mol Med (Berl) 2006;84:712–725. doi: 10.1007/s00109-006-0084-y. [DOI] [PubMed] [Google Scholar]
  • 24.Beutler B., Jiang Z., Georgel P., Crozat K., Croker B., Rutschmann S. Genetic analysis of host resistance. Toll-like receptor signaling and immunity at large. Annu Rev Immunol. 2006;24:353–389. doi: 10.1146/annurev.immunol.24.021605.090552. [DOI] [PubMed] [Google Scholar]
  • 25.Kasamatsu J., Oshiumi H., Matsumoto M., Kasahara M., Seya T. Phylogenetic and expression analysis of lamprey toll-like receptors. Dev Comp Immunol. 2010;34:855–865. doi: 10.1016/j.dci.2010.03.004. [DOI] [PubMed] [Google Scholar]
  • 26.Thoma-Uszynski S., Kiertscher S.M., Ochoa M.T., Bouis D.A., Norgard M.V., Miyake K. Activation of Toll-like receptor 2 on human dendritic cells triggers induction of IL-12, but not IL-10. J Immunol. 2000;165:3804–3810. doi: 10.4049/jimmunol.165.7.3804. [DOI] [PubMed] [Google Scholar]
  • 27.Visintin A., Mazzoni A., Spitzer J.H., Wyllie D.H., Dower S.K., Segal D.M. Regulation of Toll-like receptors in human monocytes and dendritic cells. J Immunol. 2001;166:249–255. doi: 10.4049/jimmunol.166.1.249. [DOI] [PubMed] [Google Scholar]
  • 28.Kadowaki N., Ho S., Antonenko S., Malefyt R.W., Kastelein R.A., Bazan F. Subsets of human dendritic cell precursors express different toll-like receptors and respond to different microbial antigens. J Exp Med. 2001;194:863–869. doi: 10.1084/jem.194.6.863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hornung V., Rothenfusser S., Britsch S., Krung A., Jahrsdörfer B., Giese T. Quantitative expression of toll-like receptor 1-10 mRNA in cellular subsets of human peripheral blood mononuclear cells and sensitivity to CpG oligodeoxynucleotides. J Immunol. 2002;168:4531–4537. doi: 10.4049/jimmunol.168.9.4531. [DOI] [PubMed] [Google Scholar]
  • 30.Hirayama T., Tamaki Y., Takakubo Y., Iwazaki K., Sasaki K., Ogino T. Toll-like receptors and their adaptors are regulated in macrophages after phagocytosis of lipopolysaccharide-coated titanium particles. J Orthop Res. 2011;29:984–992. doi: 10.1002/jor.21369. [DOI] [PubMed] [Google Scholar]
  • 31.Muzio M., Bosisio D., Polentarutti N., D'amico G., Stoppacciaro A., Mancinelli R. Differential expression and regulation of Toll-like receptor (TLR) in human leukocytes: Selective expression of TLR3 in dendritic cells. J Immunol. 2000;164:5998–6004. doi: 10.4049/jimmunol.164.11.5998. [DOI] [PubMed] [Google Scholar]
  • 32.Takeuchi O., Sato S., Horiuchi T., Hoshino K., Takeda K., Dong Z. Cutting edge: Role of Toll-like receptor 1 in mediating immune response to microbial lipoproteins. J Immunol. 2002;169:10–14. doi: 10.4049/jimmunol.169.1.10. [DOI] [PubMed] [Google Scholar]
  • 33.Wyllie D.H., Kiss-Toth E., Visintin A., Smith S.C., Boussouf S., Segal D.M. Evidence for an accessory protein for Toll-like receptor 1 in anti-bacterial responses. J Immunol. 2000;165:7125–7132. doi: 10.4049/jimmunol.165.12.7125. [DOI] [PubMed] [Google Scholar]
  • 34.Aliprantis A.O., Yang R.B., Mark M.R., Suggett S., Devaux B., Radolf J.D. Cell activation and apoptosis by bacterial lipoproteins through Toll-like receptor 2. Science. 1999;285:736–739. doi: 10.1126/science.285.5428.736. [DOI] [PubMed] [Google Scholar]
  • 35.Takeuchi O., Hoshino K., Kawai T., Sanjo H., Takada H., Ogawa T. Differential role of TLR2 and TLR4 in recognition of gram-positive and gram-negative cell wall components. Immunity. 1999;11:443–451. doi: 10.1016/s1074-7613(00)80119-3. [DOI] [PubMed] [Google Scholar]
  • 36.Schwadner R., Dziarski R., Wesche H., Rothe M., Kirschning C.J. Peptidoglycan- and lipoteichoic acid-induced cell activation is mediated by Toll-like receptor 2. J Biol Chem. 1999;274:17406–17409. doi: 10.1074/jbc.274.25.17406. [DOI] [PubMed] [Google Scholar]
  • 37.Mears T.K., Wang S., Lien E., Yoshimura A., Golenbock D.T., Fenton M.J. Human Toll-like receptors mediate cellular activation by Mycobacterium tuberculosis. J Immunol. 1999;163:3920–3927. [PubMed] [Google Scholar]
  • 38.Hajjar A.M., O'Mahony D.S., Ozinsky A., Underhill D.M., Aderem A., Klebanoff S.J. Cutting edge: Functional interactions between Toll-like receptor (TLR) 2 and TLR1 or TLR6 in response to phenol-soluble modulin. J Immunol. 2001;166:15–19. doi: 10.4049/jimmunol.166.1.15. [DOI] [PubMed] [Google Scholar]
  • 39.Coelho P.S., Klein A., Talvani A., Coutinho S.F., Takeuchi O., Akira S. Glycosylphosphatidylinositol-anchored mucin-like glycoproteins isolated from Trypanosoma cruzi trypomastigotes induce in vivo leukocyte recruitment dependent on MCP-1 production by IFN-γ-primed-macrophages. J Leukoc Biol. 2002;71:837–844. [PubMed] [Google Scholar]
  • 40.Opitz B., Schröder N.W., Speitzer I., Michelese K.S., Kirschning C.J., Hallatschek W. Toll-like receptor 2 mediates Tryponema glycolipid and lipoteichoic acid-induced NF-κB translation. J Biol Chem. 2001;276:22041–22047. doi: 10.1074/jbc.M010481200. [DOI] [PubMed] [Google Scholar]
  • 41.Massari P., Henneke P., Ho Y., Latz E., Golenbock D.T., Wetzler L.M. Cutting edge: immune stimulation by Neisseria porins is Toll-like receptor 2 and MyD88 dependent. J Immunol. 2002;168:1533–1537. doi: 10.4049/jimmunol.168.4.1533. [DOI] [PubMed] [Google Scholar]
  • 42.Werts C., Tapping R.I., Mathison J.C., Chuang T.H., Kravchenko V., Saint Girons I. Leptospiral lipopolysaccharide activates cells through a TLR2-dependent mechanism. Nature Immunol. 2001;2:346–352. doi: 10.1038/86354. [DOI] [PubMed] [Google Scholar]
  • 43.Hirschfeld M., Weis J.J., Toschakov V., Salkowski C.A., Cody M.J., Ward D.C. Signaling by Toll-like receptor 2 and 4 agonist results in differential gene expression in murine macrophages. Infect Immun. 2001;69:1477–1482. doi: 10.1128/IAI.69.3.1477-1482.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Underhill D.M., Ozinsky A., Hajjar A.M., Stevens A., Wilson A., Bassetti M. The Toll-like receptor 2 is recruited to macrophage phagosomes and discriminates between pathogens. Nature. 1999;401:811–815. doi: 10.1038/44605. [DOI] [PubMed] [Google Scholar]
  • 45.Alexopoulou L., Holt A.C., Medzhitov R., Flavell R.A. Recognition of double-stranded RNA and activation of NF-κB by Toll-like receptor 3. Nature. 2001;413:732–738. doi: 10.1038/35099560. [DOI] [PubMed] [Google Scholar]
  • 46.Poltorak A., He X., Smirnova I., Liu M.Y., van Huffel C., Du X. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science. 1998;282:2085–2088. doi: 10.1126/science.282.5396.2085. [DOI] [PubMed] [Google Scholar]
  • 47.Kawasaki K., Akashi S., Shimazu R., Yoshida T., Miyake K., Nishijima M. Mouse Toll-like receptor 4.MD-2 complex mediates lipopolysaccharide-mimetic signal transduction by Taxol. J Biol Chem. 2000;275:2251–2254. doi: 10.1074/jbc.275.4.2251. [DOI] [PubMed] [Google Scholar]
  • 48.Kurt-Jones E., Popova L., Kwinn L., Haynes L.M., Jones L.P., Tripp R.A. Pattern recognition receptors TLR4 and CD14 mediate response to respiratory syncytial virus. Nature Immunol. 2000;1:398–401. doi: 10.1038/80833. [DOI] [PubMed] [Google Scholar]
  • 49.Rassa J.C., Meyers J.L., Zhang Y., Kudaravalli R., Ross S. Murine retroviruses activate B cells via interaction with Toll-like receptor 4. Proc Natl Acad Sci USA. 2002;99:2281–2286. doi: 10.1073/pnas.042355399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Bulut Y., Faure E., Thomas L., Karahashi H., Michelesen K.S., Equils O. Chlamydia heat shock protein 60 activates macrophages and endothelial cells through Toll-like receptor-4 and MD2 in MyD88-dependent pathway. J Immunol. 2002;168:1435–1440. doi: 10.4049/jimmunol.168.3.1435. [DOI] [PubMed] [Google Scholar]
  • 51.Ohashi K., Burkart V., Flohé S., Kolb H. Cutting edge: Heat shock protein 60 is a putative endogenous ligand of the toll-like receptor-4 complex. J Immunol. 2000;164:558–561. doi: 10.4049/jimmunol.164.2.558. [DOI] [PubMed] [Google Scholar]
  • 52.Hayashi F., Smith K.D., Ozinsky A., Hawn T.R., Yi E.C., Goodlett D.R. The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature. 2001;410:1099–1103. doi: 10.1038/35074106. [DOI] [PubMed] [Google Scholar]
  • 53.Takaeuchi O., Kawai T., Mühlradt P.F., Morr M., Radolf J.D., Zychlinsky A. Discrimination of bacterial lipoproteins by Toll-like receptor 6. Int Immunol. 2001;13:933–940. doi: 10.1093/intimm/13.7.933. [DOI] [PubMed] [Google Scholar]
  • 54.Heil F., Hemmi H., Hochrein H., Ampenberger F., Kirschning C., Akira S. Species-specific recognition of single-stranded RNA via Toll-like receptor 7 and 8. Science. 2004;303:1526–1529. doi: 10.1126/science.1093620. [DOI] [PubMed] [Google Scholar]
  • 55.Diebold S.S., 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;303:1529–1531. doi: 10.1126/science.1093616. [DOI] [PubMed] [Google Scholar]
  • 56.Hemmi H., Takeuchi O., Kawai T., Kaisho T., Sato S., Sanjo H. Toll-like receptor recognizes bacterial DNA. Nature. 2000;408:740–745. doi: 10.1038/35047123. [DOI] [PubMed] [Google Scholar]
  • 57.Govindaraj R.G., Manavalan B., Lee G., Choi S. Molecular modeling-based evaluation of hTLR10 and identification of potential ligands in Toll-like receptor signaling. PLoS One. 2010;5:e12713. doi: 10.1371/journal.pone.0012713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Zhang D., Zhang G., Hayden M.S., Greenblatt M.B., Bussey C., Flavell R.A. Toll-like receptor that prevents infection by uropathogenic bacteria. Science. 2004;303:1522–1526. doi: 10.1126/science.1094351. [DOI] [PubMed] [Google Scholar]
  • 59.Yarovinsky F., Zhang D., Anderson J.F., Bannenberg G.L., Serhan C.N., Hayden M.S. TRL11 activation of dendritic cells by a protozoan profilin-like protein. Science. 2005;308:1626–1629. doi: 10.1126/science.1109893. [DOI] [PubMed] [Google Scholar]
  • 60.Aderem A., Ulevitch R.J. Toll-like receptors in the induction of the innate immune response. Nature. 2000;406:782–787. doi: 10.1038/35021228. [DOI] [PubMed] [Google Scholar]
  • 61.Akira S., Takeda K., Kaisho T. Toll-like receptors: critical proteins linking innate and acquired immunity. Nat Immunol. 2001;2:675–680. doi: 10.1038/90609. [DOI] [PubMed] [Google Scholar]
  • 62.Watters T.M., Kenny E.F., O'Neil L.A. Structure, function and regulation of the Toll/IL-1 receptor adaptor proteins. Immunol Cell Biol. 2007;85:411–419. doi: 10.1038/sj.icb.7100095. [DOI] [PubMed] [Google Scholar]
  • 63.O'Neill L.A., Golenbock D., Bowie A.G. The history of Toll-like receptors – redefining innate immunity. Nat Rev Immunol. 2013;13:453–460. doi: 10.1038/nri3446. [DOI] [PubMed] [Google Scholar]
  • 64.Joosten L.B., Abdollahi-Roodsaz S., Dinarello C.A., O'Neill L., Netea M.G. Toll-like receptors and chronic inflammation in rheumatic diseases: new developments. Nat Rev Rheumatol. 2016;12:344–357. doi: 10.1038/nrrheum.2016.61. [DOI] [PubMed] [Google Scholar]
  • 65.Achek A., Yesudhas D., Choi S. Toll-like receptors: promising therapeutic targets for inflammatory disease. Arch Pharm Res. 2016;39:1032–1049. doi: 10.1007/s12272-016-0806-9. [DOI] [PubMed] [Google Scholar]
  • 66.Oosting M., Cheng S.C., Bolscher J.M., Vestering-Stenger R., Platinga T.S., Verschueren I.C. Human TLR10 is an anti-inflammatory pattern-recognition receptor. Proc Natl Acad Sci USA. 2014;111:E4478–E4484. doi: 10.1073/pnas.1410293111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Huyton T., Rossjohn J., Wilce M. Toll-like receptors: Structural pieces of curved shaped puzzle. Immunol Cell Biol. 2007;85:406–410. doi: 10.1038/sj.icb.7100089. [DOI] [PubMed] [Google Scholar]
  • 68.Jin M.S., Lee J.O. Structures of the Toll-like receptor family and its ligand complexes. Immunity. 2008;29:182–191. doi: 10.1016/j.immuni.2008.07.007. [DOI] [PubMed] [Google Scholar]
  • 69.Miyake K. Roles for accessory molecules in microbial recognition by Toll-like receptors. J Endotoxin Res. 2006;12:195–204. doi: 10.1179/096805106X118807. [DOI] [PubMed] [Google Scholar]
  • 70.Jin M.S., Kim S.E., Heo J.Y., Lee M.E., Kim H.M., Paik S.G. Crystal structure of the TLR1-TLR2 heterodimer induced by binding of a tri-acylated lipopeptide. Cell. 2007;130:1071–1082. doi: 10.1016/j.cell.2007.09.008. [DOI] [PubMed] [Google Scholar]
  • 71.Park B.S., Song D.H., Kim B.S., Choi B.S., Lee H., Lee J.O. The structural basis of lipopolysaccharide recognition by the TLR4-MD2 complex. Nature. 2009;458:1191–1195. doi: 10.1038/nature07830. [DOI] [PubMed] [Google Scholar]
  • 72.Nagai Y., Kobayashi T., Motoi Y., Ishiguro K., Akashi S., Saitoh S. The radioactive 105/MD-1 complex links TLR2 and TLR4/MD-2 in antibody response to microbial membranes. J Immunol. 2005;174:7043–7049. doi: 10.4049/jimmunol.174.11.7043. [DOI] [PubMed] [Google Scholar]
  • 73.Vogl T., Tenbrock K., Ludwig S., Leukert N., Ehrhardt C., van Zoelen M.A. Mrp8 and Mrp14 are endogenous activators of Toll-like receptor 4, promoting lethal, endotoxin-induced shock. Nat Med. 2007;13:1042–1049. doi: 10.1038/nm1638. [DOI] [PubMed] [Google Scholar]
  • 74.Rallabhandi P., Nhu Q.M., Toschakov V.Y., Piao W., Medvedev A.E., Hollenberg M.D. Analysis of proteinase-activated receptor 2 and TLR4 signal transduction: A novel paradigm for receptor cooperativity. J Biol Chem. 2008;283:24314–24325. doi: 10.1074/jbc.M804800200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Babelova A., Moreth K., Tsalastra-Greul W., Zeng-Brouwers J., Eickelberg O., Young M.F. Biglycan, a danger signal that activates the NLRP3 inflammasome via toll-like and P2X receptors. J Biol Chem. 2009;284:24035–24048. doi: 10.1074/jbc.M109.014266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Tian J., Avalos A.M., Mao S.Y., Chen B., Senthil K., Wu H. Toll-like receptor 9-dependent activation by DNA-containing immune complexes is mediated by HMBG1 and RAGE. Nat Immunol. 2007;8:487–496. doi: 10.1038/ni1457. [DOI] [PubMed] [Google Scholar]
  • 77.Taylor K.R., Yamasaki K., Radek K.A., Di Nardo A., Goodarzi H., Golenbock D. Recognition of hyaluronan released in sterile injury involves a unique receptor complex dependent on Toll-like receptor 4, CD44 and MD-2. J Biol Chem. 2007;282:18265–18275. doi: 10.1074/jbc.M606352200. [DOI] [PubMed] [Google Scholar]
  • 78.O'Neill L.A., Fitzgerald K.A., Bowie A.G. The Toll-IL-1 receptor adaptor family grows to five members. Trends Immunol. 2003;24:286–290. doi: 10.1016/s1471-4906(03)00115-7. [DOI] [PubMed] [Google Scholar]
  • 79.Liew F.Y., Xu D., Brint E.K., O'Neill L.A. Negative regulation of Toll-like receptor-mediated immune responses. Nat Rev Immunol. 2005;5:446–458. doi: 10.1038/nri1630. [DOI] [PubMed] [Google Scholar]
  • 80.Kagan J.C., Su T., Hong T., Chow A., Akira S., Medzhitov R. TRAM couples endocytosis of Toll-like receptor 4 to the induction of interferon-beta. Nat Immunol. 2008;9:361–368. doi: 10.1038/ni1569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Radstake T.R., Roelofs M.F., Jenniskens Y.M., Oppers-Walgreen B., van Riel P.L., Barrera P. Expression of toll-like receptors 2 and 4 in rheumatoid synovial tissue and regulation by proinflammatory cytokines interleukin-12 and interleukin-18 via interferon-gamma. Arthritis Rheum. 2004;50:3856–3865. doi: 10.1002/art.20678. [DOI] [PubMed] [Google Scholar]
  • 82.Kyburz D., Rethage J., Seibl R., Lauener R., Gay R.E., Carson D.A. Bacterial peptidoglycans but not CpG oligodeoxynucleotides activate synovial fibroblasts by toll-like receptor signaling. Arthritis Rheum. 2003;48:642–650. doi: 10.1002/art.10848. [DOI] [PubMed] [Google Scholar]
  • 83.Brentano F., Schorr O., Gay R.E., Gay S., Kyburz D. RNA released from necrotic synovial fluid cells activates rheumatoid arthritis synovial fibroblasts via Toll-like receptor 3. Arthritis Rheum. 2005;52:2656–2665. doi: 10.1002/art.21273. [DOI] [PubMed] [Google Scholar]
  • 84.Hertz C.J., Kiertscher S.M., Godowski P.J., Bouis D.A., Norgard M.V., Roth M.D. Microbial lipopeptides stimulate dendritic cell maturation via Toll-like receptor 2. J Immunol. 2001;166:2444–2450. doi: 10.4049/jimmunol.166.4.2444. [DOI] [PubMed] [Google Scholar]
  • 85.Pierer M., Rethage J., Seibl R., Lauener R., Brentano F., Wagner U. Chemokine secretion of rheumatoid arthritis synovial fibroblasts stimulated by Toll-like receptor 2 ligands. J Immunol. 2004;172:1256–1265. doi: 10.4049/jimmunol.172.2.1256. [DOI] [PubMed] [Google Scholar]
  • 86.Schiavoni G., Mattei F., Sestili P., Borghi P., Venditti M., Morse H.C., 3rd ICSBP is essential for the development of mouse type I interferon-producing cells and for the generation and activation of CD8alpha(+) dendritic cells. J Exp Med. 2002;196:1415–1425. doi: 10.1084/jem.20021263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Iwasaki A., Medzhitov R. Toll-like receptor control of the adaptive immune responses. Nat Immunol. 2004;5:987–995. doi: 10.1038/ni1112. [DOI] [PubMed] [Google Scholar]
  • 88.Takeuchi O., Akira S. Toll-like receptors; their physiological role and signal transduction system. Int Immunopharmacol. 2001;1:625–635. doi: 10.1016/s1567-5769(01)00010-8. [DOI] [PubMed] [Google Scholar]
  • 89.Takeda K., Akira S. Roles of Toll-like receptors in innate immune responses. Genes Cells. 2001;6:733–742. doi: 10.1046/j.1365-2443.2001.00458.x. [DOI] [PubMed] [Google Scholar]
  • 90.Iwahashi M., Yamamura M., Aita T., Okamoto A., Ueno A., Ogawa N. Expression of toll-like receptor 2 on CD16+ blood monocytes and synovial tissue macrophages in rheumatoid arthritis. Arthritis Rheum. 2004;50:1457–1467. doi: 10.1002/art.20219. [DOI] [PubMed] [Google Scholar]
  • 91.Tamaki Y., Takakubo Y., Hirayama T., Konttinen Y.T., Goodman S.B., Yamakawa M. Expression of Toll-like receptor and their signaling pathway in rheumatoid arthritis. J Rheumatol. 2011;38:810–820. doi: 10.3899/jrheum.100732. [DOI] [PubMed] [Google Scholar]
  • 92.Boule M.W., Broughton C., Mackay F., Akira S., Marshak-Rothstein A., Rifkin I.R. Toll-like receptor 9-dependent and -independent dendritic cell activation by chromatin-immunoglobulin G complexes. J Exp Med. 2004;199:1631–1640. doi: 10.1084/jem.20031942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Clancy R.M., Markham A.J., Buyon J.P. Endosomal Toll-like receptors in clinically overt and silent autoimmunity. Immunol Rev. 2016;269:76–84. doi: 10.1111/imr.12383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Blease K., Kunkel S.L., Hogaboam C.M. IL-18 modulate chronic fungal asthma in murine mouse model; putative involvement of Toll-like receptor 2. Inflamm Res. 2001;50:552–560. doi: 10.1007/PL00000233. [DOI] [PubMed] [Google Scholar]
  • 95.Baker B.S., Ovigne J.M., Powles A.V., Cororan S., Fry L. Normal keratinocyte express Toll-like receptors (TLRs) 1, 2 and 5: modulation of expression in chronic plaque psoriasis. Br J Dermatol. 2003;148:670–679. doi: 10.1046/j.1365-2133.2003.05287.x. [DOI] [PubMed] [Google Scholar]
  • 96.Franchimont D., Vermeire S., El Housni H., Pierik M., Van Steen K., Gustot T. Deficient host-bacteria interaction in inflammatory bowel disease? The toll-like receptor (TLR)-4 Asp299gly polymorphism is associated with Crohn's disease and ulcerative colitis. Gut. 2004;53:987–992. doi: 10.1136/gut.2003.030205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Jackson A.C., Rossiter J.P., Lafon M. Expression of Toll-like receptor 3 in the human cerebellar cortex in rabies, herpes simplex encephalitis, and other neurological diseases. J Neurovirol. 2006;12:229–234. doi: 10.1080/13550280600848399. [DOI] [PubMed] [Google Scholar]
  • 98.Reindl M., Lutterotti A., Ingman J., Schanda K., Gassner C., Deisenhammer F. Mutations in the gene for toll-like receptor 4 and multiple sclerosis. Tissue Antigens. 2003;61:85–88. doi: 10.1034/j.1399-0039.2003.610108.x. [DOI] [PubMed] [Google Scholar]
  • 99.Cooke G.S., Segal S., Hill A.V. Tuberculosis, genetics, and environmental (TBGENENV) study group, Oxford Pneumococcal Group. Toll-like receptor 4 polymorphism and atherogenesis. N Engl J Med. 2002;347 doi: 10.1056/NEJM200212123472416. 1987–1980. [DOI] [PubMed] [Google Scholar]
  • 100.Rammi V., Madhusoodhanan R., Kosanke S., Awasthi S. A TLR-4 interacting SPA4 peptide inhibits LPS-induced lung inflammation. Innate Immun. 2013;19:596–610. doi: 10.1177/1753425912474851. [DOI] [PubMed] [Google Scholar]
  • 101.Kandimalla E.R., Bhagat L., Wang D., Yu D., Sullivan T., La Monica N. Design, synthesis and biological evaluation of novel antagonist compounds of Toll-like receptors 7, 8, and 9. Nucleic Acids Res. 2013;41:3947–3961. doi: 10.1093/nar/gkt078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Li Y., Cao H., Wang N., Xiang Y., Lu Y., Zhao K. A novel antagonist of TLR9 blocking all classes of immunostimulatory CpG-ODNs. Vaccine. 2011;29:2193–2198. doi: 10.1016/j.vaccine.2010.10.042. [DOI] [PubMed] [Google Scholar]
  • 103.Hulbert S.H., Webb C.A., Smith S.M., Sun Q. Resistance gene complexes: evolution and utilization. Annu Rev Phytopathol. 2001;39:285–312. doi: 10.1146/annurev.phyto.39.1.285. [DOI] [PubMed] [Google Scholar]
  • 104.Dangl J.L., Jones J.D. Plant pathogens and integrated defense. Nature. 2001;411:826–833. doi: 10.1038/35081161. [DOI] [PubMed] [Google Scholar]
  • 105.Harton J.A., Linhoff M.W., Zhang J., Ting J.P. Cutting edge: CATERPILLER: a large family of mammalian genes containing CARD, pyrin, nucleotide-binding, and leucine-rich repeat domains. J Immunol. 2002;169:4088–4093. doi: 10.4049/jimmunol.169.8.4088. [DOI] [PubMed] [Google Scholar]
  • 106.Inohara N., Chamaillard M., McDonald C., Nunez G. NOD-LRR proteins: Role in host-microbial interactions and inflammatory disease. Annu Rev Biochem. 2005;74:355–383. doi: 10.1146/annurev.biochem.74.082803.133347. [DOI] [PubMed] [Google Scholar]
  • 107.Ting J.P., Lovering R.C., Alnemri E.S., Bertin J., Boss J.M., Davis B.K. The NLR gene family: a standard nomenclature. Immunity. 2008;28:285–287. doi: 10.1016/j.immuni.2008.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Steimle V., Siegrist C.A., Mottet A., Lisowska-Grospierre B., Mach B. Regulation of MHC class II expression by interferon-gamma mediated by the transactivator gene CIITA. Science. 1994;265:106–109. doi: 10.1126/science.8016643. [DOI] [PubMed] [Google Scholar]
  • 109.Vinzing M., Eitel J., Lippman J., Hocke A.C., Zahlten J., Slevogt H. NAIP and Ipaf control Legionella pneumophila replication in human cells. J Immunol. 2008;180:6808–6815. doi: 10.4049/jimmunol.180.10.6808. [DOI] [PubMed] [Google Scholar]
  • 110.Inohara N., Koseki T., del Peso L., Hu Y., Yee C., Chen S. Nod1, an Apaf-1 like activator of caspase -9 and nuclear factor-kappa B. J Biol Chem. 1999;274:14560–14567. doi: 10.1074/jbc.274.21.14560. [DOI] [PubMed] [Google Scholar]
  • 111.Inohara N., Ogura Y., Chen F.F., Muto A., Nunez G. Human Nod1 confers responsiveness to bacterial lipopolysaccharides. J Biol Chem. 2001;276:2551–2554. doi: 10.1074/jbc.M009728200. [DOI] [PubMed] [Google Scholar]
  • 112.Girardin S.E., Boneca I.G., Carneiro L.A., Antignac A., Jéhanno M., Viala J. Nod1 detects a unique muropeptide from gram-negative bacterial peptidoglycan. Science. 2003;300:1584–1587. doi: 10.1126/science.1084677. [DOI] [PubMed] [Google Scholar]
  • 113.Girardin S.E., Boneca I.G., Viala J., Chamaillard M., Labigne A., Thomas G. Nod2 is a general sensor of peptidoglycan through muramyl dipeptide (MDP) detection. J Biol Chem. 2003;278:8869–8872. doi: 10.1074/jbc.C200651200. [DOI] [PubMed] [Google Scholar]
  • 114.Salem M., Ammitzboel M., Nys K., Seidelin J.B., Nielsen O.H. ATG16L1: A multifunctional susceptibility factor in Crohn disease. Autophagy. 2015;11:585–594. doi: 10.1080/15548627.2015.1017187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Conti B.J., Davis B.K., Zhang J., O'Connor W., Jr., Williams K.L., Ting J.P. CATERPILLER 16.2 (CLR16.2), a novel NBD/LRR family member that negatively regulates T cell function. J Biol Chem. 2005;280:18375–18385. doi: 10.1074/jbc.M413169200. [DOI] [PubMed] [Google Scholar]
  • 116.Schneider M., Zimmerman A.G., Roberts R.A., Zhang L., Swanson K.V., Wen H. The innate immune sensor NLRC3 attenuates Toll-like receptor signaling via modification of the signaling adaptor TRAF6 and transcription factor NF-kB. Nat Immunol. 2012;13:823–831. doi: 10.1038/ni.2378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Meissner T.B., Li A., Biswas A., Lee K.H., Bayir E., Iliopoulos D. NLR family member NLRC5 is a transcriptional regulator of MHC class I gene. Proc Natl Acad Sci USA. 2010;107:13794–13799. doi: 10.1073/pnas.1008684107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Benko S., Magalhaes J.G., Philpott D.J., Girardin S.E. NLRC5 limits the activation of inflammatory pathways. J Immunol. 2010;185:1681–1691. doi: 10.4049/jimmunol.0903900. [DOI] [PubMed] [Google Scholar]
  • 119.Biswas A., Meissner T.B., Kawai T., Kobayashi K.S. Cutting edge: Impaired MHC class I expression in mice deficient for Nlrc5/class I transactivator. J Immunol. 2012;189:516–520. doi: 10.4049/jimmunol.1200064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Amer A., Franchi L., Kanneganti T.D., Body-Malapel M., Ozören N., Brady G. Regulation of Legionella phagosome maturation and infection through flagellin and host Ipaf. J Biol Chem. 2006;281:35217–35223. doi: 10.1074/jbc.M604933200. [DOI] [PubMed] [Google Scholar]
  • 121.Franchi L., Amer A., Body-Malapel M., Kanneganti T.D., Ozören N., Jagirdar R. Cytosolic flagellin requires Ipaf for activation of caspase-1 and interleukin 1beta in salmonella-infected macrophages. Nat Immunol. 2006;7:576–582. doi: 10.1038/ni1346. [DOI] [PubMed] [Google Scholar]
  • 122.Mariathasan S., Newton K., Monack D.M., Vucic D., French D.M., Lee W.P. Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf. Nature. 2004;430:213–218. doi: 10.1038/nature02664. [DOI] [PubMed] [Google Scholar]
  • 123.Miao E.A., Alpuche-Aranda C.M., Dors M., Clark A.E., Bader M.W., Miller S.I. Cytoplasmic flagellin activates caspase-1 and secretion of interleukin 1beta via Ipaf. Nat Immunol. 2006;7:569–575. doi: 10.1038/ni1344. [DOI] [PubMed] [Google Scholar]
  • 124.Abdul-Sater A.A., Saïd-Sadier N., Lam V.M., Singh B., Pettengill M.A., Soares F. Enhancement of reactive oxygen species production and chlamydial infection by the mitochondrial Nod-like family member NLRX1. J Biol Chem. 2010;285:41637–41645. doi: 10.1074/jbc.M110.137885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Allen I.C., Moore C.B., Schneider M., Lei Y., Davis B.K., Scull M.A. NLRX1 protein attenuates inflammatory responses to infection by interfering with the RIG-I-MAVS and TRAF6-NF-κB signaling pathways. Immunity. 2011;34:854–865. doi: 10.1016/j.immuni.2011.03.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Hong M., Yoon S.I., Wilson I.A. Structure and functional characterization of the RNA-binding element of the NLRX1 innate immune modulator. Immunity. 2012;36:337–347. doi: 10.1016/j.immuni.2011.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Tattoli I., Carneiro L.A., Jéhanno M., Magalhaes J.G., Shu Y., Philpott D.J. NLRX1 is a mitochondrial NOD-like receptor that amplifies NF-kappaB and JNK pathways by inducing reactive oxygen species production. EMBO Rep. 2012;9:293–300. doi: 10.1038/sj.embor.7401161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Jin Y., Mailloux C.M., Gowan K., Riccardi S.L., LaBerge G., Bennett D.C. NALP1 in vitiligo-associated multiple autoimmune disease. N Engl J Med. 2007;356:1216–1225. doi: 10.1056/NEJMoa061592. [DOI] [PubMed] [Google Scholar]
  • 129.Bruey J.M., Bruey-Sedano N., Luciano F., Zhai D., Balpai R., Xu C. Bcl-2 and Bcl-XL regulate proinflammatory caspase-1 activation by interaction with NALP1. Cell. 2007;129:45–56. doi: 10.1016/j.cell.2007.01.045. [DOI] [PubMed] [Google Scholar]
  • 130.Eisenbarth S.C., Williams A., Colegio O.R., Meng H., Strowig T., Rongvaux A. NLRP10 is a NOD-like receptor essential to initiate adaptive immunity by dendritic cells. Nature. 2012;484:510–513. doi: 10.1038/nature11012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Krishnaswamy J.K., Singh A., Gowthaman U., Wu R., Gorrepati P., Sales Nascimento M. Coincidental loss of DOCK8 function in NLRP10-deficient and C3H/HeJ mice results in defective cell migration. Proc Natl Acad Sci USA. 2015;112:3056–3061. doi: 10.1073/pnas.1501554112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Peng H., Chang B., Lu C., Su J., Wu Y., Lv P. Nlrp2, a maternal effect gene required for early embryonic development in the mouse. PLos One. 2012;7:e30344. doi: 10.1371/journal.pone.0030344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Bruey J.M., Bruey-Sedano N., Newman R., Chandler S., Stehlik C., Reed J.C. PAN1/NALP2/PYPAF2, an inducible inflammatory mediator that regulates NF-kappaB and caspase-1 activation in macrophages. J Biol Chem. 2004;279:51897–51907. doi: 10.1074/jbc.M406741200. [DOI] [PubMed] [Google Scholar]
  • 134.Fontalba A., Gutierrez O., Fernandez-Luna J.L. NLRP2, an inhibitor of the NF-kappaB pathway, is transcriptionally activated by NF-kappaB and exhibits a nonfunctional allelic variant. J Immunol. 2007;179:8519–8524. doi: 10.4049/jimmunol.179.12.8519. [DOI] [PubMed] [Google Scholar]
  • 135.Mariathasan S., Weiss D.S., Newton K., McBride J., O'Rourke K., Roose-Girma M. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature. 2006;440:228–232. doi: 10.1038/nature04515. [DOI] [PubMed] [Google Scholar]
  • 136.Petrilli V., Papin S., Dostert C., Mayor A., Martinon F., Tschopp J. Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration. Cell Death Differ. 2007;14:1583–1589. doi: 10.1038/sj.cdd.4402195. [DOI] [PubMed] [Google Scholar]
  • 137.Eisenbarth S.C., Colegio O.R., O'Connor W., Sutterwala F.S., Flavell R.A. Crucial role for the Nalp3 inflammasome in the immunostimulatory properties of aluminium adjuvants. Nature. 2008;453:1122–1126. doi: 10.1038/nature06939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Cassel S.L., Eisenbarth S.C., Iyer S.S., Sadler J.J., Colegio O.R., Tephly L.A. The Nlrp3 inflammasome is essential for the development of silicosis. Proc Natl Acad Soc USA. 2008;105:9035–9040. doi: 10.1073/pnas.0803933105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Dostert C., Petrilli V., Van Bruggen R., Steele C., Mossman B.T., Tschopp J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science. 2008;320:674–677. doi: 10.1126/science.1156995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Hornung V., Bauernfeind F., Halle A., Samstad E.O., Kono H., Rock K.L. Silica crystals and aluminium salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol. 2008;9:847–856. doi: 10.1038/ni.1631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Cruz C.M., Rinna A., Forman H.J., Ventura A.L., Persechini P.M., Ojcius D.M. ATP activates a reactive oxygen species-dependent oxidative stress response and secretion of proinflammatory cytokines in macrophages. J Biol Chem. 2007;282:2817–2819. doi: 10.1074/jbc.M608083200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Joly S., Ma N., Sadler J.J., Soll D.R., Cassel S.L., Sutterwala F.S. Cutting edge: Candida albicans hyphae formation triggers activation of the Nlrp3 inflammasome. J Immunol. 2009;183:3578–3581. doi: 10.4049/jimmunol.0901323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Martinon F., Pétrilli V., Mayor A., Tardivel A., Tschopp J. Gout associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440:237–241. doi: 10.1038/nature04516. [DOI] [PubMed] [Google Scholar]
  • 144.Yamasaki K., Muto J., Taylor K.R., Cogen A.L., Audish D., Bertin J. NLRP3/cryopyrin is necessary for interleukin-1beta (IL-1beta) release in response to hyaluronan, an endogenous trigger of inflammation in response to injury. J Biol Chem. 2009;284:12762–12771. doi: 10.1074/jbc.M806084200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Halle A., Hornung V., Petzold G.C., Stewart C.R., Monks B.G., Reinheckel T. The NALP3 inflammasome is involved in the innate immune response to amyloid-beta. Nat Immunol. 2008;9:857–865. doi: 10.1038/ni.1636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Yazdi A.S., Guarda G., Riteau N., Drexler S.K., Tardivel A., Couillin I. Nanoparticles activate the NLR pyrin domain containing 3 (Nlrp3) inflammasome and cause pulmonary inflammation through release of IL-1α and IL-1β. Proc Natl Acad Sci USA. 2010;107:19449–19454. doi: 10.1073/pnas.1008155107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Chang B.H., Liu X., Liu J., Quan F.S., Guo Z.K., Zhang Y. Developmental expression and possible functional roles of mouse Nlrp4e in preimplantation embryos. Vitro Cell Dev Biol Anim. 2013;49:548–553. doi: 10.1007/s11626-013-9638-9. [DOI] [PubMed] [Google Scholar]
  • 148.Cui J., Li Y., Zhu L., Liu D., Songyang Z., Wang H.Y. NLRP4 negatively regulates type I interferon signaling by targeting the kinase TBK1 for degradation via the ubiquitin ligase DTX4. Nat Immunol. 2012;13:387–395. doi: 10.1038/ni.2239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Jounai N., Kobiyama K., Shiina M., Ogata K., Ishii K.J., Takeshita F. NLRP4 negatively regulates autophagic processes through an association with beclin1. J Immunol. 2011;186:1646–1655. doi: 10.4049/jimmunol.1001654. [DOI] [PubMed] [Google Scholar]
  • 150.Florentino L., Stehlik C., Oliveira V., Ariza M.E., Godzik A., Reed J.C. A novel PAAD-containing protein that modulates NF-kappa B induction by cytokine tumor necrosis factor-alpha and interleukin-1beta. J Biol Chem. 2002;277:35333–35340. doi: 10.1074/jbc.M200446200. [DOI] [PubMed] [Google Scholar]
  • 151.Fernandes R., Tsuda C., Perumalsamy A.L., Naranian T., Chong J., Acton B.M. NLRP5 mediates mitochondrial function in mouse oocytes and embryos. Biol Reprod. 2012;86:131–138. doi: 10.1095/biolreprod.111.093583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Anand P.K., Malireddi R.K., Lukens J.R., Vogel P., Bertin J., Lamkanfi M. NLRP6 negatively regulates innate immunity and host defence against bacterial pathogens. Nature. 2012;488:389–393. doi: 10.1038/nature11250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Grenier J.M., Wang L., Manji G.A., Huang W.J., Al-Garawi A., Kelly R. Functional screening of five PYPAF family members identifies PYPAF5 as a novel regulator of NF-kappaB and caspase-1. FEBS Lett. 2002;530:73–78. doi: 10.1016/s0014-5793(02)03416-6. [DOI] [PubMed] [Google Scholar]
  • 154.Murdoch S., Djuric U., Mazhar B., Seoud M., Khan R., Kuick R. Mutations in NALP7 cause recurrent hydatidiform moles and reproductive wastage in humans. Nat Genet. 2006;38:300–302. doi: 10.1038/ng1740. [DOI] [PubMed] [Google Scholar]
  • 155.Deveault C., Qian J.H., Chebaro W., Ao A., Gilbert L., Mehio A. NLRP7 mutations in women with diploid androgenetic and triploid moles: A proposed mechanism for mole formation. Hum Mol Genet. 2009;18:888–897. doi: 10.1093/hmg/ddn418. [DOI] [PubMed] [Google Scholar]
  • 156.Khare S., Dorfleutner A., Bryan N.B., Yun C., Radian A.D., de Almeida L. An NLRP7-containing inflammasome mediates recognition of microbial lipopeptides in human macrophages. Immunity. 2012;36:464–476. doi: 10.1016/j.immuni.2012.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Allen I.C., Wilson J.E., Schneider M., Lich J.D., Roberts R.A., Arthur J.C. NLRP12 suppresses colon inflammation and tumorigenesis through the negative regulation of noncanonical NF-κB signaling. Immunity. 2012;36:742–754. doi: 10.1016/j.immuni.2012.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Zamoshnikova A., Gross C.J., Schuster S., Chen K.W., Wilson A., Tacchini-Cottier F. NLRP12 is a neutrophil-specific, negative regulator of in vitro cell migration but does not modulate LPS- or infection induced NF-κB or ERK signaling. Immunobiology. 2016;221:341–346. doi: 10.1016/j.imbio.2015.10.001. [DOI] [PubMed] [Google Scholar]
  • 159.Ataide M.A., Andrade W.A., Zamboni D.S., Wang D., Souza Mdo C., Franklin B.S. Malaria-induced NLRP12/NLRP3-dependent caspase-1 activation mediates inflammation and hypersensitivity to bacterial superinfection. PLoS Pathog. 2014;10:e1003885. doi: 10.1371/journal.ppat.1003885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Westerveld G.H., Korver C.M., van Pelt A.M., Leschot N.J., van der Veen F., Repping S. Mutations in the testis-specific NALP14 gene in men suffering from spermatogenic failure. Hum Reprod. 2006;21:3178–3184. doi: 10.1093/humrep/del293. [DOI] [PubMed] [Google Scholar]
  • 161.Koonin E.V., Aravind L. The NACHT family. A new group of predicted NTPase implicated in apoptosis and MHC-transcription activation. Trends Biochem Sci. 2000;25:223–224. doi: 10.1016/s0968-0004(00)01577-2. [DOI] [PubMed] [Google Scholar]
  • 162.Bertin J., Nir W.J., Fischer C.M., Tayber O.V., Errada P.R., Grant J.R. Human CARD4 protein is a novel CED-4/Apaf-1 cell death family member that activates NF-kappaB. J Biol Chem. 1999;274:12955–12958. doi: 10.1074/jbc.274.19.12955. [DOI] [PubMed] [Google Scholar]
  • 163.Kobayashi K., Inohara N., Hernandez L.D., Galan J.E., Nunez G., Janeway C.A. RICK/Rip2/CARDIAK mediates signaling for receptors of the innate and adaptive immune system. Nature. 2002;416:194–199. doi: 10.1038/416194a. [DOI] [PubMed] [Google Scholar]
  • 164.Leifer C.A., Medvedef A.E. Molecular mechanism of regulation of Toll-like receptor signaling. J Leukoc Biol. 2016;100:927–941. doi: 10.1189/jlb.2MR0316-117RR. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Kobayashi K.S., van den Elsen P.J. NLRC5: a key regulator of MHC class I-dependent immune responses. Nat Rev Immunol. 2012;12:813–820. doi: 10.1038/nri3339. [DOI] [PubMed] [Google Scholar]
  • 166.Cooney B.J., Baker J., Brain O., Danis B., Pickulik T., Allan P. NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation. Nat Med. 2010;16:90–97. doi: 10.1038/nm.2069. [DOI] [PubMed] [Google Scholar]
  • 167.Deretic V. Autophagy as an innate immunity paradigm: expanding the scope and repertoire of pattern recognition receptors. Current Opin Immunol. 2012;24:21–31. doi: 10.1016/j.coi.2011.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Carneiro L.A., Travassos L.H. The interplay between NLRs and autophagy in immunity and inflammation. Front Immunol. 2013;11:361. doi: 10.3389/fimmu.2013.00361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Davis B.K., Wen H., Ting J.P. The inflammasome NLRs in immunity, inflammation, and associated diseases. Annu Rev Immunol. 2011;29:707–735. doi: 10.1146/annurev-immunol-031210-101405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Franchi L., Eigenbrod T., Munoz-Planillo R., Nunez G. The inflammasome: A caspase-1-activation platform that regulates immune responses and disease pathogenesis. Nat Immunol. 2009;10:241–247. doi: 10.1038/ni.1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Steimle V., Otten L.A., Zufferey M., Mach B. Complementation cloning of an MHC class II transactivator mutated in hereditary MHC class II deficiency (or bare lymphocyte syndrome) Cell. 1993;75:135–146. [PubMed] [Google Scholar]
  • 172.Roy N., Mahadevan M.S., McLean M., Schutler G., Yaraghi Z., Farahani R. The gene for neuronal apoptosis is inhibitory protein is partially deleted with spinal muscular atrophy. Cell. 1995;80:167–178. doi: 10.1016/0092-8674(95)90461-1. [DOI] [PubMed] [Google Scholar]
  • 173.Hysi P., Kabesch M., Moffatt M.F., Schedel M., Carr D., Zhang Y. NOD1 variation, immunoglobulin E and asthma. Hum Mol Genet. 2005;14:935–941. doi: 10.1093/hmg/ddi087. [DOI] [PubMed] [Google Scholar]
  • 174.McGovern D.P., Hysi P., Ahmad T., van Heel D.A., Moffatt M.F., Carey A. Association between a complex insertion/deletion polymorphism in NOD1 (CARD4) and susceptibility to inflammatory bowel disease. Hum Mol Genet. 2005;14:1245–1250. doi: 10.1093/hmg/ddi135. [DOI] [PubMed] [Google Scholar]
  • 175.Ogura Y., Bonen D.K., Inohara N., Nicolae D.L., Chen F.F., Ramos R. A frameshift mutation in NOD2 associated with susceptibility to Crohn's disease. Nature. 2001;411:603–606. doi: 10.1038/35079114. [DOI] [PubMed] [Google Scholar]
  • 176.Miceli-Richard C., Lesage S., Rybojad M., Prieur A.M., Manouvrier-Hanu S., Häfner R. CARD15 mutations in Blau syndrome. Nat Genet. 2001;29:19–20. doi: 10.1038/ng720. [DOI] [PubMed] [Google Scholar]
  • 177.Magitta N.F., Boe Wolff A.S., Johansson S., Skinningsrud B., Lie B.A., Myhr K.M. A coding polymorphism in NALP1 confers risk for autoimmune Addison's disease and type 1 diabetes. Genes Immun. 2009;10:120–124. doi: 10.1038/gene.2008.85. [DOI] [PubMed] [Google Scholar]
  • 178.Borghini S., Tassi S., Chiesa S., Caroli F., Carta S., Caorsi R. Clinical presentation and pathogenesis of cold-induced autoinflammatory disease in a family with recurrence of an NLRP12 mutation. Arthritis Rheum. 2011;63:830–839. doi: 10.1002/art.30170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Meyer E., Lim D., Pasha S., Tee L.J., Rahman F., Yates J.R. Germline mutation in NLRP2 (NALP2) in a familial imprinting disorder (Beckwith-Wiedemann Syndrome) PLoS Genet. 2009;5:e1000423. doi: 10.1371/journal.pgen.1000423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Feldman J., Prieur A.M., Quartier P., Berguin P., Certain S., Cortis E. Chronic neurological cutaneous and articular syndrome is caused by mutations in CIAS1, a gene highly expressed in polymorphonuclear cells and chondrocytes. Am J Hum Genet. 2002;71:198–203. doi: 10.1086/341357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Aksentijevich I., Nowak M., Mallah M., Chae J.J., Watford W.T., Hoffman S.R. De novo CIAS1 mutations, cytokine activation, and evidence for genetic heterogeneity in patients with neonatal-onset multisystem inflammatory disease (NOMID): a new member of the expanding family of pyrin-associated autoinflammatory diseases. Arthritis Rheum. 2002;46:3340–3348. doi: 10.1002/art.10688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Hoffman H.M., Mueller J.L., Broide D.H., Wanderer A.A., Kolodner R.D. Mutation of a new gene encoding a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle-Wells syndrome. Nat Genet. 2001;29:301–305. doi: 10.1038/ng756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Fletta P. Autoinflammatory disease: The hereditary periodic fever syndromes. Acta Biomed. 2004;75:92–99. [PubMed] [Google Scholar]
  • 184.Kastner D.L., Aksentijevich I., Goldbach-Mansky G. Autoinflammatory disease reloaded: A clinical perspective. Cell. 2010;140:784–790. doi: 10.1016/j.cell.2010.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Kufer T.A., Sansonetti P.J. NLR functions beyond pathogen recognition. Nat Immunol. 2011;12:121–128. doi: 10.1038/ni.1985. [DOI] [PubMed] [Google Scholar]
  • 186.Zhang P., Dixon M., Zucchelli M., Hambiliki F., Levkov L., Hovatta O. Expression analysis of the NLRP gene family suggests a role in human preimplantation development. PLoS One. 2008;3:e2755. doi: 10.1371/journal.pone.0002755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Bortoluci K.R., Medzhitov R. Control of infection by pyroptosis and autophagy: role of TLR and NLR. Cell Mol Life Sci. 2010;67:1643–1651. doi: 10.1007/s00018-010-0335-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Pajarinen J., Jamsen E., Konttinen Y.T., Goodman S.B. Innate immune reaction in septic and aseptic osteolysis around total hip implants. J Long Term Eff Med Implants. 2014;24:283–296. doi: 10.1615/jlongtermeffmedimplants.2014010564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Jämsen E., Kouri V.P., Ainola M., Goodman S.B., Nordström D.C., Eklund K.K. Correlations between macrophage polarizing cytokines, inflammatory mediators, osteoclast activity, and toll-like receptors in tissues around aseptically loosened hip implants. J Biomed Mater Res A. 2017;105:454–463. doi: 10.1002/jbm.a.35913. [DOI] [PubMed] [Google Scholar]
  • 190.Valladares R.D., Nich C., Zwingenberger S., Li C., Swank K.R., Gibon E. Toll-like receptors-2 and 4 are overexpressed in an experimental model of particle-induced osteolysis. J Biomed Mater Res A. 2014;102:3004–3011. doi: 10.1002/jbm.a.34972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Naganuma Y. Yamagata University Graduate School of Medicine; 2014. Analyses on immunohistochemical localization of innate immune sensors, Toll-like receptor 2 and nucleotide-binding oligomerization domain-like receptor with a pyrin domain 3, in aseptic periprosthetic tissues around loose hip joint and their response to foreign body particles; pp. 1–22. Thesis. [Google Scholar]
  • 192.St Pierre C.A., Chan M., Iwakura Y., Ayers D.C., Kurt-Jones E.A., Finberg R.W. Periprosthetic osteolysis: Characterizing the innate immune response to titanium particle. J Orthop Res. 2010;28:1418–1424. doi: 10.1002/jor.21149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Burton L., Paget D., Binder N.B., Bohnert K., Nestor B.J., Sculco T.P. Orthopaedic wear debris mediated inflammatory osteolysis is mediated in part by NALP3 inflammasome activation. J Orthop Res. 2013;31:73–80. doi: 10.1002/jor.22190. [DOI] [PubMed] [Google Scholar]
  • 194.Maitra R., Clement C.C., Scharf B., Crisi G.M., Chitta S., Paget D. Endosomal damage and TLR2 mediated inflammasome activation by alkane particles in the generation of aseptic osteolysis. Mol Immunol. 2009;47:175–184. doi: 10.1016/j.molimm.2009.09.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Cobelli N., Scharf B., Crisi M., Hardin J., Santambrogio L. Mediators of the inflammatory response to joint replacement device. Nat Rev Rheumatol. 2011;7:600–608. doi: 10.1038/nrrheum.2011.128. [DOI] [PubMed] [Google Scholar]
  • 196.Tyson-Capper A.J., Lawrence H., Holland J.P., Deehan D.J., Kirby J.A. Metal-on-metal hips: Cobalt can induce endotoxin-like response. Ann Rheum Dis. 2013;72:460–461. doi: 10.1136/annrheumdis-2012-202468. [DOI] [PubMed] [Google Scholar]
  • 197.Grandjean-Laquerriere A., Tabary O., Jacquot J., Richard D., Guenounou M., Laurent-Maquin D. Involvement of toll-like receptor 4 in the inflammatory reaction induced by hydroxyapatite particles. Biomaterials. 2007;28:400–404. doi: 10.1016/j.biomaterials.2006.09.015. [DOI] [PubMed] [Google Scholar]
  • 198.Potnis P.A., Dutta D.K., Wood S.C. Toll-like receptor 4 signaling pathway mediates proinflammatory immune response to cobalt-alloy particles. Cell Immunol. 2013;282:53–65. doi: 10.1016/j.cellimm.2013.04.003. [DOI] [PubMed] [Google Scholar]
  • 199.Lawrence H.I., Mawdesley A.E., Holland J.P., Kirby J.A., Deeban D.J., Tyson-Capper A.J. Targeting Toll-like receptor 4 prevents cobalt-mediated inflammation. Oncotarget. 2016;7:7578–7585. doi: 10.18632/oncotarget.7105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Bi Y., Seabold J.M., Kaar S.G., Ragab A.A., Goldberg V.M., Anderson J.M. Adherent endotoxin on orthopaedic wear particles stimulates cytokine production and osteoclast differentiation. J Bone Miner Res. 2001;16:2082–2091. doi: 10.1359/jbmr.2001.16.11.2082. [DOI] [PubMed] [Google Scholar]
  • 201.Ragab A.A., Van de Motter R., Lavish S.A., Goldberg V.M., Ninomiya J.T., Carlin C.R. Measurement and removal of adoherent endotoxin from titanium particles and implant surfaces. J Orthop Res. 1999;17:803–809. doi: 10.1002/jor.1100170603. [DOI] [PubMed] [Google Scholar]
  • 202.Nalepka J.L., Lee M.J., Kraay M.J., Marcus R.E., Goldberg V.M., Chen X. Lipopolysaccharide found in aseptic loosening of patients with inflammatory arthritis. Clin Orthop Relate Res. 2006;451:229–235. doi: 10.1097/01.blo.0000224050.94248.38. [DOI] [PubMed] [Google Scholar]
  • 203.Greenfield E.M., Bi Y., Ragab A.A., Goldberg V.M., Nalepka J.L., Seabold J.M. Does endotoxin contribute to aseptic loosening of orthopaedic implants? J Biomed Mater Res B Appl Biomater. 2005;72:179–185. doi: 10.1002/jbm.b.30150. [DOI] [PubMed] [Google Scholar]
  • 204.Bechtel C.P., Gebhart J.J., Tatro J.M., Kiss-Toth E., Wilkinson J.M., Greenfield E.M. Particle induced osteolysis is mediated by TIRAP/Mal in vitro. Dependence on adherent pathogen-associated molecular patterns. J Bone Joint Surg Am. 2016;98:285–294. doi: 10.2106/JBJS.O.00736. [DOI] [PubMed] [Google Scholar]
  • 205.Pajarinen J., Mackiewicz Z., Pöllänen R., Takagi M., Epstein N.J., Ma T. Titanium particles modulate expression of Toll-like receptor proteins. J Biomed Mater Res A. 2010;92:1528–1537. doi: 10.1002/jbm.a.32495. [DOI] [PubMed] [Google Scholar]
  • 206.Wasko M.K., Goodman S.B. Emperor's new clothes: Is particle disease really infected particle disease? J Orthop Res. 2016;34:1497–1504. doi: 10.1002/jor.23292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Krenn V., Moravietz L., Perino G., Kienapfel H., Ascherl R., Hassenpflug G.J. Revised histopathological consensus classification of joint implant related pathology. Pathol Res Practice. 2014;210:779–786. doi: 10.1016/j.prp.2014.09.017. [DOI] [PubMed] [Google Scholar]
  • 208.Konttinen Y.T., Pajarinen J., Takakubo Y., Gallo J., Nich C., Takagi M. Macrophage polarization and activation in response to implant debris: influence by “particle disease” and “ion disease”. J Long Term Eff Med Implant. 2014;24:267–281. doi: 10.1615/jlongtermeffmedimplants.2014011355. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Orthopaedic Translation are provided here courtesy of Chinese Speaking Orthopaedic Society

RESOURCES