J Bacteriol Virol.  2011 Dec;41(4):225-235. 10.4167/jbv.2011.41.4.225.

Toll-like Receptors and Innate Immunity

Affiliations
  • 1Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea. hayoungj@cnu.ac.kr
  • 2Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, Korea.

Abstract

Toll-like receptors (TLRs) are the best-characterized membrane-bound receptors in innate immune cells, including macrophages and dendritic cells. Upon recognition of specific ligands originating from pathogen- and modified self-derived molecules, TLRs trigger intracellular signaling cascades that involve various adaptor proteins and enzymes, resulting in the generation of proinflammatory and antimicrobial responses through the activation of transcription factors such as nuclear factor-kappaB. TLR-dependent signaling pathways are tightly regulated during innate immune responses by a variety of negative regulators. This review focuses on the newly described regulation of TLR-dependent signaling pathways, and emphasizes the roles of TLRs in innate immunity. Efforts to modulate these regulatory pathways and signaling molecules may result in the development of new therapeutic strategies through TLR-based therapy.

Keyword

Toll-like receptor; Nuclear factor-kappaB; Innate immunity

MeSH Terms

Dendritic Cells
Immunity, Innate
Ligands
Macrophages
Proteins
Toll-Like Receptors
Transcription Factors
Ligands
Proteins
Toll-Like Receptors
Transcription Factors

Figure

  • Figure 1. Toll-like receptor signaling pathways. Toll-like receptor (TLR) signaling is activated by stimulation of TLR ligands. TLR stimulation recruits MyD88 adaptor protein to the all TLRs except for TLR3. MyD88 interacts with a complex of the IRAKs and TRAF6 to activate the TAK1, which subsequently induces translocation of NF-κB and AP-1 to the nucleus through degradation of IκB proteins and activation of MAPKs, respectively. It leads to the expression of genes encoding the pro-inflammatory cytokines. Mal is also recruited to the TLR2/1, TLR2/6 and TLR4 to activate the MyD88-dependent pathway. TRIF protein is recruited to TLR3 and TLR4, which induces the interaction with a complex of TRAF3, TBK1 and IKKi to activate phosphorylation of IRF3. Activated IRF3 is dimerized and translocated into the nucleus, which induces protein expression of type I IFNs. TRIF also interacts with a TRAF6-RIP1 complex to activate NF-κB. TRAM is responsible for activation of TRIF-dependent pathway in TLR4, but not TLR3 signaling. Stimulation with ligands for TLR7, TLR8 and TLR9 forms a signaling complex consisting of MyD88, IRAK4, TRAF6, TRAF3, and IRAK1. TRAF6 and TRAF3 are responsible for activation of NF-κB (for proinflammatory cytokines) and IRF7 (for type I IFNs), respectively.

  • Figure 2. Negative regulators of Toll-like receptor signaling. Endogenous negative regulators inhibit excessive TLR signaling in diverse points. Soluble TLR2 and TLR4 function as competitors through inhibition of the interaction of TLR2 with ligand and formation of TLR4-MD2 complex, respectively. Both ST2L and SIGIRR are membrane-associated TLR regulators. ST2L binds to the MyD88 and MAL, whereas SIGIRR binds to TLR4, IRAK4 and TRAF6. Both of them inhibit MyD88-dependent pathway. Other intracellular TLR regulators include sMyD88, IRAKM, A20 and SHP: sMyD88 substitutes MyD88 to antagonize MyD88-dependent pathway through attenuation of IRAK4 recruitment; IRAKM inhibits IRAK1 phosphorylation by targeting IRAK1-IRAK4 complex; A20 is an inducible de-ubiquitination enzyme and de-ubiquitinylates TRAF6 to terminate TLR signaling; SHP functions as both a repressor of NF-κB and an inhibitor of TRAF6 ubiquitination.


Cited by  12 articles

Helicobacter pylori: Bacterial Strategy for Incipient Stage and Persistent Colonization in Human Gastric Niches
Kwang-Ho Rhee, Jin-Sik Park, Myung-Je Cho
Yonsei Med J. 2014;55(6):1453-1466.    doi: 10.3349/ymj.2014.55.6.1453.

Mitochondrial Control of Innate Immunity and Inflammation
Hyo Sun Jin, Hyun-Woo Suh, Seong-Jun Kim, Eun-Kyeong Jo
Immune Netw. 2017;17(2):77-88.    doi: 10.4110/in.2017.17.2.77.

Intracellular Signaling Pathways that Regulate Macrophage Chemokine Expression in Response to Mycobacterium abscessus
Tae Sung Kim, Hye-Mi Lee, Heekyung Yoo, Young Kil Park, Eun-Kyeong Jo
J Bacteriol Virol. 2012;42(2):121-132.    doi: 10.4167/jbv.2012.42.2.121.

Mitogen-activated Protein Kinases in Inflammation
Zahid Manzoor, Young-Sang Koh
J Bacteriol Virol. 2012;42(3):189-195.    doi: 10.4167/jbv.2012.42.3.189.

Toll-Like Receptor Ligands as Cancer Immunotherapeutics
Shee Eun Lee, Joon Haeng Rhee
J Bacteriol Virol. 2012;42(3):255-262.    doi: 10.4167/jbv.2012.42.3.255.

Bacterial 23S Ribosomal RNA, a Ligand for Toll-like Receptor 13
Zahid Manzoor, Young-Sang Koh
J Bacteriol Virol. 2012;42(4):357-358.    doi: 10.4167/jbv.2012.42.4.357.

Anti-inflammatory Activity of Carpinus tschonoskii Leaves Extract in R848-stimulated Bone Marrow-derived Macrophages and Dendritic Cells
Sung-Ha Kang, Jung-Eun Koo, Hye-Jin Hong, Vivek Bhakta Mathema, Young-Sang Koh
J Bacteriol Virol. 2012;42(1):77-82.    doi: 10.4167/jbv.2012.42.1.77.

Roles of Enteric Microbial Composition and Metabolism in Health and Diseases
Jung Mogg Kim
Korean J Gastroenterol. 2013;62(4):191-205.    doi: 10.4166/kjg.2013.62.4.191.

The Synergistic Effects of Antimicrobial Peptides on the Growth Inhibition of Salmonella Typhimurium through Imd Pathway in Drosophila Intestine
Yun-Ji Lim, Yea-Hyeon Jo, Hwa-Jung Kim, Jeong-Kyu Park
J Bacteriol Virol. 2013;43(2):120-130.    doi: 10.4167/jbv.2013.43.2.120.

Peptidylarginine Deiminase and Citrullination: Potential Therapeutic Targets for Inflammatory Diseases
Byungki Jang, Sung Jae Shin
J Bacteriol Virol. 2013;43(3):159-167.    doi: 10.4167/jbv.2013.43.3.159.

Sensing DNA Viruses and Bacteria by Intracellular DNA Sensors
Na-Rae Lee, Han-Bo Shin, Hye-In Kim, Myung-Soo Choi, Kyung-Soo Inn
J Bacteriol Virol. 2013;43(2):77-84.    doi: 10.4167/jbv.2013.43.2.77.

Acrosorium polyneurum Extract Inhibits the LPS-Induced Inflammatory Response by Impairing the MAPK and NF-κB Pathways
Zahid Manzoor, Irshad Ali, Doobyeong Chae, Young-Sang Koh
J Bacteriol Virol. 2016;46(4):288-294.    doi: 10.4167/jbv.2016.46.4.288.


Reference

1). Medzhitov R. Recognition of microorganisms and activation of the immune response. Nature. 2007; 449:819–26.
Article
2). Ishii KJ, Koyama S, Nakagawa A, Coban C, Akira S. Host innate immune receptors and beyond: making sense of microbial infections. Cell Host Microbe. 2008; 3:352–63.
Article
3). Takeda K, Akira S. Toll-like receptors in innate immunity. Int Immunol. 2005; 17:1–14.
4). Sun SC, Ley SC. New insights into NF-kappaB regulation and function. Trends Immunol. 2008; 29:469–78.
5). Li X, Jiang S, Tapping RI. Toll-like receptor signaling in cell proliferation and survival. Cytokine. 2010; 49:1–9.
Article
6). O'Neill LA. The interleukin-1 receptor/Toll-like receptor superfamily: signal transduction during inflammation and host defense. Sci STKE. 2000; 2000:re1.
7). Shibolet O, Podolsky DK. TLRs in the Gut. I V. Negative regulation of Toll-like receptors and intestinal homeostasis: addition by subtraction. Am J Physiol Gastrointest Liver Physiol. 2007; 292:G1469–73.
8). Lemaitre B, Nicolas E, Michaut L, Reichhart JM, Hoffmann JA. The dorsoventral regulatory gene cassette spatzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell. 1996; 86:973–83.
9). Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010; 11:373–84.
Article
10). Manavalan B, Basith S, Choi S. Similar Structures but Different Roles - An Updated Perspective on TLR Structures. Front Physiol. 2011; 2:41.
Article
11). Wang X, Quinn PJ. Lipopolysaccharide: Biosynthetic pathway and structure modification. Prog Lipid Res. 2010; 49:97–107.
Article
12). Cohen J. The immunopathogenesis of sepsis. Nature. 2002; 420:885–91.
Article
13). Tapping RI, Tobias PS. Mycobacterial lipoarabinomannan mediates physical interactions between TLR1 and TLR2 to induce signaling. J Endotoxin Res. 2003; 9:264–8.
Article
14). Goodridge HS, Wolf AJ, Underhill DM. Beta-glucan recognition by the innate immune system. Immunol Rev. 2009; 230:38–50.
15). Vijay-Kumar M, Gewirtz AT. Flagellin: key target of mucosal innate immunity. Mucosal Immunol. 2009; 2:197–205.
Article
16). Plattner F, Yarovinsky F, Romero S, Didry D, Carlier MF, Sher A, et al. Toxoplasma profilin is essential for host cell invasion and TLR11-dependent induction of an interleukin-12 response. Cell Host Microbe. 2008; 3:77–87.
17). Alexopoulou L, Holt AC, Medzhitov R, Flavell RA. Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3. Nature. 2001; 413:732–8.
18). Hemmi H, Kaisho T, Takeuchi O, Sato S, Sanjo H, Hoshino K, et al. Small anti-viral compounds activate immune cells via the TLR7 MyD88-dependent signaling pathway. Nat Immunol. 2002; 3:196–200.
Article
19). Diebold SS, Kaisho T, Hemmi H, Akira S, Reis e Sousa C. Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science. 2004; 303:1529–31.
Article
20). Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C, Akira S, et al. Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science. 2004; 303:1526–9.
Article
21). Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, et al. A Toll-like receptor recognizes bacterial DNA. Nature. 2000; 408:740–5.
Article
22). Carty M, Bowie AG. Evaluating the role of Toll-like receptors in diseases of the central nervous system. Biochem Pharmacol. 2011; 81:825–37.
Article
23). Jin MS, Lee JO. Structures of the toll-like receptor family and its ligand complexes. Immunity. 2008; 29:182–91.
Article
24). Latz E, Verma A, Visintin A, Gong M, Sirois CM, Klein DC, et al. Ligand-induced conformational changes allosterically activate Toll-like receptor 9. Nat Immunol. 2007; 8:772–9.
Article
25). Pålsson-McDermott EM, O'Neill LA. Building an immune system from nine domains. Biochem Soc Trans. 2007; 35:1437–44.
Article
26). Takeda K, Akira S. TLR signaling pathways. Semin Immunol. 2004; 16:3–9.
Article
27). Kawai T, Akira S. Signaling to NF-kappaB by Toll-like receptors. Trends Mol Med. 2007; 13:460–9.
28). Yamamoto M, Sato S, Hemmi H, Hoshino K, Kaisho T, Sanjo H, et al. Role of adaptor TRIF in the MyD88-independent toll-like receptor signaling pathway. Science. 2003; 301:640–3.
Article
29). Yamamoto M, Takeda K, Akira S. TIR domain-containing adaptors define the specificity of TLR signaling. Mol Immunol. 2004; 40:861–8.
Article
30). Sato S, Sugiyama M, Yamamoto M, Watanabe Y, Kawai T, Takeda K, et al. Toll/IL-1 receptor domain-containing adaptor inducing IFN-beta (TRIF) associates with TNF receptor-associated factor 6 and TANK-binding kinase 1, and activates two distinct transcription factors, NF-kappa B and IFN-regulatory factor-3, in the Toll-like receptor signaling. J Immunol. 2003; 171:4304–10.
31). Johnson AC, Li X, Pearlman E. MyD88 functions as a negative regulator of TLR3/TRIF-induced corneal inflammation by inhibiting activation of c-Jun N-terminal kinase. J Biol Chem. 2008; 283:3988–96.
Article
32). Sheedy FJ, O'Neill LA. The Troll in Toll: Mal and Tram as bridges for TLR2 and TLR4 signaling. J Leukoc Biol. 2007; 82:196–203.
Article
33). Kenny EF, Talbot S, Gong M, Golenbock DT, Bryant CE, O'Neill LA. MyD88 adaptor-like is not essential for TLR2 signaling and inhibits signaling by TLR3. J Immunol. 2009; 183:3642–51.
Article
34). Chang L, Karin M. Mammalian MAP kinase signalling cascades. Nature. 2001; 410:37–40.
Article
35). Kawai T, Akira S. TLR signaling. Cell Death Differ. 2006; 13:816–25.
Article
36). Weiss DS, Raupach B, Takeda K, Akira S, Zychlinsky A. Toll-like receptors are temporally involved in host defense. J Immunol. 2004; 172:4463–9.
Article
37). Campos MA, Closel M, Valente EP, Cardoso JE, Akira S, Alvarez-Leite JI, et al. Impaired production of proinflammatory cytokines and host resistance to acute infection with Trypanosoma cruzi in mice lacking functional myeloid differentiation factor 88. J Immunol. 2004; 172:1711–8.
38). Lawley TD, Clare S, Walker AW, Goulding D, Stabler RA, Croucher N, et al. Antibiotic treatment of clostridium difficile carrier mice triggers a supershedder state, spore-mediated transmission, and severe disease in immunocompromised hosts. Infect Immun. 2009; 77:3661–9.
39). Jarchum I, Liu M, Lipuma L, Pamer EG. Toll-like receptor 5 stimulation protects mice from acute clostridium difficile colitis. Infect Immun. 2011; 79:1498–503.
40). Takeuchi O, Hoshino K, Akira S. Cutting edge: TLR2-deficient and MyD88-deficient mice are highly susceptible to Staphylococcus aureus infection. J Immunol. 2000; 165:5392–6.
41). Gerold G, Zychlinsky A, de Diego JL. What is the role of Toll-like receptors in bacterial infections? Semin Immunol. 2007; 19:41–7.
Article
42). Miller LS, O'Connell RM, Gutierrez MA, Pietras EM, Shahangian A, Gross CE, et al. MyD88 mediates neutrophil recruitment initiated by IL-1R but not TLR2 activation in immunity against Staphylococcus aureus. Immunity. 2006; 24:79–91.
43). Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity. 2011; 34:637–50.
Article
44). Andersen-Nissen E, Hawn TR, Smith KD, Nachman A, Lampano AE, Uematsu S, et al. Cutting edge: Tlr5–/–mice are more susceptible to Escherichia coli urinary tract infection. J Immunol. 2007; 178:4717–20.
45). Feuillet V, Medjane S, Mondor I, Demaria O, Pagni PP, Galán JE, et al. Involvement of Toll-like receptor 5 in the recognition of flagellated bacteria. Proc Natl Acad Sci U S A. 2006; 103:12487–92.
Article
46). Uematsu S, Jang MH, Chevrier N, Guo Z, Kumagai Y, Yamamoto M, et al. Detection of pathogenic intestinal bacteria by Toll-like receptor 5 on intestinal CD11c+ lamina propria cells. Nat Immunol. 2006; 7:868–74.
Article
47). Arpaia N, Godec J, Lau L, Sivick KE, McLaughlin LM, Jones MB, et al. TLR signaling is required for Salmonella typhimurium virulence. Cell. 2011; 144:675–88.
48). Kumar H, Kawai T, Akira S. Pathogen recognition by the innate immune system. Int Rev Immunol. 2011; 30:16–34.
Article
49). Ku CL, Yang K, Bustamante J, Puel A, von Bernuth H, Santos OF, et al. Inherited disorders of human Tolllike receptor signaling: immunological implications. Immunol Rev. 2005; 203:10–20.
Article
50). Ku CL, von Bernuth H, Picard C, Zhang SY, Chang HH, Yang K, et al. Selective predisposition to bacterial infections in IRAK-4-deficient children: IRAK-4-dependent TLRs are otherwise redundant in protective immunity. J Exp Med. 2007; 204:2407–22.
Article
51). Casrouge A, Zhang SY, Eidenschenk C, Jouanguy E, Puel A, Yang K, et al. Herpes simplex virus encephalitis in human UNC-93B deficiency. Science. 2006; 314:308–12.
Article
52). Zhang SY, Jouanguy E, Ugolini S, Smahi A, Elain G, Romero P, et al. TLR3 deficiency in patients with herpes simplex encephalitis. Science. 2007; 317:1522–7.
Article
53). Guo Y, Audry M, Ciancanelli M, Alsina L, Azevedo J, Herman M, et al. Herpes simplex virus encephalitis in a patient with complete TLR3 deficiency: TLR3 is otherwise redundant in protective immunity. J Exp Med. 2011; 208:2083–98.
Article
54). Pérez de Diego R, Sancho-Shimizu V, Lorenzo L, Puel A, Plancoulaine S, Picard C, et al. Human TRAF3 adaptor molecule deficiency leads to impaired Toll-like receptor 3 response and susceptibility to herpes simplex encephalitis. Immunity. 2010; 33:400–11.
Article
55). Brinkmann MM, Spooner E, Hoebe K, Beutler B, Ploegh HL, Kim YM. The interaction between the ER membrane protein UNC93B and TLR3, 7, and 9 is crucial for TLR signaling. J Cell Biol. 2007; 177:265–75.
Article
56). Iwami KI, Matsuguchi T, Masuda A, Kikuchi T, Musikacharoen T, Yoshikai Y. Cutting edge: naturally occurring soluble form of mouse Toll-like receptor 4 inhibits lipopolysaccharide signaling. J Immunol. 2000; 165:6682–6.
Article
57). LeBouder E, Rey-Nores JE, Rushmere NK, Grigorov M, Lawn SD, Affolter M, et al. Soluble forms of Toll-like receptor (TLR)2 capable of modulating TLR2 signaling are present in human plasma and breast milk. J Immunol. 2003; 171:6680–9.
Article
58). Brint EK, Xu D, Liu H, Dunne A, McKenzie AN, O'Neill LA, et al. ST2 is an inhibitor of interleukin 1 receptor and Toll-like receptor 4 signaling and maintains endotoxin tolerance. Nat Immunol. 2004; 5:373–9.
Article
59). Garlanda C, Riva F, Polentarutti N, Buracchi C, Sironi M, De Bortoli M, et al. Intestinal inflammation in mice deficient in Tir8, an inhibitory member of the IL-1 receptor family. Proc Natl Acad Sci U S A. 2004; 101:3522–6.
Article
60). Bergers G, Reikerstorfer A, Braselmann S, Graninger P, Busslinger M. Alternative promoter usage of the Fosresponsive gene Fit-1 generates mRNA isoforms coding for either secreted or membrane-bound proteins related to the IL-1 receptor. EMBO J. 1994; 13:1176–88.
Article
61). Thomassen E, Renshaw BR, Sims JE. Identification and characterization of SIGIRR, a molecule representing a novel subtype of the IL-1R superfamily. Cytokine. 1999; 11:389–99.
Article
62). Wald D, Qin J, Zhao Z, Qian Y, Naramura M, Tian L, et al. SIGIRR, a negative regulator of Toll-like receptor-interleukin 1 receptor signaling. Nat Immunol. 2003; 4:920–7.
Article
63). Janssens S, Burns K, Tschopp J, Beyaert R. Regulation of interleukin-1-and lipopolysaccharide-induced NF-kappaB activation by alternative splicing of MyD88. Curr Biol. 2002; 12:467–71.
64). Janssens S, Beyaert R. Functional diversity and regulation of different interleukin-1 receptor-associated kinase (IRAK) family members. Mol Cell. 2003; 11:293–302.
Article
65). Kobayashi K, Hernandez LD, Galán JE, Janeway CA Jr, Medzhitov R, Flavell RA. IRAK-M is a negative regulator of Toll-like receptor signaling. Cell. 2002; 110:191–202.
Article
66). Opipari AW Jr, Boguski MS, Dixit VM. The A20 cDNA induced by tumor necrosis factor alpha encodes a novel type of zinc finger protein. J Biol Chem. 1990; 265:14705–8.
Article
67). Krikos A, Laherty CD, Dixit VM. Transcriptional activation of the tumor necrosis factor alpha-inducible zinc finger protein, A20, is mediated by kappa B elements. J Biol Chem. 1992; 267:17971–6.
Article
68). Boone DL, Turer EE, Lee EG, Ahmad RC, Wheeler MT, Tsui C, et al. The ubiquitin-modifying enzyme A20 is required for termination of Toll-like receptor responses. Nat Immunol. 2004; 5:1052–60.
Article
69). Seol W, Choi HS, Moore DD. An orphan nuclear hormone receptor that lacks a DNA binding domain and heterodimerizes with other receptors. Science. 1996; 272:1336–9.
Article
70). Lee YS, Chanda D, Sim J, Park YY, Choi HS. Structure and function of the atypical orphan nuclear receptor small heterodimer partner. Int Rev Cytol. 2007; 261:117–58.
Article
71). Yuk JM, Shin DM, Lee HM, Kim JJ, Kim SW, Jin HS, et al. The orphan nuclear receptor SHP acts as a negative regulator in inflammatory signaling triggered by Toll-like receptors. Nat Immunol. 2011; 12:742–51.
Article
72). Leavy O. Innate immunity: SHP regulates TLR signalling. Nat Rev Immunol. 2011; 11:502.
Full Text Links
  • JBV
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr