Reading List - UCSF Immunology Program

advertisement

INNATE IMMUNITY

Microbiology 204 Tony DeFranco

September 30, 2015

Required Reading: Discussion paper: 63; Review: 17 (or another review of your choice); additional original article: 35

Reviews:

General:

1. Janeway, CA, Jr (1992). The immune system evolved to discriminate infectious nonself from noninfectious self. Immunol. Today 13:

11-16.

2.

Janeway CA Jr, and Medzhitov R. (2002). Innate immune recognition. Ann. Rev. Immunol. 20: 197-216.

3.

Jin, MS and Lee JO. (2008). Structures of the Toll-like receptor family and its ligand complexes. Immunity 29: 182-191.

4.

Barton, G.M. and Kagan, J.C. (2009). A cell biological view of Toll-like receptor function: regulation through compartmentalization.

Nat. Rev. Immunol. 9: 535-542.

5.

Moresco, E.M.Y., LaVine, D., Beutler, B. (2011). Toll-like receptors. Current Biol. 21: R488-93.

6.

Casanova JL, Abel L, Quintana-Murci L. (2011). Human TLRs and IL-1Rs in host defense: natural insights from evolutionary, epidemiological, and clinical genetics. Annu Rev Immunol. 29: 447-491.

7.

Medzhitov R. (2008). Origin and physiological roles of inflammation. Nature 454:428-35.

8.

Elliott, M.R., Ravichandran, K.S. (2010). Clearance of apoptotic cells: implications in health and disease. J. Cell Biol. 189: 1058-1070.

9.

Dinarello, C.A. (2010) Anti-inflammatory agents: present and future. Cell 140: 935-950.

10.

Vance, R.E., Isberg, R.R., Portnoy, D.A. (2009). Patterns of pathogenesis: discrimination of pathogenic and nonpathogenic microbes by the innate immune system. Cell Host Microbe 23: 10-21,

11.

Baltimore, D. (2011). NF B is 25. Nature Immunol. 12: 683-685 (and adjacent review articles).

12.

Malynn, B., Ma, A. (2010). Ubiquitin makes its mark on immune regulation. Immunity: 33: 843-852.

13.

Nish, S., Medzhitov, R. (2011). Host defense pathways: role of redundancy and compensation in infectious disease phenotypes.

Immunity 34: 629-636.

14.

Deretic,V., Saitoh, T. and Akira, S. (2013). Autophagy in infection, inflammation, and immunity. Nat. Rev. Immunol. 13: 722-37.

15.

Moore, K.J., Sheedy, F.J., and Fisher, E.A. (2013). Macrophages in atherosclerosis: a dynamic balance. Nat. Rev. Immunol. 13: 709-21.

16.

Gay, N.J., Symmons, M.F., Gangloff, M. & Bryant, C.E. (2014). Assembly and localization of Toll-like receptor signalling complexes.

Nat. Rev. Immunol. 14: 546-558.

17.

Lamkanfi, M. and Dixit, V.M. (2014). Mechanisms and functions of inflammasomes. Cell 157: 1013-22

18.

Blander, J.M. (2014). A long awaited merger of pathways mediating host defense and programmed cell death. Nat. Rev. Immunol. 14:

601-18.

19.

Smale, S.T., Tarakhovsky, A., Natoli, G. (2014). Chromatin contributions to the regulation of innate immunity. Ann. Rev. Immunol. 32:

489-511.

20.

Carpenter, S., Ricci, E.P., Mercier, B.C., Moore, M.J. and Fitzgerald, K.A. (2014). Post-transcriptional regulation of gene expression in innate immunity. Nat. Rev. Immunol. 14: 361-372.

Anti-bacterial and anti-fungal defense

21.

Bogdan, C, Rollinghoff, M, and Diefenbach, A (2000). Reactive oxygen and reactive nitrogen intermediates in innate and specific immunity. Curr. Opin. Immunol. 12: 64-76.

22.

Lekstrom-Himes JA, and Gallin JI (2000). Immunodeficiency diseases caused by defects in phagocytes. New Engl. J. Med. 343: 1703-

1714.

23.

Ernst JD. (2000) Bacterial inhibition of phagocytosis. Cell Microbiol. 2:379-86.

24.

Walport, M.J. (2001). Complement. N. Eng. J. Med. 344: 1058-1066 and 1140-1144.

25.

Fujita, T. et al. (2004). The lectin-complement pathway—it’s role in innate immunity and evolution. Immuno. Rev. 198: 185-202.

26.

Papayannopoulos V, Zychlinsky A. (2009). NETs: a new strategy for using old weapons. Trends Immunol. 2009 30: 513-521.

27.

Serruto, D., Rappuoli, R. , Scarselli, M., Gros, P., van Strijp, J.A.G. (2010). Molecular mechanisms of complement evasion: learning from staphylococci and meningococci. Nature Rev. Microbiol. 8: 393-399.

28.

Brodsky, I.E., Medzhitov, R. (2011). Pyroptosis: macrophage suicide exposes hidden invaders. Curr. Biol. 21: R72-75.

29.

Bevins, C.L., Salzman, N.H. (2011). Paneth cells, antimicrobial peptides and maintenance of intestinal homeostasis. Nature Rev.

Microbiol. 9: 356-368.

30.

Osorio, F., Reis e Sousa, C. (2011). Myeloid C-type lectin receptors in pathogen recognition and host defense. Immunity 34: 651-664.

31.

Netea MG, Wijmenga C, O'Neill LA. (2012). Genetic variation in Toll-like receptors and disease susceptibility. Nat Immunol. 13:535-

42.

32.

Brown GD, Netea MG. (2012). Exciting developments in the immunology of fungal infections.Cell Host Microbe. 11:422-4.

33.

Philpott, D.J., Sorbara, M.T., Robertson, S.J., Croitoru, K. and Girardin, S.E. (2014). NOD proteins: regulators of inflammation in health and disease. Nat. Rev. Immunol. 14: 19-23.

34.

Von Moltke, J., Ayres, J.S., Kofoed, E.M, Chavarria-Smith, J. and Vance, R.E. (2013). Recognition of bacteria by inflammasomes.

Ann. Rev. Immunol. 31: 73-106.

35.

Broz, P., and Monack, D.M. (2013). Newly described pattern recognition receptors team up against intracellular pathogens. Nat. Rev.

Immunol. 13: 551-565.

Anti-viral defense

36.

Roulston, A. et al. (1999). Viruses and apoptosis. Ann. Rev. Microbiol. 53: 577-628.

37.

Tortorella, D., Gewurz, B.E., Furman, M.H., Schust, D.J., and Ploegh, H.L. (2000). Viral subversion of the immune system. Ann. Rev.

Immunol. 18: 861-926.

38.

Taniguchi, T., et al. (2001). IRF family of transcription factors as regulators of host defense. Ann. Rev. Immunol. 19: 623-655.

1

39.

Plasterk, RHA (2002). RNA silencing: The genome’s immune system. Science 296: 1263-65 (and accompanying articles).

40.

Liu, Y.-J. (2005). IPC: professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors. Ann. Rev. Immunol.

23: 275-306.

41.

Plantanias, L.C. (2005). Mechanisms of type I- and type II-interferon-mediated signaling. Nat. Rev. Immunol. 5: 375-386.

42.

Yoneyama M and Fujita T. (2008). Structural mechanism of RNA recognition by the RIG-I-like receptors. Immunity 29: 176-180.

43.

Ranjan P, Bowzard JB, Schwerzmann JW, Jeisy-Scott V, Fujita T, Sambhara S. (2009). Cytoplasmic nucleic acid sensors in antiviral immunity. Trends Mol Med. 15:359-68.

44.

Evans DT, Serra-Moreno R, Singh RK, Guatelli JC. (2010) BST-2/tetherin: a new component of the innate immune response to enveloped viruses. Trends Microbiol. 18:388-96.

45.

Yan, N., and Chen, Z.J. (2012). Intrinsic anti-viral immunity. Nat Immunol. 13: 214-22.

46.

Emerman, M. and Malik HS. (2010). Paleovirology—modern consequences of ancient viruses. PLoS Biology 8: e1000301.

47.

Loo, Y.-M., Gale, M. (2011). Immune signaling by RIG-I-like receptors. Immunity 34: 680-692.

48.

Hatziloannou, T. and Bieniasz, PD (2011). Anti-retroviral restriction factors. Curr. Opin. Virol. 1: 519-525.

49.

Moresco EMY and Beutler, B (2011). Resisting viral infection: the gene by gene approach. Curr. Opin. Virol. 1: 513-8.

50.

Iwasaki, A., and Pillai, P.S. (2014). Innate immunity to influenza virus infection. Nat. Rev. Immunol. 14: 315-328.

51.

Schneider, W.M., Chevillotte, M.D., and Rice, C.M. (2014). Interferon-stimulated genes: a complex web of host defenses. Ann. Rev.

Immuno. 32: 513-545.

52.

Hornung, V., Hartmann, R., Ablasser, A., and Hopfner, K.-P. (2014) OAS proteins and cGAS: unifying concepts in sensing and responding to cytosolic nucleic acids. Nat. Rev. Immunol. 14: 521-528

Original articles:

Toll-like receptors

1. Lemaitre B, Nicolas E, Michaut, L, Reichart, J-M, and Hoffman JA (1996). The dorsoventral regulatory gene cassette spatzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell 86: 973-983.

2.

Medzhitov R, Preston-Hurlburt, P, and Janeway, CA, Jr. (1997). A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature 388: 394-397.

3.

Poltorak, A, He, X, Smirnova, I, Liu, M-Y, Van Huffel, C, Du, X, Birdwell, D, Alejos, E, Silva, M, Galanos, C, Freudenberg, M,

Ricciardi-Castagnoli, P, Layton, B, and Beutler, B (1998). Defective LPS signaling in C3H/Hej and C57BL/10ScCr mice: mutations in

Tlr4 gene. Science 282: 2085-2088.

4.

Takeuchi, O, Hoshino, K, Kawai, T, Sanjo, H, Takada,o H, Ogawa, T, Takeda, K, and Akira, S. (1999). Differential roles of TLR2 and

TLR4 in recognition of gram-negative and gram-positive bacterial cell wall components. Immunity 11: 443-451.

5.

Kawai, T, Adachi, O, Ogawa, T, Takeda, K, and Akira, S. (1999). Unresponsiveness of MyD88-deficient mice to endotoxin.

Immunity 11:115-122.

6.

Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, Matsumoto M, Hoshino K, Wagner H, Takeda K, and Akira S (2000). A

Toll-like receptor recognizes bacterial DNA. Nature 408: 740-745.

7.

Alexopoulou L, Holt AC, Medzhitov R, Flavell RA. (2001). Recognition of double-stranded RNA and activation of NF-

B by Toll-like receptor 3. Nature 413:732-738.

8.

Latz E, Schoenemeyer A, Visintin A, Fitzgerald KA, Monks BG, Knetter CF, Lien E, Nilsen NJ, Espevik T, Golenbock DT, (2004).

TLR9 signals after translocating from the ER to CpG DNA in the lysosome. Nat Immunol. 5:190-8.

9.

Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C, Akira S, Lipford G, Wagner H, Bauer S. (2004). Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science. 303:1526-9.

10.

Diebold SS, Kaisho T, Hemmi H, Akira S, Reis e Sousa C. (2004). Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science 303:1529-31.

11.

Yamamoto M, Sato S, Hemmi H, Hoshino K, Kaisho T, Sanjo H, Takeuchi O, Sugiyama M, Okabe M, Takeda K, Akira S. (2003). Role of adaptor TRIF in the MyD88-independent toll-like receptor signaling pathway. Science 301: 640-3.

12.

Hoebe K, Du X, Georgel P, Janssen E, Tabeta K, Kim SO, Goode J, Lin P, Mann N, Mudd S, Crozat K, Sovath S, Han J, Beutler B.

(2003). Identification of Lps2 as a key transducer of MyD88-independent TIR signalling. Nature 424:743-8. (“Lps2” = TRIF)

13.

Yarovinsky F, Zhang D, Andersen JF, Bannenberg GL, Serhan CN, Hayden MS, Hieny S, Sutterwala FS, Flavell RA, Ghosh S, Sher A.

(2005). TLR11 activation of dendritic cells by a protozoan profilin-like protein. Science, 308:1626-1629.

14.

Kagan JC, Medzhitov R. (2006). Phosphoinositide-mediated adaptor recruitment controls Toll-like receptor signaling. Cell 125:943-55.

15.

Honda K, Ohba Y, Yanai H, Negishi H, Mizutani T, Takaoka A, Taya C, Taniguchi T. (2005). Spatiotemporal regulation of MyD88-

IRF-7 signaling for robust type-I interferon induction. Nature. 434:1035-40.

16.

LeibundGut-Landmann S, Gross O, Robinson MJ, Osorio F, Slack EC, Tsoni SV, Schweighoffer E, Tybulewicz V, Brown GD, Ruland

J, Reis e Sousa C. (2007). Syk- and CARD9-dependent coupling of innate immunity to the induction of T helper cells that produce interleukin 17. Nat Immunol. 8:630-8.

17.

Kang SJ, Liang HE, Reizis B, Locksley RM. (2008). Regulation of hierarchical clustering and activation of innate immune cells by dendritic cells. Immunity. 2008 Nov 14;29(5):819-33.

18.

Park B, Brinkmann MM, Spooner E, Lee CC, Kim YM, Ploegh HL. (2008). Proteolytic cleavage in an endolysosomal compartment is required for activation of Toll-like receptor 9. Nat Immunol. 9:1407-14.

19.

Park BS, Song DH, Kim HM, Choi BS, Lee H, Lee JO. (2009). The structural basis of lipopolysaccharide recognition by the TLR4-MD-

2 complex. Nature 458:1191-5.

20.

Lin, S-C, Lo Y-C, Wu H. (2010). Helical assembly in the MyD88-IRAK4-IRAK2 complex in TLR/IL-1R signaling. Nature 465: 885-

891.

21.

Pérez de Diego R, Sancho-Shimizu V, Lorenzo L, Puel A, Plancoulaine S, Picard C, Herman M, Cardon A, Durandy A, Bustamante J,

Vallabhapurapu S, Bravo J, Warnatz K, Chaix Y, Cascarrigny F, Lebon P, Rozenberg F, Karin M, Tardieu M, Al-Muhsen S, Jouanguy

2

E, Zhang SY, Abel L, Casanova JL. (2010). Human TRAF3 Adaptor Molecule Deficiency Leads to Impaired Toll-like Receptor 3

Response and Susceptibility to Herpes Simplex Encephalitis.

Immunity 24: 400-411.

22.

Arpaia N, Godec J, Lau L, Sivick KE, McLaughlin LM, Jones MB, Dracheva T, Peterson SN, Monack DM, Barton GM. (2011). TLR signaling is required for Salmonella typhimurium virulence. Cell 144:675-688.

Phagocytosis and killing mechanisms

23.

Shiloh MU, MacMicking JD, Nicholson S, Brause JE, Potter S, Marino M, Fang F, Dinauer M, and Nathan C (1999). Phenotype of mice and macrophages deficient in both phagocyte oxidase and inducible nitric oxide synthase. Immunity 10: 29-38.

24.

Wilson CL, Ouellette AJ, Satchell DP, Ayabe T, Lopez-Boado YS, Stratman JL, Hultgren SJ, Matrisian LM, and Parks, WC (1999).

Regulation of intestinal

-defensin activation by the metalloproteinase matrilysin in innate host defense. Science 286: 113-117.

25.

Ayabe T, Satchell DP, Wilson CL, Parks WC, Selsted, ME and Ouellette AJ (2000). Secretion of microbicidal

-defensins by intestinal

Paneth cells in response to bacteria. Nature Immunol. 1: 113-118.

26.

Reeves EP, Lu H, Jacobs HL, Messina CG, Bolsover S, Gabella G, Potma EO, Warley A, Roes J, Segal AW. (2002). Killing activity of neutrophils is mediated through activation of proteases by K+ flux. Nature. 416:291-7.

27.

Singh PK, Parsek MR, Greenberg EP, Welsh MJ. (2002). A component of innate immunity prevents bacterial biofilm development.

Nature 417:552-5.

NOD2 and the Inflammasome: intracellular recognition of bacterial infection

28.

Girardin SE, Boneca IG, Viala J, Chamaillard M, Labigne A, Thomas G, Philpott DJ, Sansonetti PJ. (2003). Nod2 is a general sensor of peptidoglycan through muramyl dipeptide (MDP) detection. J Biol Chem. 278:8869-72

29.

Inohara N, Ogura Y, Fontalba A, Gutierrez O, Pons F, Crespo J, Fukase K, Inamura S, Kusumoto S, Hashimoto M, Foster SJ, Moran

AP, Fernandez-Luna JL, Nunez G. (2003). Host recognition of bacterial muramyl dipeptide mediated through NOD2. Implications for

Crohn's disease. J Biol Chem. 278:5509-12.

30.

Kobayashi KS, Chamaillard M, Ogura Y, Henegariu O, Inohara N, Nunez G, Flavell RA. (2005). Nod2-dependent regulation of innate and adaptive immunity in the intestinal tract. Science. 307:731-4.

31.

Martinon F, Agostini L, Meylan E, Tschopp J. (2004). Identification of bacterial muramyl dipeptide as activator of the

NALP3/cryopyrin inflammasome. Curr Biol. 14:1929-1934.

32.

Martinon F, Pétrilli V, Mayor A, Tardivel A, Tschopp J. (2006). Gout-associated uric acid crystals activate the NALP3 inflammasome.

Nature 440:237-41.

33.

Woodward, J.J. Iavarone, A.T., Portnoy, D.A. (2010). c-di-AMP secreted by intracellular Listeria monocytogenes activates a host type

1 interferon response. Science 328: 1703-1705.

34.

Travassos LH, Carneiro LA, Ramjeet M, Hussey S, Kim YG, Magalhães JG, Yuan L, Soares F, Chea E, Le Bourhis L, Boneca IG,

Allaoui A, Jones NL, Nuñez G, Girardin SE, Philpott DJ. (2010). Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry. Nat Immunol. 11:55-62.

35.

Kofoed, E.M., Vance, R.E. (2011). Innate immune recognition of bacterial ligands by NAIPs determines inflammasome specificity.

Nature 477: 592-95.

36.

Ayres JS, Trinidad NJ, Vance RE. (2012). Lethal inflammasome activation by a multidrug-resistant pathobiont upon antibiotic disruption of the microbiota. Nat Medicine 18:799-806.

37.

Baroja-Mazo, A., Martín-Sánchez F, Gomez AI, Martínez CM, Amores-Iniesta J, Compan V, Barberà-Cremades M, Yagüe J, Ruiz-

Ortiz E, Antón J, Buján S, Couillin I, Brough D, Arostegui JI, Pelegrín P. (2014). The NLRP3 inflammasome is released as a particular danger signal that amplifies the inflammatory response. Nat. Immunol. 15: 738-48

38.

Franklin, B.S., Bossaller L, De Nardo D, Ratter JM, Stutz A, Engels G, Brenker C, Nordhoff M, Mirandola SR, Al-Amoudi A, Mangan

MS, Zimmer S, Monks BG, Fricke M, Schmidt RE, Espevik T, Jones B, Jarnicki AG, Hansbro PM, Busto P, Marshak-Rothstein A,

Hornemann S, Aguzzi A, Kastenmüller W, Latz E. (2014). The adaptor ASC has extracellular and prionoid activities that propagate inflammation. Nat. Immunol. 15: 727-37.

39.

Shi, J., Zhao, Y., Wang, Y., Gao, W., Ding, J., Li, P., Hu, L., and Shao, F. (2014). Inflammatory caspases are innate immune receptors for intracellular LPS. Nature 514: 187-192.

Anti-viral innate immunity

40.

Kadowaki, Antonenko S, Lau JY, and Liu YJ (2000). Natural interferon

/

-producing cells link innate and adaptive immunity. J. Exp.

Med. 192: 219-226.

41.

Li W, Li W X, Ding S W. (2002). Induction and suppression of RNA silencing by an animal virus. Science 296: 1319-21.

42.

Sheehy AM, Gaddis NC, Choi JD, Malim MH. (2002). Isolation of a human gene that inhibits HIV-1 infection and is suppressed by the viral Vif protein. Nature 418:646-50.

43.

Mangeat B, Turelli P, Caron G, Friedli M, Perrin L, Trono D. (2003). Broad antiretroviral defence by human APOBEC3G through lethal editing of nascent reverse transcripts. Nature 424: 99-103.

44.

Yoneyama M, Kikuchi M, Natsukawa T, Shinobu N, Imaizumi T, Miyagishi M, Taira K, Akira S, Fujita T. (2004). The RNA helicase

RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses.Nat Immunol. 5:730-7.

45.

Seth, R.B., Sun L., Ea C.-K., Chen, Z.J. (2005). Identification and characterization of MAVS, a mitochondrial antiviral signaling protein that activates NF-

B and IRF3. Cell 122: 669-682.

46.

Kawai T, Takahashi K, Sato S, Coban C, Kumar H, Kato H, Ishii KJ, Takeuchi O, Akira S. (2005). IPS-1, an adaptor triggering RIG-I- and Mda5-mediated type I interferon induction. Nat Immunol. 6:981-8. (NOTE: IPS-1=MAVS).

47.

Honda K, Yanai H, Negishi H, Asagiri M, Sato M, Mizutani T, Shimada N, Ohba Y, Takaoka A, Yoshida N, Taniguchi T. (2005). IRF-

7 is the master regulator of type-I interferon-dependent immune responses. Nature. 434:772-7.

48.

Li XD, Sun L, Seth RB, Pineda G, Chen ZJ. Hepatitis C virus protease NS3/4A cleaves mitochondrial antiviral signaling protein off the mitochondria to evade innate immunity. Proc Natl Acad Sci U S A. 2005 Dec 6;102(49):17717-22.

49.

Gack MU, Shin YC, Joo CH, Urano T, Liang C, Sun L, Takeuchi O, Akira S, Chen Z, Inoue S, Jung JU. (2007) TRIM25 RING-finger

E3 ubiquitin ligase is essential for RIG-I-mediated antiviral activity. Nature 446:916-920.

3

50.

Roberts TL, Idris A, Dunn JA, Kelly GM, Burnton CM, Hodgson S, Hardy LL, Garceau V, Sweet MJ, Ross IL, Hume DA, Stacey KJ.

(2009). HIN-200 proteins regulate caspase activation in response to foreign cytoplasmic DNA. Science. 323:1057-60.

51.

Hornung V, Ablasser A, Charrel-Dennis M, Bauernfeind F, Horvath G, Caffrey DR, Latz E, Fitzgerald KA. (2009). AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature 458(7237):514-8.

52.

Zelenay S, Keller AM, Whitney PG, Schraml BU, Deddouche S, Rogers NC, Schulz O, Sancho D, Reis e Sousa C. (2012). The dendritic cell receptor DNGR-1 controls endocytic handling of necrotic cell antigens to favor cross-priming of CTLs in virus-infected mice. J Clin Invest. 122:1615-27

.

53.

Ahrens S, Zelenay S, Sancho D, Hanč P, Kjær S, Feest C, Fletcher G, Durkin C, Postigo A, Skehel M, Batista F, Thompson B, Way M,

Reis e Sousa C, Schulz O. (2012). F-actin is an evolutionarily conserved damage-associated molecular pattern recognized by DNGR-1, a receptor for dead cells. Immunity 36:635-45.

54.

van de Veerdonk FL, Plantinga TS, Hoischen A, Smeekens SP, Joosten LA, Gilissen C, Arts P, Rosentul DC, Carmichael AJ, Smits-van der Graaf CA, Kullberg BJ, van der Meer JW, Lilic D, Veltman JA, Netea MG. (2011). STAT1 mutations in autosomal dominant chronic mucocutaneous candidiasis. N Engl J Med. 365:54-61.

55.

Hou, F., Sun, L., Zheng, H., Skaug, B., Jiang, Q.-X. Chen, Z.J. (2011). MAVS forms functional prion-like aggregates to activate and propagate antiviral innate immune response. Cell 146: 448-461.

56.

Bogunovic D, Byun M, Durfee LA, Abhyankar A, Sanal O, Mansouri D, Salem S, Radovanovic I, Grant AV, Adimi P, Mansouri N,

Okada S, Bryant VL, Kong XF, Kreins A, Velez MM, Boisson B, Khalilzadeh S, Ozcelik U, Darazam IA, Schoggins JW, Rice CM, Al-

Muhsen S, Behr M, Vogt G, Puel A, Bustamante J, Gros P, Huibregtse JM, Abel L, Boisson-Dupuis S, Casanova JL. (2012)

Mycobacterial disease and impaired IFN-gamma immunity in humans with inherited ISF15 deficiency. Science 337: 1684-8.

57.

Schoggins JW, Wilson SJ, Panis M, Murphy MY, Jones CT, Bieniasz P, Rice CM. (2011). A diverse range of gene products are effectors of the type 1 interferon anti-viral response. Nature 472: 481-5.

58.

Elde NC, Child SJ, Geballe AP, Malik HS. (2009). Protein kinase R reveals an evolutionary model for defeating viral mimicry. Nature

457:485-9.

59.

Odenall, C. Dixit E, Stavru F, Bierne H, Franz KM, Durbin AF, Boulant S, Gehrke L, Cossart P, Kagan JC. (2014). Diverse intracellular pathogens activate type III interferon expression from peroxisomes. Nat. Immunol. 15: 717-26.

60.

Chiu YH, Macmillan JB, Chen ZJ. (2009). RNA polymerase III detects cytosolic DNA and induces type I interferons through the RIG-I pathway. Cell 138:576-91.

61.

Burdette DL, Monroe KM, Sotelo-Troha K, Iwig JS, Eckert B, Hyodo M, Hayakawa Y, Vance RE (2011). STING is a direct innate immune sensor of cyclic di-GMP Nature 478: 515-18.

62.

Gehrke, N., Mertens C, Zillinger T, Wenzel J, Bald T, Zahn S, Tüting T, Hartmann G, Barchet W. (2013). Oxidative damage of DNA confers resistance to cytosolic nuclease TREX-1 degradation and potentiates STING-dependent immune sensing. Immunity 39: 482-

95.

63.

Sun L, Wu J, Du F, Chen X, Chen ZJ (2013). Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type 1 interferon pathway. Science 339: 786-91.

64.

Civril F, Deimling T, de Oliveria Mann CC, Ablasser A, Moldt M, Witte G, Hornung V, Hopfner KP. (2013). Structural mechanism of cytosolic DNA sensing by cGAS. Nature 498: 332-7.

65.

Ablasser, A, Goldeck M, Cavlar T, Deimling T, Witte G, Rohl I, Hopfner KP, Ludwig J, Hornung V. (2013) cGAS produces a 2’-5’linked cyclic dinucleotide second messenger that activates STING. Nature 498: 380-4.

66.

Li, XD, Wu, J, Gao D, Wang H, Sun L, Chen ZJ (2013). Pivotal roles of cGAS-cGAMP signaling in antiviral defense and immune adjuvant effects. Science 341: 1390-4.

4

Download