#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Herpesvirus Genome Recognition Induced Acetylation of Nuclear IFI16 Is Essential for Its Cytoplasmic Translocation, Inflammasome and IFN-β Responses


Herpesviruses establish a latent infection in the nucleus of specific cells and reactivation results in the nuclear viral dsDNA replication and infectious virus production. Host innate responses are initiated by the presence of viral genomes and their products, and nucleus associated IFI16 protein has recently emerged as an innate DNA sensor regulating inflammatory cytokines and type I interferon (IFN) production. IFI16 recognizes the herpesvirus genomes (KSHV, EBV, and HSV-1) in the nucleus resulting in the formation of the IFI16-ASC-Caspase-1 inflammasome complex and IL-1β production. HSV-1 genome recognition by IFI16 in the nucleus also leads to STING activation in the cytoplasm and IFN-β production. However, how IFI16 initiates inflammasome assembly and activates STING in the cytoplasm after nuclear recognition of viral genome are not known. We show that herpesvirus genome recognition in the nucleus by IFI16 leads to interaction with histone acetyltransferase-p300 and IFI16 acetylation which is essential for inflammasome assembly in the nucleus and cytoplasmic translocation, activation of STING in the cytoplasm and IFN-β production. These studies provide insight into a common molecular mechanism for the innate inflammasome assembly and STING activation response pathways that result in IL-1β and IFN-β production, respectively.


Vyšlo v časopise: Herpesvirus Genome Recognition Induced Acetylation of Nuclear IFI16 Is Essential for Its Cytoplasmic Translocation, Inflammasome and IFN-β Responses. PLoS Pathog 11(7): e32767. doi:10.1371/journal.ppat.1005019
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1005019

Souhrn

Herpesviruses establish a latent infection in the nucleus of specific cells and reactivation results in the nuclear viral dsDNA replication and infectious virus production. Host innate responses are initiated by the presence of viral genomes and their products, and nucleus associated IFI16 protein has recently emerged as an innate DNA sensor regulating inflammatory cytokines and type I interferon (IFN) production. IFI16 recognizes the herpesvirus genomes (KSHV, EBV, and HSV-1) in the nucleus resulting in the formation of the IFI16-ASC-Caspase-1 inflammasome complex and IL-1β production. HSV-1 genome recognition by IFI16 in the nucleus also leads to STING activation in the cytoplasm and IFN-β production. However, how IFI16 initiates inflammasome assembly and activates STING in the cytoplasm after nuclear recognition of viral genome are not known. We show that herpesvirus genome recognition in the nucleus by IFI16 leads to interaction with histone acetyltransferase-p300 and IFI16 acetylation which is essential for inflammasome assembly in the nucleus and cytoplasmic translocation, activation of STING in the cytoplasm and IFN-β production. These studies provide insight into a common molecular mechanism for the innate inflammasome assembly and STING activation response pathways that result in IL-1β and IFN-β production, respectively.


Zdroje

1. Ganem D (2007) Kaposi’s sarcoma-associated herpesvirus. In Field’s Virology, Fifth Edition, Knipe DM, Howley PM, eds. (Philadelphia, PA: Lippincott Williams & Wilkins).

2. Krishnan HH, Naranatt PP, Smith MS, Zeng L, et al. (2004) Concurrent expression of latent and a limited number of lytic genes with immune modulation and antiapoptotic function by Kaposi’s sarcoma-associated herpesvirus early during infection of primary endothelial and fibroblast cells and subsequent decline of lytic gene expression. J Virol 78: 3601–3620. 15016882

3. Kawai T, Akira S (2009) The roles of TLRs, RLRs and NLRs in pathogen recognition. Internat Immunol 21: 317–337.

4. Mogensen TH (2009) Pathogen recognition and inflammatory signaling in innate immune defenses. Clin Microbiol Rev 2: 240–273.

5. Fernandes-Alnemri T, Yu JW, Datta P, Wu J, Alnemri ES (2009) AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature 458: 509–513. doi: 10.1038/nature07710 19158676

6. Sharma-Walia N, Paul AG, Bottero V, Sadagopan S, Veettil MV, et al. (2010) Kaposi’s sarcoma associated herpes virus (KSHV) induced COX-2: a key factor in latency, inflammation, angiogenesis, cell survival and invasion. PLoS Pathog 6: e1000777. doi: 10.1371/journal.ppat.1000777 20169190

7. Martinon F, Burns K, Tschopp J (2002) The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell 10: 417–426. 12191486

8. Rathinam VAK, Fitzgerald KA (2011) Innate immune sensing of DNA viruses. Virology 411: 153–162. doi: 10.1016/j.virol.2011.02.003 21334037

9. Schroder K, Tschopp J (2010) The inflammasomes. Cell 140: 821–832. doi: 10.1016/j.cell.2010.01.040 20303873

10. Arend WP, Palmer G, Gabay C (2008) IL-1, IL-18, and IL-33 families of cytokines. Immunol Reviews 223: 20–38.

11. Kerur N, Veettil MV, Sharma-Walia N, Bottero V, Sadagopan S, et al. (2011) IFI16 acts as a nuclear pathogen sensor to induce the inflammasome in response to Kaposi Sarcoma-associated herpesvirus infection. Cell Host Microbe 9: 363–375. doi: 10.1016/j.chom.2011.04.008 21575908

12. Singh VV, Kerur N, Bottero V, Dutta S, Chakraborty S, et al. (2013) Kaposi’s sarcoma-associated herpesvirus latency in endothelial and B cells activates interferon gamma-inducible protein 16 (IFI16) mediated inflammasomes. J Virol 87: 4417–4431. doi: 10.1128/JVI.03282-12 23388709

13. Ansari MA, Singh VV, Dutta S, Veettil MV, Dutta D, et al. (2013) Constitutive interferon-inducible protein 16-inflammasome activation during Epstein-Barr virus latency I, II, and III in B and epithelial cells. J Virol 87: 8606–8623. doi: 10.1128/JVI.00805-13 23720728

14. Johnson KE, Chikoti L, Chandran B (2013) HSV-1 infection induces activation and subsequent inhibition of the IFI16 and NLRP3 inflammasomes. J Virol 87: 5005–5018. doi: 10.1128/JVI.00082-13 23427152

15. Li T, Diner BA, Chen J, Cristea IM (2012) Acetylation modulates cellular redistribution and DNA sensing ability of interferon-inducible protein IFI16. Proc Natl Acad Sci USA 109: 10558–10563. doi: 10.1073/pnas.1203447109 22691496

16. Orzalli MH, DeLuca NA, Knipe DM (2012) Nuclear IFI16 induction of IRF-3 signaling during herpesviral infection and degradation of IFI16 by the viral ICP0 protein. Proc Natl Acad Sci USA 109: E3008–E3017. doi: 10.1073/pnas.1211302109 23027953

17. Sathish N, Yuan Y (2011) Evasion and subversion of interferon-mediated antiviral immunity by Kaposi’s sarcoma-associated herpesvirus: an overview. J Virol 85: 10934–10944. doi: 10.1128/JVI.00687-11 21775463

18. Raghu H, Sharma-Walia N, Veettil MV, Sadagopan S, Chandran B (2009) Kaposi's Sarcoma-associated herpesvirus utilizes an actin polymerization-dependent macropinocytic pathway to enter human dermal microvascular endothelial and human umbilical vein endothelial cells. J Virol 83: 4895–4911. doi: 10.1128/JVI.02498-08 19279100

19. Lengyel P, Liu CJ (2010) The p200 family protein p204 as a modulator of cell proliferation and differentiation: a brief survey. Cell Mol Life Sci 67: 335–340. doi: 10.1007/s00018-009-0195-z 19921484

20. Lu B, Wang H, Andersson U, Tracey KJ (2013) Regulation of HMGB1 release by inflammasomes. Protein Cell 4: 163–167. doi: 10.1007/s13238-012-2118-2 23483477

21. Unterholzner L, Keating SS, Baran M, Horan KA, Jensen SB, et al. (2010) IFI16 is an innate immune sensor for intracellular DNA. Nat Immunol 11: 997–1004. doi: 10.1038/ni.1932 20890285

22. Johnson KE, Bottero V, Flaherty S, Dutta S, Singh VV, et al. (2014) IFI16 Restricts HSV-1 replication by accumulating on the HSV-1 genome, repressing HSV-1 gene expression, and directly or indirectly modulating histone modifications. PLoS Pathog 10: e1004503. doi: 10.1371/journal.ppat.1004503 25375629

23. Jin T, Perry A, Jiang J, Smith P, Curry JA, et al. (2012) Structures of the HIN domain: DNA complexes reveal ligand binding and activation mechanisms of the AIM2 inflammasome and IFI16 receptor. Immunity 36: 561–571. doi: 10.1016/j.immuni.2012.02.014 22483801

24. Brazda V, Coufal J, Liao JC, Arrowsmith CH (2012) Preferential binding of IFI16 protein to cruciform structure and superhelical DNA. Biochem Biophys Res Commun 422: 716–720. doi: 10.1016/j.bbrc.2012.05.065 22618232

25. Lieberman PM (2013) Keeping it quiet: chromatin control of gammaherpesvirus latency. Nat. Revi. Microbio 11: 863–875

26. Morrone SR, Wang T, Constantoulakis LM, Hooy RM, Delannoy MJ et al. (2013) Cooperative assembly of IFI16 filaments on dsDNA provides insights into host defense strategy. Proc Natl Acad Sci USA, 111: E62–E71. doi: 10.1073/pnas.1313577111 24367117

27. Wang X, Moore SC, Laszckzak M, Ausio J (2000) Acetylation increases the α-helical content of the histone tails of the nucleosome. J Biol Chem 275: 35013–35020. 10938086

28. Lu F, Day L, Gao SJ, Lieberman PM (2006) Acetylation of the latency-associated nuclear antigen regulates repression of Kaposi’s sarcoma-associated herpesvirus lytic transcription. J Virol 80: 5273–5282. 16699007

29. Sun C, Schattgen SA, Pisitkun JP, Hilterbrand AT, Wang LJ, et al. (2015) Evasion of innate cytosolic DNA sensing by a gammaherpesvirus facilitates establishment of latent infection. J Immunol 194: 1819–1831. doi: 10.4049/jimmunol.1402495 25595793

30. Horan KA, Hansen K, Jacobsen MR, Holm CK, Søby S, et al. (2013) Proteasomal degradation of herpes simplex virus capsids in macrophages releases DNA to the cytosol for recognition by DNA sensors. J Immunol 190: 2311–2319. doi: 10.4049/jimmunol.1202749 23345332

31. Qu y, Misaghi S, Izrael-Tomasevic A, Newton K, Gilmour LL, et al. (2012) Phosphorylation of NLRC4 is critical for inflammasome activation. Nature 490: 539–543. doi: 10.1038/nature11429 22885697

32. Hara H, Tsuchiya K, Kawamura I, Fang R, Hernandez-Cuellar E, et al. (2013) Phosphorylation of the adaptor ASC acts as a molecular switch that controls the formation of speck-like aggregates and inflammasome activity. Nat Immunol 14: 1247–1255. doi: 10.1038/ni.2749 24185614

33. Dell'Oste V, Gatti D, Gugliesi F, De Andrea M, Bawadekar M, et al. (2014) Innate nuclear sensor IFI16 translocates virions during the late stage infection and is entrapped in the egressing of in vitro human cytomegalovirus into the cytoplasm during the early stage. J Virol 88: 6970–6982. doi: 10.1128/JVI.00384-14 24696486

34. Singh VV, Dutta D, Ansari MA, Dutta S, Chandran B (2014) Kaposi’s sarcoma-associated herpesvirus induces the ATM and H2AX DNA damage response early during de novo infection of primary endothelial cells, which play roles in latency establishment. J Virol 88: 2821–2834. doi: 10.1128/JVI.03126-13 24352470

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 7
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#