#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

CTCF and Rad21 Act as Host Cell Restriction Factors for Kaposi's Sarcoma-Associated Herpesvirus (KSHV) Lytic Replication by Modulating Viral Gene Transcription


Kaposi's sarcoma-associated herpesvirus (KSHV) is a human herpesvirus that causes Kaposi's sarcoma and is associated with the development of lymphoproliferative diseases. KSHV reactivation from latency and virion production is dependent on efficient transcription of over eighty lytic cycle genes and viral DNA replication. CTCF and cohesin, cellular proteins that cooperatively regulate gene expression and mediate long-range DNA interactions, have been shown to bind at specific sites in herpesvirus genomes. CTCF and cohesin regulate KSHV gene expression during latency and may also control lytic reactivation, although their role in lytic gene expression remains incompletely characterized. Here, we analyze the dynamic changes in CTCF and cohesin binding that occur during the process of KSHV viral reactivation and virion production by high resolution chromatin immunoprecipitation and deep sequencing (ChIP-Seq) and show that both proteins dissociate from viral genomes in kinetically and spatially distinct patterns. By utilizing siRNAs to specifically deplete CTCF and Rad21, a cohesin component, we demonstrate that both proteins are potent restriction factors for KSHV replication, with cohesin knockdown leading to hundred-fold increases in viral yield. High-throughput RNA sequencing was used to characterize the transcriptional effects of CTCF and cohesin depletion, and demonstrated that both proteins have complex and global effects on KSHV lytic transcription. Specifically, both proteins act as positive factors for viral transcription initially but subsequently inhibit KSHV lytic transcription, such that their net effect is to limit KSHV RNA accumulation. Cohesin is a more potent inhibitor of KSHV transcription than CTCF but both proteins are also required for efficient transcription of a subset of KSHV genes. These data reveal novel effects of CTCF and cohesin on transcription from a relatively small genome that resemble their effects on the cellular genome by acting as gene-specific activators of some promoters, but differ in acting as global negative regulators of transcription.


Vyšlo v časopise: CTCF and Rad21 Act as Host Cell Restriction Factors for Kaposi's Sarcoma-Associated Herpesvirus (KSHV) Lytic Replication by Modulating Viral Gene Transcription. PLoS Pathog 10(1): e32767. doi:10.1371/journal.ppat.1003880
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003880

Souhrn

Kaposi's sarcoma-associated herpesvirus (KSHV) is a human herpesvirus that causes Kaposi's sarcoma and is associated with the development of lymphoproliferative diseases. KSHV reactivation from latency and virion production is dependent on efficient transcription of over eighty lytic cycle genes and viral DNA replication. CTCF and cohesin, cellular proteins that cooperatively regulate gene expression and mediate long-range DNA interactions, have been shown to bind at specific sites in herpesvirus genomes. CTCF and cohesin regulate KSHV gene expression during latency and may also control lytic reactivation, although their role in lytic gene expression remains incompletely characterized. Here, we analyze the dynamic changes in CTCF and cohesin binding that occur during the process of KSHV viral reactivation and virion production by high resolution chromatin immunoprecipitation and deep sequencing (ChIP-Seq) and show that both proteins dissociate from viral genomes in kinetically and spatially distinct patterns. By utilizing siRNAs to specifically deplete CTCF and Rad21, a cohesin component, we demonstrate that both proteins are potent restriction factors for KSHV replication, with cohesin knockdown leading to hundred-fold increases in viral yield. High-throughput RNA sequencing was used to characterize the transcriptional effects of CTCF and cohesin depletion, and demonstrated that both proteins have complex and global effects on KSHV lytic transcription. Specifically, both proteins act as positive factors for viral transcription initially but subsequently inhibit KSHV lytic transcription, such that their net effect is to limit KSHV RNA accumulation. Cohesin is a more potent inhibitor of KSHV transcription than CTCF but both proteins are also required for efficient transcription of a subset of KSHV genes. These data reveal novel effects of CTCF and cohesin on transcription from a relatively small genome that resemble their effects on the cellular genome by acting as gene-specific activators of some promoters, but differ in acting as global negative regulators of transcription.


Zdroje

1. Ganem D (2007) Kaposi's sarcoma-associated herpesvirus. In: Knipe DM, Howley PM, editors. Fields Virology. 5 ed. Philadelphia: Wolters Kluwer/Lippincott Williams and Wilkins. pp. 2847–2888.

2. HadinotoV, ShapiroM, SunCC, Thorley-LawsonDA (2009) The Dynamics of EBV Shedding Implicate a Central Role for Epithelial Cells in Amplifying Viral Output. PLoS Pathog 5: e1000496.

3. HoshinoY, KatanoH, ZouP, HohmanP, MarquesA, et al. (2009) Long-term administration of valacyclovir reduces the number of Epstein-Barr virus (EBV)-infected B cells but not the number of EBV DNA copies per B cell in healthy volunteers. J Virol 83: 11857–11861.

4. MaSD, HegdeS, YoungKH, SullivanR, RajeshD, et al. (2011) A new model of Epstein-Barr virus infection reveals an important role for early lytic viral protein expression in the development of lymphomas. J Virol 85: 165–177.

5. MaSD, YuX, MertzJE, GumperzJE, ReinheimE, et al. (2012) An Epstein-Barr Virus (EBV) Mutant with Enhanced BZLF1 Expression Causes Lymphomas with Abortive Lytic EBV Infection in a Humanized Mouse Model. J Virol 86: 7976–7987.

6. StaskusKA, SunR, MillerG, RaczP, JaslowskiA, et al. (1999) Cellular Tropism and Viral Interleukin-6 Expression Distinguish Human Herpesvirus 8†Involvement in Kaposi's Sarcoma, Primary Effusion Lymphoma, and Multicentric Castleman's Disease. J Virol 73: 4181–4187.

7. MoorePS, BoshoffC, WeissRA, ChangY (1996) Molecular mimicry of human cytokine and cytokine response pathway genes by KSHV. Science 274: 1739–1744.

8. CesarmanE, MesriEA, GershengornMC (2000) Viral G protein-coupled receptor and Kaposi's sarcoma: a model of paracrine neoplasia? J Exp Med 191: 417–422.

9. MartinDF, KuppermannBD, WolitzRA, PalestineAG, LiH, et al. (1999) Oral ganciclovir for patients with cytomegalovirus retinitis treated with a ganciclovir implant. Roche Ganciclovir Study Group. N Engl J Med 340: 1063–1070.

10. AmelioAL, McAnanyPK, BloomDC (2006) A Chromatin Insulator-Like Element in the Herpes Simplex Virus Type 1 Latency-Associated Transcript Region Binds CCCTC-Binding Factor and Displays Enhancer-Blocking and Silencing Activities. J Virol 80: 2358–2368.

11. HoldorfMM, CooperSB, YamamotoKR, MirandaJJ (2011) Occupancy of chromatin organizers in the Epstein-Barr virus genome. Virology 415: 1–5.

12. KangH, WiedmerA, YuanY, RobertsonE, LiebermanPM (2011) Coordination of KSHV latent and lytic gene control by CTCF-cohesin mediated chromosome conformation. PLoS Pathog 7: e1002140.

13. TemperaI, LiebermanPM (2010) Chromatin organization of gammaherpesvirus latent genomes. Biochim Biophys Acta 1799: 236–245.

14. GauseM, SchaafCA, DorsettD (2008) Cohesin and CTCF: cooperating to control chromosome conformation? BioEssays 30: 715–718.

15. PhillipsJE, CorcesVG (2009) CTCF: master weaver of the genome. Cell 137: 1194–1211.

16. KimTH, AbdullaevZK, SmithAD, ChingKA, LoukinovDI, et al. (2007) Analysis of the vertebrate insulator protein CTCF-binding sites in the human genome. Cell 128: 1231–1245.

17. WanLB, PanH, HannenhalliS, ChengY, MaJ, et al. (2008) Maternal depletion of CTCF reveals multiple functions during oocyte and preimplantation embryo development. Development 135: 2729–2738.

18. XieX, MikkelsenTS, GnirkeA, Lindblad-TohK, KellisM, et al. (2007) Systematic discovery of regulatory motifs in conserved regions of the human genome, including thousands of CTCF insulator sites. Proc Natl Acad Sci U S A 104: 7145–7150.

19. FilippovaGN, FagerlieS, KlenovaEM, MyersC, DehnerY, et al. (1996) An exceptionally conserved transcriptional repressor, CTCF, employs different combinations of zinc fingers to bind diverged promoter sequences of avian and mammalian c-myc oncogenes. Mol Cell Biol 16: 2802–2813.

20. BellAC, WestAG, FelsenfeldG (1999) The protein CTCF is required for the enhancer blocking activity of vertebrate insulators. Cell 98: 387–396.

21. CuddapahS, JothiR, SchonesDE, RohTY, CuiK, et al. (2009) Global analysis of the insulator binding protein CTCF in chromatin barrier regions reveals demarcation of active and repressive domains. Genome Res 19: 24–32.

22. DorsettD, MerkenschlagerM (2013) Cohesin at active genes: a unifying theme for cohesin and gene expression from model organisms to humans. Curr Opin Cell Biol 25: 327–333.

23. SchaafCA, KwakH, KoenigA, MisulovinZ, GoharaDW, et al. (2013) Genome-wide control of RNA polymerase II activity by cohesin. PLoS Genet 9: e1003382.

24. ChenHS, WikramasingheP, ShoweL, LiebermanPM (2012) Cohesins Repress Kaposi's Sarcoma-Associated Herpesvirus Immediate Early Gene Transcription during Latency. J Virol 86: 9454–9464.

25. KangH, LiebermanPM (2009) Cell cycle control of Kaposi's sarcoma-associated herpesvirus latency transcription by CTCF-cohesin interactions. J Virol 83: 6199–6210.

26. StedmanW, KangH, LinS, KissilJL, BartolomeiMS, et al. (2008) Cohesins localize with CTCF at the KSHV latency control region and at cellular c-myc and H19/Igf2 insulators. EMBO J 27: 654–666.

27. MyoungJ, GanemD (2011) Generation of a doxycycline-inducible KSHV producer cell line of endothelial origin: maintenance of tight latency with efficient reactivation upon induction. J Virol Methods 174: 12–21.

28. BruloisKF, ChangH, LeeAS, EnsserA, WongLY, et al. (2012) Construction and manipulation of a new Kaposi's sarcoma-associated herpesvirus bacterial artificial chromosome clone. J Virol 86: 9708–9720.

29. AndersS, HuberW (2010) Differential expression analysis for sequence count data. Genome Biol 11: R106.

30. NixDA, CourdySJ, BoucherKM (2008) Empirical methods for controlling false positives and estimating confidence in ChIP-Seq peaks. BMC Bioinformatics 9: 523.

31. ThorvaldsdottirH, RobinsonJT, MesirovJP (2013) Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform 14: 178–192.

32. RossettoCC, PariG (2012) KSHV PAN RNA associates with demethylases UTX and JMJD3 to activate lytic replication through a physical interaction with the virus genome. PLoS Pathog 8: e1002680.

33. ZiebarthJD, BhattacharyaA, CuiY (2013) CTCFBSDB 2.0: a database for CTCF-binding sites and genome organization. Nucleic Acids Res 41: D188–D194.

34. SchmidtD, SchwaliePC, Ross-InnesCS, HurtadoA, BrownGD, et al. (2010) A CTCF-independent role for cohesin in tissue-specific transcription. Genome Res 20: 578–588.

35. RubioED, ReissDJ, WelcshPL, DistecheCM, FilippovaGN, et al. (2008) CTCF physically links cohesin to chromatin. Proceedings of the National Academy of Sciences 105: 8309–8314.

36. WendtKS, YoshidaK, ItohT, BandoM, KochB, et al. (2008) Cohesin mediates transcriptional insulation by CCCTC-binding factor. Nature 451: 796–801.

37. BlencoweBJ, AhmadS, LeeLJ (2009) Current-generation high-throughput sequencing: deepening insights into mammalian transcriptomes. Genes Dev 23: 1379–1386.

38. DorsettD, StromL (2012) The ancient and evolving roles of cohesin in gene expression and DNA repair. Curr Biol 22: R240–250.

39. FayA, MisulovinZ, LiJ, SchaafCA, GauseM, et al. (2011) Cohesin selectively binds and regulates genes with paused RNA polymerase. Curr Biol 21: 1624–1634.

40. RhodesJM, BentleyFK, PrintCG, DorsettD, MisulovinZ, et al. (2010) Positive regulation of c-Myc by cohesin is direct, and evolutionarily conserved. Dev Biol 344: 637–649.

41. AmonW, BinneUK, BryantH, JenkinsPJ, KarsteglCE, et al. (2004) Lytic cycle gene regulation of Epstein-Barr virus. J Virol 78: 13460–13469.

42. Mavromara-NazosP, RoizmanB (1987) Activation of herpes simplex virus 1 gamma 2 genes by viral DNA replication. Virology 161: 593–598.

43. ChangJ, GanemD (2000) On the control of late gene expression in Kaposi's sarcoma-associated herpesvirus (human herpesvirus-8). J Gen Virol 81: 2039–2047.

44. TothZ, BruloisKF, WongLY, LeeHR, ChungB, et al. (2012) Negative elongation factor-mediated suppression of RNA polymerase II elongation of Kaposi's sarcoma-associated herpesvirus lytic gene expression. J Virol 86: 9696–9707.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 1
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#