#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

IFI16 Restricts HSV-1 Replication by Accumulating on the HSV-1 Genome, Repressing HSV-1 Gene Expression, and Directly or Indirectly Modulating Histone Modifications


HSV-1, a ubiquitous human pathogen that establishes a life-long infection, has evolved several mechanisms to evade host immune detection and responses. However, it is still subject to regulation by cellular factors. Recently, a host nuclear protein, IFI16, was shown to be involved in the innate defense response to HSV-1 infection. Here, we provide the first evidence that IFI16 inhibits wild-type HSV-1 replication by repressing viral gene expression independent of its roles in the immune response. We show that IFI16 binds the HSV-1 genome at the transcription start sites of several HSV-1 genes. Using a permanently IFI16-negative cell line that we generated, we demonstrate that IFI16 reduces the association of important transcription factors. IFI16 also promotes global histone modifications by increasing the markers of repressive chromatin and decreasing the markers for activating chromatin on viral and cellular genes. These insights into the role of IFI16 in HSV-1 biology suggest that stabilization of IFI16 is an attractive avenue for antiviral drug development.


Vyšlo v časopise: 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(11): e32767. doi:10.1371/journal.ppat.1004503
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004503

Souhrn

HSV-1, a ubiquitous human pathogen that establishes a life-long infection, has evolved several mechanisms to evade host immune detection and responses. However, it is still subject to regulation by cellular factors. Recently, a host nuclear protein, IFI16, was shown to be involved in the innate defense response to HSV-1 infection. Here, we provide the first evidence that IFI16 inhibits wild-type HSV-1 replication by repressing viral gene expression independent of its roles in the immune response. We show that IFI16 binds the HSV-1 genome at the transcription start sites of several HSV-1 genes. Using a permanently IFI16-negative cell line that we generated, we demonstrate that IFI16 reduces the association of important transcription factors. IFI16 also promotes global histone modifications by increasing the markers of repressive chromatin and decreasing the markers for activating chromatin on viral and cellular genes. These insights into the role of IFI16 in HSV-1 biology suggest that stabilization of IFI16 is an attractive avenue for antiviral drug development.


Zdroje

1. Knipe DM, Howley PM, editors (2006) Fields' Virology. Philadelphia: Lippincott Williams & Wilkins. 3177 p.

2. StreileinJW, DanaMR, KsanderBR (1997) Immunity causing blindness: five different paths to herpes stromal keratitis. Immunol Today 18: 443–449.

3. WhitleyRJ, GnannJW (2002) Viral encephalitis: familiar infections and emerging pathogens. Lancet 359: 507–513.

4. WhitleyRJ, SoongSJ, LinnemanCJr, LiuC, PazinG, et al. (1982) Herpes simplex encephalitis. Clinical Assessment. JAMA 247: 317–320.

5. HonessRW, RoizmanB (1974) Regulation of herpesvirus macromolecular synthesis. I. Cascade regulation of the synthesis of three groups of viral proteins. J Virol 14: 8–19.

6. HonessRW, RoizmanB (1975) Proteins specified by herpes simplex virus. XIII. Glycosylation of viral polypeptides. J Virol 16: 1308–1326.

7. KwongAD, FrenkelN (1987) Herpes simplex virus-infected cells contain a function(s) that destabilizes both host and viral mRNAs. Proc Natl Acad Sci U S A 84: 1926–1930.

8. OroskarAA, ReadGS (1989) Control of mRNA stability by the virion host shutoff function of herpes simplex virus. J Virol 63: 1897–1906.

9. DauberB, PelletierJ, SmileyJR (2011) The herpes simplex virus 1 vhs protein enhances translation of viral true late mRNAs and virus production in a cell type-dependent manner. J Virol 85: 5363–5373.

10. HeB, ChouJ, BrandimartiR, MohrI, GluzmanY, et al. (1997) Suppression of the phenotype of gamma(1)34.5- herpes simplex virus 1: failure of activated RNA-dependent protein kinase to shut off protein synthesis is associated with a deletion in the domain of the alpha47 gene. J Virol 71: 6049–6054.

11. KentJR, ZengPY, AtanasiuD, GardnerJ, FraserNW, et al. (2004) During lytic infection herpes simplex virus type 1 is associated with histones bearing modifications that correlate with active transcription. J Virol 78: 10178–10186.

12. HerreraFJ, TriezenbergSJ (2004) VP16-dependent association of chromatin-modifying coactivators and underrepresentation of histones at immediate-early gene promoters during herpes simplex virus infection. J Virol 78: 9689–9696.

13. CliffeAR, KnipeDM (2008) Herpes simplex virus ICP0 promotes both histone removal and acetylation on viral DNA during lytic infection. J Virol 82: 12030–12038.

14. HancockMH, CliffeAR, KnipeDM, SmileyJR (2010) Herpes simplex virus VP16, but not ICP0, is required to reduce histone occupancy and enhance histone acetylation on viral genomes in U2OS osteosarcoma cells. J Virol 84: 1366–1375.

15. DeshmaneSL, FraserNW (1989) During latency, herpes simplex virus type 1 DNA is associated with nucleosomes in a chromatin structure. J Virol 63: 943–947.

16. WangQY, ZhouC, JohnsonKE, ColgroveRC, CoenDM, et al. (2005) Herpesviral latency-associated transcript gene promotes assembly of heterochromatin on viral lytic-gene promoters in latent infection. Proc Natl Acad Sci U S A 102: 16055–16059.

17. SwygertSG, PetersonCL (2014) Chromatin dynamics: Interplay between remodeling enzymes and histone modifications. Biochim Biophys Acta 1839: 728–736.

18. GuH, RoizmanB (2007) Herpes simplex virus-infected cell protein 0 blocks the silencing of viral DNA by dissociating histone deacetylases from the CoREST-REST complex. Proc Natl Acad Sci U S A 104: 17134–17139.

19. JohnsonKE, ChikotiL, ChandranB (2013) HSV-1 Infection Induces Activation and Subsequent Inhibition of the IFI16 and NLRP3 Inflammasomes. J Virol 87: 5005–5018.

20. OrzalliMH, DelucaNA, KnipeDM (2012) Nuclear IFI16 induction of IRF-3 signaling during herpesviral infection and degradation of IFI16 by the viral ICP0 protein. Proc Natl Acad Sci U S A 109: E3008–3017.

21. MaulGG, EverettRD (1994) The nuclear location of PML, a cellular member of the C3HC4 zinc-binding domain protein family, is rearranged during herpes simplex virus infection by the C3HC4 viral protein ICP0. J Gen Virol 75: 1223–1233.

22. MelroeGT, DeLucaNA, KnipeDM (2004) Herpes simplex virus 1 has multiple mechanisms for blocking virus-induced interferon production. J Virol 78: 8411–8420.

23. MelroeGT, SilvaL, SchafferPA, KnipeDM (2007) Recruitment of activated IRF-3 and CBP/p300 to herpes simplex virus ICP0 nuclear foci: Potential role in blocking IFN-beta induction. Virology 360: 305–321.

24. YokotaS, YokosawaN, KubotaT, SuzutaniT, YoshidaI, et al. (2001) Herpes simplex virus type 1 suppresses the interferon signaling pathway by inhibiting phosphorylation of STATs and janus kinases during an early infection stage. Virology 286: 119–124.

25. JohnsonKE, KnipeDM (2010) Herpes simplex virus-1 infection causes the secretion of a type I interferon-antagonizing protein and inhibits signaling at or before Jak-1 activation. Virology 396: 21–29.

26. JohnsonKE, SongB, KnipeDM (2008) Role for herpes simplex virus 1 ICP27 in the inhibition of type I interferon signaling. Virology 374: 487–494.

27. CheeAV, RoizmanB (2004) Herpes simplex virus 1 gene products occlude the interferon signaling pathway at multiple sites. J Virol 78: 4185–4196.

28. UnterholznerL, KeatingSE, BaranM, HoranKA, JensenSB, et al. (2010) IFI16 is an innate immune sensor for intracellular DNA. Nat Immunol 11: 997–1004.

29. BrazdaV, CoufalJ, LiaoJC, ArrowsmithCH (2012) Preferential binding of IFI16 protein to cruciform structure and superhelical DNA. Biochem Biophys Res Commun 422: 716–720.

30. TrapaniJA, BrowneKA, DawsonMJ, RamsayRG, EddyRL, et al. (1992) A novel gene constitutively expressed in human lymphoid cells is inducible with interferon-gamma in myeloid cells. Immunogenetics 36: 369–376.

31. TrapaniJA, DawsonM, ApostolidisVA, BrowneKA (1994) Genomic organization of IFI16, an interferon-inducible gene whose expression is associated with human myeloid cell differentiation: correlation of predicted protein domains with exon organization. Immunogenetics 40: 415–424.

32. JohnstoneRW, KerryJA, TrapaniJA (1998) The human interferon-inducible protein, IFI 16, is a repressor of transcription. J Biol Chem 273: 17172–17177.

33. JohnstoneRW, WeiW, GreenwayA, TrapaniJA (2000) Functional interaction between p53 and the interferon-inducible nucleoprotein IFI 16. Oncogene 19: 6033–6042.

34. KwakJC, OngusahaPP, OuchiT, LeeSW (2003) IFI16 as a negative regulator in the regulation of p53 and p21(Waf1). J Biol Chem 278: 40899–40904.

35. TawaraH, FujiuchiN, SironiJ, MartinS, AglipayJ, et al. (2008) Loss of p53-regulatory protein IFI16 induces NBS1 leading to activation of p53-mediated checkpoint by phosphorylation of p53 SER37. Front Biosci 13: 240–248.

36. OuchiM, OuchiT (2008) Role of IFI16 in DNA damage and checkpoint. Front Biosci 13: 236–239.

37. LiT, DinerBA, ChenJ, CristeaIM (2012) Acetylation modulates cellular distribution and DNA sensing ability of interferon-inducible protein IFI16. Proc Natl Acad Sci U S A 109: 10558–10563.

38. OrzalliMH, ConwellSE, BerriosC, DeCaprioJA, KnipeDM (2013) Nuclear interferon-inducible protein 16 promotes silencing of herpesviral and transfected DNA. Proc Natl Acad Sci U S A 110: E4492–4501.

39. KerurN, VeettilMV, Sharma-WaliaN, BotteroV, SadagopanS, 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.

40. AnsariMA, SinghVV, DuttaS, VeettilMV, DuttaD, 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.

41. JakobsenMR, BakRO, AndersenA, BergRK, JensenSB, et al. (2013) IFI16 senses DNA forms of the lentiviral replication cycle and controls HIV-1 replication. Proc Natl Acad Sci U S A 110: E4571–4580.

42. SinghVV, KerurN, BotteroV, DuttaS, ChakrabortyS, 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.

43. LiT, ChenJ, CristeaIM (2013) Human cytomegalovirus tegument protein pUL83 inhibits IFI16-mediated DNA sensing for immune evasion. Cell Host Microbe 14: 591–599.

44. Cuchet-LourencoD, AndersonG, SloanE, OrrA, EverettRD (2013) The viral ubiquitin ligase ICP0 is neither sufficient nor necessary for degradation of the cellular DNA sensor IFI16 during herpes simplex virus 1 infection. J Virol 87: 13422–13432.

45. GarianoGR, Dell'OsteV, BronziniM, GattiD, LuganiniA, et al. (2012) The intracellular DNA sensor IFI16 gene acts as restriction factor for human cytomegalovirus replication. PLoS Pathog 8: e1002498.

46. LivakKJ, SchmittgenTD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25: 402–408.

47. KalamvokiM, RoizmanB (2014) HSV-1 degrades, stabilizes, requires, or is stung by STING depending on ICP0, the US3 protein kinase, and cell derivation. Proc Natl Acad Sci U S A 111: E611–617.

48. FujiuchiN, AglipayJA, OhtsukaT, MaeharaN, SahinF, et al. (2004) Requirement of IFI16 for the maximal activation of p53 induced by ionizing radiation. J Biol Chem 279: 20339–20344.

49. EverettRD, YoungDF, RandallRE, OrrA (2008) STAT-1- and IRF-3-dependent pathways are not essential for repression of ICP0-null mutant herpes simplex virus type 1 in human fibroblasts. J Virol 82: 8871–8881.

50. LiXL, BoyanapalliM, WeihuaX, KalvakolanuDV, HasselBA (1998) Induction of interferon synthesis and activation of interferon-stimulated genes by liposomal transfection reagents. J Interferon Cytokine Res 18: 947–952.

51. AltinkilicB, BrandnerG (1988) Interferon inhibits herpes simplex virus-specific translation: a reinvestigation. J Gen Virol 69: 3107–3112.

52. MittnachtS, StraubP, KirchnerH, JacobsenH (1988) Interferon treatment inhibits onset of herpes simplex virus immediate-early transcription. Virology 164: 201–210.

53. HornungV, AblasserA, Charrel-DennisM, BauernfeindF, HorvathG, et al. (2009) AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature 458: 514–518.

54. MaliP, YangL, EsveltKM, AachJ, GuellM, et al. (2013) RNA-guided human genome engineering via Cas9. Science 339: 823–826.

55. VeresA, GosisBS, DingQ, CollinsR, RagavendranA, et al. (2014) Low incidence of off-target mutations in individual CRISPR-Cas9 and TALEN targeted human stem cell clones detected by whole-genome sequencing. Cell Stem Cell 15: 27–30.

56. SmithC, GoreA, YanW, Abalde-AtristainL, LiZ, et al. (2014) Whole-genome sequencing analysis reveals high specificity of CRISPR/Cas9 and TALEN-based genome editing in human iPSCs. Cell Stem Cell 15: 12–13.

57. WuX, ScottDA, KrizAJ, ChiuAC, HsuPD, et al. (2014) Genome-wide binding of the CRISPR endonuclease Cas9 in mammalian cells. Nat Biotechnol 32: 670–676.

58. FredrikssonS, GullbergM, JarviusJ, OlssonC, PietrasK, et al. (2002) Protein detection using proximity-dependent DNA ligation assays. Nat Biotechnol 20: 473–477.

59. LesterJT, DeLucaNA (2011) Herpes simplex virus 1 ICP4 forms complexes with TFIID and mediator in virus-infected cells. J Virol 85: 5733–5744.

60. SpencerCA, KruhlakMJ, JenkinsHL, SunX, Bazett-JonesDP (2000) Mitotic transcription repression in vivo in the absence of nucleosomal chromatin condensation. J Cell Biol 150: 13–26.

61. JinT, PerryA, JiangJ, SmithP, CurryJA, 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.

62. SampathP, DelucaNA (2008) Binding of ICP4, TATA-binding protein, and RNA polymerase II to herpes simplex virus type 1 immediate-early, early, and late promoters in virus-infected cells. J Virol 82: 2339–2349.

63. MaruzuruY, FujiiH, OyamaM, Kozuka-HataH, KatoA, et al. (2013) Roles of p53 in herpes simplex virus 1 replication. J Virol 87: 9323–9332.

64. JonesKA, TjianR (1985) Sp1 binds to promoter sequences and activates herpes simplex virus ‘immediate-early’ gene transcription in vitro. Nature 317: 179–182.

65. HeS, WeintraubSJ (1998) Stepwise recruitment of components of the preinitiation complex by upstream activators in vivo. Mol Cell Biol 18: 2876–2883.

66. HallDB, StruhlK (2002) The VP16 activation domain interacts with multiple transcriptional components as determined by protein-protein cross-linking in vivo. J Biol Chem 277: 46043–46050.

67. SteinSC, Falck-PedersenE (2012) Sensing adenovirus infection: activation of interferon regulatory factor 3 in RAW 264.7 cells. J Virol 86: 4527–4537.

68. ShahA, FarooqAV, TiwariV, KimMJ, ShuklaD (2010) HSV-1 infection of human corneal epithelial cells: receptor-mediated entry and trends of re-infection. Mol Vis 16: 2476–2486.

69. MonroeKM, YangZ, JohnsonJR, GengX, DoitshG, et al. (2014) IFI16 DNA sensor is required for death of lymphoid CD4 T cells abortively infected with HIV. Science 343: 428–432.

70. CristeaIM, MoormanNJ, TerhuneSS, CuevasCD, O'KeefeES, et al. (2010) Human cytomegalovirus pUL83 stimulates activity of the viral immediate-early promoter through its interaction with the cellular IFI16 protein. J Virol 84: 7803–7814.

71. JohnstoneRW, TrapaniJA (1999) Transcription and growth regulatory functions of the HIN-200 family of proteins. Mol Cell Biol 19: 5833–5838.

72. AmraniY, TlibaO, ChoubeyD, HuangCD, KrymskayaVP, et al. (2003) IFN-gamma inhibits human airway smooth muscle cell proliferation by modulating the E2F-1/Rb pathway. Am J Physiol Lung Cell Mol Physiol 284: L1063–1071.

73. LiuCJ, ChangE, YuJ, CarlsonCS, PrazakL, et al. (2005) The interferon-inducible p204 protein acts as a transcriptional coactivator of Cbfa1 and enhances osteoblast differentiation. J Biol Chem 280: 2788–2796.

74. LuanY, YuXP, XuK, DingB, YuJ, et al. (2007) The retinoblastoma protein is an essential mediator of osteogenesis that links the p204 protein to the Cbfa1 transcription factor thereby increasing its activity. J Biol Chem 282: 16860–16870.

75. SongLL, PonomarevaL, ShenH, DuanX, AlimirahF, et al. (2010) Interferon-inducible IFI16, a negative regulator of cell growth, down-regulates expression of human telomerase reverse transcriptase (hTERT) gene. PLoS One 5: e8569.

76. KangHJ, LeeMH, KangHL, KimSH, AhnJR, et al. (2014) Differential regulation of estrogen receptor alpha expression in breast cancer cells by metastasis-associated protein 1. Cancer Res 74: 1484–1494.

77. Dell'osteV, GattiD, GugliesiF, De AndreaM, BawadekarM, et al. (2014) Innate nuclear sensor IFI16 translocates into the cytoplasm during early stage of in vitro HCMV infection and is entrapped in the egressing virions during late stage. J Virol 88: 6970–6982.

78. BarberGN (2011) STING-dependent signaling. Nat Immunol 12: 929–930.

79. AglipayJA, LeeSW, OkadaS, FujiuchiN, OhtsukaT, et al. (2003) A member of the Pyrin family, IFI16, is a novel BRCA1-associated protein involved in the p53-mediated apoptosis pathway. Oncogene 22: 8931–8938.

80. DuanX, PonomarevaL, VeerankiS, ChoubeyD (2011) IFI16 induction by glucose restriction in human fibroblasts contributes to autophagy through activation of the ATM/AMPK/p53 pathway. PLoS One 6: e19532.

81. PanD, FloresO, UmbachJL, PesolaJM, BentleyP, et al. (2014) A Neuron-Specific Host MicroRNA Targets Herpes Simplex Virus-1 ICP0 Expression and Promotes Latency. Cell Host Microbe 15: 446–456.

82. YaoF, SchafferPA (1995) An activity specified by the osteosarcoma line U2OS can substitute functionally for ICP0, a major regulatory protein of herpes simplex virus type 1. J Virol 69: 6249–6258.

83. CassadyKA, GrossM, RoizmanB (1998) The herpes simplex virus US11 protein effectively compensates for the gamma1(34.5) gene if present before activation of protein kinase R by precluding its phosphorylation and that of the alpha subunit of eukaryotic translation initiation factor 2. J Virol 72: 8620–8626.

84. Diaz-LatoudC, DiazJJ, Fabre-JoncaN, KindbeiterK, MadjarJJ, et al. (1997) Herpes simplex virus Us11 protein enhances recovery of protein synthesis and survival in heat shock treated HeLa cells. Cell Stress Chaperones 2: 119–131.

85. BenboudjemaL, MulveyM, GaoY, PimplikarSW, MohrI (2003) Association of the herpes simplex virus type 1 Us11 gene product with the cellular kinesin light-chain-related protein PAT1 results in the redistribution of both polypeptides. J Virol 77: 9192–9203.

86. Duc DodonM, MikaelianI, SergeantA, GazzoloL (2000) The herpes simplex virus 1 Us11 protein cooperates with suboptimal amounts of human immunodeficiency virus type 1 (HIV-1) Rev protein to rescue HIV-1 production. Virology 270: 43–53.

87. XingJ, WangS, LinR, MossmanKL, ZhengC (2012) Herpes simplex virus 1 tegument protein US11 downmodulates the RLR signaling pathway via direct interaction with RIG-I and MDA-5. J Virol 86: 3528–3540.

88. LussignolM, QuevalC, Bernet-CamardMF, Cotte-LaffitteJ, BeauI, et al. (2013) The herpes simplex virus 1 Us11 protein inhibits autophagy through its interaction with the protein kinase PKR. J Virol 87: 859–871.

89. Freed-PastorWA, PrivesC (2011) Dissimilar DNA binding by p53 in normal and tumor-derived cells. Cell Cycle 10: 4207.

90. Chaigne-DelalandeB, DeuveL, ReuzeauE, BasoniC, LafargeD, et al. (2006) RhoGTPases and p53 are involved in the morphological appearance and interferon-alpha response of hairy cells. Am J Pathol 168: 562–573.

91. Van MeirEG, RoemerK, DiserensAC, KikuchiT, RempelSA, et al. (1995) Single cell monitoring of growth arrest and morphological changes induced by transfer of wild-type p53 alleles to glioblastoma cells. Proc Natl Acad Sci U S A 92: 1008–1012.

92. ZambettiGP, OlsonD, LabowM, LevineAJ (1992) A mutant p53 protein is required for maintenance of the transformed phenotype in cells transformed with p53 plus ras cDNAs. Proc Natl Acad Sci U S A 89: 3952–3956.

93. SinghVV, DuttaD, AnsariMA, DuttaS, ChandranB (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.

94. SobyS, LaursenRR, OstergaardL, MelchjorsenJ (2012) HSV-1-induced chemokine expression via IFI16-dependent and IFI16-independent pathways in human monocyte-derived macrophages. Herpesviridae 3: 6.

95. KnipeDM, SpangAE (1982) Definition of a series of stages in the association of two herpesviral proteins with the cell nucleus. J Virol 43: 314–324.

96. RosenkeK, FortunatoEA (2004) Bromodeoxyuridine-labeled viral particles as a tool for visualization of the immediate-early events of human cytomegalovirus infection. J Virol 78: 7818–7822.

97. TiscorniaG, SingerO, VermaIM (2006) Production and purification of lentiviral vectors. Nat Protoc 1: 241–245.

98. BoydKE, FarnhamPJ (1999) Coexamination of site-specific transcription factor binding and promoter activity in living cells. Mol Cell Biol 19: 8393–8399.

99. ShangY, HuX, DiRenzoJ, LazarMA, BrownM (2000) Cofactor dynamics and sufficiency in estrogen receptor-regulated transcription. Cell 103: 843–852.

100. KalamvokiM, RoizmanB (2010) Interwoven roles of cyclin D3 and cdk4 recruited by ICP0 and ICP4 in the expression of herpes simplex virus genes. J Virol 84: 9709–9717.

101. CookWJ, KramerMF, WalkerRM, BurwellTJ, HolmanHA, et al. (2004) Persistent expression of chemokine and chemokine receptor RNAs at primary and latent sites of herpes simplex virus 1 infection. Virol J 1: 5.

102. McNallyBA, TrgovcichJ, MaulGG, LiuY, ZhengP (2008) A role for cytoplasmic PML in cellular resistance to viral infection. PLoS One 3: e2277.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 11
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#