#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

RNA-seq Analysis of Host and Viral Gene Expression Highlights Interaction between Varicella Zoster Virus and Keratinocyte Differentiation


Varicella zoster virus (VZV) is the etiological agent of chickenpox and shingles, diseases characterized by epidermal skin blistering. Using a calcium-induced keratinocyte differentiation model we investigated the interaction between epidermal differentiation and VZV infection. RNA-seq analysis showed that VZV infection has a profound effect on differentiating keratinocytes, altering the normal process of epidermal gene expression to generate a signature that resembles patterns of gene expression seen in both heritable and acquired skin-blistering disorders. Further investigation by real-time PCR, protein analysis and electron microscopy revealed that VZV specifically reduced expression of specific suprabasal cytokeratins and desmosomal proteins, leading to disruption of epidermal structure and function. These changes were accompanied by an upregulation of kallikreins and serine proteases. Taken together VZV infection promotes blistering and desquamation of the epidermis, both of which are necessary to the viral spread and pathogenesis. At the same time, analysis of the viral transcriptome provided evidence that VZV gene expression was significantly increased following calcium treatment of keratinocytes. Using reporter viruses and immunohistochemistry we confirmed that VZV gene and protein expression in skin is linked with cellular differentiation. These studies highlight the intimate host-pathogen interaction following VZV infection of skin and provide insight into the mechanisms by which VZV remodels the epidermal environment to promote its own replication and spread.


Vyšlo v časopise: RNA-seq Analysis of Host and Viral Gene Expression Highlights Interaction between Varicella Zoster Virus and Keratinocyte Differentiation. PLoS Pathog 10(1): e32767. doi:10.1371/journal.ppat.1003896
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003896

Souhrn

Varicella zoster virus (VZV) is the etiological agent of chickenpox and shingles, diseases characterized by epidermal skin blistering. Using a calcium-induced keratinocyte differentiation model we investigated the interaction between epidermal differentiation and VZV infection. RNA-seq analysis showed that VZV infection has a profound effect on differentiating keratinocytes, altering the normal process of epidermal gene expression to generate a signature that resembles patterns of gene expression seen in both heritable and acquired skin-blistering disorders. Further investigation by real-time PCR, protein analysis and electron microscopy revealed that VZV specifically reduced expression of specific suprabasal cytokeratins and desmosomal proteins, leading to disruption of epidermal structure and function. These changes were accompanied by an upregulation of kallikreins and serine proteases. Taken together VZV infection promotes blistering and desquamation of the epidermis, both of which are necessary to the viral spread and pathogenesis. At the same time, analysis of the viral transcriptome provided evidence that VZV gene expression was significantly increased following calcium treatment of keratinocytes. Using reporter viruses and immunohistochemistry we confirmed that VZV gene and protein expression in skin is linked with cellular differentiation. These studies highlight the intimate host-pathogen interaction following VZV infection of skin and provide insight into the mechanisms by which VZV remodels the epidermal environment to promote its own replication and spread.


Zdroje

1. EckertRL, CrishJF, RobinsonNA (1997) The epidermal keratinocyte as a model for the study of gene regulation and cell differentiation. Physiol Rev 77: 397–424.

2. FuchsE, GreenH (1980) Changes in keratin gene expression during terminal differentiation of the keratinocyte. Cell 19: 1033–1042.

3. TaylorJM, StreetTL, HaoL, CopleyR, TaylorMS, et al. (2009) Dynamic and physical clustering of gene expression during epidermal barrier formation in differentiating keratinocytes. PLoS One 4: e7651.

4. BikleDD (2004) Vitamin D regulated keratinocyte differentiation. J Cell Biochem 92: 436–444.

5. BikleDD, NgD, TuCL, OdaY, XieZ (2001) Calcium- and vitamin D-regulated keratinocyte differentiation. Mol Cell Endocrinol 177: 161–171.

6. BlanpainC, FuchsE (2009) Epidermal homeostasis: a balancing act of stem cells in the skin. Nat Rev Mol Cell Biol 10: 207–217.

7. FessingMY, AtoyanR, ShanderB, MardaryevAN, BotchkarevVVJr, et al. (2010) BMP signaling induces cell-type-specific changes in gene expression programs of human keratinocytes and fibroblasts. J Invest Dermatol 130: 398–404.

8. HildebrandJ, RutzeM, WalzN, GallinatS, WenckH, et al. (2011) A comprehensive analysis of microRNA expression during human keratinocyte differentiation in vitro and in vivo. J Invest Dermatol 131: 20–29.

9. OvaereP, LippensS, VandenabeeleP, DeclercqW (2009) The emerging roles of serine protease cascades in the epidermis. Trends Biochem Sci 34: 453–463.

10. SextonCJ, NavsariaHA, LeighIM, PowellK (1992) Replication of varicella zoster virus in primary human keratinocytes. J Med Virol 38: 260–264.

11. CheX, ZerboniL, SommerMH, ArvinAM (2006) Varicella-zoster virus open reading frame 10 is a virulence determinant in skin cells but not in T cells in vivo. J Virol 80: 3238–3248.

12. CheX, ReicheltM, SommerMH, RajamaniJ, ZerboniL, et al. (2008) Functions of the ORF9-to-ORF12 gene cluster in varicella-zoster virus replication and in the pathogenesis of skin infection. J Virol 82: 5825–5834.

13. MoffatJF, ZerboniL, KinchingtonPR, GroseC, KaneshimaH, et al. (1998) Attenuation of the vaccine Oka strain of varicella-zoster virus and role of glycoprotein C in alphaherpesvirus virulence demonstrated in the SCID-hu mouse. J Virol 72: 965–974.

14. BarrandonY, GreenH (1985) Cell size as a determinant of the clone-forming ability of human keratinocytes. Proc Natl Acad Sci U S A 82: 5390–5394.

15. HenningsH, HolbrookKA (1983) Calcium regulation of cell-cell contact and differentiation of epidermal cells in culture. An ultrastructural study. Exp Cell Res 143: 127–142.

16. Huang daW, ShermanBT, LempickiRA (2009) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4: 44–57.

17. FuchsE, HG (1980) Changes in keratin gene expression during terminal differentiation of the keratinocyte. Cell 19: 1033–1042.

18. MollR, DivoM, LangbeinL (2008) The human keratins: biology and pathology. Histochem Cell Biol 129: 705–733.

19. ReicheltM, BradyJ, ArvinAM (2009) The replication cycle of varicella-zoster virus: analysis of the kinetics of viral protein expression, genome synthesis, and virion assembly at the single-cell level. J Virol 83: 3904–3918.

20. AndreiG, van den OordJ, FitenP, OpdenakkerG, De Wolf-PeetersC, et al. (2005) Organotypic epithelial raft cultures as a model for evaluating compounds against alphaherpesviruses. Antimicrob Agents Chemother 49: 4671–4680.

21. GarrodD, ChidgeyM (2008) Desmosome structure, composition and function. Biochim Biophys Acta 1778: 572–587.

22. OldakM, SmolaH, AumailleyM, RiveroF, PfisterH, et al. (2004) The human papillomavirus type 8 E2 protein suppresses beta4-integrin expression in primary human keratinocytes. J Virol 78: 10738–10746.

23. FurioL, HovnanianA (2011) When activity requires breaking up: LEKTI proteolytic activation cascade for specific proteinase inhibition. J Invest Dermatol 131: 2169–2173.

24. ChavanasS, BodemerC, RochatA, Hamel-TeillacD, AliM, et al. (2000) Mutations in SPINK5, encoding a serine protease inhibitor, cause Netherton syndrome. Nat Genet 25: 141–142.

25. CaubetC, JoncaN, BrattsandM, GuerrinM, BernardD, et al. (2004) Degradation of corneodesmosome proteins by two serine proteases of the kallikrein family, SCTE/KLK5/hK5 and SCCE/KLK7/hK7. J Invest Dermatol 122: 1235–1244.

26. CharestJL, JenningsJM, KingWP, KowalczykAP, GarciaAJ (2009) Cadherin-mediated cell-cell contact regulates keratinocyte differentiation. J Invest Dermatol 129: 564–572.

27. FuchsE, RaghavanS (2002) Getting under the skin of epidermal morphogenesis. Nat Rev Genet 3: 199–209.

28. TaylorSL, MoffatJF (2005) Replication of varicella-zoster virus in human skin organ culture. J Virol 79: 11501–11506.

29. SchelhaasM, JansenM, HaaseI, Knebel-MorsdorfD (2003) Herpes simplex virus type 1 exhibits a tropism for basal entry in polarized epithelial cells. J Gen Virol 84: 2473–2484.

30. MurakiR, IwasakiT, SataT, SatoY, KurataT (1996) Hair follicle involvement in herpes zoster: pathway of viral spread from ganglia to skin. Virchows Arch 428: 275–280.

31. ChenJJ, ZhuZ, GershonAA, GershonMD (2004) Mannose 6-phosphate receptor dependence of varicella zoster virus infection in vitro and in the epidermis during varicella and zoster. Cell 119: 915–926.

32. StorlieJ, JacksonW, HutchinsonJ, GroseC (2006) Delayed biosynthesis of varicella-zoster virus glycoprotein C: upregulation by hexamethylene bisacetamide and retinoic acid treatment of infected cells. J Virol 80: 9544–9556.

33. StorlieJ, CarpenterJE, JacksonW, GroseC (2008) Discordant varicella-zoster virus glycoprotein C expression and localization between cultured cells and human skin vesicles. Virology 382: 171–181.

34. JonesJO, ArvinAM (2006) Inhibition of the NF-kappaB pathway by varicella-zoster virus in vitro and in human epidermal cells in vivo. J Virol 80: 5113–5124.

35. JungJY, OhJH, KimYK, ShinMH, LeeD, et al. (2012) Acute UV irradiation increases heparan sulfate proteoglycan levels in human skin. J Korean Med Sci 27: 300–306.

36. HummelM, HudsonJB, LaiminsLA (1992) Differentiation-induced and constitutive transcription of human papillomavirus type 31b in cell lines containing viral episomes. J Virol 66: 6070–6080.

37. JohnsonAS, MaronianN, VieiraJ (2005) Activation of Kaposi's sarcoma-associated herpesvirus lytic gene expression during epithelial differentiation. J Virol 79: 13769–13777.

38. UittoJ, RichardG, McGrathJA (2007) Diseases of epidermal keratins and their linker proteins. Exp Cell Res 313: 1995–2009.

39. ParamioJM, CasanovaML, SegrellesC, MittnachtS, LaneEB, et al. (1999) Modulation of cell proliferation by cytokeratins K10 and K16. Mol Cell Biol 19: 3086–3094.

40. KochPJ, RoopDR (2004) The role of keratins in epidermal development and homeostasis–going beyond the obvious. J Invest Dermatol 123: x–xi.

41. ReicheltJ, FurstenbergerG, MaginTM (2004) Loss of keratin 10 leads to mitogen-activated protein kinase (MAPK) activation, increased keratinocyte turnover, and decreased tumor formation in mice. J Invest Dermatol 123: 973–981.

42. WallaceL, Roberts-ThompsonL, ReicheltJ (2012) Deletion of K1/K10 does not impair epidermal stratification but affects desmosomal structure and nuclear integrity. J Cell Sci 125: 1750–1758.

43. DescarguesP, DeraisonC, ProstC, FraitagS, Mazereeuw-HautierJ, et al. (2006) Corneodesmosomal cadherins are preferential targets of stratum corneum trypsin- and chymotrypsin-like hyperactivity in Netherton syndrome. J Invest Dermatol 126: 1622–1632.

44. BuschkeS, StarkHJ, CerezoA, Pratzel-WunderS, BoehnkeK, et al. (2011) A decisive function of transforming growth factor-beta/Smad signaling in tissue morphogenesis and differentiation of human HaCaT keratinocytes. Mol Biol Cell 22: 782–794.

45. JonesJO, ArvinAM (2005) Viral and cellular gene transcription in fibroblasts infected with small plaque mutants of varicella-zoster virus. Antiviral Res 68: 56–65.

46. PaladiniRD, TakahashiK, BravoNS, CoulombePA (1996) Onset of re-epithelialization after skin injury correlates with a reorganization of keratin filaments in wound edge keratinocytes: defining a potential role for keratin 16. J Cell Biol 132: 381–397.

47. MarkusA, GrigoryanS, SloutskinA, YeeMB, ZhuH, et al. (2011) Varicella-zoster virus (VZV) infection of neurons derived from human embryonic stem cells: direct demonstration of axonal infection, transport of VZV, and productive neuronal infection. J Virol 85: 6220–6233.

48. NiizumaT, ZerboniL, SommerMH, ItoH, HinchliffeS, et al. (2003) Construction of varicella-zoster virus recombinants from parent Oka cosmids and demonstration that ORF65 protein is dispensable for infection of human skin and T cells in the SCID-hu mouse model. J Virol 77: 6062–6065.

49. TischerBK, KauferBB, SommerM, WussowF, ArvinAM, et al. (2007) A self-excisable infectious bacterial artificial chromosome clone of varicella-zoster virus allows analysis of the essential tegument protein encoded by ORF9. J Virol 81: 13200–13208.

50. ErazoA, KinchingtonPR (2010) Varicella-Zoster Virus Open Reading Frame 66 Protein Kinase and Its Relationship to Alphaherpesvirus US3 Kinases. Curr Top Microbiol Immunol 342: 79–98.

51. TischerBK, von EinemJ, KauferB, OsterriederN (2006) Two-step red-mediated recombination for versatile high-efficiency markerless DNA manipulation in Escherichia coli. Biotechniques 40: 191–197.

52. OjehNO, FrameJD, NavsariaHA (2001) In vitro characterization of an artificial dermal scaffold. Tissue Eng 7: 457–472.

53. LangmeadB (2010) Aligning short sequencing reads with Bowtie. Curr Protoc Bioinformatics Chapter 11: Unit 11 17.

54. TrapnellC, PachterL, SalzbergSL (2009) TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25: 1105–1111.

55. LiH, HandsakerB, WysokerA, FennellT, RuanJ, et al. (2009) The Sequence Alignment/Map format and SAMtools. Bioinformatics 25: 2078–2079.

56. GentlemanRC, CareyVJ, BatesDM, BolstadB, DettlingM, et al. (2004) Bioconductor: open software development for computational biology and bioinformatics. Genome Biol 5: R80.

57. RobinsonMD, OshlackA (2010) A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol 11: R25.

58. McCarthyDJ, ChenY, SmythGK (2012) Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res 40: 4288–4297.

59. SaeedAI, BhagabatiNK, BraistedJC, LiangW, SharovV, et al. (2006) TM4 microarray software suite. Methods Enzymol 411: 134–193.

60. SaeedAI, SharovV, WhiteJ, LiJ, LiangW, et al. (2003) TM4: a free, open-source system for microarray data management and analysis. Biotechniques 34: 374–378.

61. RobinsonJT, ThorvaldsdottirH, WincklerW, GuttmanM, LanderES, et al. (2011) Integrative genomics viewer. Nat Biotechnol 29: 24–26.

62. 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.

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#