RNA-seq analysis in equine papillomavirus type 2-positive carcinomas identifies affected pathways and potential cancer markers as well as viral gene expression and splicing events Free

Abstract

Equine papillomavirus type 2 (EcPV2) was discovered only recently, but it is found consistently in the context of genital squamous cell carcinomas (SCCs). Since neither cell cultures nor animal models exist, the characterization of this potential disease agent relies on the analysis of patient materials. To analyse the host and viral transcriptome in EcPV2-affected horses, genital tissue samples were collected from horses with EcPV2-positive lesions as well as from healthy EcPV2-negative horses. It was determined by RNA-seq analysis that there were 1957 differentially expressed (DE) host genes between the SCC and control samples. These genes were most abundantly related to DNA replication, cell cycle, extracellular matrix (ECM)–receptor interaction and focal adhesion. By comparison to other cancer studies, MMP1 and IL8 appeared to be potential marker genes for the development of SCCs. Analysis of the viral reads revealed the transcriptional activity of EcPV2 in all SCC samples. While few reads mapped to the structural viral genes, the majority of reads mapped to the non-structural early (E) genes, in particular to E6, E7 and E2/E4. Within these reads a distinct pattern of splicing events, which are essential for the expression of different genes in PV infections, was observed. Additionally, in one sample the integration of EcPV2 DNA into the host genome was detected by DNA-seq and confirmed by PCR. In conclusion, while host MMP1 and IL8 expression and the presence of EcPV2 may be useful markers in genital SCCs, further research on EcPV2-related pathomechanisms may focus on cell cycle-related genes, the viral genes E6, E7 and E2/E4, and integration events.

Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001267
2019-05-14
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/jgv/100/6/985.html?itemId=/content/journal/jgv/10.1099/jgv.0.001267&mimeType=html&fmt=ahah

References

  1. Howley PM, Schiller JT, Lowy DR. Papillomaviruses. In Fields Virology, 6th ed. Philadelphia: Wolters Kluwer / Lippincott Williams & Wilkins; 2013 pp 1662–1703
    [Google Scholar]
  2. ZurHausen H. Papillomaviruses causing cancer: evasion from host-cell control in early events in carcinogenesis. J Natl Cancer Inst 2000; 92:690–698
    [Google Scholar]
  3. Sykora S, Brandt S. Papillomavirus infection and squamous cell carcinoma in horses. Vet J 2017; 223:48–54 [View Article]
    [Google Scholar]
  4. Scase T, Brandt S, Kainzbauer C, Sykora S, Bijmholt S et al. Equus caballus papillomavirus-2 (EcPV-2): an infectious cause for equine genital cancer?. Equine Vet J 2010; 42:738–745 [View Article]
    [Google Scholar]
  5. Lange CE, Tobler K, Lehner A, Grest P, Welle MM et al. EcPV2 DNA in equine papillomas and in situ and invasive squamous cell carcinomas supports papillomavirus etiology. Vet Pathol 2013; 50:686–692 [View Article]
    [Google Scholar]
  6. Bogaert L, Willemsen A, Vanderstraeten E, Bracho MA, De Baere C et al. EcPV2 DNA in equine genital squamous cell carcinomas and normal genital mucosa. Vet Microbiol 2012; 158:33–41 [View Article]
    [Google Scholar]
  7. Knight CG, Munday JS, Rosa BV, Kiupel M. Persistent, widespread papilloma formation on the penis of a horse: a novel presentation of equine papillomavirus type 2 infection. Vet Dermatol 2011; 22:570–574 [View Article]
    [Google Scholar]
  8. Sykora S, Samek L, Schönthaler K, Palm F, Borzacchiello G et al. EcPV-2 is transcriptionally active in equine SCC but only rarely detectable in swabs and semen from healthy horses. Vet Microbiol Elsevier B.V 2012; 158:194–198 [View Article]
    [Google Scholar]
  9. Lange CE, Tobler K, Ackermann M, Favrot C. Identification of two novel equine papillomavirus sequences suggests three genera in one cluster. Vet Microbiol Elsevier B.V 2011; 149:85–90 [View Article]
    [Google Scholar]
  10. Knight CG, Dunowska M, Munday JS, Peters-Kennedy J, Rosa BV. Comparison of the levels of Equus caballus papillomavirus type 2 (EcPV-2) DNA in equine squamous cell carcinomas and non-cancerous tissues using quantitative PCR. Vet Microbiol Elsevier B.V 2013; 166:257–262 [View Article]
    [Google Scholar]
  11. Newkirk KM, Hendrix DVH, Anis EA, Rohrbach BW, Ehrhart EJ et al. Detection of papillomavirus in equine periocular and penile squamous cell carcinoma. J Vet Diagn Invest 2014; 26:131–135 [View Article]
    [Google Scholar]
  12. van den Top JGB, Harkema L, Lange C, Ensink JM, van de Lest CHA et al. Expression of p53, Ki67, EcPV2- and EcPV3 DNA, and viral genes in relation to metastasis and outcome in equine penile and preputial squamous cell carcinoma. Equine Vet J 2015; 47:188–195 [View Article]
    [Google Scholar]
  13. Zhu KW, Affolter VK, Gaynor AM, Dela Cruz FN, Pesavento PA. Equine genital squamous cell carcinoma. Vet Pathol 2015; 52:1067–1072 [View Article]
    [Google Scholar]
  14. Van den Top JGB, de Heer N, Klein WR, Ensink JM. Penile and preputial squamous cell carcinoma in the horse: a retrospective study of treatment of 77 affected horses. Equine Vet J Blackwell Publishing Ltd 2008; 40:533–537 [View Article]
    [Google Scholar]
  15. Mair TS, Walmsley JP, Phillips TJ. Surgical treatment of 45 horses affected by squamous cell carcinoma of the penis and prepuce. Equine Vet J Blackwell Publishing Ltd 2000; 32:406–410 [View Article]
    [Google Scholar]
  16. Sykora S, Jindra C, Hofer M, Steinborn R, Brandt S. Equine papillomavirus type 2: an equine equivalent to human papillomavirus 16?. Vet J Elsevier Ltd 2017; 225:3–8 [View Article]
    [Google Scholar]
  17. McBride AA, Warburton A. The role of integration in oncogenic progression of HPV-associated cancers. PLoS Pathog Public Library of Science 2017; 13:e1006211 [View Article]
    [Google Scholar]
  18. Edgar R, Domrachev M, Lash AE. Gene expression Omnibus: NCBI gene expression and hybridization array data Repository. Nucleic Acids Research 20011–4
    [Google Scholar]
  19. Nakajima K, Kaneko T, Aizu T, Nakano H, Matsuzaki Y et al. Signet-ring cutaneous squamous cell carcinoma arising on the back of the finger. Case Rep Dermatol Karger Publishers 2013; 5:215–218 [View Article]
    [Google Scholar]
  20. Espinosa de los Monteros A, Aguirre-Sanceledonio M, Ramírez GA, Castro P, Rodríguez F. Signet-ring squamous cell carcinoma in a dog. Vet Rec 2003; 153:90–92 [View Article]
    [Google Scholar]
  21. Demellawy DE, Onuma K, Alowami S. Signet ring squamous cell carcinoma-the forgotten variant: case report and review of the literature. J Cutan Pathol John Wiley & Sons, Ltd 2011; 38:306–308 [View Article]
    [Google Scholar]
  22. Mine KL, Shulzhenko N, Yambartsev A, Rochman M, Sanson GFO et al. Gene network reconstruction reveals cell cycle and antiviral genes as major drivers of cervical cancer. Nat Commun Nature Publishing Group 2013; 4:342 [View Article]
    [Google Scholar]
  23. Yuan ZQ, Nicolson L, Marchetti B, Gault EA, Campo MS et al. Transcriptional changes induced by bovine papillomavirus type 1 in equine fibroblasts. J Virol American Society for Microbiology 2008; 82:6481–6491 [View Article]
    [Google Scholar]
  24. Santegoets LAM, van Baars R, Terlou A, Heijmans-Antonissen C, Swagemakers SMA et al. Different DNA damage and cell cycle checkpoint control in low- and high-risk human papillomavirus infections of the vulva. Int J Cancer John Wiley & Sons 2012; 130:2874–2885 [View Article]
    [Google Scholar]
  25. Clarke B, Chetty R. Cell cycle aberrations in the pathogenesis of squamous cell carcinoma of the uterine cervix. Gynecol Oncol 2001; 82:238–246 [View Article]
    [Google Scholar]
  26. Moody CA, Laimins LA. Human papillomavirus oncoproteins: pathways to transformation. Nat Rev Cancer Nature Publishing Group 2010; 10:550–560 [View Article]
    [Google Scholar]
  27. Yim EK, Park JS. The role of HPV E6 and E7 oncoproteins in HPV-associated cervical carcinogenesis. Cancer Res Treat Korean Cancer Association 2005; 37:319–324 [View Article]
    [Google Scholar]
  28. Nigg EA. Cyclin-dependent protein kinases: key regulators of the eukaryotic cell cycle. Bioessays Wiley-Blackwell 1995; 17:471–480 [View Article]
    [Google Scholar]
  29. Brenna SMF, Syrjänen KJ. Regulation of Cell Cycles Is of Key Importance in Human Papillomavirus (Hpv)-Associated Cervical Carcinogenesis. Sao Paulo Medical Journal 121, 5th ed. Associação Paulista de Medicina; 2003 pp 128–132
    [Google Scholar]
  30. Bochman ML, Schwacha A. The Mcm complex: unwinding the mechanism of a replicative helicase. Microbiol Mol Biol Rev 2009; 73:652–683 [View Article]
    [Google Scholar]
  31. Griffin H, Soneji Y, Van Baars R, Arora R, Jenkins D et al. Stratification of HPV-induced cervical pathology using the virally encoded molecular marker E4 in combination with p16 or MCM. Mod Pathol 2015; 28:977–993 [View Article]
    [Google Scholar]
  32. von Knebel Doeberitz M, Reuschenbach M, Schmidt D, Bergeron C. Biomarkers for cervical cancer screening: the role of p16INK4a to highlight transforming HPV infections. Expert Rev Proteomics Taylor & Francis 2012; 9:149–163 [View Article]
    [Google Scholar]
  33. Lu P, Weaver VM, Werb Z. The extracellular matrix: a dynamic niche in cancer progression. J Cell Biol Rockefeller University Press 2012; 196:395–406 [View Article]
    [Google Scholar]
  34. Gérard C, Goldbeter A. The balance between cell cycle arrest and cell proliferation: control by the extracellular matrix and by contact inhibition. Interface Focus 2014; 4:20130075–5 [View Article]
    [Google Scholar]
  35. Ben Khalifa Y, Teissier S, Tan MKM, Phan QT, Daynac M et al. The human papillomavirus E6 oncogene represses a cell adhesion pathway and disrupts focal adhesion through degradation of TAp63β upon transformation. Lambert P, editor PLoS Pathog Public Library of Science 2011; 7:e1002256 [View Article]
    [Google Scholar]
  36. Tong X, Howley PM. The bovine papillomavirus E6 oncoprotein interacts with paxillin and disrupts the actin cytoskeleton. Proc Natl Acad Sci USA National Academy of Sciences 1997; 94:4412–4417 [View Article]
    [Google Scholar]
  37. Sarode GS, Sarode SC. E6 oncoprotein interaction with paxillin and FAK. Oral Oncol 2014; 50:e17 [View Article]
    [Google Scholar]
  38. Holloway A, Storey A. A conserved C-terminal sequence of high-risk cutaneous beta-human papillomavirus E6 proteins alters localization and signalling of β1-integrin to promote cell migration. J Gen Virol 2014; 95:123–134 [View Article]
    [Google Scholar]
  39. Kanehisa M, Goto S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res 2000; 28:27–30 [View Article]
    [Google Scholar]
  40. Kanehisa M, Goto S, Sato Y, Kawashima M, Furumichi M et al. Data, information, knowledge and principle: back to metabolism in KEGG. Nucleic Acids Res 2014; 42:D199–D205 [View Article]
    [Google Scholar]
  41. Akgül B, García-Escudero R, Ghali L, Pfister HJ, Fuchs PG et al. The E7 protein of cutaneous human papillomavirus type 8 causes invasion of human keratinocytes into the dermis in organotypic cultures of skin. Cancer Res American Association for Cancer Research 2005; 65:2216–2223 [View Article]
    [Google Scholar]
  42. Hsiao YP, Yang JH, Wu WJ, Lin MH, Sheu GT. E6 and E7 of human papillomavirus type 18 and UVB irradiation corporately regulate interleukin-6 and interleukin-8 expressions in basal cell carcinoma. Exp Dermatol 2013; 22:672–674 [View Article]
    [Google Scholar]
  43. Rajkumar T, Sabitha K, Vijayalakshmi N, Shirley S, Bose MV et al. Identification and validation of genes involved in cervical tumourigenesis. BMC Cancer BioMed Central 2011; 11:80 [View Article]
    [Google Scholar]
  44. Regezi JA, Dekker NP, Ramos DM, Li X, Macabeo-Ong M et al. Proliferation and invasion factors in HIV-associated dysplastic and nondysplastic oral warts and in oral squamous cell carcinoma: an immunohistochemical and RT-PCR evaluation. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2002; 94:724–731 [View Article]
    [Google Scholar]
  45. Ryzhakova OS, Solovyeva NI, metalloproteinases M. Matrix metalloproteinases (MMP)—MMP-1,-2,-9 and endogenous regulators of their activity in cervical carcinoma cell lines transformed by the E7 oncogene HPV16 and HPV18. Biochemistry (Moscow) Supplement Series B: Biomedical Chemistry 2012; 6:247–253 [View Article]
    [Google Scholar]
  46. Walker J, Smiley LC, Ingram D, Roman A. Expression of human papillomavirus type 16 E7 is sufficient to significantly increase expression of angiogenic factors but is not sufficient to induce endothelial cell migration. Virology 2011; 410:283–290 [View Article]
    [Google Scholar]
  47. Yuan Z, Gault EA, Campo MS, Nasir L. Upregulation of equine matrix metalloproteinase 1 by bovine papillomavirus type 1 is through the transcription factor activator protein-1. J Gen Virol 2011; 92:2608–2619 [View Article]
    [Google Scholar]
  48. Pruitt KD, Brown GR, Hiatt SM, Thibaud-Nissen F, Astashyn A et al. RefSeq: an update on mammalian reference sequences. Nucleic Acids Res Oxford University Press 2014; 42:D756–D763 [View Article]
    [Google Scholar]
  49. Westermarck J, Kähäri VM. Regulation of matrix metalloproteinase expression in tumor invasion. FASEB J 1999; 13:781–792 [View Article]
    [Google Scholar]
  50. Chen J, Xue Y, Poidinger M, Lim T, Chew SH et al. Mapping of HPV transcripts in four human cervical lesions using RNAseq suggests quantitative rearrangements during carcinogenic progression. Virology Elsevier 2014; 462-463:14–24 [View Article]
    [Google Scholar]
  51. Li Y, Wang X, Ni T, Wang F, Lu W et al. Human papillomavirus type 58 genome variations and RNA expression in cervical lesions. J Virol 2013; 87:9313–9322 [View Article]
    [Google Scholar]
  52. Xue XY, Majerciak V, Uberoi A, Kim BH, Gotte D et al. The full transcription map of mouse papillomavirus type 1 (MmuPV1) in mouse wart tissues. Hearing P, editor PLoS Pathog 2017; 13:e1006715–1006736 [View Article]
    [Google Scholar]
  53. Doorbar J. The papillomavirus life cycle. J Clin Virol 2005; 32:7–15 [View Article]
    [Google Scholar]
  54. Johansson C, Schwartz S. Regulation of human papillomavirus gene expression by splicing and polyadenylation. Nat Rev Microbiol Nature Publishing Group 2013; 11:239–251 [View Article]
    [Google Scholar]
  55. Graham SV, Faizo AAA. Control of human papillomavirus gene expression by alternative splicing. Virus Res Elsevier B.V 2017; 231:83–95 [View Article]
    [Google Scholar]
  56. Van Doorslaer K, Tan Q, Xirasagar S, Bandaru S, Gopalan V et al. The papillomavirus Episteme: a central resource for papillomavirus sequence data and analysis. Nucleic Acids Res Oxford University Press 2013; 41:D571–D578 [View Article]
    [Google Scholar]
  57. Doorbar J. The E4 protein; structure, function and patterns of expression. Virology Elsevier 2013; 445:80–98 [View Article]
    [Google Scholar]
  58. McBride AA. The papillomavirus E2 proteins. Virology 2013; 445:57–79 [View Article]
    [Google Scholar]
  59. Thorland EC, Myers SL, Gostout BS, Smith DI. Common fragile sites are preferential targets for HPV16 integrations in cervical tumors. Oncogene Nature Publishing Group 2003; 22:1225–1237 [View Article]
    [Google Scholar]
  60. Xu F, Cao M, Shi Q, Chen H, Wang Y et al. Integration of the full-length HPV16 genome in cervical cancer and CaSKi and SiHa cell lines and the possible ways of HPV integration. Virus Genes Springer US 2015; 50:210–220 [View Article]
    [Google Scholar]
  61. Ho CM, Lee BH, Chang SF, Chien TY, Huang SH et al. Integration of human papillomavirus correlates with high levels of viral oncogene transcripts in cervical carcinogenesis. Virus Res Elsevier B.V 2011; 161:124–130 [View Article]
    [Google Scholar]
  62. Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ. Basic local alignment search tool. J Mol Biol 1990; 215:403–410 [View Article]
    [Google Scholar]
  63. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics BioMed Central 2011; 12:323 [View Article]
    [Google Scholar]
  64. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 2009; 26:139–140 [View Article]
    [Google Scholar]
  65. Robinson MD, Oshlack A. A scaling normalization method for differential expression analysis of RNA-Seq data. Genome Biol BioMed Central 2010; 11:R25 [View Article]
    [Google Scholar]
  66. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA National Academy of Sciences 2005; 102:15545–15550 [View Article]
    [Google Scholar]
  67. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet 2003; 34:267–273 [View Article]
    [Google Scholar]
  68. Franceschini A, Szklarczyk D, Frankild S, Kuhn M, Simonovic M et al. String v9.1: protein-protein interaction networks, with increased coverage and integration. Nucleic Acids Res 2013; 41:D808–D815 [View Article]
    [Google Scholar]
  69. Huang DW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using David bioinformatics resources. Nature Protocols Nature Publishing Group 2009; 4:44–57 [View Article]
    [Google Scholar]
  70. Zinman GE, Naiman S, Kanfi Y, Cohen H, Bar-Joseph Z. ExpressionBlast: mining large, unstructured expression databases. Nat Methods Nature Publishing Group 2013; 10:925–926 [View Article]
    [Google Scholar]
  71. Kim D, Pertea G, Trapnell C, Pimentel H, Kelley R et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol BioMed Central 2013; 14:R36 [View Article]
    [Google Scholar]
  72. Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES et al. Integrative genomics viewer. Nat Biotechnol Nature Publishing Group 2011; 29:24–26 [View Article]
    [Google Scholar]
  73. Thorvaldsdóttir H, Robinson JT, Mesirov JP. Integrative genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform 2013; 14:178–192 [View Article]
    [Google Scholar]
  74. Paris DBBP, Kuijk EW, Roelen BAJ, Stout TAE. Establishing reference genes for use in real-time quantitative PCR analysis of early equine embryos. Reprod Fertil Dev CSIRO PUBLISHING 2011; 23:353–363 [View Article]
    [Google Scholar]
  75. Cappelli K, Felicetti M, Capomaccio S, Spinsanti G, Silvestrelli M et al. Exercise induced stress in horses: selection of the most stable reference genes for quantitative RT-PCR normalization. BMC Mol Biol BioMed Central 2008; 9:49 [View Article]
    [Google Scholar]
  76. Ståhlberg A, Rusnakova V, Forootan A, Anderova M, Kubista M. RT-qPCR work-flow for single-cell data analysis. Methods Elsevier Inc 2013; 59:80–88 [View Article]
    [Google Scholar]
  77. Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N et al. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol BioMed Central 2002; 3:research0034.1 [View Article]
    [Google Scholar]
  78. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 2014; 30:2114–2120 [View Article]
    [Google Scholar]
  79. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods Nature Publishing Group 2012; 9:357–359 [View Article]
    [Google Scholar]
  80. Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 2010; 26:841–842 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001267
Loading
/content/journal/jgv/10.1099/jgv.0.001267
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF

Most cited Most Cited RSS feed