Non-random integration of the HPV genome in cervical cancer

Martina Schmitz, Corina Driesch, Lars Jansen, Ingo B Runnebaum, Matthias Dürst, Martina Schmitz, Corina Driesch, Lars Jansen, Ingo B Runnebaum, Matthias Dürst

Abstract

HPV DNA integration into the host genome is a characteristic but not an exclusive step during cervical carcinogenesis. It is still a matter of debate whether viral integration contributes to the transformation process beyond ensuring the constitutive expression of the viral oncogenes. There is mounting evidence for a non-random distribution of integration loci and the direct involvement of cellular cancer-related genes. In this study we addressed this topic by extending the existing data set by an additional 47 HPV16 and HPV18 positive cervical carcinoma. We provide supportive evidence for previously defined integration hotspots and have revealed another cluster of integration sites within the cytogenetic band 3q28. Moreover, in the vicinity of these hotspots numerous microRNAs (miRNAs) are located and may be influenced by the integrated HPV DNA. By compiling our data and published reports 9 genes could be identified which were affected by HPV integration at least twice in independent tumors. In some tumors the viral-cellular fusion transcripts were even identical with respect to the viral donor and cellular acceptor sites used. However, the exact integration sites are likely to differ since none of the integration sites analysed thus far have shown more than a few nucleotides of homology between viral and host sequences. Therefore, DNA recombination involving large stretches of homology at the integration site can be ruled out. It is however intriguing that by sequence alignment several regions of the HPV16 genome were found to have highly homologous stretches of up to 50 nucleotides to the aforementioned genes and the integration hotspots. One common region of homologies with cellular sequences is between the viral gene E5 and L2 (nucleotides positions 4100 to 4240). We speculate that this and other regions of homology are involved in the integration process. Our observations suggest that targeted disruption, possibly also of critical cellular genes, by HPV integration remains an issue to be fully resolved.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Chromosomal hotspots for HPV integration.
Figure 1. Chromosomal hotspots for HPV integration.
Depicted are integration sites located within the cytogenetic bands 3q28 (A), 4q13.3 (B), 8q24.21 (C), 13q22.1 (D) and 17q21.2 (E). Light blue arrows: genes affected by HPV integration; red arrows: HPV fusion transcripts described in this work; grey arrows: HPV fusion transcripts described by Kraus et al. 2008; green arrows: fragile site; dark blue arrows: microRNAs.
Figure 2. Sequence homologies between the HPV16…
Figure 2. Sequence homologies between the HPV16 genome and the two adjoining genes TP63 and LEPREL1.
Three homologous stretches of up to 50 nucleotides are shown. The number of exact nucleotide matches is given in brackets (see also Sequences S1). The DNA loop between the two homologies located on LEPREL1 comprises 102.689 nucleotides; the second loop 269.968 nucleotides. Grey dots refer to the approximate location of the corresponding viral-cellular fusion transcripts detected in tumors D3829, T2107 and T4335 (from left to right).

References

    1. Munoz N, Bosch FX, de Sanjose S, Herrero R, Castellsague X, et al. Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med. 2003;348:518–527.
    1. Durst M, Kleinheinz A, Hotz M, Gissmann L. The physical state of human papillomavirus type 16 DNA in benign and malignant genital tumours. J Gen Virol 66 (Pt. 1985;7):1515–1522.
    1. Wentzensen N, Vinokurova S, von Knebel Doeberitz M. Systematic review of genomic integration sites of human papillomavirus genomes in epithelial dysplasia and invasive cancer of the female lower genital tract. Cancer Res. 2004;64:3878–3884.
    1. Pett M, Coleman N. Integration of high-risk human papillomavirus: a key event in cervical carcinogenesis? J Pathol. 2007;212:356–367.
    1. Kalantari M, Blennow E, Hagmar B, Johansson B. Physical state of HPV16 and chromosomal mapping of the integrated form in cervical carcinomas. Diagn Mol Pathol. 2001;10:46–54.
    1. Klaes R, Woerner SM, Ridder R, Wentzensen N, Duerst M, et al. Detection of high-risk cervical intraepithelial neoplasia and cervical cancer by amplification of transcripts derived from integrated papillomavirus oncogenes. Cancer Res. 1999;59:6132–6136.
    1. Vinokurova S, Wentzensen N, Kraus I, Klaes R, Driesch C, et al. Type-dependent integration frequency of human papillomavirus genomes in cervical lesions. Cancer Research. 2008;68:307–313.
    1. Khan MJ, Castle PE, Lorincz AT, Wacholder S, Sherman M, et al. The elevated 10-year risk of cervical precancer and cancer in women with human papillomavirus (HPV) type 16 or 18 and the possible utility of type-specific HPV testing in clinical practice. Journal of the National Cancer Institute. 2005;97:1072–1079.
    1. Romanczuk H, Howley PM. Disruption of either the E1 or the E2 regulatory gene of human papillomavirus type 16 increases viral immortalization capacity. Proceedings of the National Academy of Sciences of the United States of America. 1992;89:3159–3163.
    1. Baker CC, Phelps WC, Lindgren V, Braun MJ, Gonda MA, et al. Structural and transcriptional analysis of human papillomavirus type 16 sequences in cervical carcinoma cell lines. Journal of virology. 1987;61:962–971.
    1. Hafner N, Driesch C, Gajda M, Jansen L, Kirchmayr R, et al. Integration of the HPV16 genome does not invariably result in high levels of viral oncogene transcripts. Oncogene. 2008;27:1610–1617.
    1. Peter M, Stransky N, Couturier J, Hupe P, Barillot E, et al. Frequent genomic structural alterations at HPV insertion sites in cervical carcinoma. J Pathol. 2010;221:320–330.
    1. Reuter S, Bartelmann M, Vogt M, Geisen C, Napierski I, et al. APM-1, a novel human gene, identified by aberrant co-transcription with papillomavirus oncogenes in a cervical carcinoma cell line, encodes a BTB/POZ-zinc finger protein with growth inhibitory activity. Embo J. 1998;17:215–222.
    1. Schmitz M, Driesch C, Beer-Grondke K, Jansen L, Runnebaum IB, et al. Loss of gene function as a consequence of human papillomavirus DNA integration. International journal of cancer Journal international du cancer. 2012.
    1. Ferber MJ, Thorland EC, Brink AA, Rapp AK, Phillips LA, et al. Preferential integration of human papillomavirus type 18 near the c-myc locus in cervical carcinoma. Oncogene. 2003;22:7233–7242.
    1. Popescu NC, DiPaolo JA. Integration of human papillomavirus 16 DNA and genomic rearrangements in immortalized human keratinocyte lines. Cancer Research. 1990;50:1316–1323.
    1. Peter M, Rosty C, Couturier J, Radvanyi F, Teshima H, et al. MYC activation associated with the integration of HPV DNA at the MYC locus in genital tumors. Oncogene. 2006;25:5985–5993.
    1. Koopman LA, Szuhai K, van Eendenburg JD, Bezrookove V, Kenter GG, et al. Recurrent integration of human papillomaviruses 16, 45, and 67 near translocation breakpoints in new cervical cancer cell lines. Cancer Res. 1999;59:5615–5624.
    1. Thorland EC, Myers SL, Gostout BS, Smith DI. Common fragile sites are preferential targets for HPV16 integrations in cervical tumors. Oncogene. 2003;22:1225–1237.
    1. Ziegert C, Wentzensen N, Vinokurova S, Kisseljov F, Einenkel J, et al. A comprehensive analysis of HPV integration loci in anogenital lesions combining transcript and genome-based amplification techniques. Oncogene. 2003;22:3977–3984.
    1. Kraus I, Driesch C, Vinokurova S, Hovig E, Schneider A, et al. The majority of viral-cellular fusion transcripts in cervical carcinomas cotranscribe cellular sequences of known or predicted genes. Cancer Res. 2008;68:2514–2522.
    1. Iorio MV, Croce CM. MicroRNAs in cancer: small molecules with a huge impact. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2009;27:5848–5856.
    1. Calin GA, Croce CM. MicroRNAs and chromosomal abnormalities in cancer cells. Oncogene. 2006;25:6202–6210.
    1. Lee JW, Choi CH, Choi JJ, Park YA, Kim SJ, et al. Altered MicroRNA expression in cervical carcinomas. Clinical cancer research : an official journal of the American Association for Cancer Research. 2008;14:2535–2542.
    1. Lui WO, Pourmand N, Patterson BK, Fire A. Patterns of known and novel small RNAs in human cervical cancer. Cancer Research. 2007;67:6031–6043.
    1. Cheng AM, Byrom MW, Shelton J, Ford LP. Antisense inhibition of human miRNAs and indications for an involvement of miRNA in cell growth and apoptosis. Nucleic acids research. 2005;33:1290–1297.
    1. Martinez I, Gardiner AS, Board KF, Monzon FA, Edwards RP, et al. Human papillomavirus type 16 reduces the expression of microRNA-218 in cervical carcinoma cells. Oncogene. 2008;27:2575–2582.
    1. Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:2257–2261.
    1. Wang X, Wang HK, McCoy JP, Banerjee NS, Rader JS, et al. Oncogenic HPV infection interrupts the expression of tumor-suppressive miR-34a through viral oncoprotein E6. RNA. 2009;15:637–647.
    1. Chang TC, Wentzel EA, Kent OA, Ramachandran K, Mullendore M, et al. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Molecular cell. 2007;26:745–752.
    1. Cho WC. MicroRNAs: potential biomarkers for cancer diagnosis, prognosis and targets for therapy. The international journal of biochemistry & cell biology. 2010;42:1273–1281.
    1. Ueda T, Volinia S, Okumura H, Shimizu M, Taccioli C, et al. Relation between microRNA expression and progression and prognosis of gastric cancer: a microRNA expression analysis. The lancet oncology. 2010;11:136–146.
    1. You J. Papillomavirus interaction with cellular chromatin. Biochimica et biophysica acta. 2010;1799:192–199.
    1. Dey A, Chitsaz F, Abbasi A, Misteli T, Ozato K. The double bromodomain protein Brd4 binds to acetylated chromatin during interphase and mitosis. Proceedings of the National Academy of Sciences of the United States of America. 2003;100:8758–8763.
    1. Choi YW, Bae SM, Kim YW, Lee HN, Park TC, et al. Gene expression profiles in squamous cell cervical carcinoma using array-based comparative genomic hybridization analysis. International journal of gynecological cancer : official journal of the International Gynecological Cancer Society. 2007;17:687–696.
    1. Prazeres H, Torres J, Rodrigues F, Pinto M, Pastoriza MC, et al. Chromosomal, epigenetic and microRNA-mediated inactivation of LRP1B, a modulator of the extracellular environment of thyroid cancer cells. Oncogene. 2011;30:1302–1317.
    1. Lu YJ, Wu CS, Li HP, Liu HP, Lu CY, et al. Aberrant methylation impairs low density lipoprotein receptor-related protein 1B tumor suppressor function in gastric cancer. Genes, Chromosomes & Cancer. 2010;49:412–424.
    1. Liu CX, Musco S, Lisitsina NM, Yaklichkin SY, Lisitsyn NA. Genomic organization of a new candidate tumor suppressor gene, LRP1B. Genomics. 2000;69:271–274.
    1. Nagayama K, Kohno T, Sato M, Arai Y, Minna JD, et al. Homozygous deletion scanning of the lung cancer genome at a 100-kb resolution. Genes, Chromosomes & Cancer. 2007;46:1000–1010.
    1. Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 2008;455:1069–1075.
    1. Kohno T, Otsuka A, Girard L, Sato M, Iwakawa R, et al. A catalog of genes homozygously deleted in human lung cancer and the candidacy of PTPRD as a tumor suppressor gene. Genes, Chromosomes & Cancer. 2010;49:342–352.
    1. Sonoda I, Imoto I, Inoue J, Shibata T, Shimada Y, et al. Frequent silencing of low density lipoprotein receptor-related protein 1B (LRP1B) expression by genetic and epigenetic mechanisms in esophageal squamous cell carcinoma. Cancer Research. 2004;64:3741–3747.
    1. Nakagawa T, Pimkhaokham A, Suzuki E, Omura K, Inazawa J, et al. Genetic or epigenetic silencing of low density lipoprotein receptor-related protein 1B expression in oral squamous cell carcinoma. Cancer science. 2006;97:1070–1074.
    1. Li Y, Knisely JM, Lu W, McCormick LM, Wang J, et al. Low density lipoprotein (LDL) receptor-related protein 1B impairs urokinase receptor regeneration on the cell surface and inhibits cell migration. The Journal of biological chemistry. 2002;277:42366–42371.
    1. Shah R, Smith P, Purdie C, Quinlan P, Baker L, et al. The prolyl 3-hydroxylases P3H2 and P3H3 are novel targets for epigenetic silencing in breast cancer. British journal of cancer. 2009;100:1687–1696.
    1. Vasilescu F, Ceausu M, Tanase C, Stanculescu R, Vladescu T, et al. P53, p63 and Ki-67 assessment in HPV-induced cervical neoplasia. Romanian journal of morphology and embryology = Revue roumaine de morphologie et embryologie. 2009;50:357–361.
    1. Nishi H, Isaka K, Sagawa Y, Usuda S, Fujito A, et al. Mutation and transcription analyses of the p63 gene in cervical carcinoma. International journal of oncology. 1999;15:1149–1153.
    1. Santamarina-Fojo S, Haudenschild C, Amar M. The role of hepatic lipase in lipoprotein metabolism and atherosclerosis. Current opinion in lipidology. 1998;9:211–219.
    1. Sauermann M, Sahin O, Sultmann H, Hahne F, Blaszkiewicz S, et al. Reduced expression of vacuole membrane protein 1 affects the invasion capacity of tumor cells. Oncogene. 2008;27:1320–1326.
    1. Chan JA, Krichevsky AM, Kosik KS. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Research. 2005;65:6029–6033.
    1. Liu M, Wu H, Liu T, Li Y, Wang F, et al. Regulation of the cell cycle gene, BTG2, by miR-21 in human laryngeal carcinoma. Cell research. 2009;19:828–837.
    1. Schmitz M, Scheungraber C, Herrmann J, Teller K, Gajda M, et al. Quantitative multiplex PCR assay for the detection of the seven clinically most relevant high-risk HPV types. Journal of clinical virology : the official publication of the Pan American Society for Clinical Virology. 2009;44:302–307.
    1. Dall KL, Scarpini CG, Roberts I, Winder DM, Stanley MA, et al. Characterization of naturally occurring HPV16 integration sites isolated from cervical keratinocytes under noncompetitive conditions. Cancer Res. 2008;68:8249–8259.
    1. Matovina M, Sabol I, Grubisic G, Gasperov NM, Grce M. Identification of human papillomavirus type 16 integration sites in high-grade precancerous cervical lesions. Gynecol Oncol. 2009;113:120–127.
    1. Wentzensen N, Ridder R, Klaes R, Vinokurova S, Schaefer U, et al. Characterization of viral-cellular fusion transcripts in a large series of HPV16 and 18 positive anogenital lesions. Oncogene. 2002;21:419–426.

Source: PubMed

3
Suscribir