Stratification of HPV-induced cervical pathology using the virally encoded molecular marker E4 in combination with p16 or MCM

Heather Griffin, Yasmina Soneji, Romy Van Baars, Rupali Arora, David Jenkins, Miekel van de Sandt, Zhonglin Wu, Wim Quint, Robert Jach, Krzysztof Okon, Hubert Huras, Albert Singer, John Doorbar, Heather Griffin, Yasmina Soneji, Romy Van Baars, Rupali Arora, David Jenkins, Miekel van de Sandt, Zhonglin Wu, Wim Quint, Robert Jach, Krzysztof Okon, Hubert Huras, Albert Singer, John Doorbar

Abstract

High-risk human papillomavirus (HPV) types cause cervical lesions of varying severity, ranging from transient productive infections to high-grade neoplasia. Disease stratification requires the examination of lesional pathology, and possibly also the detection of biomarkers. P16(INK4a) and MCM are established surrogates of high-risk HPV E6/E7 activity, and can be extensively expressed in high-grade lesions. Here we have combined these two cellular biomarkers with detection of the abundant HPV-encoded E4 protein in order to identify both productive and transforming lesions. This approach has allowed us to distinguish true papillomavirus infections from similar pathologies, and has allowed us to divide the heterogeneous CIN2 category into those that are CIN1-like and express E4, and those that more closely resemble nonproductive CIN3. To achieve this, 530 lesional areas were evaluated according to standard pathology criteria and by using a multiple staining approach that allows us to superimpose biomarker patterns either singly or in combination onto an annotated hematoxylin and eosin (H&E) image. Conventional grading of neoplasia was established by review panel, and compared directly with the composite molecular pathology visualized on the same tissue section. The detection of E4 coincided with the onset of vacuolation, becoming abundant in koilocytes as the MCM marker declined and cells lost their defined nuclear margins as visualized by standard H&E staining. Of the dual marker approaches, p16(INK4a) and E4 appeared most promising, with E4 generally identifying areas of low-grade disease even when p16(INK4a) was present. Extensive p16(INK4a) expression usually coincided with an absence of E4 expression or its focal retention in sporadic cells within the lesion. Our results suggest that a straightforward molecular evaluation of HPV life-cycle deregulation in cervical neoplasia may help improve disease stratification, and that this can be achieved using complementary molecular biomarker pairs such as MCM/E4 or, more promisingly, p16(INK4a)/E4 as an adjunct to conventional pathology.

Figures

Figure 1. Overlay of biomarker patterns onto…
Figure 1. Overlay of biomarker patterns onto annotated Hemotoxylin and Eosin Pathology
A. Individual tissue sections were subject to immunofluorescence staining to detect the biomarkers E4 using a TVG405-Alexa 488 conjugate (green), MCM using an anti-mouse Alexa 594 secondary antibody (red) and cellular DNA using 4′,6-Diamidino-2-Phenylindole (DAPI; blue). Individual stains were recorded digitally at high resolution (lower panels) before the section was processed for p16INK4a staining and development in 3_Amino_9_ethylcarbazole (AEC; brown). The AEC image was digitally captured, before the tissue section was cleared of the AEC substrate and stained with Carazzis (X2) Hemotoxylin and Eosin. All H&E stained images were then examined independently by three pathologists, and regions with discrete pathology phenotypes recorded. The primary categories of HPV-associated pathology comprised CIN1, CIN2 and CIN3, with the general term ‘non-CIN’ being used to encompass a variety of non HPV-associated pathologies such as inflammation and metaplasia. A ‘normal’ classification was given when there was no histological abnormality. B. The 3-colour immunofluorescence stain is shown on the left. Single channel coloured images (collected as described in A) were extracted from the immunofluorescence image or from the p16INK4a AEC stain and were superimposed onto the H&E images (under the heading ‘Pathology overlay’). The simple dual marker molecular pathologies (i.e. not overlayed onto the H&E image) are shown on the right to reveal the relative distributions of E4/MCM and E4/ p16INK4a (under the heading ‘Dual marker molecular pathology). The image shown is typical of those used to prepare the more detailed images of neoplasias used in subsequent figures.
Figure 2. Distribution of the HPV_E4, p16…
Figure 2. Distribution of the HPV_E4, p16INK4a and MCM2 biomarkers in lesions unambiguously classified as CIN1
A. Biomarker patterns typically associated with CIN1. The E4/MCM (green/red) biomarker patterns are shown in the immunofluorescence image to the left of the figure. To facilitate comparison with lesional pathology, the E4/MCM (green/red) and p16INK4a (brown) biomarkers are overlayed onto (and shown alongside) the basic H & E stain in the central part of the figure. The E4/MCM (green/red) and E4/ p16INK4a (green/brown) biomarker overlays are shown separately to the right of the figure so that their relative distributions can be observed. The Biomarker patterns seen in HPV associated CIN1 are described below.
  1. i) Most CIN1 are productive infections, with MCM (red) reaching and extending into the E4 expressing layers (green). In this lesion, MCM and p16INK4a have broadly similar distributions.

  2. ii) Although p16INK4a (brown) and MCM (red) expression can have different distributions, MCM again extends into the E4-positive layers (green). In this lesion, MCM extends higher into the epithelial layers than p16INK4a.

  3. iii) A small number of consensus CIN1 (6/33) failed to show E4 (green) biomarker expression. p16INK4a (brown) expression extends into the upper epithelial layers and is more extensive than MCM (red).

B. Correlation of Biomarker patterns with Pathology. The E4/MCM (green/red) biomarker distribution in consensus CIN1 are shown as immunofluorescence images to the left of the figure, and overlayed onto H & E pathology towards the centre. The higher magnification shown on the right allows correlation of specific pathology characteristics with the accumulation of E4 and the decline of MCM.
  1. i) In the majority of CIN1, the E4 (green) biomarker accumulates during the process of koilocyte formation. Arrows labelled 1 to 4 show the progressive vacuolation that accompanies E4 accumulation (beginning at arrow 2 and highest at arrow 4), and the loss of the MCM biomarker (lowest at arrow 4).

  2. ii) Despite some differences in cell morphology and tissue architecture amongst CIN1, the distribution of biomarkers described above (i) is conserved.

  3. iii) In one (out of 33) consensus CIN1, koilocyte-like cells we present but were devoid of markers of productive infection, including the E4 (green) biomarker and associated MCM (red) staining.

Figure 3. Relevance of the HPV_E4, p16…
Figure 3. Relevance of the HPV_E4, p16INK4a and MCM biomarkers in consensus CIN3 lesions
A. Biomarker Patterns Typically Associated with CIN3. The E4/MCM (green/red) and p16INK4a (brown) biomarker images are formatted as outlined in Fig.2A. The biomarker patterns seen in HPV-associated CIN3 are described below.
  1. i) In the majority of CIN3, the E4 (green) biomarker is absent, and both the MCM (red) and p16INK4a (brown) biomarkers extend uniformly through the full thickness of the epithelium.

  2. ii) Despite differences in lesion size and morphology, the biomarker patterns described above (i) were broadly similar in other CIN3.

B. Expression of the E4 biomarker is an occasional occurrence in CIN3.
  1. i) In a small number of CIN3 (4/68), focal areas of E4 (green) were apparent. Both p16INK4a (brown) and MCM (red) extended through the full thickness of the lesion with evidence of nuclear crowding as seen in Fig 3A. Panel A is shown enlarged in Fig. 3C.

  2. ii) In one case (out of 68), the E4 (green) biomarker was extensive, but was confined to regions showing low-grade pathology that lacked strong p16INK4a (brown) biomarker staining. Panel B is shown enlarged in Fig. 3B.

C. Pathology associated with E4 expression in CIN3. The three images to the left show an enlargement of Fig. 3B(i) panel A. Although vacuolated cells are sometimes apparent in CIN3 (arrows marked ‘v’), they are not necessarily associated with expression of the E4 biomarker (Fig. 2B(i)). The pathology associated with E4 expression in CIN3 is more clearly shown in the images to the right, which is an enlargement of panel B from Fig.3B(ii). This region of low-grade pathology was contained within the CIN3 area. The pattern of vacuolation and koilocyte formation is comparable to that seen in CIN1 (Fig. 2B), with E4 expression beginning at the first sign of vacuolation (arrows labelled 1) and becoming prominent as the MCM biomarker is lost (arrows labelled 2).
Figure 4. Expression of HPV_E4 along with…
Figure 4. Expression of HPV_E4 along with p16INK4a and MCM suggests two categories within the consensus CIN2 group
A. HPV_E4-positivity amongst consensus CIN2. The E4/MCM (green/red) and p16INK4a (brown) biomarker images are formatted as outlined in Fig.2A. The biomarker patterns seen in E4-positive HPV-associated CIN2 are described below.
  1. i) High cell density and the expression of MCM precedes the accumulation of E4 in a subset of cells showing evidence of vacuolation. P16INK4a and MCM expression extend throughout the full thickness of the epithelium. Panel A is enlarged in Fig.5.

  2. ii) Despite differences in epithelial thickness, the biomarker pattern is preserved in other consensus CIN2.

  3. iii) As in CIN1, some CIN2 show a more extensive expression of MCM into the upper epithelial layers when compared to p16INK4a. E4 expression is limited to cells close to the epithelial surface. Panel B is enlarged in Fig.5 to illustrate the correlation between pathology and biomarker patterns.

B. HPV_E4 negativity amongst consensus CIN2.
  1. i) Loss of the E4 biomarker in a proportion of CIN2, can be associated with extensive expression of the p16INK4a /MCM biomarkers throughout the thickness of the epithelium, as well as an absence of obvious differentiation at the level of pathology. Panel C is enlarged in Fig. 5.

  2. ii) MCM can extend closer throughout the epithelium more robustly than p16INK4a without the expression of the E4 biomarker. The panel shown in D is enlarged in Fig. 5 to show the absence of key pathology features apparent in the E4-positive CIN2.

Figure 5. Expression of the HPV_E4 biomarker…
Figure 5. Expression of the HPV_E4 biomarker in CIN2 is associated with discrete regions of CIN1-like pathology
A.
  1. i) Pathology associated with E4 expression in CIN2. The six images show enlargements of the regions that are boxed in Fig. 4A(i) and (iii) (H&E images). The pattern of vacuolation and koilocyte formation is similar to that seen in CIN1 (Fig. 2B), with E4 expression, vacuolation and MCM decline coinciding closely (arrows 1&2).

B.
  1. ii) The six images show an enlargement of the regions that are boxed in Fig 4B(i) and (ii). In these lesions, vacuolated cells are sometimes apparent (arrows marked ‘v’), but are not necessarily associated with expression of the E4 biomarker (Fig. 4B(i) and (ii)).

Figure 6. Division of Cervical Pathologies according…
Figure 6. Division of Cervical Pathologies according to the Presence and Distribution of the Molecular Markers E4 and p16INK4a
A. Lesional areas where there was total agreement amongst the panel of pathologists. The columns in each graph show the individual diagnostic opinions provided by the pathologist panel after review of the H&E stained slides, as either non-CIN, CIN1, CIN2 or CIN3. Graphs shown in A include only lesional areas where there was total agreement amongst the pathologist panel. This standard pathology-grading is stratified according to whether the diagnosed areas were subsequently found to be HPV-E4-positive (green edged columns/left-most graph) or E4-negative (red edged columns/right-most graph), and to what extent the p16INK4a expression extended through the epithelium. Lesional areas showing full thickness p16 staining are indicated as dark brown columns, with lower levels of staining being shown in lighter shades of brown. Lesional areas that lacked p16INK4a staining are indicated by white columns. In general, the patterns fit with our current model of life-cycle deregulation in high-grade neoplasia, with an absence of both markers in the ‘total-agreement’ non-CIN group. As described in the text, E4 expression in CIN3 (marked by an asterisk) was typically sporadic and in a small number of cells close to the epithelial surface. B. All lesional areas including those where there was disagreement amongst the panel of pathologists. The columns show the individual diagnostic opinions provided by the pathologist panel on all lesional areas, irrespective of whether there was agreement or disagreement between individual pathologists. Labelling is as described in ‘A’ above. Because the H&E-based diagnostic opinion often differed between the individual pathologists, the pattern of the p16INK4a and E4 staining typical of non-CIN, CIN1, CIN2 and CIN3 (shown in Fig.6A) is less apparent, with some evidence of virus infection in the ‘proposed’ non-CIN group, and an absence of biomarker staining in some ‘proposed’CIN1.
Figure 7. Molecular Principles Underlying the use…
Figure 7. Molecular Principles Underlying the use of p16, MCM and E4 as HPV-Associated Disease Biomarkers
A. In uninfected epithelium, the cellular MCM protein (red) is usually detectable at low levels only in the basal and parabasal cell layers as a result of cell-cycle stimulation by growth factors. This facilitates the phosphorylation of pRb by cyclin-dependent kinases, the release of the E2F transcription factor, and the regulated expression of MCM. During normal metaplasia or wound healing, MCM may also be detected in the upper epithelial layers. The cellular p16INK4a protein is also stimulated by E2F, but does not usually accumulate to detectable levels in uninfected epithelium. It provides feedback-regulation on the activity of cyclin-dependent kinases. p16INK4a is sometimes visualised as a weak cytoplasmic stain in cells undergoing senescence (pale brown). The HPV-encoded E4 protein is never expressed in uninfected epithelium and E4 antibodies show no reactivity with cellular proteins. B. (i) In HPV-infected epithelial tissue, the high-risk E6 & E7 genes (red) are expressed together from the viral early promoter (PE), and function to drive cell-cycle entry in order to allow cell proliferation and genome amplification. The high-risk E4 gene (green) is expressed from a spliced mRNA, and becomes abundant following the activation of the viral late promoter (PL) as the infected cell exits the cell cycle and commits to true differentiation. (ii) E6 and E7 are expressed at low level in the cell, but the consequences of their presence can be visualised by alterations in the presence of p16INK4a and MCM. The association of E7 with pRb leads to E2F release irrespective of growth factor stimulation. This allows MCM and also p16INK4a to accumulate to higher levels than are typically seen in uninfected epithelium where expression is dependent on cyclin-dependent kinase activation. The E7 protein also acts to increase the transcription of p16INK4a as a result of epigenetic modification of the p16INK4a promoter. In this context, p16INK4a and MCM can be used with caution as surrogate markers of E6/E7 deregulation. The viral E4 protein becomes abundant in the upper layers of HPV-infected epithelium as a result of viral late promoter activation and the cleavage of the full-length E4 protein by calpain. Calpain-cleavage exposes a C-terminal multimerization motif in E4, which allows its assembly into amyloid-like fibres. The high-level accumulation of E4-amyloid is thought to coincide with progression of the infected cell through the G2 phase of the cell cycle and eventually to cell-cycle exit, explaining its appearance as MCM levels decline.

References

    1. Bosch FX, Broker TR, Forman D, et al. Comprehensive control of human papillomavirus infections and related diseases. Vaccine. 2013;31(Suppl 8):I1–31.
    1. Ferlay J, Shin HR, Bray F, et al. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010;127:2893–917.
    1. Doorbar J, Quint W, Banks L, et al. The biology and life-cycle of human papillomaviruses. Vaccine. 2012;30(Suppl 5):F55–70.
    1. Westra WH. The morphologic profile of HPV-related head and neck squamous carcinoma: implications for diagnosis, prognosis, and clinical management. Head Neck Pathol. 2012;6(Suppl 1):S48–54.
    1. de Villiers EM, Fauquet C, Broker TR, Bernard HU, zur Hausen H. Classification of papillomaviruses. Virology. 2004;324:17–27.
    1. Peh WL, Middleton K, Christensen N, et al. Life cycle heterogeneity in animal models of human papillomavirus-associated disease. Journal of Virology. 2002;76:10401–16.
    1. Cubie HA. Diseases associated with human papillomavirus infection. Virology. 2013;445:21–34.
    1. Jenkins D. Histopathology and cytopathology of cervical cancer. Dis Markers. 2007;23:199–212.
    1. Arwert EN, Hoste E, Watt FM. Epithelial stem cells, wound healing and cancer. Nat Rev Cancer. 2012;12:170–80.
    1. Kurman RJ, Ellenson LH, Ronnett BM. Blaustein’s Pathology of the Female Genital Tract. Springer US; 2010.
    1. Pyeon D, Pearce SM, Lank SM, Ahlquist P, Lambert PF. Establishment of human papillomavirus infection requires cell cycle progression. PLoS Pathog. 2009;5:e1000318.
    1. Middleton K, Peh W, Southern S, et al. Organization of human papillomavirus productive cycle during neoplastic progression provides a basis for selection of diagnostic markers. J Virol. 2003;77:10186–201.
    1. Bodily J, Laimins LA. Persistence of human papillomavirus infection: keys to malignant progression. Trends in microbiology. 2011;19:33–9.
    1. Isaacson Wechsler E, Wang Q, Roberts I, et al. Reconstruction of human papillomavirus type 16-mediated early-stage neoplasia implicates E6/E7 deregulation and the loss of contact inhibition in neoplastic progression. J Virol. 2012;86:6358–64.
    1. Hafner N, Driesch C, Gajda M, et al. Integration of the HPV16 genome does not invariably result in high levels of viral oncogene transcripts. Oncogene. 2008;27:1610–7.
    1. Khleif SN, DeGregori J, Yee CL, et al. Inhibition of cyclin D-CDK4/CDK6 activity is associated with an E2F-mediated induction of cyclin kinase inhibitor activity. Proc Natl Acad Sci U S A. 1996;93:4350–4.
    1. Chen HC, Schiffman M, Lin CY, et al. Persistence of type-specific human papillomavirus infection and increased long-term risk of cervical cancer. J Natl Cancer Inst. 2011;103:1387–96.
    1. Geraets DT, van Doorn LJ, Kleter B, et al. Long-term follow-up of HPV16-positive women: persistence of the same genetic variant and low prevalence of variant co-infections. PLoS One. 2013;8:e80382.
    1. Quint KD, de Koning MN, Quint WG, Pirog EC. Progression of cervical low grade squamous intraepithelial lesions: in search of prognostic biomarkers. Eur J Obstet Gynecol Reprod Biol. 2013;170:501–6.
    1. Cantor SB, Atkinson EN, Cardenas-Turanzas M, et al. Natural history of cervical intraepithelial neoplasia: a meta-analysis. Acta Cytol. 2005;49:405–15.
    1. Doorbar J, Cubie H. Molecular basis for advances in cervical screening. Mol Diagn. 2005;9:129–42.
    1. Roman A, Munger K. The papillomavirus E7 proteins. Virology. 2013;445:138–68.
    1. Tornesello ML, Buonaguro L, Giorgi-Rossi P, Buonaguro FM. Viral and cellular biomarkers in the diagnosis of cervical intraepithelial neoplasia and cancer. BioMed research international. 2013;2013:519619.
    1. von Knebel Doeberitz M, Reuschenbach M, Schmidt D, Bergeron C. Biomarkers for cervical cancer screening: the role of p16(INK4a) to highlight transforming HPV infections. Expert review of proteomics. 2012;9:149–63.
    1. Ikenberg H, Bergeron C, Schmidt D, et al. Screening for cervical cancer precursors with p16/Ki-67 dual-stained cytology: results of the PALMS study. J Natl Cancer Inst. 2013;105:1550–7.
    1. McLaughlin-Drubin ME, Crum CP, Munger K. Human papillomavirus E7 oncoprotein induces KDM6A and KDM6B histone demethylase expression and causes epigenetic reprogramming. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:2130–5.
    1. Doorbar J. Papillomavirus life cycle organization and biomarker selection. Disease Markers. 2007;23:297–313.
    1. Griffin H, Wu Z, Marnane R, et al. E4 antibodies facilitate detection and type-assignment of active HPV infection in cervical disease. PLoS One. 2012;7:e49974.
    1. van der Marel J, van Baars R, Quint W, et al. The impact of human papillomavirus genotype on colposcopic appearance: a cross-sectional analysis. BJOG. 2014
    1. Kleter B, van Doorn LJ, ter Schegget J, et al. Novel short-fragment PCR assay for highly sensitive broad-spectrum detection of anogenital human papillomaviruses. The American journal of pathology. 1998;153:1731–9.
    1. Kleter B, van Doorn LJ, Schrauwen L, et al. Development and clinical evaluation of a highly sensitive PCR-reverse hybridization line probe assay for detection and identification of anogenital human papillomavirus. J Clin Microbiol. 1999;37:2508–17.
    1. Quint W, Jenkins D, Molijn A, et al. One virus, one lesion--individual components of CIN lesions contain a specific HPV type. The Journal of pathology. 2012;227:62–71.
    1. Doorbar J, Foo C, Coleman N, et al. Characterization of events during the late stages of HPV16 infection in vivo using high-affinity synthetic Fabs to E4. Virology. 1997;238:40–52.
    1. de Sanjose S, Quint WG, Alemany L, et al. Human papillomavirus genotype attribution in invasive cervical cancer: a retrospective cross-sectional worldwide study. Lancet Oncol. 2010;11:1048–56.
    1. Kalantari M, Garcia-Carranca A, Morales-Vazquez CD, et al. Laser capture microdissection of cervical human papillomavirus infections: copy number of the virus in cancerous and normal tissue and heterogeneous DNA methylation. Virology. 2009;390:261–7.
    1. Guan P, Howell-Jones R, Li N, et al. Human papillomavirus types in 115,789 HPV-positive women: a meta-analysis from cervical infection to cancer. International journal of cancer Journal international du cancer. 2012;131:2349–59.
    1. Davy CE, Jackson DJ, Raj K, et al. Human Papillomavirus Type 16 E1^E4-Induced G2 Arrest Is Associated with Cytoplasmic Retention of Active Cdk1/Cyclin B1 Complexes. J Virol. 2005;79:3998–4011.
    1. Carreon JD, Sherman ME, Guillen D, et al. CIN2 is a much less reproducible and less valid diagnosis than CIN3: results from a histological review of population-based cervical samples. Int J Gynecol Pathol. 2007;26:441–6.
    1. Cuzick J, Bergeron C, von Knebel Doeberitz M, et al. New technologies and procedures for cervical cancer screening. Vaccine. 2012;30(Suppl 5):F107–16.
    1. Bulkmans NW, Rozendaal L, Voorhorst FJ, Snijders PJ, Meijer CJ. Long-term protective effect of high-risk human papillomavirus testing in population-based cervical screening. Br J Cancer. 2005;92:1800–2.
    1. Elfstrom KM, Smelov V, Johansson AL, et al. Long-term HPV type-specific risks for ASCUS and LSIL: A 14-year follow-up of a randomized primary HPV screening trial. Int J Cancer. 2014
    1. Ronco G, Dillner J, Elfstrom KM, et al. Efficacy of HPV-based screening for prevention of invasive cervical cancer: follow-up of four European randomised controlled trials. Lancet. 2014;383:524–32.
    1. Kocken M, Uijterwaal MH, de Vries AL, et al. High-risk human papillomavirus testing versus cytology in predicting post-treatment disease in women treated for high-grade cervical disease: a systematic review and meta-analysis. Gynecol Oncol. 2012;125:500–7.
    1. Litjens RJ, Hopman AH, van de Vijver KK, et al. Molecular biomarkers in cervical cancer diagnosis: a critical appraisal. Expert opinion on medical diagnostics. 2013;7:365–77.
    1. Maiorano D, Lutzmann M, Mechali M. MCM proteins and DNA replication. Curr Opin Cell Biol. 2006;18:130–6.
    1. Murphy N, Ring M, Heffron CC, et al. Quantitation of CDC6 and MCM5 mRNA in cervical intraepithelial neoplasia and invasive squamous cell carcinoma of the cervix. Mod Pathol. 2005;18:844–9.
    1. Murphy N, Ring M, Heffron CC, et al. p16INK4A, CDC6, and MCM5: predictive biomarkers in cervical preinvasive neoplasia and cervical cancer. J Clin Pathol. 2005;58:525–34.
    1. McCluggage WG, Walsh MY, Thornton CM, et al. Inter- and intra-observer variation in the histopathological reporting of cervical squamous intraepithelial lesions using a modified Bethesda grading system. British journal of obstetrics and gynaecology. 1998;105:206–10.
    1. Piaton E, Advenier AS, Carre C, et al. p16(INK4a) /Ki-67 dual labelling as a marker for the presence of high-grade cancer cells or disease progression in urinary cytopathology. Cytopathology. 2013;24:327–34.
    1. McIntosh PB, Martin SR, Jackson DJ, et al. Structural analysis reveals an amyloid form of the human papillomavirus type 16 E1^E4 protein and provides a molecular basis for its accumulation. Journal of Virology. 2008;82:8196–203.
    1. Khan J, Davy CE, McIntosh PB, et al. Role of calpain in the formation of human papillomavirus type 16 E1^E4 amyloid fibers and reorganization of the keratin network. Journal of Virology. 2011;85:9984–97.
    1. Wang Q, Kennedy A, Das P, et al. Phosphorylation of the human papillomavirus type 16 E1^E4 protein at T57 by ERK triggers a structural change that enhances keratin binding and protein stability. J Virol. 2009;83:3668–83.
    1. Doorbar J. The E4 protein; structure, function and patterns of expression. Virology. 2013;445:80–98.
    1. Davy C, Doorbar J. G2/M cell cycle arrest in the life cycle of viruses. Virology. 2007;368:219–26.
    1. Ortoft G, Henriksen T, Hansen E, Petersen L. After conisation of the cervix, the perinatal mortality as a result of preterm delivery increases in subsequent pregnancy. BJOG. 2011;117:258–67.
    1. Armarnik S, Sheiner E, Piura B, et al. Obstetric outcome following cervical conization. Arch Gynecol Obstet. 2011;283:765–9.
    1. Kinney W, Hunt WC, Dinkelspiel H, et al. Cervical excisional treatment of young women: A population-based study. Gynecol Oncol. 2014;132:628–35.
    1. Henderson D, Hall L, Prpic N, et al. The selection and characterization of antibodies to minichromosome maintenance proteins that highlight cervical dysplasia. J Immunol Methods. 2011;370:1–13.
    1. Galgano MT, Castle PE, Atkins KA, et al. Using biomarkers as objective standards in the diagnosis of cervical biopsies. Am J Surg Pathol. 2010;34:1077–87.
    1. Stoler MH, Schiffman M, Atypical Squamous Cells of Undetermined Significance-Low-grade Squamous Intraepithelial Lesion Triage Study G Interobserver reproducibility of cervical cytologic and histologic interpretations: realistic estimates from the ASCUS-LSIL Triage Study. JAMA. 2001;285:1500–5.
    1. Vrdoljak-Mozetic D, Krasevic M, Versa Ostojic D, et al. HPV16 genotype, p16/Ki-67 dual staining and koilocytic morphology as potential predictors of the clinical outcome for cervical low-grade squamous intraepithelial lesions. Cytopathology. 2013

Source: PubMed

3
Se inscrever