Human Papillomavirus and Cellular Pathways: Hits and Targets
Abstract
:1. Introduction
2. Signaling Pathways and HPV
2.1. p53
2.2. pRb and Pocket Proteins
2.3. EGFR
2.4. PI3K/Akt/mTOR
2.5. JNK/ERK/ AP-1
2.5.1. JNK
2.5.2. ERK
2.5.3. AP-1
2.6. Autophagy
3. EMT
3.1. E-Cadherin
3.2. Wnt/β-Catenin Pathway
4. Immunology and Inflammation
4.1. NF-κB Pathway
4.2. JAK/STAT Pathway
4.3. TGF-β/TNF-α
5. miRNAs
6. DNA Damage Response
7. DNA Methylation
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Zur Hausen, H. Papillomaviruses in the causation of human cancers-a brief historical account. Virology 2009, 384, 260–265. [Google Scholar] [CrossRef] [Green Version]
- Giuliano, A.R.; Nyitray, A.G.; Kreimer, A.R.; Pierce Campbell, C.M.; Goodman, M.T.; Sudenga, S.L.; Monsonego, J.; Franceschi, S. EUROGIN 2014 roadmap: Differences in human papillomavirus infection natural history, transmission and human papillomavirus-related cancer incidence by gender and anatomic site of infection. Int. J. Cancer 2015, 136, 2752–2760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Villiers, E.M.; Fauquet, C.; Broker, T.R.; Bernard, H.U.; Zur Hausen, H. Classification of papillomaviruses. Virology 2004, 324, 17–27. [Google Scholar] [CrossRef] [Green Version]
- Harden, M.E.; Munger, K. Human papillomavirus molecular biology. Mutat. Res. Rev. Mutat. Res. 2017, 772, 3–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Favre, M.; Orth, G.; Croissant, O.; Yaniv, M. Human papillomavirus DNA: Physical map. Proc. Natl. Acad. Sci. USA 1975, 72, 4810–4814. [Google Scholar] [CrossRef] [Green Version]
- Zheng, Z.M.; Baker, C.C. Papillomavirus genome structure, expression, and post-transcriptional regulation. Front. Biosci. 2006, 11, 2286–2302. [Google Scholar] [CrossRef] [Green Version]
- Ribeiro, A.L.; Caodaglio, A.S.; Sichero, L. Regulation of HPV transcription. Clinics 2018, 73, e486s. [Google Scholar] [CrossRef] [PubMed]
- Gupta, S.; Kumar, P.; Das, B.C. HPV: Molecular pathways and targets. Curr. Probl. Cancer 2018, 42, 161–174. [Google Scholar] [CrossRef] [PubMed]
- Doorbar, J.; Egawa, N.; Griffin, H.; Kranjec, C.; Murakami, I. Human papillomavirus molecular biology and disease association. Rev. Med. Virol. 2015, 25, 2–23. [Google Scholar] [CrossRef] [Green Version]
- Sahasrabuddhe, V.V.; Luhn, P.; Wentzensen, N. Human papillomavirus and cervical cancer: Biomarkers for improved prevention efforts. Future Microbiol. 2011, 6, 1083–1098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sabatini, M.E.; Chiocca, S. Human papillomavirus as a driver of head and neck cancers. Br. J. Cancer 2020, 122, 306–314. [Google Scholar] [CrossRef]
- Martinez-Zapien, D.; Ruiz, F.X.; Poirson, J.; Mitschler, A.; Ramirez, J.; Forster, A.; Cousido-Siah, A.; Masson, M.; Pol, S.V.; Podjarny, A.; et al. Structure of the E6/E6AP/p53 complex required for HPV-mediated degradation of p53. Nature 2016, 529, 541–545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zilfou, J.T.; Lowe, S.W. Tumor suppressive functions of p53. Cold Spring Harb. Perspect. Biol. 2009, 1, a001883. [Google Scholar] [CrossRef] [PubMed]
- Muller, P.A.J.; Vousden, K.H. P53 mutations in cancer. Nat. Cell Biol. 2013, 15, 2–8. [Google Scholar] [CrossRef] [PubMed]
- Scheffner, M.; Werness, B.A.; Huibregtse, J.M.; Levine, A.J.; Howley, P.M. The E6 oncoprotein encoded by human papillomavirus types 16 and 18 promotes the degradation of p53. Cell 1990, 63, 1129–1136. [Google Scholar] [CrossRef]
- Oh, S.T.; Longworth, M.S.; Laimins, L.A. Roles of the E6 and E7 Proteins in the Life Cycle of Low-Risk Human Papillomavirus Type 11. J. Virol. 2004, 78, 2620–2626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thomas, J.T.; Laimins, L.A. Human Papillomavirus Oncoproteins E6 and E7 Independently Abrogate the Mitotic Spindle Checkpoint. J. Virol. 1998, 72, 1131–1137. [Google Scholar] [CrossRef] [Green Version]
- Thompson, D.A.; Belinsky, G.; Chang, T.H.T.; Jones, D.L.; Schlegel, R.; Münger, K. The human papillomavirus-16 E6 oncoprotein decreases the vigilance of mitotic checkpoints. Oncogene 1997, 15, 3025–3035. [Google Scholar] [CrossRef] [Green Version]
- Celegato, M.; Messa, L.; Goracci, L.; Mercorelli, B.; Bertagnin, C.; Spyrakis, F.; Suarez, I.; Cousido-Siah, A.; Travé, G.; Banks, L.; et al. A novel small-molecule inhibitor of the human papillomavirus E6-p53 interaction that reactivates p53 function and blocks cancer cells growth. Cancer Lett. 2020, 470, 115–125. [Google Scholar] [CrossRef]
- Citro, S.; Bellini, A.; Medda, A.; Sabatini, M.E.; Tagliabue, M.; Chu, F.; Chiocca, S. Human Papilloma Virus Increases ΔNp63α Expression in Head and Neck Squamous Cell Carcinoma. Front. Cell. Infect. Microbiol. 2020, 10. [Google Scholar] [CrossRef]
- Citro, S.; Bellini, A.; Miccolo, C.; Ghiani, L.; Carey, T.E.; Chiocca, S. Synergistic antitumour activity of HDAC inhibitor SAHA and EGFR inhibitor gefitinib in head and neck cancer: A key role for ΔNp63α. Br. J. Cancer 2019, 120, 658–667. [Google Scholar] [CrossRef] [Green Version]
- Dyson, N.; Howley, P.M.; Münger, K.; Harlow, E. The human papilloma virus-16 E7 oncoprotein is able to bind to the retinoblastoma gene product. Science 1989, 243, 934–937. [Google Scholar] [CrossRef] [PubMed]
- Hu, T.; Ferril, S.C.; Snider, A.M.; Barbosa, M.S. In vivo analysis of HPV E7 protein association with pRb, p107 and p130. Int. J. Oncol. 1995, 6, 167–174. [Google Scholar] [CrossRef] [PubMed]
- Davies, R.; Hicks, R.; Crook, T.; Morris, J.; Vousden, K. Human papillomavirus type 16 E7 associates with a histone H1 kinase and with p107 through sequences necessary for transformation. J. Virol. 1993, 67, 2521–2528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Smith-Mccune, K.; Kalman, D.; Robbins, C.; Shivakumar, S.; Yuschenkoff, L.; Bishop, J.M. Intranuclear localization of human papillomavirus 16 E7 during transformation and preferential binding of E7 to the Rb family member p130. Proc. Natl. Acad. Sci. USA 1999, 96, 6999–7004. [Google Scholar] [CrossRef] [Green Version]
- Kouzarides, T. Transcriptional control by the retinoblastoma protein. Semin. Cancer Biol. 1995, 6, 91–98. [Google Scholar] [CrossRef]
- Sherr, C.J.; Roberts, J.M. CDK inhibitors: Positive and negative regulators of G1-phase progression. Genes Dev. 1999, 13, 1501–1512. [Google Scholar] [CrossRef] [Green Version]
- Tommasino, M.; Crawford, L. Human Papillomavirus E6 and E7: Proteins which deregulate the cell cycle. BioEssays 1995, 17, 509–518. [Google Scholar] [CrossRef]
- Jones, D.L.; Münger, K. Analysis of the p53-mediated G1 growth arrest pathway in cells expressing the human papillomavirus type 16 E7 oncoprotein. J. Virol. 1997, 71, 2905–2912. [Google Scholar] [CrossRef] [Green Version]
- Reinstein, E.; Scheffner, M.; Oren, M.; Ciechanover, A.; Schwartz, A. Degradation of the E7 human papillomavirus oncoprotein by the ubiquitin-proteasome system: Targeting via ubiquitination of the N-terminal residue. Oncogene 2000, 19, 5944–5950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Munger, K.; Werness, B.A.; Dyson, N.; Phelps, W.C.; Harlow, E.; Howley, P.M. Complex formation of c-myc papillomavirus E7 proteins with the retinoblastoma tumor suppressor gene product. EMBO J. 1989, 8, 4099–4105. [Google Scholar] [CrossRef]
- Storey, A.; Pim, D.; Murray, A.; Osborn, K.; Banks, L.; Crawford, L. Comparison of the in vitro transforming activities of human papillomavirus types. EMBO J. 1988, 7, 1815–1820. [Google Scholar] [CrossRef]
- Roskoski, R. The ErbB/HER family of protein-tyrosine kinases and cancer. Pharmacol. Res. 2014, 79, 34–74. [Google Scholar] [CrossRef]
- Cha, D.; O’brien, P.; O’toole, E.A.; Woodley, D.T.; Hudson, L.G. Enhanced Modulation of Keratinocyte Motility by Transforming Growth Factor-a (TGF-a) Relative to Epidermal Growth Factor (EGF). J. Invest. Dermatol. 1996, 106, 590–597. [Google Scholar] [CrossRef] [Green Version]
- Yarden, Y. The EGFR family and its ligands in human cancer: Signalling mechanisms and therapeutic opportunities. Eur. J. Cancer 2001, 37, 3–8. [Google Scholar] [CrossRef]
- Franklin, W.A.; Veve, R.; Hirsch, F.R.; Helfrich, B.A.; Bunn, P.A. Epidermal growth factor receptor family in lung cancer and premalignancy. Semin. Oncol. 2002, 29, 3–14. [Google Scholar] [CrossRef]
- Hirsch, F.R.; Scagliotti, G.V.; Langer, C.J.; Varella-Garcia, M.; Franklin, W.A. Epidermal growth factor family of receptors in preneoplasia and lung cancer: Perspectives for targeted therapies. Lung Cancer 2003, 41, 29–42. [Google Scholar] [CrossRef]
- Makinoshima, H.; Takita, M.; Matsumoto, S.; Yagishita, A.; Owada, S.; Esumi, H.; Tsuchihara, K. Epidermal growth factor receptor (EGFR) signaling regulates global metabolic pathways in EGFR-mutated lung adenocarcinoma. J. Biol. Chem. 2014, 289, 20813–20823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Babic, I.; Anderson, E.S.; Tanaka, K.; Guo, D.; Masui, K.; Li, B.; Zhu, S.; Gu, Y.; Villa, G.R.; Akhavan, D.; et al. EGFR mutation-induced alternative splicing of max contributes to growth of glycolytic tumors in brain cancer. Cell Metab. 2013, 17, 1000–1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Makinoshima, H.; Takita, M.; Saruwatari, K.; Umemura, S.; Obata, Y.; Ishii, G.; Matsumoto, S.; Sugiyama, E.; Ochiai, A.; Abe, R.; et al. Signaling through the phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) axis is responsible for aerobic glycolysis mediated by glucose transporter in epidermal growth factor receptor (EGFR)-mutated lung adenocarcinoma. J. Biol. Chem. 2015, 290, 17495–17504. [Google Scholar] [CrossRef] [Green Version]
- Pfeiffer, D.; Stellwag, B.; Pfeiffer, A.; Borlinghaus, P.; Meier, W.; Scheidel, P. Clinical implications of the epidermal growth factor receptor in the squamous cell carcinoma of the uterine cervix. Gynecol. Oncol. 1989, 33, 146–150. [Google Scholar] [CrossRef]
- Ilahi, N.E.; Bhatti, A. Impact of HPV E5 on viral life cycle via EGFR signaling. Microb. Pathog. 2020, 139, 103923. [Google Scholar] [CrossRef]
- Kim, S.H.; Juhnn, Y.S.; Kang, S.; Park, S.W.; Sung, M.W.; Bang, Y.J.; Song, Y.S. Human papillomavirus 16 E5 up-regulates the expression of vascular endothelial growth factor through the activation of epidermal growth factor receptor, MEK/ERK1,2 and PI3K/Akt. Cell. Mol. Life Sci. 2006, 63, 930–938. [Google Scholar] [CrossRef]
- Kim, S.H.; Oh, J.M.; No, J.H.; Bang, Y.J.; Juhnn, Y.S.; Song, Y.S. Involvement of NF-κB and AP-1 in COX-2 upregulation by human papillomavirus 16 E5 oncoprotein. Carcinogenesis 2009, 30, 753–757. [Google Scholar] [CrossRef] [PubMed]
- Straight, S.W.; Hinkle, P.M.; Jewers, R.J.; McCance, D.J. The E5 oncoprotein of human papillomavirus type 16 transforms fibroblasts and effects the downregulation of the epidermal growth factor receptor in keratinocytes. J. Virol. 1993, 67, 4521–4532. [Google Scholar] [CrossRef] [Green Version]
- Crusius, K.; Auvinen, E.; Steuer, B.; Gaissert, H.; Alonso, A. The human papillomavirus type 16 E5-protein modulates ligand-dependent activation of the EGF receptor family in the human epithelial cell line HaCaT. Exp. Cell Res. 1998, 241, 76–83. [Google Scholar] [CrossRef]
- Topography of Early HPV 16 Transcription in High-Grade Genital Precancers-PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/2547317/ (accessed on 23 December 2020).
- Dassonville, O.; Formento, J.L.; Francoual, M.; Ramaioli, A.; Santini, J.; Schneider, M.; Demard, F.; Milano, G. Expression of epidermal growth factor receptor and survival in upper aerodigestive tract cancer. J. Clin. Oncol. 1993, 11, 1873–1878. [Google Scholar] [CrossRef]
- Buchholz, T.A.; Tu, X.; Ang, K.K.; Esteva, F.J.; Kuerer, H.M.; Pusztai, L.; Cristofanilli, M.; Singletary, S.E.; Hortobagyi, G.N.; Sahin, A.A. Epidermal growth factor receptor expression correlates with poor survival in patients who have breast carcinoma treated with doxorubicin-based neoadjuvant chemotherapy. Cancer 2005, 104, 676–681. [Google Scholar] [CrossRef] [PubMed]
- Sheridan, M.T.; O’Dwyer, T.; Seymour, C.B.; Mothersill, C.E. Potential indicators of radiosensitivity in squamous cell carcinoma of the head and neck. Radiat. Oncol. Investig. 1997, 5, 180–186. [Google Scholar] [CrossRef]
- Balaban, N.; Moni, J.; Shannon, M.; Dang, L.; Murphy, E.; Goldkorn, T. The effect of ionizing radiation on signal transduction: Antibodies to EGF receptor sensitize A431 cells to radiation. Biochim. Biophys. Acta Mol. Cell Res. 1996, 1314, 147–156. [Google Scholar] [CrossRef] [Green Version]
- Mehra, R.; Cohen, R.B.; Burtness, B.A. The role of cetuximab for the treatment of squamous cell carcinoma of the head and neck. Clin. Adv. Hematol. Oncol. 2008, 6, 742–750. [Google Scholar] [PubMed]
- Vermorken, J.B.; Psyrri, A.; Mesía, R.; Peyrade, F.; Beier, F.; De Blas, B.; Celik, I.; Licitra, L. Impact of tumor HPV status on outcome in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck receiving chemotherapy with or without cetuximab: Retrospective analysis of the phase iii extreme trial. Ann. Oncol. 2014, 25, 801–807. [Google Scholar] [CrossRef]
- Rosenthal, D.I.; Harari, P.M.; Giralt, J.; Bell, D.; Raben, D.; Liu, J.; Schulten, J.; Ang, K.K.; Bonner, J.A. Association of Human Papillomavirus and p16 Status with Outcomes in the IMCL-9815 Phase III Registration Trial for Patients with Locoregionally Advanced Oropharyngeal Squamous Cell Carcinoma of the Head and Neck Treated with Radiotherapy with or Without Cetuximab. J. Clin. Oncol. 2016, 34, 1300–1308. [Google Scholar] [CrossRef] [PubMed]
- Rodon, J.; Dienstmann, R.; Serra, V.; Tabernero, J. Development of PI3K inhibitors: Lessons learned from early clinical trials. Nat. Rev. Clin. Oncol. 2013, 10, 143–153. [Google Scholar] [CrossRef]
- Manning, B.D.; Toker, A. AKT/PKB Signaling: Navigating the Network. Cell 2017, 169, 381–405. [Google Scholar] [CrossRef] [Green Version]
- Saxton, R.A.; Sabatini, D.M. mTOR Signaling in Growth, Metabolism, and Disease. Cell 2017, 168, 960–976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lim, H.J.; Crowe, P.; Yang, J.L. Current clinical regulation of PI3K/PTEN/Akt/mTOR signalling in treatment of human cancer. J. Cancer Res. Clin. Oncol. 2015, 141, 671–689. [Google Scholar] [CrossRef]
- Lawrence, M.S.; Sougnez, C.; Lichtenstein, L.; Cibulskis, K.; Lander, E.; Gabriel, S.B.; Getz, G.; Ally, A.; Balasundaram, M.; Birol, I.; et al. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 2015, 517, 576–582. [Google Scholar] [CrossRef] [Green Version]
- Gillison, M.L.; Akagi, K.; Xiao, W.; Jiang, B.; Pickard, R.K.L.; Li, J.; Swanson, B.J.; Agrawal, A.D.; Zucker, M.; Stache-Crain, B.; et al. Human papillomavirus and the landscape of secondary genetic alterations in oral cancers. Genome Res. 2019, 29, 1–17. [Google Scholar] [CrossRef] [Green Version]
- Pim, D.; Massimi, P.; Dilworth, S.M.; Banks, L. Activation of the protein kinase B pathway by the HPV-16 E7 oncoprotein occurs through a mechanism involving interaction with PP2A. Oncogene 2005, 24, 7830–7838. [Google Scholar] [CrossRef] [Green Version]
- Menges, C.W.; Baglia, L.A.; Lapoint, R.; McCance, D.J. Human papillomavirus type 16 E7 up-regulates AKT activity through the retinoblastoma protein. Cancer Res. 2006, 66, 5555–5559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Contreras-Paredes, A.; De la Cruz-Hernández, E.; Martínez-Ramírez, I.; Dueñas-González, A.; Lizano, M. E6 variants of human papillomavirus 18 differentially modulate the protein kinase B/phosphatidylinositol 3-kinase (akt/PI3K) signaling pathway. Virology 2009, 383, 78–85. [Google Scholar] [CrossRef] [Green Version]
- Lu, Z.; Hu, X.; Li, Y.; Zheng, L.; Zhou, Y.; Jiang, H.; Ning, T.; Basang, Z.; Zhang, C.; Ke, Y. Human papillomavirus 16 E6 oncoprotein interferences with insulin signaling pathway by binding to tuberin. J. Biol. Chem. 2004, 279, 35664–35670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Callejas-Valera, J.L.; Iglesias-Bartolome, R.; Amornphimoltham, P.; Palacios-Garcia, J.; Martin, D.; Califano, J.A.; Molinolo, A.A.; Gutkind, J.S. mTOR inhibition prevents rapid-onset of carcinogen-induced malignancies in a novel inducible HPV-16 E6/E7 mouse model. Carcinogenesis 2016, 37, 1014–1025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Z.; Valera, J.C.; Zhao, X.; Chen, Q.; Silvio Gutkind, J. mTOR co-targeting strategies for head and neck cancer therapy. Cancer Metastasis Rev. 2017, 36, 491–502. [Google Scholar] [CrossRef] [Green Version]
- Day, T.A.; Shirai, K.; O’Brien, P.E.; Matheus, M.G.; Godwin, K.; Sood, A.J.; Kompelli, A.; Vick, J.A.; Martin, D.; Vitale-Cross, L.; et al. Inhibition of mTOR Signaling and Clinical Activity of Rapamycin in Head and Neck Cancer in a Window of Opportunity Trial. Clin. Cancer Res. 2019, 25, 1156–1164. [Google Scholar] [CrossRef] [Green Version]
- Harsha, C.; Banik, K.; Ang, H.L.; Girisa, S.; Vikkurthi, R.; Parama, D.; Rana, V.; Shabnam, B.; Khatoon, E.; Kumar, A.P.; et al. Targeting akt/mtor in oral cancer: Mechanisms and advances in clinical trials. Int. J. Mol. Sci. 2020, 21, 3285. [Google Scholar] [CrossRef]
- Zeke, A.; Misheva, M.; Reményi, A.; Bogoyevitch, M.A. JNK Signaling: Regulation and Functions Based on Complex Protein-Protein Partnerships. Microbiol. Mol. Biol. Rev. 2016, 80, 793–835. [Google Scholar] [CrossRef] [Green Version]
- Bode, A.M.; Dong, Z. The functional contrariety of JNK. Molecular Carcinogenesis 2007, 46, 591–598. [Google Scholar] [CrossRef] [Green Version]
- Hammouda, M.; Ford, A.; Liu, Y.; Zhang, J. The JNK Signaling Pathway in Inflammatory Skin Disorders and Cancer. Cells 2020, 9, 857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kyriakis, J.M.; Avruch, J. Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation. Physiol. Rev. 2001, 81, 807–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, X.; Destrument, A.; Tournier, C. Physiological roles of MKK4 and MKK7: Insights from animal models. Biochim. Biophys. Acta Mol. Cell Res. 2007, 1773, 1349–1357. [Google Scholar] [CrossRef] [PubMed]
- Chadee, D.N.; Kyriakis, J.M. Activation of SAPK/JNKs in vitro. Methods Mol. Biol. 2010, 661, 59–73. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.R.; Tan, T.H. The c-Jun N-terminal kinase pathway and apoptotic signaling (review). Int. J. Oncol. 2000, 16, 651–662. [Google Scholar] [CrossRef]
- Morgan, E.L.; Scarth, J.A.; Patterson, M.R.; Wasson, C.W.; Hemingway, G.C.; Barba-Moreno, D.; Macdonald, A. E6-mediated activation of JNK drives EGFR signalling to promote proliferation and viral oncoprotein expression in cervical cancer. Cell Death Differ. 2020. [Google Scholar] [CrossRef]
- Kohno, M.; Pouyssegur, J. Targeting the ERK signaling pathway in cancer therapy. Ann. Med. 2006, 38, 200–211. [Google Scholar] [CrossRef]
- Pages, G.; Lenormand, P.; L’Allemain, G.; Chambard, J.C.; Meloche, S.; Pouyssegur, J. Mitogen-activated protein kinases p42(mapk) and p44(mapk) are required for fibroblast proliferation. Proc. Natl. Acad. Sci. USA 1993, 90, 8319–8323. [Google Scholar] [CrossRef] [Green Version]
- Sun, Y.; Liu, W.Z.; Liu, T.; Feng, X.; Yang, N.; Zhou, H.F. Signaling pathway of MAPK/ERK in cell proliferation, differentiation, migration, senescence and apoptosis. J. Recept. Signal Transduct. 2015, 35, 600–604. [Google Scholar] [CrossRef]
- Lewis, T.S.; Shapiro, P.S.; Ahn, N.G. Signal transduction through MAP kinase cascades. Adv. Cancer Res. 1998, 74, 137–139. [Google Scholar] [CrossRef]
- Hoshino, R.; Chatani, Y.; Yamori, T.; Tsuruo, T.; Oka, H.; Yoshida, O.; Shimada, Y.; Ari-I, S.; Wada, H.; Fujimoto, J.; et al. Constitutive activation of the 41-/43-kDa mitogen-activated protein kinase signaling pathway in human tumors. Oncogene 1999, 18, 813–822. [Google Scholar] [CrossRef] [Green Version]
- Gioeli, D.; Mandell, J.W.; Petroni, G.R.; Frierson, H.F.; Weber, M.J. Activation of Mitogen-Activated Protein Kinase Associated with Prostate Cancer Progression. Cancer Res. 1999, 59, 279–284. [Google Scholar] [PubMed]
- Sebolt-Leopold, J.S.; Herrera, R. Targeting the mitogen-activated protein kinase cascade to treat cancer. Nat. Rev. Cancer 2004, 4, 937–947. [Google Scholar] [CrossRef] [PubMed]
- Xie, J.; Wang, S.; Li, Z.; Ao, C.; Wang, J.; Wang, L.; Peng, X.; Zeng, K. 5-aminolevulinic acid photodynamic therapy reduces HPV viral load via autophagy and apoptosis by modulating Ras/Raf/MEK/ERK and PI3K/AKT pathways in HeLa cells. J. Photochem. Photobiol. B Biol. 2019, 194, 46–55. [Google Scholar] [CrossRef]
- Morales-Garcia, V.; Contreras-Paredes, A.; Martinez-Abundis, E.; Gomez-Crisostomo, N.P.; Lizano, M.; Hernandez-Landero, F.; Cruz-Hernandez, E. The high-risk HPV E6 proteins modify the activity of the eIF4E protein via the MEK/ERK and AKT/PKB pathways. FEBS Open Biol. 2020, 10, 2541–2552. [Google Scholar] [CrossRef]
- Liu, F.; Lin, B.; Liu, X.; Zhang, W.; Zhang, E.; Hu, L.; Ma, Y.; Li, X.; Tang, X. ERK signaling pathway is involved in HPV-16 E6 but not E7 oncoprotein-induced HIF-1α protein accumulation in NSCLC cells. Oncol. Res. 2016, 23, 109–118. [Google Scholar] [CrossRef] [PubMed]
- Bowser, B.S.; Alam, S.; Meyers, C. Treatment of a Human Papillomavirus Type 31b-Positive Cell Line with Benzo[a]Pyrene Increases Viral Titer through Activation of the Erk1/2 Signaling Pathway. J. Virol. 2011, 85, 4982–4992. [Google Scholar] [CrossRef] [Green Version]
- Eferl, R.; Wagner, E.F. AP-1: A double-edged sword in tumorigenesis. Nat. Rev. Cancer 2003, 3, 859–868. [Google Scholar] [CrossRef]
- Gazon, H.; Barbeau, B.; Mesnard, J.M.; Peloponese, J.M. Hijacking of the AP-1 signaling pathway during development of ATL. Front. Microbiol. 2018, 8, 2686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nakabeppu, Y.; Ryder, K.; Nathans, D. DNA binding activities of three murine Jun proteins: Stimulation by Fos. Cell 1988, 55, 907–915. [Google Scholar] [CrossRef]
- Rauscher, F.J.; Cohen, D.R.; Curran, T.; Bos, T.J.; Vogt, P.K.; Bohmann, D.; Tjian, R.; Franza, B.R. Fos-associated protein p39 is the product of the jun proto-oncogene. Science 1988, 240, 1010–1016. [Google Scholar] [CrossRef] [PubMed]
- Angel, P.; Karin, M. The role of Jun, Fos and the AP-1 complex in cell-proliferation and transformation. BBA Rev. Cancer 1991, 1072, 129–157. [Google Scholar] [CrossRef]
- Hibi, M.; Lin, A.; Smeal, T.; Minden, A.; Karin, M. Identification of an oncoprotein- and UV-responsive protein kinase that binds and potentiates the c-Jun activation domain. Genes Dev. 1993, 7, 2135–2148. [Google Scholar] [CrossRef] [Green Version]
- Monje, P.; Hernández-Losa, J.; Lyons, R.J.; Castellone, M.D.; Gutkind, J.S. Regulation of the transcriptional activity of c-Fos by ERK: A novel role for the prolyl isomerase Pin1. J. Biol. Chem. 2005, 280, 35081–35084. [Google Scholar] [CrossRef] [Green Version]
- Monje, P.; Marinissen, M.J.; Gutkind, J.S. Phosphorylation of the Carboxyl-Terminal Transactivation Domain of c-Fos by Extracellular Signal-Regulated Kinase Mediates the Transcriptional Activation of AP-1 and Cellular Transformation Induced by Platelet-Derived Growth Factor. Mol. Cell. Biol. 2003, 23, 7030–7043. [Google Scholar] [CrossRef] [Green Version]
- Treisman, R. Ternary complex factors: Growth factor regulated transcriptional activators. Curr. Opin. Genet. Dev. 1994, 4, 96–101. [Google Scholar] [CrossRef]
- Murphy, L.O.; Smith, S.; Chen, R.H.; Fingar, D.C.; Blenis, J. Molecular, interpretation of ERK signal duration by immediate early gene products. Nat. Cell Biol. 2002, 4, 556–564. [Google Scholar] [CrossRef] [PubMed]
- Steger, G.; Rehtanz, M.; Schnabel, C. Identification of a promoter in position 56 within the long control region of human papillomavirus type 18. Arch. Virol. 2001, 146, 2069–2084. [Google Scholar] [CrossRef]
- Rösl, F.; Das, B.C.; Lengert, M.; Geletneky, K.; zur Hausen, H. Antioxidant-induced changes of the AP-1 transcription complex are paralleled by a selective suppression of human papillomavirus transcription. J. Virol. 1997, 71, 362–370. [Google Scholar] [CrossRef] [Green Version]
- Soto, U.; Das, B.C.; Lengert, M.; Finzer, P.; Zur Hausen, H.; Rösl, F. Conversion of HPV 18 positive non-tumorigenic HeLa-fibroblast hybrids to invasive growth involves loss of TNF-α mediated repression of viral transcription and modification of the AP-1 transcription complex. Oncogene 1999, 18, 3187–3198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dhandapani, K.M.; Mahesh, V.B.; Brann, D.W. Curcumin suppresses growth and chemoresistance of human glioblastoma cells via AP-1 and NFκB transcription factors. J. Neurochem. 2007, 102, 522–538. [Google Scholar] [CrossRef]
- Muñoz, J.P.; Carrillo-Beltrán, D.; Aedo-Aguilera, V.; Calaf, G.M.; León, O.; Maldonado, E.; Tapia, J.C.; Boccardo, E.; Ozbun, M.A.; Aguayo, F. Tobacco Exposure Enhances Human Papillomavirus 16 Oncogene Expression via EGFR/PI3K/Akt/c-Jun Signaling Pathway in Cervical Cancer Cells. Front. Microbiol. 2018, 9. [Google Scholar] [CrossRef]
- Mahata, S.; Bharti, A.C.; Shukla, S.; Tyagi, A.; Husain, S.A.; Das, B.C. Berberine modulates AP-1 activity to suppress HPV transcription and downstream signaling to induce growth arrest and apoptosis in cervical cancer cells. Mol. Cancer 2011, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Warowicka, A.; Nawrot, R.; Goździcka-Józefiak, A. Antiviral activity of berberine. Arch. Virol. 2020, 165, 1935–1945. [Google Scholar] [CrossRef] [PubMed]
- Gross, A.S.; Graef, M. Mechanisms of Autophagy in Metabolic Stress Response. J. Mol. Biol. 2020, 432, 28–52. [Google Scholar] [CrossRef]
- Levy, J.M.M.; Towers, C.G.; Thorburn, A. Targeting autophagy in cancer. Nat. Rev. Cancer 2017, 17, 528–542. [Google Scholar] [CrossRef]
- AM, C.; SW, R.; B, L. Autophagy in Human Health and Disease. N. Engl. J. Med. 2013, 368, 1845–1846. [Google Scholar] [CrossRef]
- Hamasaki, M.; Furuta, N.; Matsuda, A.; Nezu, A.; Yamamoto, A.; Fujita, N.; Oomori, H.; Noda, T.; Haraguchi, T.; Hiraoka, Y.; et al. Autophagosomes form at ER-mitochondria contact sites. Nature 2013, 495, 389–393. [Google Scholar] [CrossRef]
- Stanley, R.E.; Ragusa, M.J.; Hurley, J.H. The beginning of the end: How scaffolds nucleate autophagosome biogenesis. Trends Cell Biol. 2014, 24, 73–81. [Google Scholar] [CrossRef] [Green Version]
- Mizushima, N.; Yoshimori, T.; Levine, B. Methods in Mammalian Autophagy Research. Cell 2010, 140, 313–326. [Google Scholar] [CrossRef] [Green Version]
- Liang, C.; Lee, J.S.; Inn, K.S.; Gack, M.U.; Li, Q.; Roberts, E.A.; Vergne, I.; Deretic, V.; Feng, P.; Akazawa, C.; et al. Beclin1-binding UVRAG targets the class C Vps complex to coordinate autophagosome maturation and endocytic trafficking. Nat. Cell Biol. 2008, 10, 776–787. [Google Scholar] [CrossRef] [Green Version]
- Mattoscio, D.; Medda, A.; Chiocca, S. Human papilloma virus and autophagy. Int. J. Mol. Sci. 2018, 19, 1775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mattoscio, D.; Casadio, C.; Miccolo, C.; Maffini, F.; Raimondi, A.; Tacchetti, C.; Gheit, T.; Tagliabue, M.; Galimberti, V.E.; De Lorenzi, F.; et al. Autophagy regulates UBC9 levels during viral-mediated tumorigenesis. PLOS Pathog. 2017, 13, e1006262. [Google Scholar] [CrossRef]
- Antonioli, M.; Pagni, B.; Vescovo, T.; Ellis, R.; Cosway, B.; Rollo, F.; Bordoni, V.; Agrati, C.; Labus, M.; Covello, R.; et al. HPV sensitizes OPSCC cells to cisplatin-induced apoptosis by inhibiting autophagy through E7-mediated degradation of AMBRA1. Autophagy 2020, 1–13. [Google Scholar] [CrossRef]
- Hatakeyama, H.; Mizumachi, T.; Sakashita, T.; Kano, S.; Homma, A.; Fukuda, S. Epithelial-mesenchymal transition in human papillomavirus-positive and -negative oropharyngeal squamous cell carcinoma. Oncol. Rep. 2014, 32, 2673–2679. [Google Scholar] [CrossRef]
- Lefevre, M.; Rousseau, A.; Rayon, T.; Dalstein, V.; Clavel, C.; Beby-Defaux, A.; Pretet, J.L.; Soussan, P.; Polette, M.; Lacau Saint Guily, J.; et al. Epithelial to mesenchymal transition and HPV infection in squamous cell oropharyngeal carcinomas: The papillophar study. Br. J. Cancer 2017, 116, 362–369. [Google Scholar] [CrossRef] [Green Version]
- Ihler, F.; Gratz, R.; Wolff, H.A.; Weiss, B.G.; Bertlich, M.; Kitz, J.; Salinas, G.; Rave-Fränk, M.; Canis, M. Epithelial-Mesenchymal Transition during Metastasis of HPV-Negative Pharyngeal Squamous Cell Carcinoma. Biomed. Res. Int. 2018, 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Invest. 2009, 119, 1420–1428. [Google Scholar] [CrossRef] [Green Version]
- Graves, C.A.; Abboodi, F.F.; Tomar, S.; Wells, J.; Pirisi, L. The translational significance of epithelial-mesenchymal transition in head and neck cancer. Clin. Transl. Med. 2014, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- van der Heijden, M.; Essers, P.B.M.; Verhagen, C.V.M.; Willems, S.M.; Sanders, J.; de Roest, R.H.; Vossen, D.M.; Leemans, C.R.; Verheij, M.; Brakenhoff, R.H.; et al. Epithelial-to-mesenchymal transition is a prognostic marker for patient outcome in advanced stage HNSCC patients treated with chemoradiotherapy. Radiother. Oncol. 2020, 147, 186–194. [Google Scholar] [CrossRef]
- Mirantes, C.; Espinosa, I.; Ferrer, I.; Dolcet, X.; Prat, J.; Matias-Guiu, X. Epithelial-to-mesenchymal transition and stem cells in endometrial cancer. Hum. Pathol. 2013, 44, 1973–1981. [Google Scholar] [CrossRef]
- Campo, L.; Zhang, C.; Breuer, E.K. EMT-inducing molecular factors in gynecological cancers. Biomed. Res. Int. 2015, 2015, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Rodrigues, I.S.; Lavorato-Rocha, A.M.; De M Maia, B.; Stiepcich, M.M.A.; De Carvalho, F.M.; Baiocchi, G.; Soares, F.A.; Rocha, R.M. Epithelial-mesenchymal transition-like events in vulvar cancer and its relation with HPV. Br. J. Cancer 2013, 109, 184–194. [Google Scholar] [CrossRef] [Green Version]
- Hu, D.; Zhou, J.; Wang, F.; Shi, H.; Li, Y.; Li, B. HPV-16 E6/E7 promotes cell migration and invasion in cervical cancer via regulating cadherin switch in vitro and in vivo. Arch. Gynecol. Obstet. 2015, 292, 1345–1354. [Google Scholar] [CrossRef]
- Semb, H.; Christofori, G. The tumor-suppressor function of E-cadherin. Am. J. Hum. Genet. 1998, 63, 1588–1593. [Google Scholar] [CrossRef] [Green Version]
- Loh, C.Y.; Chai, J.Y.; Tang, T.F.; Wong, W.F.; Sethi, G.; Shanmugam, M.K.; Chong, P.P.; Looi, C.Y. The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells 2019, 8, 1118. [Google Scholar] [CrossRef] [Green Version]
- Maître, J.L.; Heisenberg, C.P. Three functions of cadherins in cell adhesion. Curr. Biol. 2013, 23, 626–633. [Google Scholar] [CrossRef] [Green Version]
- Perrais, M.; Chen, X.; Perez-Moreno, M.; Gumbiner, B.M. E-cadherin homophilic ligation inhibits cell growth and epidermal growth factor receptor signaling independently of other cell interactions. Mol. Biol. Cell 2007, 18, 2013–2025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, J.; Li, X.; Yin, X.; Zhang, J.; Shi, B. Association of low expression of e-cadherin and β-catenin with the progression of early stage human squamous cervical cancer. Oncol. Lett. 2019, 17, 5729–5739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Laurson, J.; Khan, S.; Chung, R.; Cross, K.; Raj, K. Epigenetic repression of E-cadherin by human papillomavirus 16 E7 protein. Carcinogenesis 2010, 31, 918–926. [Google Scholar] [CrossRef] [Green Version]
- Kramer, B.; Hock, C.; Schultz, J.D.; Lammert, A.; Kuhlin, B.; Birk, R.; Hormann, K.; Aderhold, C. Impact of small molecules on β-catenin and E-cadherin expression in HPV16-positive and -negative squamous cell carcinomas. Anticancer Res. 2017, 37, 2845–2852. [Google Scholar] [CrossRef] [Green Version]
- MacDonald, B.T.; Tamai, K.; He, X. Wnt/β-Catenin Signaling: Components, Mechanisms, and Diseases. Dev. Cell 2009, 17, 9–26. [Google Scholar] [CrossRef] [Green Version]
- Bathaie, S.Z.; Faridi, N.; Nasimian, A.; Heidarzadeh, H.; Tamanoi, F. How Phytochemicals Prevent Chemical Carcinogens and/or Suppress Tumor Growth. Enzymes 2015, 37, 1–42. [Google Scholar] [CrossRef] [PubMed]
- Chakraborty, B.; Mukhopadhyay, D.; Roychowdhury, A.; Basu, M.; Alam, N.; Chatterjee, K.; Chakrabarti, J.; Panda, C.K. Differential Wnt-β- catenin pathway activation in HPV positive and negative oral epithelium is transmitted during head and neck tumorigenesis: Clinical implications. Med. Microbiol. Immunol. 2020. [Google Scholar] [CrossRef]
- Ayala-Calvillo, E.; Mojica-Vázquez, L.H.; García-Carrancá, A.; González-Maya, L. Wnt/p-catenin pathway activation and silencing of the APC gene in HPV-positive human cervical cancer-derived cells. Mol. Med. Rep. 2018, 17, 200–208. [Google Scholar] [CrossRef] [PubMed]
- Polakis, P. Wnt signaling in cancer. Cold Spring Harb. Perspect. Biol. 2012, 4, 9. [Google Scholar] [CrossRef] [Green Version]
- Rampias, T.; Boutati, E.; Pectasides, E.; Sasaki, C.; Kountourakis, P.; Weinberger, P.; Psyrri, A. Activation of Wnt signaling pathway by human papillomavirus E6 and E7 oncogenes in HPV16-positive oropharyngeal squamous carcinoma cells. Mol. Cancer Res. 2010, 8, 433–443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tormoen, G.W.; Crittenden, M.R.; Gough, M.J. Role of the immunosuppressive microenvironment in immunotherapy. Adv. Radiat. Oncol. 2018, 3, 520–526. [Google Scholar] [CrossRef] [Green Version]
- Paijens, S.T.; Vledder, A.; de Bruyn, M.; Nijman, H.W. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell. Mol. Immunol. 2020. [Google Scholar] [CrossRef]
- Jewett, A.; Kos, J.; Kaur, K.; Safaei, T.; Sutanto, C.; Chen, W.; Wong, P.; Namagerdi, A.K.; Fang, C.; Fong, Y.; et al. Natural Killer Cells: Diverse Functions in Tumor Immunity and Defects in Pre-neoplastic and Neoplastic Stages of Tumorigenesis. Mol. Ther. Oncolytics 2020, 16, 41–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Whiteside, T.L. The tumor microenvironment and its role in promoting tumor growth. Oncogene 2008, 27, 5904–5912. [Google Scholar] [CrossRef] [Green Version]
- Saloura, V.; Izumchenko, E.; Zuo, Z.; Bao, R.; Korzinkin, M.; Ozerov, I.; Zhavoronkov, A.; Sidransky, D.; Bedi, A.; Hoque, M.O.; et al. Immune profiles in primary squamous cell carcinoma of the head and neck. Oral Oncol. 2019, 96, 77–88. [Google Scholar] [CrossRef] [PubMed]
- Farhood, B.; Najafi, M.; Mortezaee, K. CD8+ cytotoxic T lymphocytes in cancer immunotherapy: A review. J. Cell. Physiol. 2019, 234, 8509–8521. [Google Scholar] [CrossRef]
- Wang, M.; Wang, J.; Wang, R.; Jiao, S.; Wang, S.; Zhang, J.; Zhang, M. Identification of a monoclonal antibody that targets PD-1 in a manner requiring PD-1 Asn58 glycosylation. Commun. Biol. 2019, 2. [Google Scholar] [CrossRef] [Green Version]
- Messina, J.L.; Fenstermacher, D.A.; Eschrich, S.; Qu, X.; Berglund, A.E.; Lloyd, M.C.; Schell, M.J.; Sondak, V.K.; Weber, J.S.; Mulé, J.J. 12-chemokine gene signature identifies lymph node-like structures in melanoma: Potential for patient selection for immunotherapy? Sci. Rep. 2012, 2. [Google Scholar] [CrossRef] [Green Version]
- Vandermark, E.R.; Deluca, K.A.; Gardner, C.R.; Marker, D.F.; Schreiner, C.N.; Strickland, D.A.; Wilton, K.M.; Mondal, S.; Woodworth, C.D. Human papillomavirus type 16 E6 and E 7 proteins alter NF-kB in cultured cervical epithelial cells and inhibition of NF-kB promotes cell growth and immortalization. Virology 2012, 425, 53–60. [Google Scholar] [CrossRef] [Green Version]
- Tilborghs, S.; Corthouts, J.; Verhoeven, Y.; Arias, D.; Rolfo, C.; Trinh, X.B.; van Dam, P.A. The role of Nuclear Factor-kappa B signaling in human cervical cancer. Crit. Rev. Oncol. Hematol. 2017, 120, 141–150. [Google Scholar] [CrossRef] [PubMed]
- Lin, Y.; Bai, L.; Chen, W.; Xu, S. The NF-κB activation pathways, emerging molecular targets for cancer prevention and therapy. Expert Opin. Ther. Targets 2010, 14, 45–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Byg, L.M.; Vidlund, J.; Vasiljevic, N.; Clausen, D.; Forslund, O.; Norrild, B. NF-κB signalling is attenuated by the E7 protein from cutaneous human papillomaviruses. Virus Res. 2012, 169, 48–53. [Google Scholar] [CrossRef]
- Xu, M.; Katzenellenbogen, R.A.; Grandori, C.; Galloway, D.A. NFX1 Plays a Role in Human Papillomavirus Type 16 E6 Activation of NFκB Activity. J. Virol. 2010, 84, 11461–11469. [Google Scholar] [CrossRef] [Green Version]
- An, J.; Mo, D.; Liu, H.; Veena, M.S.; Srivatsan, E.S.; Massoumi, R.; Rettig, M.B. Inactivation of the CYLD Deubiquitinase by HPV E6 Mediates Hypoxia-Induced NF-κB Activation. Cancer Cell 2008, 14, 394–407. [Google Scholar] [CrossRef] [Green Version]
- Guttridge, D.C.; Albanese, C.; Reuther, J.Y.; Pestell, R.G.; Baldwin, A.S. NF-κB Controls Cell Growth and Differentiation through Transcriptional Regulation of Cyclin D1. Mol. Cell. Biol. 1999, 19, 5785–5799. [Google Scholar] [CrossRef] [Green Version]
- Rebhandl, S.; Huemer, M.; Greil, R.; Geisberger, R. AID/APOBEC deaminases and cancer. Oncoscience 2015, 2, 320–333. [Google Scholar] [CrossRef] [Green Version]
- Gutiérrez-Hoya, A.; Soto-Cruz, I. Role of the JAK/STAT Pathway in Cervical Cancer: Its Relationship with HPV E6/E7 Oncoproteins. Cells 2020, 9, 2297. [Google Scholar] [CrossRef] [PubMed]
- Hong, S.; Laimins, L.A. The JAK-STAT Transcriptional Regulator, STAT-5, Activates the ATM DNA Damage Pathway to Induce HPV 31 Genome Amplification upon Epithelial Differentiation. PLoS Pathog. 2013, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barnard, P.; McMillan, N.A.J. The human papillomavirus E7 oncoprotein abrogates signaling mediated by interferon-α. Virology 1999, 259, 305–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morgan, E.L.; Macdonald, A. Manipulation of JAK/STAT signalling by high-risk HPVs: Potential therapeutic targets for HPV-associated malignancies. Viruses 2020, 12, 977. [Google Scholar] [CrossRef]
- Morgan, E.L.; Macdonald, A. Autocrine STAT3 activation in hpv positive cervical cancer through a virus-driven Rac1— NFκB—IL-6 signalling axis. PLoS Pathog. 2019, 15. [Google Scholar] [CrossRef] [Green Version]
- Morgan, E.L.; Wasson, C.W.; Hanson, L.; Kealy, D.; Pentland, I.; McGuire, V.; Scarpini, C.; Coleman, N.; Arthur, J.S.C.; Parish, J.L.; et al. STAT3 activation by E6 is essential for the differentiation-dependent HPV18 life cycle. PLoS Pathog. 2018, 14. [Google Scholar] [CrossRef]
- Furtek, S.L.; Backos, D.S.; Matheson, C.J.; Reigan, P. Strategies and Approaches of Targeting STAT3 for Cancer Treatment. ACS Chem. Biol. 2016, 11, 308–318. [Google Scholar] [CrossRef]
- Lau, Y.T.K.; Ramaiyer, M.; Johnson, D.E.; Grandis, J.R. Targeting STAT3 in cancer with nucleotide therapeutics. Cancers 2019, 11, 1681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gaykalova, D.A.; Manola, J.B.; Ozawa, H.; Zizkova, V.; Morton, K.; Bishop, J.A.; Sharma, R.; Zhang, C.; Michailidi, C.; Considine, M.; et al. NF-κB and stat3 transcription factor signatures differentiate HPV-positive and HPV-negative head and neck squamous cell carcinoma. Int. J. Cancer 2015, 137, 1879–1889. [Google Scholar] [CrossRef] [PubMed]
- Barbisan, G.; Pérez, L.O.; Contreras, A.; Golijow, C.D. TNF-α and IL-10 promoter polymorphisms, HPV infection, and cervical cancer risk. Tumor Biol. 2012, 33, 1549–1556. [Google Scholar] [CrossRef] [PubMed]
- Boccardo, E.; Noya, F.; Broker, T.R.; Chow, L.T.; Villa, L.L. HPV-18 confers resistance to TNF-α in organotypic cultures of human keratinocytes. Virology 2004, 328, 233–243. [Google Scholar] [CrossRef] [Green Version]
- Villa, L.L.; Vieira, K.B.L.; Pei, X. -F.; Schlegel, R. Differential effect of tumor necrosis factor on proliferation of primary human keratinocytes and cell lines containing human papillomavirus types 16 and 18. Mol. Carcinog. 1992, 6, 5–9. [Google Scholar] [CrossRef] [PubMed]
- Dong, W.; Sun, S.; Cao, X.; Cui, Y.; Chen, A.; Li, X.; Zhang, J.; Cao, J.; Wang, Y. Exposure to TNF-α combined with TGF-β induces carcinogenesis in vitro via NF-κB/Twist axis. Oncol. Rep. 2017, 37, 1873–1882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Polz-Dacewicz, M.; Strycharz-Dudziak, M.; Dworzański, J.; Stec, A.; Kocot, J. Salivary and serum IL-10, TNF-α, TGF-β, VEGF levels in oropharyngeal squamous cell carcinoma and correlation with HPV and EBV infections. Infect. Agent. Cancer 2016, 11. [Google Scholar] [CrossRef] [Green Version]
- Sannigrahi, M.K.; Sharma, R.; Singh, V.; Panda, N.K.; Rattan, V.; Khullar, M. DNA methylation regulated microRNAs in HPV-16-induced head and neck squamous cell carcinoma (HNSCC). Mol. Cell. Biochem. 2018, 448, 321–333. [Google Scholar] [CrossRef]
- John, K.; Wu, J.; Lee, B.W.; Farah, C.S. MicroRNAs in head and neck cancer. Int. J. Dent. 2013, 2013, 650218. [Google Scholar] [CrossRef]
- Lajer, C.B.; Garnæs, E.; Friis-Hansen, L.; Norrild, B.; Therkildsen, M.H.; Glud, M.; Rossing, M.; Lajer, H.; Svane, D.; Skotte, L.; et al. The role of miRNAs in human papilloma virus (HPV)-associated cancers: Bridging between HPV-related head and neck cancer and cervical cancer. Br. J. Cancer 2012, 106, 1526–1534. [Google Scholar] [CrossRef] [PubMed]
- Sadri Nahand, J.; Moghoofei, M.; Salmaninejad, A.; Bahmanpour, Z.; Karimzadeh, M.; Nasiri, M.; Mirzaei, H.R.; Pourhanifeh, M.H.; Bokharaei-Salim, F.; Mirzaei, H.; et al. Pathogenic role of exosomes and microRNAs in HPV-mediated inflammation and cervical cancer: A review. Int. J. Cancer 2020, 146, 305–320. [Google Scholar] [CrossRef]
- Hasanzadeh, M.; Movahedi, M.; Rejali, M.; Maleki, F.; Moetamani-Ahmadi, M.; Seifi, S.; Hosseini, Z.; Khazaei, M.; Amerizadeh, F.; Ferns, G.A.; et al. The potential prognostic and therapeutic application of tissue and circulating microRNAs in cervical cancer. J. Cell. Physiol. 2019, 234, 1289–1294. [Google Scholar] [CrossRef]
- Shishodia, G.; Shukla, S.; Srivastava, Y.; Masaldan, S.; Mehta, S.; Bhambhani, S.; Sharma, S.; Mehrotra, R.; Das, B.C.; Bharti, A.C. Alterations in microRNAs miR-21 and let-7a correlate with aberrant STAT3 signaling and downstream effects during cervical carcinogenesis. Mol. Cancer 2015, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sannigrahi, M.K.; Sharma, R.; Singh, V.; Panda, N.K.; Rattan, V.; Khullar, M. Role of host miRNA Hsa-miR-139-3p in HPV-16–induced carcinomas. Clin. Cancer Res. 2017, 23, 3884–3895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, H.; Zhang, D.; Chen, Q.; Hong, Y. Plasma expression of miRNA-21,-214, -34a, and -200a in patients with persistent HPV infection and cervical lesions. BMC Cancer 2019, 19. [Google Scholar] [CrossRef]
- Eades, G.; Yao, Y.; Yang, M.; Zhang, Y.; Chumsri, S.; Zhou, Q. miR-200a regulates SIRT1 expression and Epithelial to Mesenchymal Transition (EMT)-like transformation in mammary epithelial cells. J. Biol. Chem. 2011, 286, 25992–26002. [Google Scholar] [CrossRef] [Green Version]
- Božinović, K.; Sabol, I.; Dediol, E.; Milutin Gašperov, N.; Manojlović, S.; Vojtechova, Z.; Tachezy, R.; Grce, M. Genome-wide miRNA profiling reinforces the importance of miR-9 in human papillomavirus associated oral and oropharyngeal head and neck cancer. Sci. Rep. 2019, 9. [Google Scholar] [CrossRef]
- Zeng, K.; Zhang, W.; Hu, X. Progress of research in miR-218 and cervical cancer. Chin. Ger. J. Clin. Oncol. 2013, 12, 399–402. [Google Scholar] [CrossRef]
- HPV E6/p53 Mediated Down-Regulation of miR-34a Inhibits Warburg Effect Through Targeting LDHA in Cervical Cancer-PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/27186405/ (accessed on 26 December 2020).
- Spriggs, C.C.; Blanco, L.Z.; Maniar, K.P.; Laimins, L.A. Expression of HPV-induced DNA damage repair factors correlates with CIN progression. Int. J. Gynecol. Pathol. 2019, 38, 1–10. [Google Scholar] [CrossRef]
- Moody, C.A.; Laimins, L.A. Human papillomaviruses activate the ATM DNA damage pathway for viral genome amplification upon differentiation. PLoS Pathog. 2009, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Banerjee, N.S.; Moore, D.; Parker, C.J.; Broker, T.R.; Chow, L.T. Targeting dna damage response as a strategy to treat hpv infections. Int. J. Mol. Sci. 2019, 20, 5455. [Google Scholar] [CrossRef] [Green Version]
- Zhao, J.; Guo, Z.; Wang, Q.; Si, T.; Pei, S.; Qu, H.; Shang, L.; Yang, Y.; Wang, L. HPV infection associated DNA damage correlated with cervical precancerous lesions and cancer in the highest area of cervical cancer mortality, Longnan, China. Cancer Manag. Res. 2019, 11, 7197–7210. [Google Scholar] [CrossRef] [Green Version]
- Cortés Gutiérrez, E.I.; García-Vielma, C.; Aguilar-Lemarroy, A.; Vallejo-Ruíz, V.; Piña-Sánchez, P.; Zapata-Benavides, P.; Gosálvez, J. Expression of the HPV18/E6 oncoprotein induces DNA damage. Eur. J. Histochem. 2017, 61, 106–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nickson, C.M.; Moori, P.; Carter, R.J.; Rubbi, C.P.; Parsons, J.L. Misregulation of DNA damage repair pathways in HPV-positive head and neck squamous cell carcinoma contributes to cellular radiosensitivity. Oncotarget 2017, 8, 29963–29975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Verhees, F.; Legemaate, D.; Demers, I.; Jacobs, R.; Haakma, W.E.; Rousch, M.; Kremer, B.; Speel, E.J. The antiviral agent cidofovir induces DNA damage and mitotic catastrophe in HPV-positive and-negative head and neck squamous cell carcinomas in vitro. Cancers 2019, 11, 919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ekanayake Weeramange, C.; Tang, K.D.; Vasani, S.; Langton-Lockton, J.; Kenny, L.; Punyadeera, C. DNA Methylation Changes in Human Papillomavirus-Driven Head and Neck Cancers. Cells 2020, 9, 1359. [Google Scholar] [CrossRef] [PubMed]
- Hsu, Y.W.; Huang, R.L.; Su, P.H.; Chen, Y.C.; Wang, H.C.; Liao, C.C.; Lai, H.C. Genotype-specific methylation of HPV in cervical intraepithelial neoplasia. J. Gynecol. Oncol. 2017, 28. [Google Scholar] [CrossRef] [Green Version]
- Roy, D.; Tiirikainen, M. Diagnostic Power of DNA Methylation Classifiers for Early Detection of Cancer. Trends Cancer 2020, 6, 78–81. [Google Scholar] [CrossRef]
- Kim, K.; Garner-Hamrick, P.A.; Fisher, C.; Lee, D.; Lambert, P.F. Methylation Patterns of Papillomavirus DNA, Its Influence on E2 Function, and Implications in Viral Infection. J. Virol. 2003, 77, 12450–12459. [Google Scholar] [CrossRef] [Green Version]
- Amaro-Filho, S.M.; Pereira Chaves, C.B.; Felix, S.P.; Basto, D.L.; de Almeida, L.M.; Moreira, M.A.M. HPV DNA methylation at the early promoter and E1/E2 integrity: A comparison between HPV16, HPV18 and HPV45 in cervical cancer. Papillomavirus Res. 2018, 5, 172–179. [Google Scholar] [CrossRef]
- von Knebel Doeberitz, M.; Prigge, E.S. Role of DNA methylation in HPV associated lesions. Papillomavirus Res. 2019, 7, 180–183. [Google Scholar] [CrossRef]
- Clarke, M.A.; Gradissimo, A.; Schiffman, M.; Lam, J.; Sollecito, C.C.; Fetterman, B.; Lorey, T.; Poitras, N.; Raine-Bennett, T.R.; Castle, P.E.; et al. Human papillomavirus DNA methylation as a biomarker for cervical precancer: Consistency across 12 genotypes and potential impact on management of hpv-positive women. Clin. Cancer Res. 2018, 24, 2194–2202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Giuliano, A.R.; Nedjai, B.; Lorincz, A.T.; Schell, M.J.; Rahman, S.; Banwait, R.; Boulware, D.; Sirak, B.; Martin-Gomez, L.; Abrahamsen, M.; et al. Methylation of HPV 16 and EPB41L3 in oral gargles: Associations with oropharyngeal cancer detection and tumor characteristics. Int. J. Cancer 2020, 146, 1018–1030. [Google Scholar] [CrossRef] [PubMed]
- Harper, D.M.; DeMars, L.R. HPV vaccines–A review of the first decade. Gynecol. Oncol. 2017, 146, 196–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Low Risk | High Risk | |
---|---|---|
HPV Type | 1, 6, 10, 11, 32, 42, 44 | 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 66, 68 |
Associated Disease | Anogenital warts Cutaneous warts Recurrent respiratory papillomatosis Heck’s disease | Intraepithelial neoplasia Invasive carcinoma: HNSCC, Cervical cancer, Anogenital cancers, Non-melanoma skin cancer |
miRNA | Viral Oncoprotein | Pathways Involved | Role in Tumorigenesis | References |
---|---|---|---|---|
miR-200a | HPV-16 E6/E7 | EMT | Downregulated | Wang et al., 2019 [175] |
Downregulation prevents EMT inhibition | Eades et al., 2011 [176] | |||
miR-9 | HPV-16 E6 | Cell metabolism | Upregulated in recurring HNSCC and cervical cancer | Božinović et al., 2019 [177] |
miR-7a, miR-21 | HPV-16 E6 | JAK/STAT | Maintain STAT3 activated in HPV-positive cells | Shishodia et al., 2015 [173] |
miR-29 | HPV-16 E6/E7 | Often downregulated. | Božinović et al., 2019 [177] | |
miR-218 | HPV-16 E6 | PI3K/Akt pathway, cell-cycle | Upregulates expression of the LAMB3 epithelial cell marker. | Zheng et al., 2013 [178] |
miR-34a | HPV-16 E6 | p53-dependent pathway | Downregulated | Zhang et al., 2016 [179] |
Hsa-miR-139-3p | HPV-16 E1, E6/E7 | p-53, cell-cycle | Upregulation restores p53 expression and inhibits E6/E7. | Sannigrahi et al., 2017 [174] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Medda, A.; Duca, D.; Chiocca, S. Human Papillomavirus and Cellular Pathways: Hits and Targets. Pathogens 2021, 10, 262. https://doi.org/10.3390/pathogens10030262
Medda A, Duca D, Chiocca S. Human Papillomavirus and Cellular Pathways: Hits and Targets. Pathogens. 2021; 10(3):262. https://doi.org/10.3390/pathogens10030262
Chicago/Turabian StyleMedda, Alessandro, Daria Duca, and Susanna Chiocca. 2021. "Human Papillomavirus and Cellular Pathways: Hits and Targets" Pathogens 10, no. 3: 262. https://doi.org/10.3390/pathogens10030262
APA StyleMedda, A., Duca, D., & Chiocca, S. (2021). Human Papillomavirus and Cellular Pathways: Hits and Targets. Pathogens, 10(3), 262. https://doi.org/10.3390/pathogens10030262