Activation of OSM-STAT3 Epigenetically Regulates Tumor-Promoting Transcriptional Programs in Cervical Cancer
Abstract
:Simple Summary
Abstract
1. Introduction
2. Materials and Methods
2.1. Single-Cell RNA-Sequencing Data Analysis
2.2. Cell-Cell Interaction Analysis
2.3. The Human Protein Atlas
2.4. Cell Cuture
2.5. Flow Cytometry
2.6. RNA Extraction and Real-Time Quantitative PCR
2.7. RNA-Seq Analysis
2.8. Omni-Assay for Transposase-Accessible Chromatin (ATAC) Protocol
2.9. ATAC-Seq Analysis
2.10. ATAC-Seq Data Visualization
2.11. Motif Enrichment Analysis
2.12. Public Expression Profiles
2.13. Stactistical Analysis
3. Results
3.1. OSM–OSMR Interaction between Epithelial Cell and Macrophage in Cervical Cancer
3.2. High OSMR Expression Is Correlated with Tumor Progression Characteristics of Cervical Cancer
3.3. Regulation of OSM–OSMR Signlaling by STAT3 in Cervical Cancer Cells
3.4. Gene Signatures Regulated by OSM-STAT3 Activity in Cervical Cancer
3.5. Chromatin Remodeling by OSM-induced STAT3 Activity in Cervical Cancer
3.6. OSM-STAT3 Gene Expression Signature Is Associated with Poor Prognosis in Cervical Cancer Patients
4. Discussion
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Torre, L.A.; Islami, F.; Siegel, R.L.; Ward, E.M.; Jemal, A. Global Cancer in Women: Burden and Trends. Cancer Epidemiol. Biomark. Prev. 2017, 26, 444–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Watson, M.; Saraiya, M.; Benard, V.; Coughlin, S.S.; Flowers, L.; Cokkinides, V.; Schwenn, M.; Huang, Y.; Giuliano, A. Burden of cervical cancer in the United States, 1998–2003. Cancer 2008, 113, 2855–2864. [Google Scholar] [CrossRef] [PubMed]
- Crosbie, E.J.; Einstein, M.H.; Franceschi, S.; Kitchener, H.C. Human papillomavirus and cervical cancer. Lancet 2013, 382, 889–899. [Google Scholar] [CrossRef]
- Steenbergen, R.D.M.; Snijders, P.J.F.; Heideman, D.A.M.; Meijer, C.J.L.M. Clinical implications of (epi)genetic changes in HPV-induced cervical precancerous lesions. Nat. Rev. Cancer 2014, 14, 395–405. [Google Scholar] [CrossRef]
- Schiffman, M.; Doorbar, J.; Wentzensen, N.; De Sanjosé, S.; Fakhry, C.; Monk, B.J.; Stanley, M.A.; Franceschi, S. Carcinogenic human papillomavirus infection. Nat. Rev. Dis. Prim. 2016, 2, 16086. [Google Scholar] [CrossRef]
- Vink, M.A.; Bogaards, J.A.; van Kemenade, F.J.; de Melker, H.E.; Meijer, C.J.L.M.; Berkhof, J. Clinical Progression of High-Grade Cervical Intraepithelial Neoplasia: Estimating the Time to Preclinical Cervical Cancer From Doubly Censored National Registry Data. Am. J. Epidemiol. 2013, 178, 1161–1169. [Google Scholar] [CrossRef] [Green Version]
- Campos, N.G.; Castle, P.E.; Wright, T.C.; Kim, J.J. Cervical cancer screening in low-resource settings: A cost-effectiveness framework for valuing tradeoffs between test performance and program coverage. Int. J. Cancer 2015, 137, 2208–2219. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Rashmi, R.; Inkman, M.; Jayachandran, K.; Ruiz, F.; Waters, M.R.; Grigsby, P.W.; Markovina, S.; Schwarz, J.K. Integrating imaging and RNA-seq improves outcome prediction in cervical cancer. J. Clin. Investig. 2021, 131, e139232. [Google Scholar] [CrossRef]
- Uyar, D.; Rader, J. Genomics of Cervical Cancer and the Role of Human Papillomavirus Pathobiology. Clin. Chem. 2014, 60, 144–146. [Google Scholar] [CrossRef]
- Bushweller, J.H. Targeting transcription factors in cancer—From undruggable to reality. Nat. Rev. Cancer 2019, 19, 611–624. [Google Scholar] [CrossRef] [PubMed]
- Chen, A.; Koehler, A.N. Transcription Factor Inhibition: Lessons Learned and Emerging Targets. Trends Mol. Med. 2020, 26, 508–518. [Google Scholar] [CrossRef] [PubMed]
- Arkin, M.R.; Tang, Y.; Wells, J.A. Small-Molecule Inhibitors of Protein-Protein Interactions: Progressing toward the Reality. Chem. Biol. 2014, 21, 1102–1114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Darnell, J.E., Jr.; Kerr, I.M.; Stark, G.R. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 1994, 264, 1415–1421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brooks, A.J.; Putoczki, T. JAK-STAT Signalling Pathway in Cancer. Cancers 2020, 12, 1971. [Google Scholar] [CrossRef]
- Huynh, J.; Chand, A.; Gough, D.; Ernst, M. Therapeutically exploiting STAT3 activity in cancer—Using tissue repair as a road map. Nat. Rev. Cancer 2019, 19, 82–96. [Google Scholar] [CrossRef]
- Johnson, D.E.; O’Keefe, R.A.; Grandis, J.R. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat. Rev. Clin. Oncol. 2018, 15, 234–248. [Google Scholar] [CrossRef]
- Lin, W.-H.; Chang, Y.-W.; Hong, M.-X.; Hsu, T.-C.; Lee, K.-C.; Lin, C.; Lee, J.-L. STAT3 phosphorylation at Ser727 and Tyr705 differentially regulates the EMT–MET switch and cancer metastasis. Oncogene 2021, 40, 791–805. [Google Scholar] [CrossRef]
- Jones, S.A.; Jenkins, B.J. Recent insights into targeting the IL-6 cytokine family in inflammatory diseases and cancer. Nat. Rev. Immunol. 2018, 18, 773–789. [Google Scholar] [CrossRef]
- Li, N.; Grivennikov, S.I.; Karin, M. The Unholy Trinity: Inflammation, Cytokines, and STAT3 Shape The Cancer Microenvironment. Cancer Cell 2011, 19, 429–431. [Google Scholar] [CrossRef]
- Siveen, K.S.; Sikka, S.; Surana, R.; Dai, X.; Zhang, J.; Kumar, A.P.; Tan, B.K.; Sethi, G.; Bishayee, A. Targeting the STAT3 signaling pathway in cancer: Role of synthetic and natural inhibitors. Biochim. Biophys. Acta Rev. Cancer 2014, 1845, 136–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schindler, C.; Darnell, J.E., Jr. Transcriptional responses to polypeptide ligands: The JAK-STAT pathway. Annu. Rev. Biochem. 1995, 64, 621–652. [Google Scholar] [CrossRef] [PubMed]
- Abaurrea, A.; Araujo, A.M.; Caffarel, M.M. The Role of the IL-6 Cytokine Family in Epithelial–Mesenchymal Plasticity in Cancer Progression. Int. J. Mol. Sci. 2021, 22, 8334. [Google Scholar] [CrossRef] [PubMed]
- Tanaka, M.; Miyahima, A. Oncostatin M, a multifunctional cytokine. In Reviews of Physiology, Biochemistry and Pharmacology; Springer: Berlin/Heidelberg, Germany, 2003; Volume 149, pp. 39–52. [Google Scholar] [CrossRef]
- Zarling, J.M.; Shoyab, M.; Marquardt, H.; Hanson, M.B.; Lioubin, M.N.; Todaro, G.J. Oncostatin M: A growth regulator produced by differentiated histiocytic lymphoma cells. Proc. Natl. Acad. Sci. USA 1986, 83, 9739–9743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Araujo, A.M.; Abaurrea, A.; Azcoaga, P.; López-Velazco, J.I.; Manzano, S.; Rodriguez, J.; Rezola, R.; Egia-Mendikute, L.; Valdés-Mora, F.; Flores, J.M.; et al. Stromal oncostatin M cytokine promotes breast cancer progression by reprogramming the tumor microenvironment. J. Clin. Investig. 2022, 132, e165107. [Google Scholar] [CrossRef]
- Junk, D.J.; Bryson, B.L.; Smigiel, J.M.; Parameswaran, N.; Bartel, C.A.; Jackson, M.W. Oncostatin M promotes cancer cell plasticity through cooperative STAT3-SMAD3 signaling. Oncogene 2017, 36, 4001–4013. [Google Scholar] [CrossRef] [Green Version]
- Lee, B.Y.; Hogg, E.K.J.; Below, C.R.; Kononov, A.; Blanco-Gomez, A.; Heider, F.; Xu, J.; Hutton, C.; Zhang, X.; Scheidt, T.; et al. Heterocellular OSM-OSMR signalling reprograms fibroblasts to promote pancreatic cancer growth and metastasis. Nat. Commun. 2021, 12, 7336. [Google Scholar] [CrossRef]
- West, N.R.; Owens, B.M.J.; Hegazy, A.N. The oncostatin M-stromal cell axis in health and disease. Scand. J. Immunol. 2018, 88, e12694. [Google Scholar] [CrossRef]
- Kan, C.E.; Cipriano, R.; Jackson, M.W. c-MYC functions as a molecular switch to alter the response of human mammary epithelial cells to oncostatin M. Cancer Res. 2011, 71, 6930–6939. [Google Scholar] [CrossRef]
- Serrano, M.; Lin, A.W.; McCurrach, M.E.; Beach, D.; Lowe, S.W. Oncogenic ras Provokes Premature Cell Senescence Associated with Accumulation of p53 and p16INK4a. Cell 1997, 88, 593–602. [Google Scholar] [CrossRef] [Green Version]
- Caffarel, M.M.; Coleman, N. Oncostatin M receptor is a novel therapeutic target in cervical squamous cell carcinoma. J. Pathol. 2014, 232, 386–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kucia-Tran, J.A.; Tulkki, V.; Smith, S.; Scarpini, C.G.; Hughes, K.; Araujo, A.M.; Yan, K.Y.M.; Botthof, J.; Pérez-Gómez, E.; Quintanilla, M.; et al. Overexpression of the oncostatin-M receptor in cervical squamous cell carcinoma is associated with epithelial–mesenchymal transition and poor overall survival. Br. J. Cancer 2016, 115, 212–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Caffarel, M.M.; Chattopadhyay, A.; Araujo, A.M.; Bauer, J.; Scarpini, C.G.; Coleman, N. Tissue transglutaminase mediates the pro-malignant effects of oncostatin M receptor over-expression in cervical squamous cell carcinoma. J. Pathol. 2013, 231, 168–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kucia-Tran, J.A.; Tulkki, V.; Scarpini, C.G.; Smith, S.; Wallberg, M.; Paez-Ribes, M.; Araujo, A.M.; Botthoff, J.; Feeney, M.; Hughes, K.; et al. Anti-oncostatin M antibody inhibits the pro-malignant effects of oncostatin M receptor overexpression in squamous cell carcinoma. J. Pathol. 2018, 244, 283–295. [Google Scholar] [CrossRef] [PubMed]
- Winder, D.M.; Chattopadhyay, A.; Muralidhar, B.; Bauer, J.; English, W.R.; Zhang, X.; Karagavriilidou, K.; Roberts, I.; Pett, M.R.; Murphy, G.; et al. Overexpression of the oncostatin M receptor in cervical squamous cell carcinoma cells is associated with a pro-angiogenic phenotype and increased cell motility and invasiveness. J. Pathol. 2011, 225, 448–462. [Google Scholar] [CrossRef]
- Bai, L.; Zhou, H.; Xu, R.; Zhao, Y.; Chinnaswamy, K.; McEachern, D.; Chen, J.; Yang, C.-Y.; Liu, Z.; Wang, M.; et al. A Potent and Selective Small-Molecule Degrader of STAT3 Achieves Complete Tumor Regression In Vivo. Cancer Cell 2019, 36, 498–511. [Google Scholar] [CrossRef] [PubMed]
- Li, C.; Guo, L.; Li, S.; Hua, K. Single-cell transcriptomics reveals the landscape of intra-tumoral heterogeneity and transcriptional activities of ECs in CC. Mol. Ther. Nucleic Acids 2021, 24, 682–694. [Google Scholar] [CrossRef]
- Stuart, T.; Butler, A.; Hoffman, P.; Hafemeister, C.; Papalexi, E.; Mauck, W.M., III; Hao, Y.; Stoeckius, M.; Smibert, P.; Satija, R. Comprehensive Integration of Single-Cell Data. Cell 2019, 177, 1888–1902. [Google Scholar] [CrossRef]
- Efremova, M.; Vento-Tormo, M.; Teichmann, S.A.; Vento-Tormo, R. CellPhoneDB: Inferring cell–cell communication from combined expression of multi-subunit ligand–receptor complexes. Nat. Protoc. 2020, 15, 1484–1506. [Google Scholar] [CrossRef]
- Dobin, A.; Davis, C.A.; Schlesinger, F.; Drenkow, J.; Zaleski, C.; Jha, S.; Batut, P.; Chaisson, M.; Gingeras, T.R. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 2013, 29, 15–21. [Google Scholar] [CrossRef]
- Love, M.I.; Huber, W.; Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014, 15, 550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Buenrostro, J.D.; Wu, B.; Chang, H.Y.; Greenleaf, W.J. ATAC-seq: A Method for Assaying Chromatin Accessibility Genome-Wide. Curr. Protoc. Mol. Biol. 2015, 109, 21–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Robinson, M.D.; McCarthy, D.J.; Smyth, G.K. edgeR: A Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 2010, 26, 139–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ramírez, F.; Ryan, D.P.; Grüning, B.; Bhardwaj, V.; Kilpert, F.; Richter, A.S.; Heyne, S.; Dündar, F.; Manke, T. deepTools2: A next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 2016, 44, 160–165. [Google Scholar] [CrossRef] [PubMed]
- Tang, Z.; Kang, B.; Li, C.; Chen, T.; Zhang, Z. GEPIA2: An enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res. 2019, 47, 556–560. [Google Scholar] [CrossRef] [Green Version]
- Jing, X.; Yang, F.; Shao, C.; Wei, K.; Xie, M.; Shen, H.; Shu, Y. Role of hypoxia in cancer therapy by regulating the tumor microenvironment. Mol. Cancer 2019, 18, 157. [Google Scholar] [CrossRef] [Green Version]
- MacCarthy-Morrogh, L.; Martin, P. The hallmarks of cancer are also the hallmarks of wound healing. Sci. Signal 2020, 13, 8690. [Google Scholar] [CrossRef]
- Weis, S.M.; Cheresh, D.A. Tumor angiogenesis: Molecular pathways and therapeutic targets. Nat. Med. 2011, 17, 1359–1370. [Google Scholar] [CrossRef]
- Klein, K.; Stoiber, D.; Sexl, V.; Witalisz-Siepracka, A. Untwining Anti-Tumor and Immunosuppressive Effects of JAK Inhibitors—A Strategy for Hematological Malignancies? Cancers 2021, 13, 2611. [Google Scholar] [CrossRef]
- Mesa, R.A.; Yasothan, U.; Kirkpatrick, P. Ruxolitinib. Nat. Rev. Drug Discov. 2012, 11, 103–104. [Google Scholar] [CrossRef]
- Wingelhofer, B.; Neubauer, H.A.; Valent, P.; Han, X.; Constantinescu, S.N.; Gunning, P.T.; Müller, M.; Moriggl, R. Implications of STAT3 and STAT5 signaling on gene regulation and chromatin remodeling in hematopoietic cancer. Leukemia 2018, 32, 1713–1726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cangul, H. Hypoxia upregulates the expression of the NDRG1 gene leading to its overexpression in various human cancers. BMC Genet. 2004, 5, 27. [Google Scholar] [CrossRef] [Green Version]
- Daskalaki, I.; Gkikas, I.; Tavernarakis, N. Hypoxia and Selective Autophagy in Cancer Development and Therapy. Front. Cell Dev. Biol. 2018, 6, 104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Menendez, M.T.; Teygong, C.; Wade, K.; Florimond, C.; Blader, I.J. siRNA Screening Identifies the Host Hexokinase 2 (HK2) Gene as an Important Hypoxia-Inducible Transcription Factor 1 (HIF-1) Target Gene in Toxoplasma gondii-Infected Cells. mBio 2015, 6, e00462-15. [Google Scholar] [CrossRef] [Green Version]
- Song, Y.; Zheng, S.; Wang, J.; Long, H.; Fang, L.; Wang, G.; Li, Z.; Que, T.; Liu, Y.; Li, Y.; et al. Hypoxia-induced PLOD2 promotes proliferation, migration and invasion via PI3K/Akt signaling in glioma. Oncotarget 2017, 8, 41947–41962. [Google Scholar] [CrossRef] [Green Version]
- Xu, F.; Zhang, J.; Hu, G.; Liu, L.; Liang, W. Hypoxia and TGF-β1 induced PLOD2 expression improve the migration and invasion of cervical cancer cells by promoting epithelial-to-mesenchymal transition (EMT) and focal adhesion formation. Cancer Cell Int. 2017, 17, 54. [Google Scholar] [CrossRef]
- Bhagwat, A.S.; Vakoc, C.R. Targeting Transcription Factors in Cancer. Trends Cancer 2015, 1, 53–65. [Google Scholar] [CrossRef] [Green Version]
- Xie, K.; Tan, K.; Naylor, M.J. Transcription Factors as Novel Therapeutic Targets and Drivers of Prostate Cancer Progression. Front. Oncol. 2022, 12, 854151. [Google Scholar] [CrossRef]
- 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]
- Chernosky, N.M.; Tamagno, I. The Role of the Innate Immune System in Cancer Dormancy and Relapse. Cancers 2021, 13, 5621. [Google Scholar] [CrossRef]
- Junk, D.J.; Bryson, B.L.; Jackson, M.W. HiJAK’d Signaling; the STAT3 Paradox in Senescence and Cancer Progression. Cancers 2014, 6, 741–755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vlaicu, P.; Mertins, P.; Mayr, T.; Widschwendter, P.; Ataseven, B.; Högel, B.; Eiermann, W.; Knyazev, P.; Ullrich, A. Monocytes/macrophages support mammary tumor invasivity by co-secreting lineage-specific EGFR ligands and a STAT3 activator. BMC Cancer 2013, 13, 197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Muthusami, S.; Sabanayagam, R.; Periyasamy, L.; Muruganantham, B.; Park, W.Y. A review on the role of epidermal growth factor signaling in the development, progression and treatment of cervical cancer. Int. J. Biol. Macromol. 2021, 194, 179–187. [Google Scholar] [CrossRef] [PubMed]
- Soonthornthum, T.; Arias-Pulido, H.; Joste, N.; Lomo, L.; Muller, C.; Rutledge, T.; Verschraegen, C. Epidermal growth factor receptor as a biomarker for cervical cancer. Ann. Oncol. 2011, 22, 2166–2178. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.W.; Kim, Y.T.; Kim, D.K.; Song, C.H.; Lee, J.W. Expression of Epidermal Growth Factor Receptor in Carcinoma of the Cervix. Gynecol. Oncol. 1996, 60, 283–287. [Google Scholar] [CrossRef] [PubMed]
- Mo, X.-T.; Leung, T.H.-Y.; Tang, H.W.-M.; Siu, M.K.-Y.; Wan, P.K.-T.; Chan, K.K.-L.; Cheung, A.N.-Y.; Ngan, H.Y.-S. CD109 mediates tumorigenicity and cancer aggressiveness via regulation of EGFR and STAT3 signalling in cervical squamous cell carcinoma. Br. J. Cancer 2020, 123, 833–843. [Google Scholar] [CrossRef]
- Babon, J.J.; Varghese, L.N.; Nicola, N.A. Inhibition of IL-6 family cytokines by SOCS3. Semin. Immunol. 2014, 26, 13–19. [Google Scholar] [CrossRef] [Green Version]
- Salminen, A.; Kaarniranta, K.; Kauppinen, A. Insulin/IGF-1 signaling promotes immunosuppression via the STAT3 pathway: Impact on the aging process and age-related diseases. Inflamm. Res. 2021, 70, 1043–1061. [Google Scholar] [CrossRef]
- Comità, S.; Femmino, S.; Thairi, C.; Alloatti, G.; Boengler, K.; Pagliaro, P.; Penna, C. Regulation of STAT3 and its role in cardioprotection by conditioning: Focus on non-genomic roles targeting mitochondrial function. Basic Res. Cardiol. 2021, 116, 56. [Google Scholar] [CrossRef]
- Yang, R.; Rincon, M. Mitochondrial Stat3, the Need for Design Thinking. Int. J. Biol. Sci. 2016, 12, 532–544. [Google Scholar] [CrossRef]
- Rankin, E.B.; Nam, J.-M.; Giaccia, A.J. Hypoxia: Signaling the Metastatic Cascade. Trends Cancer 2016, 2, 295–304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hanahan, D.; Weinberg, R.A. The Hallmarks of Cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kabakov, A.E.; Yakimova, A.O. Hypoxia-Induced Cancer Cell Responses Driving Radioresistance of Hypoxic Tumors: Approaches to Targeting and Radiosensitizing. Cancers 2021, 13, 1102. [Google Scholar] [CrossRef]
- Lin, S.H.; Koong, A.C. Breathing New Life Into Hypoxia-Targeted Therapies for Non-Small Cell Lung Cancer. J. Natl. Cancer Inst. 2017, 110, 1–2. [Google Scholar] [CrossRef] [PubMed]
- Unruh, A.; Ressel, A.; Mohamed, H.G.; Johnson, R.S.; Nadrowitz, R.; Richter, E.; Katschinski, D.M.; Wenger, R.H. The hypoxia-inducible factor-1α is a negative factor for tumor therapy. Oncogene 2003, 22, 3213–3220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fyles, A.; Milosevic, M.; Hedley, D.; Pintilie, M.; Levin, W.; Manchul, L.; Hill, R.P. Tumor Hypoxia Has Independent Predictor Impact Only in Patients With Node-Negative Cervix Cancer. J. Clin. Oncol. 2002, 20, 680–687. [Google Scholar] [CrossRef]
- Lyng, H.; Malinen, E. Hypoxia in cervical cancer: From biology to imaging. Clin. Transl. Imaging 2017, 5, 373–388. [Google Scholar] [CrossRef]
- Lyng, H.; Sundfør, K.; Tropé, C.; Rofstad, E.K. Disease control of uterine cervical cancer: Relationships to tumor oxygen tension, vascular density, cell density, and frequency of mitosis and apoptosis measured before treatment and during radiotherapy. Clin. Cancer Res. 2000, 6, 1104–1112. [Google Scholar]
- Hockel, M.; Schlenger, K.; Aral, B.; Mitze, M.; Schaffer, U.; Vaupel, P. Association between tumor hypoxia and malignant progression in advanced cancer of the uterine cervix. Cancer Res. 1996, 56, 4509–4515. [Google Scholar]
Characteristics | GSE6791 | GSE29570 |
---|---|---|
No. | 20 | 45 |
Age | ||
≤45 | 7 (35%) | 19 (42%) |
>45 | 13 (65%) | 26 (58%) |
Stage | ||
IB | 16 (80%) | 25 (56%) |
II/III | 3 (15%) | 16 (36%) |
IV | 1 (5%) | 4 (8%) |
Histology | ||
SCC | 20 (100%) | 41 (91%) |
ACC | 3 (7%) | |
ASCC | 1 (2%) |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Noh, J.; You, C.; Kang, K.; Kang, K. Activation of OSM-STAT3 Epigenetically Regulates Tumor-Promoting Transcriptional Programs in Cervical Cancer. Cancers 2022, 14, 6090. https://doi.org/10.3390/cancers14246090
Noh J, You C, Kang K, Kang K. Activation of OSM-STAT3 Epigenetically Regulates Tumor-Promoting Transcriptional Programs in Cervical Cancer. Cancers. 2022; 14(24):6090. https://doi.org/10.3390/cancers14246090
Chicago/Turabian StyleNoh, Junho, Chaelin You, Keunsoo Kang, and Kyuho Kang. 2022. "Activation of OSM-STAT3 Epigenetically Regulates Tumor-Promoting Transcriptional Programs in Cervical Cancer" Cancers 14, no. 24: 6090. https://doi.org/10.3390/cancers14246090
APA StyleNoh, J., You, C., Kang, K., & Kang, K. (2022). Activation of OSM-STAT3 Epigenetically Regulates Tumor-Promoting Transcriptional Programs in Cervical Cancer. Cancers, 14(24), 6090. https://doi.org/10.3390/cancers14246090