Identification of key genes and construction of regulatory network for the progression of cervical cancer

Gene Reports ◽  
2020 ◽  
Vol 21 ◽  
pp. 100965
Author(s):  
Monika Rajput ◽  
Mukesh Kumar ◽  
Mayuri Kumari ◽  
Atanu Bhattacharjee ◽  
Aanchal Anant Awasthi
2021 ◽  
Vol 22 (5) ◽  
pp. 2442
Author(s):  
Qun Wang ◽  
Aurelia Vattai ◽  
Theresa Vilsmaier ◽  
Till Kaltofen ◽  
Alexander Steger ◽  
...  

Cervical cancer is primarily caused by the infection of high-risk human papillomavirus (hrHPV). Moreover, tumor immune microenvironment plays a significant role in the tumorigenesis of cervical cancer. Therefore, it is necessary to comprehensively identify predictive biomarkers from immunogenomics associated with cervical cancer prognosis. The Cancer Genome Atlas (TCGA) public database has stored abundant sequencing or microarray data, and clinical data, offering a feasible and reliable approach for this study. In the present study, gene profile and clinical data were downloaded from TCGA, and the Immunology Database and Analysis Portal (ImmPort) database. Wilcoxon-test was used to compare the difference in gene expression. Univariate analysis was adopted to identify immune-related genes (IRGs) and transcription factors (TFs) correlated with survival. A prognostic prediction model was established by multivariate cox analysis. The regulatory network was constructed and visualized by correlation analysis and Cytoscape, respectively. Gene functional enrichment analysis was performed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). A total of 204 differentially expressed IRGs were identified, and 22 of them were significantly associated with the survival of cervical cancer. These 22 IRGs were actively involved in the JAK-STAT pathway. A prognostic model based on 10 IRGs (APOD, TFRC, GRN, CSK, HDAC1, NFATC4, BMP6, IL17RD, IL3RA, and LEPR) performed moderately and steadily in squamous cell carcinoma (SCC) patients with FIGO stage I, regardless of the age and grade. Taken together, a risk score model consisting of 10 novel genes capable of predicting survival in SCC patients was identified. Moreover, the regulatory network of IRGs associated with survival (SIRGs) and their TFs provided potential molecular targets.


2019 ◽  
Vol 15 (11) ◽  
pp. e1007435 ◽  
Author(s):  
Jiajun Zhang ◽  
Wenbo Zhu ◽  
Qianliang Wang ◽  
Jiayu Gu ◽  
L. Frank Huang ◽  
...  

2020 ◽  
Vol 39 (7) ◽  
pp. 1328-1346 ◽  
Author(s):  
Jinhui Liu ◽  
Jing Yang ◽  
Feng Gao ◽  
Siyue Li ◽  
Sipei Nie ◽  
...  

2020 ◽  
Vol Volume 13 ◽  
pp. 157-165
Author(s):  
Bingqi Wang ◽  
Min Wang ◽  
Xianping Li ◽  
Min Yang ◽  
Lei Liu

2013 ◽  
Vol 23 (08) ◽  
pp. 1350138 ◽  
Author(s):  
HONGWEI YIN ◽  
XIAOYONG XIAO ◽  
XIAOQING WEN ◽  
TIANSHOU ZHOU

In the development of Drosophila wing disc, morphogen Dpp, which is a signaling molecule from a local region and disperses into anterior and posterior compartments, builds up a gradient with precise pattern information. Experiments have demonstrated that the key genes (brk, dad, omb and sal) and phosphorylated protein (pMad), which are activated by Dpp signaling molecules and form the gradients of the corresponding proteins of these genes, direct and control the spatial pattern of the wing disc. However, the regulatory network of these genes are in complex and nonlinear interaction with upstream regulators and downstream targets. In this paper, the mathematical model is built according to the regulatory relationships of these key genes. The stabilities of the gradients of these corresponding proteins are investigated. Furthermore, numerical simulations show that these gradients are robust with respect to some major reaction rates in this regulatory network.


Author(s):  
Musalula Sinkala ◽  
Mildred Zulu ◽  
Panji Nkhoma ◽  
Doris Kafita ◽  
Ephraim Zulu ◽  
...  

Cervical cancer has remained the most prevalent and lethal malignancy among women worldwide and accounted for over 250,000 deaths in 2019. Nearly ninety-five per cent of cervical cancer cases are associated with persistent infection with high-risk Human Papillomavirus (HPV), and seventy per cent of these are associated with viral integration in the host genome. HPV-infection imparts specific changes in the regulatory network of infected cancer cells that are of diagnostic, prognostic and therapeutic importance. Here, we conducted a systems-level analysis of the regulatory network changes, and the associated regulatory proteins thereof, in HPV-positive cervical cancer. We applied functional pathway analysis to show that HPV-positive cancers are characterised by perturbations of numerous cellular processes, predominantly in those linked to the cell cycle, mitosis, cytokine and immune cell signalling. Using computational predictions, we revealed that HPV-positive cervical cancers are regulated by transcription factors including, SOX2, E2F, NANOG, OCT4, and MYC, which control various processes such as the renewal of cancer stem cells, and the proliferation and differentiation of tumour cells. Through the analysis of upstream regulatory kinases, we identified the mitogen-activated protein kinases; among others, MAPK1, MAPK3 and MAPK8, and the cyclin-dependent kinases; among others, CDK1, CDK2 and CD4, as the key kinases that control the biological processes in HPV-positive cervical cancers. Taken together, we uncover a landscape of the key regulatory pathways and proteins in HPV-positive cervical cancers, all of which may provide attractive drug targets for future therapeutics.


2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Heng Zou ◽  
Huijia Chen ◽  
Shuaibin Liu ◽  
Xiaoling Gan

Abstract Background Circular RNAs (circRNAs) are increasingly implicated in regulating human carcinogenesis. Previous work showed the oncogenic activity of circ_0018289 in cervical cancer. However, the molecular basis underlying the modulation of circ_0018289 in cervical carcinogenesis is still not fully understood. Methods The levels of circ_0018289, microRNA (miR)-183-5p, and transmembrane p24 trafficking protein 5 (TMED5) were measured by quantitative real-time polymerase chain reaction (qRT-PCR) or western blot assay. Ribonuclease (RNase) R and subcellular localization assays were used to characterize circ_0018289. Cell proliferation was detected by the Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2′-deoxyuridine (Edu) assays. Cell apoptosis and tube formation were assessed by flow cytometry and tube formation assays, respectively. A dual-luciferase reporter assay was performed to confirm the direct relationship between miR-183-5p and circ_0018289 or TMED5. The role of circ_0018289 in tumor growth was gauged by mouse xenograft experiments. Results Circ_0018289 was overexpressed in cervical cancer tissues and cells. Circ_0018289 silencing impeded cell proliferation, enhanced cell apoptosis, and suppressed angiogenesis in vitro, as well as diminished tumor growth in vivo. Mechanistically, circ_0018289 targeted and regulated miR-183-5p by binding to miR-183-5p, and circ_0018289 regulated cervical cancer development and angiogenesis partially through miR-183-5p. Moreover, TMED5 was directly targeted and inhibited by miR-183-5p through the perfect complementary sites in TMED5 3′UTR, and TMED5 knockdown phenocopied miR-183-5p overexpression in suppressing cervical cancer development and angiogenesis. Furthermore, circ_0018289 induced TMED5 expression by competitively binding to shared miR-183-5p. Conclusion Our observations identified the circ_0018289/miR-183-5p/TMED5 regulatory network as a novel molecular basis underlying the modulation of cervical carcinogenesis.


Sign in / Sign up

Export Citation Format

Share Document