trying...
4 1 0 1 MCID_676f085df306d7202f067694
32908931
Jin-Han Gu[author] Gu, Jin Han[Full Author Name] gu, jin han[Author]
trying2... trying...
32908931 2021 05 19 2021 05 19 2314-6141 2020 2020 BioMed research international Biomed Res Int The Expression and Potential Role of Tubulin Alpha 1b in Wilms' Tumor. 9809347 9809347 9809347 10.1155/2020/9809347 We explored the difference in expression of tubulin alpha 1b (TUBA1B) between Wilms' tumor (WT) and normal tissues (NT) from in-house patients and databases, to determine TUBA1B expression in WT and the predictive pathways of coexpressed genes. In-house RNA-sequencing data were performed with WT and NT from three patients from our institute. Other four RNA-sequencing and microarray data were also downloaded from multiple public databases. The TUBA1B expression between WT and NT was analyzed by Student's t -test and meta-analysis. The correlation between the expression of TUBA1B and other genes in each study was analyzed. Genes with p < 0.05 and r > 0.5 were considered as the coexpressing genes of TUBA1B. Overlapping the coexpressed genes of the five studies, including three in-house patients (3 WT vs. 3 NT), GTEx-TARGET (126 WT vs. 51 NT), GSE2172 (18 WT vs. 3 NT), GSE11024 (27 WT vs. 12 NT), and GSE73209 (32 WT vs. 6 NT), were performed with limma and VennDiagram packages in R software. The website of WEB-based GEne SeT AnaLysis toolkit were used to analyze the gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional annotations for the overlapped genes. The results showed that the relative expression of TUBA1B in WT tissues from in-house three patients was 280.0086, 141.7589, and 303.8292 and that in NT was 16.5836, 104.8141, and 12.79 (3 WT vs. 3 NT, p = 0.0285, ROC = 100%, SMD = 2.74). Student's t -test and meta-analysis in all studies revealed that the expression of TUBA1B was upregulated in WT tissues compared to that in NT (p < 0.05, SMD = 2.89, sROC = 0.98). Finally, the research identified the expression of TUBA1B in WT tissues was significantly upregulated than that in NT. The coexpressed genes of TUBA1B were enriched in the pathway of DNA replication, mismatch repair, cell cycle, pathogenic Escherichia coli infection, and spliceosome. Copyright © 2020 Qiong-Qian Xu et al. Xu Qiong-Qian QQ Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China. Qin Li-Ting LT Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China. Liang Song-Wu SW Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China. Chen Peng P Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China. Gu Jin-Han JH Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China. Huang Zhi-Guang ZG Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China. Yang Xia X Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China. Gao Li L Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China. Wang Shi-Shuo SS Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China. Luo Yi-Ge YG Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China. Liu Lin-Le-Yi LL Department of First Clinical Medical College, Guangxi Medical University, Nanning 530021, China. Wang Jun J Department of First Clinical Medical College, Guangxi Medical University, Nanning 530021, China. Lin Jin-Yan JY Department of First Clinical Medical College, Guangxi Medical University, Nanning 530021, China. Chen Gang G 0000-0003-2402-2987 Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China. Chen Jia-Bo JB 0000-0003-1757-3481 Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China. eng Journal Article 2020 08 25 United States Biomed Res Int 101600173 0 TUBA1B protein, human 0 Tubulin IM Gene Expression Regulation, Neoplastic Humans Kidney Neoplasms genetics Tubulin genetics Up-Regulation Wilms Tumor genetics The authors declare that there is no conflict of interest regarding the publication of this paper. 2020 5 22 2020 7 23 2020 9 10 5 36 2020 9 11 6 0 2021 5 20 6 0 2020 8 25 epublish 32908931 PMC7468616 10.1155/2020/9809347 Spreafico F., Ferrari A., Mascarin M., et al. Wilms tumor, medulloblastoma, and rhabdomyosarcoma in adult patients: lessons learned from the pediatric experience. Cancer Metastasis Reviews. 2019;38(4):683–694. doi: 10.1007/s10555-019-09831-3. 10.1007/s10555-019-09831-3 31797181 Fu W., Zhuo Z., Hua R.-X., et al. Association of KRAS and NRAS gene polymorphisms with Wilms tumor risk: a four-center case-control study. Aging. 2019;11(5):1551–1563. doi: 10.18632/aging.101855. 10.18632/aging.101855 PMC6428095 30860980 Otto G. Kidney cancer: targeting Wilms tumour. Nature Reviews. Nephrology. 2017;13(11):p. 662. doi: 10.1038/nrneph.2017.131. 10.1038/nrneph.2017.131 28890538 Treger T. D., Chowdhury T., Pritchard-Jones K., Behjati S. The genetic changes of Wilms tumour. Nature Reviews. Nephrology. 2019;15(4):240–251. doi: 10.1038/s41581-019-0112-0. 10.1038/s41581-019-0112-0 30705419 Zhu J., Jia W., Wu C., Fu W. Base Excision Repair Gene Polymorphisms and Wilms Tumor Susceptibility. Base Excision Repair Gene Polymorphisms and Wilms Tumor Susceptibility. 2018;33:88–93. doi: 10.1016/j.ebiom.2018.06.018. 10.1016/j.ebiom.2018.06.018 PMC6085508 29937070 Lin Z., Gasic I., Chandrasekaran V., et al. TTC5 mediates autoregulation of tubulin via mRNA degradation. Science. 2020;367(6473):100–104. doi: 10.1126/science.aaz4352. 10.1126/science.aaz4352 PMC6942541 31727855 Kim N. D., Park E. S., Kim Y. H., et al. Structure-based virtual screening of novel tubulin inhibitors and their characterization as anti-mitotic agents. Bioorganic & Medicinal Chemistry. 2010;18(19):7092–7100. doi: 10.1016/j.bmc.2010.07.072. 10.1016/j.bmc.2010.07.072 20810285 Reader J., Harper A. K., Legesse T., et al. EP4 and Class III β-Tubulin Expression in Uterine Smooth Muscle Tumors: Implications for Prognosis and Treatment. Cancers. 2019;11(10):p. 1590. doi: 10.3390/cancers11101590. 10.3390/cancers11101590 PMC6826612 31635323 Cook A. D., Manka S. W., Wang S., Moores C. A., Atherton J. A microtubule RELION-based pipeline for cryo-EM image processing. Journal of Structural Biology. 2020;209(1):p. 107402. doi: 10.1016/j.jsb.2019.10.004. 10.1016/j.jsb.2019.10.004 PMC6961209 31610239 Tuszynski J. A., Friesen D., Freedman H., et al. Microtubules as Sub-Cellular Memristors. Scientific reports. 2020;10(1):p. 2108. doi: 10.1038/s41598-020-58820-y. 10.1038/s41598-020-58820-y PMC7005844 32034179 Consortium TG. The Genotype-Tissue Expression (GTEx) pilot analysis: Multitissue gene regulation in humans. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. 2015;348(6235):648–660. doi: 10.1126/science.1262110. 10.1126/science.1262110 PMC4547484 25954001 Barrett T., Wilhite S. E., Ledoux P., et al. NCBI GEO: archive for functional genomics data sets—update. Nucleic Acids Research. 2012;41:D991–D995. doi: 10.1093/nar/gks1193. 10.1093/nar/gks1193 PMC3531084 23193258 Kodama Y., Shumway M., Leinonen R., Collaboration INSD The Sequence Read Archive: explosive growth of sequencing data. Nucleic Acids Research. 2011;40(D1):D54–D56. doi: 10.1093/nar/gkr854. 10.1093/nar/gkr854 PMC3245110 22009675 Rhodes D. R., Kalyana-Sundaram S., Mahavisno V., et al. Oncomine 3.0: genes, pathways, and networks in a collection of 18,000 cancer gene expression profiles. Neoplasia. 2007;9(2):166–180. doi: 10.1593/neo.07112. 10.1593/neo.07112 PMC1813932 17356713 Athar A., Füllgrabe A., George N., et al. ArrayExpress update – from bulk to single-cell expression data. ArrayExpress update - from bulk to single-cell expression data. 2019;47(D1):D711–D715. doi: 10.1093/nar/gky964. 10.1093/nar/gky964 PMC6323929 30357387 Ritchie M. E., Phipson B., Wu D., et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Research. 2015;43(7):p. e47. doi: 10.1093/nar/gkv007. 10.1093/nar/gkv007 PMC4402510 25605792 Chen H., Boutros P. C. VennDiagram: a package for the generation of highly-customizable Venn and Euler diagrams in R. BMC bioinformatics. 2011;12(1):p. 35. doi: 10.1186/1471-2105-12-35. 10.1186/1471-2105-12-35 PMC3041657 21269502 Liao Y., Wang J., Jaehnig E. J., Shi Z., Zhang B. WebGestalt 2019: gene set analysis toolkit with revamped UIs and APIs. Nucleic Acids Research. 2019;47(W1):W199–W205. doi: 10.1093/nar/gkz401. 10.1093/nar/gkz401 PMC6602449 31114916 Dezső Z., Oestreicher J., Weaver A., et al. Gene expression profiling reveals epithelial mesenchymal transition (EMT) genes can selectively differentiate eribulin sensitive breast cancer cells. PLos One. 2014;9(8):p. e106131. doi: 10.1371/journal.pone.0106131. 10.1371/journal.pone.0106131 PMC4149521 25171249 Yen T. J., Machlin P. S., Cleveland D. W. Autoregulated instability of β-tubulin mRNAs by recognition of the nascent amino terminus of βtubulin. Nature. 1988;334(6183):580–585. doi: 10.1038/334580a0. 10.1038/334580a0 3405308 Cleveland D. W. Autoregulated control of tubilin synthesis in animal cells. Current opinion in cell biology. 1989;1(1):10–14. doi: 10.1016/s0955-0674(89)80030-4. 10.1016/s0955-0674(89)80030-4 2629858 Gasic I., Boswell S. A., Mitchison T. J. Tubulin mRNA stability is sensitive to change in microtubule dynamics caused by multiple physiological and toxic cues. PLoS Biology. 2019;17(4, article e3000225) doi: 10.1371/journal.pbio.3000225. 10.1371/journal.pbio.3000225 PMC6474637 30964857 Cheung C. H., Wu S. Y., Lee T. R., et al. Cancer cells acquire mitotic drug resistance properties through beta I-tubulin mutations and alterations in the expression of beta-tubulin isotypes. PLoS One. 2010;5(9, article e12564) doi: 10.1371/journal.pone.0012564. 10.1371/journal.pone.0012564 PMC2933234 20838440 Lu C., Zhang J., He S., et al. Increased α-tubulin1b expression indicates poor prognosis and resistance to chemotherapy in hepatocellular carcinoma. Digestive Diseases and Sciences. 2013;58(9):2713–2720. doi: 10.1007/s10620-013-2692-z. 10.1007/s10620-013-2692-z 23625295 Blenk S., Engelmann J. C., Pinkert S., et al. Explorative data analysis of MCL reveals gene expression networks implicated in survival and prognosis supported by explorative CGH analysis. BMC Cancer. 2008;8(1):p. 106. doi: 10.1186/1471-2407-8-106. 10.1186/1471-2407-8-106 PMC2442114 18416826 Pritchard-Jones K., Perotti D. WARNING: G-401 and SK-NEP-1 cell lines are not Wilms tumor cell lines. Pediatric Blood & Cancer. 2019;66(7, article e27741) doi: 10.1002/pbc.27741. 10.1002/pbc.27741 30924592 Stroup E. K., Budhipramono Y. Y. A., Hwang T. H., et al. WT‐CLS1is a rhabdoid tumor cell line and can be inhibited bymiR‐16. Cancer reports. 2019;2(3) doi: 10.1002/cnr2.1110. 10.1002/cnr2.1110 32802817 2022 04 16 2288-6575 99 2 2020 Aug Annals of surgical treatment and research Ann Surg Treat Res Nomogram for predicting overall survival in children with neuroblastoma based on SEER database. 118 126 118-126 10.4174/astr.2020.99.2.118 This study was performed to establish and validate a nomogram for predicting the overall survival in children with neuroblastoma. The latest clinical data of neuroblastoma in Surveillance, Epidemiology, and End Results (SEER) database was extracted from 2000 to 2016. The cases included were randomly divided into training and validation cohorts. The survival curves were drawn with a Kaplan-Meier estimator to investigate the influences of certain single factors on overall survival. Also, least absolute shrinkage and selection operator regression was applied to further select the prognostic variables for neuroblastoma. Additionally, receiver operating characteristic (ROC) curves and calibration curves were used to evaluate the accuracy of the nomogram. In total, 1,262 patients were collected and 8 independent prognostic factors were achieved, including patients' age, sex, race, tumor grade, radiotherapy, chemotherapy, tumor site, and tumor size. Then we constructed a nomogram by using the data of the training cohort with 886 cases. Subsequently, the nomogram was validated internally and externally with 886 and 376 cases, respectively. The internal validation revealed that the area under the curves (AUC) of ROC curves of 1-, 3-, and 5-year overall survival were 0.69, 0.78, and 0.81, respectively. Accordingly, the external validation also showed that the AUC of 1-, 3-, and 5-year overall survival were all ≥0.69. Both methods of validation demonstrated that the predictive calibration curves were consistent with standard curves. The nomogram possess the potential to be a new tool in predicting the survival rate of neuroblastoma patients. Copyright © 2020, the Korean Surgical Society. Liang Song-Wu SW 0000-0002-5664-8776 Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China. Chen Gang G 0000-0002-4864-1451 Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China. Luo Yi-Ge YG 0000-0002-2929-4482 Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China. Chen Peng P 0000-0001-5893-8120 Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China. Gu Jin-Han JH 0000-0003-1663-6532 Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China. Xu Qiong-Qian QQ 0000-0003-0924-2658 Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China. Dang Yi-Wu YW 0000-0002-7793-1239 Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China. Qin Li-Ting LT 0000-0003-0213-2499 Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China. Lu Hui-Ping HP 0000-0001-7765-0763 Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China. Huang Wen-Ting WT 0000-0002-8635-1500 Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China. Huang Zhi-Guang ZG 0000-0003-4457-9491 Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China. Gao Li L 0000-0003-4418-0528 Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China. Chen Jia-Bo JB 0000-0003-1757-3481 Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China. eng Journal Article 2020 07 31 Korea (South) Ann Surg Treat Res 101622895 2288-6575 Neuroblastoma Nomograms Prognosis Risk factors SEER program Conflict of Interest: No potential conflict of interest relevant to this article was reported. 2019 12 26 2020 5 25 2020 6 5 2020 8 18 6 0 2020 8 18 6 0 2020 8 18 6 1 2020 8 1 ppublish 32802817 PMC7406400 10.4174/astr.2020.99.2.118 Jubierre L, Jiménez C, Rovira E, Soriano A, Sábado C, Gros L, et al. Targeting of epigenetic regulators in neuroblastoma. Exp Mol Med. 2018;50:51. PMC5938021 29700278 Rickman DS, Schulte JH, Eilers M. The expanding world of N-MYC-driven tumors. Cancer Discov. 2018;8:150–163. 29358508 Meena JP, Gupta AK. Neuroblastoma in a developing country: miles to go. Indian J Pediatr. 2019;86:403–405. 30915646 Zareifar S, Shakibazad N, Zekavat OR, Bordbar M, Shahriari M. Successful treatment of refractory metastatic neuroblastoma with panobinostat in combination with chemotherapy agents and iodine-131-meta-iodobenzylguanidine therapy. J Oncol Pharm Pract. 2020;26:481–486. 31156056 Herd F, Basta NO, McNally RJQ, Tweddle DA. A systematic review of re-induction chemotherapy for children with relapsed high-risk neuroblastoma. Eur J Cancer. 2019;111:50–58. PMC6458963 30822684 Swift CC, Eklund MJ, Kraveka JM, Alazraki AL. Updates in diagnosis, management, and treatment of neuroblastoma. Radiographics. 2018;38:566–580. 29528815 PDQ Pediatric Treatment Editorial Board. PDQ Cancer Information Summaries. Bethesda (MD): National Cancer Institute (US); 2002. Neuroblastoma Treatment (PDQ®): Health Professional Version. Valter K, Zhivotovsky B, Gogvadze V. Cell death-based treatment of neuroblastoma. Cell Death Dis. 2018;9:113. PMC5833874 29371588 Pastor ER, Mousa SA. Current management of neuroblastoma and future direction. Crit Rev Oncol Hematol. 2019;138:38–43. 31092383 Han Y, Ye X, Cheng J, Zhang S, Feng W, Han Z, et al. Integrative analysis based on survival associated co-expression gene modules for predicting Neuroblastoma patients' survival time. Biol Direct. 2019;14:4. PMC6375203 30760313 Yao W, Li K, Dong KR, Zheng S, Xiao XM. Long-term prognosis of low-risk neuroblastoma treated by surgery alone: an experience from a single institution of China. World J Pediatr. 2019;15:148–152. 30446974 Pinto NR, Applebaum MA, Volchenboum SL, Matthay KK, London WB, Ambros PF, et al. Advances in risk classification and treatment strategies for neuroblastoma. J Clin Oncol. 2015;33:3008–3017. PMC4567703 26304901 Morgenstern DA, Bagatell R, Cohn SL, Hogarty MD, Maris JM, Moreno L, et al. The challenge of defining “ultra-high-risk” neuroblastoma. Pediatr Blood Cancer. 2019;66:e27556. 30479064 Park JR, Bagatell R, Cohn SL, Pearson AD, Villablanca JG, Berthold F, et al. Revisions to the international neuroblastoma response criteria: a consensus statement from the National Cancer Institute clinical trials planning meeting. J Clin Oncol. 2017;35:2580–2587. PMC5676955 28471719 Matthay KK, Maris JM, Schleiermacher G, Nakagawara A, Mackall CL, Diller L, et al. Neuroblastoma. Nat Rev Dis Primers. 2016;2:16078. 27830764 Cohn SL, Pearson AD, London WB, Monclair T, Ambros PF, Brodeur GM, et al. The International Neuroblastoma Risk Group (INRG) classification system: an INRG Task Force report. J Clin Oncol. 2009;27:289–297. PMC2650388 19047291 Brodeur GM, Pritchard J, Berthold F, Carlsen NL, Castel V, Castelberry RP, et al. Revisions of the international criteria for neuroblastoma diagnosis, staging, and response to treatment. J Clin Oncol. 1993;11:1466–1477. 8336186 Hu CY, Pan ZY, Yang J, Chu XH, Zhang J, Tao XJ, et al. Nomograms for predicting long-term overall survival and cancer-specific survival in lip squamous cell carcinoma: a population-based study. Cancer Med. 2019;8:4032–4042. PMC6639254 31112373 Liu G, Liu Q, Sun SR. Nomograms for estimating survival in patients with papillary thyroid cancer after surgery. Cancer Manag Res. 2019;11:3535–3544. PMC6497889 31114382 Huang L, Balavarca Y, van der Geest L, Lemmens V, Van Eycken L, De Schutter H, et al. Development and validation of a prognostic model to predict the prognosis of patients who underwent chemotherapy and resection of pancreatic adenocarcinoma: a large international population-based cohort study. BMC Med. 2019;17:66. PMC6432746 30905320 Guo CX, Shen YN, Zhang Q, Zhang XZ, Wang JL, Gao SL, et al. Prediction of postoperative pancreatic fistula using a nomogram based on the updated definition. Ann Surg Treat Res. 2020;98:72–81. PMC7002880 32051815 Mohler JL, Antonarakis ES, Armstrong AJ, D'Amico AV, Davis BJ, Dorff T, et al. Prostate Cancer, Version 2.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2019;17:479–505. 31085757 Xie Y, Xu H, Fang F, Li Z, Zhou H, Pan J, et al. A 3-protein expression signature of neuroblastoma for outcome prediction. Am J Surg Pathol. 2018;42:1027–1035. 29794872 Li X, Meng Y. A prognostic nomogram for neuroblastoma in children. PeerJ. 2019;7:e7316. PMC6626656 31338261 Tibshirani R. The lasso method for variable selection in the Cox model. Stat Med. 1997;16:385–395. 9044528 Hu J, Wang T, Zhang KH, Jiang YP, Xu S, Chen SH, et al. Pretreatment risk management of a novel nomogram model for prediction of thoracoabdominal extrahepatic metastasis in primary hepatic carcinoma. J Transl Med. 2019;17:117. PMC6454745 30961629 Jiang Y, Wang W, Chen C, Zhang X, Zha X, Lv W, et al. Radiomics signature on computed tomography imaging: association with lymph node metastasis in patients with gastric cancer. Front Oncol. 2019;9:340. PMC6498894 31106158 London WB, Castel V, Monclair T, Ambros PF, Pearson AD, Cohn SL, et al. Clinical and biologic features predictive of survival after relapse of neuroblastoma: a report from the International Neuroblastoma Risk Group project. J Clin Oncol. 2011;29:3286–3292. PMC3158599 21768459 Louis CU, Shohet JM. Neuroblastoma: molecular pathogenesis and therapy. Annu Rev Med. 2015;66:49–63. PMC4418018 25386934 32214060 2020 12 18 2020 12 18 1643-3750 26 2020 Mar 26 Medical science monitor : international medical journal of experimental and clinical research Med Sci Monit RNA-Sequencing, Connectivity Mapping, and Molecular Docking to Investigate Ligand-Protein Binding for Potential Drug Candidates for the Treatment of Wilms Tumor. e920725 e920725 10.12659/MSM.920725 BACKGROUND Wilms tumor, or nephroblastoma, is a malignant pediatric embryonal renal tumor that has a poor prognosis. This study aimed to use bioinformatics data, RNA-sequencing, connectivity mapping, molecular docking, and ligand-protein binding to identify potential targets for drug therapy in Wilms tumor. MATERIAL AND METHODS Wilms tumor and non-tumor samples were obtained from high throughput gene expression databases, and differentially expressed genes (DEGs) were analyzed using the voom method in the limma package. The overlapping DEGs were obtained from the intersecting drug target genes using the Connectivity Map (CMap) database, and systemsDock was used for molecular docking. Gene databases were searched for gene expression profiles for complementary analysis, analysis of clinical significance, and prognosis analysis to refine the study. RESULTS From 177 cases of Wilms tumor, there were 648 upregulated genes and 342 down-regulated genes. Gene Ontology (GO) enrichment analysis showed that the identified DEGs that affected the cell cycle. After obtaining 21 candidate drugs, there were seven overlapping genes with 75 drug target genes and DEGs. Molecular docking results showed that relatively high scores were obtained when retinoic acid and the cyclin-dependent kinase inhibitor, alsterpaullone, were docked to the overlapping genes. There were significant standardized mean differences for three overlapping genes, CDK2, MAP4K4, and CRABP2. However, four upregulated overlapping genes, CDK2, MAP4K4, CRABP2, and SIRT1 had no prognostic significance. CONCLUSIONS RNA-sequencing, connectivity mapping, and molecular docking to investigate ligand-protein binding identified retinoic acid and alsterpaullone as potential drug candidates for the treatment of Wilms tumor. Luo Jia-Yuan JY Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland). Yan Shi-Bai SB Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland). Chen Gang G Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland). Chen Peng P Department of Pediatric Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland). Liang Song-Wu SW Department of Pediatric Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland). Xu Qiong-Qian QQ Department of Pediatric Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland). Gu Jin-Han JH Department of Pediatric Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland). Huang Zhi-Guang ZG Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland). Qin Li-Ting LT Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland). Lu Hui-Ping HP Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland). Mo Wei-Jia WJ Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland). Luo Yi-Ge YG Department of Pediatric Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland). Chen Jia-Bo JB Department of Pediatric Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland). eng Journal Article 2020 03 26 United States Med Sci Monit 9609063 1234-1010 0 Antineoplastic Agents 0 Ligands IM Antineoplastic Agents pharmacology therapeutic use Drug Evaluation, Preclinical Gene Expression Regulation, Neoplastic Gene Ontology Humans Kaplan-Meier Estimate Ligands Molecular Docking Simulation Prognosis Protein Binding ROC Curve Sequence Analysis, RNA Wilms Tumor drug therapy genetics Conflict of interest . None.2020 3 28 6 0 2020 3 28 6 0 2020 12 19 6 0 2020 3 26 epublish 32214060 PMC7119447 10.12659/MSM.920725 920725 Fu W, Zhuo Z, Hua RX, et al. Association of KRAS and NRAS gene polymorphisms with Wilms tumor risk: a four-center case-control study. Aging (Albany NY) 2019;11:1551–63. PMC6428095 30860980 Bu Q, He H, Fan D, et al. Association between loss of heterozygosity of chromosome 16q and survival in Wilms’ tumor: A meta-analysis. Pathol Res Pract. 2018;214:1772–77. 30143352 Cao X, Liu DH, Zhou Y, et al. Histone deacetylase 5 promotes Wilms’ tumor cell proliferation through the upregulation of c-Met. Mol Med Rep. 2016;13:2745–50. 26847592 Pan Z, Bu Q, You H, et al. Determining the optimal cutoff point for lymph node density and its impact on overall survival in children with Wilms’ tumor. Cancer Manag Res. 2019;11:759–66. PMC6339645 30697068 Elgendy A, Shehata S, Medhat Zaki A, Shehata S. National survey on the management of Wilms tumor. J Pediatr Hematol Oncol. 2018;41:280–85. 30102647 Yongfeng C, Xuemei W, Mei Y, Jungang X. [Expression of the Fra-1 gene in the peripheral blood of children with Wilms tumor]. Zhongguo Dang Dai Er Ke Za Zhi. 2019;21(2):161–64. [in Chinese] PMC7389833 30782279 Fuxing L, Jinshui L, Zhiqiang P, Lanhui C. [Efficacy and hygienic and economic analysis of comprehensive treatment for nephroblastoma in children]. Chin J of Clinical Rational Drug Use. 2018;11:163–64. [in Chinese] Zhu J, Fu W, Jia W, et al. Association between NER pathway gene polymorphisms and Wilms tumor risk. Mol Ther Nucleic Acids. 2018;12:854–60. PMC6118157 30161024 Zhu J, Jia W, Wu C, et al. Base excision repair gene polymorphisms and Wilms tumor susceptibility. EBioMedicine. 2018;33:88–93. PMC6085508 29937070 Wang X, Song P, Huang C, et al. Weighted gene co-expression network analysis for identifying hub genes in association with prognosis in Wilms tumor. Mol Med Rep. 2019;19:2041–50. PMC6390024 30664180 Diets IJ, Hoyer J, Ekici AB, et al. TRIM28 haploinsufficiency predisposes to Wilms tumor. Int J Cancer. 2019;145:941–51. 30694527 Liu K, He B, Xu J, et al. MiR-483-5p targets MKNK1 to suppress Wilms’ tumor cell proliferation and apoptosis in vitro and in vivo. Med Sci Monit. 2019;25:1459–68. PMC6398281 30798328 Fu W, Li L, Xiong SW, et al. MiR-423 rs6505162 C>A polymorphism contributes to decreased Wilms tumor risk. J Cancer. 2018;9:2460–65. PMC6036889 30026843 Chen YT, Xie JY, Sun Q, Mo WJ. Novel drug candidates for treating esophageal carcinoma: A study on differentially expressed genes, using connectivity mapping and molecular docking. Int J Oncol. 2019;54:152–66. PMC6254996 30387840 Pang JS, Li ZK, Lin P, et al. The underlying molecular mechanism and potential drugs for treatment in papillary renal cell carcinoma: A study based on TCGA and Cmap datasets. Oncol Rep. 2019;41:2089–102. PMC6412146 30816528 He J, Yan H, Cai H, et al. Statistically controlled identification of differentially expressed genes in one-to-one cell line comparisons of the CMAP database for drug repositioning. J Transl Med. 2017;15:198. PMC5622488 28962576 Jeong J, Kim H, Choi J. In silico molecular docking and in vivo validation with caenorhabditis elegans to discover molecular initiating events in adverse outcome pathway framework: Case study on endocrine-disrupting chemicals with estrogen and androgen receptors. Int J Mol Sci. 2019;20(5) pii: E1209. PMC6429066 30857347 Luo H, Jing B, Xia Y, et al. WP1130 reveals USP24 as a novel target in T-cell acute lymphoblastic leukemia. Cancer Cell Int. 2019;19:56. PMC6415346 30911287 Liu Z, Wang F, Zhou ZW, et al. Alisertib induces G2/M arrest, apoptosis, and autophagy via PI3K/Akt/mTOR- and p38 MAPK-mediated pathways in human glioblastoma cells. Am J Transl Res. 2017;9:845–73. PMC5375982 28386317 Shukla A, Tyagi R, Meena S, et al. 2D- and 3D-QSAR modelling, molecular docking and in vitro evaluation studies on 18beta-glycyrrhetinic acid derivatives against triple-negative breast cancer cell line. J Biomol Struct Dyn. 2019;22:1–18. 30686140 Abd El-Karim SS, Syam YM, El Kerdawy AM, Abdelghany TM. New thiazol-hydrazono-coumarin hybrids targeting human cervical cancer cells: Synthesis, CDK2 inhibition, QSAR and molecular docking studies. Bioorg Chem. 2019;86:80–96. 30685646 Chaube U, Chhatbar D, Bhatt H. 3D-QSAR, molecular dynamics simulations and molecular docking studies of benzoxazepine moiety as mTOR inhibitor for the treatment of lung cancer. Bioorg Med Chem Lett. 2016;26:864–74. 26764189 Cesaro S, Mauro M, Sattin G, et al. PAI-1 and protein C as early markers of veno-occlusive disease in patients treated for Wilms tumor. Pediatr Blood Cancer. 2019;66:e27695. 30865371 Le Rouzic MA, Mansuy L, Galloy MA, et al. Agreement between clinicoradiological signs at diagnosis and radiohistological analysis after neoadjuvant chemotherapy of suspected Wilms tumor rupture: Consequences on therapeutic choices. Pediatr Blood Cancer. 2019;66:e27674. 30773805 Su H, Wang X, Song J, et al. MicroRNA-539 inhibits the progression of Wilms’ Tumor through downregulation of JAG1 and Notch1/3. Cancer Biomark. 2019;24:125–33. PMC6398546 30530967 Jing P, Zou J, Weng K, Peng P. The PI3K/AKT axis modulates AATF activity in Wilms’ tumor cells. FEBS Open Bio. 2018;8(10):1615–23. PMC6168685 30338213 Li H, Huang D, Hang S. Salidroside inhibits the growth, migration and invasion of Wilms’ tumor cells through down-regulation of miR-891b. Life Sci. 2019;222:60–68. 30822424 Jia W, Deng F, Fu W, et al. Curcumin suppresses wilms’ tumor metastasis by inhibiting RECK methylation. Biomed Pharmacother. 2019;111:1204–12. 30841433 Groener JB, Gelen D, Mogler C, et al. BRAF V600E and retinoic acid in radioiodine-refractory papillary thyroid cancer. Horm Metab Res. 2019;51:69–75. 30396219 Zhang Y, Peng L, Chu J, et al. PH and redox dual-responsive copolymer micelles with surface charge reversal for co-delivery of all-trans-retinoic acid and paclitaxel for cancer combination chemotherapy. Int J Nanomedicine. 2018;13:6499–515. PMC6199233 30410335 Bouriez D, Giraud J, Gronnier C, Varon C. Efficiency of all-trans retinoic acid on gastric cancer: A narrative literature review. Int J Mol Sci. 2018;19(11) pii: E3388. PMC6275086 30380687 Wang K, Baldwin GS, Nikfarjam M, He H. Antitumor effects of all-trans retinoic acid and its synergism with gemcitabine are associated with downregulation of p21-activated kinases in pancreatic cancer. Am J Physiol Gastrointest Liver Physiol. 2019;316:G632–40. 30844294 Ghaffari H, Varner JD, Petzold LR. Analysis of the role of thrombomodulin in all-trans retinoic acid treatment of coagulation disorders in cancer patients. Theor Biol Med Model. 2019;16:3. PMC6376718 30764845 Zirn B, Samans B, Spangenberg C, et al. All-trans retinoic acid treatment of Wilms tumor cells reverses expression of genes associated with high risk and relapse in vivo. Oncogene. 2005;24:5246–51. 15897880 Yin P, Zheng N, Dong J, et al. Alsterpaullone induces apoptosis of HepG2 cells via a p38 mitogen-activated protein kinase signaling pathway. Oncol Lett. 2019;17:1177–83. PMC6312958 30655881 Cui C, Wang Y, Wang Y, et al. Alsterpaullone, a cyclin-dependent kinase inhibitor, mediated toxicity in HeLa cells through apoptosis-inducing effect. J Anal Methods Chem. 2013;2013 602091. PMC3610382 23577282 Lahusen T, De Siervi A, Kunick C, Senderowicz AM. Alsterpaullone, a novel cyclin-dependent kinase inhibitor, induces apoptosis by activation of caspase-9 due to perturbation in mitochondrial membrane potential. Mol Carcinog. 2003;36:183–94. 12669310 Bae T, Weon KY, Lee JW, et al. Restoration of paclitaxel resistance by CDK1 intervention in drug resistant ovarian cancer. Carcinogenesis. 2015;36:1561–71. 26442525 31612488 2021 02 02 2021 02 02 1097-4652 235 4 2020 Apr Journal of cellular physiology J Cell Physiol A novel risk signature that combines 10 long noncoding RNAs to predict neuroblastoma prognosis. 3823 3834 3823-3834 10.1002/jcp.29277 Neuroblastoma (NBL) is the most frequently encountered extracranial solid neoplasm and impacts significantly on the survival of patients, especially in cases of advanced tumor stage or relapse. A long noncoding RNA (lncRNA) signature to predict the survival of patients with NBL is proposed in this paper. Differentially expressed lncRNA (DElncRNA) was selected using the Limma plus Voom package in R based on the RNA-sequencing data downloaded from the Therapeutically Applicable Research To Generate Effective Treatments database and Genotype-Tissue Expression database. Univariate cox regression analysis, least absolute shrinkage and selection operator regression analysis, and multivariate cox regression analysis were conducted to identify candidate DElncRNAs for the risk signature. Consequently, 10 DElncRNAs were designated as candidate DElncRNAs for the risk signature. Time-dependent receiver operating characteristic curves and Kapan-Meier survival curves confirmed the efficacy of the risk signature in predicting the survival of patients with NBL (area under the curve = 0.941; p ≤ .001). One of the DElncRNA constituent subparts (LINC01010) was significantly associated with the survival outcome of patients with NBL in GSE62564 (p = .004). Thus, a risk signature comprising 10 DElncRNAs was identified as effective for individual risk stratification and the survival prediction outcomes of patients with NBL. © 2019 Wiley Periodicals, Inc. Gao Li L Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China. Lin Peng P Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China. Chen Peng P Department of Pediatric Surgery, First calculated using the following formula Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China. Gao Rui-Zhi RZ Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China. Yang Hong H Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China. He Yun Y Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China. Chen Jia-Bo JB Department of Pediatric Surgery, First calculated using the following formula Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China. Luo Yi-Ge YG Department of Pediatric Surgery, First calculated using the following formula Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China. Xu Qiong-Qian QQ Department of Pediatric Surgery, First calculated using the following formula Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China. Liang Song-Wu SW Department of Pediatric Surgery, First calculated using the following formula Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China. Gu Jin-Han JH Department of Pediatric Surgery, First calculated using the following formula Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China. Huang Zhi-Guang ZG Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China. Dang Yi-Wu YW Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China. Chen Gang G 0000-0002-4864-1451 Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China. eng Journal Article Research Support, Non-U.S. Gov't 2019 10 14 United States J Cell Physiol 0050222 0021-9541 0 Biomarkers, Tumor 0 RNA, Long Noncoding IM Biomarkers, Tumor genetics Female Gene Expression Regulation, Neoplastic genetics Humans Infant Kaplan-Meier Estimate Male Neoplasm Recurrence, Local genetics pathology Neuroblastoma genetics pathology Prognosis RNA, Long Noncoding classification genetics Risk Factors Sequence Analysis, RNA Transcriptome Therapeutically Applicable Research to Generate Effective Treatments long noncoding RNA neuroblastoma prognosis risk signature 2019 5 28 2019 9 27 2019 10 16 6 0 2021 2 3 6 0 2019 10 16 6 0 ppublish 31612488 10.1002/jcp.29277 REFERENCES Barone, G., Anderson, J., Pearson, A. D. J., Petrie, K., & Chesler, L. (2013). New strategies in neuroblastoma: Therapeutic targeting of MYCN and ALK. Clinical Cancer Research, 19(21), 5814-5821. https://doi.org/10.1158/1078-0432.ccr-13-0680 Benjamini, Y., & Hochberg, Y. (1995). Controlling for the false discovery rate: A practical and power approach to multiple testing. Journal of the Royal Statistical Society, 57(1), 289-300. https://doi.org/org/10.1111/j.2517-6161.1995.tb02031.x Bin, X., Hongjian, Y., Xiping, Z., Chen, B., Shifeng, Y., & Binbin, T. (2018). Research progresses in roles of LncRNA and its relationships with breast cancer. Cancer Cell International, 18, 179. https://doi.org/10.1186/s12935-018-0674-0 Brodeur, G., Seeger, R., Schwab, M., Varmus, H., & Bishop, J. (1984). Amplification of N-myc in untreated human neuroblastomas correlates with advanced disease stage. Science, 224(4653), 1121-1124. Carithers, L. J., & Moore, H. M. (2015). The genotype-tissue expression (GTEx) project. Biopreservation and Biobanking, 13(5), 307-308. https://doi.org/10.1089/bio.2015.29031.hmm Chen, L. P., Wang, H., Zhang, Y., Chen, Q. X., Lin, T. S., Liu, Z. Q., & Zhou, Y. Y. (2019). Robust analysis of novel mRNA-lncRNA cross talk based on ceRNA hypothesis uncovers carcinogenic mechanism and promotes diagnostic accuracy in esophageal cancer. Cancer Management and Research, 11, 347-358. https://doi.org/10.2147/cmar.s183310 Fan, C. N., Ma, L., & Liu, N. (2018). Systematic analysis of lncRNA-miRNA-mRNA competing endogenous RNA network identifies four-lncRNA signature as a prognostic biomarker for breast cancer. Journal of Translational Medicine, 16(1), 264. https://doi.org/10.1186/s12967-018-1640-2 Fatica, A., & Bozzoni, I. (2014). Long non-coding RNAs: New players in cell differentiation and development. Nature Reviews Genetics, 15(1), 7-21. https://doi.org/10.1038/nrg3606 Fusco, P., Esposito, M., & Tonini, G. (2018). Chromosome instability in neuroblastoma. Oncology Letters, 16(6), 6887-6894. https://doi.org/10.3892/ol.2018.9545 Gao, J., Kwan, P. W., & Shi, D. (2010). Sparse kernel learning with LASSO and Bayesian inference algorithm. Neural Networks, 23(2), 257-264. https://doi.org/10.1016/j.neunet.2009.07.001 Gao, L., Xie, Z.-C., Pang, J.-S., Li, T.-T., & Chen, G. (2019). A novel alternative splicing-based prediction model for uteri corpus endometrial carcinoma. Aging, 11(1), 263-283. https://doi.org/10.18632/aging.101753 Guan, S., Lu, J., Zhao, Y., Yu, Y., Li, H., Chen, Z., … Yang, J. (2018). MELK is a novel therapeutic target in high-risk neuroblastoma. Oncotarget, 9(2), 2591-2602. https://doi.org/10.18632/oncotarget.23515 Huang, M., & Weiss, W. A. (2013). Neuroblastoma and MYCN. Cold Spring Harbor Perspectives in Medicine, 3(10), a014415. https://doi.org/10.1101/cshperspect.a014415 Li, C. H., & Chen, Y. (2013). Targeting long non-coding RNAs in cancers: Progress and prospects. The International Journal of Biochemistry & Cell Biology, 45(8), 1895-1910. https://doi.org/10.1016/j.biocel.2013.05.030 Li, H., Zhao, X., Li, C., Sheng, C., & Bai, Z. (2019). Integrated analysis of lncRNA-associated ceRNA network reveals potential biomarkers for the prognosis of hepatitis B virus-related hepatocellular carcinoma. Cancer Management and Research, 11, 877-897. https://doi.org/10.2147/cmar.s186561 Li, Y., Zhang, J., Huo, C., Ding, N., Li, J., Xiao, J., … Xu, J. (2017). Dynamic organization of lncRNA and circular RNA regulators collectively controlled cardiac differentiation in humans. EBioMedicine, 24, 137-146. https://doi.org/10.1016/j.ebiom.2017.09.015 Lin, P., Guo, Y. N., Shi, L., Li, X., Yang, H., He, Y., … Chen, G. (2019). Development of a prognostic index based on an immunogenomic landscape analysis of papillary thyroid cancer. Aging, 11(2), 480-500. https://doi.org/10.18632/aging.101754 Lin, P., He, R., Ma, F., Liang, L., He, Y., Yang, H., … Chen, G. (2018). Systematic analysis of survival-associated alternative splicing signatures in gastrointestinal pan-adenocarcinomas. EBioMedicine, 34, 46-60. https://doi.org/10.1016/j.ebiom.2018.07.040 Lin, R. J., Lin, Y. C., Chen, J., Kuo, H. H., Chen, Y. Y., Diccianni, M. B., … Yu, A. L. (2010). microRNA signature and expression of Dicer and Drosha can predict prognosis and delineate risk groups in neuroblastoma. Cancer Research, 70(20), 7841-7850. https://doi.org/10.1158/0008-5472.CAN-10-0970 Liu, S., Xu, Y., & Zhang, S. (2018). LINGO1, C7orf31 and VEGFA are prognostic genes of primary glioblastoma: Analysis of gene expression microarray. Neoplasma, 65(4), 532-541. https://doi.org/10.4149/neo_2018_170722N496 Liu, X., Zhang, Z., Zhang, B., Zheng, Y., Zheng, C., Liu, B., … Dong, R. (2018). Circulating tumor cells detection in neuroblastoma patients by EpCAM-independent enrichment and immunostaining-fluorescence in situ hybridization. EBioMedicine, 35, 244-250. https://doi.org/10.1016/j.ebiom.2018.08.005 Maris, J. M. (2010). Recent advances in neuroblastoma. New England Journal of Medicine, 362(23), 2202-2211. https://doi.org/10.1056/NEJMra0804577 Maris, J. M., Hogarty, M. D., Bagatell, R., & Cohn, S. L. (2007). Neuroblastoma. The Lancet, 369(9579), 2106-2120. https://doi.org/10.1016/s0140-6736(07)60983-0 Matouk, I. J., DeGroot, N., Mezan, S., Ayesh, S., Abu-lail, R., Hochberg, A., & Galun, E. (2007). The H19 non-coding RNA is essential for human tumor growth. PLoS One, 2(9), e845. https://doi.org/10.1371/journal.pone.0000845 Mazar, J., Rosado, A., Shelley, J., Marchica, J., & Westmoreland, T. J. (2017). The long non-coding RNA GAS5 differentially regulates cell cycle arrest and apoptosis through activation of BRCA1 and p53 in human neuroblastoma. Oncotarget, 8(4), 6589-6607. https://doi.org/10.18632/oncotarget.14244 Miller, R. W., L Young, J., Jr., & Novakovic, B. (1995). Childhood cancer. Cancer, 75(Suppl 1), 395-405. Mohlin, S. A., Wigerup, C., & Påhlman, S. (2011). Neuroblastoma aggressiveness in relation to sympathetic neuronal differentiation stage. Seminars in Cancer Biology, 21(4), 276-282. https://doi.org/10.1016/j.semcancer.2011.09.002. O'Brien, E. M., Selfe, J. L., Martins, A. S., Walters, Z. S., & Shipley, J. M. (2018). The long non-coding RNA MYCNOS-01 regulates MYCN protein levels and affects growth of MYCN-amplified rhabdomyosarcoma and neuroblastoma cells. BMC Cancer, 18(1), 217. https://doi.org/10.1186/s12885-018-4129-8 Speleman, F., Park, J. R., & Henderson, T. O. (2016). Neuroblastoma: A tough nut to crack. American Society of Clinical Oncology Educational Book, 35, e548-e557. https://doi.org/10.14694/edbk_159169 Stafman, L., & Beierle, E. (2016). Cell proliferation in neuroblastoma. Cancers, 8(1), 13. https://doi.org/10.3390/cancers8010013 Tang, W., Dong, K., Li, K., Dong, R., & Zheng, S. (2016). MEG3, HCN3 and linc01105 influence the proliferation and apoptosis of neuroblastoma cells via the HIF-1α and p53 pathways. Scientific Reports, 6, 36268. https://doi.org/10.1038/srep36268 Wang, P., Jin, M., Sun, C. H., Yang, L., Li, Y. S., Wang, X., & Sun, Y. N (2018). A three-lncRNA expression signature predicts survival in head and neck squamous cell carcinoma (HNSCC). Bioscience Reports, 38(6), BSR20181528. https://doi.org/10.1042/bsr20181528 Wu, W. J., Shen, Y., Sui, J., Li, C. Y., Yang, S., Xu, S. Y., … Liang, G. Y. (2018). Integrated analysis of long noncoding RNA competing interactions revealed potential biomarkers in cervical cancer: Based on a public database. Molecular Medicine Reports, 17(6), 7845-7858. https://doi.org/10.3892/mmr.2018.8846 Wu, B., Wang, K., Fei, J., Bao, Y., Wang, X., Song, Z., … Zhong, Z. (2018). Novel three lncRNA signature predicts survival in patients with pancreatic cancer. Oncology Reports, 40(6), 3427-3437. https://doi.org/10.3892/or.2018.6761 Wu, Y., Wang, P. S., Wang, B. G., Xu, L., Fang, W. X., Che, X. F., … Li, Z. (2018). Genomewide identification of a novel six-LncRNA signature to improve prognosis prediction in resectable hepatocellular carcinoma. Cancer Medicine, 7(12), 6219-6233. https://doi.org/10.1002/cam4.1854 Xia, E., Bhandari, A., Shen, Y., Zhou, X., & Wang, O. (2018). lncRNA LINC00673 induces proliferation, metastasis and epithelial-mesenchymal transition in thyroid carcinoma via Kruppel-like factor 2. International Journal of Oncology, 53(5), 1927-1938. https://doi.org/10.3892/ijo.2018.4524 Yang, X., He, J., Chang, Y., Luo, A., Luo, A., Zhang, J., … Xu, L. (2018). HOTAIR gene polymorphisms contribute to increased neuroblastoma susceptibility in Chinese children. Cancer, 124(12), 2599-2606. https://doi.org/10.1002/cncr.31353 Yang, H., Lin, P., Wu, H. Y., Li, H. Y., He, Y., Dang, Y. W., … Chen, G. (2018). Genomic analysis of small nucleolar RNAs identifies distinct molecular and prognostic signature in hepatocellular carcinoma. Oncology Reports, 40(6), 3346-3358. https://doi.org/10.3892/or.2018.6715 Yang, T., Zhang, Z., Zhang, J., Tan, T., Yang, J., Pan, J., … Zhou, Y. (2018). The rs2147578 C > G polymorphism in the Inc-LAMC2-1:1 gene is associated with increased neuroblastoma risk in the Henan children. BMC Cancer, 18(1), 948. https://doi.org/10.1186/s12885-018-4847-y. 948 You, X., Yang, S., Sui, J., Wu, W., Liu, T., Xu, S., … Yao, Y. (2018). Molecular characterization of papillary thyroid carcinoma: A potential three-lncRNA prognostic signature. Cancer Management and Research, 10, 4297-4310. https://doi.org/10.2147/cmar.s174874 Zaatiti, H., Abdallah, J., Nasr, Z., Khazen, G., Sandler, A., & Abou-Antoun, T. (2018). Tumorigenic proteins upregulated in the MYCN-amplified IMR-32 human neuroblastoma cells promote proliferation and migration. International Journal of Oncology, 52(3), 787-803. https://doi.org/10.3892/ijo.2018.4236 Zhang, Z., Liu, Q., Wang, P., Li, J., He, T., Ouyang, Y., … Wang, W. (2018). Development and internal validation of a nine-lncRNA prognostic signature for prediction of overall survival in colorectal cancer patients. PeerJ, 6, e6061. https://doi.org/10.7717/peerj.6061 Zhang, R., Lin, P., Yang, X., He, R. Q., Wu, H. Y., Dang, Y. W., … Chen, G. (2018). Survival associated alternative splicing events in diffuse large B-cell lymphoma. American Journal of Translational Research, 10(8), 2636-2647. Zhang, J., Zhuo, Z., Li, W., Zhu, J., He, J., & Su, J. (2018). XRCC1 gene polymorphisms and risk of neuroblastoma in Chinese children. Aging, 10(10), 2944-2953. https://doi.org/10.18632/aging.101601 Zhong, X., Liu, Y., Liu, H., Zhang, Y., Wang, L., & Zhang, H. (2018). Identification of potential prognostic genes for neuroblastoma. Frontiers in Genetics, 9, 589. https://doi.org/10.3389/fgene.2018.00589 Zhuo, Z. J., Zhang, R., Zhang, J., Zhu, J., Yang, T., Zou, Y., … Xia, H. (2018). Associations between lncRNA MEG3 polymorphisms and neuroblastoma risk in Chinese children. Aging, 10(3), 481-491. https://doi.org/10.18632/aging.101406 Zhou, Y., Zhang, X., & Klibanski, A. (2012). MEG3 noncoding RNA: A tumor suppressor. Journal of Molecular Endocrinology, 48(3), R45-R53. https://doi.org/10.1530/jme-12-0008 trying2...
Publications by Jin-Han Gu | LitMetric
Publications by authors named "Jin-Han Gu"
We explored the difference in expression of tubulin alpha 1b (TUBA1B) between Wilms' tumor (WT) and normal tissues (NT) from in-house patients and databases, to determine TUBA1B expression in WT and the predictive pathways of coexpressed genes. In-house RNA-sequencing data were performed with WT and NT from three patients from our institute. Other four RNA-sequencing and microarray data were also downloaded from multiple public databases.
View Article and Find Full Text PDF
Ann Surg Treat Res
August 2020
Purpose : This study was performed to establish and validate a nomogram for predicting the overall survival in children with neuroblastoma.Methods : The latest clinical data of neuroblastoma in Surveillance, Epidemiology, and End Results (SEER) database was extracted from 2000 to 2016. The cases included were randomly divided into training and validation cohorts.
View Article and Find Full Text PDF
BACKGROUND Wilms tumor, or nephroblastoma, is a malignant pediatric embryonal renal tumor that has a poor prognosis. This study aimed to use bioinformatics data, RNA-sequencing, connectivity mapping, molecular docking, and ligand-protein binding to identify potential targets for drug therapy in Wilms tumor. MATERIAL AND METHODS Wilms tumor and non-tumor samples were obtained from high throughput gene expression databases, and differentially expressed genes (DEGs) were analyzed using the voom method in the limma package.
View Article and Find Full Text PDF
J Cell Physiol
April 2020
Neuroblastoma (NBL) is the most frequently encountered extracranial solid neoplasm and impacts significantly on the survival of patients, especially in cases of advanced tumor stage or relapse. A long noncoding RNA (lncRNA) signature to predict the survival of patients with NBL is proposed in this paper. Differentially expressed lncRNA (DElncRNA) was selected using the Limma plus Voom package in R based on the RNA-sequencing data downloaded from the Therapeutically Applicable Research To Generate Effective Treatments database and Genotype-Tissue Expression database.
View Article and Find Full Text PDF