Welcome to visit Zhongnan Medical Journal Press Series journal website!

Home Articles Vol 37,2024 No.8 Detail

Expression of ADAM12, MUC3A and MUC17 in gastric cancer and analysis of survival prognosis

Published on Sep. 04, 2024Total Views: 179 times Total Downloads: 99 times Download Mobile

Author: LI Yuxin GUO Tangxi WEI Yongchang

Affiliation: Department of Gastrointestinal Tumors Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China

Keywords: ADAM12 MUC3A MUC17 Gastric cancer Survival prognosis Targeted therapy

DOI: 10.12173/j.issn.1004-4337.202403010

Reference: Li YX, Guo TX, Wei YC. Expression of ADAM12, MUC3A and MUC17 in gastric cancer and analysis of survival prognosis[J]. Journal of Mathematical Medicine, 2024, 37(8): 600-609. DOI: 10.12173/j.issn.1004-4337.202403010[Article in Chinese]

  • Abstract
  • Full-text
  • References
Abstract

Objective  To investigate the expression of a disintergin and metalloprotease 12 (ADAM12), mucin 3A (MUC3A) and mucin 17 (MUC17) in gastric cancer and the relationship between them and survival prognosis.

Methods  Information from The Cancer Genome Atlas (TCGA) and Gene Expression Profiling Interactive Analysis 2 (GEPIA2) databases was collected, and R 4.2.2 as well as SPSS software were used to analyse the differential expression, survival-prognosis relationship, pan-cancer expression, and correlation between different clinicopathological features and expressions of ADAM12, MUC3A and MUC17 genes in gastric cancer. The expression levels of the three genes in normal tissues were analysed by Human Protein Atlas (HPA) database. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were used to evaluate the potential functions of the three genes. The expression of ADAM12, MUC3A and MUC17 in tumour tissues and paracancerous tissues was further verified using quantitative real-time polymerase chain reaction (q-PCR).

Results  Bioinformatics analysis and q-PCR results showed that the expression of ADAM12, MUC3A and MUC17 were low in normal tissues and all upregulated in gastric cancer (P<0.05), and ADAM12 was accompanied by a poor prognosis in gastric cancer (P<0.05). The three genes were enriched in tumor-related pathways and associated with the development of gastric cancer.

Conclusion  ADAM12, MUC3A and MUC17 show high expression in gastric cancer, of which ADAM12 is an influencing factor in the prognosis of gastric cancer. All three genes are associated with the progression of tumors and may be potential therapeutic targets for gastric cancer.

Full-text
Please download the PDF version to read the full text: download
References

1.Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries[J]. CA Cancer J Clin, 2021, 71(3): 209-249. DOI: 10.3322/caac.21660.

2.Thrift AP, Wenker TN, El-Serag HB. Global burden of gastric cancer: epidemiological trends, risk factors, screening and prevention[J]. Nat Rev Clin Oncol, 2023, 20(5): 338-349. DOI: 10.1038/s41571-023-00747-0.

3.Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012[J]. Int J Cancer, 2015, 136(5): E359-E386. DOI: 10.1002/ijc.29210.

4.Joshi SS, Badgwell BD. Current treatment and recent progress in gastric cancer[J]. CA Cancer J Clin, 2021, 71(3): 264-279. DOI: 10.3322/caac.21657.

5.Nyren-Erickson EK, Jones JM, Srivastava DK, et al. A disintegrin and metalloproteinase-12 (ADAM12): function, roles in disease progression, and clinical implications[J]. Biochim Biophys Acta, 2013, 1830(10): 4445-4455. DOI: 10.1016/j.bbagen.2013.05.011.

6.Wang M, Lu S, Zhu Y, et al. ADAM12 is an effective marker in the second trimester of pregnancy for prenatal screening of Down syndrome[J]. Prenat Diagn, 2010, 30(6): 561-564. DOI: 10.1002/pd.2523.

7.Ren K, Ruan Y, Tang J, et al. Association of ADAM12 gene polymorphisms with knee osteoarthritis susceptibility[J]. Oncotarget, 2017, 8(44): 77710-77721. DOI: 10.18632/oncotarget.20772.

8.Brockhausen I, Melamed J. Mucins as anti-cancer targets: perspectives of the glycobiologist[J]. Glycoconj J, 2021, 38(4): 459-474. DOI: 10.1007/s10719-021-09986-8.

9.Yonezawa S, Higashi M, Yamada N, et al. Mucins in human neoplasms: clinical pathology, gene expression and diagnostic application[J]. Pathol Int, 2011, 61(12): 697-716. DOI: 10.1111/j.1440-1827.2011.02734.x.

10.Gum JR Jr, Crawley SC, Hicks JW, et al. MUC17, a novel membrane-tethered mucin[J]. Biochem Biophys Res Commun, 2002, 291(3): 466-475. DOI: 10.1006/bbrc.2002.6475.

11.Qiu H, Cao S, Xu R. Cancer incidence, mortality, and burden in China: a time-trend analysis and comparison with the United States and United Kingdom based on the global epidemiological data released in 2020[J]. Cancer Commun (Lond), 2021, 41(10): 1037-1048. DOI: 10.1002/cac2.12197.

12.Guan WL, He Y, Xu RH. Gastric cancer treatment: recent progress and future perspectives[J]. J Hematol Oncol, 2023, 16(1): 57. DOI: 10.1186/s13045-023-01451-3.

13.Liu Z, Zhou Z, Dang Q, et al. Immunosuppression in tumor immune microenvironment and its optimization from CAR-T cell therapy[J]. Theranostics, 2022, 12(14): 6273-6290. DOI: 10.7150/thno.76854.

14.Liu G, Rui W, Zhao X, et al. Enhancing CAR-T cell efficacy in solid tumors by targeting the tumor microenvironment[J]. Cell Mol Immunol, 2021, 18(5): 1085-1095. DOI: 10.1038/s41423-021-00655-2.

15.Waarts MR, Stonestrom AJ, Park YC, et al. Targeting mutations in cancer[J]. J Clin Invest, 2022, 132(8): e154943. DOI: 10.1172/JCI154943.

16.Edwards DR, Handsley MM, Pennington CJ. The ADAM metalloproteinases[J]. Mol Aspects Med, 2008, 29(5): 258-289. DOI: 10.1016/j.mam.2008.08.001.

17.Primakoff P, Myles DG. The ADAM gene family: surface proteins with adhesion and protease activity[J]. Trends Genet, 2000, 16(2): 83-87. DOI: 10.1016/s0168-9525(99)01926-5.

18.Chang Z, Duan Q, Yu C, et al. Proteomics and biochemical analyses of secreted proteins revealed a novel mechanism by which ADAM12S regulates the migration of gastric cancer cells[J]. J Proteome Res, 2022, 21(9): 2160-2172. DOI: 10.1021/acs.jproteome.2c00221.

19.Wang R, Godet I, Yang Y, et al. Hypoxia-inducible factor-dependent ADAM12 expression mediates breast cancer invasion and metastasis[J]. Proc Natl Acad Sci USA, 2021, 118(19): e2020490118. DOI: 10.1073/pnas.2020490118.

20.Roy R, Dagher A, Butterfield C, et al. ADAM12 is a novel regulator of tumor angiogenesis via STAT3 signaling[J]. Mol Cancer Res, 2017, 15(11): 1608-1622. DOI: 10.1158/1541-7786.MCR-17-0188.

21.Wang G, Romero Y, Thevarajan I, et al. ADAM12 abrogation alters immune cell infiltration and improves response to checkpoint blockade therapy in the T11 murine model of triple-negative breast cancer[J]. Oncoimmunology, 2022, 12(1): 2158006. DOI: 10.1080/2162402X.2022.2158006.

22.Piotrowski KB, Blasco LP, Samsøe-Petersen J, et al. ADAM12 expression is upregulated in cancer cells upon radiation and constitutes a prognostic factor in rectal cancer patients following radiotherapy[J]. Cancer Gene Ther, 2023, 30(10): 1369-1381. DOI: 10.1038/s41417-023-00643-w.

23.Linden SK, Sutton P, Karlsson NG, et al. Mucins in the mucosal barrier to infection[J]. Mucosal Immunol, 2008, 1(3): 183-197. DOI: 10.1038/mi.2008.5.

24.Merikallio H, Pincikova T, Kotortsi I, et al. Mucins 3A and 3B are expressed in the epithelium of human large airway[J]. Int J Mol Sci, 2023, 24(17): 13546. DOI: 10.3390/ijms241713546.

25.Kufe DW. Mucins in cancer: function, prognosis and therapy[J]. Nat Rev Cancer, 2009, 9(12): 874-885. DOI: 10.1038/nrc2761.

26.Su W, Feng B, Hu L, et al. MUC3A promotes the progression of colorectal cancer through the PI3K/Akt/mTOR pathway[J]. BMC Cancer, 2022, 22(1): 602. DOI: 10.1186/s12885-022-09709-8.

27.Sun Y, Sun X, You C, et al. MUC3A promotes non-small cell lung cancer progression via activating the NFκB pathway and attenuates radiosensitivity[J]. Int J Biol Sci, 2021, 17(10): 2523-2536. DOI: 10.7150/ijbs.59430.

28.Luo Y, Ma S, Sun Y, et al. MUC3A induces PD-L1 and reduces tyrosine kinase inhibitors effects in EGFR-mutant non-small cell lung cancer[J]. Int J Biol Sci, 2021, 17(7): 1671-1681. DOI: 10.7150/ijbs.57964.

29.Schneider H, Pelaseyed T, Svensson F, et al. Study of mucin turnover in the small intestine by in vivo labeling[J]. Sci Rep, 2018, 8(1): 5760. DOI: 10.1038/s41598-018-24148-x.

30.Resta-Lenert S, Das S, Batra SK, et al. Muc17 protects intestinal epithelial cells from enteroinvasive E. coli infection by promoting epithelial barrier integrity[J]. Am J Physiol Gastrointest Liver Physiol, 2011, 300(6): G1144-G1155. DOI: 10.1152/ajpgi.00138.2010.

31.Almasmoum H. The roles of transmembrane mucins located on chromosome 7q22.1 in colorectal cancer[J]. Cancer Manag Res, 2021, 13: 3271-3280. DOI: 10.2147/CMAR.S299089.

32.Cox KE, Liu S, Lwin TM, et al. The mucin family of proteins: candidates as potential biomarkers for colon cancer[J]. Cancers (Basel), 2023, 15(5): 1491. DOI: 10.3390/cancers15051491.

33.Yuan C, Yao X, Dai P, et al. Genomic alterations dissection revealed MUC4 mutation as a potential driver in lung adenocarcinoma local recurrence[J]. Transl Lung Cancer Res, 2023, 12(5): 985-998. DOI: 10.21037/tlcr-22-793.

34.Lin S, Ruan H, Qin L, et al. Acquired resistance to EGFR-TKIs in NSCLC mediates epigenetic downregulation of MUC17 by facilitating NF-κB activity via UHRF1/DNMT1 complex[J]. Int J Biol Sci, 2023, 19(3): 832-851. DOI: 10.7150/ijbs.75963.

35.汪泽慧, 张军. 黏蛋白型O-糖基化与结直肠癌基础和临床研究进展[J]. 胃肠病学, 2023, 28(3): 181-185. [Wang ZH, Zhang J. Progress of basic and clinical research on mucin-type O-glycosylation and colorectal cancer[J]. Chinese Journal of Gastroenterology, 2023, 28(3): 181-185.] DOI: 10.3969/j.issn.1008-7125.2023.03.005.

Popular papers
Last 6 months