Original Articles

Vol. 36 No. 6 (2025): Turkish Journal of Gastroenterology

m6A Methylation Regulator RBM15-Mediated Upregulation of ITGBL1 mRNA Stability Aggravates Colon Adenocarcinoma Progression by Remodeling the Tumor Microenvironment

Main Article Content

Jie Zhu
Dengliang Liu
Yingying Zou

Abstract

Background/Aims: Colon adenocarcinoma (COAD) is a prevalent malignant tumor of the digestive system. Previous research has indicated that RNA N6-methyladenosine (m6A) methyltransferase RNA-binding motif protein-15 (RBM15) is involved in various cancers. We aimed to investigate the function of RBM15 in COAD progression and its underlying molecular mechanism.


Materials and Methods: TIMER and UALCAN databases were applied to analyze the relationship between COAD and Integrin β-like 1 protein (ITGBL1) or RBM15. RT-qPCR and Western blot were used to analyze ITGBL1, M2-type macrophage markers, EMT-related markers, and RBM15 expression. CCK-8, colony formation, and transwell experiments detected cell viability, proliferation, migration, and invasion. The effect of ITGBL1 on COAD tumor growth was examined using a xenograft tumor model. The effects of COAD cells on macrophage polarization and the proliferation and apoptosis of CD8+ T cells were analyzed using flow cytometry analysis. Relationships between RBM15 and ITGBL1 were validated using MeRIP and dual-luciferase reporter assay.


Results: ITGBL1 and RBM15 contents were elevated in COAD. ITGBL1 knockdown could hinder COAD cell proliferation, migration, invasion, M2-type macrophage polarization, and lymphocyte immunity. Meanwhile, the lack of RBM15 dampened tumor growth in vivo. Mechanistically, RBM15 could increase ITGBL1 expression by m6A methylation.


Conclusion: RBM15 could promote COAD progression by regulating ITGBL1 mRNA stability, providing a promising biomarker and a potential target for COAD.

Cite this article as: Zhu J, Liu D, Zou Y. M6A methylation regulator RBM15-mediated upregulation of ITGBL1 mRNA stability
aggravates colon adenocarcinoma progression by remodeling the tumor microenvironment. Turk J Gastroenterol.
2025;36(6):343-356.

Article Details

References

1. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin. 2024;74(1):12-49. [CrossRef]

2. Aktan Ç, Tekin F, Oruç N, Özütemiz Ö. CHRM3-associated miRNAs may play a role in bile acid-induced proliferation of H508 colon cancer cells. Turk J Gastroenterol. 2023;34(3):298-307. [CrossRef]

3. Hu B, Chen Z, Yao F, Li B. Construction of a risk model for colon cancer prognosis based on ubiquitin-related genes. Turk J Gastroenterol. 2023;34(5):449-456. [CrossRef]

4. Alvizo-Rodríguez CR, Flores-López BA, Ayala-Madrigal ML, et al. Interaction of CCND2, CDKN1A, and POLD3 Variants in Mexican Patients with Colorectal Cancer. Turk J Gastroenterol. 2022;33(6):525-531.

5. Liu Y, Duan L, Li L, et al. A Network Pharmacology-based Mechanism of the traditional Chinese medicine Formula Li Kun Zhi Ji acting on Colon Cancer. Lett Drug Des Discov. 2023;20(9):1274-1283. [CrossRef]

6. Achilli P, Crippa J, Grass F, et al. Survival impact of adjuvant chemotherapy in patients with stage IIA colon cancer: analysis of the National Cancer Database. Int J Cancer. 2021;148(1):161-169. [CrossRef]

7. Cha JH, Chan LC, Song MS, Hung MC. New approaches on cancer immunotherapy. Cold Spring Harb Perspect Med. 2020;10(8). [CrossRef]

8. Hugo W, Zaretsky JM, Sun L, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell. 2017;168(3):542. [CrossRef]

9. Cheli Y, Tulic MK, El Hachem N, et al. ITGBL1 is a new immunomodulator that favors development of melanoma tumors by inhibiting natural killer cells cytotoxicity. Mol Cancer. 2021;20(1):12. [CrossRef]

10. Yin FY, Qi HP, Qiao H, Lv XH, Tan HH. ITGBL1 promotes gastric cancer cell proliferation and invasion via Akt signal pathway. Front Biosci (Landmark Ed). 2021;26(4):682-691. [CrossRef]

11. Du T, Zhang K, Zhang Z, Guo A, Yu G, Xu Y. ITGBL1 transcriptionally inhibited by JDP2 promotes the development of pancreatic cancer through the TGF-beta/Smad pathway. Braz J Med Biol Res. 2022;55:e11989. [CrossRef]

12. Qiu X, Feng JR, Qiu J, et al. ITGBL1 promotes migration, invasion and predicts a poor prognosis in colorectal cancer. Biomed Pharmacother. 2018;104:172-180. [CrossRef]

13. Matsuyama T, Ishikawa T, Takahashi N, et al. Transcriptomic expression profiling identifies ITGBL1, an epithelial to mesenchymal transition (EMT)-associated gene, is a promising recurrence prediction biomarker in colorectal cancer. Mol Cancer. 2019;18(1):19. [CrossRef]

14. Qi L, Ding Y. Construction of key signal regulatory network in metastatic colorectal cancer. Oncotarget. 2018;9(5):6086-6094. [CrossRef]

15. Liu J, Harada BT, He C. Regulation of gene expression by N(6)-methyladenosine in cancer. Trends Cell Biol. 2019;29(6):487-499. [CrossRef]

16. Tian S, Lai J, Yu T, Li Q, Chen Q. Regulation of gene expression associated with the N6-methyladenosine (m6A) enzyme system and its significance in cancer. Front Oncol. 2020;10:623634. [CrossRef]

17. Liu T, Li C, Jin L, Li C, Wang L. The prognostic value of m6A RNA methylation regulators in colon adenocarcinoma. Med Sci Monit. 2019;25:9435-9445. [CrossRef]

18. Zhang Z, Mei Y, Hou M. Knockdown RBM15 inhibits colorectal cancer cell proliferation and metastasis via N6-methyladenosine (m6A) modification of MyD88 mRNA. Cancer Biother Radiopharm. 2022;37(10):976-986. [CrossRef]

19. Chen J. Regulatory mechanism of RNA binding motif protein 15-mediated N(6) methyladenosine modification in proliferation, invasion, and migration of colorectal cancer cells. Environ Toxicol. 2023;38(11):2545-2559. [CrossRef]

20. Zhao Z, Ju Q, Ji J, Li Y, Zhao Y. N6-methyladenosine methylation regulator RBM15 is a potential prognostic biomarker and promotes cell proliferation in pancreatic adenocarcinoma. Front Mol Biosci. 2022;9:842833. [CrossRef]

21. Dong H, Zhang H, Mao X, Liu S, Xu W, Zhang Y. RBM15 promates the proliferation, migration and invasion of pancreatic cancer cell lines. Cancers. 2023;15(4). [CrossRef]

22. Pisibon C, Ouertani A, Bertolotto C, Ballotti R, Cheli Y. Immune checkpoints in cancers: from signaling to the clinic. Cancers. 2021;13(18). [CrossRef]

23. Blank CU, Haanen JB, Ribas A, Schumacher TN. Cancer immunology. The “cancer immunogram”. Science (New York, NY). 2016;352(6286):658-660. [CrossRef]

24. Song EK, Jeon J, Jang DG, et al. ITGBL1 modulates integrin activity to promote cartilage formation and protect against arthritis. Sci Transl Med. 2018;10(462). [CrossRef]

25. Akhtar N, Streuli CH. An integrin-ILK-microtubule network orients cell polarity and lumen formation in glandular epithelium. Nat Cell Biol. 2013;15(1):17-27. [CrossRef]

26. Cortez AJ, Kujawa KA, Wilk AM, et al. Evaluation of the role of ITGBL1 in ovarian cancer. Cancers. 2020;12(9). [CrossRef]

27. Bai Z, Zhou Y, Ye Z, Xiong J, Lan H, Wang F. Tumor-infiltrating lymphocytes in colorectal cancer: the fundamental indication and application on immunotherapy. Front Immunol. 2021;12:808964. [CrossRef]

28. Tay C, Tanaka A, Sakaguchi S. Tumor-infiltrating regulatory T cells as targets of cancer immunotherapy. Cancer Cell. 2023;41(3):450-465. [CrossRef]

29. An Y, Duan H. The role of m6A RNA methylation in cancer metabolism. Mol Cancer. 2022;21(1):14. [CrossRef]

30. Hiriart E, Gruffat H, Buisson M, et al. Interaction of the Epstein-Barr virus mRNA export factor EB2 with human Spen proteins SHARP, OTT1, and a novel member of the family, OTT3, links Spen proteins with splicing regulation and mRNA export. J Biol Chem. 2005;280(44):36935-36945. [CrossRef]

Similar Articles

<< < 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 > >> 

You may also start an advanced similarity search for this article.