Skip to main content
Top
Published in: BMC Cancer 1/2022

Open Access 01-12-2022 | Colorectal Cancer | Research

MUC3A promotes the progression of colorectal cancer through the PI3K/Akt/mTOR pathway

Authors: Wei Su, Baijie Feng, Lina Hu, Xianzhi Guo, Minghua Yu

Published in: BMC Cancer | Issue 1/2022

Login to get access

Abstract

Mucin 3A (MUC3A) is overexpressed in colorectal cancer (CRC) and associated with poor prognosis, but the related mechanism remains unclear. Our study found that MUC3A promotes the progression of CRC by activating the PI3K/Akt/mTOR signaling pathway. Knockout of MUC3A significantly inhibited the proliferation of CRC cells and induced G1 phase arrest by upregulating p21 protein, an important cell cycle regulator. Moreover, knockout of MUC3A significantly inhibited invasion ability and enhanced the sensitivity to the chemotherapeutic agent 5-FU. Furthermore, we found that knockout of MUC3A repressed the PI3K/Akt/mTOR pathway through RNA-seq. Treatment with the PI3K/Akt/mTOR pathway inhibitor rapamycin successfully eliminated the difference in proliferation, invasion and chemoresistance between MUC3A knockout cells and control cells. Our study suggests that MUC3A is a potential oncogene that promotes the proliferation, invasion, and chemotherapy resistance of CRC. Moreover, CRC patients with high expression of MUC3A may benefit from rapamycin treatment. 
Appendix
Available only for authorised users
Literature
1.
go back to reference Sung H, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209–49.CrossRef Sung H, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209–49.CrossRef
3.
go back to reference Yatsuoka T, et al. Lymph node metastasis of colorectal cancer with submucosal invasion. Gan To Kagaku Ryoho. 2013;40(12):2041–3.PubMed Yatsuoka T, et al. Lymph node metastasis of colorectal cancer with submucosal invasion. Gan To Kagaku Ryoho. 2013;40(12):2041–3.PubMed
4.
go back to reference Shi Y, et al. Dragon (RGMb) induces oxaliplatin resistance in colon cancer cells. Oncotarget. 2016;7(30):48027–37.CrossRef Shi Y, et al. Dragon (RGMb) induces oxaliplatin resistance in colon cancer cells. Oncotarget. 2016;7(30):48027–37.CrossRef
5.
go back to reference Miura K, et al. 5-fu metabolism in cancer and orally-administrable 5-fu drugs. Cancers (Basel). 2010;2(3):1717–30.CrossRef Miura K, et al. 5-fu metabolism in cancer and orally-administrable 5-fu drugs. Cancers (Basel). 2010;2(3):1717–30.CrossRef
6.
go back to reference Gum JR Jr, et al. MUC3 human intestinal mucin. Analysis of gene structure, the carboxyl terminus, and a novel upstream repetitive region. J Biol Chem. 1997;272(42):26678–86.CrossRef Gum JR Jr, et al. MUC3 human intestinal mucin. Analysis of gene structure, the carboxyl terminus, and a novel upstream repetitive region. J Biol Chem. 1997;272(42):26678–86.CrossRef
7.
go back to reference Kyo K, et al. Associations of distinct variants of the intestinal mucin gene MUC3A with ulcerative colitis and Crohn’s disease. J Hum Genet. 2001;46(1):5–20.CrossRef Kyo K, et al. Associations of distinct variants of the intestinal mucin gene MUC3A with ulcerative colitis and Crohn’s disease. J Hum Genet. 2001;46(1):5–20.CrossRef
8.
go back to reference Korbut E, et al. Molecular Profile of Barrett’s Esophagus and Gastroesophageal Reflux Disease in the Development of Translational Physiological and Pharmacological Studies. Int J Mol Sci. 2020;21(17):6436.CrossRef Korbut E, et al. Molecular Profile of Barrett’s Esophagus and Gastroesophageal Reflux Disease in the Development of Translational Physiological and Pharmacological Studies. Int J Mol Sci. 2020;21(17):6436.CrossRef
9.
go back to reference Ho SB, et al. Cysteine-rich domains of muc3 intestinal mucin promote cell migration, inhibit apoptosis, and accelerate wound healing. Gastroenterology. 2006;131(5):1501–17.CrossRef Ho SB, et al. Cysteine-rich domains of muc3 intestinal mucin promote cell migration, inhibit apoptosis, and accelerate wound healing. Gastroenterology. 2006;131(5):1501–17.CrossRef
10.
go back to reference Zimmer KP, et al. TGFalpha-associated MUC2 and MUC3 expression of the gastric epithelium in Menetrier’s disease during remission of ulcerative colitis. Gut. 2011;60(11):1607–8.CrossRef Zimmer KP, et al. TGFalpha-associated MUC2 and MUC3 expression of the gastric epithelium in Menetrier’s disease during remission of ulcerative colitis. Gut. 2011;60(11):1607–8.CrossRef
11.
go back to reference Niu T, et al. Increased expression of MUC3A is associated with poor prognosis in localized clear-cell renal cell carcinoma. Oncotarget. 2016;7(31):50017–26.CrossRef Niu T, et al. Increased expression of MUC3A is associated with poor prognosis in localized clear-cell renal cell carcinoma. Oncotarget. 2016;7(31):50017–26.CrossRef
12.
go back to reference Sotoudeh M, et al. MSLN (Mesothelin), ANTXR1 (TEM8), and MUC3A are the potent antigenic targets for CAR T cell therapy of gastric adenocarcinoma. J Cell Biochem. 2019;120(4):5010–7.CrossRef Sotoudeh M, et al. MSLN (Mesothelin), ANTXR1 (TEM8), and MUC3A are the potent antigenic targets for CAR T cell therapy of gastric adenocarcinoma. J Cell Biochem. 2019;120(4):5010–7.CrossRef
13.
go back to reference Luo Y, et al. MUC3A induces PD-L1 and reduces tyrosine kinase inhibitors effects in EGFR-mutant non-small cell lung cancer. Int J Biol Sci. 2021;17(7):1671–81.CrossRef Luo Y, et al. MUC3A induces PD-L1 and reduces tyrosine kinase inhibitors effects in EGFR-mutant non-small cell lung cancer. Int J Biol Sci. 2021;17(7):1671–81.CrossRef
14.
go back to reference Byrd JC, Bresalier RS. Mucins and mucin binding proteins in colorectal cancer. Cancer Metastasis Rev. 2004;23(1–2):77–99.CrossRef Byrd JC, Bresalier RS. Mucins and mucin binding proteins in colorectal cancer. Cancer Metastasis Rev. 2004;23(1–2):77–99.CrossRef
15.
go back to reference Sun Y, et al. MUC3A promotes non-small cell lung cancer progression via activating the NFkappaB pathway and attenuates radiosensitivity. Int J Biol Sci. 2021;17(10):2523–36.CrossRef Sun Y, et al. MUC3A promotes non-small cell lung cancer progression via activating the NFkappaB pathway and attenuates radiosensitivity. Int J Biol Sci. 2021;17(10):2523–36.CrossRef
16.
go back to reference Yang Z, et al. SIRT6/HIF-1alpha axis promotes papillary thyroid cancer progression by inducing epithelial-mesenchymal transition. Cancer Cell Int. 2019;19:17.CrossRef Yang Z, et al. SIRT6/HIF-1alpha axis promotes papillary thyroid cancer progression by inducing epithelial-mesenchymal transition. Cancer Cell Int. 2019;19:17.CrossRef
17.
go back to reference Bailie L, Loughrey MB, Coleman HG. Lifestyle Risk Factors for Serrated Colorectal Polyps: A Systematic Review and Meta-analysis. Gastroenterology. 2017;152(1):92–104.CrossRef Bailie L, Loughrey MB, Coleman HG. Lifestyle Risk Factors for Serrated Colorectal Polyps: A Systematic Review and Meta-analysis. Gastroenterology. 2017;152(1):92–104.CrossRef
18.
go back to reference Jung AY, et al. DNA methyltransferase and alcohol dehydrogenase: gene-nutrient interactions in relation to risk of colorectal polyps. Cancer Epidemiol Biomarkers Prev. 2008;17(2):330–8.CrossRef Jung AY, et al. DNA methyltransferase and alcohol dehydrogenase: gene-nutrient interactions in relation to risk of colorectal polyps. Cancer Epidemiol Biomarkers Prev. 2008;17(2):330–8.CrossRef
19.
go back to reference Dekker E, Rex DK. Advances in CRC Prevention: Screening and Surveillance. Gastroenterology. 2018;154(7):1970–84.CrossRef Dekker E, Rex DK. Advances in CRC Prevention: Screening and Surveillance. Gastroenterology. 2018;154(7):1970–84.CrossRef
20.
go back to reference Duncan TJ, et al. The role of MUC1 and MUC3 in the biology and prognosis of colorectal cancer. World J Surg Oncol. 2007;5:31.CrossRef Duncan TJ, et al. The role of MUC1 and MUC3 in the biology and prognosis of colorectal cancer. World J Surg Oncol. 2007;5:31.CrossRef
21.
go back to reference Barr AR, et al. DNA damage during S-phase mediates the proliferation-quiescence decision in the subsequent G1 via p21 expression. Nat Commun. 2017;8:14728.CrossRef Barr AR, et al. DNA damage during S-phase mediates the proliferation-quiescence decision in the subsequent G1 via p21 expression. Nat Commun. 2017;8:14728.CrossRef
22.
go back to reference Cannell IG, et al. A Pleiotropic RNA-Binding Protein Controls Distinct Cell Cycle Checkpoints to Drive Resistance of p53-Defective Tumors to Chemotherapy. Cancer Cell. 2015;28(5):623–37.CrossRef Cannell IG, et al. A Pleiotropic RNA-Binding Protein Controls Distinct Cell Cycle Checkpoints to Drive Resistance of p53-Defective Tumors to Chemotherapy. Cancer Cell. 2015;28(5):623–37.CrossRef
23.
go back to reference Alzahrani AS. PI3K/Akt/mTOR inhibitors in cancer: At the bench and bedside. Semin Cancer Biol. 2019;59:125–32.CrossRef Alzahrani AS. PI3K/Akt/mTOR inhibitors in cancer: At the bench and bedside. Semin Cancer Biol. 2019;59:125–32.CrossRef
24.
go back to reference Wen Y, et al. HER-2/neu promotes androgen-independent survival and growth of prostate cancer cells through the Akt pathway. Cancer Res. 2000;60(24):6841–5.PubMed Wen Y, et al. HER-2/neu promotes androgen-independent survival and growth of prostate cancer cells through the Akt pathway. Cancer Res. 2000;60(24):6841–5.PubMed
25.
go back to reference Spokoini R, et al. Glycogen synthase kinase-3 plays a central role in mediating glucocorticoid-induced apoptosis. Mol Endocrinol. 2010;24(6):1136–50.CrossRef Spokoini R, et al. Glycogen synthase kinase-3 plays a central role in mediating glucocorticoid-induced apoptosis. Mol Endocrinol. 2010;24(6):1136–50.CrossRef
26.
go back to reference Murugan AK. mTOR: Role in cancer, metastasis and drug resistance. Semin Cancer Biol. 2019;59:92–111.CrossRef Murugan AK. mTOR: Role in cancer, metastasis and drug resistance. Semin Cancer Biol. 2019;59:92–111.CrossRef
27.
go back to reference Hua H, et al. Targeting mTOR for cancer therapy. J Hematol Oncol. 2019;12(1):71.CrossRef Hua H, et al. Targeting mTOR for cancer therapy. J Hematol Oncol. 2019;12(1):71.CrossRef
28.
go back to reference Benjamin D, et al. Rapamycin passes the torch: a new generation of mTOR inhibitors. Nat Rev Drug Discov. 2011;10(11):868–80.CrossRef Benjamin D, et al. Rapamycin passes the torch: a new generation of mTOR inhibitors. Nat Rev Drug Discov. 2011;10(11):868–80.CrossRef
Metadata
Title
MUC3A promotes the progression of colorectal cancer through the PI3K/Akt/mTOR pathway
Authors
Wei Su
Baijie Feng
Lina Hu
Xianzhi Guo
Minghua Yu
Publication date
01-12-2022
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2022
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-022-09709-8

Other articles of this Issue 1/2022

BMC Cancer 1/2022 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine