ERMP1 Facilitates The Malignant Characteristics of Colorectal Cancer Cells through Modulating PI3K/AKT/β-Catenin Pathway and Localization of GRP78

Document Type : Original Article

Authors

1 Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran

2 Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran

3 Autophagy Research Center, Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran

Abstract

Objective: Endoplasmic reticulum-metallopeptidase 1 (ERMP1) is involved in cellular response to oxidative stress.
However, its functional role in proliferation and progression of cancer cells remains unknown. The focus of this study
was to investigate the molecular-mechanisms in which ERMP1 modulates the proliferation and progression of colorectal
cancer (CRC) cells under normal and environment stress conditions.
Materials and Methods: In this experimental study, ERMP1 expression was evaluated using reverse transcriptionquantitative
polymerase chain reaction (RT-qPCR) in CRC cells. ERMP1 was knocked down using lentiviral transduction
of ERMP1-specific shRNA into HCT116 cells. ERMP1 was also upregulated using lipofectamine transfection of
ERMP1-overexpressing vector into SW48 cells. To evaluate the role of ERMP1 in the cellular and environmental stress
conditions, ERMP1-downregulated cells were exposed to stressful conditions including starvation, serum free medium,
and treatment with redox or chemotherapy agents for 72 hours. The expression of AKT, p-AKT, phospho-mammalian
target of rapamycin (p-mTOR), β-catenin, p-β-catenin, E-cadherin, and Glucose-regulating protein 78 (GRP78) proteins
was evaluated by western blotting. The expression of ERMP1, CYCLIN D, and c-MYC was evaluated by RT-qPCR. The
cell surface localization of GRP78, cell cycle distribution, and apoptosis were determined by Flow cytometry.
Results: ERMP1 knock-down reduced the cellular proliferation, inactivated the PI3K/AKT pathway, prompted the
G1 arrest, and attenuated the free β-catenin and CYCLIN D expression. Opposite results were obtained in ERMP1-
overexpressed cells. Knock-down of ERMP1 also reduced the GRP78 localization at the cell surface. Various
environmental stress conditions differently affected the ERMP1-downregulated cells.
Conclusion: ERMP1 functioned as an oncogene in CRC cells by promoting malignant characteristics. The
phosphoinositide 3-kinases (PI3K)/AKT/β-catenin pathway and localization of GRP78 were closely related to the effects
of ERMP1. Consequently, ERMP1 might be regarded as a promising target in therapeutic strategies related to CRC.

Keywords

Main Subjects


  1. Grandi A, Santi A, Campagnoli S, Parri M, De Camilli E, Song C, et al. ERMP1, a novel potential oncogene involved in UPR and oxidative stress defense, is highly expressed in human cancer. Oncotarget. 2016; 7(39): 63596-63610.
  2. Garcia-Rudaz C, Luna F, Tapia V, Kerr B, Colgin L, Galimi F, et al. Fxna, a novel gene differentially expressed in the rat ovary at the time of folliculogenesis, is required for normal ovarian histogenesis. Development (Cambridge, England). 2007; 134(5): 945-957.
  3. Wu J, Liu S, Liu G, Dombkowski A, Abrams J, Martin-Trevino R, et al. Identification and functional analysis of 9p24 amplified genes in human breast cancer. Oncogene. 2012; 31(3): 333-341.
  4. Biller LH, Schrag D. Diagnosis and treatment of metastatic colorectal cancer: a review. JAMA. 2021; 325(7): 669-685.
  5. Sakata S, Larson DW. Targeted therapy for colorectal cancer. Surg Oncol Clin N Am. 2022; 31(2): 255-264.
  6. Shiu RP, Pouyssegur J, Pastan I. Glucose depletion accounts for the induction of two transformation-sensitive membrane proteinsin Rous sarcoma virus-transformed chick embryo fibroblasts. Proc Natl Acad Sci USA. 1977; 74(9): 3840-3844.
  7. Farshbaf M, Khosroushahi AY, Mojarad-Jabali S, Zarebkohan A, Valizadeh H, Walker PR. Cell surface GRP78: An emerging imaging marker and therapeutic target for cancer. J Control Release. 2020; 328: 932-941.
  8. Qu J, Zhang L, Li L, Su Y. miR-148b functions as a tumor suppressor by targeting endoplasmic reticulum metallo protease 1 in human endometrial cancer cells. Oncol Res. 2018; 27(1): 81-88.
  9. Lu H, Hu J, Li J, Lu W, Deng X, Wang X. miR-328-3p overexpression attenuates the malignant proliferation and invasion of liver cancer via targeting endoplasmic reticulum metallo protease 1 to inhibit AKT phosphorylation. Ann Transl Med. 2020; 8(12): 754.
  10. Porta C, Paglino C, Mosca A. Targeting PI3K/Akt/mTOR signaling in cancer. Front Oncol. 2014; 4: 64
  11. Lien EC, Lyssiotis CA, Cantley LC. Metabolic reprogramming by the PI3K-Akt-mTOR pathway in cancer. Recent Results Cancer Res. 2016; 207: 39-72.
  12. Saavedra-García P, Roman-Trufero M, Al-Sadah HA, Blighe K, López-Jiménez E, Christoforou M, et al. Systems level profiling of chemotherapy-induced stress resolution in cancer cells reveals druggable trade-offs. Proc Natl Acad Sci USA. 2021; 118(17): e2018229118.
  13. Al-Akra L, Bae DH, Leck LYW, Richardson DR, Jansson PJ. The biochemical and molecular mechanisms involved in the role of tumor micro-environment stress in development of drug resistance. Biochim Biophys Acta Gen Subj. 2019; 1863(9): 1390-1397.
  14. Xiao Z, Osipyan A, Song S, Chen D, Schut RA, van Merkerk R, et al. Thieno [2, 3-d] pyrimidine-2, 4 (1 H, 3 H)-dione derivative inhibits d-dopachrome tautomerase activity and suppresses the proliferation of non-small cell lung cancer cells. J Med Chem. 2022; 65(3): 2059-2077.
  15. Sun Y, Campisi J, Higano C, Beer TM, Porter P, Coleman I, et al. Treatment-induced damage to the tumor microenvironment promotes prostate cancer therapy resistance through WNT16B. Nat Med. 2012; 18(9): 1359-1368.
  16. Tiligada E. Chemotherapy: induction of stress responses. Endocr Relat Cancer. 2006; 13 Suppl 1: S115-S124.
  17. Xiang XY, Yang XC, Su J, Kang JS, Wu Y, Xue YN, et al. Inhibition of autophagic flux by ROS promotes apoptosis during DTT-induced ER/oxidative stress in HeLa cells. Oncol Rep. 2016; 35(6): 3471- 3479.
  18. Tsai CW, Yang MD, Hsia TC, Chang WS, Hsu CM, Hsieh YH, et al. Dithiothreitol enhanced arsenic-trioxide-induced cell apoptosis in cultured oral cancer cells via mitochondrial dysfunction and endoplasmic reticulum stress. Environ Toxicol. 2017; 32(1): 17-27.
  19. Mal’tseva VN, Goltyaev MV, Novoselov SV, Varlamova EG. Effects of sodium selenite and dithiothreitol on expression of endoplasmic reticulum selenoproteins and apoptosis markers in MSF7 breast adenocarcinoma cells. Mol Biol (Mosk). 2022; 56(1): 135-146.
  20. Tan Z, Chan YJA, Chua YJK, Rutledge SD, Pavelka N, Cimini D, et al. Environmental stresses induce karyotypic instability in colorectal cancer cells. Mol Biol Cell. 2019; 30(1): 42-55.
  21. Anderson AR, Weaver AM, Cummings PT, Quaranta V. Tumor morphology and phenotypic evolution driven by selective pressure from the microenvironment. Cell. 2006; 127(5): 905-915.
  22. Pàez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Viñals F, et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell. 2009; 15(3): 220-231.
  23. Vasudev NS, Reynolds AR. Anti-angiogenic therapy for cancer: current progress, unresolved questions and future directions. Angiogenesis. 2014; 17(3): 471-494.
  24. Narayanankutty A. PI3K/ Akt/ mTOR pathway as a therapeutic target for colorectal cancer: a review of preclinical and clinical evidence. Curr Drug Targets. 2019; 20(12): 1217-1226.
  25. Karimi Roshan M, Soltani A, Soleimani A, Rezaie Kahkhaie K, Afshari AR, Soukhtanloo M. Role of AKT and mTOR signaling pathways in the induction of epithelial-mesenchymal transition (EMT) process. Biochimie. 2019; 165: 229-234.
  26. Yan J, Wong N, Hung C, Chen WX, Tang D. Contactin-1 reduces Ecadherin expression via activating AKT in lung cancer. PLoS One. 2013; 8(5): e65463.
  27. He R, Du S, Lei T, Xie X, Wang Y. Glycogen synthase kinase 3β in tumorigenesis and oncotherapy (Review). Oncol Rep. 2020; 44(6): 2373-2385.
  28. Alves M, Borges DP, Kimberly A, Martins Neto F, Oliveira AC, de Sousa JC, et al. Glycogen synthase kinase-3 beta expression correlates with worse overall survival in non-small cell lung cancer-Aclinicopathological series. Front Oncol. 2021; 11: 621050.
  29. Liu J, Lam JB, Chow KH, Xu A, Lam KS, Moon RT, et al. Adiponectin stimulates Wnt inhibitory factor-1 expression through epigenetic regulations involving the transcription factor specificity protein 1. Carcinogenesis. 2008; 29(11): 2195-2202.
  30. Yun SH, Park JI. PGC-1α regulates cell proliferation and invasion via AKT/GSK-3β/β-catenin pathway in human colorectal cancer SW620 and SW480 cells. Anticancer Res. 2020; 40(2): 653- 664.
  31. Chen JS, Huang JQ, Luo B, Dong SH, Wang RC, Jiang ZK, et al. PIK3CD induces cell growth and invasion by activating AKT/ GSK-3β/β-catenin signaling in colorectal cancer. Cancer Sci. 2019; 110(3): 997-1011.
  32. Prossomariti A, Piazzi G, Alquati C, Ricciardiello L. Are Wnt/β- catenin and PI3K/AKT/mTORC1 distinct pathways in colorectal cancer? Cell Mol Gastroenterol Hepatol. 2020; 10(3): 491-506.
  33. Gonzalez-Avila G, Sommer B, Mendoza-Posada DA, Ramos C, Garcia-Hernandez AA, Falfan-Valencia R. Matrix metalloproteinases participation in the metastatic process and their diagnostic and therapeutic applications in cancer. Crit Rev Oncol Hematol. 2019; 137: 57-83.
  34. Hong KO, Kim JH, Hong JS, Yoon HJ, Lee JI, Hong SP, et al. Inhibition of Akt activity induces the mesenchymal-to-epithelial reverting transition with restoring E-cadherin expression in KB and KOSCC- 25B oral squamous cell carcinoma cells. J Exp Clin Cancer Res. 2009; 28(1): 28.
  35. García-Gutiérrez L, Delgado MD, León J. MYC oncogene contributions to release of cell cycle brakes. Genes (Basel). 2019; 10(3): 244.
  36. Huang H, Weng H, Zhou H, Qu L. Attacking C-MYC: targeted and combined therapies for cancer. Curr Pharm Des. 2014; 20(42): 6543-6554.
  37. Xi J, Chen Y, Huang S, Cui F, Wang X. Suppression of GRP78 sensitizes human colorectal cancer cells to oxaliplatin by downregulation of CD24. Oncol Lett. 2018; 15(6): 9861-9867.
  38. Liu R, Li X, Gao W, Zhou Y, Wey S, Mitra SK, et al. Monoclonal antibody against cell surface GRP78 as a novel agent in suppressing PI3K/AKT signaling, tumor growth, and metastasis. Clin Cancer Res. 2013; 19(24): 6802-6811.
  39. Fu R, Yang P, Wu HL, Li ZW, Li ZY. GRP78 secreted by colon cancer cells facilitates cell proliferation via PI3K/Akt signaling. Asian Pac J Cancer Prev. 2014; 15(17): 7245-7249.
  40. Li Z, Wang Y, Wu H, Zhang L, Yang P, Li Z. GRP78 enhances the glutamine metabolism to support cell survival from glucose deficiency by modulating the β-catenin signaling. Oncotarget. 2014; 5(14): 53.