1
|
Jin H, Liang G, Huang W, Wang Z, Wu L, Li Y. AGR2 activates the TGF-β/Smad signaling pathway to promote epithelial-mesenchymal transition, invasion, and metastasis in nasopharyngeal carcinoma. Eur Arch Otorhinolaryngol 2025; 282:2411-2418. [PMID: 40119906 DOI: 10.1007/s00405-025-09328-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/03/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Anterior gradient 2 protein (AGR2) is associated with tumorigenesis and metastasis in different cancers. However, its role in nasopharyngeal carcinoma (NPC) remains unknown. This study aimed to explore the effect of AGR2 on epithelial-mesenchymal transition (EMT) in NPC and its underlying mechanisms. METHODS AGR2 expression was analyzed in cancerous and para-cancerous tissues from ten NPC patients using RT-qPCR. Western blotting was used to determine the AGR2 protein levels in two NPC cell lines and a nasopharyngeal epithelial cell line. AGR2 was overexpressed or knocked out in NPC cells and its effects on cell viability, migration, invasion, and EMT markers were evaluated in vitro. RESULT AGR2 expression was significantly higher in NPC tissues compared to adjacent normal tissues. Similarly, NPC cell lines exhibited increased AGR2 levels compared to the nasopharyngeal epithelial cell line. AGR2 knockout significantly reduced cell viability, migration, and invasion. It also decreased N-cadherin protein levels while increasing E-cadherin, α-SMA, and vimentin expression. Conversely, AGR2 overexpression produced the opposite effects. Furthermore, AGR2 deletion inactivated the TGF-β/Smad signaling pathway. CONCLUSION AGR2 promotes tumor progression and EMT in NPC through activation of the TGF-β/Smad signaling pathway. These findings suggest that AGR2 may serve as a potential biomarker and therapeutic target for NPC.
Collapse
Affiliation(s)
- Hui Jin
- Department of Otolaryngology, Wuhan Third Hospital, No. 241 Pengliuyang Road, Wuchang District, Wuhan City, 430000, Hubei Province, China
| | - Gengtian Liang
- Department of Otolaryngology, Wuhan Third Hospital, No. 241 Pengliuyang Road, Wuchang District, Wuhan City, 430000, Hubei Province, China.
| | - Wenxia Huang
- Department of Otolaryngology, Wuhan Third Hospital, No. 241 Pengliuyang Road, Wuchang District, Wuhan City, 430000, Hubei Province, China
| | - Zhen Wang
- Department of Otolaryngology, Wuhan Third Hospital, No. 241 Pengliuyang Road, Wuchang District, Wuhan City, 430000, Hubei Province, China
| | - Longjun Wu
- Department of Otolaryngology, Wuhan Third Hospital, No. 241 Pengliuyang Road, Wuchang District, Wuhan City, 430000, Hubei Province, China
| | - Yaping Li
- Department of Cardiology, Wuhan Third Hospital, Wuhan, 430000, Hubei Province, China
| |
Collapse
|
2
|
Zhang Y, Zhang J. AGR2 facilitates teratoma progression by regulating glycolysis via the AnXA2/EGFR axis. Exp Cell Res 2024; 442:114228. [PMID: 39197578 DOI: 10.1016/j.yexcr.2024.114228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/07/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024]
Abstract
Anterior gradient-2 (AGR2) is highly expressed in several tumors and plays an important role in tumor development. However, the biological function of AGR2 in teratomas has not yet been thoroughly studied. In this study, AGR2 was found to be upregulated in teratoma tissues and in human testicular teratoma cell lines by Western blotting and qRT-PCR assays. A DNA Methylation-Specific PCR assay demonstrated that AGR2 upregulation resulted from hypomethylated AGR2 in teratoma cells. NCC-IT and NT2-D1 cells were transfected with pcDNA-AGR2 or sh-AGR2 to obtain AGR2-overexpressed or -silenced cells, and cell proliferation, invasion and glycolysis were determined using CCK-8, 5-ethynyl-2'-deoxyuridine (EdU), Transwell assays, and commercial kits. The results revealed that overexpression of AGR2 promoted teratoma cell proliferation and invasion and elevated glycolysis levels evidencing by the increase in lactate secretion, glucose consumption, ATP levels and the expression of glycolysis-related proteins, while knockdown of AGR2 showed the opposite results. The interactions between AGR2 and annexin A2 (AnXA2), as well as between AnXA2 and epidermal growth factor receptor (EGFR) were verified by co-immunoprecipitation assay. Mechanistic studies revealed that AGR2 interacts with AnXA2 and increases the level of AnXA2 to recruit more AnXA2 to EGFR, there by promoting EGFR expression. A series of rescue experiments showed that knockdown of AnXA2 or EGFR weakened the promotional effects of AGR2 overexpression on the proliferation, invasion, and glycolysis of teratoma cells. Finally, tumorigenicity assays were performed using NT2-D1 cells stably transfected with either LV-NC-shRNA or LV-shAGR2. The results showed that AGR2 knockdown significantly inhibited teratoma tumor growth in vivo. In conclusion, our data suggested that AGR2 facilitates glycolysis in teratomas through promoting EGFR expression by interacting with AnXA2, thereby promoting teratoma cells proliferation and invasion.
Collapse
Affiliation(s)
- Yahong Zhang
- Department of Gynecology, Baoji People's Hospital, No. 24 Xinhua Lane, Jing'er Road, Baoji, 721000, Shaanxi Province, China
| | - Jing Zhang
- Department of Gynecology, Baoji Central Hospital, No. 8 Jiangtan Road, Weibin District, Baoji, 721008, Shaanxi Province, China.
| |
Collapse
|
3
|
Qu S, Jia W, Nie Y, Shi W, Chen C, Zhao Z, Song W. AGR2: The Covert Driver and New Dawn of Hepatobiliary and Pancreatic Cancer Treatment. Biomolecules 2024; 14:743. [PMID: 39062458 PMCID: PMC11275012 DOI: 10.3390/biom14070743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/04/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
The anterior gradient protein 2 (AGR2) plays a crucial role in facilitating the formation of protein disulfide bonds within the endoplasmic reticulum (ER). Research suggests that AGR2 can function as an oncogene, with its heightened expression linked to the advancement of hepatobiliary and pancreatic cancers through invasion and metastasis. Notably, AGR2 not only serves as a pro-oncogenic agent but also as a downstream targeting protein, indirectly fostering cancer progression. This comprehensive review delves into the established functions and expression patterns of AGR2, emphasizing its pivotal role in cancer progression, particularly in hepatobiliary and pancreatic malignancies. Furthermore, AGR2 emerges as a potential cancer prognostic marker and a promising target for immunotherapy, offering novel avenues for the treatment of hepatobiliary and pancreatic cancers and enhancing patient outcomes.
Collapse
Affiliation(s)
- Shen Qu
- Xi’an Medical University, Xi’an 710021, China; (S.Q.); (W.J.); (W.S.); (C.C.)
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (Y.N.); (Z.Z.)
| | - Weili Jia
- Xi’an Medical University, Xi’an 710021, China; (S.Q.); (W.J.); (W.S.); (C.C.)
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (Y.N.); (Z.Z.)
| | - Ye Nie
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (Y.N.); (Z.Z.)
| | - Wen Shi
- Xi’an Medical University, Xi’an 710021, China; (S.Q.); (W.J.); (W.S.); (C.C.)
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (Y.N.); (Z.Z.)
| | - Chao Chen
- Xi’an Medical University, Xi’an 710021, China; (S.Q.); (W.J.); (W.S.); (C.C.)
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (Y.N.); (Z.Z.)
| | - Zihao Zhao
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (Y.N.); (Z.Z.)
| | - Wenjie Song
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (Y.N.); (Z.Z.)
| |
Collapse
|
4
|
Fu M, Deng F, Chen J, Fu L, Lei J, Xu T, Chen Y, Zhou J, Gao Q, Ding H. Current data and future perspectives on DNA methylation in ovarian cancer (Review). Int J Oncol 2024; 64:62. [PMID: 38757340 PMCID: PMC11095605 DOI: 10.3892/ijo.2024.5650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
Ovarian cancer (OC) represents the most prevalent malignancy of the female reproductive system. Its distinguishing features include a high aggressiveness, substantial morbidity and mortality, and a lack of apparent symptoms, which collectively pose significant challenges for early detection. Given that aberrant DNA methylation events leading to altered gene expression are characteristic of numerous tumor types, there has been extensive research into epigenetic mechanisms, particularly DNA methylation, in human cancers. In the context of OC, DNA methylation is often associated with the regulation of critical genes, such as BRCA1/2 and Ras‑association domain family 1A. Methylation modifications within the promoter regions of these genes not only contribute to the pathogenesis of OC, but also induce medication resistance and influence the prognosis of patients with OC. As such, a more in‑depth understanding of DNA methylation underpinning carcinogenesis could potentially facilitate the development of more effective therapeutic approaches for this intricate disease. The present review focuses on classical tumor suppressor genes, oncogenes, signaling pathways and associated microRNAs in an aim to elucidate the influence of DNA methylation on the development and progression of OC. The advantages and limitations of employing DNA methylation in the diagnosis, treatment and prevention of OC are also discussed. On the whole, the present literature review indicates that the DNA methylation of specific genes could potentially serve as a prognostic biomarker for OC and a therapeutic target for personalized treatment strategies. Further investigations in this field may yield more efficacious diagnostic and therapeutic alternatives for patients with OC.
Collapse
Affiliation(s)
- Mengyu Fu
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Fengying Deng
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jie Chen
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Li Fu
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jiahui Lei
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Ting Xu
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Department of Gynecology and Obstetrics, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215100, P.R. China
| | - Youguo Chen
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jinhua Zhou
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Qinqin Gao
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Hongmei Ding
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
5
|
Zheng C, Mao Y, Ye J, Zhang M, Chen Y. Function and mechanism of exogenous AGR2 in colorectal cancer cells. Heliyon 2024; 10:e28175. [PMID: 38560175 PMCID: PMC10981063 DOI: 10.1016/j.heliyon.2024.e28175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
Background Anterior gradient 2 (AGR2) is highly enriched in several malignant tumors and can boost tumor metastasis. Whereas, AGR2 role in colorectal cancer (CRC) is not clear. Methods AGR2 expression in the GEPIA database was studied, and the results were confirmed by Western blot in CRC cell lines (SW480, SW620, and HT-29). The impact of AGR2 on the multiplication, migration, invasion and EMT of CRC cells were studied by CCK-8 assay, as well as clone formation, wound healing and transwell assays. The protein concent related to the AKT/β-catenin signaling pathway were accessed via Western blot. Results AGR2 concent in CRC tissues was notablely boosted versus normal colorectal tissues. Exogenous AGR2 boosted the multiplication of CRC cells. In addition, exogenous AGR2 induced EMT, which demonstrated that ZEB1, N-cadherin, Vimentin, Slug, Snail protein concent boosted and E-cadherin protein abated in CRC cells. In terms of mechanism, exogenous AGR2 upgulated p-AKT/AKT, p-GSK3β/GSK3β and β-catenin concent. Exogenous AGR2 combined with AKT agonist IGF- Ⅰ can further enhance the multiplication, migration and invasion of CRC cells. Conclusion Exogenous AGR2 enhances the multiplication of CRC cells and induces EMT process, the mechanism of which is related to AKT/β-catenin signal pathway.
Collapse
Affiliation(s)
- Chao Zheng
- Department of General Surgery, The People's Hospital of Yuhuan, Taizhou, 317600, Zhejiang, China
| | - Yu Mao
- Department of General Surgery, The People's Hospital of Yuhuan, Taizhou, 317600, Zhejiang, China
| | - Jianping Ye
- Department of General Surgery, The People's Hospital of Yuhuan, Taizhou, 317600, Zhejiang, China
| | - Miaolong Zhang
- Department of General Surgery, The People's Hospital of Yuhuan, Taizhou, 317600, Zhejiang, China
| | - Yongfeng Chen
- Department of General Surgery, The People's Hospital of Yuhuan, Taizhou, 317600, Zhejiang, China
| |
Collapse
|
6
|
Kwok H, Li H, Yang J, Deng J, Lee NC, Au TW, Sit AK, Hsin MK, Ma SK, Cheung LW, Girard L, Fujimoto J, Wistuba II, Gao B, Minna JD, Lam DC. Single-cell transcriptomic analysis uncovers intratumoral heterogeneity and drug-tolerant persister in ALK-rearranged lung adenocarcinoma. Cancer Commun (Lond) 2023; 43:951-955. [PMID: 37272226 PMCID: PMC10397560 DOI: 10.1002/cac2.12449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 04/21/2023] [Accepted: 05/26/2023] [Indexed: 06/06/2023] Open
Affiliation(s)
- Hoi‐Hin Kwok
- Department of Medicine, Li Ka Shing Faculty of MedicineUniversity of Hong KongHong Kong SARP. R. China
| | - Huiyu Li
- Nancy B. and Jake L. Hamon Center for Therapeutic Oncology ResearchUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Jiashuang Yang
- Department of Medicine, Li Ka Shing Faculty of MedicineUniversity of Hong KongHong Kong SARP. R. China
| | - Junyang Deng
- Department of Medicine, Li Ka Shing Faculty of MedicineUniversity of Hong KongHong Kong SARP. R. China
| | - Nerissa Chui‐Mei Lee
- Department of Medicine, Li Ka Shing Faculty of MedicineUniversity of Hong KongHong Kong SARP. R. China
| | - Timmy Wing‐Kuk Au
- Cardiothoracic Surgical DepartmentQueen Mary HospitalHong Kong SARP. R. China
| | - Alva Ko‐Yung Sit
- Cardiothoracic Surgical DepartmentQueen Mary HospitalHong Kong SARP. R. China
| | | | - Stephanie Kwai‐Yee Ma
- School of Biomedical Sciences, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARP. R. China
| | - Lydia Wai‐Ting Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARP. R. China
| | - Luc Girard
- Nancy B. and Jake L. Hamon Center for Therapeutic Oncology ResearchUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Junya Fujimoto
- Department of Translational Molecular Pathology, Division of Pathology/Lab MedicineUniversity of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Ignacio Ivan Wistuba
- Department of Translational Molecular Pathology, Division of Pathology/Lab MedicineUniversity of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Boning Gao
- Nancy B. and Jake L. Hamon Center for Therapeutic Oncology ResearchUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - John Dorrance Minna
- Nancy B. and Jake L. Hamon Center for Therapeutic Oncology ResearchUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - David Chi‐Leung Lam
- Department of Medicine, Li Ka Shing Faculty of MedicineUniversity of Hong KongHong Kong SARP. R. China
| |
Collapse
|
7
|
Gene expression related to lung cancer altered by PHMG-p treatment in PBTE cells. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00319-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
8
|
Cai W, Jing M, Wen J, Guo H, Xue Z. Epigenetic Alterations of DNA Methylation and miRNA Contribution to Lung Adenocarcinoma. Front Genet 2022; 13:817552. [PMID: 35711943 PMCID: PMC9194831 DOI: 10.3389/fgene.2022.817552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 04/26/2022] [Indexed: 12/24/2022] Open
Abstract
This study focused on the epigenetic alterations of DNA methylation and miRNAs for lung adenocarcinoma (LUAD) diagnosis and treatment using bioinformatics analyses. DNA methylation data and mRNA and miRNA expression microarray data were obtained from The Cancer Genome Atlas (TCGA) database. The differentially methylated genes (DMGs), differentially expressed genes (DEGs), and differentially expressed miRNAs were analyzed by using the limma package. The DAVID database performed GO and KEGG pathway enrichment analyses. Using STRING and Cytoscape, we constructed the protein-protein interaction (PPI) network and achieved visualization. The online analysis tool CMap was used to identify potential small-molecule drugs for LUAD. In LUAD, 607 high miRNA-targeting downregulated genes and 925 low miRNA-targeting upregulated genes, as well as 284 hypermethylated low-expression genes and 315 hypomethylated high-expression genes, were obtained. They were mainly enriched in terms of pathways in cancer, neuroactive ligand-receptor interaction, cAMP signaling pathway, and cytosolic DNA-sensing pathway. In addition, 40 upregulated and 84 downregulated genes were regulated by both aberrant alternations of DNA methylation and miRNAs. Five small-molecule drugs were identified as a potential treatment for LUAD, and five hub genes (SLC2A1, PAX6, LEP, KLF4, and FGF10) were found in PPI, and two of them (SLC2A1 and KLF4) may be related to the prognosis of LUAD. In summary, our study identified a series of differentially expressed genes associated with epigenetic alterations of DNA methylation and miRNA in LUAD. Five small-molecule drugs and five hub genes may be promising drugs and targets for LUAD treatment.
Collapse
Affiliation(s)
- Wenhan Cai
- Medical School of Chinese PLA, Beijing, China
| | - Miao Jing
- Medical School of Chinese PLA, Beijing, China
| | - Jiaxin Wen
- Department of Thoracic Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Hua Guo
- Medical School of Chinese PLA, Beijing, China
| | - Zhiqiang Xue
- Department of Thoracic Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|