1
|
Chen Y, Deng S, Xu J, Yan Y, Lan S, Guo M. Research status and hotspots on the mechanisms of liver X receptor in cancer progression: A bibliometric analysis. Medicine (Baltimore) 2024; 103:e37126. [PMID: 38552096 PMCID: PMC10977575 DOI: 10.1097/md.0000000000037126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/10/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND The mechanism of liver X receptor in cancer has been gradually revealed in recent years. This study is committed to analyzing the current research status of the mechanism of liver × receptor in cancer progression by using bibliometric methods and to explore the development trend of liver × receptor related research in the future, in order to provide some reference for further exploration in this field. METHODS The Web of Science core collection database was used to carry out the original data retrieval. Excel software was used for data statistics. Vosviewer and CiteSpace software were used to analyze the publication situation, cooperation network, reference co-citation, keyword and term co-occurrence, term bursts, and cluster analysis, and draw visual maps. RESULTS A total of 631 publications meeting the research criteria were included by December 2022, with an average of 32.5 citations per paper. The main research fields were molecular biology, oncology and cell biology, and the papers were mainly published in journals about molecular, biology and immunology. Cell is the journal with the highest citation. The United States is the most influential country, the University of California, Los Angeles is the main research institution, and Gustafsson, Jan-ake is the author with the highest output. In reference co-citation clustering, cluster#2 "cancer development" is the main cluster, and the period from 2014 to 2018 is an important stage of relevant theoretical progress. "Tumor microenvironment" with high burst and novelty became the most noteworthy term in term burst. CONCLUSION Using bibliometric methods to reveal the current status of LXR and cancer mechanisms, and making predictions of possible future hotspots based on the analysis of the current situation, the translation of LXR anti-cancer research to clinical applications, the impact on the tumor microenvironment as a whole and more immune pathways, and the formation of a systematic cognition of the effects of more cancer cell lines and oncogenic signaling crosstalk, which is a possible direction for future research.
Collapse
Affiliation(s)
- Yukun Chen
- Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Siqi Deng
- Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jiexia Xu
- Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yu Yan
- Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Shuwen Lan
- Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Mingzhang Guo
- Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
2
|
Long Y, Wu J, Shen Y, Gan C, Zhang C, Wang G, Jing J, Zhang C, Pan W. CAPG is a novel biomarker for early gastric cancer and is involved in the Wnt/β-catenin signaling pathway. Cell Death Discov 2024; 10:15. [PMID: 38191512 PMCID: PMC10774411 DOI: 10.1038/s41420-023-01767-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 01/10/2024] Open
Abstract
Past studies have shown that the Gelsolin-like actin-capping protein (CAPG) regulates cell migration and proliferation and is strongly associated with tumor progression. We present the first study of the mechanism of action of CAPG in early gastric cancer (EGC). We demonstrate that CAPG expression is upregulated in gastric cancer (GC) especially EGC. CAPG promotes GC proliferation, migration, invasion, and metastasis in vivo and in vitro. More importantly, CAPG plays a role in GC by involving the Wnt/β-catenin signaling pathway. Our findings suggest that CAPG may function as a novel biomarker for EGC.
Collapse
Affiliation(s)
- Yan Long
- Cancer Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou, Zhejiang, China
| | - JiaQi Wu
- Department of Gastroenterology, Affiliated Hospital of Hangzhou Normal University, 310015, Hangzhou, China
| | - Yu Shen
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenxiao Gan
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuandong Zhang
- The Medical College of QingDao University, Qingdao, Shandong, China
| | - Gang Wang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Jiyong Jing
- Department of Medical Education and Simulation Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China
| | - Chenjing Zhang
- Cancer Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou, Zhejiang, China.
| | - Wensheng Pan
- Cancer Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Han N, Yuan M, Yan L, Tang H. Emerging Insights into Liver X Receptor α in the Tumorigenesis and Therapeutics of Human Cancers. Biomolecules 2023; 13:1184. [PMID: 37627249 PMCID: PMC10452869 DOI: 10.3390/biom13081184] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Liver X receptor α (LXRα), a member of the nuclear receptor superfamily, is identified as a protein activated by ligands that interacts with the promoters of specific genes. It regulates cholesterol, bile acid, and lipid metabolism in normal physiological processes, and it participates in the development of some related diseases. However, many studies have demonstrated that LXRα is also involved in regulating numerous human malignancies. Aberrant LXRα expression is emerging as a fundamental and pivotal factor in cancer cell proliferation, invasion, apoptosis, and metastasis. Herein, we outline the expression levels of LXRα between tumor tissues and normal tissues via the Oncomine and Tumor Immune Estimation Resource (TIMER) 2.0 databases; summarize emerging insights into the roles of LXRα in the development, progression, and treatment of different human cancers and their diversified mechanisms; and highlight that LXRα can be a biomarker and therapeutic target in diverse cancers.
Collapse
Affiliation(s)
- Ning Han
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Man Yuan
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Libo Yan
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
4
|
Feng Z, Ding H, Peng Z, Hu K. Downregulated KDM6A mediates gastric carcinogenesis via Wnt/β-catenin signaling pathway mediated epithelial-to-mesenchymal transition. Pathol Res Pract 2023; 245:154461. [PMID: 37060821 DOI: 10.1016/j.prp.2023.154461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/29/2023] [Accepted: 04/10/2023] [Indexed: 04/17/2023]
Abstract
This study explored the connection between KDM6A expression and patient prognosis and the mechanism of KDM6A's role in developing GC (GC). From the immunohistochemical Analysis of 107 GC patients' tumors, we discovered that patients with reduced KDM6A expression had a shorter survival time. There was a correlation between KDM6A expression and the degree of differentiation of tumor tissue, T stage, N stage, and TNM stage. KDM6A gene expression was positively connected with the expression level of E-cadherin and negatively connected with the expression level of N-cadherin and vimentin in vitro tests. KDM6A gene suppression prevented GC cell proliferation, migration, and invasion, whereas high KDM6A gene expression promoted these processes. Second, low expression of KDM6A down-regulates GSK3β, p-GSK3β, up-regulates C-Myc, CyclinD1, and promotes β-catenin protein expression in the nucleus, while the high expression does the opposite. Then, we used ICG001 to block the Wnt/β-catenin signal transduction pathway, and the results revealed that ICG001 could reduce the promoting effect of low KDM6A expression on aggressiveness and EMT in GC cells. KDM6A down-regulation stimulates the proliferation of GC cells, while ICG001 reverses this action in vivo tests. Patients whose KDM6A expression was found to be low had a poor prognosis, as this study found. The EMT is inhibited by regulating theWnt/β-catenin signaling by KDM6A, which reduces GC cell proliferation, migration, and invasion. KDM6A may be a viable target for GC in clinical therapy.
Collapse
Affiliation(s)
- Zhenyou Feng
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Huiming Ding
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Zhiwei Peng
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Kongwang Hu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230032, China; Fuyang Hospital Affiliated to Anhui Medical University, Fuyang 236000, China.
| |
Collapse
|
5
|
Han S, Jin X, Hu T, Chi F. LAPTM5 regulated by FOXP3 promotes the malignant phenotypes of breast cancer through activating the Wnt/β‑catenin pathway. Oncol Rep 2023; 49:60. [PMID: 36799186 PMCID: PMC9942259 DOI: 10.3892/or.2023.8497] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 12/22/2022] [Indexed: 02/12/2023] Open
Abstract
Breast cancer remains the most common malignancy and the leading cause of cancer‑associated mortality in women worldwide. Lysosomal protein transmembrane 5 (LAPTM5), a lysosomal membrane protein, plays an important role in several human malignancies. However, the biological functions and mechanism of LAPTM5 in breast cancer remain unclear. In the present study, the potential tumor‑promoting effect of LAPTM5 was predicted by bioinformatics analysis. LAPTM5 was highly expressed in breast cancer clinical specimens. Moreover, in vitro studies demonstrated that cell proliferation, migration and invasion, as well as the process of epithelial‑mesenchymal transition (EMT) were promoted by LAPTM5 overexpression and were suppressed by LAPTM5 downregulation in vitro. The tumor‑promoting effects of LAPTM5 were also confirmed by xenograft tumor assay in vivo. It was found that the tumor‑promoting effects of LAPTM5 were partly dependent on the activation of the Wnt/β‑catenin signaling pathway. Furthermore, dual‑luciferase and chromatin immunoprecipitation assays verified that the transcription factor forkhead box protein 3 (FOXP3) directly bound to the promoter of LAPTM5 and negatively regulated its expression. Taken together, the present findings indicated that LAPTM5, negatively regulated by FOXP3, promoted the malignant phenotypes of breast cancer through activating the Wnt/β‑catenin signaling pathway.
Collapse
Affiliation(s)
- Sijia Han
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Xueying Jin
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Tianyu Hu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Feng Chi
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China,Correspondence to: Dr Feng Chi, Department of Oncology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, Liaoning 110022, P.R. China, E-mail: and
| |
Collapse
|
6
|
Peng N, Zhang Z, Wang Y, Yang M, Fan J, Wang Q, Deng L, Chen D, Cai Y, Li Q, Wang X, Li W. Down-regulated LINC00115 inhibits prostate cancer cell proliferation and invasion via targeting miR-212-5p/FZD5/Wnt/β-catenin axis. J Cell Mol Med 2021; 25:10627-10637. [PMID: 34697900 PMCID: PMC8581327 DOI: 10.1111/jcmm.17000] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 01/21/2023] Open
Abstract
Prostate cancer is the second most frequent malignancy in men worldwide, and its incidence is increasing. Therefore, it is urgently required to clarify the underlying mechanisms of prostate cancer. Although the long non‐coding RNA LINC00115 was identified as an oncogene in several cancers, the expression and function of LINC00115 in prostate cancer have not been explored. Our results showed that LINC00115 was significantly up‐regulated in prostate cancer tissues, which was significantly associated with a poor prognosis for prostate cancer patients. Functional studies showed that knockdown LINC00115 inhibited cell proliferation and invasion. In addition, LINC00115 served as a competing endogenous RNA (ceRNA) through sponging miR‐212‐5p to release Frizzled Family Receptor 5 (FZD5) expression. The expression of miR‐212‐5p was noticeably low in tumour tissues, and FZD5 expression level was down‐regulated with the knockdown of LINC00115. Knockdown LINC00115 inhibited the Wnt/β‑catenin signalling pathway by inhibiting the expression of FZD5. Rescue experiments further showed that LINC00115 inhibits prostate cancer cell proliferation and invasion via targeting miR‐212‐5p/ FZD5/ Wnt/β‐catenin axis. The present study provided clues that LINC00115 may be a promising novel therapeutic target for prostate cancer patients.
Collapse
Affiliation(s)
- Naixiong Peng
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Zejian Zhang
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Yaomin Wang
- Wake Forest University, Winston-Salem, NC, USA
| | - Minlong Yang
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Jiqing Fan
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Qinjun Wang
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Ling Deng
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Dong Chen
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Yuefeng Cai
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Qihui Li
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Xisheng Wang
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Wei Li
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| |
Collapse
|
7
|
Eskandari M, Mellati AA. Liver X Receptor as a Possible Drug Target for Blood-Brain Barrier Integrity. Adv Pharm Bull 2021; 12:466-475. [PMID: 35935038 PMCID: PMC9348539 DOI: 10.34172/apb.2022.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/13/2021] [Indexed: 12/04/2022] Open
Abstract
Purpose: blood-brain barrier (BBB) is made of specialized cells that are responsible for the selective passage of substances directed to the brain. The integrated BBB is essential for precise controlling of the different substances passage as well as protecting the brain from various damages. In this article, we attempted to explain the role of liver X receptor (LXR) in maintaining BBB integrity as a possible drug target.
Methods: In this study, various databases, including PubMed, Google Scholar, and Scopus were searched using the following keywords: blood-brain barrier, BBB, liver X receptor, and LXR until July, 2020. Additionally, contents close to the subject of our study were surveyed.
Results: LXR is a receptor the roles of which in various diseases have been investigated. LXR can affect maintaining BBB by affecting various ways such as ATP-binding cassette transporter A1 (ABCA1), matrix metalloproteinase-9 (MMP9), insulin-like growth factor 1 (IGF1), nuclear factor-kappa B (NF-κB) signaling, mitogen-activated protein kinase (MAPK), tight junction molecules, both signal transducer and activator of transcription 1 (STAT1), Wnt/β-catenin Signaling, transforming growth factor beta (TGF-β) signaling, and expressions of Smad 2/3 and Snail.
Conclusion: LXR could possibly be used either as a target for drug delivery to brain tissue or as a target for maintaining the BBB integrity in different diseases; thereby the drug will be conducted to tissues, other than the brain. If it is verified that only LXRα is necessary for protecting BBB, some specific LXRα ligands must be found and then used in medication.
Collapse
Affiliation(s)
- Mahsa Eskandari
- Medical school, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Awsat Mellati
- Zanjan Metabolic Disease Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
8
|
Zhang X, Zhang D, Sun X, Li S, Sun Y, Zhai H. Tumor Suppressor Gene XEDAR Promotes Differentiation and Suppresses Proliferation and Migration of Gastric Cancer Cells Through Upregulating the RELA/LXRα Axis and Deactivating the Wnt/β-Catenin Pathway. Cell Transplant 2021; 30:963689721996346. [PMID: 33637015 PMCID: PMC7923976 DOI: 10.1177/0963689721996346] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 01/19/2021] [Accepted: 02/01/2021] [Indexed: 01/06/2023] Open
Abstract
X-linked ectodermal dysplasia receptor (XEDAR) is a new member of the tumor necrosis factor receptor (TNFR) family that induces cell death. The purpose of this study is to determine the tumor-suppressive potential of XEDAR in the development and differentiation of gastric cancer (GC). XEDAR levels were analyzed in human GC tissues and adjacent normal tissues by immunohistochemistry (IHC), quantitative real-time reverse transcription PCR (RT-qPCR), and Western blot analysis. We found that XEDAR expression was significantly downregulated in GC tissues and further decreased in low differentiated GC tissues. Overexpression of XEDAR in MKN45 and MGC803 cells suppressed the ability of cell proliferation and migration, whereas silencing XEDAR showed the opposite effect. Additionally, XEDAR silencing resulted in the upregulation of the differentiation molecular markers β-catenin, CD44 and Cyclin D1 at the protein levels, whereas XEDAR overexpression showed the opposite effect. Notably, XEDAR positively regulated the expression of liver X receptor alpha (LXRα) through upregulating the RELA gene that was characterized as a transcription factor of LXRα in this study. Inhibition of LXRα by GSK2033 or activation of the Wnt/β-catenin pathway by Wnt agonist 1 impaired the effect of XEDAR overexpression on differentiation of MKN45 cells. Moreover, inhibition of RELA mediated by siRNA could promote cell proliferation/migration and rescue the effect of XEDAR overexpression on cell behaviors and expression of genes. Subsequently, overexpression of XEDAR suppressed the growth of GC cells in vivo. Taken together, our findings showed that XEDAR could promote differentiation and suppress proliferation and invasion of GC cells.
Collapse
Affiliation(s)
- Xinwu Zhang
- Department of General Surgery, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Di Zhang
- Department of General Surgery, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaoli Sun
- Department of General Surgery, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Shunle Li
- Department of General Surgery, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yun Sun
- Department of General Surgery, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Hongjun Zhai
- Department of General Surgery, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
9
|
SERPINH1 regulates EMT and gastric cancer metastasis via the Wnt/β-catenin signaling pathway. Aging (Albany NY) 2020; 12:3574-3593. [PMID: 32091407 PMCID: PMC7066881 DOI: 10.18632/aging.102831] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/22/2020] [Indexed: 12/12/2022]
Abstract
In this study, we investigated the role of SERPINH1 in gastric cancer (GC) progression. GC patient tissues show significantly higher SERPINH1 mRNA and protein levels than normal gastric mucosal tissues. GC patients with high SERPINH1 expression are associated with lymph node metastasis and poor prognosis. SERPINH1 mRNA levels negatively correlate with E-cadherin mRNA levels and positively correlate with levels of N-cadherin, MMP2, and MMP9 mRNA levels. This suggests SERPINH1 regulates epithelial to mesenchymal transition (EMT). SERPINH1 expression was significantly higher in the HGC-27, AGS, MGC-803, and SGC-7901 GC cell lines than in the GES-1 normal gastric mucosal cell line. In SERPINH1-silenced SGC-7901 cells, survival, colony formation, migration and invasion were all reduced, whereas they were all enhanced in SERPINH1-overexpressing MGC-803 cells. Levels of WNT/β-catenin signaling pathway proteins, including β-catenin, Wnt2, GSK-3β, p-GSK-3β, NF-κB P65, Snail1, Slug and TWIST, were all reduced in SERPINH1-silenced SGC-7901 cells, and increased in the SERPINH1-overexpressing MGC-803 cells. Inhibition of SERPINH1 protein using Co1003 significantly decreased survival, invasion, and migration of GC cells. SERPINH1 thus appears to regulate EMT and GC progression via the Wnt/β-catenin pathway, making SERPINH1 a potential prognostic biomarker and therapeutic target in GC patients.
Collapse
|