1
|
Li H, Yang Z, Huang J, Lin L, Shi D, Chu Y, Wu D, Cai Y, Li B, Lu J, Guo Q. CALCR interaction with ANTXR1 drives gastric tumor growth and metastasis via AKT signaling pathway. Sci Rep 2025; 15:11826. [PMID: 40195530 PMCID: PMC11977274 DOI: 10.1038/s41598-025-96310-1] [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: 12/18/2024] [Accepted: 03/27/2025] [Indexed: 04/09/2025] Open
Abstract
This study investigates the role of CALCR, a G-protein-coupled receptor, in gastric cancer (GC) progression and its interaction with ANTXR1. A total of 121 patients with gastric cancer were enrolled from the Department of General Surgery, Anyang Tumor Hospital, Anyang City, Henan Province, China. 218 tumor tissues and corresponding para-carcinoma tissues were collected from 109 patients, while adjacent tissues were retained from the remaining 12 cases. Kaplan-Meier analysis evaluated the prognostic value of m6A-related genes in GC. Immunohistochemistry (IHC) was conducted to evaluate CALCR expression. Quantitative real-time PCR (qRT-PCR), Western blot analysis, CCK-8 assays, flow cytometry and transwell assays were used to assess CALCR's role in cell proliferation, apoptosis, migration, and invasion. Co-immunoprecipitation experiments were performed to explore the interaction between CALCR and ANTXR1. Statistical analyses were conducted using SPSS 25.0 and GraphPad Prism 8.0, with p < 0.05 considered significant. IHC staining revealed that 53.2% (n = 58) of the tumor tissues exhibited high CALCR expression, compared to only 6.6% (n = 8) of the para-carcinoma tissues (p < 0.001). CALCR knockdown in GC cell lines significantly reduced proliferation (p < 0.01), increased apoptosis (p < 0.01), and inhibited migration and invasion (p < 0.001). In a nude mouse model, CALCR knockdown resulted in significantly reduced tumor growth and metastasis (p < 0.05). Co-immunoprecipitation showed that CALCR interacts with ANTXR1, leading to decreased AKT phosphorylation. CALCR is a crucial factor in GC progression, presenting a potential prognostic marker and therapeutic target. Targeting the CALCR-ANTXR1 axis and AKT pathway offers new avenues for GC treatment.
Collapse
Affiliation(s)
- Hongbo Li
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zihan Yang
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jingbo Huang
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Lele Lin
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310009, China
| | - Dike Shi
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310009, China
| | - Yiming Chu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310009, China
| | - Dan Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310009, China
| | - Yanna Cai
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Baozhong Li
- Department of General Surgery, Anyang Tumor Hospital, Henan Medical Key Laboratory of Precise Prevention and Treatment of Esophageal Cancer, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, 455000, China
| | - Junyang Lu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Qingqu Guo
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310009, China.
| |
Collapse
|
2
|
Franco LS, Arunachalam S, Chauhan A, Kareff SA, Hallenbeck PL. Elevated expression of ANTXR1 gene in tumors is a poor prognostic biomarker for patients with bladder cancer. Front Mol Biosci 2025; 11:1520223. [PMID: 39917181 PMCID: PMC11798775 DOI: 10.3389/fmolb.2024.1520223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/27/2024] [Indexed: 02/09/2025] Open
Abstract
The TEM8 protein coded by the ANTXR1 gene represents an emerging biomarker in solid tumors. In addition to the various roles TEM8 plays in oncogenesis, including angiogenesis, epithelial-to-mesenchymal transition, and cell migration, it has also been shown that the overexpression of the ANTXR1 gene in solid tumors correlates with poor prognostic indicators in several solid tumor histologies. As such, TEM8 has been identified as the target of novel oncologic therapies. It is especially attractive given its selective expression on the surface of solid tumor cells and associated stromal cells, such as cancer stem cells, invasive cancer cells, and immune cells, such as macrophages, angiogenic endothelial cells, pericytes, and cancer-associated fibroblasts. Furthermore, TEM8 plays this unique role as a mostly non-mutated gene in solid cancers. Here, we demonstrate that elevated expression of ANTXR1 in bladder cancer showed a statistical difference not only in overall survival (OS) but in progression-free survival (PFS), confirming the prognostic biomarker power of ANTXR1 expression.
Collapse
Affiliation(s)
- L. S. Franco
- Seneca Therapeutics, Inc., Blue Bell, PA, United States
| | | | - A. Chauhan
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL, United States
| | - S. A. Kareff
- Lynn Cancer Institute, Boca Raton Regional Hospital, Boca Raton, FL, United States
| | | |
Collapse
|
3
|
Shen X, Xu S, Zheng Z, Liang W, Guo J. The regulatory role of tRNA-derived small RNAs in the prognosis of gastric cancer. Cell Signal 2024; 125:111511. [PMID: 39551416 DOI: 10.1016/j.cellsig.2024.111511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024]
Abstract
In recent years, tRNA-derived small RNAs (tsRNAs) including tRNA-derived stress-induced RNAs (tiRNAs) and tRNA-derived fragments (tRFs), with specific structure and enriched in body fluids, have been found to have specific biological functions. In this paper, the biogenesis, classification, subcellular localization, and biological functions of tsRNAs were summarized. It has been proved that tsRNAs affected tumor cells in proliferation, apoptosis, migration and invasion, and played roles in regulating the occurrence and development of various tumors. In gastric cancer (GC), the imbalance of tsRNAs, such as tRF-33-P4R8YP9LON4VDP, tRF-17-WS7K092, tRF-23-Q99P9P9NDD and others, was closely related to the clinicopathological characteristics of GC patients. Some tsRNAs, such as tRF-23-Q99P9P9NDD, tRF-31-U5YKFN8DYDZDD, and tRF-27-FDXXE6XRK45 promoted the proliferation, migration and invasion of GC cells. Other tsRNAs, such as tRF-41-YDLBRY73W0K5KKOVD, tRF-18-79MP9PO4, and tRF-Glu-TTC-027 inhibited the proliferation, migration and invasion of GC cells. The tsRNAs played roles in the occurrence of GC were through several signaling pathways, such as phosphoinositide 3-kinase (PI3K)-AKT serine/threonine kinase (AKT), Wnt-β-Catenin, and mitogen-activated protein kinase (MAPK) pathways. These findings may provide new strategies for the diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Xiaoban Shen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Shiyi Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Zhinuo Zheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Wei Liang
- Department of Laboratory Medicine, The First Affiliated Hospital of Ningbo University, Ningbo 315211, China.
| | - Junming Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
4
|
Kundu P, Jain R, Kanuri NN, Arimappamagan A, Santosh V, Kondaiah P. DNA Methylation in Recurrent Glioblastomas: Increased TEM8 Expression Activates the Src/PI3K/AKT/GSK-3β/B-Catenin Pathway. Cancer Genomics Proteomics 2024; 21:485-501. [PMID: 39191501 PMCID: PMC11363927 DOI: 10.21873/cgp.20466] [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: 04/01/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND/AIM Glioblastomas (GBM) are infiltrative malignant brain tumors which mostly recur within a year's time following surgical resection and chemo-radiation therapy. Studies on glioblastoma cells following radio-chemotherapy, have been demonstrated to induce trans-differentiation, cellular plasticity, activation of DNA damage response and stemness. As glioblastomas are heterogenous tumors that develop treatment resistance and plasticity, we investigated if there exist genome-wide DNA methylation changes in recurrent tumors. MATERIALS AND METHODS Utilizing genome-wide DNA methylation arrays, we compared the DNA methylation profile of 11 primary (first occurrence) tumors with 13 recurrent (relapsed) GBM, to delineate the contribution of epigenetic changes associated with therapy exposure, therapy resistance, and relapse of disease. RESULTS Our data revealed 1,224 hypermethylated- and 526 hypomethylated-probes in recurrent glioblastomas compared to primary disease. We found differential methylation of solute carrier and ion channel genes, interleukin receptor/ligand genes, tumor-suppressor genes and genes associated with metastasis. We functionally characterized one such hypomethylated-up-regulated gene, namely anthrax toxin receptor 1/tumor endothelial marker 8 (ANTXR1/TEM8), whose expression was validated to be significantly up-regulated in recurrent glioblastomas compared to primary tumors and confirmed by immunohistochemistry. Using overexpression and knockdown approaches, we showed that TEM8 induces proliferation, invasion, migration, and chemo-radioresistance in glioblastoma cells. Additionally, we demonstrated a novel mechanism of β-catenin stabilization and activation of the β-catenin transcriptional program due to TEM8 overexpression via a Src/PI3K/AKT/GSK3β/β-catenin pathway. CONCLUSION We report genome-wide DNA methylation changes in recurrent GBM and suggest involvement of the TEM8 gene in GBM recurrence and progression.
Collapse
Affiliation(s)
- Paramita Kundu
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
- Breast Cancer Now Toby Robins Research Centre, Department of Breast Research, The Institute of Cancer Research, London, U.K
| | - Ruchi Jain
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
- Al Jalila Genomics Centre, Al Jalila Children's Hospital, Dubai, United Arab Emirates
| | - Nandaki Nag Kanuri
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | | | - Vani Santosh
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Paturu Kondaiah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India;
| |
Collapse
|
5
|
Jahantab MB, Salehi M, Koushki M, Farrokhi Yekta R, Amiri-Dashatan N, Rezaei-Tavirani M. Modelling of miRNA-mRNA Network to Identify Gene Signatures with Diagnostic and Prognostic Value in Gastric Cancer: Evidence from In-Silico and In-Vitro Studies. Rep Biochem Mol Biol 2024; 13:281-300. [PMID: 39995653 PMCID: PMC11847593 DOI: 10.61186/rbmb.13.2.281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/08/2024] [Indexed: 02/26/2025]
Abstract
Background Gastric cancer (GC) is a prevalent malignancy with high recurrence. Advances in systems biology have identified molecular pathways and biomarkers. This study focuses on discovering gene and miRNA biomarkers for diagnosing and predicting survival in GC patients. Methods Three sets of genes (GSE19826, GSE81948, and GSE112369) and two sets of miRNA expression (GSE26595, GSE78775) were obtained from the Gene Expression Omnibus (GEO), and subsequently, differentially expressed genes (DEGs) and miRNAs (DEMs) were identified. Functional pathway enrichment, DEG-miR-TF-protein-protein interaction network, DEM-mRNA network, ROC curve, and survival analyses were performed. Finally, qRT-PCR was applied to validate our results. Results From the high-throughput profiling studies of GC, we investigated 10 candidate mRNA and 7 candidate miRNAs as potential biomarkers. Expression analysis of these hubs revealed that 5 miRNAs (including miR-141-3p, miR-204-5p, miR-338-3p, miR-609, and miR-369-5p) were significantly upregulated compared to the controls. The genes with the highest degree included 6 upregulated and 4 downregulated genes in tumor samples compared to controls. The expression of miR-141-3p, miR-204-5p, SESTD1, and ANTXR1 were verified in vitro from these hub DEMs and DEGs. The findings indicated a decrease in the expression of miR-141-3p and miR-204-5p and increased expression of SESTD1 and ANTXR1 in GC cell lines compared to the GES-1 cell line. Conclusions The current investigation successfully recognized a set of prospective miRNAs and genes that may serve as potential biomarkers for GC's early diagnosis and prognosis.
Collapse
Affiliation(s)
- Mohammad Bagher Jahantab
- Clinical Research Development Unit, Shahid Jalil Hospital, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Mohammad Salehi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Mehdi Koushki
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Reyhaneh Farrokhi Yekta
- Proteomics Research Center, School of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Nasrin Amiri-Dashatan
- Proteomics Research Center, School of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Zanjan Metabolic Diseases Research Center, Health and Metabolic Diseases Research Institute, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, School of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Yang L, Liu YN, Gu Y, Guo Q. Deltonin enhances gastric carcinoma cell apoptosis and chemosensitivity to cisplatin via inhibiting PI3K/AKT/mTOR and MAPK signaling. World J Gastrointest Oncol 2023; 15:1739-1755. [PMID: 37969408 PMCID: PMC10631430 DOI: 10.4251/wjgo.v15.i10.1739] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/23/2023] [Accepted: 07/19/2023] [Indexed: 10/10/2023] Open
Abstract
BACKGROUND As an active ingredient derived from Dioscorea zingiberensis C.H. Wright, deltonin has been reported to show anti-cancer effects in a variety of malignancies. AIM To investigate the role and mechanism of action of deltonin in promoting gastric carcinoma (GC) cell apoptosis and chemosensitivity to cisplatin. METHODS The GC cell lines AGS, HGC-27, and MKN-45 were treated with deltonin and then subjected to flow cytometry and 3-(4,5-dimethylthiazol-2-yl)-3,5-diphenyltetrazolium bromide assays for cell apoptosis and viability determination. Western blot analysis was conducted to examine alterations in the expression of apoptosis-related proteins (Bax, Bid, Bad, and Fas), DNA repair-associated proteins (Rad51 and MDM2), and phosphatidylinositol 3-kinase/protein kinase B/mammalian target of the rapamycin (PI3K/AKT/mTOR) and p38-mitogen-activated protein kinase (MAPK) axis proteins. Additionally, the influence of deltonin on GC cell chemosensitivity to cisplatin was evaluated both in vitro and in vivo. RESULTS Deltonin treatment weakened viability, enhanced apoptosis, and dampened DNA repair in GC cell lines in a dose-dependent pattern. Furthermore, deltonin mitigated PI3K, AKT, mTOR, and p38-MAPK phosphorylation. HS-173, an inhibitor of PI3K, attenuated GC cell viability and abolished deltonin inhibition of GC cell viability and PI3K/AKT/mTOR and p38-MAPK pathway activation. Deltonin also promoted the chemosensitivity of GC cells to cisplatin via repressing GC cell proliferation and growth and accelerating apoptosis. CONCLUSION Deltonin can boost the chemosensitivity of GC cells to cisplatin via inactivating p38-MAPK and PI3K/AKT/mTOR signaling.
Collapse
Affiliation(s)
- Lin Yang
- Intensive Care Unit, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Ya-Nan Liu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Yi Gu
- Nursing Department of Obstetrics and Gynecology, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Qi Guo
- Department of Radiotherapy, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
7
|
ANTXR1 as a potential sensor of extracellular mechanical cues. Acta Biomater 2023; 158:80-86. [PMID: 36638946 DOI: 10.1016/j.actbio.2023.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/18/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
Cell adhesion molecules mediate cell-cell or cell-matrix interactions, some of which are mechanical sensors, such as integrins. Emerging evidence indicates that anthrax toxin receptor 1 (ANTXR1), a newly identified cell adhesion molecule, can also sense extracellular mechanical signals such as hydrostatic pressure and extracellular matrix (ECM) rigidity. ANTXR1 can interact with ECM through connecting intracellular cytoskeleton and ECM molecules (just like integrins) to regulate numerous biological processes, such as cell adhesion, cell migration or ECM homeostasis. Although with high structural similarity to integrins, its functions and downstream signal transduction are independent from those of integrins. In this perspective, based on existing evidence in literature, we analyzed the structural and functional evidence that ANTXR1 can act as a potential sensor for extracellular mechanical cues. To our knowledge, this is the first in-depth overview of ANTXR1 from the perspective of mechanobiology. STATEMENT OF SIGNIFICANCE: An overview of ANTXR1 from the perspective of mechanobiology; An analysis of mechanical sensitivity of ANTXR1 in structure and function; A summary of existing evidence of ANTXR1 as a potential mechanosensor.
Collapse
|
8
|
XU JINGYAO, HAO SHUANGLI, HAN KAIYUE, YANG WANXI, DENG HONG. How is the AKT/mTOR pathway involved in cell migration and invasion? BIOCELL 2023. [DOI: 10.32604/biocell.2023.026618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
9
|
Lu J, Zhu D, Zhang X, Wang J, Cao H, Li L. The crucial role of LncRNA MIR210HG involved in the regulation of human cancer and other disease. Clin Transl Oncol 2023; 25:137-150. [PMID: 36088513 DOI: 10.1007/s12094-022-02943-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/30/2022] [Indexed: 01/07/2023]
Abstract
Long noncoding RNAs (lncRNAs) have evoked considerable interest in recent years due to their critical functions in the regulation of disease processes. Abnormal expression of lncRNAs is found in multiple diseases, and lncRNAs have been exploited for diverse medical applications. The lncRNA MIR210HG is a recently discovered lncRNA that is widely dysregulated in human disease. MIR210HG was described to have biological functions with potential roles in disease development, including cell proliferation, invasion, migration, and energy metabolism. And MIR210HG dysregulation was confirmed to have promising clinical values in disease diagnosis, treatment, and prognosis. In this review, we systematically summarize the expression profiles, roles, underlying mechanisms, and clinical applications of MIR210HG in human disease.
Collapse
Affiliation(s)
- Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Shangcheng District, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Danhua Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Shangcheng District, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Xiaoqian Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Shangcheng District, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Jie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Shangcheng District, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Hongcui Cao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Shangcheng District, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Shangcheng District, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
10
|
Luo D, Wang H, Wang Q, Liang W, Liu B, Xue D, Yang Y, Ma B. Senecavirus A as an Oncolytic Virus: Prospects, Challenges and Development Directions. Front Oncol 2022; 12:839536. [PMID: 35371972 PMCID: PMC8968071 DOI: 10.3389/fonc.2022.839536] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Oncolytic viruses have the capacity to selectively kill infected tumor cells and trigger protective immunity. As such, oncolytic virotherapy has become a promising immunotherapy strategy against cancer. A variety of viruses from different families have been proven to have oncolytic potential. Senecavirus A (SVA) was the first picornavirus to be tested in humans for its oncolytic potential and was shown to penetrate solid tumors through the vascular system. SVA displays several properties that make it a suitable model, such as its inability to integrate into human genome DNA and the absence of any viral-encoded oncogenes. In addition, genetic engineering of SVA based on the manipulation of infectious clones facilitates the development of recombinant viruses with improved therapeutic indexes to satisfy the criteria of safety and efficacy regulations. This review summarizes the current knowledge and strategies of genetic engineering for SVA, and addresses the current challenges and future directions of SVA as an oncolytic agent.
Collapse
Affiliation(s)
- Dankun Luo
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haiwei Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Qiang Wang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenping Liang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Liu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongbo Xue
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Yang
- Departments of Biochemistry and Molecular Biology and Oncology, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Biao Ma
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
11
|
Bagaria J, Kim KO, Bagyinszky E, An SSA, Baek JH. Discriminating Potential Genetic Markers for Complete Response and Non-Complete Response Patients to Neoadjuvant Chemotherapy with Locally Advanced Rectal Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19074008. [PMID: 35409691 PMCID: PMC8997875 DOI: 10.3390/ijerph19074008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 02/07/2023]
Abstract
Background: Neoadjuvant chemoradiotherapy (nCRT) prior to surgery is considered standard therapy for locally advanced rectal cancer. Unfortunately, most patients with rectal cancer are resistant to radiotherapy. This might be a genetic cause. The role of certain rectal cancer-causing genes has not been completely elucidated. This study aims to investigate the genes responsible for locally advanced rectal cancer patients not reacting to radiotherapy. Methods: Whole exome sequencing of the DNA samples was performed on the samples. Bioinformatic analysis on the subjects was established. Individual genetic information was screened to identify differently expressed genes that more frequently appeared in non-complete response (NCR) compared to complete response (CR) patients after nCRT. All variations were verified by Sanger sequencing. Results: Genotyping information and pathway analyses of the samples indicated genes such as FLCN, CALML5, and ANTXR1 to be commonly mutated in CR group, whereas genes such as GALNTL14, CNKSR1, ACD, and CUL3 were more commonly mutated in the NCR group. Chi-square test revealed some significant variants (<0.05) such as rs3744124 (FLCN), rs28365986 (ANTXR1), rs10904516 (CALML5), rs3738952 (CUL3), rs13394 and rs2293013 (PIH1D1), rs2274531 (GPA33), rs4963048 (BRSK2), rs17883366 (IL3RA), rs2297575 (PSMD5), rs2288101 (GALNT14), and rs11954652 (DCTN4). Conclusion: Identifying an array of genes that separate NCRs from CRs would lead to finding genetic biomarkers for early detection of rectal cancer patients that are resistant to nCRT. A further investigation to validate the significance of genetic biomarkers to segregate NCRs from CRs should be performed with a larger CRC dataset. Protein expression levels, as well as transcriptomic analysis, would also help us understand the mechanism of how these genes could play a role in preventing radiation therapy to patients. This would be essential to prevent redundant radiation therapy.
Collapse
Affiliation(s)
- Jaya Bagaria
- Department of Bionano Technology, Gachon University, Seongnam-si 13120, Korea; (J.B.); (E.B.)
| | - Kyung-Ok Kim
- Gachon Medical Research Institute, Gil Medical Center, Gachon University, Incheon 21565, Korea;
| | - Eva Bagyinszky
- Department of Bionano Technology, Gachon University, Seongnam-si 13120, Korea; (J.B.); (E.B.)
| | - Seong Soo A. An
- Department of Bionano Technology, Gachon University, Seongnam-si 13120, Korea; (J.B.); (E.B.)
- Correspondence: (S.S.A.A.); (J.-H.B.); Tel.: +82-10-4344-9633 (S.S.A.A.); +82-10-5248-6656 (J.-H.B.)
| | - Jeong-Heum Baek
- Division of Colon and Rectal Surgery, Department of Surgery, Gil Medical Center, College of Medicine, Gachon University, Incheon 21565, Korea
- Correspondence: (S.S.A.A.); (J.-H.B.); Tel.: +82-10-4344-9633 (S.S.A.A.); +82-10-5248-6656 (J.-H.B.)
| |
Collapse
|
12
|
Gao F, Li R, Wei PF, Ou L, Li M, Bai Y, Luo WJ, Fan Z. Synergistic anticancer effects of everolimus (RAD001) and Rhein on gastric cancer cells via phosphoinositide-3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway. Bioengineered 2022; 13:6332-6342. [PMID: 35209807 PMCID: PMC8973710 DOI: 10.1080/21655979.2021.2005988] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/09/2021] [Accepted: 11/09/2021] [Indexed: 01/26/2023] Open
Abstract
Everolimus (RAD001) is a mTOR inhibitor and is widely used for the treatment of gastric cancer (GC). Evidence suggests that Rhein has anticancer effect on GC. But the synergistic effect and mechanism of RAD001 and Rhein combination on GC is not clear. The current study aims to clarify the combination of RAD001 and Rhein in GC treatment. We found Rhein dose-dependently repressed MGC-803 cell viability (50% inhibition concentration (IC50) value = 94.26 μM). Rhein (80 μM) significantly suppressed GC cell proliferation and invasion. RAD001 dose-dependently repressed MGC-803 cells viability (IC50 value = 45.41 nM). The combination of Rhein and RAD001 repressed MGC-803 cells viability, invasion, and proliferation compared to the administration of Rhein or RAD001 alone. Protein levels of epithelial-mesenchymal transition (EMT)-related molecules E-cadherin, N-cadherin and Vimentin expressions were significantly affected by the combination of Rhein and RAD001. The combination of Rhein and RAD001 significantly facilitated cell apoptosis and up-regulated expressions of cell apoptosis and cycle-related protein p53, cyclin-dependent kinase 4 (CDK4) and cyclin D1 compared to the administration of Rhein or RAD001 alone. Moreover, the combination of Rhein and RAD001 repressed the expressions of phosphorylation-phosphoinositide-3-kinase (p-PI3K), p-protein kinase B (p-AKT) and p-mammalian target of rapamycin (p-mTOR). Finally, the combination of RAD001 and Rhein significantly decreased tumor weight and volume, suppressed the expressions of p-PI3K, p-Akt and p-mTOR, and repressed cell proliferation marker Ki-67 expression, which exerted synergistic cancer prevention in GC in vivo. Overall, the combination of Rhein and RAD001 exert synergistic cancer prevention in GC via PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Feng Gao
- Teaching and Research Office of Chinese Pharmacy, The College of Pharmacy of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Rui Li
- Department of Emergency, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Pei-Feng Wei
- The Office of Drug Clinical Trial Institution, The Second Affiliated Hospital of Shaanxi University of traditional Chinese Medicine, Xianyang, Shaanxi, China
| | - Li Ou
- Teaching and Research Office of Chinese Pharmacy, The College of Pharmacy of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Min Li
- Teaching and Research Office of Chinese Pharmacy, The College of Pharmacy of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yang Bai
- The Office of Drug Clinical Trial Institution, The Second Affiliated Hospital of Shaanxi University of traditional Chinese Medicine, Xianyang, Shaanxi, China
| | - Wen-Jia Luo
- Teaching and Research Office of Chinese Pharmacy, The College of Pharmacy of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Zheng Fan
- Department of Gastroenterology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang712000, Shaanxi, China
| |
Collapse
|
13
|
|
14
|
Dong Z, Zhang J, Niu L, Hou G, Gao Z, Yang Q. miR-381-3p Involves in Glioma Progression by Suppressing Tumor-Promoter Factor ANTXR1. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:4883509. [PMID: 34956398 PMCID: PMC8702332 DOI: 10.1155/2021/4883509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/25/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023]
Abstract
Accumulating studies revealed association between development of glioma and miRNA dysregulation. A case in point is miR-381-3p, but its mechanism in glioma is unclear yet. In this work, we confirmed that overexpressed miR-381-3p repressed biological functions of glioma cells. Additionally, we also discovered that upregulated anthrax toxin receptor 1 (ANTXR1) was negatively mediated by miR-381-3p. We further proved that miR-381-3p-targeted ANTXR1 was able to counteract the suppression of miR-381-3p on biological functions of glioma. We concluded that miR-381-3p and ANTXR1 were both important factors in modulating glioma progression. miR-381-3p/ANTXR1 axis is expected to be a molecular target for glioma.
Collapse
Affiliation(s)
- Zhiqiang Dong
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou 730030, China
| | - Jinglong Zhang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou 730030, China
| | - Liang Niu
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou 730030, China
| | - Guokuo Hou
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou 730030, China
| | - Zhenshan Gao
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou 730030, China
| | - Qiang Yang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou 730030, China
| |
Collapse
|
15
|
Si T, Ning X, Chen H, Hu Z, Dun L, Zheng N, Huang P, Yang L, Yi P. ANTXR1 as a potential prognostic biomarker for hepatitis B virus-related hepatocellular carcinoma identified by a weighted gene correlation network analysis. J Gastrointest Oncol 2021; 12:3079-3092. [PMID: 35070431 PMCID: PMC8748048 DOI: 10.21037/jgo-21-764] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/17/2021] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND With high incidence and mortality rates, hepatocellular carcinoma (HCC) is one of the most prevalent malignant tumors worldwide. Chronic hepatitis B virus (HBV) infection is a leading cause of HCC, especially for Asians and blacks. However, the molecular mechanisms underlying HBV-related HCC are unclear. This study sought to identify novel prognostic biomarkers and explore the potential pathogenesis of HBV-related HCC. METHODS The gene expression profiles and corresponding clinical information of HCC from The Cancer Genome Atlas Liver Hepatocellular Carcinoma data set were analyzed by a weighted gene co-expression network analysis. Correlations between the co-expression modules and clinical traits were calculated. Next, key modules associated with HBV infection were identified. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were conducted for the genes in the key modules. The hub genes were identified based on the protein-protein interaction (PPI) network via the Cytoscape. Finally, an overall survival (OS) analysis was performed. RESULTS The two modules (i.e., the brown and yellow modules) most relevant to HBV infection were constructed. A functional enrichment analysis revealed that the genes in the two modules were mainly enriched in HCC-related pathways, such as the phosphatidylinositol-3-kinase and protein kinase B signaling pathway, focal adhesion, human papillomavirus infection, the Rap1 signaling pathway, and the cyclic guanosine monophosphate-dependent protein kinase (cGMP-PKG) signaling pathway. Ten hub genes [i.e., COL3A1, ANTXR1, COL14A1, THBS2, ADAMTS2, AEBP1, PRELP, EMILIN1, DCN and PODN] in the brown module, and 10 hub genes [i.e., USP34, SEC24C, ZNF770, STAG1, TSTD2, PKD1P6, CCNK, GFT2I, NT5C2 and SMG6] in the yellow module were identified. Among the hub genes, ANTXR1 (Anthrax-toxin receptor 1) was significantly correlated with HBV-related HCC patients' OS. CONCLUSIONS ANTXR1 represents a potential therapeutic target for HBV-related HCC. This study offers novel insights into the molecular mechanisms of HBV-induced tumorigenesis, which needs to be further validated by basic experiments and large-scale cohort studies.
Collapse
Affiliation(s)
- Tao Si
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Xuejian Ning
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Haihui Chen
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Zhengguo Hu
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Linglu Dun
- Department of Neurology Laboratory, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Na Zheng
- Department of Neurology Laboratory, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Ping Huang
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Liu Yang
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Ping Yi
- Department of Neurology Laboratory, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| |
Collapse
|
16
|
Ding C, Liu J, Zhang J, Wan Y, Hu L, Charwudzi A, Zhan H, Meng Y, Zheng H, Wang H, Wang Y, Gao L, Hu X, Li J, Xiong S. Tumor Endothelial Marker 8 Promotes Proliferation and Metastasis via the Wnt/β-Catenin Signaling Pathway in Lung Adenocarcinoma. Front Oncol 2021; 11:712371. [PMID: 34722259 PMCID: PMC8552045 DOI: 10.3389/fonc.2021.712371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/28/2021] [Indexed: 01/15/2023] Open
Abstract
Tumor endothelial marker 8 (TEM8), also known as ANTXR1, was highly expressed in cancers, and was identified as a biomarker for early diagnosis and prognosis in some cancers. However, the clinical role and molecular mechanisms of TEM8 in lung adenocarcinoma (LUAD) are still unclear. The present study aimed to explore its clinical value and the molecular mechanisms of TEM8 underlying the progression of LUAD. Our study found the elevation of TEM8 in LUAD cell lines and tissues. What’s more, we observed that the TEM8 expression level was associated with tumor size, primary tumor, and AJCC stage, and LUAD patients with high TEM8 expression usually have a poor prognosis. Then, we conducted a series of experiments by the strategy of loss-of-function and gain-of-function, and our results suggested that the knockdown of TEM8 suppressed proliferation, migration, and invasion and induced apoptosis in LUAD whereas overexpression of TEM8 had the opposite effect. Molecular mechanistic investigation showed that TEM8 exerted its promoting effects mainly through activating the Wnt/β-catenin signaling pathway. In short, our findings suggested that TEM8 played a crucial role in the progression of LUAD by activating the Wnt/β-catenin signaling pathway and could serve as a potential therapeutic target for LUAD.
Collapse
Affiliation(s)
- Chen Ding
- Department of Hematology/Oncology Lab, The Second Hospital of Anhui Medical University, Hefei, China
| | - Jun Liu
- Department of Hematology/Oncology Lab, The Second Hospital of Anhui Medical University, Hefei, China
| | - Jiali Zhang
- Department of Hematology/Oncology Lab, The Second Hospital of Anhui Medical University, Hefei, China
| | - Yang Wan
- Department of Hematology/Oncology Lab, The Second Hospital of Anhui Medical University, Hefei, China
| | - Linhui Hu
- Department of Hematology/Oncology Lab, The Second Hospital of Anhui Medical University, Hefei, China
| | - Alice Charwudzi
- Department of Hematology/Oncology Lab, The Second Hospital of Anhui Medical University, Hefei, China
| | - Heqin Zhan
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Ye Meng
- Department of Hematology/Oncology Lab, The Second Hospital of Anhui Medical University, Hefei, China
| | - Huimin Zheng
- Department of Hematology/Oncology Lab, The Second Hospital of Anhui Medical University, Hefei, China
| | - HuiPing Wang
- Department of Hematology/Oncology Lab, The Second Hospital of Anhui Medical University, Hefei, China
| | - Youliang Wang
- Laboratory of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Lihua Gao
- Laboratory of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Xianwen Hu
- Laboratory of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Jingrong Li
- Department of Emergency, The Second Hospital of Anhui Medical University, Hefei, China
| | - Shudao Xiong
- Department of Hematology/Oncology Lab, The Second Hospital of Anhui Medical University, Hefei, China.,Center of Hematology Research, Anhui Medical University, Hefei, China
| |
Collapse
|
17
|
Li M, Fang L, Kwantwi LB, He G, Luo W, Yang L, Huang Y, Yin S, Cai Y, Ma W, Zhan H, Tong Z, Zhang L, Liang C, Yin Y. N-Myc promotes angiogenesis and therapeutic resistance of prostate cancer by TEM8. Med Oncol 2021; 38:127. [PMID: 34523032 PMCID: PMC8440287 DOI: 10.1007/s12032-021-01575-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/01/2021] [Indexed: 11/24/2022]
Abstract
Although patients with early localized prostate cancer can survive longer, castration-resistant prostate cancer (CRPC) has gradually emerged with the use of androgen deprivation therapy (ADT). N-Myc and TEM8 play a vital role in the progression of several cancer types. However, the underlying mechanism of how N-Myc and TEM8 promote the progression of prostate cancer remains unclear. In this study, the expression of N-Myc and TEM8 was detected in benign prostatic hyperplasia (BPH) and prostate cancer (PCa) tissues by immunohistochemistry (IHC). LNCaP cell lines were maintained in RPMI 1640 medium supplemented with 10% charcoal-stripped fetal bovine serum. Subsequently, R language software was used to verify our results. Tubule formation assay of human umbilical vein endothelial cell (HUVEC) was conducted to examine the effect of N-Myc and TEM8 overexpression on angiogenesis in prostate cancer cells. IHC results showed a positive correlation between the expression of N-Myc and TEM8 in prostate cancer tissues. Further analysis showed that N-Myc and TEM8 were associated with clinicopathological features and poor prognosis in prostate cancer patients. Moreover, the overexpression of N-Myc and TEM8 promoted proliferation of prostate cancer cells and angiogenesis. Additionally, N-Myc and TEM8 overexpression was associated with therapeutic resistance. We further found that N-Myc promoted angiogenesis and therapeutic resistance in prostate cancer via TEM8. Hence, targeting N-Myc/TEM8 pathway in prostate cancer would be a novel therapeutic strategy to enhance the treatment of prostate cancer patients.
Collapse
Affiliation(s)
- Mingfeng Li
- Department of Pathology, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Linna Fang
- Department of Pathology, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Louis Boafo Kwantwi
- Department of Pathology, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Guifang He
- Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China
| | - Wenwu Luo
- Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China
| | - Lijie Yang
- Department of Pathology, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Yuhang Huang
- Department of Pathology, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Shiyuan Yin
- Department of Pathology, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Yongping Cai
- Department of Pathology, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China.,Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China
| | - Wei Ma
- Department of Pathology, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China.,Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China
| | - Heqin Zhan
- Department of Pathology, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China.,Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China
| | - Zhuting Tong
- Department of Radiotherapy, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China
| | - Li Zhang
- Department of Urology, First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, People's Republic of China
| | - Chaozhao Liang
- Department of Urology, First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, People's Republic of China.
| | - Yu Yin
- Department of Pathology, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China. .,Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China.
| |
Collapse
|
18
|
Sun KR, Lv HF, Chen BB, Nie CY, Zhao J, Chen XB. Latest therapeutic target for gastric cancer: Anthrax toxin receptor 1. World J Gastrointest Oncol 2021; 13:216-222. [PMID: 33889273 PMCID: PMC8040068 DOI: 10.4251/wjgo.v13.i4.216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/23/2021] [Accepted: 03/13/2021] [Indexed: 02/06/2023] Open
Abstract
Anthrax toxin receptor 1 (ANTXR1), also known as tumor endothelial marker 8, is a highly conserved cell surface protein overexpressed in tumor-infiltrating vessels. It was first found in vascular endothelial cells of human colorectal cancer. Although our understanding of its physiological function is limited, it has been found that ANTXR1 binds collagen and promotes migration of endothelial cells in vitro. ANTXR1 is upregulated in vessels of different tumor types in mice and humans, and is also expressed by tumor cells themselves in some tumors, such as gastric, lung, intestinal and breast cancer. Developmental angiogenesis and wound healing were not disturbed in ANTXR1 knockout mice, but compared with wild-type mice, growth of melanoma was impaired after ANTXR1 knockout, indicating that host-derived ANTXR1 can promote tumor growth on the basis of immune activity. Previous studies have shown that ANTXR1 vaccines or sublethal doses of anthrax toxin can inhibit angiogenesis, slow tumor growth and prolong survival. These studies suggest that ANTXR1 is necessary for tumor rather than physiological angiogenesis. It has been found that ANTXR1 plays an important role in tumor angiogenesisas well as in the growth and metastasis of many kinds of tumors. This article reviews the physiological function of ANTXR1 and its role in different kinds of cancer.
Collapse
Affiliation(s)
- Ke-Ran Sun
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Hui-Fang Lv
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Bei-Bei Chen
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Cai-Yun Nie
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Jing Zhao
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Xiao-Bing Chen
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450000, Henan Province, China
| |
Collapse
|
19
|
Huang X, Zhang J, Zheng Y. ANTXR1 Is a Prognostic Biomarker and Correlates With Stromal and Immune Cell Infiltration in Gastric Cancer. Front Mol Biosci 2020; 7:598221. [PMID: 33385012 PMCID: PMC7770144 DOI: 10.3389/fmolb.2020.598221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer (GC) is a complex and heterogeneous disease, making it difficult to ascertain the optimal therapeutic approach for individual GC patients. Stromal and immune cell infiltration in GC has a strong correlation with clinical outcomes; however, the underlying mechanisms that drive immunosuppression remain vastly undiscovered. Recent studies validated that anthrax toxin receptor 1 (ANTXR1) is aberrantly expressed in several cancers and holds promise as a new therapeutic target for cancer. However, its immunological roles in GC are still unclear. Here, we show that we identify the distinct stromal and immune cell infiltration in GC between the high and low ANTXR1 expression group by analyzing genomic data. Clinically, ANTXR1 is highly expressed in GC and correlates with adverse clinicopathological characteristics. Additionally, high ANTXR1 expression is linked to markedly poor clinical outcomes and resistance to chemotherapy, whereas the low ANTXR1 expression group is correlated with better outcomes and response to chemotherapy in GC patients. We further revealed the differential landscape of somatic tumor mutation burden (TMB) between the two groups and observed that patients with high ANTXR1 expression suffered from a lower TMB, potentially leading to less sensitivity to checkpoint therapy. Molecularly, results displayed that ANTXR1 is an immunosuppressive element, which may perform its function via promoting the secretion of immunosuppressive factors that play a significant role in modulating tumor-associated fibroblast transformation, M2 macrophage polarization, and T cell exhaustion. Gene set enrichment analysis revealed that cancer-related pathways including epithelial-to-mesenchymal transition, focal adhesion, and transforming growth factor-β (TGF-β) signaling pathways were enriched in high ANTXR1 expression tumors. Our work suggests that ANTXR1 could not only serve as a valuable prognostic biomarker in GC but also be deemed as a potential immunotherapeutic target and useful biomarker of sensitivity to chemotherapy.
Collapse
Affiliation(s)
- Xiaodong Huang
- Department of General Surgery, The Third People's Hospital of Hubei Province, Wuhan, China
| | - Jie Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yongbin Zheng
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
20
|
Amido-Bridged Nucleic Acid-Modified Antisense Oligonucleotides Targeting SYT13 to Treat Peritoneal Metastasis of Gastric Cancer. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 22:791-802. [PMID: 33230476 PMCID: PMC7644579 DOI: 10.1016/j.omtn.2020.10.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023]
Abstract
Patients with peritoneal metastasis of gastric cancer have dismal prognosis, mainly because of inefficient systemic delivery of drugs to peritoneal tumors. We aimed to develop an intraperitoneal treatment strategy using amido-bridged nucleic acid (AmNA)-modified antisense oligonucleotides (ASOs) targeting synaptotagmin XIII (SYT13) and to identify the function of SYT13 in gastric cancer cells. We screened 71 candidate oligonucleotide sequences according to SYT13-knockdown efficacy, in vitro activity, and off-target effects. We evaluated the effects of SYT13 knockdown on cellular functions and signaling pathways, as well as the effects of intraperitoneal administration to mice of AmNA-modified anti-SYT13 ASOs. We selected the ASOs (designated hSYT13-4378 and hSYT13-4733) with the highest knockdown efficiencies and lowest off-target effects and determined their abilities to inhibit cellular functions associated with the metastatic potential of gastric cancer cells. We found that SYT13 interfered with focal adhesion kinase (FAK)-mediated intracellular signals. Intraperitoneal administration of hSYT13-4378 and hSYT13-4733 in a mouse xenograft model of metastasis inhibited the formation of peritoneal nodules and significantly increased survival. Reversible, dose- and sequence-dependent liver damage was induced by ASO treatment without causing abnormal morphological and histological changes in the brain. Intra-abdominal administration of AmNA-modified anti-SYT13 ASOs represents a promising strategy for treating peritoneal metastasis of gastric cancer.
Collapse
|
21
|
Chen Q, Hu L, Chen K. Construction of a Nomogram Based on a Hypoxia-Related lncRNA Signature to Improve the Prediction of Gastric Cancer Prognosis. Front Genet 2020; 11:570325. [PMID: 33193668 PMCID: PMC7641644 DOI: 10.3389/fgene.2020.570325] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/29/2020] [Indexed: 12/24/2022] Open
Abstract
Background Gastric cancer is one of the most common malignant tumors and has a poor prognosis. Hypoxia is related to the poor prognosis of cancer patients. We searched for hypoxia-related long non-coding RNAs (lncRNAs) to predict both overall survival (OS) and disease-free survival (DFS) of gastric cancer patients. Methods We obtained hypoxia-related lncRNA expression profiles and clinical follow-up data of patients with gastric cancer from The Cancer Genome Atlas and the Molecular Signatures Database. The patients were randomly divided into a training group, test group and combined group. The hypoxia-related prognostic signature was constructed by Lasso regression and Cox regression models, the prognoses in different groups were compared by Kaplan-Meier (K-M) analysis, and the accuracy of the prognostic model was assessed by receiver operating characteristic (ROC) analysis. Results A hypoxia-related prognostic signature comprising 10 lncRNAs was constructed to predict both OS and DFS in gastric cancer. In the training, test and combined groups, patients were divided into high- and low-risk groups according to the formula. Kaplan-Meier analysis showed that patients in the high-risk group have poor prognoses, and the difference was significant in the subgroup analyses. Receiver operating characteristic analysis revealed that the predictive power of the model prediction is more accurate than that of standard benchmarks. The signature differed across Helicobacter pylori (Hp) status and T stages. Multivariate Cox analysis showed that the signature is an independent risk factor for both OS and DFS. A clinically predictive nomogram combining the lncRNA signature and clinical features was constructed; the nomogram accurately predicted both OS and DFS and had high clinical application value. Weighted correlation network analysis combined with enrichment analysis showed that the primary pathways were the PI3K-Akt, JAK-STAT, and IL-17 signaling pathways. The target genes NOX4, COL8A1, and CHST1 were associated with poor prognosis in the Gene Expression Profiling Interactive Analysis, Gene Expression Omnibus, and K-M Plotter databases. Conclusions Our 10-lncRNA prognostic signature and nomogram are accurate, reliable tools for predicting both OS and DFS in gastric cancer.
Collapse
Affiliation(s)
- Qian Chen
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Lang Hu
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Kaihua Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
22
|
Lv X, Li P, Wang J, Gao H, Hei Y, Zhang J, Li S. hsa_circ_0000520 influences herceptin resistance in gastric cancer cells through PI3K-Akt signaling pathway. J Clin Lab Anal 2020; 34:e23449. [PMID: 32701211 PMCID: PMC7595902 DOI: 10.1002/jcla.23449] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/22/2022] Open
Abstract
Background To investigate whether hsa_circ_0000520 affects Herceptin resistance in gastric cancer by regulating the PI3K‐AKT signaling. Methods The expression of hsa_circ_0000520 was detected by qRT‐PCR in gastric cancer tissues and cell lines. A Herceptin‐resistant gastric cancer cell was established. PcDNA and pcDNA‐hsa_circ_0000520 were transfected into NCI‐N87R cells and treated with Herceptin at a concentration of 10 μg/mL for 24 hours. MTT tested cell proliferation, and apoptosis was measured by flow cytometry. IGF‐1 treatment was used to activate PI3K‐Akt signaling. The expression levels of related proteins were detected. Results The expression of hsa_circ_0000520 was reduced in gastric cancer tissues and cell lines, and hsa_circ_0000520 in NCI‐N87R cells was significantly lower than that of NCI‐N87 cells. Compared with the CON group, the cell viability of the Herceptin group was significantly reduced, the apoptosis rate was significantly increased, the level of Bax protein was significantly increased, and the levels of Bcl‐2, p‐PI3K, and p‐Akt protein were significantly reduced. Compared with the Herceptin + pcDNA group, the cell viability of the Herceptin + hsa_circ_0000520 group was significantly reduced, the apoptosis rate was significantly increased, the level of Bax protein was significantly increased, and the levels of p‐PI3K and p‐Akt proteins were significantly reduced. After IGF‐1 treatment, the cell viability was significantly increased, the apoptosis rate was significantly reduced, the level of Bax protein was significantly reduced, and the level of Bcl‐2 protein was significantly increased. Conclusion Hsa_circ_0000520 overexpression may reverse the Herceptin resistance of gastric cancer cells by inhibiting the PI3K‐Akt signaling pathway.
Collapse
Affiliation(s)
- Xukun Lv
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Linqing, China
| | - Peizhe Li
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Linqing, China
| | - Jinkai Wang
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Linqing, China
| | - Hengling Gao
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Linqing, China
| | - Yingrui Hei
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Linqing, China
| | - Jianxian Zhang
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Linqing, China
| | - Shuliang Li
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Linqing, China
| |
Collapse
|
23
|
MicroRNAs and Their Influence on the ZEB Family: Mechanistic Aspects and Therapeutic Applications in Cancer Therapy. Biomolecules 2020; 10:biom10071040. [PMID: 32664703 PMCID: PMC7407563 DOI: 10.3390/biom10071040] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 02/07/2023] Open
Abstract
Molecular signaling pathways involved in cancer have been intensively studied due to their crucial role in cancer cell growth and dissemination. Among them, zinc finger E-box binding homeobox-1 (ZEB1) and -2 (ZEB2) are molecules that play vital roles in signaling pathways to ensure the survival of tumor cells, particularly through enhancing cell proliferation, promoting cell migration and invasion, and triggering drug resistance. Importantly, ZEB proteins are regulated by microRNAs (miRs). In this review, we demonstrate the impact that miRs have on cancer therapy, through their targeting of ZEB proteins. MiRs are able to act as onco-suppressor factors and inhibit the malignancy of tumor cells through ZEB1/2 down-regulation. This can lead to an inhibition of epithelial-mesenchymal transition (EMT) mechanism, therefore reducing metastasis. Additionally, miRs are able to inhibit ZEB1/2-mediated drug resistance and immunosuppression. Additionally, we explore the upstream modulators of miRs such as long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), as these regulators can influence the inhibitory effect of miRs on ZEB proteins and cancer progression.
Collapse
|
24
|
Cai C, Dang W, Liu S, Huang L, Li Y, Li G, Yan S, Jiang C, Song X, Hu Y, Gu J. Anthrax toxin receptor 1/tumor endothelial marker 8 promotes gastric cancer progression through activation of the PI3K/AKT/mTOR signaling pathway. Cancer Sci 2020; 111:1132-1145. [PMID: 31977138 PMCID: PMC7156833 DOI: 10.1111/cas.14326] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 12/25/2019] [Accepted: 01/06/2020] [Indexed: 01/18/2023] Open
Abstract
Anthrax toxin receptor 1 (ANTXR1), a type I transmembrane protein, is one of the receptors that facilitates the entrance of anthrax toxin into cells. Previous studies have confirmed the pivotal role of ANTXR1 in progression and tumorigenesis of diverse cancer types. However, the biological function of ANTXR1 in gastric cancer (GC) is still unknown. The present study aimed to investigate the role of ANTXR1 in GC and illuminate the potential molecular mechanisms. Bioinformatics analysis found that ANTXR1 expression was significantly upregulated in GC tissue and its overexpression was associated with poor prognosis of GC patients. Moreover, we confirmed the upregulation of ANTXR1 in GC cell lines and GC tissue by quantitative PCR, western blot analysis, and immunohistochemical analysis. Additionally, high protein expression level of ANTXR1 was positively associated with several clinicopathological parameters in GC patients. In our study, a series of in vitro and in vivo assays were undertaken through strategies of loss/gain-of-function and rescue assays. Consequently, our results indicated that ANTXR1 induced proliferation, cell cycle progression, invasion and migration, and tumorigenicity and induced suppressed apoptosis in GC. Mechanistic investigation indicated that ANTXR1 exerted its promoting effects on GC through activation of the PI3K/AKT/mTOR signaling pathway. In conclusion, our findings suggested that ANTXR1 plays a crucial role in the development and progression of GC and could serve as a novel prognostic biomarker and potential therapeutic target for GC.
Collapse
Affiliation(s)
- Chen Cai
- Department of General SurgeryXinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Biliary Tract Disease ResearchShanghaiChina
| | - Wei Dang
- Department of General SurgeryXinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Biliary Tract Disease ResearchShanghaiChina
| | - Shilei Liu
- Department of General SurgeryXinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Biliary Tract Disease ResearchShanghaiChina
| | - Ling Huang
- Department of General SurgeryXinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Biliary Tract Disease ResearchShanghaiChina
| | - Yang Li
- Department of General SurgeryXinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Biliary Tract Disease ResearchShanghaiChina
| | - Guoqiang Li
- Department of General SurgeryXinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Biliary Tract Disease ResearchShanghaiChina
| | - Siyuan Yan
- Department of General SurgeryXinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Biliary Tract Disease ResearchShanghaiChina
| | - Chengkai Jiang
- Department of General SurgeryXinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Biliary Tract Disease ResearchShanghaiChina
| | - Xiaoling Song
- Department of General SurgeryXinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Biliary Tract Disease ResearchShanghaiChina
| | - Yunping Hu
- Department of General SurgeryXinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Biliary Tract Disease ResearchShanghaiChina
| | - Jun Gu
- Department of General SurgeryXinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Biliary Tract Disease ResearchShanghaiChina
| |
Collapse
|