1
|
Wei Z, Ge D, Bu J, Wang X, Zhong T, Gongye X, Zhang B, Yan F, He C, Guo R, Li J, Jin Z. STC2 regulates the proliferation, migration and glycolysis of glioma cells through modulating ITGB2. Metab Brain Dis 2025; 40:156. [PMID: 40126711 DOI: 10.1007/s11011-025-01571-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/04/2025] [Indexed: 03/26/2025]
Abstract
Glioma is a common and aggressive primary malignant brain tumor. However, the progression mechanism of glioma has not been well revealed. In this study, we intend to detect the function and related mechanism of STC2 in glioma. Gene Expression Profiling Interactive Analysis database was used to detect STC2 and ITGB2 expression in glioma samples, as well as the relationship between STC2 and other genes. The relationship between STC2 and ITGB2 was confirmed by co-immunoprecipitation assay. The biology function of glioma cells was determined by cell counting kit-8, colony formation, transwell, ELISA and western blot assays. We discovered that STC2 was highly expressed in glioma samples and cell lines. Knocked down of STC2 inhibited cell proliferation, invasion, migration and glycolysis. Further analysis demonstrated the interaction between ITGB2 and STC2 as well as its involvement in STC2-regulated proliferation, invasion, migration and glycolysis. In summary, our data afforded novel insights into understanding the regulatory mechanism of STC2 and suggested that the STC2/ITGB2 axis might be a potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Zefeng Wei
- Center for Reproductive Medicine, Affiliated Hospital of Jining Medical University, Jining, 272029, P. R. China
| | - Dengfeng Ge
- Department of Cardiothoracic Surgery, Jiangsu Province Hospital of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
| | - Jie Bu
- Jining Medical University, Jining, 272067, P. R. China
| | - Xuenan Wang
- Center for Reproductive Medicine, Affiliated Hospital of Jining Medical University, Jining, 272029, P. R. China
| | - Tao Zhong
- Jining Medical University, Jining, 272067, P. R. China
| | | | - Bin Zhang
- Department of Cardiothoracic Surgery, Jiangsu Province Hospital of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
| | - Feng Yan
- The People's Hospital of Qufu, Jining, 273100, P. R. China
| | - Chunyan He
- Jining Medical University, Jining, 272067, P. R. China
| | - Runhan Guo
- Jining Medical University, Jining, 272067, P. R. China
| | - Jiayi Li
- Jining Medical University, Jining, 272067, P. R. China
| | - Zhenzhen Jin
- Center for Reproductive Medicine, Affiliated Hospital of Jining Medical University, Jining, 272029, P. R. China.
| |
Collapse
|
2
|
Wen B, Chen J, Ding T, Mao Z, Jin R, Wang Y, Shi M, Zhao L, Yang A, Qin X, Chen X. Development and experimental validation of hypoxia-related gene signatures for osteosarcoma diagnosis and prognosis based on WGCNA and machine learning. Sci Rep 2024; 14:18734. [PMID: 39134603 PMCID: PMC11319349 DOI: 10.1038/s41598-024-69638-3] [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: 01/21/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant tumour of the bone with high mortality. Here, we comprehensively analysed the hypoxia signalling in OS and further constructed novel hypoxia-related gene signatures for OS prediction and prognosis. This study employed Gene Set Enrichment Analysis (GSEA), Weighted correlation network analysis (WGCNA) and Least absolute shrinkage and selection operator (LASSO) analyses to identify Stanniocalcin 2 (STC2) and Transmembrane Protein 45A (TMEM45A) as the diagnostic biomarkers, which further assessed by Receiver Operating Characteristic (ROC), decision curve analysis (DCA), and calibration curves in training and test dataset. Univariate and multivariate Cox regression analyses were used to construct the prognostic model. STC2 and metastasis were devised to forge the OS risk model. The nomogram, risk score, Kaplan Meier plot, ROC, DCA, and calibration curves results certified the excellent performance of the prognostic model. The expression level of STC2 and TMEM45A was validated in external datasets and cell lines. In immune cell infiltration analysis, cancer-associated fibroblasts (CAFs) were significantly higher in the low-risk group. And the immune infiltration of CAFs was negatively associated with the expression of STC2 (P < 0.05). Pan-cancer analysis revealed that the expression level of STC2 was significantly higher in Esophageal carcinoma (ESCA), Head and Neck squamous cell carcinoma (HNSC), Kidney renal clear cell carcinoma (KIRC), Lung squamous cell carcinoma (LUSC), and Stomach adenocarcinoma (STAD). Additionally, the higher expression of STC2 was associated with the poor outcome in those cancers. In summary, this study identified STC2 and TMEM45A as novel markers for the diagnosis and prognosis of osteosarcoma, and STC2 was shown to correlate with immune infiltration of CAFs negatively.
Collapse
Affiliation(s)
- Bo Wen
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
- Department of Orthopedics, No. 945 Hospital of the PLA Joint Logistics Support Force, Yaan, 625000, Sichuan, China
| | - Jian Chen
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Tianqi Ding
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Zhiyou Mao
- Department of Orthopedics, No. 945 Hospital of the PLA Joint Logistics Support Force, Yaan, 625000, Sichuan, China
| | - Rong Jin
- Department of Orthopedics, No. 945 Hospital of the PLA Joint Logistics Support Force, Yaan, 625000, Sichuan, China
| | - Yirui Wang
- Department of Cardiology, No. 945 Hospital of the PLA Joint Logistics Support Force, Yaan, 625000, Sichuan, China
| | - Meiqin Shi
- Department of Orthopedics, No. 945 Hospital of the PLA Joint Logistics Support Force, Yaan, 625000, Sichuan, China
| | - Lixun Zhao
- Department of Orthopedics, No. 945 Hospital of the PLA Joint Logistics Support Force, Yaan, 625000, Sichuan, China
| | - Asang Yang
- Department of Orthopedics, No. 945 Hospital of the PLA Joint Logistics Support Force, Yaan, 625000, Sichuan, China
| | - Xianyun Qin
- Department of Orthopedics, No. 945 Hospital of the PLA Joint Logistics Support Force, Yaan, 625000, Sichuan, China.
| | - Xuewei Chen
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China.
| |
Collapse
|
3
|
Jiang K, Yin X, Zhang Q, Yin J, Tang Q, Xu M, Wu L, Shen Y, Zhou Z, Yu H, Yan S. STC2 activates PRMT5 to induce radioresistance through DNA damage repair and ferroptosis pathways in esophageal squamous cell carcinoma. Redox Biol 2023; 60:102626. [PMID: 36764215 PMCID: PMC9929488 DOI: 10.1016/j.redox.2023.102626] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/01/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Radioresistance is the major reason for the failure of radiotherapy in esophageal squamous cell carcinoma (ESCC). Previous evidence indicated that stanniocalcin 2 (STC2) participates in various biological processes of malignant tumors. However, researches on its effect on radioresistance in cancers are limited. In this study, STC2 was screened out by RNA-sequencing and bioinformatics analyses as a potential prognosis predictor of ESCC radiosensitivity and then was determined to facilitate radioresistance. We found that STC2 expression is increased in ESCC tissues compared to adjacent normal tissues, and a higher level of STC2 is associated with poor prognosis. Also, STC2 mRNA and protein expression levels were higher in radioresistant cells than in their parental cells. Further investigation revealed that STC2 could interact with protein methyltransferase 5 (PRMT5) and activate PRMT5, thus leading to the increased expression of symmetric dimethylation of histone H4 on Arg 3 (H4R3me2s). Mechanistically, STC2 can promote DDR through the homologous recombination and non-homologous end joining pathways by activating PRMT5. Meanwhile, STC2 can participate in SLC7A11-mediated ferroptosis in a PRMT5-dependent manner. Finally, these results were validated through in vivo experiments. These findings uncovered that STC2 might be an attractive therapeutic target to overcome ESCC radioresistance.
Collapse
Affiliation(s)
- Kan Jiang
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University Cancer Center, Zhejiang, 310003, Hangzhou, China
| | - Xin Yin
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University Cancer Center, Zhejiang, 310003, Hangzhou, China
| | - Qingyi Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Yin
- Department of Colorectal Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Qiuying Tang
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University Cancer Center, Zhejiang, 310003, Hangzhou, China
| | - Mengyou Xu
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Lingyun Wu
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University Cancer Center, Zhejiang, 310003, Hangzhou, China
| | - Yifan Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziyang Zhou
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University Cancer Center, Zhejiang, 310003, Hangzhou, China
| | - Hao Yu
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University Cancer Center, Zhejiang, 310003, Hangzhou, China
| | - Senxiang Yan
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University Cancer Center, Zhejiang, 310003, Hangzhou, China.
| |
Collapse
|
4
|
Sengun S, Korkmaz H, Ciris M, Yüceer RO, Boyluboy SM, Kiran M. Diagnostic and prognostic value of Stanniocalcin 1 expression in papillary thyroid cancer. Endocrine 2022; 78:95-103. [PMID: 35788886 DOI: 10.1007/s12020-022-03126-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 06/22/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE To evaluate the potential role of immunohistochemical changes in stanniocalcin 1 (STC1) and stanniocalcin 2 (STC2) expressions in papillary thyroid cancer (PTC) tissues in the disease's diagnosis and to investigate their relationship with classical clinicopathological prognostic factors. METHODS The study included 100 patients with PTC. Normal thyroid tissue adjacent to the tumor was taken as the control group. Clinicopathological prognostic features at the time of diagnosis of patients were recorded. STC1 and STC2 expressions of tumor tissue and adjacent normal tissue were determined immunohistochemically. RESULTS The sensitivity of STC1 in the diagnosis of PTC was 93%, the specificity was 94%, positive predictive value (PPV) 93.9%, and negative predictive value (NPV) 93.1%. It was determined that the STC1 staining score in tumor tissue was positively correlated with the disease TNM stage score (r = 0.259, p = 0.009) and the increase in STC1 staining score were independent risk factors that increased the risk of lymph node metastasis (R2 = 0.398, p < 0.001). While 21% of the tumor tissues were stained with STC2, none of the normal thyroid tissues adjacent to the tumor tissue showed any staining with STC2. No correlation was found between STC2 immunohistochemical staining of tumor tissue and clinicopathological risk factors for the disease. CONCLUSION Increased STC1 expression in thyroid lesions may be helpful in diagnosing PTC. In addition, since increased STC1 expression in PTC tissues is associated with the risk of lymph node metastasis, it may be an efficient marker for predicting the prognosis of the disease.
Collapse
Affiliation(s)
- Sevinç Sengun
- Department of Internal Medicine, Faculty of Medicine, Suleyman Demirel University, 32260, Cunur, Isparta, Turkey
| | - Hakan Korkmaz
- Department of Internal Medicine, Division of Endocrinology, Faculty of Medicine, Suleyman Demirel University, 32260, Cunur, Isparta, Turkey.
| | - Metin Ciris
- Department of Pathology, Faculty of Medicine, Suleyman Demirel University, 32260, Cunur, Isparta, Turkey
| | - Ramazan Oguz Yüceer
- Department of Pathology, Faculty of Medicine, Suleyman Demirel University, 32260, Cunur, Isparta, Turkey
| | - Serife Mehtap Boyluboy
- Department of Internal Medicine, Division of Endocrinology, Faculty of Medicine, Suleyman Demirel University, 32260, Cunur, Isparta, Turkey
| | - Mehmet Kiran
- Department of Pathology, Faculty of Medicine, Suleyman Demirel University, 32260, Cunur, Isparta, Turkey
| |
Collapse
|
5
|
Stanniocalcin 2 (STC2): a universal tumour biomarker and a potential therapeutical target. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:161. [PMID: 35501821 PMCID: PMC9063168 DOI: 10.1186/s13046-022-02370-w] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/19/2022] [Indexed: 12/24/2022]
Abstract
Stanniocalcin 2 (STC2) is a glycoprotein which is expressed in a broad spectrum of tumour cells and tumour tissues derived from human breast, colorectum, stomach, esophagus, prostate, kidney, liver, bone, ovary, lung and so forth. The expression of STC2 is regulated at both transcriptional and post-transcriptional levels; particularly, STC2 is significantly stimulated under various stress conditions like ER stress, hypoxia and nutrient deprivation. Biologically, STC2 facilitates cells dealing with stress conditions and prevents apoptosis. Importantly, STC2 also promotes the development of acquired resistance to chemo- and radio- therapies. In addition, multiple groups have reported that STC2 overexpression promotes cell proliferation, migration and immune response. Therefore, the overexpression of STC2 is positively correlated with tumour growth, invasion, metastasis and patients' prognosis, highlighting its potential as a biomarker and a therapeutic target. This review focuses on discussing the regulation, biological functions and clinical importance of STC2 in human cancers. Future perspectives in this field will also be discussed.
Collapse
|
6
|
Li J, Zhang Z, Feng X, Shen Z, Sun J, Zhang X, Bu F, Xu M, Tan C, Wang Z. Stanniocalcin-2 promotes cell EMT and glycolysis via activating ITGB2/FAK/SOX6 signaling pathway in nasopharyngeal carcinoma. Cell Biol Toxicol 2022; 38:259-272. [PMID: 33797657 PMCID: PMC8986754 DOI: 10.1007/s10565-021-09600-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/03/2021] [Indexed: 12/18/2022]
Abstract
Stanniocalcin-2 (STC2) has been proved to regulate a variety of signaling pathways including cell growth, metastasis, and therapeutic resistance. However, the role of STC2 in the regulation of nasopharyngeal carcinoma (NPC) remains poorly understood. In this study, we investigated the regulatory function of STC2 on epithelial-mesenchymal transition (EMT) and glycolysis traits in NPC and revealed the underlying molecular mechanisms. We found that STC2 was highly expressed in primary nasopharyngeal carcinoma tissues and lymph node metastatic tissues. Silencing of STC2 inhibited cell proliferation, invasion, and glycolysis. Further analyses for the clinical samples demonstrated that STC2 expression was associated with the poor clinical progression. Moreover, we demonstrated the interaction of ITGB2 with STC2 and its involvement in STC2-mediated ITGB2/FAK/SOX6 axis. Collectively, our results provide new insights into understanding the regulatory mechanism of STC2 and suggest that the STC2/ITGB2/FAK/SOX6 signaling axis may be a potential therapeutic target for NPC.
Collapse
Affiliation(s)
- Jingquan Li
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
- Clinical Research Unit of Shanghai municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Shanghai, 200071 China
| | - Zihao Zhang
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032 Anhui China
| | - Xu Feng
- Department of Pathology and Tissue Bank, Fudan University Shanghai Cancer Center, 270 Dong’an Road, Shanghai, 200032 China
| | - Zhuqing Shen
- Department of Pharmacy, Eye & ENT Hospital, Fudan University, Shanghai, 200031 China
| | - Ji Sun
- Department of Pharmacy, Eye & ENT Hospital, Fudan University, Shanghai, 200031 China
| | - Xiuwen Zhang
- Department of Pharmacy, Eye & ENT Hospital, Fudan University, Shanghai, 200031 China
| | - Fengjiao Bu
- Department of Pharmacy, Eye & ENT Hospital, Fudan University, Shanghai, 200031 China
| | - Midie Xu
- Department of Pathology and Tissue Bank, Fudan University Shanghai Cancer Center, 270 Dong’an Road, Shanghai, 200032 China
| | - Cong Tan
- Department of Pathology and Tissue Bank, Fudan University Shanghai Cancer Center, 270 Dong’an Road, Shanghai, 200032 China
| | - Ziliang Wang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
- Clinical Research Unit of Shanghai municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Shanghai, 200071 China
| |
Collapse
|
7
|
OUP accepted manuscript. Carcinogenesis 2022; 43:671-681. [DOI: 10.1093/carcin/bgac030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/15/2022] [Accepted: 03/28/2022] [Indexed: 11/14/2022] Open
|
8
|
Li S, Huang Q, Li D, Lv L, Li Y, Wu Z. The significance of Stanniocalcin 2 in malignancies and mechanisms. Bioengineered 2021; 12:7276-7285. [PMID: 34612765 PMCID: PMC8806499 DOI: 10.1080/21655979.2021.1977551] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Human stanniocalcin 2 (STC2) is an ortholog of fish stanniocalcins (STCs) and is widely expressed in various organs and tissues. The gene is localized on chromosome 5q33 or 5q35. STC2 has been implicated in glucose homeostasis and phosphorus metabolism. It is also reported to be implicated in various malignancies. STC2 was found to be implicated in breast cancer and gynecologic cancers, suggesting hormone-specific or -dependent activities in these malignancies. Moreover, it was reported to be involved in gastrointestinal tumors, including esophageal, gastric, colorectal, and liver cancers, and respiratory cancers, including laryngeal and lung cancers. It also influenced renal carcinoma and prostate cancer. Notably, as a secreted phosphoprotein, STC2 was detectable in serum and possessed promising predictive value in several malignancies. This review aims to improve the understanding of the role of STC2 in patient diagnosis and prognosis, and tumor development and progression, as well as the mechanisms involved.
Collapse
Affiliation(s)
- Shasha Li
- Department of Hepatobiliary Disease, Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Qian Huang
- Department of Hepatobiliary Disease, Fuzong Clinical College, Fujian Medical University, Fuzhou, China
| | - Dongliang Li
- Department of Hepatobiliary Disease, Dongfang Hospital, Xiamen University, Fuzhou, China.,Department of Hepatobiliary Disease, Fuzong Clinical College, Fujian Medical University, Fuzhou, China
| | - Lizhi Lv
- Department of Hepatobiliary Disease, Dongfang Hospital, Xiamen University, Fuzhou, China.,Department of Hepatobiliary Disease, Fuzong Clinical College, Fujian Medical University, Fuzhou, China
| | - Yi Li
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Zhixian Wu
- Department of Hepatobiliary Disease, Dongfang Hospital, Xiamen University, Fuzhou, China.,Department of Hepatobiliary Disease, Fuzong Clinical College, Fujian Medical University, Fuzhou, China
| |
Collapse
|
9
|
Zhang Y, Chen P, Zhou Q, Wang H, Hua Q, Wang J, Zhong H. A Novel Immune-Related Prognostic Signature in Head and Neck Squamous Cell Carcinoma. Front Genet 2021; 12:570336. [PMID: 34220923 PMCID: PMC8249947 DOI: 10.3389/fgene.2021.570336] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 04/20/2021] [Indexed: 02/05/2023] Open
Abstract
The immune response within the tumor microenvironment plays a key role in tumorigenesis and determines the clinical outcomes of head and neck squamous cell carcinoma (HNSCC). However, to date, very limited robust and reliable immunological biomarkers have been developed that are capable of estimating prognosis in HNSCC patients. In this study, we aimed to identify the effects of novel immune-related gene signatures (IRGs) that can predict HNSCC prognosis. Based on gene expression profiles and clinical data of HNSCC patient cohorts from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database, a total of 439 highly variable expressed immune-related genes (including 239 upregulated and 200 downregulated genes) were identified by using differential gene expression analysis. Pathway enrichment analysis indicated that these immune-related differentially expressed genes were enriched in inflammatory functions. After process screening in the training TCGA cohort, six immune-related genes (PLAU, STC2, TNFRSF4, PDGFA, DKK1, and CHGB) were significantly associated with overall survival (OS) based on the LASSO Cox regression model. Integrating these genes with clinicopathological features, a multivariable model was built and suggested better performance in determining patients’ OS in the testing cohort, and the independent validation cohort. In conclusion, a well-established model encompassing both immune-related gene signatures and clinicopathological factors would serve as a promising tool for the prognostic prediction of HNSCC.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Otolaryngology-Head and Neck Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Ping Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qiang Zhou
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hongyan Wang
- Department of Otolaryngology-Head and Neck Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Qingquan Hua
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jie Wang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongliang Zhong
- Department of Neurosurgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Exploration of prognostic index based on immune-related genes in patients with liver hepatocellular carcinoma. Biosci Rep 2021; 40:225490. [PMID: 32579175 PMCID: PMC7327182 DOI: 10.1042/bsr20194240] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 02/08/2023] Open
Abstract
The present study aimed to screen the immune-related genes (IRGs) in patients with liver hepatocellular carcinoma (LIHC) and construct a synthetic index for indicating the prognostic outcomes. The bioinformatic analysis was performed on the data of 374 cancer tissues and 50 normal tissues, which were downloaded from TCGA database. We observed that 17 differentially expressed IRGs were significantly associated with survival in LIHC patients. These LIHC-specific IRGs were validated with function analysis and molecular characteristics. Cox analysis was applied for constructing a RiskScore for predicting the survival. The RiskScore involved six IRGs and corresponding coefficients, which was calculated with the following formula: RiskScore = [Expression level of FABP5 *(0.064)] + [Expression level of TRAF3 * (0.198)] + [Expression level of CSPG5 * (0.416)] + [Expression level of IL17D * (0.197)] + [Expression level of STC2 * (0.036)] + [Expression level of BRD8 * (0.140)]. The RiskScore was positively associated with the poor survival, which was verified with the dataset from ICGC database. Further analysis revealed that the RiskScore was independent of any other clinical feature, while it was linked with the infiltration levels of six types of immune cells. Our study reported the survival-associated IRGs in LIHC and then constructed IRGs-based RiskScore as prognostic indicator for screening patients with high risk of short survival. Both the screened IRGs and IRGs-based RiskScore were clinically significant, which may be informative for promoting the individualized immunotherapy against LIHC.
Collapse
|
11
|
Li J, Yang Z, Huang S, Li D. BIRC7 and STC2 Expression Are Associated With Tumorigenesis and Poor Outcome in Extrahepatic Cholangiocarcinoma. Technol Cancer Res Treat 2020; 19:1533033820971676. [PMID: 33234031 PMCID: PMC7705185 DOI: 10.1177/1533033820971676] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background: Extrahepatic cholangiocarcinoma (EHCC) is a highly aggressive epithelial malignancy and has a poor prognosis for the insensitivity to therapies and difficulty in detection. Novel targets and biomarkers are urgently needed to develop for functional, diagnostic and prognostic application on EHCC. Methods: Immunohistochemical staining technique using the EnVision antibody complex was performed on the samples obtained from 100 EHCC, 30 peritumoral extrahepatic biliary tract (EHBT), 10 EHBT adenomas and 15 normal EHBT tissues. Results: The positive rates of BIRC7 and STC2 expression in tissues obtained from peritumoral EHBT, EHBT adenomas and normal EHBT were significantly lower than those in EHCC tissues. BIRC7 and STC2 proteins were expressed at significantly higher levels in patients with lymph node metastasis, invasion of adjacent tissues, and higher TNM stage (III and/or IV) and unable to undergo resection (biopsy only). Kaplan-Meier survival curves indicated that significantly decreased overall survival rate in patients with positive-BIRC7 or positive-STC2 expression compared with patients of negative-BIRC7 or negative-STC2 expression, respectively. Cox-proportional regression analysis demonstrated that positive-BIRC7 and positive-STC2 expression, along with poor differentiation of EHCC, tumor size >3 cm, lymph node metastasis, invasion of adjacent tissues and unable to undergo resection are independent prognostic factors of EHCC patients. Conclusions: The levels of BIRC7 and STC2 expression were correlated with clinicopathological characteristics of EHCC, and positive expression of BIRC7 and STC2 are associated with progression and poor clinical outcomes of EHCC. BIRC7 and STC2 might be a potential biomarker for EHCC in clinic.
Collapse
Affiliation(s)
- Jiequn Li
- Department of Liver Transplantation, 70566The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhulin Yang
- Department of General Surgery, 70566The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shengfu Huang
- Department of General Surgery, 70566The Second Xiangya Hospital, Central South University, Changsha, China
| | - Daiqiang Li
- Department of Pathology, 70566The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Wu ZL, Deng YJ, Zhang GZ, Ren EH, Yuan WH, Xie QQ. Development of a novel immune-related genes prognostic signature for osteosarcoma. Sci Rep 2020; 10:18402. [PMID: 33110201 PMCID: PMC7591524 DOI: 10.1038/s41598-020-75573-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022] Open
Abstract
Immune-related genes (IRGs) are responsible for osteosarcoma (OS) initiation and development. We aimed to develop an optimal IRGs-based signature to assess of OS prognosis. Sample gene expression profiles and clinical information were downloaded from the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) and Genotype-Tissue Expression (GTEx) databases. IRGs were obtained from the ImmPort database. R software was used to screen differentially expressed IRGs (DEIRGs) and functional correlation analysis. DEIRGs were analyzed by univariate Cox regression and iterative LASSO Cox regression analysis to develop an optimal prognostic signature, and the signature was further verified by independent cohort (GSE39055) and clinical correlation analysis. The analyses yielded 604 DEIRGs and 10 hub IRGs. A prognostic signature consisting of 13 IRGs was constructed, which strikingly correlated with OS overall survival and distant metastasis (p < 0.05, p < 0.01), and clinical subgroup showed that the signature's prognostic ability was independent of clinicopathological factors. Univariate and multivariate Cox regression analyses also supported its prognostic value. In conclusion, we developed an IRGs signature that is a prognostic indicator in OS patients, and the signature might serve as potential prognostic indicator to identify outcome of OS and facilitate personalized management of the high-risk patients.
Collapse
Affiliation(s)
- Zuo-Long Wu
- Guanghe Traditional Chinese and Western Medicine Hospital, Lanzhou, 730000, Gansu, China
- Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Ya-Jun Deng
- Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Guang-Zhi Zhang
- Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - En-Hui Ren
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, No.29 Tongren Road, Xining, 810000, Qinghai, China
- Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Wen-Hua Yuan
- Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Qi-Qi Xie
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, No.29 Tongren Road, Xining, 810000, Qinghai, China.
- Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
13
|
Song W, Ren J, Wang WJ, Wang CT, Fu T. Genome-wide methylation and expression profiling identify a novel epigenetic signature in gastrointestinal pan-adenocarcinomas. Epigenomics 2020; 12:907-920. [PMID: 32166971 DOI: 10.2217/epi-2020-0036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/04/2020] [Indexed: 02/06/2023] Open
Abstract
Aim: To identify methylation-driven genes and establish a novel epigenetic signature for gastrointestinal (GI) pan-adenocarcinomas. Materials & methods: Methylation and RNA-seq data for GI adenocarcinomas were downloaded from the Cancer Genome Atlas database. A methylation-driven gene signature was established by multivariate Cox regression analysis. We developed a prognostic nomogram using a combination of methylation-driven gene risk score and clinicopathological variables. A joint survival analysis based on gene expression and methylation was conducted to further investigate the prognostic role of methylation-driven genes. Results: An epigenetic signature was established based on five methylation-driven genes. We also established a prognostic nomogram based on methylation-driven gene risk score and clinicopathologic factors, with a favorable predictive ability. Joint survival analysis revealed that 28 methylation-driven genes could be independent prognostic factors for overall survival for GI adenocarcinomas. Conclusion: An epigenetic signature was established that effectively predicts the overall survival for GI adenocarcinomas across anatomic boundaries.
Collapse
Affiliation(s)
- Wei Song
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Jun Ren
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Wen-Jie Wang
- Department of Radio-Oncology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215001, China
| | - Chun-Tao Wang
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Tao Fu
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| |
Collapse
|
14
|
Gong S, Xu M, Zhang Y, Shan Y, Zhang H. The Prognostic Signature and Potential Target Genes of Six Long Non-coding RNA in Laryngeal Squamous Cell Carcinoma. Front Genet 2020; 11:413. [PMID: 32411183 PMCID: PMC7198905 DOI: 10.3389/fgene.2020.00413] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/01/2020] [Indexed: 12/11/2022] Open
Abstract
Studies have shown that long non-coding RNA (lncRNA) may act as the carcinogenic factor or tumor suppressor of laryngeal squamous cell carcinoma (LSCC). This study aims to identify the prognostic value and potential target protein-coding genes (PCGs) of lncRNAs in LSCC. The LSCC datasets were collected from The Cancer Genome Atlas (TCGA). Statistical and bioinformatic methods were used to establish and evaluate the prognostic model, identify the correlation between lncRNAs and clinical characteristics, and screen for PCGs co-expressed with lncRNAs. Weighted gene co-expression network analysis (WGCNA) identified PCG modules associated with clinical characteristics. The expression of lncRNAs and PCGs was analyzed using our LSCC patients by RT-qPCR. LINC02154, LINC00528, SPRY4-AS1, TTTY14, LNCSRLR, and KLHL7-DT were selected to establish the prognostic model. The overall survival (OS) of low-risk patients forecasted by the model was significantly better than high-risk patients. Receiver operating characteristic (ROC) curve and concordance index (C-index) validated the accuracy of the prognostic model. Chi-square test showed that six lncRNAs were associated with one of the clinical characteristics, i.e., gender, clinical stage, T and N stage, respectively. WGCNA identified PCG modules associated with gender, clinical stage, T and N stage. We took the intersection of the PCG modules of WGCNA, the differentially expressed PCGs between LSCC and normal samples, and the PCGs co-expressed with six lncRNAs. The intersection PCGs survival analysis showed that four PCGs, i.e., STC2, TSPAN9, SMS, and TCEA3 affected the OS of LSCC. More importantly, the differential expression of six lncRNAs and four PCGs between LSCC and normal samples was verified by our LSCC patients. In conclusion, we successfully established a prognostic model based on six-lncRNA RiskScore and initially screened the potential target PCGs of six lncRNAs for further basic and clinical research.
Collapse
Affiliation(s)
- Shiqi Gong
- Department of Otolaryngology-Head and Neck Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Meng Xu
- Department of Radiation Oncology, The First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Yiyun Zhang
- Department of Otolaryngology-Head and Neck Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yamin Shan
- Department of Otolaryngology-Head and Neck Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Zhang
- Department of Otolaryngology-Head and Neck Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
15
|
Abstract
BACKGROUND Several studies have explored the prognostic value of stanniocalcin 2 (STC2) in various cancers, but obtained inconsistent results. Therefore, this meta-analysis was performed to determine the prognostic and clinicopathologic significance of STC2 in various cancers. METHODS Eligible studies were identified by searching the online databases PubMed, Embase, Web of Science, and the China National Knowledge Infrastructure up to March 2019. Hazard ratios (HRs) with 95% confidence intervals (CIs) and were calculated to clarify the correlation between STC2 expression and prognosis of different cancers. Odds ratios (ORs) with 95% CI were selected to appraise the correlation between STC2 with clinicopathologic characteristics of patients with cancer. RESULTS A total of 16 eligible studies with 4074 patients with cancer were included in our meta-analysis. The results showed that high STC2 expression can predict poor overall survival (OS) for cancer (HR = 1.48, 95% CI: 1.15-1.90, P = .002). Subgroup analysis found that high STC2 expression was associated with worse OS in Asian (HR = 1.85, 95% CI: 1.35-2.55), the reported directly from articles group (HR = 1.39, 95% CI: 1.05-1.84), survival curves group (HR = 1.93, 95% CI: 1.36-2.74), and gastric cancer (HR = 1.43, 95% CI: 1.04-1.95). Furthermore, high STC2 expression was significantly related to advanced T stage (OR = 1.83, 95% CI: 1.17-2.86, P = .008), lymph node metastasis (OR = 2.29, 95% CI: 1.51-3.45, P < .001), lymphatic invasion (OR = 2.15, 95% CI: 1.53-3.02, P < .001), venous invasion (OR = 1.97, 95% CI: 1.30-2.99, P = .001), and more advanced clinical stage (OR = 2.36, 95% CI: 1.74-3.19, P < .001) CONCLUSION:: Elevated expression of STC2 suggested a poor prognosis in patients with cancer and may serve as a new tumor marker to monitor cancer development and progression.
Collapse
Affiliation(s)
- Lixia Hu
- Department of Oncology, The Second People's Hospital of Hefei
| | - Yanyan Zha
- Department of Oncology, The Second People's Hospital of Hefei
| | - Fanliang Kong
- Department of Oncology, The Second People's Hospital of Hefei
| | - Yueyin Pan
- Department of Oncology, Anhui Province Hospital, Hefei, Anhui, China
| |
Collapse
|
16
|
Li Q, Zhou X, Fang Z, Pan Z. Effect of STC2 gene silencing on colorectal cancer cells. Mol Med Rep 2019; 20:977-984. [PMID: 31173256 PMCID: PMC6625197 DOI: 10.3892/mmr.2019.10332] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 03/27/2019] [Indexed: 02/06/2023] Open
Abstract
Stanniocalcin 2 (STC2), a secretory glycoprotein hormone, regulates many biological processes including cell proliferation, apoptosis, tumorigenesis and atherosclerosis. However, the effect of STC2 on proliferation, migration and epithelial‑mesenchymal transition (EMT) progression in human colorectal cancer (CRC) cells remains poorly understood. The expression level of STC2 was determined by quantitative real‑time polymerase chain reaction (qPCR) and western blot analysis. Cell Counting Kit‑8 (CCK‑8) was used to detect the viability of SW480 cells. The invasion and migration of cells were identified by wound healing and Transwell assays. The mRNA and protein expression levels of β‑catenin, matrix metalloproteinase (MMP)‑2, MMP‑9, E‑cadherin and vimentin were assessed by qPCR and western blot analysis. In the present study, it was demonstrated that STC2 was highly expressed in the CRC cell lines. After silencing of STC2, the cell viability, migration and invasion were significantly reduced. Silencing of STC2 in the CRC Sw480 cells increased the expression of E‑cadherin and decreased the expression of vimentin, MMP‑2 and MMP‑9, compared to those in the normal and empty vector group. Furthermore, the expression of β‑catenin in the STC2 gene silenced group was suppressed, and the expression of β‑catenin was reversed by Wnt activator, SB216763. These results demonstrated that STC2 participates in the development and progression of CRC by promoting CRC cell proliferation, survival and migration and activating the Wnt/β‑catenin signaling pathway.
Collapse
Affiliation(s)
- Qianyuan Li
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xiukou Zhou
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Zhengyu Fang
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Zhiyun Pan
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
17
|
Xue K, Li J, Nan S, Zhao X, Xu C. Downregulation of LINC00460 decreases STC2 and promotes autophagy of head and neck squamous cell carcinoma by up-regulating microRNA-206. Life Sci 2019; 231:116459. [DOI: 10.1016/j.lfs.2019.05.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/05/2019] [Accepted: 05/07/2019] [Indexed: 11/26/2022]
|
18
|
He H, Qie S, Guo Q, Chen S, Zou C, Lu T, Su Y, Zong J, Xu H, He D, Xu Y, Chen B, Pan J, Sang N, Lin S. Stanniocalcin 2 (STC2) expression promotes post-radiation survival, migration and invasion of nasopharyngeal carcinoma cells. Cancer Manag Res 2019; 11:6411-6424. [PMID: 31372045 PMCID: PMC6636319 DOI: 10.2147/cmar.s197607] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 06/06/2019] [Indexed: 12/26/2022] Open
Abstract
Background: Stanniocalcin 2 (STC2) expression is upregulated under multiple stress conditions including hypoxia, nutrient starvation and radiation. Overexpression of STC2 correlates with tumor progression and poor prognosis. Purpose: We previously demonstrated that overexpression of STC2 in nasopharyngeal carcinomas (NPC) positively correlates with radiation resistance and tumor metastasis, two major clinical obstacles to the improvement of NPC management. However, it remains elusive whether STC2 expression is a critical contributing factor for post-radiation survival and metastasis of NPC cells. Materials and methods: Using the radiation resistant CNE2 cell line as a model, we examined the importance of STC2 expression for post-radiation survival, migration and invasion. Here, we report the establishment of STC2 knockout lines (CNE2-STC2-KO) using the CRISPR/Cas9-based genome editing technique. Results: Compared with the parental line, STC2-KO cells showed similar proliferation and morphology in normal culture conditions, and loss of STC2 did not compromise the cell tumorigenicity in nude mice model. However, STC2-KO lines demonstrated increased sensitivity to X-radiation under either normoxic or hypoxic conditions. Particularly, upon X-radiation, parental CNE2 cells only slightly whereas STC2-KO cells remarkably decreased the migration and invasion ability. Cell cycle analysis revealed that loss of STC2 accumulated cells in G1 and G2/M phases but decreased S-population. Conclusion: These data indicate that the expression of STC2, which can be stimulated by metabolic or therapeutic stresses, is one important factor to promote survival and metastasis of post-radiation NPC cells. Therefore, targeting STC2 or relative downstream pathways may provide novel strategies to overcome radiation resistance and metastasis of NPC.
Collapse
Affiliation(s)
- Huocong He
- Department of Radiation Biology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Shuo Qie
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia, PA 19104, USA.,Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Qiaojuan Guo
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Shuyang Chen
- Department of Biology, Drexel University College of Arts & Sciences, Philadelphia, PA 19104, USA.,Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Changyan Zou
- Department of Radiation Biology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Tianzhu Lu
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Ying Su
- Department of Radiation Biology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Jingfeng Zong
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Hanchuan Xu
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Dan He
- Department of Biology, Drexel University College of Arts & Sciences, Philadelphia, PA 19104, USA
| | - Yun Xu
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Bijuan Chen
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Jianji Pan
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Nianli Sang
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia, PA 19104, USA.,Department of Biology, Drexel University College of Arts & Sciences, Philadelphia, PA 19104, USA
| | - Shaojun Lin
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| |
Collapse
|
19
|
Zhang C, Chen S, Ma X, Yang Q, Su F, Shu X, Xie W, Feng M, Xiong B. Upregulation of STC2 in colorectal cancer and its clinicopathological significance. Onco Targets Ther 2019; 12:1249-1258. [PMID: 30863092 PMCID: PMC6389002 DOI: 10.2147/ott.s191609] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Stanniocalcin 2 (STC2) is a glycoprotein hormone involved in many biological processes and a secretory protein that regulates malignant tumor progression. The aim of the present study was to further explore the clinicopathological significance and prognostic role of STC2 in colorectal cancer (CRC). Methods In this study, STC2 expression was first investigated in Gene Expression Omnibus and The Cancer Genome Atlas, and then validated with the data from our medical center. Univariate and multivariate analyses were performed to assess the association between prognostic factors and survival outcome. Results In Gene Expression Omnibus and The Cancer Genome Atlas databases, bioinformatics analysis confirmed that STC2 was significantly increased in CRC compared with that in normal tissues (P<0.01), and CRC patients with high STC2 expression had a shorter overall survival. By analyzing data from our medical center, the results also showed that STC2 expression of CRC tissues was higher than that in normal tissues, whether the transcriptional or protein levels. In the CRC tissues, high STC2 expression was significantly correlated with lymph node metastasis (P=0.047), distant metastasis (P=0.040), and advanced clinical stage (P=0.047). Moreover, Kaplan–Meier analyses indicated that high STC2 expression predicted a worse prognosis, and multivariate Cox regression analysis revealed that STC2 was an independent prognostic factor for overall survival (HR =1.976, 95% CI: 1.092–3.576, P=0.024) in patients with CRC. Conclusion Our results suggested that STC2 played an important role in CRC progression and prognosis, and could be a useful biomarker for survival prediction.
Collapse
Affiliation(s)
- Chunxiao Zhang
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| | - Shuangqian Chen
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| | - Xiang Ma
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| | - Qian Yang
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| | - Fei Su
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| | - Xiang Shu
- Department of Technology, Wuhan Hesheng Medical Technological Company, Wuhan 430071, China
| | - Wei Xie
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| | - Maohui Feng
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| | - Bin Xiong
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| |
Collapse
|
20
|
Wang J, Sahengbieke S, Xu X, Zhang L, Xu X, Sun L, Deng Q, Wang D, Chen D, Pan Y, Liu Z, Yu S. Gene expression analyses identify a relationship between stanniocalcin 2 and the malignant behavior of colorectal cancer. Onco Targets Ther 2018; 11:7155-7168. [PMID: 30425508 PMCID: PMC6203107 DOI: 10.2147/ott.s167780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Colorectal cancer (CRC) is one of the main causes of cancer-related death worldwide. Stanniocalcin 2 (STC2), a secreted glycoprotein, has been suggested to exert various functions in progression of many cancers. However, the precise biological role in CRC is not fully understood. Therefore, this study based on several public datasets aims at investigating the roles of STC2 in CRC. Methods We used The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases to evaluate the STC2 expression and its clinical significance in CRC. Cell migration and invasion by STC2 overexpression and knockdown were assessed using Transwell migration and Matrigel invasion assays. We next performed RNAseq analysis on SW480 cells with or without STC2 overexpression. Differentially expressed genes were selected by using fold-change >5 and P-value <0.05. Results In this study, we found that STC2 level was significantly higher in CRC than that in adjacent noncancerous tissues from TCGA and GEO. Tumors with high mRNA levels of STC2 were more common in patients with rectal cancer, left-sided CRC, advanced T-stage (T3-T4), positive lymph node involvement and advanced AJCC-stage (III-IV) from TCGA. STC2 displayed the negative correlation with the expressions of epithelial cell markers, while it was positively correlated with the expressions of mesenchymal cell markers, MMPs and the epithelial-mesenchymal transition (EMT)-related transcriptional factors. Furthermore, we found that STC2 promoted cell migration and invasion in vitro. And a group of differentially expressed genes, which were modulated by STC2, were identified from RNAseq analyses. Conclusion Our study demonstrates that STC2 is overexpressed in CRC compared with normal tissues, and promotes CRC cell migration and invasion. Our data suggest that STC2 may be used as a potential biomarker for clinical application and target therapy in future.
Collapse
Affiliation(s)
- Jian Wang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China,
| | - Sana Sahengbieke
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Xiaoping Xu
- Department of Anorectal Surgery, Yuhang District First People's Hospital, Hangzhou, Zhejiang Province, People's Republic of China
| | - Lei Zhang
- Department of Anorectal Surgery, Yuhang District First People's Hospital, Hangzhou, Zhejiang Province, People's Republic of China
| | - Xiaoming Xu
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Lifeng Sun
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China,
| | - Qun Deng
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China,
| | - Da Wang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China,
| | - Dong Chen
- Department of Anorectal Surgery, Yuhang District First People's Hospital, Hangzhou, Zhejiang Province, People's Republic of China
| | - Yuan Pan
- Department of Anorectal Surgery, Yuhang District First People's Hospital, Hangzhou, Zhejiang Province, People's Republic of China
| | - Zhaohui Liu
- Department of Anorectal Surgery, Yuhang District First People's Hospital, Hangzhou, Zhejiang Province, People's Republic of China
| | - Shaojun Yu
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China,
| |
Collapse
|
21
|
Zhao M, Xie W, Tsai SH, Hein TW, Rocke BA, Kuo L, Rosa RH. Intravitreal Stanniocalcin-1 Enhances New Blood Vessel Growth in a Rat Model of Laser-Induced Choroidal Neovascularization. Invest Ophthalmol Vis Sci 2018; 59:1125-1133. [PMID: 29490350 PMCID: PMC5830987 DOI: 10.1167/iovs.17-23083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Purpose The purpose of this study was to investigate the impact of stanniocalcin-1 (STC-1), a photoreceptor-protective glycoprotein, on the development of choroidal neovascularization (CNV) in relation to VEGF and its main receptor (VEGFR2) expression after laser injury. Methods In rats, CNV was induced by laser photocoagulation in both eyes, followed by intravitreal injection of STC-1 in the right eye and vehicle or denatured STC-1 injection in the left eye as control. Two weeks after laser injury, fundus autofluorescence (FAF) imaging and fundus fluorescein angiography (FFA) were performed. Fluorescein leakage from CNV was graded using a defined scale system. The size of CNV was quantified with spectral domain optical coherence tomography (SD-OCT), fluorescein-labeled choroid-sclera flat mounts, and hematoxylin-eosin staining. Protein expressions were evaluated by Western blot. Results Photocoagulation produced a well-circumscribed area of CNV. With STC-1 treatment, CNV lesions assessed by FAF were increased by 50% in both intensity and area. The CNV lesions were also increased with SD-OCT, flat-mount, and histologic analyses. FFA disclosed enhanced fluorescein leakage in CNV lesions in STC-1 treated eyes. The STC-1 protein was detected in the choroidal tissue and its level was increased with CNV lesions in correlation with VEGF and VEGFR2 expressions. Intravitreal administration of STC-1 significantly increased choroidal expression of both VEGF and VEGFR2 proteins. Conclusions Chorodial tissue expresses STC-1, which seemingly acts as a stress response protein by enhancing pathological new blood vessel growth in laser-induced CNV. It is likely that STC-1 promotes CNV development via VEGF signaling.
Collapse
Affiliation(s)
- Min Zhao
- Ophthalmic Vascular Research Program, Department of Ophthalmology, Scott & White Eye Institute, Temple, Texas, United States.,Department of Medical Physiology, Texas A&M University Health Science Center, Temple, Texas, United States
| | - Wankun Xie
- Ophthalmic Vascular Research Program, Department of Ophthalmology, Scott & White Eye Institute, Temple, Texas, United States.,Department of Medical Physiology, Texas A&M University Health Science Center, Temple, Texas, United States
| | - Shu-Huai Tsai
- Department of Medical Physiology, Texas A&M University Health Science Center, Temple, Texas, United States
| | - Travis W Hein
- Ophthalmic Vascular Research Program, Department of Ophthalmology, Scott & White Eye Institute, Temple, Texas, United States.,Department of Medical Physiology, Texas A&M University Health Science Center, Temple, Texas, United States
| | - Brent A Rocke
- Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, Texas, United States
| | - Lih Kuo
- Ophthalmic Vascular Research Program, Department of Ophthalmology, Scott & White Eye Institute, Temple, Texas, United States.,Department of Medical Physiology, Texas A&M University Health Science Center, Temple, Texas, United States
| | - Robert H Rosa
- Ophthalmic Vascular Research Program, Department of Ophthalmology, Scott & White Eye Institute, Temple, Texas, United States.,Department of Medical Physiology, Texas A&M University Health Science Center, Temple, Texas, United States
| |
Collapse
|
22
|
Brantley KD, Kjærsgaard A, Cronin-Fenton D, Yacoub R, Nielsen AS, Lauridsen KL, Hamilton-Dutoit S, Lash TL. Stanniocalcin Expression as a Predictor of Late Breast Cancer Recurrence. Cancer Epidemiol Biomarkers Prev 2018; 27:653-659. [PMID: 29593009 DOI: 10.1158/1055-9965.epi-17-0905] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/12/2017] [Accepted: 03/20/2018] [Indexed: 11/16/2022] Open
Abstract
Background: Expression of human paracrine hormones stanniocalcin 1 (STC1) and stanniocalcin 2 (STC2) may potentiate late breast cancer recurrence. We tested the hypothesis that expression of STC1 and STC2 in primary breast tumors is more strongly associated with late versus early recurrences.Methods: A total of 541 estrogen receptor-positive, tamoxifen-treated (ER+/TAM+) and 300 ER-negative, tamoxifen-untreated (ER-/TAM-) breast cancer patients who experienced recurrence within 10 years of primary diagnosis and matched recurrence-free controls were selected from a cohort of 11,251 Danish breast cancer patients diagnosed with stage I, II, or III breast cancer during 1985 to 2001. The association between IHC expression of STC1 and STC2 in primary breast tumor tissue microarrays and breast cancer recurrence was evaluated within median time to recurrence quintiles.Results: The association between STC1 expression, dichotomized as positive or negative, and recurrence was strongly positive for the final time quintile (6-10 years postdiagnosis) in the ER+/TAM+ group [aOR = 2.70; 95% confidence interval (CI): 1.22-5.98]. Regression of the log ORs relating dichotomous STC1 and STC2 expression to recurrence by median time to recurrence (year) resulted in a relatively large positive effect estimate for STC1 (β = 0.16; 95% CI, -0.03-0.36) and a near-null positive effect estimate for STC2 (β = 0.04; 95% CI, -0.14-0.21).Conclusions: Our results suggest a stronger association between primary tumor STC1 expression and late recurrence, as opposed to early recurrence, although no clear trend was apparent.Impact: STC1 expression in the primary tumor may potentiate late recurrences, suggesting dormancy pathways that merit further investigation. Cancer Epidemiol Biomarkers Prev; 27(6); 653-9. ©2018 AACR.
Collapse
Affiliation(s)
- Kristen D Brantley
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia.
| | - Anders Kjærsgaard
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Rami Yacoub
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Anja S Nielsen
- Institute of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | - Timothy L Lash
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia.,Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark.,Winship Cancer Institute, Atlanta, Georgia
| |
Collapse
|
23
|
Yang S, Ji Q, Chang B, Wang Y, Zhu Y, Li D, Huang C, Wang Y, Sun G, Zhang L, Guan Q, Xiang J, Wei W, Lu Z, Liao T, Meng J, Wang Z, Ma B, Zhou L, Wang Y, Yang G. STC2 promotes head and neck squamous cell carcinoma metastasis through modulating the PI3K/AKT/Snail signaling. Oncotarget 2018; 8:5976-5991. [PMID: 27863406 PMCID: PMC5351606 DOI: 10.18632/oncotarget.13355] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 10/14/2016] [Indexed: 12/14/2022] Open
Abstract
The mammalian peptide hormone stanniocalcin 2 (STC2) plays an oncogenic role in many human cancers. However, the exact function of STC2 in human head and neck squamous cell carcinoma (HNSCC) is unclear. We aimed to examine the function and clinical significance of STC2 in HNSCC. Using in vitro and in vivo assays, we show that overexpression of STC2 suppressed cell apoptosis, promoted cell proliferation, migration, invasion, and cell cycle arrest at the G1/S transition. By contrast, silencing of STC2 inhibited these activities. We further show that STC2 upregulated the phosphorylation of AKT and enhanced HNSCC metastasis via Snail-mediated increase of vimentin and decrease of E-cadherin. These responses were blocked by silencing of STC2/Snail expression or inhibition of pAKT activity. Furthermore, clinical data indicate that high STC2 expression was associated with high levels of pAKT and Snail in tumor samples from HNSCC patients with regional lymph node metastasis (P < 0.01). Thus, we conclude that STC2 controls HNSCC metastasis via the PI3K/AKT/Snail signaling axis and that targeted therapy against STC2 may be a novel strategy to effectively treat patients with metastatic HNSCC.
Collapse
Affiliation(s)
- Shuwen Yang
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Qinghai Ji
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Bin Chang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yan Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yongxue Zhu
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Duanshu Li
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Caiping Huang
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yulong Wang
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Guohua Sun
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ling Zhang
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Qing Guan
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jun Xiang
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wenjun Wei
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhongwu Lu
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Tian Liao
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Jiao Meng
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Ziliang Wang
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Ben Ma
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Li Zhou
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yu Wang
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Gong Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, China
| |
Collapse
|
24
|
Chen B, Zeng X, He Y, Wang X, Liang Z, Liu J, Zhang P, Zhu H, Xu N, Liang S. STC2 promotes the epithelial-mesenchymal transition of colorectal cancer cells through AKT-ERK signaling pathways. Oncotarget 2018; 7:71400-71416. [PMID: 27662663 PMCID: PMC5342087 DOI: 10.18632/oncotarget.12147] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 09/13/2016] [Indexed: 02/05/2023] Open
Abstract
The STC2 protein involves in carcinogenesis and progression of many cancers. It remains unclear how STC2 regulates the epithelial-mesenchymal transition (EMT) process and colorectal cancer (CRC) development. Here we systematically investigated STC2-activated early occurrence of EMT and CRC cell migration in vitro, clinical associations of STC2 with CRC development and patient survival. The secretion and expression level of STC2 were both greatly increased in EMT cells and CRC cells compared with the normal epithelial NCM460 cells. And the conditioned media from EMT cells stimulated epithelia and colon cancer cells to obtain EMT characteristics. STC2 overexpression promoted CRC cell growth and cell migration in vitro, and STC2 enhanced tumor growth in a mouse CRC-xenograft model. Corresponding to EMT marker expression changes, several critical signaling pathway molecules including pERK, pAKT, PI3K and Ras were remarkably increased either in NCM460 cells transfected with STC2 plasmids or in cells induced with exogenous STC2 protein. However blocking AKT-ERK signaling pathways attenuated STC2-activated EMT process. Furthermore the elevated STC2 expressions were also confirmed in 77 clinical tumor tissues and sera from CRC patients, and the increased STC2 in tumor tissues and sera correlated with tumor pathologic stage and poor survival for CRC patients. In conclusion, STC2 promotes CRC tumorigenesis and EMT progression through activating ERK/MEK and PI3K/AKT signaling pathways. STC2 protein is also a potential tumor biomarker for CRC diagnosis and prognosis.
Collapse
Affiliation(s)
- Bing Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Xiao Zeng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Yu He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Xixi Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Ziwei Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Jingjing Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Peng Zhang
- Department of Urinary Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, P. R. China
| | - Hongxia Zhu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China.,Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, Cancer Institute & Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, 100034, P. R. China
| | - Ningzhi Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China.,Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, Cancer Institute & Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, 100034, P. R. China
| | - Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| |
Collapse
|
25
|
Yokoi K, Yamashita K, Ishii S, Tanaka T, Nishizawa N, Tsutsui A, Miura H, Katoh H, Yamanashi T, Naito M, Sato T, Nakamura T, Watanabe M. Comprehensive molecular exploration identified promoter DNA methylation of the CRBP1 gene as a determinant of radiation sensitivity in rectal cancer. Br J Cancer 2017; 116:1046-1056. [PMID: 28291773 PMCID: PMC5396119 DOI: 10.1038/bjc.2017.65] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/14/2017] [Accepted: 02/20/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Neoadjuvant chemoradiotherapy (NCRT) for advanced rectal cancer (RC) is a well-evidenced therapy; however, some RC patients have no therapeutic response. Patient selection for NCRT so that non-responsive patients are excluded has been subjective. To date, no molecular markers indicating radiation sensitivity have been reported. METHODS We irradiated six colorectal cancer (CRC) cell lines and identified HCT116 cells as radiation-sensitive and HCT15 and DLD-1 cells as radiation resistant. Using a microarray, we selected candidate radiation sensitivity marker genes by choosing genes whose expression was consistent with a radiation-resistant or sensitive cell phenotype. RESULTS Among candidate genes, cellular retinol binding protein 1 (CRBP1) was of particular interest because it was not only induced in HCT116 cells by tentative 10 Gy radiation treatments, but also its expression was increased in HCT116-derived radiation-resistant cells vs parental cells. Forced expression of CRBP1 decreased the viability of both HCT15 and DLD-1 cells in response to radiation therapy. We also confirmed that CRBP1 was epigenetically silenced by hypermethylation of its promoter DNA, and that the quantitative methylation value of CRBP1 significantly correlated with histological response in RC patients with NCRT (P=0.031). CONCLUSIONS Our study identified CRBP1 as a radiation-sensitive predictor in RC.
Collapse
Affiliation(s)
- K Yokoi
- Department of Surgery, Kitasato University School of Medicine, Kitasato, 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - K Yamashita
- Department of Surgery, Kitasato University School of Medicine, Kitasato, 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - S Ishii
- Department of Surgery, Kitasato University School of Medicine, Kitasato, 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - T Tanaka
- Department of Surgery, Kitasato University School of Medicine, Kitasato, 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - N Nishizawa
- Department of Surgery, Kitasato University School of Medicine, Kitasato, 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - A Tsutsui
- Department of Surgery, Kitasato University School of Medicine, Kitasato, 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - H Miura
- Department of Surgery, Kitasato University School of Medicine, Kitasato, 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - H Katoh
- Department of Surgery, Kitasato University School of Medicine, Kitasato, 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - T Yamanashi
- Department of Surgery, Kitasato University School of Medicine, Kitasato, 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - M Naito
- Department of Surgery, Kitasato University School of Medicine, Kitasato, 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - T Sato
- Department of Surgery, Kitasato University School of Medicine, Kitasato, 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - T Nakamura
- Department of Surgery, Kitasato University School of Medicine, Kitasato, 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - M Watanabe
- Department of Surgery, Kitasato University School of Medicine, Kitasato, 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| |
Collapse
|
26
|
Mokarram P, Albokashy M, Zarghooni M, Moosavi MA, Sepehri Z, Chen QM, Hudecki A, Sargazi A, Alizadeh J, Moghadam AR, Hashemi M, Movassagh H, Klonisch T, Owji AA, Łos MJ, Ghavami S. New frontiers in the treatment of colorectal cancer: Autophagy and the unfolded protein response as promising targets. Autophagy 2017; 13:781-819. [PMID: 28358273 PMCID: PMC5446063 DOI: 10.1080/15548627.2017.1290751] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC), despite numerous therapeutic and screening attempts, still remains a major life-threatening malignancy. CRC etiology entails both genetic and environmental factors. Macroautophagy/autophagy and the unfolded protein response (UPR) are fundamental mechanisms involved in the regulation of cellular responses to environmental and genetic stresses. Both pathways are interconnected and regulate cellular responses to apoptotic stimuli. In this review, we address the epidemiology and risk factors of CRC, including genetic mutations leading to the occurrence of the disease. Next, we discuss mutations of genes related to autophagy and the UPR in CRC. Then, we discuss how autophagy and the UPR are involved in the regulation of CRC and how they associate with obesity and inflammatory responses in CRC. Finally, we provide perspectives for the modulation of autophagy and the UPR as new therapeutic options for CRC treatment.
Collapse
Affiliation(s)
- Pooneh Mokarram
- a Colorectal Research Center and Department of Biochemistry , School of Medicine, Shiraz University of Medical Sciences , Shiraz , Iran
| | - Mohammed Albokashy
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | - Maryam Zarghooni
- c Zabol University of Medical Sciences , Zabol , Iran.,d University of Toronto Alumni , Toronto , ON , Canada
| | - Mohammad Amin Moosavi
- e Department of Molecular Medicine , Institute of Medical Biotechnology, National Institute for Genetic Engineering and Biotechnology , Tehran , Iran
| | - Zahra Sepehri
- c Zabol University of Medical Sciences , Zabol , Iran
| | - Qi Min Chen
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | | | | | - Javad Alizadeh
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | - Adel Rezaei Moghadam
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | - Mohammad Hashemi
- g Department of Clinical Biochemistry , School of Medicine, Zahedan University of Medical Sciences , Zahedan , Iran
| | - Hesam Movassagh
- h Department of Immunology , Rady Faculty of Health Sciences, College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | - Thomas Klonisch
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada
| | - Ali Akbar Owji
- i Department of Clinical Biochemistry , School of Medicine, Shiraz Medical University , Shiraz , Iran
| | - Marek J Łos
- j Małopolska Centre of Biotechnology , Jagiellonian University , Krakow , Poland ; LinkoCare Life Sciences AB , Sweden
| | - Saeid Ghavami
- b Department of Human Anatomy and Cell Science , Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba , Winnipeg , MB , Canada.,k Health Policy Research Center , Shiraz University of Medical Sciences , Shiraz , Iran
| |
Collapse
|
27
|
Fjeldbo CS, Aarnes EK, Malinen E, Kristensen GB, Lyng H. Identification and Validation of Reference Genes for RT-qPCR Studies of Hypoxia in Squamous Cervical Cancer Patients. PLoS One 2016; 11:e0156259. [PMID: 27244197 PMCID: PMC4887009 DOI: 10.1371/journal.pone.0156259] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 05/11/2016] [Indexed: 12/26/2022] Open
Abstract
Hypoxia is an adverse factor in cervical cancer, and hypoxia-related gene expression could be a powerful biomarker for identifying the aggressive hypoxic tumors. Reverse transcription quantitative PCR (RT-qPCR) is a valuable method for gene expression studies, but suitable reference genes for data normalization that are independent of hypoxia status and clinical parameters of cervical tumors are lacking. In the present work, we aimed to identify reference genes for RT-qPCR studies of hypoxia in squamous cervical cancer. From 422 candidate reference genes selected from the literature, we used Illumina array-based expression profiles to identify 182 genes not affected by hypoxia in cervical cancer, i.e. genes regulated by hypoxia in eight cervical cancer cell lines or correlating with the hypoxia-associated dynamic contrast-enhanced magnetic resonance imaging parameter ABrix in 42 patients, were excluded. Among the 182 genes, nine candidates (CHCHD1, GNB2L1, IPO8, LASP1, RPL27A, RPS12, SOD1, SRSF9, TMBIM6) that were not associated with tumor volume, stage, lymph node involvement or disease progression in array data of 150 patients, were selected for further testing by RT-qPCR. geNorm and NormFinder analyses of RT-qPCR data of 74 patients identified CHCHD1, SRSF9 and TMBIM6 as the optimal set of reference genes, with stable expression both overall and across patient subgroups with different hypoxia status (ABrix) and clinical parameters. The suitability of the three reference genes were validated in studies of the hypoxia-induced genes DDIT3, ERO1A, and STC2. After normalization, the RT-qPCR data of these genes showed a significant correlation with Illumina expression (P<0.001, n = 74) and ABrix (P<0.05, n = 32), and the STC2 data were associated with clinical outcome, in accordance with the Illumina data. Thus, CHCHD1, SRSF9 and TMBIM6 seem to be suitable reference genes for studying hypoxia-related gene expression in squamous cervical cancer samples by RT-qPCR. Moreover, STC2 is a promising prognostic hypoxia biomarker in cervical cancer.
Collapse
Affiliation(s)
- Christina S. Fjeldbo
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Eva-Katrine Aarnes
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Eirik Malinen
- Department of Medical Physics, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | - Gunnar B. Kristensen
- Department of Gynaecologic Oncology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Heidi Lyng
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- * E-mail:
| |
Collapse
|
28
|
Yuan Q, Zhan L, Zhang LL, Wang Q, Liu J, Jiang ZY, Hu XM, Yuan XC. Stanniocalcin 2 induces oxaliplatin resistance in colorectal cancer cells by upregulating P-glycoprotein. Can J Physiol Pharmacol 2016; 94:929-35. [PMID: 27245421 DOI: 10.1139/cjpp-2015-0530] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multidrug resistance (MDR) limits the anticancer effects of chemotherapy in patients with metastatic colorectal cancer (CRC). Oxaliplatin is a common component of combinational therapeutic regimens administered to patients with metastatic CRC; however, it is also used as a constituent of adjuvant therapy for patients at a risk of recurrent disease. In the present study, we investigated the role of stanniocalcin 2 (STC2) in chemoresistance. STC2 knockdown sensitized chemoresistant CRC cells to oxaliplatin. Moreover, the expression of exogenous STC2 in chemonaïve CRC cells induced oxaliplatin resistance. We confirmed that STC2 upregulated P-glycoprotein (P-gp) expression in CRC cells. Furthermore, shRNA against phosphoinositide 3-kinase (PI3K) or Akt inhibited the action of STC2 on P-gp upregulation and MDR in CRC. To our knowledge, this is the first report to demonstrate the induction of oxaliplatin resistance in CRC cells in response to STC2 stimulation of P-gp via the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Qiong Yuan
- a Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China.,c Drug Research Base of Cardiovascular and Cerebral Vascular, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Lin Zhan
- a Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Li-Li Zhang
- b Department of Histology and Embryology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Qiang Wang
- d Department of Immunology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Juan Liu
- a Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Zhen-Yu Jiang
- a Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Xia-Min Hu
- a Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China.,c Drug Research Base of Cardiovascular and Cerebral Vascular, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Xin-Chu Yuan
- b Department of Histology and Embryology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| |
Collapse
|
29
|
Bagordakis E, Sawazaki-Calone I, Macedo CCS, Carnielli CM, de Oliveira CE, Rodrigues PC, Rangel ALCA, Dos Santos JN, Risteli J, Graner E, Salo T, Paes Leme AF, Coletta RD. Secretome profiling of oral squamous cell carcinoma-associated fibroblasts reveals organization and disassembly of extracellular matrix and collagen metabolic process signatures. Tumour Biol 2016; 37:9045-57. [PMID: 26762409 DOI: 10.1007/s13277-015-4629-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 12/10/2015] [Indexed: 01/23/2023] Open
Abstract
An important role has been attributed to cancer-associated fibroblasts (CAFs) in the tumorigenesis of oral squamous cell carcinoma (OSCC), the most common tumor of the oral cavity. Previous studies demonstrated that CAF-secreted molecules promote the proliferation and invasion of OSCC cells, inducing a more aggressive phenotype. In this study, we searched for differences in the secretome of CAFs and normal oral fibroblasts (NOF) using mass spectrometry-based proteomics and biological network analysis. Comparison of the secretome profiles revealed that upregulated proteins involved mainly in extracellular matrix organization and disassembly and collagen metabolism. Among the upregulated proteins were fibronectin type III domain-containing 1 (FNDC1), serpin peptidase inhibitor type 1 (SERPINE1), and stanniocalcin 2 (STC2), the upregulation of which was validated by quantitative PCR and ELISA in an independent set of CAF cell lines. The transition of transforming growth factor beta 1 (TGF-β1)-mediating NOFs into CAFs was accompanied by significant upregulation of FNDC1, SERPINE1, and STC2, confirming the participation of these proteins in the CAF-derived secretome. Type I collagen, the main constituent of the connective tissue, was also associated with several upregulated biological processes. The immunoexpression of type I collagen N-terminal propeptide (PINP) was significantly correlated in vivo with CAFs in the tumor front and was associated with significantly shortened survival of OSCC patients. Presence of CAFs in the tumor stroma was also an independent prognostic factor for OSCC disease-free survival. These results demonstrate the value of secretome profiling for evaluating the role of CAFs in the tumor microenvironment and identify potential novel therapeutic targets such as FNDC1, SERPINE1, and STC2. Furthermore, type I collagen expression by CAFs, represented by PINP levels, may be a prognostic marker of OSCC outcome.
Collapse
Affiliation(s)
- Elizabete Bagordakis
- Department of Oral Diagnosis, School of Dentistry, State University of Campinas, Av. Limeira 901, CEP 13414-018, Piracicaba, SP, Brazil
| | - Iris Sawazaki-Calone
- Oral Pathology and Oral Medicine, Dentistry School, Western Paraná State University, Cascavel, PR, Brazil
| | - Carolina Carneiro Soares Macedo
- Department of Oral Diagnosis, School of Dentistry, State University of Campinas, Av. Limeira 901, CEP 13414-018, Piracicaba, SP, Brazil
| | - Carolina M Carnielli
- Brazilian Biociences National Laboratory-CNPEM, CEP 13083-970, Campinas, SP, Brazil
| | - Carine Ervolino de Oliveira
- Department of Oral Diagnosis, School of Dentistry, State University of Campinas, Av. Limeira 901, CEP 13414-018, Piracicaba, SP, Brazil
| | - Priscila Campioni Rodrigues
- Department of Oral Diagnosis, School of Dentistry, State University of Campinas, Av. Limeira 901, CEP 13414-018, Piracicaba, SP, Brazil
| | - Ana Lucia C A Rangel
- Oral Pathology and Oral Medicine, Dentistry School, Western Paraná State University, Cascavel, PR, Brazil
| | - Jean Nunes Dos Santos
- Laboratory of Surgical Pathology, Dental School, Federal University of Bahia-UFBA, Salvador, BA, Brazil
| | - Juha Risteli
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital, Oulu, Finland
| | - Edgard Graner
- Department of Oral Diagnosis, School of Dentistry, State University of Campinas, Av. Limeira 901, CEP 13414-018, Piracicaba, SP, Brazil
| | - Tuula Salo
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital, Oulu, Finland.,Oral and Maxillofacial Diseases Unit, University of Helsinki, Helsinki, Finland.,Helsinki University Hospital, Helsinki, Finland
| | | | - Ricardo D Coletta
- Department of Oral Diagnosis, School of Dentistry, State University of Campinas, Av. Limeira 901, CEP 13414-018, Piracicaba, SP, Brazil.
| |
Collapse
|
30
|
Wang Y, Gao Y, Cheng H, Yang G, Tan W. Stanniocalcin 2 promotes cell proliferation and cisplatin resistance in cervical cancer. Biochem Biophys Res Commun 2015; 466:362-8. [PMID: 26361149 DOI: 10.1016/j.bbrc.2015.09.029] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 09/05/2015] [Indexed: 12/14/2022]
Abstract
Cervical cancer is one of the most common carcinomas in the female reproductive system. Treatment of cervical cancer involves surgical removal and chemotherapy. Resistance to platinum-based chemotherapy drugs including cisplatin has increasingly become an important problem in the treatment of cervical cancer patients. We found in this study that stanniocalcin 2 (STC2) expression was upregulated in both cervical cancer tissues and cell lines. The levels of STC2 expression in cervical cancer cell lines were positively correlated with the rate of cell proliferation. Furthermore, in cisplatin resistant cervical cancer cells, the levels of STC2 expression were significantly elevated. Modulation of STC2 expression by siRNA or overexpression in cisplatin resistant cells resulted in altered cell survival, apoptosis, and cisplatin resistance. Finally, we found that there was significant difference in the activity of the MAPK signaling pathway between cisplatin sensitive and resistant cervical cancer cells, and that STC2 could regulate the activity of the MAPK signaling pathway.
Collapse
Affiliation(s)
- Yuxia Wang
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Ying Gao
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Hairong Cheng
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Guichun Yang
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Wenhua Tan
- Department of Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, China.
| |
Collapse
|
31
|
Wu J, Lai M, Shao C, Wang J, Wei JJ. STC2 overexpression mediated by HMGA2 is a biomarker for aggressiveness of high-grade serous ovarian cancer. Oncol Rep 2015; 34:1494-502. [PMID: 26165228 DOI: 10.3892/or.2015.4120] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 06/17/2015] [Indexed: 12/14/2022] Open
Abstract
High-grade serous cancer (HGSC) is a lethal form of ovarian cancer due to invasion and early metastasis. Gain of epithelial-mesenchymal transition (EMT) contributes to the aggressiveness of HGSC. High-mobility gene group A2 (HMGA2), an architectural transcription factor, plays a major role in HGSC through the regulation of EMT gene expression. Based on the gene profiling analysis, we found that the potent EMT gene, stanniocalcin 2 (STC2), was highly correlated with HMGA2 expression. In the present study, we demonstrated that STC2 was directly regulated by HMGA2 at the transcriptional level. Overexpressing STC2 in vitro directly enhanced cell migration and invasion. To investigate the correlation of STC2 and HMGA2 expression and the potential biomarker for ovarian cancer, three independent large cohorts of ovarian cancer (cohort 1=278, cohort 2=150 and cohort 3=95 cases) were examined in the present study. The results showed that the expression of HMGA2 and STC2 was positively correlated. Furthermore, STC2 expression was significantly associated with tumor grade and histotype. HGSC had significantly higher levels of STC2 expression and was inversely correlated with patient survival. These findings suggested that STC2 is an important new biomarker that can be used for HGSC.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| | - Maode Lai
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| | - Changshun Shao
- Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA
| | - Jian Wang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Jian-Jun Wei
- Department of Pathology, Northwestern University School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
32
|
Hou J, Wang Z, Xu H, Yang L, Yu X, Yang Z, Deng Y, Meng J, Feng Y, Guo X, Yang G. Stanniocalicin 2 suppresses breast cancer cell migration and invasion via the PKC/claudin-1-mediated signaling. PLoS One 2015; 10:e0122179. [PMID: 25830567 PMCID: PMC4382185 DOI: 10.1371/journal.pone.0122179] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 02/08/2015] [Indexed: 12/14/2022] Open
Abstract
Stanniocalcin (STC), a glycoprotein hormone, is expressed in a wide variety of tissues to regulate Ca2+ and PO4- homeostasis. STC2, a member of STC family, has been reported to be associated with tumor development. In this study, we investigated whether the expression of STC2 is associated with migration and invasion of breast cancer cells. We found that breast cancer cell line 231 HM transfected with STC2 shRNA displayed high motility, fibroblast morphology, and enhanced cell migration and invasion. Introduction of STC2 in 231 cells reduced cell migration and invasion. In response to irradiation, silencing of STC2 in 231 HM cells reduced apoptosis, whereas overexpression of STC2 in 231 cells promoted apoptosis, compared with in control cells. Mechanistic study showed that STC2 negatively regulated PKC to control the expression of Claudin-1, which subsequently induced the expressions of EMT-related factors including ZEB1, ZO-1, Slug, Twist, and MMP9. Suppression of PKC activity by using a PKC inhibitor (Go 6983) restored the normal motility of STC2-silenced cells. Furthermore, in vivo animal assay showed that STC2 inhibited tumorigenesis and metastasis of breast cancer cells. Collectively, these results indicate that STC2 may inhibit EMT at least partially through the PKC/Claudin-1-mediated signaling in human breast cancer cells. Thus, STC2 may be exploited as a biomarker for metastasis and targeted therapy in human breast cancer.
Collapse
Affiliation(s)
- Jing Hou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Cancer Institute, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ziliang Wang
- Cancer Institute, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Han Xu
- Breast surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lina Yang
- Cancer Institute, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoli Yu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhaozhi Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yun Deng
- Cancer Institute, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiao Meng
- Cancer Institute, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Feng
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaomao Guo
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Gong Yang
- Cancer Institute, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| |
Collapse
|
33
|
Fang Z, Tian Z, Luo K, Song H, Yi J. Clinical significance of stanniocalcin expression in tissue and serum of gastric cancer patients. Chin J Cancer Res 2014; 26:602-10. [PMID: 25400427 DOI: 10.3978/j.issn.1000-9604.2014.10.08] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 09/25/2014] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Stanniocalcin (STC) has been recognized as a potential biomarker in a variety of cancers. The aim of this study was to examine STC1 and STC2 expression in tumor and serum samples from gastric cancer (GC) patients. METHODS A total of 83 GC patients treated with radical resection were enrolled in this study. Immunohistochemistry was used to detect STC protein expression in paired tumor and adjacent normal tissues. Serum STC levels were determined by enzyme-linked immunosorbent assay (ELISA). The receiver operating characteristics (ROC) curve was constructed to describe diagnostic specificity and sensitivity. RESULTS Both of STC1 and STC2 protein expression were upregulated in GC tissues compared with that in normal ones. Moreover, the high/moderate of STC1 protein was significantly associated with lymph metastasis, clinical stage and adverse 3-year progression-free survival (PFS). In addition, serum STC1 and STC2 expression in GC patients were much higher than that in patients with benign gastric disease, which decreased at postoperative 7-10 days. The sensitivity of serum STC protein also showed superiority over CEA and CA19-9. CONCLUSIONS STC upregulation plays an important role in GC development, and serum STC1 and STC2 might function as promising tumor markers for GC diagnosis and prognosis.
Collapse
Affiliation(s)
- Zheng Fang
- 1 Department of General Surgery, 101 Hospital of People's Liberation Army, Wuxi 214044, China ; 2 Department of Medical Oncology, 3 Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Zhiqiang Tian
- 1 Department of General Surgery, 101 Hospital of People's Liberation Army, Wuxi 214044, China ; 2 Department of Medical Oncology, 3 Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Kunlun Luo
- 1 Department of General Surgery, 101 Hospital of People's Liberation Army, Wuxi 214044, China ; 2 Department of Medical Oncology, 3 Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Haizhu Song
- 1 Department of General Surgery, 101 Hospital of People's Liberation Army, Wuxi 214044, China ; 2 Department of Medical Oncology, 3 Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Jun Yi
- 1 Department of General Surgery, 101 Hospital of People's Liberation Army, Wuxi 214044, China ; 2 Department of Medical Oncology, 3 Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| |
Collapse
|
34
|
Na SS, Aldonza MB, Sung HJ, Kim YI, Son YS, Cho S, Cho JY. Stanniocalcin-2 (STC2): A potential lung cancer biomarker promotes lung cancer metastasis and progression. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1854:668-76. [PMID: 25463045 DOI: 10.1016/j.bbapap.2014.11.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 10/29/2014] [Accepted: 11/05/2014] [Indexed: 12/14/2022]
Abstract
The homodimeric glycoprotein, stanniocalcin 2 (STC2) is previously known to be involved in the regulation of calcium and phosphate transport in the kidney and also reported to play multiple roles in several cancers. However, its function and clinical significance in lung cancer have never been reported and still remain uncertain. Here, we investigated the possibility of STC2 as a lung cancer biomarker and identified its potential role in lung cancer cell growth, metastasis and progression. Proteomic analysis of secretome of primary cultured lung cancer cells revealed higher expression of STC2 in cancers compared to that of adjacent normal cells. RT-PCR and Western blot analyses showed higher mRNA and protein expressions of STC2 in lung cancer tissues compared to the adjacent normal tissues. Knockdown of STC2 in H460 lung cancer cells slowed down cell growth progression and colony formation. Further analysis revealed suppression of migration, invasion and delayed G0/G1 cell cycle progression in the STC2 knockdown cells. STC2 knockdown also attenuated the H202-induced oxidative stress on H460 cell viability with a subsequent increase in intracellular ROS levels, which suggest a protective role of STC2 in redox regulatory system of lung cancer. These findings suggest that STC2 can be a potential lung cancer biomarker and plays a positive role in lung cancer metastasis and progression. This article is part of a Special Issue entitled: Medical Proteomics.
Collapse
Affiliation(s)
- Sang-su Na
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Republic of Korea; Department of Physical Therapy, College of Rehabilitation, Daegu University, Daegu, Republic of Korea
| | - Mark Borris Aldonza
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Hye-Jin Sung
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Yong-In Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Yeon Sung Son
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Sukki Cho
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seoungnam-si, Gyeonggi-do, Republic of Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Republic of Korea.
| |
Collapse
|
35
|
ZHANG ZHENHAI, WU YAGUANG, QIN CHENGKUN, RONG ZHONGHOU, SU ZHONGXUE, XIAN GUOZHE. Stanniocalcin 2 expression predicts poor prognosis of hepatocellular carcinoma. Oncol Lett 2014; 8:2160-2164. [PMID: 25289096 PMCID: PMC4186577 DOI: 10.3892/ol.2014.2520] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 08/12/2014] [Indexed: 12/14/2022] Open
Abstract
Previous studies have shown that the expression level of stanniocalcin 2 (STC2) is associated with tumor progression. However, to date, the association between STC2 and clinicopathological factors in hepatocellular carcinoma (HCC) has not been investigated. The clinical significance of STC2 was investigated in 30 fresh HCC samples using western blot analysis and in 240 HCC tissues using immunohistochemical analysis. The level of STC2 in cancerous tissue was higher than in the matched non-cancerous tissues. Using immunohistochemistry, the STC2-positive group exhibited a higher incidence of lymph node metastasis and venous invasion compared with the STC2-negative group. Kaplan-Meier survival analysis revealed that the positive expression of STC2 correlated with poor overall survival (OS) and disease-free survival of HCC patients (P<0.01). STC2 expression was observed to be an independent prognostic factor for OS in HCC patients by multivariate analysis (hazard ratio, 2.39; 95% confidence interval, 1.04-5.89; P=0.013). These data suggest that STC2 expression may be a useful indicator of poor prognosis in HCC patients.
Collapse
Affiliation(s)
- ZHEN-HAI ZHANG
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - YA-GUANG WU
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - CHENG-KUN QIN
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - ZHONG-HOU RONG
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - ZHONG-XUE SU
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - GUO-ZHE XIAN
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
36
|
Clinical utility of measuring expression levels of Stanniocalcin 2 in patients with colorectal cancer. Med Oncol 2014; 31:237. [PMID: 25234931 DOI: 10.1007/s12032-014-0237-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 09/04/2014] [Indexed: 12/11/2022]
|
37
|
Expression of stanniocalcin-1 and stanniocalcin-2 in laryngeal squamous cell carcinoma and correlations with clinical and pathological parameters. PLoS One 2014; 9:e95466. [PMID: 24743310 PMCID: PMC3990672 DOI: 10.1371/journal.pone.0095466] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 03/27/2014] [Indexed: 12/14/2022] Open
Abstract
Background Stanniocalcin-1 (STC1) and stanniocalcin-2 (STC2) are secreted glycoprotein hormones involved in various types of human malignancies. The roles of STC1 and STC2 in laryngeal squamous cell carcinoma (LSCC) remain unknown. We investigated correlations between STC1 and STC2 expression and clinicopathological or prognostic factors in LSCC. Methods Pre-surgical peripheral blood samples were collected between 2012 and 2013 from 62 patients with LSCC. Quantitative RT-PCR analysis was performed to examine mRNA levels of STC1 and STC2. Immunohistochemistry was performed to retrospectively analyze 90 paraffin-embedded LSCC tissue samples, which were obtained from patients who received surgery between 2006 and 2009. These patients did not have histories of treatment or malignancies. Univariate analysis of patient survival was performed by the Kaplan–Meier method. Multivariate analyses were performed with the Cox proportional hazards model. Results The relative mRNA levels of STC1 and STC2 in peripheral blood were significantly greater in LSCC patients than those of healthy volunteers (both P<0.05). STC2 protein expression in tumor tissues was associated with invasion into the thyroid cartilage, T-Stage, lymphatic metastasis, clinical stage, and pathological differentiation (all P<0.05). In addition, STC2 protein expression was an independent prognostic factor for overall survival in patients with LSCC (P = 0.025). In contrast, STC1 expression only correlated with clinical stage (P = 0.026) and was not an independent or significant prognostic factor. Conclusions Circulating STC1 and STC2 mRNA are potentially useful blood markers for LSCC. Our results strongly suggest that the STC2 protein, but not STC1, may be a valuable biomarker for LSCC malignancies and a prognostic marker for poor outcome following surgery. Future studies should examine STC2 as a novel molecular target for the treatment of LSCC.
Collapse
|
38
|
Survival analyses correlate stanniocalcin 2 overexpression to poor prognosis of nasopharyngeal carcinomas. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:26. [PMID: 24606961 PMCID: PMC4015363 DOI: 10.1186/1756-9966-33-26] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 03/03/2014] [Indexed: 12/14/2022]
Abstract
Background Stanniocalcin 2 (STC2) is overexpressed in several types of human cancers, and its overexpression positively correlates to tumor progression and poor prognosis. However, the clinical significance of STC2 overexpression in nasopharyngeal carcinomas (NPC) has not been investigated. This study examined STC2 expression in a cohort of 94 NPC samples, and explored its value in clinical diagnosis and prognosis. Methods Tumor samples from 94 patients diagnosed in 2008 were studied. All samples were obtained prior to treatment start. All cases were clinically diagnosed and pathologically confirmed to be poorly differentiated or undifferentiated NPC without distant metastasis, and have been treated with radical radiation therapy and followed-up for five years. Survival analyses were performed. Results Of the 94 NPC samples, STC2 overexpression (STC2+) was detected in 65 samples (69.1%). Overall survival rate of STC2 (+) patients is significantly lower than that of patients with normal STC2 levels (72.2% vs. 96.4%, respectively, P = 0.049). Moreover, STC2 (+) is also strongly predictive of a low progression-free survival and distant metastasis-free survival (63.0% vs 92.9%. P = 0.007; and 77.0% vs 96.4%. P = 0.028). Of the 54 patients treated with IMRT, residual tumors were found in 54.8% of STC2 positive patients (17/31), but only in 17.4% of STC2 negative ones (4/23), suggesting STC2 overexpression predicts a higher risk of residual tumors after IMRT. Conclusions STC2 overexpression correlates to poor prognosis for NPC and may be useful as a novel biomarker to predict NPC responses to radiation. Whether STC2 promotes NPC progression and metastasis remains to be investigated.
Collapse
|
39
|
Miyazaki S, Kikuchi H, Iino I, Uehara T, Setoguchi T, Fujita T, Hiramatsu Y, Ohta M, Kamiya K, Kitagawa K, Kitagawa M, Baba S, Konno H. Anti-VEGF antibody therapy induces tumor hypoxia and stanniocalcin 2 expression and potentiates growth of human colon cancer xenografts. Int J Cancer 2014; 135:295-307. [PMID: 24375080 DOI: 10.1002/ijc.28686] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 11/20/2013] [Accepted: 12/10/2013] [Indexed: 01/24/2023]
Abstract
Tumor angiogenesis plays a critical role in colorectal cancer progression. Recent randomized clinical trials have revealed the additive effect of bevacizumab, a humanized monoclonal antibody against vascular endothelial growth factor (VEGF)-A, to conventional chemotherapy in the improved survival of patients with metastatic colorectal cancer. However, a number of preclinical reports indicate the development of resistance to anti-angiogenic therapy. In this study, we addressed the effects of anti-VEGF antibodies on the growth and malignant behavior of colorectal cancer cells. TK-4, a solid tumor strain derived from a colon cancer patient, was subcutaneously or orthotopically implanted into nude mice. Short-term administration of anti-VEGF antibodies inhibited the growth of cecal tumors at day 14 by suppressing mitosis, but prolonged treatment resulted in the recovery of cellular proliferation and suppression of apoptosis at day 35. Intratumoral hypoxia induced by anti-VEGF antibody treatment resulted in activation of hypoxia inducible factor-1α protein and an increased number of aldehyde dehydrogenase 1-positive tumor cells. In microarray analysis, stanniocalcin 2 (STC2) was the most highly upregulated gene in anti-VEGF antibody-treated tumors. In vitro analyses showed that the growth and migration of SW480 colon cancer cells under hypoxic conditions were significantly inhibited by knockdown of STC2. In vivo serial transplantation of TK-4 revealed that long-term administration of anti-VEGF antibodies increased the tumorigenicity of colon cancers and accelerated tumor growth when transplanted into secondary recipient mice. Our data provide a potential molecular explanation for the limited clinical effectiveness of anti-VEGF antibodies.
Collapse
Affiliation(s)
- Shinichiro Miyazaki
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Arigami T, Uenosono Y, Ishigami S, Yanagita S, Hagihara T, Haraguchi N, Matsushita D, Hirahara T, Okumura H, Uchikado Y, Nakajo A, Hokita S, Natsugoe S. Clinical significance of stanniocalcin 2 expression as a predictor of tumor progression in gastric cancer. Oncol Rep 2013; 30:2838-44. [PMID: 24100594 DOI: 10.3892/or.2013.2775] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 09/02/2013] [Indexed: 11/06/2022] Open
Abstract
Stanniocalcin 2 (STC2) is a glycoprotein hormone that plays an important role in calcium and phosphate homeostasis. Furthermore, recent studies have demonstrated that STC2 expression in the primary site is correlated with tumor progression in several types of malignancies. However, few reports have investigated the clinical significance of STC2 expression in the blood of patients with gastric cancer. Therefore, we examined STC2 expression as a molecular blood marker for detection of circulating tumor cells (CTCs) and assessed the relationship between STC2 expression and clinico-pathological features including prognosis in patients with gastric cancer. Quantitative PCR assay was used to assess STC2 mRNA expression in 4 gastric cancer cell lines and in blood specimens from 93 patients with gastric cancer and 22 healthy volunteers. The numbers of STC2 mRNA copies were significantly higher in the gastric cancer cell lines and in blood from patients with gastric cancer than in blood from healthy volunteers (P=0.0002 and P=0.01, respectively). STC2 expression was positive in 43 (46.2%) of the 93 patients with gastric cancer, and its expression was significantly correlated with age, depth of tumor invasion, lymph node metastasis, stage and venous invasion (P=0.023, P=0.045, P=0.035, P=0.007 and P=0.027, respectively). The 5-year survival rate was significantly lower in patients with STC2 expression compared to patients without STC2 expression (P=0.014). Our results indicate that STC2 could be a useful molecular blood marker for predicting tumor progression by monitoring CTCs in patients with gastric cancer.
Collapse
Affiliation(s)
- Takaaki Arigami
- Department of Digestive Surgery, Breast and Thyroid Surgery, Field of Oncology, Course of Advanced Therapeutics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Law AYS, Wong CKC. Stanniocalcin-1 and -2 promote angiogenic sprouting in HUVECs via VEGF/VEGFR2 and angiopoietin signaling pathways. Mol Cell Endocrinol 2013; 374:73-81. [PMID: 23664860 DOI: 10.1016/j.mce.2013.04.024] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 03/27/2013] [Accepted: 04/26/2013] [Indexed: 12/15/2022]
Abstract
The members of stanniocalcins (STCs: STC-1 and STC-2) family are known to be involved in tumor progression and metastasis. Although current evidences suggest the involvement of STCs in vascular biology, the functional roles of STCs in angiogenesis have not yet been elucidated. The objective of this study was to decipher the roles of STCs in angiogenesis of human umbilical vascular endothelial cells (HUVECs). We prepared STC1 or STC2 lentiviral particles to transduce the cells to reveal their effects on the processes of cell proliferation, migration and tube formation. The stimulatory effects of STCs on these processes were demonstrated, supporting the notion of STCs in angiogenesis. To dissect the molecular components involved, STC1 or STC2 transduction led to significant increases in the expression levels of cell cycle regulators (i.e. cyclin-D and phospho-retinoblastoma), matrix metalloproteinase (MMP)-2 but a decrease of tissue inhibitors of metalloproteases (TIMP)-1. The expression levels of the cell adhesion/junctional proteins vimentin and VE-cadherin, were significantly induced. Moreover the transduction induced both mRNA and protein levels of eNOS, VEGF and VEGFR2 (KDR mRNA and pKDR), highlighting the stimulatory effects of STCs on VEGF-signaling pathway. Furthermore STC2 transduction but not STC1, activated angiopoietin (Ang)-2 pathway. Taken together, STC1 and STC2 play positive roles in angiogenic sprouting. The action of STC1 was mediated via VEGF/VEGFR2 pathway while STC2 were mediated via VEGF/VEGFR2 and Ang-2 pathways.
Collapse
Affiliation(s)
- Alice Y S Law
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | | |
Collapse
|
42
|
Wang YY, Li L, Zhao ZS, Wang HJ. Clinical utility of measuring expression levels of KAP1, TIMP1 and STC2 in peripheral blood of patients with gastric cancer. World J Surg Oncol 2013; 11:81. [PMID: 23548070 PMCID: PMC3620905 DOI: 10.1186/1477-7819-11-81] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 03/09/2013] [Indexed: 12/18/2022] Open
Abstract
Background We examined preoperative kinesin II-associated protein (KAP1), TIMP metallopeptidase inhibitor 1 (TIMP1) and stanniocalcin 2 (STC2) expression levels in patients with gastric cancers to assess their clinical application for diagnosing and monitoring diseases. Methods Real-time reverse transcription-polymerase chain reaction was used to detect the expression levels of KAP1, TIMP1, STC2, talin 2 (TLN2), sushi-repeat-containing protein, X-linked 2 (SRPX2) and secreted protein, acidic, cysteine-rich (SPARC) in the patients’ peripheral blood karyocytes. The data were analyzed with receiver operating characteristics (ROC) curves. Results A total of 112 patients with gastric cancer, 42 patients with recurrence and 107 healthy volunteers were recruited. There were significant correlations between KAP1, TIMP1 and STC2 levels, and TNM tumor stages and distant metastases. The area under the ROC curves (AUC) of KAP1 was 0.803 ± 0.040 (P = 0.0001), the AUC of TIMP1 was 0.767 ± 0.043 (P = 0.0001) and the AUC of STC2 was 0.769 ± 0.045 (P = 0.0001), thus differentiating preoperative gastric cancer patients from healthy volunteers by ROC curve analysis. The AUC of STC2 was 0.739 ± 0.070 (P = 0.004) and the AUC of KAP1 was 0.418 ± 0.088 (P = 0.319), thus differentiating recurrence of gastric cancer from healthy volunteers by ROC curve analysis. High TIMP1 and STC2 expression levels were suspected to be poor prognostic factors of disease recurrence in patients with gastric cancer. Conclusions KAP1, TIMP1 and STC2 expression levels may be potential biomarkers for the screening, diagnosis, prognosis and surveillance of gastric cancer.
Collapse
Affiliation(s)
- Yuan-Yu Wang
- Department of Pathology, Zhejiang Provincial People's Hospital, 158 shangtang road, Hangzhou, Zhejiang, 310014, PR China
| | | | | | | |
Collapse
|
43
|
Jiang J, Westberg JA, Andersson LC. Stanniocalcin 2, forms a complex with heme oxygenase 1, binds hemin and is a heat shock protein. Biochem Biophys Res Commun 2012; 421:274-9. [PMID: 22503972 DOI: 10.1016/j.bbrc.2012.03.151] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 03/30/2012] [Indexed: 11/25/2022]
Abstract
Stanniocalcin 2 (STC2) is a homolog of stanniocalcin 1, a 56kD glycoprotein hormone that originally was found to confer calcitonin-like activity in fish. Human STC2 is expressed in various tissues such as kidney, spleen, heart, and pancreas. STC2 has been demonstrated to be induced by different kinds of stress and display cytoprotective activity, but the molecular mechanism is poorly understood. Heme oxygenase 1 (HO1) degrades heme to biliverdin, carbon monoxide and free iron, and is a stress-responsive protein. Using yeast two-hybrid screening we identified HO1 as a binding partner of STC2. The interaction was validated by in vivo co-immunoprecipitation and immunofluorescence. The binding site for HO1 was located to amino acids 181-200 of STC2. We also found that STC2 binds hemin via a consensus heme regulatory motif. Moreover, STC2 expression was induced by heat shock in HEK293 cells. Taken together, our findings point to three novel functions of STC2, and suggest that STC2 interacts with HO1 to form a eukaryotic 'stressosome' involved in the degradation of heme.
Collapse
Affiliation(s)
- Ji Jiang
- Department of Pathology, Haartman Institute, University of Helsinki and HUSLAB, PO Box 21, Haartmaninkatu 3, FI-00014 Helsinki, Finland.
| | | | | |
Collapse
|
44
|
Yeung BHY, Law AYS, Wong CKC. Evolution and roles of stanniocalcin. Mol Cell Endocrinol 2012; 349:272-80. [PMID: 22115958 DOI: 10.1016/j.mce.2011.11.007] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 11/07/2011] [Indexed: 12/11/2022]
Abstract
In fish, stanniocalcin-1 (STC1) is a key endocrine factor that acts on gill, intestine and kidney to regulate serum calcium and phosphate homeostasis. The recent identification and study of mammalian STCs (STC1 and STC2) revealed that the hormones are made in virtually all tissues and they act primarily as paracrine/autocrine factors to regulate various biological functions. Based on their ubiquitous expression patterns and generally undetectable levels in blood serum, it is unlikely that the mammalian STCs play important roles in serum Ca(2+)/P(i) homeostasis. However current evidences still support the local action of STCs in Ca(2+) and P(i) transport, probably via their action on Ca(2+)-channels and Na(+)/P(i) co-transporter. At present, information about the sequence, expression and distribution of the STC receptor(s) is lacking. However, recent emerging evidence hints the involvement of STC1 and STC2 in the sub-cellular functions of mitochondria and endoplasmic reticulum respectively, particularly responding to oxidative stress and unfolded protein response. With increasing evidence that demonstrates the local actions of STCs, the focus of the research has been moved to cellular inflammation and carcinogenesis. This review integrates the information available on STCs in fish and mammals, focusing mainly on their embryonic origin, tissue distribution, their potential regulatory mechanisms and the modes of action, and their physiological and pathophysiological functions, particularly in cancer biology.
Collapse
Affiliation(s)
- B H Y Yeung
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | | | | |
Collapse
|
45
|
Akagi T, Hijiya N, Inomata M, Shiraishi N, Moriyama M, Kitano S. Visinin-like protein-1 overexpression is an indicator of lymph node metastasis and poor prognosis in colorectal cancer patients. Int J Cancer 2011; 131:1307-17. [PMID: 22052372 DOI: 10.1002/ijc.27341] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 10/26/2011] [Indexed: 12/16/2022]
Abstract
Lymph node metastasis is an important factor determining outcome from colorectal cancer (CRC). Identification of molecular markers useful to predict lymph node metastasis is urgently needed. Our objective was to identify genes useful for characterization and prediction of lymph node metastasis in CRC. Gene expression profiles of cancer were determined by human U133 Plus 2.0 GeneChip® in 24 CRC patients, and patients with and without lymph node metastasis were compared. We focused on the visinin-like protein-1 (VSNL-1) gene and evaluated VSNL-1 mRNA expression levels with reverse transcriptase-polymerase chain reaction and immunohistochemical methods. Immunohistochemical evaluation of VSNL-1 mRNA expression was also performed in 143 other CRC patients to determine clinicopathological significance of VSNL-1. Twenty-four novel discriminating genes showed expression significantly different between patients with and without lymph node metastasis. Mean level of VSNL-1 mRNA expression in tumor tissue with lymph node metastasis was significantly higher than that in tissue without lymph node metastasis. Immunohistochemical examination demonstrated immunoreactivity for VSNL-1 in cytoplasm of the cancer cells with lymph node metastasis. High VSNL-1 expression was significantly associated with lymphatic invasion in stage II disease (p = 0.0061) and number of lymph node metastases in stage III disease (p = 0.0461). Patients with high VSNL-1 expression had significantly poorer prognosis than those with low expression in stage III disease (p = 0.045). This study is the first to demonstrate a prognostic role for VSNL-1 at the mRNA level, suggesting the possible usefulness of VSNL-1 as a predictor of lymph node metastasis and poor prognosis in CRC.
Collapse
Affiliation(s)
- Tomonori Akagi
- Department of Surgery I, Oita University Faculty of Medicine, Hasama-machi, Oita, Japan.
| | | | | | | | | | | |
Collapse
|
46
|
Dondeti VR, Wubbenhorst B, Lal P, Gordan JD, D'Andrea K, Attiyeh EF, Simon MC, Nathanson KL. Integrative genomic analyses of sporadic clear cell renal cell carcinoma define disease subtypes and potential new therapeutic targets. Cancer Res 2011; 72:112-21. [PMID: 22094876 DOI: 10.1158/0008-5472.can-11-1698] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Sporadic clear cell renal cell carcinoma (ccRCC), the most common type of adult kidney cancer, is often associated with genomic copy number aberrations on chromosomes 3p and 5q. Aberrations on chromosome 3p are associated with inactivation of the tumor suppressor gene von-Hippel Lindau (VHL), which activates the hypoxia-inducible factors HIF1α and HIF2α. In contrast, ccRCC genes on chromosome 5q remain to be defined. In this study, we conducted an integrated analysis of high-density copy number and gene expression data for 54 sporadic ccRCC tumors that identified the secreted glycoprotein STC2 (stanniocalcin 2) and the proteoglycan VCAN (versican) as potential 5q oncogenes in ccRCCs. In functional assays, STC2 and VCAN each promoted tumorigenesis by inhibiting cell death. Using the same approach, we also investigated the two VHL-deficient subtypes of ccRCC, which express both HIF1α and HIF2α (H1H2) or only HIF2α (H2). This analysis revealed a distinct pattern of genomic aberrations in each group, with the H1H2 group displaying, on average, a more aberrant genome than the H2 group. Together our findings provide a significant advance in understanding ccRCCs by offering a molecular definition of two subtypes with distinct characteristics as well as two potential chromosome 5q oncogenes, the overexpression of which is sufficient to promote tumorigenesis by limiting cell death.
Collapse
Affiliation(s)
- Vijay R Dondeti
- Department of Medicine, Abramson Family Cancer Research Institute, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
The regulation of cellular Ca(2+) homeostasis is essential for innumerable physiological and pathological processes. Stanniocalcin 1, a secreted glycoprotein hormone originally described in fish, is a well-established endocrine regulator of gill Ca(2+) uptake during hypercalcemia. While there are two mammalian Stanniocalcin homologs (STC1 and STC2), their precise molecular functions remain unknown. Notably, STC2 is a prosurvival component of the unfolded protein response. Here, we demonstrate a cell-intrinsic role for STC2 in the regulation of store-operated Ca(2+) entry (SOCE). Fibroblasts cultured from Stc2 knockout mice accumulate higher levels of cytosolic Ca(2+) following endoplasmic reticulum (ER) Ca(2+) store depletion, specifically due to an increase in extracellular Ca(2+) influx through store-operated Ca(2+) channels (SOC). The knockdown of STC2 expression in a hippocampal cell line also potentiates SOCE, and the overexpression of STC2 attenuates SOCE. Moreover, STC2 interacts with the ER Ca(2+) sensor STIM1, which activates SOCs following ER store depletion. These results define a novel molecular function for STC2 as a negative modulator of SOCE and provide the first direct evidence for the regulation of Ca(2+) homeostasis by mammalian STC2. Furthermore, our findings implicate the modulation of SOCE through STC2 expression as one of the prosurvival measures of the unfolded protein response.
Collapse
|
48
|
Law AYS, Wong CKC. Stanniocalcin-2 promotes epithelial-mesenchymal transition and invasiveness in hypoxic human ovarian cancer cells. Exp Cell Res 2010; 316:3425-34. [PMID: 20619259 DOI: 10.1016/j.yexcr.2010.06.026] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Revised: 06/11/2010] [Accepted: 06/29/2010] [Indexed: 12/24/2022]
Abstract
The human glycoprotein, stanniocalcin-2 (STC2) is a HIF-1 target gene that is found to be associated with tumor development. The relationship of the prognostic outcome to the level of its expression in cancer tissues is controversial; however experimental evidence suggests that STC2 is a positive regulator of cancer progression. In the present study, we investigated if the expression of STC2 in hypoxic cells is associated with cancer invasion and metastasis. We studied the epithelial-mesenchymal transition (EMT) markers in STC2-silenced and over-expressed SKOV3 cells maintained in hypoxic condition. Western blot and immunocytochemical analysis revealed that the stable expression of exogenous STC2 promoted EMT, as revealed by the increase of N-cadherin/vimentin but a decrease of E-cadherin levels. This observation was further confirmed by colony formation assay where the STC2 stably transfected cells showed high degree of motility with fibroblast morphology under hypoxic condition. In conducting invasion assay in hypoxia, the STC2 stably transfected cells showed high degree of invasiveness. This observation was correlated with the significant increase of MMP2 and MMP9 expression in the STC2 stably transfected cells. In HUVEC/SKOV3 co-culture invasion study, endothelial invasion was found to be enhanced by the seeding of STC2 stably transfected cells in the lower compartment. These observations were possibly mediated by an increase of ROS and activated ERK1/2 levels in the cells. Collectively, the finding provides the first evidence that STC2 is a positive regulator in tumor progression at hypoxia.
Collapse
Affiliation(s)
- Alice Y S Law
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | | |
Collapse
|
49
|
Yokobori T, Mimori K, Ishii H, Iwatsuki M, Tanaka F, Kamohara Y, Ieta K, Kita Y, Doki Y, Kuwano H, Mori M. Clinical Significance of Stanniocalcin 2 as a Prognostic Marker in Gastric Cancer. Ann Surg Oncol 2010; 17:2601-7. [DOI: 10.1245/s10434-010-1086-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2010] [Indexed: 11/18/2022]
|
50
|
Roch GJ, Sherwood NM. Genomics Reveal Ancient Forms of Stanniocalcin in Amphioxus and Tunicate. Integr Comp Biol 2010; 50:86-97. [DOI: 10.1093/icb/icq010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|