1
|
Identification of candidate genes encoding tumor-specific neoantigens in early- and late-stage colon adenocarcinoma. Aging (Albany NY) 2021; 13:4024-4044. [PMID: 33428592 PMCID: PMC7906157 DOI: 10.18632/aging.202370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/31/2020] [Indexed: 12/24/2022]
Abstract
Colon adenocarcinoma (COAD) is one of the most common gastrointestinal malignant tumors and is characterized by a high mortality rate. Here, we integrated whole-exome and RNA sequencing data from The Cancer Genome Atlas and investigated the mutational spectra of COAD-overexpressed genes to define clinically relevant diagnostic/prognostic signatures and to unmask functional relationships with both tumor-infiltrating immune cells and regulatory miRNAs. We identified 24 recurrently mutated genes (frequency > 5%) encoding putative COAD-specific neoantigens. Five of them (NEB, DNAH2, ABCA12, CENPF and CELSR1) had not been previously reported as COAD biomarkers. Through machine learning-based feature selection, four early-stage-related (COL11A1, TG, SOX9, and DNAH2) and four late-stage-related (COL11A1, SOX9, TG and BRCA2) candidate neoantigen-encoding genes were selected as diagnostic signatures. They respectively showed 100% and 97% accuracy in predicting early- and late-stage patients, and an 8-gene signature had excellent prognostic performance predicting disease-free survival (DFS) in COAD patients. We also found significant correlations between the 24 candidate neoantigen genes and the abundance and/or activation status of 22 tumor-infiltrating immune cell types and 56 regulatory miRNAs. Our novel neoantigen-based signatures may improve diagnostic and prognostic accuracy and help design targeted immunotherapies for COAD treatment.
Collapse
|
2
|
Li S, Zhang J, Qian S, Wu X, Sun L, Ling T, Jin Y, Li W, Sun L, Lai M, Xu F. S100A8 promotes epithelial-mesenchymal transition and metastasis under TGF-β/USF2 axis in colorectal cancer. Cancer Commun (Lond) 2021; 41:154-170. [PMID: 33389821 PMCID: PMC7896751 DOI: 10.1002/cac2.12130] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/25/2020] [Accepted: 12/20/2020] [Indexed: 12/12/2022] Open
Abstract
Background The transforming growth factor‐β (TGF‐β) pathway plays a pivotal role in inducing epithelial‐mesenchymal transition (EMT), which is a key step in cancer invasion and metastasis. However, the regulatory mechanism of TGF‐β in inducing EMT in colorectal cancer (CRC) has not been fully elucidated. In previous studies, it was found that S100A8 may regulate EMT. This study aimed to clarify the role of S100A8 in TGF‐β‐induced EMT and explore the underlying mechanism in CRC. Methods S100A8 and upstream transcription factor 2 (USF2) expression was detected by immunohistochemistry in 412 CRC tissues. Kaplan‐Meier survival analysis was performed. In vitro, Western blot, and migration and invasion assays were performed to investigate the effects of S100A8 and USF2 on TGF‐β‐induced EMT. Mouse metastasis models were used to determine in vivo metastasis ability. Luciferase reporter and chromatin immunoprecipitation assay were used to explore the role of USF2 on S100A8 transcription. Results During TGF‐β‐induced EMT in CRC cells, S100A8 and the transcription factor USF2 were upregulated. S100A8 promoted cell migration and invasion and EMT. USF2 transcriptionally regulated S100A8 expression by directly binding to its promoter region. Furthermore, TGF‐β enhanced the USF2/S100A8 signaling axis of CRC cells whereas extracellular S100A8 inhibited the USF2/S100A8 axis of CRC cells. S100A8 expression in tumor cells was associated with poor overall survival in CRC. USF2 expression was positively related to S100A8 expression in tumor cells but negatively related to S100A8‐positive stromal cells. Conclusions TGF‐β was found to promote EMT and metastasis through the USF2/S100A8 axis in CRC while extracellular S100A8 suppressed the USF2/S100A8 axis. USF2 was identified as an important switch on the intracellular and extracellular S100A8 feedback loop.
Collapse
Affiliation(s)
- Si Li
- Department of Pathology and Pathophysiology, and Department of General Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China
| | - Jun Zhang
- Department of Pathology and Pathophysiology, and Department of General Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China
| | - Senmi Qian
- Department of Pathology and Pathophysiology, and Department of General Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China
| | - Xuesong Wu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China
| | - Liang Sun
- Department of Pathology and Pathophysiology, and Department of General Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China
| | - Tianyi Ling
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China
| | - Yao Jin
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China
| | - Wenxiao Li
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China
| | - Lichao Sun
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Maode Lai
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China
| | - Fangying Xu
- Department of Pathology and Pathophysiology, and Department of General Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China
| |
Collapse
|
3
|
Liu J, Zhang Y, Li Q, Wang Y. Transgelins: Cytoskeletal Associated Proteins Implicated in the Metastasis of Colorectal Cancer. Front Cell Dev Biol 2020; 8:573859. [PMID: 33117801 PMCID: PMC7575706 DOI: 10.3389/fcell.2020.573859] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/07/2020] [Indexed: 12/20/2022] Open
Abstract
Transgelins, including transgelin-1 (T-1), transgelin-2 (T-2), and transgelin-3 (T-3), are a family of actin-binding proteins (ABPs) that can alter the structure and morphology of the cytoskeleton. These proteins function by regulating migration, proliferation and apoptosis in many different cancers. Several studies have shown that in various types of tumor cells, including colorectal cancer (CRC) cells, and in the tumor microenvironment, the expression and biological effects of transgelins are diverse and may transform during tumor progression. Previous researches have demonstrated that transgelin levels are positively correlated with metastasis in CRC, and down-regulating their expression can inhibit this process. In advanced disease, T-1 is a tumor activator with increasing expression, and T-2 expression increases with the progression of CRC. Finally, T-3 is only expressed in neurons and is not associated with CRC. This evidence suggests that T-1 and T-2 are potential biomarkers and therapeutic targets for CRC metastasis.
Collapse
Affiliation(s)
- Jingwen Liu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yingru Zhang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
4
|
Zhu S, Zhao G, Zhao X, Zhan X, Cai M, Geng C, Pu Q, Zhao Q, Fu Q, Huang C, Wang Q. Elevated soluble E-cadherin during the epithelial-mesenchymal transition process and as a diagnostic marker in colorectal cancer. Gene 2020; 754:144899. [PMID: 32544494 DOI: 10.1016/j.gene.2020.144899] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/20/2020] [Accepted: 06/10/2020] [Indexed: 01/06/2023]
Abstract
Epithelial-mesenchymal transition (EMT) plays a crucial role in colorectal cancer (CRC) metastasis. Soluble E-cadherin (sE-cadherin) is a peptide degradation product of the E-cadherin, a key epithelial molecule of EMT. However, it is not known if elevated levels of sE-cadherin also occur during EMT. And the study of sE-cadherin in colorectal cancer is rare. The purpose of the study was to evaluate the relationship between sE-cadherin and EMT in CRC and to evaluate the diagnostic value of sE-cadherin as a serum marker for CRC. Transforming growth factor-β1 (TGF-β1) was used to induce EMT in HT29 and SW480 cells. The cells treated with TGF-β1 showed morphological and biological behavior changes consistent with EMT. Western blot and ELISA showed the levels of sE-cadherin were increased during EMT in CRC cells. In addition, we intravenously injected luciferase-labeled SW480 cells into nude mice to construct CRC metastasis model. Following the elongation of time, the fluorescence intensity of the experimental group was gradually increased. Correspondingly, the serum concentration of sE-cadherin also increased during CRC metastasis in mice. Furthermore, compared to healthy subjects, significantly higher levels of serum sE-cadherin were also observed in CRC patients and correlated with clinicopathological features. For discriminating CRC from healthy controls, the area under the receiver operating characteristic (ROC) curve (AUC) of sE-cadherin was 0.853, while the optimal cut-off point was set at 5928.16 ng/ml, the diagnostic sensitivity was 73.9% and the specificity was 80%. Compared with current commercial biomarkers (CEA, CA19-9 and CA125), the diagnostic performance of sE-cadherin was highest. Combined sE-cadherin and CEA raised the sensitivity to 82.4%. Serum sE-cadherin level can be used as a potential diagnostic biomarker of CRC.
Collapse
Affiliation(s)
- Shuzhen Zhu
- Department of Clinical Laboratory, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, Shandong 250012, China
| | - Guanghui Zhao
- Department of Clinical Laboratory, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, Shandong 250012, China
| | - Xiaoyun Zhao
- Department of Clinical Laboratory, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, Shandong 250012, China
| | - Xiaohong Zhan
- Department of Pathology, The Affiliated Hospital of Qingdao University, Jiangsu Road, Qingdao, Shandong 266035, China
| | - Meijuan Cai
- Department of Clinical Laboratory, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, Shandong 250012, China
| | - Congcong Geng
- Department of Clinical Laboratory, Qilu Hospital(Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong 266035, China
| | - Qian Pu
- Department of General Surgery, Qilu Hospital(Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong 266035, China
| | - Qianqian Zhao
- Department of Pathology, Qilu Hospital(Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong 266035, China
| | - Qingsong Fu
- Medical Experimental Center, Qilu Hospital(Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong 266035, China
| | - Chunxiao Huang
- Department of Clinical Laboratory, Qilu Hospital(Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong 266035, China
| | - Qian Wang
- Department of Clinical Laboratory, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, Shandong 250012, China.
| |
Collapse
|
5
|
Jin L, Shen F, Weinfeld M, Sergi C. Insulin Growth Factor Binding Protein 7 (IGFBP7)-Related Cancer and IGFBP3 and IGFBP7 Crosstalk. Front Oncol 2020; 10:727. [PMID: 32500027 PMCID: PMC7242731 DOI: 10.3389/fonc.2020.00727] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/16/2020] [Indexed: 12/17/2022] Open
Abstract
The insulin/insulin-like growth factors (IGFs) have crucial tasks in the growth, differentiation, and proliferation of healthy and pernicious cells. They are involved in coordinated complexes, including receptors, ligands, binding proteins, and proteases. However, the systems can become dysregulated in tumorigenesis. Insulin-like growth factor-binding protein 7 (IGFBP7) is a protein belonging to the IGFBP superfamily (also termed GFBP-related proteins). Numerous studies have provided evidence that IGFBP3 and IGFBP7 are involved in a variety of cancers, including hepatocellular carcinoma (HCC), breast cancer, gastroesophageal cancer, colon cancer, prostate cancer, among many others. Still, very few suggest an interaction between these two molecules. In studying several cancer types in our laboratories, we found that both proteins share some crucial signaling pathways. The objective of this review is to present a comprehensive overview of the relationship between IGFBP7 and cancer, as well as highlighting IGFBP3 crosstalk with IGFBP7 reported in recent studies.
Collapse
Affiliation(s)
- Li Jin
- Department of Laboratory Medicine, Shiyan Taihe Hospital, College of Biomedical Engineering, Hubei University of Medicine, Shiyan, China
| | - Fan Shen
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Michael Weinfeld
- Division of Experimental Oncology, Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Consolato Sergi
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
- Department of Orthopedics, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- Key Laboratory of Fermentation Engineering, National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, China
- Stollery Children's Hospital, University Alberta Hospital, Edmonton, AB, Canada
| |
Collapse
|
6
|
Li S, Xu F, Li H, Zhang J, Zhong A, Huang B, Lai M. S100A8 + stroma cells predict a good prognosis and inhibit aggressiveness in colorectal carcinoma. Oncoimmunology 2016; 6:e1260213. [PMID: 28197382 DOI: 10.1080/2162402x.2016.1260213] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/04/2016] [Accepted: 11/07/2016] [Indexed: 01/07/2023] Open
Abstract
Gene microarray and bioinformatic analysis showed that S100A8 was more abundant in the stroma surrounding tumor buddings (TBs) than in the stroma surrounding primary tumor cells in colorectal carcinomas. Here, S100A8+ cells in 419 colorectal carcinoma samples were stained by immunohistochemistry and counted using Image-pro plus 6.0. TBs were also counted and biomarkers associated with the epithelial-mesenchymal transition and apoptosis were assessed by immunohistochemistry. We evaluated the association between S100A8+ cells and clinico-pathological variables as well as survival. Migration and invasion as well as biomarkers of the epithelial-mesenchymal transition and apoptosis were tested in CRC cells, treated with graded concentrations of recombinant human S100A8 protein. We found that the density of S100A8+ cells in the tumor invasive front (S100A8+TIF) clearly distinguished patients with 5-y survival from those who did not survive (p = 0.01). The S100A8+-associated tumor budding (SATB) index determined by the S100A8+TIF and TB was an independent predictor of overall survival (p = 0.001) other than the S100A8+TIF or TB alone. Migration and invasion properties of CRC cells were inhibited by recombinant human S100A8 treatment. The particular S100A8+ cells in the stroma were associated with important biomarkers of the epithelial-mesenchymal transition (E-cadherin and SNAIL) and apoptosis (BCL2). In conclusion, S100A8+ cells in the stroma predict a good prognosis in colorectal carcinoma. An index combining S100A8+ cells and TB independently predicts survival. Recombinant human S100A8 inhibited CRC cell migration and invasion, which was involved in epithelial-mesenchymal transition (E-cadherin and SNAIL) and apoptosis (BCL2).
Collapse
Affiliation(s)
- Si Li
- Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China
| | - Fangying Xu
- Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China
| | - Hui Li
- Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China
| | - Jing Zhang
- Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China
| | - Anjing Zhong
- Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China
| | - Bin Huang
- Department of Pathology, The First Peoples Hospital of Xiaoshan , Hangzhou, Xiaoshan, China
| | - Maode Lai
- Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China
| |
Collapse
|
7
|
Ruan W, Kang Z, Li Y, Sun T, Wang L, Liang L, Lai M, Wu T. Interaction between IGFBP7 and insulin: a theoretical and experimental study. Sci Rep 2016; 6:19586. [PMID: 27101796 PMCID: PMC4840315 DOI: 10.1038/srep19586] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 10/14/2015] [Indexed: 12/13/2022] Open
Abstract
Insulin-like growth factor binding protein 7 (IGFBP7) can bind to insulin with high affinity which inhibits the early steps of insulin action. Lack of recognition mechanism impairs our understanding of insulin regulation before it binds to insulin receptor. Here we combine computational simulations with experimental methods to investigate the interaction between IGFBP7 and insulin. Molecular dynamics simulations indicated that His200 and Arg198 in IGFBP7 were key residues. Verified by experimental data, the interaction remained strong in single mutation systems R198E and H200F but became weak in double mutation system R198E-H200F relative to that in wild-type IGFBP7. The results and methods in present study could be adopted in future research of discovery of drugs by disrupting protein-protein interactions in insulin signaling. Nevertheless, the accuracy, reproducibility, and costs of free-energy calculation are still problems that need to be addressed before computational methods can become standard binding prediction tools in discovery pipelines.
Collapse
Affiliation(s)
- Wenjing Ruan
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou 310058, P. R. China.,Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, P.R. China
| | - Zhengzhong Kang
- Soft Matter Research Center and Department of Chemistry, Zhejiang University, Hangzhou 310027, P. R. China
| | - Youzhao Li
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou 310058, P. R. China
| | - Tianyang Sun
- Soft Matter Research Center and Department of Chemistry, Zhejiang University, Hangzhou 310027, P. R. China
| | - Lipei Wang
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou 310058, P. R. China
| | - Lijun Liang
- Soft Matter Research Center and Department of Chemistry, Zhejiang University, Hangzhou 310027, P. R. China
| | - Maode Lai
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou 310058, P. R. China
| | - Tao Wu
- Soft Matter Research Center and Department of Chemistry, Zhejiang University, Hangzhou 310027, P. R. China
| |
Collapse
|
8
|
IGFBP-rP1 suppresses epithelial-mesenchymal transition and metastasis in colorectal cancer. Cell Death Dis 2015; 6:e1695. [PMID: 25789970 PMCID: PMC4385937 DOI: 10.1038/cddis.2015.59] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 02/02/2015] [Accepted: 02/05/2015] [Indexed: 11/08/2022]
Abstract
Epithelial-mesenchymal transition (EMT) was initially recognized during organogenesis and has recently been reported to be involved in promoting cancer invasion and metastasis. Cooperation of transforming growth factor-β (TGF-β) and other signaling pathways, such as Ras and Wnt, is essential to inducing EMT, but the molecular mechanisms remain to be fully determined. Here, we reported that insulin-like growth factor binding protein-related protein 1 (IGFBP-rP1), a potential tumor suppressor, controls EMT in colorectal cancer progression. We revealed the inhibitory role of IGFBP-rP1 through analyses of clinical colorectal cancer samples and various EMT and metastasis models in vitro and in vivo. Moreover, we demonstrated that IGFBP-rP1 suppresses EMT and tumor metastasis by repressing TGF-β-mediated EMT through the Smad signaling cascade. These data establish that IGFBP-rP1 functions as a suppressor of EMT and metastasis in colorectal cancer.
Collapse
|
9
|
Wang N, Chen Y, Yang X, Jiang Y. Selenium-binding protein 1 is associated with the degree of colorectal cancer differentiation and is regulated by histone modification. Oncol Rep 2014; 31:2506-14. [PMID: 24737289 DOI: 10.3892/or.2014.3141] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 02/24/2014] [Indexed: 01/05/2023] Open
Abstract
The aim of the present study was to examine the regulation of selenium binding protein 1 (SELENBP1) expression in colorectal cancer (CRC). Samples of cancer tissue and adjacent normal mucosa were collected from 83 CRC patients, and analyzed for SELENBP1 expression by 2D-DIGE, immunoblotting, RT-PCR and immunostaining. Expression levels of SELENBP1, carcinoembryonic antigen (CEA) and alkaline phosphatase (AKP) were determined in cultures of human colon cancer cell lines (SW480, SW620 and HT29) folllowing treatment with i) sodium butyrate (NaB, 2 mM), a differentiation inducer; ii) Trichostatin A (TSA, 0.3 µM), a histone deacetylase inhibitor; or iii) 5'-aza-2'-deoxycytidine (5-Aza-dC, 5 µM), a DNA methylation inhibitor. SELENBP1 expression was found to be downregulated (2.54-fold) in the CRC samples as determined by 2D-DIGE and confirmed by immunoblotting and RT-PCR. SELENBP1 expression was correlated with the degree of differentiation, but not with TNM stage or lymph node metastasis, and was higher in benign polyps (1.97±0.57) than in CRC tissues (0.96±0.59). In the CRC cell lines, NaB treatment led to the upregulation of SELENBP1, CEA and AKP when compared with the untreated cells (2.24- to 4.82-fold). SELENBP1 was also upregulated in cells treated with TSA alone (1.25- to 3.64-fold), or in combination with 5-Aza-dC (1.32- to 4.13-fold). In CRC, the downregulated SELENBP1 expression was reactivated by inducing differentiation. Therefore, SELENBP1 is a potential pharmacological target for individualized CRC treatment.
Collapse
Affiliation(s)
- Ning Wang
- Department of General Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yang Chen
- Central Experimental Laboratory, The First Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xinghua Yang
- Department of General Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yi Jiang
- Central Experimental Laboratory, The First Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
10
|
Zhu S, Xu F, Zhang J, Ruan W, Lai M. Insulin-like growth factor binding protein-related protein 1 and cancer. Clin Chim Acta 2014; 431:23-32. [PMID: 24513543 DOI: 10.1016/j.cca.2014.01.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Revised: 01/23/2014] [Accepted: 01/24/2014] [Indexed: 12/11/2022]
Abstract
Insulin-like growth factor binding protein-related protein 1 (IGFBP-rP1) belongs to the IGFBP family whose members have a conserved structural homology. It has a low affinity for IGFs and a high affinity for insulin, suggesting that IGFBP-rP1 may have a biological function distinct from other members of the family. IGFBP-rP1 is ubiquitously expressed in normal human tissues and has diverse biological functions, regulating cell proliferation, apoptosis and senescence; it may also have a key role in vascular biology. Increasing evidence suggests that IGFBP-rP1 acts as a tumor suppressor. It elicits its biological effects by both insulin/IGF-dependent and -independent mechanisms. This paper provides a brief overview of the structure and regulation of IGFBP-rP1 and its various biological functions in cancer, as well as the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Shuzhen Zhu
- Department of Pathology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058 Zhejiang, China; Department of Clinical Laboratory, Qilu Hospital, Shandong University, 107 Wenhuaxi Road, Jinan 250012, China.
| | - Fangying Xu
- Department of Pathology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058 Zhejiang, China.
| | - Jing Zhang
- Department of Pathology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058 Zhejiang, China.
| | - Wenjing Ruan
- Department of Respiratory Diseases, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchundong Road, Hangzhou 310016, China.
| | - Maode Lai
- Department of Pathology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058 Zhejiang, China.
| |
Collapse
|
11
|
Identification of IGFBP-7 by urinary proteomics as a novel prognostic marker in early acute kidney injury. Kidney Int 2013; 85:909-19. [PMID: 24067438 DOI: 10.1038/ki.2013.363] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 07/31/2013] [Accepted: 08/01/2013] [Indexed: 11/08/2022]
Abstract
Early diagnosis of acute kidney injury (AKI) and accurate prognostic stratification is a prerequisite for optimal medical management. To identify novel prognostic markers of AKI, urine was collected on the first day of AKI in critically ill patients. Twelve patients with early recovery and 12 matching patients with late/non-recovery were selected and their proteome analyzed by gel electrophoresis and mass spectrometry. We identified eight prognostic candidates including α-1 microglobulin, α-1 antitrypsin, apolipoprotein D, calreticulin, cathepsin D, CD59, insulin-like growth factor-binding protein 7 (IGFBP-7), and neutrophil gelatinase-associated lipocalin (NGAL). Subsequent quantification by ELISA showed that IGFBP-7 was the most potent predictor of renal recovery. IGFBP-7 and NGAL were then chosen for further analyses in an independent verification group of 28 patients with and 12 control patients without AKI. IGFBP-7 and NGAL discriminated between early and late/non-recovery patients and patients with and without AKI. Significant upregulation of the urinary markers predicted mortality (IGFBP-7: AUC 0.68; NGAL: AUC 0.81), recovery (IGFBP-7: AUC 0.74; NGAL: AUC 0.70), and severity of AKI (IGFBP-7: AUC 0.77; NGAL: AUC 0.69), and were associated with the duration of AKI. IGFBP-7 was a more accurate predictor of renal outcome than NGAL. Thus, IGFBP-7 is a novel prognostic urinary marker that warrants further investigation.
Collapse
|
12
|
Zhan Y, Wang J, Ma Y, Liu Z, Xu H, Lu S, Lu B. Serum insulin-like, growth factor binding protein-related protein 1 (IGFBP-rP1) and endometrial cancer risk in Chinese women. Int J Cancer 2012; 132:411-6. [PMID: 22544761 DOI: 10.1002/ijc.27622] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 03/28/2012] [Indexed: 11/08/2022]
Abstract
Hyperinsulinemia and the metabolic syndrome confer increased risks of endometrial carcinoma. The roles of insulin, and, insulin-like growth factor-binding proteins (IGFBPs) in the etiology of endometrial carcinoma, remain unclear. We recruited 206 patients with endometrial carcinoma and 350 healthy women to a case-control study of fasting insulin and IGFBP-related protein 1 (IGFBP-rP1) in a Chinese tertiary centre. Patients with endometrial carcinoma had higher insulin concentrations (14.8 ± 16.7 vs. 8.1 ± 9.4 μU/mL; p < 0.001) and lower IGFBP-rP1 levels (17.5 ± 17.2 vs. 22.4 ± 22.8 μg/L; p = 0.018) than controls. High insulin and IGFBP-rP1 levels were both positively and negatively associated with endometrial cancer (odds ratio for the highest tertile versus the lowest tertile: insulin: 4.11; 95% CI = 2.61-6.47; IGFBP-rP1: 0.38; 95% CI = 0.24-0.60). Logistic regression analysis confirmed the associations between endometrial carcinoma and fasting insulin or IGFBP-rP1 after adjustments for age, BMI, serum glucose, cholesterol, triglycerides and high-density lipoprotein cholesterol (odds ratio for the highest tertile versus the lowest tertile: insulin: 2.13; 95% CI = 1.30-3.49; IGFBP-rP1: 0.57; 95% CI = 0.34-0.94). Hyperinsulinemia and high IGFBP-rP1 levels confer altered risks for endometrial carcinoma.
Collapse
Affiliation(s)
- Yan Zhan
- Department of Clinical Laboratory, The Affiliated Women's Hospital, School of Medicine, Zhejiang University, China
| | | | | | | | | | | | | |
Collapse
|
13
|
Hamed MAA, Ahmed SAA, Khaled HM. Efficiency of diagnostic biomarkers among colonic schistosomiasis Egyptian patients. Mem Inst Oswaldo Cruz 2011; 106:322-9. [PMID: 21655820 DOI: 10.1590/s0074-02762011000300011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 01/05/2011] [Indexed: 12/13/2022] Open
Abstract
The schistosomal parasite plays a critical role in the development of malignant lesions in different organs. The pathogenesis of cancer is currently under intense investigation to identify reliable prognostic indices for disease detection. The objective of this paper is to evaluate certain biochemical parameters as diagnostic tools to efficiently differentiate between colonic carcinoma and colonic carcinoma associated with schistosomal infection among Egyptian patients. The parameters under investigation are interleukin 2 (IL-2), tumour necrosis factor alpha (TNF-α), carcinoembryonic antigen (CEA) levels, tissue telomerase, pyruvate kinase (PK), glucose-6-phosphate dehydrogenase (G-6-PD) and lactate dehydrogenase (LDH) enzyme activities. The results revealed a significant elevation in the level of the tumour markers IL-2, TNF-α and CEA as well as the activities of LDH, telomerase and G-6-PD among non-bilharzial and bilharzial colonic cancer groups, with a more potent effect in bilharzial infection-associated colonic cancer. A significant inhibition in PK activity was recorded in the same manner as compared to normal tissues. The efficacy of this biomarker was also evaluated through detecting sensitivity, specificity, negative and positive predictive values. In conclusion, schistosomal colonic carcinoma patients displayed more drastic changes in all parameters under investigation. The combination of the selected parameters succeeded in serving as biomarkers to differentiate between the two malignant types.
Collapse
|