1
|
Zhang Y, Li Z, Chen X, Huang Y, Zou B, Xu Y. Prognostic significance of FAT10 expression in malignant tumors: a systematic review and meta-analysis. Future Oncol 2024; 20:1505-1514. [PMID: 38864667 PMCID: PMC11441062 DOI: 10.1080/14796694.2024.2357531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 05/16/2024] [Indexed: 06/13/2024] Open
Abstract
Aim: FAT10, a ubiquitin-like modifier protein, influences apoptosis, DNA damage response and tumor growth, with unclear effects on cancer prognosis.Methods: We reviewed FAT10 expression's impact on malignancy prognosis through a systematic review and meta-analysis, including studies up to September 2023 from PubMed, EMBASE and Web of Science.Results: From 18 studies involving 2513 patients, FAT10 overexpression significantly reduced overall and disease-free survival across various tumors, indicating correlations with advanced disease stage, poor differentiation, lymph node metastasis and larger tumor size.Conclusion: FAT10's overexpression suggests a negative prognostic value in cancer, meriting further investigation.PROSPERO Registration Number: CRD42023431287.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Zheng Li
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Xi Chen
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Ying Huang
- College of Management, Sichuan Agricultural University, Chengdu, 611130, P.R. China
| | - Bingwen Zou
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yong Xu
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, P.R. China
| |
Collapse
|
2
|
Liu Y, Yang R, Zhang Y, Zhu Y, Bao W. ANGPTL4 functions as an oncogene through regulation of the ETV5/CDH5/AKT/MMP9 axis to promote angiogenesis in ovarian cancer. J Ovarian Res 2022; 15:131. [PMID: 36517864 PMCID: PMC9749186 DOI: 10.1186/s13048-022-01060-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 11/15/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Angiopoietin-like 4 (ANGPTL4) is highly expressed in a variety of neoplasms and promotes cancer progression. Nevertheless, the mechanism of ANGPTL4 in ovarian cancer (OC) metastasis remains unclear. This study aimeds to explore whether ANGPTL4 regulates OC progression and elucidate the underlying mechanism. METHODS ANGPTL4 expression in clinical patient tumor samples was determined by immunohistochemistry (IHC) and high-throughput sequencing. ANGPTL4 knockdown (KD) and the addition of exogeneous cANGPTL4 protein were used to investigate its function. An in vivo xenograft tumor experiment was performed by intraperitoneal injection of SKOV3 cells transfected with short hairpin RNAs (shRNAs) targeting ANGPTL4 in nude mice. Western blotting and qRT-PCR were used to detect the levels of ANGPTL4, CDH5, p-AKT, AKT, ETV5, MMP2 and MMP9 in SKOV3 and HO8910 cells transfected with sh-ANGPTL4 or shRNAs targeting ETV5. RESULTS Increased levels of ANGPTL4 were associated with poor prognosis and metastasis in OC and induced the angiogenesis and metastasis of OC cells both in vivo and in vitro. This tumorigenic effect was dependent on CDH5, and the expression levels of ANGPTL4 and CDH5 in human OC werepositively correlated. In addition, CDH5 activated p-AKT, and upregulated the expression of MMP2 and MMP9. We also found that the expression of ETV5 was upregulated by ANGPTL4, which could bind the promoter region of CDH5, leading to increased CDH5 expression. CONCLUSION Our data indicated that an increase in the ANGPTL4 level results in increased ETV5 expression in OC, leading to metastasis via activation of the CDH5/AKT/MMP9 signaling pathway.
Collapse
Affiliation(s)
- Yinping Liu
- Qingpu Branch of Zhongshan Hospital, Fudan University, 1158 Gongyuandong Road, Qingpu District, 201700, Shanghai, P. R. China
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 85 Wujin Road, Hongkou, 200080, Shanghai, P. R. China
| | - Rui Yang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 85 Wujin Road, Hongkou, 200080, Shanghai, P. R. China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 85 Wujin Road, Hongkou, 200080, Shanghai, P. R. China
| | - Yaping Zhu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 85 Wujin Road, Hongkou, 200080, Shanghai, P. R. China.
| | - Wei Bao
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 85 Wujin Road, Hongkou, 200080, Shanghai, P. R. China.
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 85 Wujin Road, Hongkou, 201620, Shanghai, P.R. China.
| |
Collapse
|
3
|
Chou CL, Chen TJ, Li WS, Lee SW, Yang CC, Tian YF, Lin CY, He HL, Wu HC, Shiue YL, Li CF, Kuo YH. Upregulated Ubiquitin D is a Favorable Prognostic Indicator for Rectal Cancer Patients Undergoing Preoperative Concurrent Chemoradiotherapy. Onco Targets Ther 2022; 15:1171-1181. [PMID: 36238133 PMCID: PMC9553428 DOI: 10.2147/ott.s378666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/23/2022] [Indexed: 11/06/2022] Open
Abstract
PURPOSE For locally advanced rectal cancer, neoadjuvant concurrent chemoradiotherapy (CCRT) allows tumor downstaging and makes curative radical proctectomy possible. However, we lack a genetic biomarker to predict cancer prognosis or treatment response. We investigated the association between ubiquitin D (UBD) expression and clinical outcomes in rectal cancer patients receiving CCRT. PATIENTS AND METHODS We analyzed the genes associated with the protein modification process (GO:0036211) and identified the UBD gene as the most relevant among the top 7 differentially expressed genes associated with CCRT resistance. We collected tissue specimens from 172 rectal cancer patients who had received CCRT followed by a curative proctectomy. We examine the relationship between UBD expression and patient characteristics, pathological findings, and patient survival, such as metastasis-free survival (MeFS) and disease-specific survival. RESULTS Upregulated UBD expression was associated with lower pre-CCRT tumor T stage (P = 0.009), lower post-CCRT tumor T stage (P < 0.001), lower post-CCRT nodal stage (P < 0.001), less vascular invasion (P = 0.015), and better tumor regression (P < 0.001). Using univariate analysis, we found that high UBD expression was correlated with better disease-free survival (DFS) (P < 0.0001), local recurrence-free survival (LRFS) (P < 0.0001) and MeFS (P < 0.0001). Moreover, multivariate analysis demonstrated that high UBD expression was associated with superior DFS (P < 0.001), LRFS (P = 0.01), and MeFS (P = 0.004). CONCLUSION UBD upregulation was linked to better clinical prognosis, favorable pathological features, and good treatment response in rectal cancer patients undergoing CCRT. These results suggest UBD is a biomarker for rectal cancer.
Collapse
Affiliation(s)
- Chia-Lin Chou
- Department of Medical Technology, Chung Hwa University of Medical Technology, Tainan, 717, Taiwan
- Division of Colon and Rectal Surgery, Department of Surgery, Chi Mei Medical Center, Tainan, 710, Taiwan
| | - Tzu-Ju Chen
- Department of Medical Technology, Chung Hwa University of Medical Technology, Tainan, 717, Taiwan
- Department of Pathology, Chi Mei Medical Center, Tainan, 710, Taiwan
- Department of Clinical Pathology, Chi Mei Medical Center, Tainan, 710, Taiwan
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Wan-Shan Li
- Department of Medical Technology, Chung Hwa University of Medical Technology, Tainan, 717, Taiwan
- Department of Pathology, Chi Mei Medical Center, Tainan, 710, Taiwan
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Sung-Wei Lee
- Department of Radiation Oncology, Chi Mei Medical Center, Liouying, 736, Taiwan
| | - Ching-Chieh Yang
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, 710, Taiwan
- College of Pharmacy and Science, Chia Nan University, Tainan, Taiwan
| | - Yu-Feng Tian
- Division of Colon and Rectal Surgery, Department of Surgery, Chi Mei Medical Center, Tainan, 710, Taiwan
| | - Cheng-Yi Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, 710, Taiwan
| | - Hong-Lin He
- Department of Pathology, E-DA Hospital & E-DA Cancer Hospital, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Hung-Chang Wu
- College of Pharmacy and Science, Chia Nan University, Tainan, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Yow-Ling Shiue
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
- Institute of Precision Medicine, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Chien-Feng Li
- Institute of Precision Medicine, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
- Department of Medical Research, Chi Mei Medical Center, Tainan, 710, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Yu-Hsuan Kuo
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
- College of Pharmacy and Science, Chia Nan University, Tainan, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
- Institute of Precision Medicine, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| |
Collapse
|
4
|
Ploypetch S, Roytrakul S, Jaresitthikunchai J, Phaonakrop N, Teewasutrakul P, Rungsipipat A, Suriyaphol G. Salivary proteomics in monitoring the therapeutic response of canine oral melanoma. PLoS One 2021; 16:e0256167. [PMID: 34411146 PMCID: PMC8376060 DOI: 10.1371/journal.pone.0256167] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/30/2021] [Indexed: 11/18/2022] Open
Abstract
Saliva biomarkers are suitable for monitoring the therapeutic response of canine oral melanoma (COM), because saliva directly contacts the tumor, and saliva collection is non-invasive, convenient and cost effective. The present study aimed to investigate novel biomarkers from the salivary proteome of COM treated with surgery and a chemotherapy drug, carboplatin, 1-6 times, using a liquid chromatography-tandem mass spectrometry approach. The expression of a potential salivary biomarker, ubiquitin D (UBD), was observed and verified by western blot analysis. A significantly increased ratio of free UBD (fUBD) to conjugated UBD (cUBD) was shown in the pre-surgery stage (PreS) in OM dogs with short-term survival (STS) (less than 12 months after surgery) compared with that with long-term survival (more than 12 months after surgery). In dogs with STS, the ratio was also shown to be augmented in PreS compared with that after surgery, followed by treatment with carboplatin twice, 4 and 5 times [After treatment (AT)2, AT4 and AT5]. In addition, the expression of fUBD was enhanced in PreS compared with that of AT2 in the STS group. In conclusion, this study revealed that a ratio of fUBD to cUBD in PreS was plausibly shown to be a potential prognostic biomarker for survival in dogs with OM.
Collapse
Affiliation(s)
- Sekkarin Ploypetch
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Companion Animal Cancer Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Janthima Jaresitthikunchai
- Functional Proteomics Technology Laboratory, Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Narumon Phaonakrop
- Functional Proteomics Technology Laboratory, Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Patharakrit Teewasutrakul
- Companion Animal Cancer Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Oncology Clinic, Faculty of Veterinary Science, Small Animal Teaching Hospital, Chulalongkorn University, Bangkok, Thailand
| | - Anudep Rungsipipat
- Companion Animal Cancer Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Gunnaporn Suriyaphol
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Companion Animal Cancer Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
5
|
Su H, Qin M, Liu Q, Jin B, Shi X, Xiang Z. Ubiquitin-Like Protein UBD Promotes Cell Proliferation in Colorectal Cancer by Facilitating p53 Degradation. Front Oncol 2021; 11:691347. [PMID: 34350116 PMCID: PMC8327751 DOI: 10.3389/fonc.2021.691347] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/30/2021] [Indexed: 11/25/2022] Open
Abstract
Purpose Ubiquitin D (UBD) is a member of the ubiquitin-like modifier (UBL) family and is highly expressed in a variety of cancers including colorectal cancer (CRC). However, the mechanisms of its regulatory roles in CRC are largely elusive. In this study, we revealed the effect of UBD on the proliferation of CRC. Methods The expression of UBD in clinical tissue samples of CRC and seven CRC cell lines was detected using qRT-PCR, immunohistochemistry (IHC) and Western blotting. CCK-8, colony formation, EdU and flow cytometry assays were used to detect the functional changes of CRC cells transfected with UBD stable expression plasmids in vitro. A xenograft model was constructed to assess the effect of UBD on the growth of CRC cells in vivo. The connection between UBD and p53 was analyzed using Western blotting, immunoprecipitation, proteasome inhibition assay and Cycloheximide (CHX) chase assay. Results UBD was overexpressed in CRC tumor tissues compared with nontumor tissues, and its overexpression was positively associated with the tumor size and TNM stage of CRC patients. Functionally, UBD significantly accelerated CRC cell viability and proliferation in vitro and promoted tumorigenesis in vivo. Mechanistically, UBD interacted with p53 in CRC cells, downregulated the expression of p53 by regulating its degradation, shortened the p53 half-life, thereby further affecting the decrease in p21 and the increase in Cyclin D1, Cyclin E, CDK2, CDK4 and CDK6. Moreover, in vivo experiments showed that UBD-induced tumor growth in nude mice was dependent on a decrease in p53. Conclusions Our study proved that UBD mediates the degradation of p53, thereby facilitating the growth of CRC cells and ultimately promoting the progression of CRC. Therefore, UBD may be a potential therapeutic target and a promising prognostic biomarker for CRC.
Collapse
Affiliation(s)
- Hongbin Su
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mengdi Qin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiang Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bo Jin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xianjun Shi
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zheng Xiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Xiang S, Shao X, Cao J, Yang B, He Q, Ying M. FAT10: Function and Relationship with Cancer. Curr Mol Pharmacol 2021; 13:182-191. [PMID: 31729307 DOI: 10.2174/1874467212666191113130312] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/20/2019] [Accepted: 10/22/2019] [Indexed: 11/22/2022]
Abstract
Posttranslational protein modifications are known to be extensively involved in cancer, and a growing number of studies have revealed that the ubiquitin-like modifier FAT10 is directly involved in cancer development. FAT10 was found to be highly upregulated in various cancer types, such as glioma, hepatocellular carcinoma, breast cancer and gastrointestinal cancer. Protein FAT10ylation and interactions with FAT10 lead to the functional change of proteins, including proteasomal degradation, subcellular delocalization and stabilization, eventually having significant effects on cancer cell proliferation, invasion, metastasis and even tumorigenesis. In this review, we summarized the current knowledge on FAT10 and discussed its biological functions in cancer, as well as potential therapeutic strategies based on the FAT10 pathway.
Collapse
Affiliation(s)
- Senfeng Xiang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xuejing Shao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ji Cao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Meidan Ying
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
7
|
Aichem A, Groettrup M. The ubiquitin-like modifier FAT10 - much more than a proteasome-targeting signal. J Cell Sci 2020; 133:133/14/jcs246041. [PMID: 32719056 DOI: 10.1242/jcs.246041] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human leukocyte antigen (HLA)-F adjacent transcript 10 (FAT10) also called ubiquitin D (UBD) is a member of the ubiquitin-like modifier (ULM) family. The FAT10 gene is localized in the MHC class I locus and FAT10 protein expression is mainly restricted to cells and organs of the immune system. In all other cell types and tissues, FAT10 expression is highly inducible by the pro-inflammatory cytokines interferon (IFN)-γ and tumor necrosis factor (TNF). Besides ubiquitin, FAT10 is the only ULM which directly targets its substrates for degradation by the 26S proteasome. This poses the question as to why two ULMs sharing the proteasome-targeting function have evolved and how they differ from each other. This Review summarizes the current knowledge of the special structure of FAT10 and highlights its differences from ubiquitin. We discuss how these differences might result in differential outcomes concerning proteasomal degradation mechanisms and non-covalent target interactions. Moreover, recent insights about the structural and functional impact of FAT10 interacting with specific non-covalent interaction partners are reviewed.
Collapse
Affiliation(s)
- Annette Aichem
- Biotechnology Institute Thurgau at the University of Konstanz, CH-8280 Kreuzlingen, Switzerland.,Division of Immunology, Department of Biology, University of Konstanz, D-78457 Konstanz, Germany
| | - Marcus Groettrup
- Biotechnology Institute Thurgau at the University of Konstanz, CH-8280 Kreuzlingen, Switzerland .,Division of Immunology, Department of Biology, University of Konstanz, D-78457 Konstanz, Germany
| |
Collapse
|
8
|
Zhang X, Sun H, Chen W, He X. Elevated expression of AGGF1 predicts poor prognosis and promotes the metastasis of colorectal cancer. BMC Cancer 2019; 19:1252. [PMID: 31881864 PMCID: PMC6935059 DOI: 10.1186/s12885-019-6474-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/17/2019] [Indexed: 12/17/2022] Open
Abstract
Background Angiogenic factor with G-patch and FHA domains 1 (AGGF1) can promote angiogenesis and increasing evidence has highlighted the important roles of AGGF1 in tumorigenesis. However, the differential expression as well as the biological functions of AGGF1 in colorectal cancer (CRC) remain to be established. The purpose of the present study is therefore to identify the effect of AGGF1 on prognosis and metastasis in CRC patients. Methods The expression level of AGGF1 in CRC was examined by qPCR, western blot and immunohistochemistry in a tissue microarray containing 236 CRC specimens and paired normal mucosae. And the effect of AGGF1 on CRC cell malignance was investigated in our established stable AGGF1 upregulated and knockdown CRC cell lines. Results The expression level of AGGF1 in CRC tissue was not significantly different to that in adjacent normal mucosa at the mRNA level. However, at the protein level, AGGF1 expression in CRC tissues was significantly higher than in paired normal mucosa, which showed a clear association with TNM stage, AJCC stage, vascular invasion, and differentiation. Further, we revealed an apparent correlation between AGGF1 expression and poorer disease-free survival and overall survival of CRC patients. In addition, we discovered that AGGF1 significantly promoted CRC cell wound healing, migration, and invasion in vitro and distant metastasis in vivo. Conclusions Our study demonstrates the aberrant overexpression of AGGF1 in CRC and provides a basis on which to explore the application of AGGF1 as a potential therapeutic target for CRC patients, especially for CRC patients with distant metastasis.
Collapse
Affiliation(s)
- Xin Zhang
- Department of pathology, Zhejiang Provincial Peoples' Hospital, Peoples' Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Huimin Sun
- Department of pathology, Weifang People's Hospital, Weifang, 261041, China
| | - Wanyuan Chen
- Department of pathology, Zhejiang Provincial Peoples' Hospital, Peoples' Hospital of Hangzhou Medical College, Hangzhou, 310014, China.
| | - Xianglei He
- Department of pathology, Zhejiang Provincial Peoples' Hospital, Peoples' Hospital of Hangzhou Medical College, Hangzhou, 310014, China.
| |
Collapse
|
9
|
Zhuang M, Zhao S, Jiang Z, Wang S, Sun P, Quan J, Yan D, Wang X. MALAT1 sponges miR-106b-5p to promote the invasion and metastasis of colorectal cancer via SLAIN2 enhanced microtubules mobility. EBioMedicine 2019; 41:286-298. [PMID: 30797712 PMCID: PMC6444028 DOI: 10.1016/j.ebiom.2018.12.049] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/06/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023] Open
Abstract
Background The low expression of miR93/25 (members of miR-106b~25 cluster) promoted the invasion and metastasis of colon cancer cells, which predicted poor survival. However, the role of miR-106b-5p, the member of miR-106b~25 cluster, in colorectal cancer (CRC) remains unclear. Methods Bioinformatics methods were used to predict the potential pairs of lncRNA-miRNA-mRNA. In situ hybridization and qPCR were used to evaluate the expression of MALAT1 and miR-106b-5p in the paraffin-embedded normal and CRC tissues. Kaplan–Meier analysis with the log-rank test was used for survival analyses. Immunohistochemistry staining was applied to investigate the expression of SLAIN2. Fluorescence recovery after photobleaching assay was applied to observe the microtubule (MT) mobility. In vitro and in vivo invasion and metastasis assays were used to explore the function of MALAT1/miR-106b-5p/SLAIN2 in the progression of CRC. Findings miR-106b-5p was identified as a suppressor in CRC. Functionally, ectopic or silencing the expression of miR-106b-5p inhibited or promoted the invasion and metastasis of CRC cells in vitro and in vivo. The long non-coding RNA MALAT1 regulated the miR-106b-5p expression and further mediated the mobility of SLAIN2-related MTs by functioning as a competing endogenous RNA in vitro and in vivo, which resulted in the progression of CRC. Clinically, low miR-106b-5p expression predicted poor survival of CRC patients, especially in combination with high MALAT1/ SLAIN2 expression. Interpretation miR-106b-5p served as a suppressor in combination with MALAT1/miR-106b-5p/SLAIN2, which might be a group of potential prognostic biomarkers in the prognosis of CRC. Fund This work was supported by National Program Project for Precision Medicine in National Research and Development Plan of China (2016YFC0905300), National Natural Science Foundation of China (81572930), National Key Research and Development Program of the Ministry of Science and Technology of China (2016YFC0905303, 2016YFC1303200), Beijing Science and Technology Program (D17110002617004), Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences (2018PT32012), CAMS Innovation Fund for Medical Sciences (CIFMS) (2016-I2M-1-001), Incentive Fund for Academic Leaders of Oncology Hospital, Chinese Academy of Medical Sciences (RC2016003), and Beijing Hope Run Special Fund from Cancer Foundation of China (LC2017A19). The project of Shanghai Jiaotong Univversity (YG2017QN30).
Collapse
Affiliation(s)
- Meng Zhuang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Senlin Zhao
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zheng Jiang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Song Wang
- Department of Colorectal Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Peng Sun
- Department of Colorectal Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jichuan Quan
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Dongwang Yan
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Xishan Wang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
10
|
Li C, Dai L, Zhang J, Zhang Y, Lin Y, Cheng L, Tian H, Zhang X, Wang Q, Yang Q, Wang Y, Shi G, Cheng F, Su X, Yang Y, Zhang S, Yu D, Wei Y, Deng H. Follistatin-like protein 5 inhibits hepatocellular carcinoma progression by inducing caspase-dependent apoptosis and regulating Bcl-2 family proteins. J Cell Mol Med 2018; 22:6190-6201. [PMID: 30255547 PMCID: PMC6237577 DOI: 10.1111/jcmm.13906] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 08/18/2018] [Indexed: 02/05/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common and deadly malignant tumors in the world, especially in China. Follistatin‐like protein 5 (FSTL5) is a member of the FSTL family, which is involved in cell proliferation, migration, differentiation, and embryo development. We aimed to investigate the function and underlying mechanism of FSTL5 in HCC. FSTL5 expression was determined by immunohistochemistry staining in a liver cancer tissue microarray (TMA) and the correlation between FSTL5 and the prognosis of HCC patients was analysed. Further proliferation assay, colony formation assay, flow cytometry, and xenograft tumor model were performed to investigate the bioeffects of FSTL5 in HCC in vitro and in vivo. We found that FSTL5 expression was downregulated in HCC tissues and positively correlated with the prognosis of patients with HCC at tumor node metastasis stage I/II. Overexpression of FSTL5 efficiently impaired HCC growth both in vivo and in vitro with an exogenous manner. Mechanistic investigation demonstrated that FSTL5 promoted HCC cell apoptosis in a caspase‐dependent manner and regulated Bcl‐2 family proteins. These results indicate that FSTL5 may be a potential novel target for HCC treatment, and a biomarker for tumor prognosis.
Collapse
Affiliation(s)
- Chunlei Li
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Biochemistry, Faculty of Basic Medicine, Chongqing Three Gorges Medical College, Wanzhou, Chongqing, China
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junfeng Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Yujing Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Lin
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lin Cheng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongwei Tian
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingnan Wang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qianmei Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuan Wang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Gang Shi
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fuyi Cheng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaolan Su
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shuang Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dechao Yu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
11
|
Mi Y, Zhao S, Zhou C, Weng J, Li J, Wang Z, Sun H, Tang H, Zhang X, Sun X, Peng Z, Wen Y. Downregulation of homeobox gene Barx2 increases gastric cancer proliferation and metastasis and predicts poor patient outcomes. Oncotarget 2018; 7:60593-60608. [PMID: 27533254 PMCID: PMC5312404 DOI: 10.18632/oncotarget.11260] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 07/19/2016] [Indexed: 01/09/2023] Open
Abstract
Barx2 is a Bar family homeodomain transcription factor shown to play a critical role in cell adhesion and cytoskeleton remodeling, key processes in carcinogenesis and metastasis. Using quantitative real-time PCR, Western blotting, and immunohistochemistry, we found that Barx2 is expressed at lower levels in human gastric cancer (GC) tissues than in adjacent normal mucosa. In a multivariate analysis, Barx2 expression emerged as an independent prognostic factor for disease-free and overall survival. Kaplan-Meier survival analysis showed a trend toward even shorter overall survival in the patient group with Barx2-negative tumors, independent of advanced UICC stage and tumor relapse. Using in vitro and in vivo assays, we demonstrated that under normal conditions Barx2 inhibited GC cell proliferation and invasiveness through inhibition of the Wnt/β-catenin signaling pathway. These findings indicate that reduction or loss of Barx2 dis-inhibits GC cell proliferation and invasion, and that reduction in Barx2 could serve as an independent prognostic biomarker for poor outcome in GC patients.
Collapse
Affiliation(s)
- Yushuai Mi
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 200080 Shanghai, China
| | - Senlin Zhao
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 200080 Shanghai, China.,Department of Oncology and Department of Clinical and Experimental Medicine, Linkoping University, SE-581 85 Linkoping, Sweden
| | - Chongzhi Zhou
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 200080 Shanghai, China
| | - Junyong Weng
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 200080 Shanghai, China
| | - Jikun Li
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 200080 Shanghai, China
| | - Zhanshan Wang
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 200080 Shanghai, China
| | - Huimin Sun
- Department of Pathology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 200080 Shanghai, China
| | - Huamei Tang
- Department of Pathology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 200080 Shanghai, China
| | - Xin Zhang
- Department of Pathology, Zhejiang Provincial People's Hospital, 310014 Hangzhou Zhejiang, China
| | - Xiaofeng Sun
- Department of Oncology and Department of Clinical and Experimental Medicine, Linkoping University, SE-581 85 Linkoping, Sweden
| | - Zhihai Peng
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 200080 Shanghai, China
| | - Yugang Wen
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 200080 Shanghai, China.,Department of Oncology and Department of Clinical and Experimental Medicine, Linkoping University, SE-581 85 Linkoping, Sweden
| |
Collapse
|
12
|
Zhang J, Liu L, Sun Y, Xiang J, Zhou D, Wang L, Xu H, Yang X, Du N, Zhang M, Yan Q, Xi X. MicroRNA-520g promotes epithelial ovarian cancer progression and chemoresistance via DAPK2 repression. Oncotarget 2018; 7:26516-34. [PMID: 27049921 PMCID: PMC5041996 DOI: 10.18632/oncotarget.8530] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 02/18/2016] [Indexed: 11/25/2022] Open
Abstract
The lack of efficient tumor progression and chemoresistance indicators leads to high mortality in epithelial ovarian cancer (EOC) patients. Dysregulated miR-520g expression is involved in these processes in hepatic and colorectal cancers. In this study, we found that miR-520g expression gradually increased across normal, benign, borderline and EOC tissues. High miR-520g expression promoted tumor progression and chemoresistance to platinum-based chemotherapy, and reduced survival in EOC patients. miR-520g upregulation increased EOC cell proliferation, induced cell cycle transition and promoted cell invasion, while miR-520g downregulation inhibited tumor-related functions. In vivo, overexpression or downregulation of miR-520g respectively generated larger or smaller subcutaneous xenografts in nude mice. Death-associated protein kinase 2 (DAPK2) was a direct target of miR-520g. In 116 EOC tissue samples, miR-520g expression was significantly lower following DAPK2 overexpression. DAPK2 overexpression or miR-520g knockdown reduced EOC cell proliferation, invasion, wound healing and chemoresistance. This study suggests that miR-520g contributes to tumor progression and drug resistance by post-transcriptionally downregulating DAPK2, and that miR-520g may be a valuable therapeutic target in patients with EOC.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Lei Liu
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yunyan Sun
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Jiandong Xiang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Dongmei Zhou
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Li Wang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Huali Xu
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Xiaoming Yang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Na Du
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Meng Zhang
- Department of Pathology, Fudan University Affiliated Shanghai Cancer Center, Shanghai, China
| | - Qin Yan
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Xiaowei Xi
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| |
Collapse
|
13
|
Peng JH, Fang YJ, Li CX, Ou QJ, Jiang W, Lu SX, Lu ZH, Li PX, Yun JP, Zhang RX, Pan ZZ, Wan DS. A scoring system based on artificial neural network for predicting 10-year survival in stage II A colon cancer patients after radical surgery. Oncotarget 2017; 7:22939-47. [PMID: 27008710 PMCID: PMC5008413 DOI: 10.18632/oncotarget.8217] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 03/10/2016] [Indexed: 01/03/2023] Open
Abstract
Nearly 20% patients with stage II A colon cancer will develop recurrent disease post-operatively. The present study aims to develop a scoring system based on Artificial Neural Network (ANN) model for predicting 10-year survival outcome. The clinical and molecular data of 117 stage II A colon cancer patients from Sun Yat-sen University Cancer Center were used for training set and test set; poor pathological grading (score 49), reduced expression of TGFBR2 (score 33), over-expression of TGF-β (score 45), MAPK (score 32), pin1 (score 100), β-catenin in tumor tissue (score 50) and reduced expression of TGF-β in normal mucosa (score 22) were selected as the prognostic risk predictors. According to the developed scoring system, the patients were divided into 3 subgroups, which were supposed with higher, moderate and lower risk levels. As a result, for the 3 subgroups, the 10-year overall survival (OS) rates were 16.7%, 62.9% and 100% (P < 0.001); and the 10-year disease free survival (DFS) rates were 16.7%, 61.8% and 98.8% (P < 0.001) respectively. It showed that this scoring system for stage II A colon cancer could help to predict long-term survival and screen out high-risk individuals for more vigorous treatment.
Collapse
Affiliation(s)
- Jian-Hong Peng
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, P.R. China
| | - Yu-Jing Fang
- Department of Colorectal Surgery, Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, P. R. China
| | - Cai-Xia Li
- School of Mathematics and Computational Science,Sun Yat-sen University, Guangzhou, P.R. China,.,Guangdong Provincial Key Laboratory of Computational Science, Sun Yat-sen University, Guangzhou, P.R. China
| | - Qing-Jian Ou
- Department of Colorectal Surgery, Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, P. R. China
| | - Wu Jiang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, P.R. China
| | - Shi-Xun Lu
- Department of pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, P. R. China
| | - Zhen-Hai Lu
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, P.R. China
| | - Pei-Xing Li
- School of Mathematics and Computational Science,Sun Yat-sen University, Guangzhou, P.R. China,.,Guangdong Provincial Key Laboratory of Computational Science, Sun Yat-sen University, Guangzhou, P.R. China
| | - Jing-Ping Yun
- Department of pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, P. R. China
| | - Rong-Xin Zhang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, P.R. China
| | - Zhi-Zhong Pan
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, P.R. China
| | - De Sen Wan
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, P.R. China
| |
Collapse
|
14
|
Wang Z, Zhu WG, Xu X. Ubiquitin-like modifications in the DNA damage response. Mutat Res 2017; 803-805:56-75. [PMID: 28734548 DOI: 10.1016/j.mrfmmm.2017.07.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 06/03/2017] [Accepted: 07/03/2017] [Indexed: 12/14/2022]
Abstract
Genomic DNA is damaged at an extremely high frequency by both endogenous and environmental factors. An improper response to DNA damage can lead to genome instability, accelerate the aging process and ultimately cause various human diseases, including cancers and neurodegenerative disorders. The mechanisms that underlie the cellular DNA damage response (DDR) are complex and are regulated at many levels, including at the level of post-translational modification (PTM). Since the discovery of ubiquitin in 1975 and ubiquitylation as a form of PTM in the early 1980s, a number of ubiquitin-like modifiers (UBLs) have been identified, including small ubiquitin-like modifiers (SUMOs), neural precursor cell expressed, developmentally down-regulated 8 (NEDD8), interferon-stimulated gene 15 (ISG15), human leukocyte antigen (HLA)-F adjacent transcript 10 (FAT10), ubiquitin-fold modifier 1 (UFRM1), URM1 ubiquitin-related modifier-1 (URM1), autophagy-related protein 12 (ATG12), autophagy-related protein 8 (ATG8), fan ubiquitin-like protein 1 (FUB1) and histone mono-ubiquitylation 1 (HUB1). All of these modifiers have known roles in the cellular response to various forms of stress, and delineating their underlying molecular mechanisms and functions is fundamental in enhancing our understanding of human disease and longevity. To date, however, the molecular mechanisms and functions of these UBLs in the DDR remain largely unknown. This review summarizes the current status of PTMs by UBLs in the DDR and their implication in cancer diagnosis, therapy and drug discovery.
Collapse
Affiliation(s)
- Zhifeng Wang
- Guangdong Key Laboratory of Genome Stability & Disease Prevention, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, China
| | - Wei-Guo Zhu
- Guangdong Key Laboratory of Genome Stability & Disease Prevention, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, China
| | - Xingzhi Xu
- Guangdong Key Laboratory of Genome Stability & Disease Prevention, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, China; Beijing Key Laboratory of DNA Damage Response, Capital Normal University College of Life Sciences, Beijing 100048, China.
| |
Collapse
|
15
|
Bourroul GM, Fragoso HJ, Gomes JWF, Bourroul VSO, Oshima CTF, Gomes TS, Saba GT, Palma RT, Waisberg J. The destruction complex of beta-catenin in colorectal carcinoma and colonic adenoma. EINSTEIN-SAO PAULO 2017; 14:135-42. [PMID: 27462886 PMCID: PMC4943346 DOI: 10.1590/s1679-45082016ao3678] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/03/2016] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE To evaluate the destruction complex of beta-catenin by the expression of the proteins beta-catetenin, adenomatous polyposis coli, GSK3β, axin and ubiquitin in colorectal carcinoma and colonic adenoma. METHODS Tissue samples from 64 patients with colorectal carcinoma and 53 patients with colonic adenoma were analyzed. Tissue microarray blocks and slides were prepared and subjected to immunohistochemistry with polyclonal antibodies in carcinoma, adjacent non-neoplastic mucosa, and adenoma tissues. The immunoreactivity was evaluated by the percentage of positive stained cells and by the intensity assessed through of the stained grade of proteins in the cytoplasm and nucleus of cells. In the statistical analysis, the Spearman correlation coefficient, Student's t, χ2, Mann-Whitney, and McNemar tests, and univariate logistic regression analysis were used. RESULTS In colorectal carcinoma, the expressions of beta-catenin and adenomatous polyposis coli proteins were significantly higher than in colonic adenomas (p<0.001 and p<0.0001, respectively). The immunoreactivity of GSK3β, axin 1 and ubiquitin proteins was significantly higher (p=0.03, p=0.039 and p=0.03, respectively) in colorectal carcinoma than in the colonic adenoma and adjacent non-neoplastic mucosa. The immunohistochemistry staining of these proteins did not show significant differences with the clinical and pathological characteristics of colorectal cancer and colonic adenoma. CONCLUSIONS These results suggest that, in adenomas, the lower expression of the beta-catenin, axin 1 and GSK3β proteins indicated that the destruction complex of beta-catenin was maintained, while in colorectal carcinoma, the increased expression of beta-catenin, GSK3β, axin 1, and ubiquitin proteins indicated that the destruction complex of beta-catenin was disrupted. OBJETIVO Avaliar o complexo de destruição da betacatenina no carcinoma colorretal e no adenoma do colo pela expressão das proteínas betacatenina, adenomatous polyposis coli, GSK3β, axina e ubiquitina. MÉTODOS Amostras de tecidos de 64 doentes com carcinoma colorretal e de 53 pacientes com adenoma do colo foram analisadas. Blocos de tecidos foram submetidos ao estudo imuno-histoquímico com anticorpos policlonais nos tecidos do carcinoma, mucosa não neoplásica adjacente e adenoma. A imunorreatividade foi avaliada pela porcentagem de positividade de células coradas e pela intensidade do grau de coloração das proteínas no citoplasma e no núcleo das células. Na análise estatística, foram utilizados o coeficiente de correlação de Spearman, os testes t de Student, χ2, Mann-Whitney e de McNemar, e a análise de regressão logística univariada. RESULTADOS No carcinoma colorretal, as expressões da betacatenina e da adenomatous polyposis coli foram significativamente maiores do que em adenomas do colo (p<0,001 e p<0,0001, respectivamente). A imunorreatividade das proteínas GSK3β, axina 1 e ubiquitina foi significativamente maior (p=0,03, p=0,039 e p=0,03, respectivamente) no carcinoma colorretal do que no adenoma e na mucosa não neoplásica adjacente. A coloração imuno-histoquímica dessas proteínas não apresentou diferenças significantes em relação às características clinicopatológicas do câncer colorretal e do adenoma. CONCLUSÕES Em adenomas, as menores expressões de betacatenina, axina 1 e GSK3β indicaram que o complexo de destruição da betacatenina estava conservado, enquanto que, no carcinoma colorretal, o aumento das expressões da betacatenina, GSK3β, 1 axina, e ubiquitina indicaram que o complexo de destruição de betacatenina estava alterado.
Collapse
Affiliation(s)
| | - Hélio José Fragoso
- Hospital do Servidor Público Estadual "Francisco Morato de Oliveira", São Paulo, SP, Brazil
| | | | | | | | - Thiago Simão Gomes
- Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | - Rogério Tadeu Palma
- Hospital do Servidor Público Estadual "Francisco Morato de Oliveira", São Paulo, SP, Brazil
| | - Jaques Waisberg
- Hospital do Servidor Público Estadual "Francisco Morato de Oliveira", São Paulo, SP, Brazil
| |
Collapse
|
16
|
Cheng L, Yang Q, Li C, Dai L, Yang Y, Wang Q, Ding Y, Zhang J, Liu L, Zhang S, Fan P, Hu X, Xiang R, Yu D, Wei Y, Deng H. DDA1, a novel oncogene, promotes lung cancer progression through regulation of cell cycle. J Cell Mol Med 2017; 21:1532-1544. [PMID: 28211159 PMCID: PMC5542901 DOI: 10.1111/jcmm.13084] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 12/07/2016] [Indexed: 02/05/2023] Open
Abstract
Lung cancer is globally widespread and associated with high morbidity and mortality. DDA1 (DET1 and DDB1 associated 1) was first discovered and registered in the GenBank database by our colleagues. DDA1, an evolutionarily conserved gene, might have significant functions. Recent reports have demonstrated that DDA1 is linked to the ubiquitin–proteasome pathway and facilitates the degradation of target proteins. However, the function of DDA1 in lung cancer was previously unknown. This study aimed to investigate whether DDA1 contributes to tumorigenesis and progression of lung cancer. We found that the expression of DDA1 in normal lung cells and tissue was significantly lower than that in lung cancer and was associated with poor prognosis. DDA1 overexpression promoted proliferation of lung tumour cells and facilitated cell cycle progression in vitro and subcutaneous xenograft tumour progression in vivo. Mechanistically, this was associated with the regulation of S phase and cyclins including cyclin D1/D3/E1. These results indicate that DDA1 promotes lung cancer progression, potentially through promoting cyclins and cell cycle progression. Therefore, DDA1 may be a potential novel target for lung cancer treatment, and a biomarker for tumour prognosis.
Collapse
Affiliation(s)
- Lin Cheng
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qianmei Yang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Can Li
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Dai
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Yang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingnan Wang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu Ding
- Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junfeng Zhang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Liu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shuang Zhang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ping Fan
- Huaxi Biobank, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xun Hu
- Huaxi Biobank, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rong Xiang
- Department of Immunology, Nankai University School of Medicine, Tianjin, China
| | - Dechao Yu
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
17
|
Mi Y, Zhao S, Zhang W, Zhang D, Weng J, Huang K, Sun H, Tang H, Zhang X, Sun X, Peng Z, Wen Y. Down-regulation of Barx2 predicts poor survival in colorectal cancer. Biochem Biophys Res Commun 2016; 478:67-73. [DOI: 10.1016/j.bbrc.2016.07.091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 07/20/2016] [Indexed: 01/20/2023]
|
18
|
The ubiquitin-like modifier FAT10 in cancer development. Int J Biochem Cell Biol 2016; 79:451-461. [PMID: 27393295 DOI: 10.1016/j.biocel.2016.07.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/30/2016] [Accepted: 07/01/2016] [Indexed: 12/13/2022]
Abstract
During the last years it has emerged that the ubiquitin-like modifier FAT10 is directly involved in cancer development. FAT10 expression is highly up-regulated by pro-inflammatory cytokines IFN-γ and TNF-α in all cell types and tissues and it was also found to be up-regulated in many cancer types such as glioma, colorectal, liver or gastric cancer. While pro-inflammatory cytokines within the tumor microenvironment probably contribute to FAT10 overexpression, an increasing body of evidence argues that pro-malignant capacities of FAT10 itself largely underlie its broad and intense overexpression in tumor tissues. FAT10 thereby regulates pathways involved in cancer development such as the NF-κB- or Wnt-signaling. Moreover, FAT10 directly interacts with and influences downstream targets such as MAD2, p53 or β-catenin, leading to enhanced survival, proliferation, invasion and metastasis formation of cancer cells but also of non-malignant cells. In this review we will provide an overview of the regulation of FAT10 expression as well as its function in carcinogenesis.
Collapse
|
19
|
Zhao S, Tang H, Yan D, Fan J, Sun H, Wen Y, Yu F, Cui F, Zhang D, Xue Y, Liu C, Yue B, Chen J, Wang J, Wang X, Zhang M, Yu Y, Jiang W, Liu X, Mi Y, Zhou Z, Qin X, Peng Z. DDA1 promotes stage IIB-IIC colon cancer progression by activating NFκB/CSN2/GSK-3β signaling. Oncotarget 2016; 7:19794-812. [PMID: 26942699 PMCID: PMC4991419 DOI: 10.18632/oncotarget.7847] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 02/06/2016] [Indexed: 02/05/2023] Open
Abstract
Conventional high-recurrence risk factors are not sufficient to predict post-operative risk of tumor recurrence or sensitivity to 5-fluorouracil (5-FU)-based chemotherapy for stage II colon cancer. DDA1, an evolutionarily conserved gene located at 19p13.11, may be involved in the activation of nuclear factor kappaB (NFκB). This study aimed to investigate whether DDA1 contributes to tumorigenesis and progression of stage II colon cancer via activation of the NFκB pathway. We found that positive expression of DDA1 alone or in combination with p65 nuclear translocation correlated with increased risk of tumor recurrence in patients with stage IIB-IIC colon cancer. DDA1 overexpression in colon cancer lines promoted cell proliferation, facilitated cell cycle progression, inhibited 5-FU-induced apoptosis, enhanced invasion, and induced the epithelial-mesenchymal transition. Suppression of DDA1 inhibited tumor progression, and reduced tumor growth in vivo. We also demonstrated that DDA1-mediated tumor progression is associated with the activation of the NFκB/COP9 signalosome 2(CSN2)/glycogen synthase kinase3β (GSK3β) pathway. These results indicate that DDA1 promotes colon cancer progression through activation of NFκB/CSN2/GSK3β signaling. DDA1, together with NFκB activation status, may serve as a sensitive biomarker for tumor recurrence risk and prognosis in patients with stage IIB-IIC colon cancers.
Collapse
Affiliation(s)
- Senlin Zhao
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Huamei Tang
- Department of Pathology, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Dongwang Yan
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Junwei Fan
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Hongcheng Sun
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Yugang Wen
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Fudong Yu
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Feifei Cui
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Dongyuan Zhang
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Yingming Xue
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Chenchen Liu
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Ben Yue
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Jian Chen
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Jingtao Wang
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Xiao Wang
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Meng Zhang
- Department of Pathology, Fudan University Affiliated Shanghai Cancer Center, Shanghai, China
| | - Yang Yu
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Weiliang Jiang
- Department of Gastroenterology, Shanghai First People's Hospital, Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Xisheng Liu
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Yushuai Mi
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Zongguang Zhou
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xuebin Qin
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, USA
| | - Zhihai Peng
- Department of General Surgery, Shanghai First People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
20
|
Yu G, Fang W, Xia T, Chen Y, Gao Y, Jiao X, Huang S, Wang J, Li Z, Xie K. Metformin potentiates rapamycin and cisplatin in gastric cancer in mice. Oncotarget 2016; 6:12748-62. [PMID: 25909163 PMCID: PMC4494971 DOI: 10.18632/oncotarget.3327] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 02/09/2015] [Indexed: 12/22/2022] Open
Abstract
Here we showed that pAMPKα and PTEN were down-regulated and p-mTOR, p-S6, p-4EBP1, MMP7, and DCN1 were up-regulated in human gastric cancer tissue samples as compared to that in the noncancerous tissues. Metformin inhibited tumor growth in mice. Also it enhanced cisplatin- or rapamycin-induced reduction of tumor growth as compared with treatment of either drug alone. In addition to activation of AMPK and suppression of the mTOR pathway, a series of increased and decreased genes expression were induced by metformin, including PTEN, MMP7, and FN1. We suggest that metformin could potentially be used for the treatment of gastric cancer especially in combination with cisplatin or rapamycin.
Collapse
Affiliation(s)
- Guanzhen Yu
- Department of Medical Oncology, Changzheng Hospital, Shanghai 200070, China.,Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Wenzheng Fang
- Department of Medical Oncology, Changzheng Hospital, Shanghai 200070, China.,Department of Oncology, Fuzhou General Hospital, Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350025, China
| | - Tian Xia
- Department of Gastroenterology, Changhai Hospital, Shanghai 200433, China.,Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ying Chen
- Department of Pathology, Changhai Hospital, Shanghai 200433, China
| | - Yunshu Gao
- Department of Oncology, Qingdao, Shandong 266000, China
| | - Xiaodong Jiao
- Department of Medical Oncology, Changzheng Hospital, Shanghai 200070, China
| | - Suyun Huang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Jiejun Wang
- Department of Medical Oncology, Changzheng Hospital, Shanghai 200070, China
| | - Zhaosheng Li
- Department of Gastroenterology, Changhai Hospital, Shanghai 200433, China
| | - Keping Xie
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|