1
|
Xu B, Qiu T, Yang R, Qiang J, Yang Y, Zhou M, Li X, Dong J, Lu Y, Dong Z. Oxymatrine inhibits migration and invasion of esophageal squamous cell carcinoma cell lines via the MEK1/ERK/β-catenin pathway. Chem Biol Interact 2024; 404:111270. [PMID: 39419199 DOI: 10.1016/j.cbi.2024.111270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
Esophageal, cancer is a prevalent malignant tumour of the digestive system in China, and esophageal squamous cell carcinoma (ESCC) accounts for 90 % of all esophageal cancer cases. Currently, the primary treatment involves surgical resection combined with postoperative radiotherapy. In this study, we used two ESCC cell lines to determine whether oxymatrine (OMT) inhibits ESCC, whether the mechanism involves the MEK1/ERK/β-catenin pathway, and how OMT modulates this pathway to affect the development of ESCC. The effects of OMT treatment were monitored with Cell Counting Kit-8 (CCK-8) assays as well as with clony formation, migration and invasion, wound healing, Hoechst 33258, and Western blot analyses. The relationship between OMT and the target was also evaluated by molecular docking and cell stability experiments. These findings suggest that ESCC development and metastasis may be inhibited by OMT and that OMT targets MEK1 through the ERK/β-catenin/EMT pathway to suppress ESCC cell migration and invasion. In addition, in vivo studies confirmed that OMT can inhibit the growth of ESCC cell lines in NOG mice without causing damage to other organs. In conclusion, in vitro experiments, revealed that OMT prevents the migration and invasiveness of ESCC cells by inhibiting the ERK/β-catenin/EMT pathway and thus targeting MAP2K1 (MEK1) in ESCC.
Collapse
Affiliation(s)
- Baoshi Xu
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Tian Qiu
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London, E1 4NS, UK
| | - Rongrong Yang
- Department of Oncology, The Second People's Hospital of Lianyungang (The Oncology Hospital of Lianyungang), Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222000, China
| | - Jingchao Qiang
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yongliang Yang
- Department of Oncology, The Second People's Hospital of Lianyungang (The Oncology Hospital of Lianyungang), Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222000, China
| | - Mengyuan Zhou
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Xing Li
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jingquan Dong
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yingzhi Lu
- Department of Oncology, The Second People's Hospital of Lianyungang (The Oncology Hospital of Lianyungang), Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222000, China.
| | - Zibo Dong
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
2
|
Majer AD, Hua X, Katona BW. Menin in Cancer. Genes (Basel) 2024; 15:1231. [PMID: 39336822 PMCID: PMC11431421 DOI: 10.3390/genes15091231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
The protein menin is encoded by the MEN1 gene and primarily serves as a nuclear scaffold protein, regulating gene expression through its interaction with and regulation of chromatin modifiers and transcription factors. While the scope of menin's functions continues to expand, one area of growing investigation is the role of menin in cancer. Menin is increasingly recognized for its dual function as either a tumor suppressor or a tumor promoter in a highly tumor-dependent and context-specific manner. While menin serves as a suppressor of neuroendocrine tumor growth, as seen in the cancer risk syndrome multiple endocrine neoplasia type 1 (MEN1) syndrome caused by pathogenic germline variants in MEN1, recent data demonstrate that menin also suppresses cholangiocarcinoma, pancreatic ductal adenocarcinoma, gastric adenocarcinoma, lung adenocarcinoma, and melanoma. On the other hand, menin can also serve as a tumor promoter in leukemia, colorectal cancer, ovarian and endometrial cancers, Ewing sarcoma, and gliomas. Moreover, menin can either suppress or promote tumorigenesis in the breast and prostate depending on hormone receptor status and may also have mixed roles in hepatocellular carcinoma. Here, we review the rapidly expanding literature on the role and function of menin across a broad array of different cancer types, outlining tumor-specific differences in menin's function and mechanism of action, as well as identifying its therapeutic potential and highlighting areas for future investigation.
Collapse
Affiliation(s)
- Ariana D Majer
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xianxin Hua
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bryson W Katona
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
3
|
McClurg DP, Sanghera C, Mukherjee S, Fitzgerald RC, Jones CM. A systematic review of circulating predictive and prognostic biomarkers to aid the personalised use of radiotherapy in the radical treatment of patients with oesophageal cancer. Radiother Oncol 2024; 195:110224. [PMID: 38479442 DOI: 10.1016/j.radonc.2024.110224] [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: 01/12/2024] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND The availability of circulating biomarkers that are predictive of treatment response or prognostic of overall outcome could enable the personalised and adaptive use of radiotherapy (RT) in patients with oesophageal adenocarcinoma (OAC) and squamous cell carcinoma (OSCC). METHODS A systematic review was carried out following Preferred Reporting Items for Systematic Reviews guidance. Medline, EMBASE, PubMed, Cochrane Library, CINAHL, Scopus and the Web of Science databases were searched for studies published between January 2005-February 2023 relating to circulating biomarkers evaluated in the context of neoadjuvant or definitive RT delivered for OAC/OSCC. Study quality was assessed using predefined criteria. RESULTS A total of 3012 studies were screened and 57 subsequently included, across which 61 biomarkers were reported. A majority (43/57,75.4%) of studies were of Asian origin and retrospective (40/57, 70.2%), with most (52/57, 91.2%) biomarkers reported in the context of patients with OSCC. There was marked inter-study heterogeneity in patient populations, treatment characteristics, biomarker measurement and the cut points used to define biomarker positivity. Nevertheless, there is evidence for the prognostic and predictive value of circulating tumour DNA and numerous miRNAs in OAC and OSCC, as well as for the prognostic and predictive value of circulating levels of CYFRA21.1 in OSCC. CONCLUSIONS There is consistent evidence for the potential predictive and prognostic value of a small number of biomarkers in OSCC and OAC, though these data are insufficient for translation to current clinical practice. Well-designed prospective studies are now required to validate their role in stratified and personalised RT treatment approaches.
Collapse
Affiliation(s)
- Dylan P McClurg
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Chandan Sanghera
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Somnath Mukherjee
- Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Christopher M Jones
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Department of Oncology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
4
|
Zhang WT, Wang YJ, Zhang GX, Zhang YH, Gao SS. Diagnostic value of circulating microRNAs for esophageal cancer: a meta-analysis based on Asian data. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2023; 115:504-514. [PMID: 35040334 DOI: 10.17235/reed.2022.8348/2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND OBJECTIVE esophageal cancer (EC) is one of the most common gastrointestinal malignant diseases. We conducted a comprehensive meta-analysis to explore the clinical applicability of circulating microRNA for the diagnosis of EC. METHODS as of September 10, 2021, a comprehensive literature search was conducted on PubMed, Embase, Web of Science, Cochrane Library, Wanfang Database, and China National Knowledge Infrastructure (CNKI) to identify eligible studies. The sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), and area under the curve (AUC) were pooled to evaluate the test performance. The potential sources of heterogeneity were analyzed by subgroup analysis. Deeks' funnel plot was used to assess publication bias. RESULTS 85 studies from 50 articles were included in the current meta-analysis. The overall pooled sensitivity was 0.82 (95 % CI, 0.79-0.84), specificity was 0.84 (95 % CI, 0.81-0.86), PLR was 4.9 (95 % CI, 4.2-5.9), NLR was 0.22 (95 % CI, 0.19-0.25), DOR was 22 (95 % CI, 17-29) and AUC was 0.89 (95 % CI, 0.86-0.92), respectively. Subgroup analysis suggested that miRNA clusters with a large sample size showed better diagnostic accuracy. Publication bias was not found. CONCLUSIONS circulating miRNAs can be used as a potential non-invasive biomarker for the diagnosis of EC in Asian populations.
Collapse
|
5
|
The crucial choice of reference genes: identification of miR-191-5p for normalization of miRNAs expression in bone marrow mesenchymal stromal cell and HS27a/HS5 cell lines. Sci Rep 2020; 10:17728. [PMID: 33082452 PMCID: PMC7576785 DOI: 10.1038/s41598-020-74685-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
Bone marrow mesenchymal stromal cells (BM-MSCs) have a critical role in tissue regeneration and in the hematopoietic niche due to their differentiation and self-renewal capacities. These mechanisms are finely tuned partly by small non-coding microRNA implicated in post-transcriptional regulation. The easiest way to quantify them is RT-qPCR followed by normalization on validated reference genes (RGs). This study identified appropriate RG for normalization of miRNA expression in BM-MSCs and HS27a and HS5 cell lines in various conditions including normoxia, hypoxia, co-culture, as model for the hematopoietic niche and after induced differentiation as model for regenerative medicine. Six candidates, namely miR-16-5p, miR-34b-3p, miR-103a-3p, miR-191-5p, let-7a-5p and RNU6A were selected and their expression verified by RT-qPCR. Next, a ranking on stability of the RG candidates were performed with two algorithms geNorm and RefFinder and the optimal number of RGs needed to normalize was determined. Our results indicate miR-191-5p as the most stable miRNA in all conditions but also that RNU6a, usually used as RG is the less stable gene. This study demonstrates the interest of rigorously evaluating candidate miRNAs as reference genes and the importance of the normalization process to study the expression of miRNAs in BM-MSCs or derived cell lines.
Collapse
|
6
|
Wang S, Liu N, Tang Q, Sheng H, Long S, Wu W. MicroRNA-24 in Cancer: A Double Side Medal With Opposite Properties. Front Oncol 2020; 10:553714. [PMID: 33123467 PMCID: PMC7566899 DOI: 10.3389/fonc.2020.553714] [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: 04/20/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022] Open
Abstract
MicroRNA-24 (miR-24) has been widely studied in a variety of human cancers, which plays different roles in specific type of cancers. In the present review, we summarized the recent surveys regarding the role of miR-24 in different human cancers. On the one hand, miR-24 was reported to be down-regulated in some types of cancer, indicating its role as a tumor suppressor. On the other hand, it has shown that miR-24 was up-regulated in some other types of cancer, even in the same type of cancer, suggesting the role of miR-24 being as an oncogene. Firstly, miR-24 was dysregualted in human cancers, which is related to the clinical performance of cancer patients. Thus miR-24 could be used as a potential non-invasive diagnostic marker in human cancers. Secondly, miR-24 was associated with the tumor initiation and progression, being as a promoter or inhibitor. Therefore, miR-24 might be an effective prognostic biomarker in different type of cancers. Lastly, the abnormal expression of miR-24 was involved in the chemo- and radio- therapies of cancer patients, indicating the role of miR-24 being as a predictive biomarker to cancer treatment. Totally, miR-24 contributes to tumorigenesis, tumor progression, and tumor therapy, which closely related to clinic. The present review shows that miR-24 plays a double role in human cancers and provides plenty of evidences to apply miR-24 as a potential novel therapeutic target in treating human cancers.
Collapse
Affiliation(s)
- Sumei Wang
- Department of Oncology, Clinical and Basic Research Team of Traditional Chinese Medicine Prevention and Treatment of Non-Small Cell Lung Cancer, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Nayan Liu
- Department of Oncology, Clinical and Basic Research Team of Traditional Chinese Medicine Prevention and Treatment of Non-Small Cell Lung Cancer, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
- Guangdong Pharmaceutical University, Guangzhou, China
| | - Qing Tang
- Department of Oncology, Clinical and Basic Research Team of Traditional Chinese Medicine Prevention and Treatment of Non-Small Cell Lung Cancer, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Honghao Sheng
- Department of Oncology, Clinical and Basic Research Team of Traditional Chinese Medicine Prevention and Treatment of Non-Small Cell Lung Cancer, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Shunqin Long
- Department of Oncology, Clinical and Basic Research Team of Traditional Chinese Medicine Prevention and Treatment of Non-Small Cell Lung Cancer, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Wanyin Wu
- Department of Oncology, Clinical and Basic Research Team of Traditional Chinese Medicine Prevention and Treatment of Non-Small Cell Lung Cancer, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| |
Collapse
|
7
|
Abstract
PURPOSE Esophageal squamous cell carcinoma (ESCC) is one of the predominant types of esophageal cancer with poor prognosis which shows high prevalence in eastern countries. Studying microRNAs that were considered for their capabilities such as tissue-specific expression and involvement in different cell features may be informative in the field of diagnostic and prognostic tumor markers. The expression levels of miR-27a and miR-24-2 have been reported to be dysregulated in various cancers and contribute in tumorigenesis and progression; thus, evaluating their expressional behavior and its association with tumor states alteration in ESCC could potentially be helpful. METHODS The study was conducted on 30 fresh specimens including tumor and normal counterparts' tissues of ESCC. After the extraction of total RNA, complementary DNA synthesis was performed by the use of linear specific primers. Eventually, real-time polymerase chain reaction was carried out for the measurement of microRNAs expression level. RESULTS According to the obtained data, miR 27a and miR-24-2 were significantly upregulated (~2.5 fold, p < 0.05) in tumor specimens compared with their normal adjacent tissue; Moreover, upregulation of miR-27a and 24-2 showed cooperative relationship while analyzed. However, there was no correlation between clinicopathological features and microRNAs upregulation. CONCLUSIONS The results of this study show that miR-27a and miR-24-2 cooperatively upregulate in ESCC and suggest that these microRNAs can be introduced as a candidate for further study in the field of screening and prognostic biomarkers.
Collapse
|
8
|
Bellavia D, Salamanna F, Raimondi L, De Luca A, Carina V, Costa V, Alessandro R, Fini M, Giavaresi G. Deregulated miRNAs in osteoporosis: effects in bone metastasis. Cell Mol Life Sci 2019; 76:3723-3744. [PMID: 31147752 PMCID: PMC11105262 DOI: 10.1007/s00018-019-03162-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/22/2019] [Accepted: 05/28/2019] [Indexed: 12/19/2022]
Abstract
Starting from their role exerted on osteoblast and osteoclast differentiation and activity pathways, microRNAs (miRNAs) have been recently identified as regulators of different processes in bone homeostasis. For this purpose, in a recent review, we highlighted, as deregulated miRNAs could be involved in different bone diseases such as osteoporosis. In addition, recent studies supported the concept that osteoporosis-induced bone alterations might offer a receptive site for cancer cells to form bone metastases, However, to date, no data on specific-shared miRNAs between osteoporosis and bone metastases have been considered and described to clarify the evidence of this link. The main goal of this review is to underline as deregulated miRNAs in osteoporosis may have specific roles in the development of bone metastases. The review showed that several circulating osteoporotic miRNAs could facilitate tumor progression and bone-metastasis formation in several tumor types, i.e., breast cancer, prostate cancer, non-small-cell lung cancer, esophageal squamous cell carcinoma, and multiple myeloma. In detail, serum up-regulation of pro-osteoporotic miRNAs, as well as serum down-regulation of anti-osteoporotic miRNAs are common features of all these tumors and are able to promote bone metastasis. These results are of key importance and could help researcher and clinicians to establish new therapeutic strategies connected with deregulation of circulating miRNAs and able to interfere with pathogenic processes of osteoporosis, tumor progressions, and bone-metastasis formation.
Collapse
Affiliation(s)
| | - F Salamanna
- Laboratory of Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - L Raimondi
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - A De Luca
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - V Carina
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - V Costa
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - R Alessandro
- Section of Biology and Genetics, Department of BioMedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), University of Palermo, 90133, Palermo, Italy
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council, Palermo, Italy
| | - M Fini
- Laboratory of Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - G Giavaresi
- Laboratory of Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
9
|
Yao C, Liu HN, Wu H, Chen YJ, Li Y, Fang Y, Shen XZ, Liu TT. Diagnostic and Prognostic Value of Circulating MicroRNAs for Esophageal Squamous Cell Carcinoma: a Systematic Review and Meta-analysis. J Cancer 2018; 9:2876-2884. [PMID: 30123356 PMCID: PMC6096380 DOI: 10.7150/jca.25351] [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: 02/04/2018] [Accepted: 06/11/2018] [Indexed: 01/08/2023] Open
Abstract
Background and Aim: MicroRNAs, dysregulated in the circulation of esophageal squamous cell carcinoma (ESCC) patient, have been assumed to be with great potential in the diagnosis and prognosis of esophageal cancer. We aimed to review previous articles on ESCC. Methods: A search of electronic databases was performed before Nov 12, 2017. We summarized the identification of microRNA imbalance in the blood of ESCC compared with the healthy controls, with the objective to evaluate the efficiency of microRNAs in diagnosing and forecasting ESCC. Results: A total of 35 studies investigating plasma or serum microRNAs were included in the meta-analysis. Based on the consequences of the quality assessment of each study, the articles involved were appropriate for quantitative synthesis. For diagnostic meta-analysis. The overall pooled sensitivity, specificity, and area under the curve of circulating microRNA is 0.794 (95% CI: 0.765 - 0.820), 0.779 (95%CI: 0.746 - 0.808), 0.86 (95%CI: 0.82 - 0.88). The diagnostic value of each microRNA was calculated respectively. For prognostic meta-analysis, the overall pooled hazard ratios of higher microRNA expression in circulation was 1.34 (95% CI: 1.14-1.58), which could significantly predict poorer survival in ESCC. Conclusions: Circulating microRNAs distinguish patients with ESCC from healthy controls with high sensitivity and specificity, compared to other invasive currently used screening methods. Simultaneously, there was prognostic value for the prognosis of ESCC.
Collapse
Affiliation(s)
- Can Yao
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Hai-Ning Liu
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Hao Wu
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yan-Jie Chen
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yu Li
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Ying Fang
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Xi-Zhong Shen
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China.,Shanghai Institute of Liver Diseases, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Tao-Tao Liu
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China
| |
Collapse
|
10
|
Komatsu S, Ichikawa D, Kawaguchi T, Takeshita H, Miyamae M, Ohashi T, Okajima W, Imamura T, Kiuchi J, Arita T, Konishi H, Shiozaki A, Fujiwara H, Okamoto K, Otsuji E. Plasma microRNA profiles: identification of miR-23a as a novel biomarker for chemoresistance in esophageal squamous cell carcinoma. Oncotarget 2018; 7:62034-62048. [PMID: 27566562 PMCID: PMC5308709 DOI: 10.18632/oncotarget.11500] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 08/10/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND This study aims to explore novel microRNAs in plasma for predicting chemoresistance in preoperative chemotherapy of patients with esophageal squamous cell carcinoma (ESCC) using a microRNA array-based approach. RESULTS (1) Four candidate microRNAs (miR-223, 103a, 23b and 23a), which were highly expressed in the pretreatment plasma of patients with a low histopathologic response, were selected. (2) In a large-scale validation analysis by quantitative RT–PCR, plasma levels of miR-223, miR-23b and miR-23a were significantly higher in patients with a low histopathologic response than in those with a high histopathologic response (p = 0.0345, p = 0.0125 and p = 0.0114). (3) Of all candidate microRNAs, miR-23a expression of pretreatment ESCC tumor tissues was significantly higher in ESCC patients with a low histopathologic response than in those with a high histopathologic response (p = 0.0278). (4) After overexpressing each candidate in ESCC cells, miR-23a induced significant chemoresistance to both 5-fluorouracil and cisplatin, and miR-223 to cisplatin in vitro. (5) A high level of plasma miR-23a, which tended to correlate with lymphatic invasion (p = 0.0808) and deep depth of invasion (p = 0.0658), was an independent risk factor for chemoresistance in ESCC (p = 0.0222; odds ratio: 12.4; range 1.46–105). MATERIALS AND METHODS We used the Toray® 3D-Gene microRNA array-based approach to compare plasma microRNA levels between patients with a high or a low histopathologic response to chemotherapy. All patients underwent a preoperative chemotherapy regimen with cisplatin plus 5-fluorouracil. CONCLUSIONS Plasma miR-23a might be a useful biomarker for predicting chemoresistance in ESCC patients.
Collapse
Affiliation(s)
- Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Daisuke Ichikawa
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Tsutomu Kawaguchi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hiroki Takeshita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Mahito Miyamae
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Takuma Ohashi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Wataru Okajima
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Taisuke Imamura
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Jun Kiuchi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Kazuma Okamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kawaramachihirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
11
|
Fong LY, Taccioli C, Jing R, Smalley KJ, Alder H, Jiang Y, Fadda P, Farber JL, Croce CM. MicroRNA dysregulation and esophageal cancer development depend on the extent of zinc dietary deficiency. Oncotarget 2017; 7:10723-38. [PMID: 26918602 PMCID: PMC4905434 DOI: 10.18632/oncotarget.7561] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/08/2016] [Indexed: 12/21/2022] Open
Abstract
Zinc deficiency (ZD) increases the risk of esophageal squamous cell carcinoma (ESCC), and marginal ZD is prevalent in humans. In rats, marked-ZD (3 mg Zn/kg diet) induces a proliferative esophagus with a 5-microRNA signature (miR-31, -223, -21, -146b, -146a) and promotes ESCC. Here we report that moderate and mild-ZD (6 and 12 mg Zn/kg diet) also induced esophageal hyperplasia, albeit less pronounced than induced by marked-ZD, with a 2-microRNA signature (miR-31, -146a). On exposure to an environmental carcinogen, ∼16% of moderate/mild-ZD rats developed ESCC, a cancer incidence significantly greater than for Zn-sufficient rats (0%) (P ≤ 0.05), but lower than marked-ZD rats (68%) (P < 0.001). Importantly, the high ESCC, marked-ZD esophagus had a 15-microRNA signature, resembling the human ESCC miRNAome, with miR-223, miR-21, and miR-31 as the top-up-regulated species. This signature discriminated it from the low ESCC, moderate/mild-ZD esophagus, with a 2-microRNA signature (miR-31, miR-223). Additionally, Fbxw7, Pdcd4, and Stk40 (tumor-suppressor targets of miR-223, -21, and -31) were downregulated in marked-ZD cohort. Bioinformatics analysis predicted functional relationships of the 3 tumor-suppressors with other cancer-related genes. Thus, microRNA dysregulation and ESCC progression depend on the extent of dietary Zn deficiency. Our findings suggest that even moderate ZD may promote esophageal cancer and dietary Zn has preventive properties against ESCC. Additionally, the deficiency-associated miR-223, miR-21, and miR-31 may be useful therapeutic targets in ESCC.
Collapse
Affiliation(s)
- Louise Y Fong
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Cristian Taccioli
- Animal Medicine, Production and Health Department, University of Padua, Padua, Italy
| | - Ruiyan Jing
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Karl J Smalley
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Hansjuerg Alder
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Yubao Jiang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Paolo Fadda
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - John L Farber
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Carlo M Croce
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
12
|
Kennedy L, Hargrove L, Demieville J, Francis N, Seils R, Villamaria S, Francis H. Recent Advances in Understanding Cholangiocarcinoma. F1000Res 2017; 6:1818. [PMID: 29067165 PMCID: PMC5635438 DOI: 10.12688/f1000research.12118.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/10/2017] [Indexed: 12/13/2022] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive malignancy that arises from damaged epithelial cells, cholangiocytes, and possibly de-differentiated hepatocytes. CCA has a poor overall survival rate and limited therapeutic options. Based on this data, it is imperative that new diagnostic and therapeutic interventions be developed. Recent work has attempted to understand the pathological mechanisms driving CCA progression. Specifically, recent publications have delved into the role of cancer stem cells (CSCs), mesenchymal stem cells (MSCs), and microRNAs (miRNAs) during CCA pathology. CSCs are a specific subset of cells within the tumor environment that are derived from a cell with stem-like properties and have been shown to influence recurrence and chemoresistance during CCA. MSCs are known for their anti-inflammatory activity and have been postulated to influence malignancy during CCA, but little is known about their exact functions. miRNAs exert various functions via gene regulation at both the transcriptional and the translational levels, giving miRNAs diverse roles in CCA progression. Additionally, current miRNA-based therapeutic approaches are in clinical trials for various liver diseases, giving hope for similar approaches for CCA. However, the interactions among these three factors in the context of CCA are unknown. In this review, we focus on recently published data (within the last 3 years) that discuss the role of CSCs, MSCs, and miRNAs and their possible interactions during CCA pathogenesis.
Collapse
Affiliation(s)
- Lindsey Kennedy
- Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, TX, USA.,Research, Central Texas Veterans Health Care System, Temple, TX, USA
| | - Laura Hargrove
- Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, TX, USA
| | | | - Nicole Francis
- Baylor Scott & White Health Digestive Disease Research Center, Temple, TX, USA
| | - Rowan Seils
- Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, TX, USA
| | - Sara Villamaria
- Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, TX, USA
| | - Heather Francis
- Department of Internal Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, TX, USA.,Research, Central Texas Veterans Health Care System, Temple, TX, USA.,Baylor Scott & White Health Digestive Disease Research Center, Temple, TX, USA
| |
Collapse
|
13
|
Zhang D, Zhang W, Liu W, Mao Y, Fu Z, Liu J, Huang W, Zhang Z, An D, Li B. Human papillomavirus infection increases the chemoradiation response of esophageal squamous cell carcinoma based on P53 mutation. Radiother Oncol 2017. [DOI: 10.1016/j.radonc.2017.06.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
14
|
Ehrlich L, Hall C, Venter J, Dostal D, Bernuzzi F, Invernizzi P, Meng F, Trzeciakowski JP, Zhou T, Standeford H, Alpini G, Lairmore TC, Glaser S. miR-24 Inhibition Increases Menin Expression and Decreases Cholangiocarcinoma Proliferation. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:570-580. [PMID: 28087162 DOI: 10.1016/j.ajpath.2016.10.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/04/2016] [Accepted: 10/25/2016] [Indexed: 12/15/2022]
Abstract
Menin (MEN1) is a tumor-suppressor protein in neuroendocrine tissue. Therefore, we tested the novel hypothesis that menin regulates cholangiocarcinoma proliferation. Menin and miR-24 expression levels were measured in the following intrahepatic and extrahepatic cholangiocarcinoma (CCA) cell lines, Mz-ChA-1, TFK-1, SG231, CCLP, HuCCT-1, and HuH-28, as well as the nonmalignant human intrahepatic biliary line, H69. miR-24 miRNA and menin protein levels were manipulated in vitro in Mz-ChA-1 cell lines. Markers of proliferation and angiogenesis (Ki-67, vascular endothelial growth factors A/C, vascular endothelial growth factor receptors 2/3, angiopoietin 1/2, and angiopoietin receptors 1/2) were evaluated. Mz-ChA-1 cells were injected into the flanks of nude mice and treated with miR-24 inhibitor or inhibitor scramble. Menin expression was decreased in advanced CCA specimens, whereas miR-24 expression was increased in CCA. Menin overexpression decreased proliferation, angiogenesis, migration, and invasion. Inhibition of miR-24 increased menin protein expression while decreasing proliferation, angiogenesis, migration, and invasion. miR-24 was shown to negatively regulate menin expression by luciferase assay. Tumor burden and expression of proliferative and angiogenic markers was decreased in the miR-24 inhibited tumor group compared to controls. Interestingly, treated tumors were more fibrotic than the control group. miR-24-dependent expression of menin may be important in the regulation of nonmalignant and CCA proliferation and may be an additional therapeutic tool for managing CCA progression.
Collapse
Affiliation(s)
- Laurent Ehrlich
- Department of Medicine, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas; Division of Gastroenterology and Medical Physiology, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas
| | - Chad Hall
- Division of Surgery, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas
| | - Julie Venter
- Department of Medicine, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas
| | - David Dostal
- Division of Gastroenterology and Medical Physiology, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas
| | - Francesca Bernuzzi
- Department of Medicine and Surgery, Program for Autoimmune Liver Diseases, International Center for Digestive Diseases, University of Milan-Bicocca, Milan, Italy; Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano (Milan), Italy
| | - Pietro Invernizzi
- Department of Medicine and Surgery, Program for Autoimmune Liver Diseases, International Center for Digestive Diseases, University of Milan-Bicocca, Milan, Italy; Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano (Milan), Italy
| | - Fanyin Meng
- Department of Medicine, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas; Division of Gastroenterology and Medical Physiology, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas; Research Section, Central Texas Veterans Health Care System, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas; Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas
| | - Jerome P Trzeciakowski
- Division of Gastroenterology and Medical Physiology, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas
| | - Tianhao Zhou
- Department of Medicine, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas
| | - Holly Standeford
- Research Section, Central Texas Veterans Health Care System, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas
| | - Gianfranco Alpini
- Department of Medicine, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas; Research Section, Central Texas Veterans Health Care System, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas; Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas
| | - Terry C Lairmore
- Division of Surgery, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas
| | - Shannon Glaser
- Department of Medicine, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas; Research Section, Central Texas Veterans Health Care System, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas; Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White and Texas A&M University Health Science Center, Temple, Texas.
| |
Collapse
|
15
|
Abstract
MicroRNAs (miRNA) are 22-nucleotide non-coding RNAs that post-transcriptionally regulate gene expression by base pairing to partially complementary sequences in the 3'-untranslated region of their target messenger RNA. Altered miRNA expression also changes the expression of oncogenes and tumor suppressors, affecting the proliferation, apoptosis, motility and invasibility of gastrointestinal cancer cells, including the cells of esophageal squamous cell carcinoma (ESCC). It has been suggested that various miRNA expression profiles may provide useful biomarkers and therapeutic targets, but to date few studies have been published on the role of miRNA in ESCC. In this review we summarize the identification and characterization of miRNAs involved in ESCC and discuss their potential as biomarkers and therapeutic targets.
Collapse
|
16
|
Wang C, Li Q, Liu F, Chen X, Nesa EU, Guan S, Liu B, Han L, Tan B, Wang D, Chen P, Liu X, Zhang H, Sun Y, Cheng Y. Serum miR-1297: a promising diagnostic biomarker in esophageal squamous cell carcinoma. Biomarkers 2016; 21:517-22. [PMID: 27152453 DOI: 10.3109/1354750x.2016.1160291] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We aimed to value the diagnostic potential of serum miR-1297 in esophageal squamous cell cancer (ESCC). Its expression level was detected in 156 pairs of patients with ESCC and healthy volunteers using quantitative real-time polymerase chain reaction (qRT-PCR) method. It was statistically decreased in ESCC patients compared with healthy controls. AUC based on serum miR-1297 was 0.840 ± 0.035 in discovery group and 0.837 ± 0.034 in validation group. Further analysis on early-stage patients revealed that the AUC was 0.819 ± 0.053 in discovery group and 0.814 ± 0.044 in validation group. Its sensitivity and specificity were promising. In conclusion, serum miR-1297 can serve as an ideal indicator for the diagnosis of ESCC.
Collapse
Affiliation(s)
- Cong Wang
- a Department of Radiation Oncology , Qilu Hospital of Shandong University , Jinan , Shandong , People's Republic of China
| | - Qingbao Li
- b Department of Cardiac Surgery , Shandong Provincial Hospital Affiliated to Shandong University , Jinan , Shandong , People's Republic of China
| | - Fang Liu
- c Department of Imaging , Shandong Medical College , Jinan , Shandong , People's Republic of China
| | - Xuan Chen
- a Department of Radiation Oncology , Qilu Hospital of Shandong University , Jinan , Shandong , People's Republic of China
| | - Effat Un Nesa
- a Department of Radiation Oncology , Qilu Hospital of Shandong University , Jinan , Shandong , People's Republic of China
| | - Shanghui Guan
- a Department of Radiation Oncology , Qilu Hospital of Shandong University , Jinan , Shandong , People's Republic of China
| | - Bowen Liu
- a Department of Radiation Oncology , Qilu Hospital of Shandong University , Jinan , Shandong , People's Republic of China
| | - Lihui Han
- a Department of Radiation Oncology , Qilu Hospital of Shandong University , Jinan , Shandong , People's Republic of China
| | - Bingxu Tan
- a Department of Radiation Oncology , Qilu Hospital of Shandong University , Jinan , Shandong , People's Republic of China
| | - Ding Wang
- d Department of Clinical Laboratory , Qilu Hospital of Shandong University , Jinan , Shandong , People's Republic of China
| | - Pengxiang Chen
- a Department of Radiation Oncology , Qilu Hospital of Shandong University , Jinan , Shandong , People's Republic of China
| | - Xiaoyue Liu
- a Department of Radiation Oncology , Qilu Hospital of Shandong University , Jinan , Shandong , People's Republic of China
| | - Han Zhang
- a Department of Radiation Oncology , Qilu Hospital of Shandong University , Jinan , Shandong , People's Republic of China
| | - Ying Sun
- a Department of Radiation Oncology , Qilu Hospital of Shandong University , Jinan , Shandong , People's Republic of China
| | - Yufeng Cheng
- a Department of Radiation Oncology , Qilu Hospital of Shandong University , Jinan , Shandong , People's Republic of China
| |
Collapse
|
17
|
DONG WEI, LI BAOSHENG, WANG JUAN, SONG YIPENG, ZHANG ZICHENG, FU CHENGRUI, ZHANG PEILIANG. Diagnostic and predictive significance of serum microRNA-7 in esophageal squamous cell carcinoma. Oncol Rep 2015; 35:1449-56. [DOI: 10.3892/or.2015.4499] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 11/05/2015] [Indexed: 11/06/2022] Open
|
18
|
Guan S, Wang C, Chen X, Liu B, Tan B, Liu F, Wang D, Han L, Wang L, Huang X, Wang J, Yao B, Shi J, Chen P, Nesa EU, Song Q, Cheng Y. MiR-613: a novel diagnostic and prognostic biomarker for patients with esophageal squamous cell carcinoma. Tumour Biol 2015; 37:4383-91. [PMID: 26499784 DOI: 10.1007/s13277-015-4271-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 10/15/2015] [Indexed: 12/29/2022] Open
Abstract
MicroRNA-613 (miR-613) plays important roles in tumorigenesis and cancer progression. We aimed to evaluate its expression level and potential for diagnosis and prognosis in esophageal squamous cell cancer (ESCC). We examined miR-613 expression in 60 pairs of ESCC cancerous and matched paracancerous tissues, serum samples from 75 ESCC patients and 75 healthy volunteers, and 105 formalin-fixed paraffin-embedded (FFPE) tissue samples using quantitative reverse transcription polymerase chain reaction. Receiver-operating characteristic (ROC) curve analysis, Kaplan-Meier method, and Cox regression were applied to analyze its diagnostic and prognostic value. MiR-613 was significantly decreased in ESCC tissue compared with paracancerous tissue (P < 0.001). Moreover, the expression level of miR-613 was significantly reduced with increased T stage of ESCC. Statistically significant difference between ESCC patients and healthy controls in expression level of miR-613 (0.89 ± 0.73 vs. 1.71 ± 1.03, P < 0.001) was found. The area under the ROC curve (AUC) based on serum miR-613 was 0.767 ± 0.040. We also performed analysis on early-stage patients and revealed that the AUC value was 0.728 ± 0.052 (P < 0.001). The Kaplan-Meier curve revealed that the downregulation of miR-613 was related to worse overall survival (OS) and progression-free survival (PFS) of ESCC patients (P = 0.018 and P = 0.035, respectively). Furthermore, the multivariate analysis identified miR-613 to be an independent prognostic factor for OS and PFS (P = 0.031 and P = 0.006, respectively) In conclusion, miR-613 is significantly reduced in cancerous tissue and serum samples of ESCC patients. It can serve as an ideal indicator for the diagnosis and prognosis of ESCC.
Collapse
Affiliation(s)
- Shanghui Guan
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Cong Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Xuan Chen
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Bowen Liu
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Bingxu Tan
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Fang Liu
- Department of Image, Shandong Medical College, Jinan, Shandong, 250002, People's Republic of China
| | - Ding Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Lihui Han
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Lu Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Xiaochen Huang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Jiangfeng Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Bin Yao
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Jialei Shi
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Pengxiang Chen
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Effat Un Nesa
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Qingxu Song
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China.
| |
Collapse
|
19
|
Schwarzenbach H, da Silva AM, Calin G, Pantel K. Data Normalization Strategies for MicroRNA Quantification. Clin Chem 2015; 61:1333-42. [PMID: 26408530 DOI: 10.1373/clinchem.2015.239459] [Citation(s) in RCA: 346] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 08/25/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Different technologies, such as quantitative real-time PCR or microarrays, have been developed to measure microRNA (miRNA) expression levels. Quantification of miRNA transcripts implicates data normalization using endogenous and exogenous reference genes for data correction. However, there is no consensus about an optimal normalization strategy. The choice of a reference gene remains problematic and can have a serious impact on the actual available transcript levels and, consequently, on the biological interpretation of data. CONTENT In this review article we discuss the reliability of the use of small RNAs, commonly reported in the literature as miRNA expression normalizers, and compare different strategies used for data normalization. SUMMARY A workflow strategy is proposed for normalization of miRNA expression data in an attempt to provide a basis for the establishment of a global standard procedure that will allow comparison across studies.
Collapse
Affiliation(s)
- Heidi Schwarzenbach
- Department of Tumour Biology, Center of Experimental Medicine, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreia Machado da Silva
- Department of Experimental Therapeutics and The Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX; Instituto de Investigação em Saúde, Universidade do Porto, Porto, Portugal; INEB, Institute of Biomedical Engineering, Universidade do Porto, Porto, Portugal
| | - George Calin
- Department of Experimental Therapeutics and The Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Klaus Pantel
- Department of Tumour Biology, Center of Experimental Medicine, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany;
| |
Collapse
|