1
|
Doghish AS, El-Husseiny AA, Abdelmaksoud NM, El-Mahdy HA, Elsakka EGE, Abdel Mageed SS, Mahmoud AMA, Raouf AA, Elballal MS, El-Dakroury WA, AbdelRazek MMM, Noshy M, El-Husseiny HM, Abulsoud AI. The interplay of signaling pathways and miRNAs in the pathogenesis and targeted therapy of esophageal cancer. Pathol Res Pract 2023; 246:154529. [PMID: 37196470 DOI: 10.1016/j.prp.2023.154529] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023]
Abstract
Globally, esophageal cancer (EC) is the 6th leading cause of cancer-related deaths and the second deadliest gastrointestinal cancer. Multiple genetic and epigenetic factors, such as microRNAs (miRNAs), influence its onset and progression. miRNAs are short nucleic acid molecules that can regulate multiple cellular processes by regulating gene expression. Therefore, EC initiation, progression, apoptosis evasions, invasion capacity, promotion, angiogenesis, and epithelial-mesenchymal transition (EMT) enhancement are associated with miRNA expression dysregulation. Wnt/-catenin signaling, Mammalian target of rapamycin (mTOR)/P-gp, phosphoinositide-3-kinase (PI3K)/AKT/c-Myc, epidermal growth factor receptor (EGFR), and transforming growth factor (TGF)-β signaling are crucial pathways in EC that are controlled by miRNAs. This review was conducted to provide an up-to-date assessment of the role of microRNAs in EC pathogenesis and their modulatory effects on responses to various EC treatment modalities.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| | - Ahmed A El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt; Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City 11829, Cairo, Egypt
| | - Nourhan M Abdelmaksoud
- Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| | - Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Abdulla M A Mahmoud
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ahmed Amr Raouf
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mohamed M M AbdelRazek
- Department of Pharmacognosy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mina Noshy
- Clinical Pharmacy Department, Faculty of Pharmacy, King Salman International University (KSIU), SouthSinai, Ras Sudr 46612, Egypt
| | - Hussein M El-Husseiny
- Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan; Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt; Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| |
Collapse
|
2
|
Noncoding RNAs in esophageal cancer: A glimpse into implications for therapy resistance. Pharmacol Res 2023; 188:106678. [PMID: 36709789 DOI: 10.1016/j.phrs.2023.106678] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/09/2023] [Accepted: 01/25/2023] [Indexed: 01/27/2023]
Abstract
Esophageal cancer (EC) is one of the most common malignancies of the digestive system and has a high morbidity and mortality worldwide. Chemotherapy in combination with radiotherapy is one of the most important treatment modalities for EC. Chemoradiotherapy is currently acknowledged worldwide as being the standard treatment for locally advanced or unresectable disease. Unfortunately, due to the existence of therapy resistance, a number of EC patients fail to benefit from drug or irradiation treatment, which ultimately leads to poor outcomes. Considerable efforts have been made to explore the mechanisms underlying the therapy resistance of EC. Notably, noncoding RNAs (ncRNAs), including microRNAs (miRNAs), long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs), are current research areas for the modulation of therapy responses and may serve as new targets to overcome treatment resistance in EC. Herein, we summarized the mechanisms by which ncRNAs are involved in drug and radiation resistance in EC and highlighted their role in promoting or repressing treatment resistance. Additionally, we discussed the clinical relevance of ncRNAs, which may serve as potential therapeutic targets and predictive biomarkers for EC.
Collapse
|
3
|
Mo Y, Leung LL, Mak CSL, Wang X, Chan WS, Hui LMN, Tang HWM, Siu MKY, Sharma R, Xu D, Tsui SKW, Ngan HYS, Yung MMH, Chan KKL, Chan DW. Tumor-secreted exosomal miR-141 activates tumor-stroma interactions and controls premetastatic niche formation in ovarian cancer metastasis. Mol Cancer 2023; 22:4. [PMID: 36624516 PMCID: PMC9827705 DOI: 10.1186/s12943-022-01703-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Metastatic colonization is one of the critical steps in tumor metastasis. A pre-metastatic niche is required for metastatic colonization and is determined by tumor-stroma interactions, yet the mechanistic underpinnings remain incompletely understood. METHODS PCR-based miRNome profiling, qPCR, immunofluorescent analyses evaluated the expression of exosomal miR-141 and cell-to-cell communication. LC-MS/MS proteomic profiling and Dual-Luciferase analyses identified YAP1 as the direct target of miR-141. Human cytokine profiling, ChIP, luciferase reporter assays, and subcellular fractionation analyses confirmed YAP1 in modulating GROα production. A series of in vitro tumorigenic assays, an ex vivo model and Yap1 stromal conditional knockout (cKO) mouse model demonstrated the roles of miR-141/YAP1/GROα/CXCR1/2 signaling cascade. RNAi, CRISPR/Cas9 and CRISPRi systems were used for gene silencing. Blood sera, OvCa tumor tissue samples, and tissue array were included for clinical correlations. RESULTS Hsa-miR-141-3p (miR-141), an exosomal miRNA, is highly secreted by ovarian cancer cells and reprograms stromal fibroblasts into proinflammatory cancer-associated fibroblasts (CAFs), facilitating metastatic colonization. A mechanistic study showed that miR-141 targeted YAP1, a critical effector of the Hippo pathway, reducing the nuclear YAP1/TAZ ratio and enhancing GROα production from stromal fibroblasts. Stromal-specific knockout (cKO) of Yap1 in murine models shaped the GROα-enriched microenvironment, facilitating in vivo tumor colonization, but this effect was reversed after Cxcr1/2 depletion in OvCa cells. The YAP1/GROα correlation was demonstrated in clinical samples, highlighting the clinical relevance of this research and providing a potential therapeutic intervention for impeding premetastatic niche formation and metastatic progression of ovarian cancers. CONCLUSIONS This study uncovers miR-141 as an OvCa-derived exosomal microRNA mediating the tumor-stroma interactions and the formation of tumor-promoting stromal niche through activating YAP1/GROα/CXCRs signaling cascade, providing new insight into therapy for OvCa patients with peritoneal metastases.
Collapse
Affiliation(s)
- Yulan Mo
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Leanne L. Leung
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Celia S. L. Mak
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Xueyu Wang
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Wai-Sun Chan
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Lynn M. N. Hui
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Hermit W. M. Tang
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Michelle K. Y. Siu
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Rakesh Sharma
- grid.194645.b0000000121742757Centre for PanorOmic Sciences Proteomics and Metabolomics Core, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Dakang Xu
- grid.16821.3c0000 0004 0368 8293Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Stephen K. W. Tsui
- grid.10784.3a0000 0004 1937 0482School of Biomedical Sciences, The Chinese University of Hong Kong, SAR Hong Kong, China
| | - Hextan Y. S. Ngan
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Mingo M. H. Yung
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Karen K. L. Chan
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - David W. Chan
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China ,grid.10784.3a0000 0004 1937 0482School of Biomedical Sciences, The Chinese University of Hong Kong, SAR Hong Kong, China ,grid.511521.3School of Medicine, The Chinese University of Hong Kong-Shenzhen, Shenzhen, 518172 China
| |
Collapse
|
4
|
Negahdary M, Angnes L. Application of electrochemical biosensors for the detection of microRNAs (miRNAs) related to cancer. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214565] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
5
|
Chang N, Ge N, Zhao Y, Yang L, Qin W, Cui Y. Hsa_circ_0007142 contributes to cisplatin resistance in esophageal squamous cell carcinoma via miR‐494‐3p/LASP1 axis. J Clin Lab Anal 2022; 36:e24304. [PMID: 35312115 PMCID: PMC9102771 DOI: 10.1002/jcla.24304] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 01/21/2023] Open
Abstract
Background Methods Results Conclusion
Collapse
Affiliation(s)
- Na Chang
- Department of Cancer Radiotherapy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Science and Technology of China (Anhui Provincial Cancer Hospital) Hefei Anhui China
| | - Ning Ge
- Department of Cancer Radiotherapy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Science and Technology of China (Anhui Provincial Cancer Hospital) Hefei Anhui China
| | - Yufei Zhao
- Department of Cancer Radiotherapy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Science and Technology of China (Anhui Provincial Cancer Hospital) Hefei Anhui China
| | - Liu Yang
- Department of Cancer Radiotherapy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Science and Technology of China (Anhui Provincial Cancer Hospital) Hefei Anhui China
| | - Wei Qin
- Department of Science and Education The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Science and Technology of China (Anhui provincial Cancer Hospital) Hefei Anhui China
| | - Yayun Cui
- Department of Cancer Radiotherapy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Science and Technology of China (Anhui Provincial Cancer Hospital) Hefei Anhui China
| |
Collapse
|
6
|
Yes-associated protein 1 exerts its tumor-promoting effects and increases cisplatin resistance in tongue squamous cell carcinoma cells by dysregulating Hippo signal pathway. Anticancer Drugs 2022; 33:352-361. [DOI: 10.1097/cad.0000000000001269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
7
|
Li Y, Zu L, Wu H, Zhang F, Fan Y, Pan H, Du X, Guo F, Zhou Q. MiR-192/NKRF axis confers lung cancer cell chemoresistance to cisplatin via the NF-κB pathway. Thorac Cancer 2021; 13:430-441. [PMID: 34953057 PMCID: PMC8807278 DOI: 10.1111/1759-7714.14278] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 02/05/2023] Open
Abstract
Background Chemoresistance influences the therapeutic effect of cisplatin and remains a major obstacle to its clinical use. MicroRNAs are associated with drug resistance of various tumors. However, the association between microRNAs and cisplatin in lung cancer remains largely unclear. Methods MicroRNA expression profile was identified by microRNA microarray between the lung cancer cisplatin‐sensitive cell line A549 (A549) and cisplatin‐resistant cell line A549/DDP (A549/DDP) and confirmed by quantitative real‐time‐PCR (qRT‐PCR). In vitro loss‐ and gain‐of‐function studies were performed to reveal the biological function of miR‐192 and related mechanism of the microRNA‐192/NKRF axis in lung cancer cell cisplatin resistance. Results Increased miR‐192 expression was detected in A549/DDP cells compared to A549. High miR‐192 expression significantly suppressed apoptosis, enhanced proliferation, and conferred resistance to cisplatin in lung cancer cells. NF‐κB repressing factor (NKRF), which is involved in the regulation of the NF‐κB signaling pathway, was identified as a direct target of miR‐192. Overexpression of miR‐192 significantly increased the nuclear protein amount and transcriptional activation of NF‐κB and expression of cIAP1, cIAP2, Bcl‐xl and XIAP, whereas decreased miR‐192 expression did the opposite. Inhibition of the NF‐κB signal pathway by curcumin reversed the effect of upregulation of miR‐192 on proliferation, apoptosis and cisplatin‐resistance in lung cancer cells. These results indicated that miR‐192/ NKRF axis enhances the cisplatin resistance of lung cancer cells through activating the NF‐κB pathway in vitro. Conclusions MiR‐192 plays a crucial role in cisplatin‐resistance of lung cancer cells. Thus, MiR‐192 may represent a therapeutic target for overcoming resistance to cisplatin‐based chemotherapy in lung cancer.
Collapse
Affiliation(s)
- Yang Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Lingling Zu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Heng Wu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Fang Zhang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yaguang Fan
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongli Pan
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinxin Du
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Fengjie Guo
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Qinghua Zhou
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China.,Sichuan Lung Cancer Institute, Sichuan Lung Cancer Center, West China Hospital, Chengdu, Sichuan University, China
| |
Collapse
|
8
|
Wei L, Sun J, Zhang N, Shen Y, Wang T, Li Z, Yang M. Novel Implications of MicroRNAs, Long Non-coding RNAs and Circular RNAs in Drug Resistance of Esophageal Cancer. Front Cell Dev Biol 2021; 9:764313. [PMID: 34881242 PMCID: PMC8645845 DOI: 10.3389/fcell.2021.764313] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022] Open
Abstract
Esophageal cancer is the eighth most common malignancy and the sixth leading cause of cancer-related deaths worldwide. Chemotherapy based on platinum drugs, 5-fluorouracil, adriamycin, paclitaxel, gemcitabine, and vinorelbine, as well as targeted treatment and immunotherapy with immune checkpoint inhibitors improved the prognosis in a portion of patients with advanced esophageal cancer. Unfortunately, a number of esophageal cancer patients develop drug resistance, resulting in poor outcomes. Multiple mechanisms contributing to drug resistance of esophageal cancer have been reported. Notably, non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), have been identified to play crucial roles in modulating esophageal cancer drug resistance. In the present review, we highlight the underlying mechanisms how miRNAs, lncRNAs, and circRNAs impact the drug resistance of esophageal cancer. Several miRNAs, lncRNAs, and circRNAs may have potential clinical implications as novel biomarkers and therapeutic targets for esophageal cancer.
Collapse
Affiliation(s)
- Ling Wei
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jujie Sun
- Department of Pathology, Shandong Cancer Hospital and Institute, Jinan, China
| | - Nasha Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| | - Yue Shen
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Teng Wang
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zengjun Li
- Department of Endoscopy, Shandong Cancer Hospital and Institute, Jinan, China
| | - Ming Yang
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
9
|
Dashti F, Mirazimi SMA, Rabiei N, Fathazam R, Rabiei N, Piroozmand H, Vosough M, Rahimian N, Hamblin MR, Mirzaei H. The role of non-coding RNAs in chemotherapy for gastrointestinal cancers. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:892-926. [PMID: 34760336 PMCID: PMC8551789 DOI: 10.1016/j.omtn.2021.10.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gastrointestinal (GI) cancers, including colorectal, gastric, hepatic, esophageal, and pancreatic tumors, are responsible for large numbers of deaths around the world. Chemotherapy is the most common approach used to treat advanced GI cancer. However, chemoresistance has emerged as a critical challenge that prevents successful tumor elimination, leading to metastasis and recurrence. Chemoresistance mechanisms are complex, and many factors and pathways are involved. Among these factors, non-coding RNAs (ncRNAs) are critical regulators of GI tumor development and subsequently can induce resistance to chemotherapy. This occurs because ncRNAs can target multiple signaling pathways, affect downstream genes, and modulate proliferation, apoptosis, tumor cell migration, and autophagy. ncRNAs can also induce cancer stem cell features and affect the epithelial-mesenchymal transition. Thus, ncRNAs could possibly act as new targets in chemotherapy combinations to treat GI cancer and to predict treatment response.
Collapse
Affiliation(s)
- Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Nikta Rabiei
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Fathazam
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negin Rabiei
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Haleh Piroozmand
- Faculty of Veterinary Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
10
|
Andrikopoulou A, Shalit A, Zografos E, Koutsoukos K, Korakiti AM, Liontos M, Dimopoulos MA, Zagouri F. MicroRNAs as Potential Predictors of Response to CDK4/6 Inhibitor Treatment. Cancers (Basel) 2021; 13:cancers13164114. [PMID: 34439268 PMCID: PMC8391635 DOI: 10.3390/cancers13164114] [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: 06/30/2021] [Revised: 08/07/2021] [Accepted: 08/11/2021] [Indexed: 01/07/2023] Open
Abstract
Simple Summary MicroRNAs are endogenous non-coding 20–22 nucleotide long RNAs that play a fundamental role in the post-transcriptional control of gene expression. Consequently, microRNAs are involved in multiple biological processes of cancer and could be used as biomarkers with prognostic and predictive significance. Cyclin-dependent kinase 4/6 (CDK4/6) inhibitors have become a mainstay of treatment for patients with advanced hormone receptor-positive (HR) breast cancer. Despite the initial high response rates, approximately 10% of patients demonstrate primary resistance to CDK4/6 inhibitors while acquired resistance is almost inevitable. Considering the fundamental role of miRNAs in tumorigenesis, we aimed to explore the potential involvement of microRNAs in response to CDK4/6 inhibition in solid tumors. A number of microRNAs were shown to confer resistance or sensitivity to CDK4/6 inhibitors in preclinical studies, although this remains to be proved in human studies. Abstract Cyclin-dependent kinase 4/6 (CDK4/6) inhibitors have emerged as novel treatment options in the management of advanced or metastatic breast cancer. MicroRNAs are endogenous non-coding 19–22-nucleotide-long RNAs that regulate gene expression in development and tumorigenesis. Herein, we systematically review all microRNAs associated with response to CDK4/6 inhibitors in solid tumors and hematological malignancies. Eligible articles were identified by a search of the MEDLINE and ClinicalTrials.gov databases for the period up to1 January 2021; the algorithm consisted of a predefined combination of the words “microRNAs”, “cancer” and “CDK 4/6 inhibitors”. Overall, 15 studies were retrieved. Six microRNAs (miR-126, miR-326, miR3613-3p, miR-29b-3p, miR-497 and miR-17-92) were associated with sensitivity to CDK4/6 inhibitors. Conversely, six microRNAs (miR-193b, miR-432-5p, miR-200a, miR-223, Let-7a and miR-21) conferred resistance to treatment with CDK4/6 inhibitors. An additional number of microRNAs (miR-124a, miR9, miR200b and miR-106b) were shown to mediate cellular response to CDK4/6 inhibitors without affecting sensitivity to treatment. Collectively, our review provides evidence that microRNAs could serve as predictive biomarkers for treatment with CDK4/6 inhibitors. Moreover, microRNA-targeted therapy could potentially maximize sensitivity to CDK4/6 inhibition.
Collapse
Affiliation(s)
- Angeliki Andrikopoulou
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, 11528 Athens, Greece; (A.A.); (E.Z.); (K.K.); (A.-M.K.); (M.L.); (M.-A.D.)
| | - Almog Shalit
- Medical School, National and Kapodistrian University of Athens, 80 Vasilissis Sofias Avenue, 11528 Athens, Greece;
| | - Eleni Zografos
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, 11528 Athens, Greece; (A.A.); (E.Z.); (K.K.); (A.-M.K.); (M.L.); (M.-A.D.)
| | - Konstantinos Koutsoukos
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, 11528 Athens, Greece; (A.A.); (E.Z.); (K.K.); (A.-M.K.); (M.L.); (M.-A.D.)
| | - Anna-Maria Korakiti
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, 11528 Athens, Greece; (A.A.); (E.Z.); (K.K.); (A.-M.K.); (M.L.); (M.-A.D.)
| | - Michalis Liontos
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, 11528 Athens, Greece; (A.A.); (E.Z.); (K.K.); (A.-M.K.); (M.L.); (M.-A.D.)
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, 11528 Athens, Greece; (A.A.); (E.Z.); (K.K.); (A.-M.K.); (M.L.); (M.-A.D.)
| | - Flora Zagouri
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, 11528 Athens, Greece; (A.A.); (E.Z.); (K.K.); (A.-M.K.); (M.L.); (M.-A.D.)
- Correspondence: ; Tel.: +30-21-0338-1554; Fax: +30-21-3216-2511
| |
Collapse
|
11
|
Lin D, Chen X, Tan L. The predictive value of microRNAs for pathological response after neoadjuvant treatment in esophageal squamous cell carcinoma: a systematic review. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:420. [PMID: 33842641 PMCID: PMC8033340 DOI: 10.21037/atm-20-3000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neoadjuvant treatment followed by esophagectomy has been the standard strategy for resectable locally advanced esophageal squamous cell carcinoma (ESCC). Pathological response after neoadjuvant treatment is of vital importance in the determination of long-term survival. Due to the involvement of microRNAs (miRNAs) in ESCC, some studies have proposed miRNA models to predict the pathological response. We aimed to summarize current studies on the predictive value of the miRNA models. We searched the relevant studies on PubMed, Web of Science and Cochrane Library up to February 14, 2020, using the following search term: (esophageal OR esophagus OR oesophageal OR oesophagus) AND (miR OR miRNA OR microRNA) AND (neoadjuvant OR preoperative OR induction). The initial search retrieved 206 studies. We briefly summarized the involvement of miRNAs in the origin, development and chemo- and radioresistance in ESCC. Then, 9 studies were enrolled in the systematic review. A great heterogeneity was observed across these studies. Of the 6 studies with diagnostic tests, the area under curve varied a lot. Although much evidence demonstrated the correlation between miRNAs and pathological response after in ESCC, the current studies has not established any promising models. A well-designed prospective study is essential to investigate the potential predictive models for pathological response after neoadjuvant treatment in ESCC.
Collapse
Affiliation(s)
- Dong Lin
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Xiaosang Chen
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
12
|
Singh S, Raza W, Parveen S, Meena A, Luqman S. Flavonoid display ability to target microRNAs in cancer pathogenesis. Biochem Pharmacol 2021; 189:114409. [PMID: 33428895 DOI: 10.1016/j.bcp.2021.114409] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/01/2021] [Accepted: 01/05/2021] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs) are non-coding, conserved, single-stranded nucleotide sequences involved in physiological and developmental processes. Recent evidence suggests an association between miRNAs' deregulation with initiation, promotion, progression, and drug resistance in cancer cells. Besides, miRNAs are known to regulate the epithelial-mesenchymal transition, angiogenesis, autophagy, and senescence in different cancer types. Previous reports proposed that apart from the antioxidant potential, flavonoids play an essential role in miRNAs modulation associated with changes in cancer-related proteins, tumor suppressor genes, and oncogenes. Thus, flavonoids can suppress proliferation, help in the development of drug sensitivity, suppress metastasis and angiogenesis by modulating miRNAs expression. In the present review, we summarize the role of miRNAs in cancer, drug resistance, and the chemopreventive potential of flavonoids mediated by miRNAs. The potential of flavonoids to modulate miRNAs expression in different cancer types demonstrate their selectivity and importance as regulators of carcinogenesis. Flavonoids as chemopreventive agents targeting miRNAs are extensively studied in vitro, in vivo, and pre-clinical studies, but their efficiency in targeting miRNAs in clinical studies is less investigated. The evidence presented in this review highlights the potential of flavonoids in cancer prevention/treatment by regulating miRNAs, although further investigations are required to validate and establish their clinical usefulness.
Collapse
Affiliation(s)
- Shilpi Singh
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Waseem Raza
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India; Jawahar Lal Nehru University, New Delhi 110067, India
| | - Shahnaz Parveen
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Abha Meena
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Suaib Luqman
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India.
| |
Collapse
|
13
|
Phatak P, Noe M, Asrani K, Chesnick IE, Greenwald BD, Donahue JM. MicroRNA-141-3p regulates cellular proliferation, migration, and invasion in esophageal cancer by targeting tuberous sclerosis complex 1. Mol Carcinog 2020; 60:125-137. [PMID: 33382472 DOI: 10.1002/mc.23274] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/20/2020] [Accepted: 11/27/2020] [Indexed: 12/18/2022]
Abstract
MicroRNA (miR)-141-3p, which functions as an oncogene in multiple malignancies, has been shown to be highly overexpressed in esophageal cancer cells in our previous work. miR-141-3p is predicted to bind the messenger RNA (mRNA) of tuberous sclerosis complex 1 (TSC1), a tumor suppressor, with high affinity. In this study, we investigated the expression and functional interaction between miR-141-3p and TSC1 in esophageal cancer cells. Experiments were conducted in four esophageal cancer lines and in tumor cells isolated from human esophageal cancer specimens by laser capture microdissection. miR-141-3p expression was measured by real time and droplet digital PCR. Biotinylated RNA pull-down and luciferase reporter assays were used to assess binding. miR-141-3p function was tested by assessing proliferation, migration, invasion, and induction of autophagy following its silencing. We found that miR-141-3p levels were increased in TE7, OE33, and TE10 esophageal cancer cells compared to FLO-1 cells, with similar heterogeneity observed in human esophageal cancer specimens. Silencing of miR-141-3p led to increased TSC1 protein expression in these cells and was associated with increased TSC1 translation. Binding studies reveal that miR-141-3p binds to each of the predicted binding sites in the 3'-untranslated region of TSC1 mRNA. Following miR-141-3p silencing, TE7, OE33, and TE10 cells exhibited decreased proliferation, migration, and invasion, as well as enhanced autophagy. Importantly, these phenotypic effects were replicated by overexpression of TSC1 alone in these cells. Our results indicate that miR-141-3p functions in an oncogenic capacity in a subset of esophageal cancer cells, in part by suppressing TSC1 expression.
Collapse
Affiliation(s)
- Pornima Phatak
- Birmingham Veterans Affairs Medical Center, Birmingham, Alabama, USA.,Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, USA
| | - Michael Noe
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Kaushal Asrani
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | - Bruce D Greenwald
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - James M Donahue
- Birmingham Veterans Affairs Medical Center, Birmingham, Alabama, USA.,Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, USA
| |
Collapse
|
14
|
Yao W, Guo P, Mu Q, Wang Y. Exosome-Derived Circ-PVT1 Contributes to Cisplatin Resistance by Regulating Autophagy, Invasion, and Apoptosis Via miR-30a-5p/YAP1 Axis in Gastric Cancer Cells. Cancer Biother Radiopharm 2020; 36:347-359. [PMID: 32799541 DOI: 10.1089/cbr.2020.3578] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background: Emerging studies manifested that exosomal RNAs had pivotal roles in human cancer therapies. This article aimed to research the regulatory mechanism of exosomal circRNA-plasmacytoma variant translocation 1 (circ-PVT1) in cisplatin (DDP) resistance of gastric cancer (GC). Methods: Exosomes were isolated by ExoQuick® method and ultracentrifugation and then identified through transmission electron microscope and the examination of exosome markers. Related proteins were detected using Western blot. Quantitative real-time polymerase chain reaction (qRT-PCR) was applied for measuring circ-PVT1, microRNA-30a-5p (miR-30a-5p), and Yes-associated protein 1 (YAP1) expression. The half inhibitory concentration (IC50) of DDP was assessed by 3-(4, 5-dimethylthiazol-2-y1)-2, 5-diphenyl tetrazolium bromide (MTT). Cell apoptosis and invasion were, respectively, determined using flow cytometry and transwell assay. Target relationship was confirmed by dual-luciferase reporter assay. The impact of circ-PVT1 on DDP resistance was explored via xenograft tumor assay. Results: Exosomal circ-PVT1 was upregulated while miR-30a-5p was downregulated in DDP-resistant GC serums and cells. Circ-PVT1 knockdown repressed DDP resistance in DDP-resistant GC cells via promoting apoptosis and decreasing invasion or autophagy by negatively targeting miR-30a-5p. YAP1 was a direct target of miR-30a-5p. MiR-30a-5p overexpression inhibited DDP resistance via reducing YAP1. Circ-PVT1 modulated YAP1 expression by targeting miR-30a-5p. Circ-PVT1 depression expedited DDP sensitivity of GC via miR-30a-5p/YAP1 axis in vivo. Conclusion: Exosomal circ-PVT1 facilitated DDP resistance via modulating autophagy, invasion and apoptosis by miR-30a-5p/YAP1 axis in GC cells. Exosomal circ-PVT1 might be a prospective indicator in DDP therapy of GC.
Collapse
Affiliation(s)
- Wenjuan Yao
- Department of Clinical Laboratory, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Peng Guo
- Department of Hepatology and Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Qiming Mu
- Department of Clinical Laboratory, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Yuecheng Wang
- Department of Gastroenterology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
15
|
Businello G, Parente P, Mastracci L, Pennelli G, Traverso G, Milione M, Bellan E, Michelotto M, Kotsafti A, Grillo F, Fassan M. The Pathologic and Molecular Landscape of Esophageal Squamous Cell Carcinogenesis. Cancers (Basel) 2020; 12:2160. [PMID: 32759723 PMCID: PMC7465394 DOI: 10.3390/cancers12082160] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/27/2020] [Accepted: 08/01/2020] [Indexed: 02/07/2023] Open
Abstract
Esophageal squamous cell carcinoma represents the most common histotype of epithelial neoplasm occurring within esophageal mucosa worldwide. Despite the comprehensive molecular characterization of this entity, to date no significant targeted therapy has been introduced into clinical practice. In this review, we describe the molecular landscape of esophageal squamous cell carcinoma based on the most recent literature. Moreover, we focus on other rare variants and on the relationship with head and neck squamous cell carcinomas.
Collapse
Affiliation(s)
- Gianluca Businello
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (G.B.); (G.P.); (E.B.); (M.M.)
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo FG, Italy;
| | - Luca Mastracci
- Anatomic Pathology, Ospedale Policlinico San Martino IRCCS, 16132 Genova, Italy; (L.M.); (F.G.)
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, 16132 Genova, Italy;
| | - Gianmaria Pennelli
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (G.B.); (G.P.); (E.B.); (M.M.)
| | | | - Massimo Milione
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, 16132 Genova, Italy;
| | - Elena Bellan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (G.B.); (G.P.); (E.B.); (M.M.)
| | - Mauro Michelotto
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (G.B.); (G.P.); (E.B.); (M.M.)
| | - Andromachi Kotsafti
- Laboratory of Advanced Translational Research, Veneto Institute of Oncology IOV–IRCCS, 35128 Padua, Italy;
| | - Federica Grillo
- Anatomic Pathology, Ospedale Policlinico San Martino IRCCS, 16132 Genova, Italy; (L.M.); (F.G.)
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, 16132 Genova, Italy;
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (G.B.); (G.P.); (E.B.); (M.M.)
| |
Collapse
|
16
|
Cui F, Hao ZX, Li J, Zhang YL, Li XK, He JX. SOX2 mediates cisplatin resistance in small-cell lung cancer with downregulated expression of hsa-miR-340-5p. Mol Genet Genomic Med 2020; 8:e1195. [PMID: 32130794 PMCID: PMC7216814 DOI: 10.1002/mgg3.1195] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 01/19/2020] [Accepted: 02/14/2020] [Indexed: 12/12/2022] Open
Abstract
Background This study is aimed to unravel the genetic factors associated with microRNA (miRNA) expression in regulating sex‐determining region Y‐box 2 (SOX2)‐mediated cisplatin resistance in small‐cell lung cancer (SCLC). Methods The relevance of SOX2 expression in SCLC was analyzed in a panel of SCLC cells by quantitative real‐time PCR (qPCR) and western blot (WB). We selected DMS114 cell line, in which SOX2 was amplified via lentiviral vector‐mediated transfection of the SOX2 genes and tested for the half‐maximal inhibitory concentration (IC50) by MTS assay. High‐throughput sequencing and screening of differentially expressed miRNAs between SOX2‐overexpressing and normal control cells were performed. Finally, miRanda software was used to verify the miRNAs bound with SOX2 and qPCR was used to identify the expression of miRNAs which were binding with SOX2. Results Cisplatin‐resistant SOX2‐overexpressing DMS114 cell lines were successfully developed, showing a statistically significant increase in SOX2 expression by qPCR and WB. Our results showed a typically higher IC50 value in SOX2‐overexpressing cells compared with the negative controls. The high‐throughput sequencing analysis revealed that 68 miRNAs were upregulated and 24 miRNAs were downregulated in the SOX2‐overexpressing cells. The 24 downregulated miRNAs were further verified. Of them, a cancer‐related miRNA, hsa‐miR‐340‐5p, showed a higher binding affinity with SOX2 in network regulation mapping, which was also found to be markedly downregulated under qPCR analysis. Conclusion We demonstrated that downregulated expression of hsa‐miR‐340‐5p may affect cisplatin resistance by mediating SOX2 expression in SCLC cells, which may provide a potential target for the therapy of chemoresistant SCLCs.
Collapse
Affiliation(s)
- Fei Cui
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhe-Xue Hao
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jin Li
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ya-Lei Zhang
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xu-Kai Li
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian-Xing He
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
17
|
Zhang JH, Xia HB. Lentiviral-Mediated Overexpression of MicroRNA-141 Promotes Cell Proliferation and Inhibits Apoptosis in Human Esophageal Squamous Cell Carcinoma. Recent Pat Anticancer Drug Discov 2020; 14:170-176. [PMID: 30599110 DOI: 10.2174/1574892814666181231142136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 12/20/2018] [Accepted: 12/26/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Esophageal Carcinoma (EC) is the eighth most common cancer worldwide. Numerous studies have highlighted a vital role of microRNAs (miRNAs) in the development of EC. However, the mechanism of microRNA (miRNA)-141 in Esophageal Squamous Cell Carcinoma (ESCC) remains unknown. OBJECTIVE In this study, we explored the effects of miRNA-141 on EC cell proliferation, apoptosis, xenograft tumour growth and their possible mechanisms. METHODS A lentivirus-vector-expressing miRNA-141 was constructed, and a TE-1 cell line of ESCC with a stable expression of miRNA-141 was transfected and screened. The miRNA-141 expression level was detected using qRT-PCR. Effects of miRNA-141 overexpression on cell proliferation and apoptosis were detected using MTT and flow cytometry, respectively. Using a dual-luciferase reporter assay, a direct interaction between miRNA-141 and the 3'-Untranslated Region (UTR) of YAP1 and SOX17 was confirmed. Tumour xenograft experiment in nude mice was used to detect the tumour growth, and the effects of miRNA-141 overexpression on YAP1 and SOX17 were analysed using Western blot. RESULTS We found that miRNA-141 was highly expressed in TE-1 cells, and miRNA-141 overexpression promoted cell proliferation and inhibited apoptosis. Moreover, the miRNA-141 group showed significantly increased tumour growth ability, luciferase activities and expression levels of YAP1 and SOX17 in the miRNA-141group were significantly down-regulated. CONCLUSION miRNA-141 promotes cell proliferation and inhibits apoptosis in ESCC by downregulating the expression level of YAP1 and SOX17, indicating that miRNA-141 may be a potential molecular target for the treatment of ESCC.
Collapse
Affiliation(s)
- Jun-He Zhang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, Shaanxi, China.,Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Hai-Bin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, Shaanxi, China
| |
Collapse
|
18
|
Bao J, Li X, Li Y, Huang C, Meng X, Li J. MicroRNA-141-5p Acts as a Tumor Suppressor via Targeting RAB32 in Chronic Myeloid Leukemia. Front Pharmacol 2020; 10:1545. [PMID: 32038235 PMCID: PMC6987442 DOI: 10.3389/fphar.2019.01545] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/29/2019] [Indexed: 12/11/2022] Open
Abstract
MicroRNA-141-5p (miR-141-5p), an important member of the miR-200 family, has been reported to be involved in cellular proliferation, migration, invasion, and drug resistance in different kinds of human malignant tumors. However, the role and function of miR-141-5p in chronic myeloid leukemia (CML) are unclear. In this current study, we found that the level of miR-141-5p was significantly decreased in peripheral blood cells from CML patients compared with normal blood cells and human leukemic cell line (K562 cells) compared with normal CD34+ cells, but was remarkably elevated in patients after treatment with nilotinib or imatinib. Suppression of miR-141-5p promoted K562 cell proliferation and migration in vitro. As expected, overexpression of miR-141-5p weakened K562 cell proliferation, migration, and promoted cell apoptosis. A xenograft model in nude mice showed that overexpression of miR-141-5p markedly suppressed tumor growth in vivo. Mechanistic studies suggested that RAB32 was the potential target of miR-141-5p, and silencing of RAB32 suppressed the proliferation and migration of K562 cells and promoted cell apoptosis. Taken together, our study demonstrates that miR-141-5p plays an important role in the activation of K562 cells in vitro and may act as a tumor suppressor via targeting RAB32 in the development of CML.
Collapse
Affiliation(s)
- Jing Bao
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China.,Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaofeng Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Yuhuan Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Cheng Huang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Xiaoming Meng
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Jun Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| |
Collapse
|
19
|
Jiang W, Zhao X, Yang W. MiR-647 promotes cisplatin-induced cell apoptosis via downregulating IGF2 in non-small cell lung cancer. Minerva Med 2019; 112:312-313. [PMID: 31726809 DOI: 10.23736/s0026-4806.19.06240-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Wei Jiang
- Department of Infectious Diseases, Tianjin First Center Hospital, Tianjin, China
| | - Xuequn Zhao
- Department of Infectious Diseases, Tianjin First Center Hospital, Tianjin, China
| | - Wenjie Yang
- Department of Infectious Diseases, Tianjin First Center Hospital, Tianjin, China -
| |
Collapse
|
20
|
Liu X, Pan CG, Luo ZQ. High expression of NFAT2 contributes to carboplatin resistance in lung cancer. Exp Mol Pathol 2019; 110:104290. [DOI: 10.1016/j.yexmp.2019.104290] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 11/30/2022]
|
21
|
Feng W, Su Z, Yin Q, Zong W, Shen X, Ju S. ncRNAs associated with drug resistance and the therapy of digestive system neoplasms. J Cell Physiol 2019; 234:19143-19157. [PMID: 30941775 DOI: 10.1002/jcp.28551] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/25/2019] [Accepted: 03/05/2019] [Indexed: 12/19/2022]
Abstract
Digestive system cancer remains a common cancer and the main cause of cancer-related death worldwide. Drug resistance is a major challenge in the therapy of digestive system cancer, and represents a primary obstacle in the treatment of cancer by restricting the efficiency of both traditional chemotherapy and biological therapies. Existing studies indicate that noncoding RNAs play an important role in the evolution and progression of drug resistance in digestive system cancer, mainly by modulating drug transporter-related proteins, DNA damage repair, cell-cycle-related proteins, cell apoptosis-related proteins, drug target-related proteins, and the tumor microenvironment. In this review, we address the potential mechanisms of ncRNAs underlying drug resistance in digestive system tumors and discuss the possible application of ncRNAs against drug resistance in digestive system tumors.
Collapse
Affiliation(s)
- Wei Feng
- Center of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Zhangyao Su
- School of Medicine, Nantong University, Nantong, China
| | - Qingqing Yin
- Center of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Wei Zong
- Center of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Xianjuan Shen
- Clinical Medical Research Center, Affiliated Hospital of Nantong University, Nantong, China
| | - Shaoqing Ju
- Center of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
22
|
The role and mechanisms of action of microRNAs in cancer drug resistance. Clin Epigenetics 2019; 11:25. [PMID: 30744689 PMCID: PMC6371621 DOI: 10.1186/s13148-018-0587-8] [Citation(s) in RCA: 478] [Impact Index Per Article: 79.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 11/19/2018] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs with a length of about 19–25 nt, which can regulate various target genes and are thus involved in the regulation of a variety of biological and pathological processes, including the formation and development of cancer. Drug resistance in cancer chemotherapy is one of the main obstacles to curing this malignant disease. Statistical data indicate that over 90% of the mortality of patients with cancer is related to drug resistance. Drug resistance of cancer chemotherapy can be caused by many mechanisms, such as decreased antitumor drug uptake, modified drug targets, altered cell cycle checkpoints, or increased DNA damage repair, among others. In recent years, many studies have shown that miRNAs are involved in the drug resistance of tumor cells by targeting drug-resistance-related genes or influencing genes related to cell proliferation, cell cycle, and apoptosis. A single miRNA often targets a number of genes, and its regulatory effect is tissue-specific. In this review, we emphasize the miRNAs that are involved in the regulation of drug resistance among different cancers and probe the mechanisms of the deregulated expression of miRNAs. The molecular targets of miRNAs and their underlying signaling pathways are also explored comprehensively. A holistic understanding of the functions of miRNAs in drug resistance will help us develop better strategies to regulate them efficiently and will finally pave the way toward better translation of miRNAs into clinics, developing them into a promising approach in cancer therapy.
Collapse
|
23
|
Baradaran B, Shahbazi R, Khordadmehr M. Dysregulation of key microRNAs in pancreatic cancer development. Biomed Pharmacother 2018; 109:1008-1015. [PMID: 30551350 DOI: 10.1016/j.biopha.2018.10.177] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/22/2018] [Accepted: 10/30/2018] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer (PC) is mentioned as one of the fourth major cause of cancer-related deaths and also is considered as one of the most malignancies worldwide. Sadly, widely metastasis is frequently observed at the time of PC detection and there are, thereby, almost poor prognosis and ineffective treatment in PC patients. microRNAs (miRNAs), a group of short non-coding RNAs, regulate various cellular and developmental mechanisms, such as cell growth, proliferation, apoptosis, differentiation and angiogenesis. Also, they have essential roles even on the progression of different human and animal diseases. In recent years, extensive studies confirmed the important role of miRNAs in various steps of PC developments, including; tumor initiation, invasion and metastasis, which can use valuably for cancer detection, prognosis and therapy. Therefore, the present study reviewed the new recent investigations in miRNAs involvement in the biology of PC associated with their clinical implications.
Collapse
Affiliation(s)
- Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roya Shahbazi
- Department of Pathology, Faculty of Veterinary Medicine, University of Tabriz, 51665-1647, Tabriz, Iran
| | - Monireh Khordadmehr
- Department of Pathology, Faculty of Veterinary Medicine, University of Tabriz, 51665-1647, Tabriz, Iran.
| |
Collapse
|
24
|
Liu H, Du S, Lei T, Wang H, He X, Tong R, Wang Y. Multifaceted regulation and functions of YAP/TAZ in tumors (Review). Oncol Rep 2018; 40:16-28. [PMID: 29749524 PMCID: PMC6059739 DOI: 10.3892/or.2018.6423] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 04/19/2018] [Indexed: 12/14/2022] Open
Abstract
The Hippo pathway, initially identified through screenings for mutant tumor suppressors in Drosophila, is an evolutionarily conserved signaling pathway that controls organ size by regulating cell proliferation and apoptosis. Abnormal regulation of the Hippo pathway may lead to cancer in mammals. As the major downstream effectors of the Hippo pathway, unphosphorylated Yes-associated protein (YAP) and its homolog transcriptional co-activator TAZ (also called WWTR1) (hereafter called YAP/TAZ) are translocated into the nucleus. In the nucleus, in order to induce target gene expression, YAP/TAZ bind to the TEA domain (TEAD) proteins, and this binding subsequently promotes cell proliferation and inhibits apoptosis. In contrast, as key regulators of tumorigenesis and development, YAP/TAZ are phosphorylated and regulated by multiple molecules and pathways including Lats1/2 of Hippo, Wnt and G-protein-coupled receptor (GPCR) signaling, with a regulatory role in cell physiology, tumor cell development and pathological abnormalities simultaneously. In particular, the crucial role of YAP/TAZ in tumors ensures their potential as targets in designing anticancer drugs. To date, mounting research has elucidated the suppression of YAP/TAZ via effective inhibitors, which significantly highlights their application in cancer treatment. In the present review, we focus on the functions of YAP/TAZ in cancer, discuss their potential as new therapeutic target for tumor treatment, and provide valuable suggestions for further study in this field.
Collapse
Affiliation(s)
- Huirong Liu
- Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Suya Du
- School of Medicine, University of Electronic Science and Technology of China Chengdu, Sichuan 610054, P.R. China
| | - Tiantian Lei
- School of Medicine, University of Electronic Science and Technology of China Chengdu, Sichuan 610054, P.R. China
| | - Hailian Wang
- Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Xia He
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Rongsheng Tong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Yi Wang
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
25
|
Vrana D, Hlavac V, Brynychova V, Vaclavikova R, Neoral C, Vrba J, Aujesky R, Matzenauer M, Melichar B, Soucek P. ABC Transporters and Their Role in the Neoadjuvant Treatment of Esophageal Cancer. Int J Mol Sci 2018; 19:E868. [PMID: 29543757 PMCID: PMC5877729 DOI: 10.3390/ijms19030868] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 03/07/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022] Open
Abstract
The prognosis of esophageal cancer (EC) is poor, despite considerable effort of both experimental scientists and clinicians. The tri-modality treatment consisting of neoadjuvant chemoradiation followed by surgery has remained the gold standard over decades, unfortunately, without significant progress in recent years. Suitable prognostic factors indicating which patients will benefit from this tri-modality treatment are missing. Some patients rapidly progress on the neoadjuvant chemoradiotherapy, which is thus useless and sometimes even harmful. At the same time, other patients achieve complete remission on neoadjuvant chemoradiotherapy and subsequent surgery may increase their risk of morbidity and mortality. The prognosis of patients ranges from excellent to extremely poor. Considering these differences, the role of drug metabolizing enzymes and transporters, among other factors, in the EC response to chemotherapy may be more important compared, for example, with pancreatic cancer where all patients progress on chemotherapy regardless of the treatment or disease stage. This review surveys published literature describing the potential role of ATP-binding cassette transporters, the genetic polymorphisms, epigenetic regulations, and phenotypic changes in the prognosis and therapy of EC. The review provides knowledge base for further research of potential predictive biomarkers that will allow the stratification of patients into defined groups for optimal therapeutic outcome.
Collapse
Affiliation(s)
- David Vrana
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University, Hnevotinska 976/3, 77515 Olomouc, Czech Republic.
| | - Viktor Hlavac
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 76, 32300 Pilsen, Czech Republic.
| | - Veronika Brynychova
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 76, 32300 Pilsen, Czech Republic.
| | - Radka Vaclavikova
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 76, 32300 Pilsen, Czech Republic.
| | - Cestmir Neoral
- Department of Surgery, Faculty of Medicine and Dentistry, Palacky University, Hnevotinska 976/3, 77515 Olomouc, Czech Republic.
| | - Jiri Vrba
- Department of Surgery, Faculty of Medicine and Dentistry, Palacky University, Hnevotinska 976/3, 77515 Olomouc, Czech Republic.
| | - Rene Aujesky
- Department of Surgery, Faculty of Medicine and Dentistry, Palacky University, Hnevotinska 976/3, 77515 Olomouc, Czech Republic.
| | - Marcel Matzenauer
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University, Hnevotinska 976/3, 77515 Olomouc, Czech Republic.
| | - Bohuslav Melichar
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University, Hnevotinska 976/3, 77515 Olomouc, Czech Republic.
| | - Pavel Soucek
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 76, 32300 Pilsen, Czech Republic.
- Department of Surgery, Faculty Hospital Pilsen, Alej Svobody 80, 30460 Pilsen, Czech Republic.
| |
Collapse
|
26
|
Abstract
Our understanding of the epigenetic changes occurring in gastrointestinal cancers has gained tremendous advancements in recent years, and some epigenetic biomarkers are already translated into the clinics for cancer diagnostics. In parallel, pharmacoepigenetics and pharmacoepigenomics of solid tumors are relevant novel, but emerging and promising fields. Areas covered: A comprehensive review of the literature to summarize and update the emerging field of pharmacoepigenetics and pharmacoepigenomics of gastrointestinal cancers. Expert commentary: Several epigenetic modifications have been proposed to account for interindividual variations in drug response in gastrointestinal cancers. Similarly, single-agent or combined strategies with high doses of drugs that target epigenetic modifications (epi-drugs) were scarcely tolerated by the patients, and current research has moved to their combination with standard therapies to achieve chemosensitization, radiosensitization, and immune modulation of cancerous cells. In parallel, recent genome-wide technologies are revealing the pathways that are epigenetically deregulated during cancer-acquired resistance, including those targeted by non-coding RNAs. Indeed, novel, less toxic, and more specific molecules are under investigation to specifically target those pathways. The field is rapidly expanding and gathering together information coming from these investigations has the potential to lead to clinical applications in the coming new years.
Collapse
Affiliation(s)
- Angela Lopomo
- a Department of Translational Research and New Technologies in Medicine and Surgery, Laboratory of Medical Genetics , University of Pisa, Medical School , Pisa , Italy
| | - Fabio Coppedè
- a Department of Translational Research and New Technologies in Medicine and Surgery, Laboratory of Medical Genetics , University of Pisa, Medical School , Pisa , Italy
| |
Collapse
|
27
|
Yang W, Ma J, Zhou W, Zhou X, Cao B, Zhang H, Zhao Q, Fan D, Hong L. Molecular mechanisms and clinical implications of miRNAs in drug resistance of esophageal cancer. Expert Rev Gastroenterol Hepatol 2017; 11:1151-1163. [PMID: 28838272 DOI: 10.1080/17474124.2017.1372189] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
With the increasing incidence of esophageal cancer, drug resistance is becoming a major obstacle to successful cancer therapy since chemotherapy is regarded as a curative approach to inhibit cancer cell proliferation. Despite the great progress in anticancer treatment achieved during the last decades, the mechanisms of multidrug resistance have not been completely elucidated. Recently, accumulating studies and pre-clinical reports highlighted the role of miRNAs in the drug resistance of esophageal cancer. Areas covered: In this review, we mainly summarized the current advances of miRNAs in esophageal cancer and the mechanisms underlying drug resistance. We also reviewed the potential role of miRNAs as biomarkers for predicting drug response and prognosis. Finally, we envisaged the future orientation and challenges in translating the existing knowledge of drug resistance related miRNAs into clinical applications. Expert commentary: Based on the current knowledge of certain miRNAs, we believe that miRNAs would be helpful to overcome the drug resistance and provide personalized treatment for patients with esophageal cancer. The aims of this study were to provide a comprehensive summary on the emerging role of miRNAs in the drug resistance of esophageal cancer and attract broad attention of more researchers on this field.
Collapse
Affiliation(s)
- Wanli Yang
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Jiaojiao Ma
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Wei Zhou
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Xin Zhou
- b The First Brigade of Student , Fourth Military Medical University , Xi'an , China
| | - Bo Cao
- b The First Brigade of Student , Fourth Military Medical University , Xi'an , China
| | - Hongwei Zhang
- c Department of Digestive Surgery , Xijing Hospital, Fourth Military Medical University , Xi'an , China
| | - Qingchuan Zhao
- c Department of Digestive Surgery , Xijing Hospital, Fourth Military Medical University , Xi'an , China
| | - Daiming Fan
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Liu Hong
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| |
Collapse
|
28
|
Pang L, Lu J, Huang J, Xu C, Li H, Yuan G, Cheng X, Chen J. Upregulation of miR-146a increases cisplatin sensitivity of the non-small cell lung cancer A549 cell line by targeting JNK-2. Oncol Lett 2017; 14:7745-7752. [PMID: 29344219 PMCID: PMC5755143 DOI: 10.3892/ol.2017.7242] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 08/10/2017] [Indexed: 12/12/2022] Open
Abstract
The aim of the present study was to investigate the effects of microRNA (miR-)146a on the cisplatin sensitivity of the non-small cell lung cancer (NSCLC) A549 cell line and study the underlying molecular mechanism. The differences in expression of miRNAs between A549 and A549/cisplatin (A549/DDP) cells were determined, and miR-146a was selected to study its effect on cisplatin sensitivity of A549/DDP cells. miR-146a mimic and inhibitor transient transfection systems were constructed using vectors, and A549/DDP cells were infected with miR-146a mimic and inhibitor to investigate growth, apoptosis and migration. The directed target of miR-146a was determined and the underlying molecular mechanism was validated in the present study. The results of the present study demonstrated that miR-146a was downregulated in NSCLC A549/DDP cells, compared with A549 cells. The overexpression of miR-146a induced apoptosis and inhibited the growth and invasion of A549/DDP cells, which resulted in increased cisplatin sensitivity in NSCLC cells. The JNK2 gene was determined as the direct target of miR-146a, and may be activated by the overexpression of miR-146a. Additionally, JNK2 activated the expression of p53 and inhibited B cell lymphoma 2. The upregulation of miR-146a increased cisplatin sensitivity of the A549 cell line by targeting JNK2, which may provide a novel method for treating NSCLC cisplatin resistance.
Collapse
Affiliation(s)
- Linrong Pang
- Department of Chemoradiotherapy Center, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Jinger Lu
- Department of Endocrinology, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Jia Huang
- Department of Chemoradiotherapy Center, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Caihong Xu
- Department of Chemoradiotherapy Center, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Hui Li
- Department of Chemoradiotherapy Center, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Guangbo Yuan
- Department of Chemoradiotherapy Center, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Xiaochun Cheng
- Department of Chemoradiotherapy Center, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Jun Chen
- Department of Chemoradiotherapy Center, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
29
|
miR-221 Mediates Chemoresistance of Esophageal Adenocarcinoma by Direct Targeting of DKK2 Expression. Ann Surg 2017; 264:804-814. [PMID: 27501171 DOI: 10.1097/sla.0000000000001928] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Chemoresistance is a main obstacle to effective esophageal cancer (EC) therapy. We hypothesize that altered expression of microRNAs (miRNAs) play a role in EC cancer progression and resistance to 5-fluorouracil (5-FU) based chemotherapeutic strategies. METHODS Four pairs of esophageal adenocarcinoma (EAC) cell lines and corresponding 5-FU resistant variants were established. The expression levels of miRNAs previously shown to be involved in the general regulation of stem cell pathways were analyzed by qRT-PCR. The effects of selected miRNAs on proliferation, apoptosis, and chemosensitivity were evaluated both in vitro and in vivo. We identified a particular miRNA and analyzed its putative target genes in 14 pairs of human EC tumor specimens with surrounding normal tissue by qRT-PCR as well as Wnt pathway associated genes by immunohistochemistry in another 45 EAC tumor samples. RESULTS MiR-221 was overexpressed in 5-FU resistant EC cell lines as well as in human EAC tissue. DKK2 was identified as a target gene for miR-221. Knockdown of miR-221 in 5-FU resistant cells resulted in reduced cell proliferation, increased apoptosis, restored chemosensitivity, and led to inactivation of the Wnt/β-catenin pathway mediated by alteration in DKK2 expression. Moreover, miR-221 reduction resulted in alteration of EMT-associated genes such as E-cadherin and vimentin as well as significantly slower xenograft tumor growth in nude mice. RT profiler analysis identified a substantial dysregulation of 4 Wnt/β-catenin signaling and chemoresistance target genes as a result of miR-221 modulation: CDH1, CD44, MYC, and ABCG2. CONCLUSION MiR-221 controls 5-FU resistance of EC partly via modulation of Wnt/β-catenin-EMT pathways by direct targeting of DKK2 expression. MiR-221 may serve as a prognostic marker and therapeutic target for patients with 5-FU resistant EAC.
Collapse
|
30
|
Meng X, Chen X, Lu P, Ma W, Yue D, Song L, Fan Q. miR-202 Promotes Cell Apoptosis in Esophageal Squamous Cell Carcinoma by Targeting HSF2. Oncol Res 2017; 25:215-223. [PMID: 28277193 PMCID: PMC7840838 DOI: 10.3727/096504016x14732772150541] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common malignant cancers with high mortality around the world. However, the regulatory mechanism of ESCC carcinogenesis is not completely known. Here we demonstrate the novel role of miR-202 in regulating ESCC cell apoptosis. The analysis of data obtained from the GEO database showed that the expression of miR-202 is aberrantly decreased in tumor tissue from ESCC patients and cultured ESCC cell lines. After transfection with miR-202 mimic or inhibitor, the apoptotic capacity of ESCC cells was significantly increased by miR-202 overexpression but reduced by miR-202 repression. We then identified HSF2 as a direct target of miR-202 with the binding site on the 3′-UTR of HSF2 mRNA in ESCC cells. The apoptosis of ESCC cells induced by the miR-202 mimic could be repressed by HSF2 overexpression. Further studies indicated that HSF2 overexpression strongly upregulated the expression of Hsp70 at both the mRNA and protein levels. In addition, HSF2/Hsp70 suppressed ESCC cell apoptosis by preventing caspase 3 activation. In conclusion, miR-202 is a potential tumor suppressor in human ESCC and acts by regulating the apoptosis of ESCC cells by targeting HSF2, in which caspase 3 activation is involved. This might provide a novel therapeutic target for human ESCC.
Collapse
Affiliation(s)
- Xiangrui Meng
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | | | | | | | | | | | | |
Collapse
|
31
|
Biersack B. Interactions between anticancer active platinum complexes and non-coding RNAs/microRNAs. Noncoding RNA Res 2017; 2:1-17. [PMID: 30159416 PMCID: PMC6096430 DOI: 10.1016/j.ncrna.2016.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/07/2016] [Accepted: 10/07/2016] [Indexed: 12/13/2022] Open
Abstract
Platinum(II) complexes such as cisplatin, carboplatin and oxaliplatin are clinically approved for the therapy of various solid tumors. Challenging pathogenic properties of cancer cells and the response of cancers towards platinum-based drugs are strongly influenced by non-coding small RNA molecules, the microRNAs (miRNAs). Both increased platinum activity and formation of tumor resistance towards platinum drugs are controlled by miRNAs. This review gives an overview of the interactions between platinum-based drugs and miRNAs, and their influence on platinum activity in various cancer types is discussed.
Collapse
Key Words
- 5-FU, 5-fluorouracil
- Anticancer drugs
- CBDCA, cyclobutane-1,1-dicarboxylate
- Carboplatin
- Cisplatin
- DACH, 1,2-diaminocyclohexane
- DDP, cisplatin
- EGCG, (−)-epigallocatechin-3-gallate
- EOX, epirubicin/oxaliplatin/xeloda
- FOLFOX, folinate/5-FU/oxaliplatin
- GC, gemcitabine/cisplatin, gastric cancer
- LNA, locked nucleic acid
- MVAC, methotrexate/vinblastine/adriamycin/cisplatin
- MicroRNA
- Oxaliplatin
- Platinum complexes
- XELOX, xeloda/oxaliplatin
- dTTP, deoxythymidine triphosphate
Collapse
|
32
|
Azarnezhad A, Mehdipour P. Cancer Genetics at a Glance: The Comprehensive Insights. CANCER GENETICS AND PSYCHOTHERAPY 2017:79-389. [DOI: 10.1007/978-3-319-64550-6_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
33
|
Zang Y, Tai Y, Wan B, Jia X. miR-200a-3p promotes the proliferation of human esophageal cancer cells by post-transcriptionally regulating cytoplasmic collapsin response mediator protein-1. Int J Mol Med 2016; 38:1558-1564. [DOI: 10.3892/ijmm.2016.2758] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 08/12/2016] [Indexed: 11/05/2022] Open
|
34
|
Hemmatzadeh M, Mohammadi H, Karimi M, Musavishenas MH, Baradaran B. Differential role of microRNAs in the pathogenesis and treatment of Esophageal cancer. Biomed Pharmacother 2016; 82:509-19. [PMID: 27470391 DOI: 10.1016/j.biopha.2016.05.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 05/06/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022] Open
Abstract
Esophageal cancer (EC) is the most invasive disease associated with inclusive poor prognosis. EC usually is found as either adenocarcinoma (EAC) or squamous cell carcinomas (ESCC). ESCC forms in squamous cells and highly occurs in the upper third of the esophagus. EAC appears in glandular cells and ordinarily develops in the lower one third of the esophagus near the stomach. Barrett's esophagus (BE) is a metaplastic precursor of EAC. There is a persistent need for improving our understanding of the molecular basis of this disease. MicroRNAs (miRNAs) demonstrate an uncovered class of small, non-coding RNAs that can negatively regulate the protein coding gene, and are associated with approximately all known physiological and pathological processes, especially cancer. MiRNAs can affect cancer pathogenesis, playing a crucial role as either oncogenes or tumor suppressors. The recent emergence of observations on the role of miRNAs in cancer and their functions has induced many investigations to examine their relevance to esophageal cancer. In esophageal cancer, miRNA dysregulation plays a crucial role in cancer prognosis and in patients' responsiveness to neo-adjuvant and adjuvant therapies. In this review, the oncogenic, tumor suppressive, and drug resistance related roles of miRNAs, and their involvement in the pathogenesis and treatment of esophageal cancer were summarized.
Collapse
Affiliation(s)
- Maryam Hemmatzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Tabriz University of Medical Sciences, International Branch (Aras), Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Karimi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Tabriz University of Medical Sciences, International Branch (Aras), Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hossein Musavishenas
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Tabriz University of Medical Sciences, International Branch (Aras), Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
35
|
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
|
36
|
Jia J, Li C, Luo S, Liu-Smith F, Yang J, Wang X, Wang N, Lai B, Lei T, Wang Q, Xiao S, Shao Y, Zheng Y. Yes-Associated Protein Contributes to the Development of Human Cutaneous Squamous Cell Carcinoma via Activation of RAS. J Invest Dermatol 2016; 136:1267-1277. [PMID: 26902922 DOI: 10.1016/j.jid.2016.02.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 01/06/2016] [Accepted: 02/02/2016] [Indexed: 11/17/2022]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is one of the most common skin malignant tumors with an increasing incidence. Studies have shown that Yes-associated protein (YAP) participates in the development of a variety of tumors as an oncogene, but to our knowledge its role in cSCC has not been reported. In this study, we used immunohistochemistry to show that YAP expression was elevated in cSCC samples of different stages versus in normal skin and that it was well correlated with the progression of the disease. Down-regulation of YAP in cSCC cell lines A431 and SCL-1 inhibited cell proliferation by inducing growth arrest during the G1/S phase transition, promoted apoptosis, and reduced invasion and migration abilities in vitro. Conversely, overexpression of YAP promoted cell proliferation and protected cells against basal and chemotherapy-induced apoptosis. These oncogenic effects of YAP were associated with activation of the RAS protein and its downstream AKT and ERK. Using a mouse xenograft model, we further showed that YAP depletion inhibited cSCC tumor growth in vivo. Our results suggested that YAP is involved in the carcinogenesis and development of cSCC and that it may serve as a biomarker or therapeutic target of this disease.
Collapse
Affiliation(s)
- Jinjing Jia
- Department of Dermatology, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Changji Li
- Department of Dermatology, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Suju Luo
- Department of Dermatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Feng Liu-Smith
- Departments of Epidemiology and Medicine, University of California Irvine, Irvine, CA, USA
| | - Jiao Yang
- Department of Dermatology, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Xin Wang
- Department of Dermatology, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Nanping Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Baochang Lai
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Ting Lei
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China; Department of Pathology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Qiongyu Wang
- Department of Dermatology, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Shengxiang Xiao
- Department of Dermatology, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Yongping Shao
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China; Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China.
| | - Yan Zheng
- Department of Dermatology, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
37
|
Zhao J, Li X, Yang Y, Zhu D, Zhang C, Liu D, Wu K, Zhao S. Effect of YAP1 silencing on esophageal cancer. Onco Targets Ther 2016; 9:3137-46. [PMID: 27307755 PMCID: PMC4888714 DOI: 10.2147/ott.s102338] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Background YAP1, the nuclear effector of the Hippo pathway, has become an attractive target for treatment of malignancies and is a candidate oncogene in esophageal cancer (EC). We hypothesized that knockdown of YAP1 could suppress EC and could be used for targeted therapy. However, there are few reports of the effect of YAP1 knockdown in EC. Materials and methods Quantitative real-time polymerase chain reaction and Western blot assays were performed to determine the expression levels of YAP1 mRNA and protein in primary EC tissue samples, EC cell lines, and controls. Immunohistochemistry was also performed to detect YAP1 protein expression in primary EC tumor and matched nontumor control tissues. YAP1-knockdown cell lines were constructed using short-hairpin RNA, and MTT, flow cytometry, and transwell chamber assays were used to analyze the effect of YAP1 knockdown on EC cell proliferation, apoptosis, and invasion. In vivo tumor formation assays were used to investigate the antitumor effect of YAP1 knockdown. Results We found that YAP1 mRNA and protein were upregulated in EC and that YAP1 expression correlated significantly with metastasis and tumor stage. We also found that YAP1 knockdown repressed cell proliferation and invasion and promoted apoptosis of EC cell lines. In addition, animal experiments revealed that YAP1 knockdown suppressed the growth of esophageal tumors in vivo. Conclusion Collectively, these data confirm our hypothesis that YAP1 knockdown suppresses EC and suggest that YAP1 knockdown could be exploited in the targeted gene therapy of EC in the future.
Collapse
Affiliation(s)
- Jia Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, People's Republic of China; Key Thoracic Tumour Experimental Laboratory of Zhengzhou, Zhengzhou, Henan, People's Republic of China
| | - Xiangnan Li
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, People's Republic of China; Key Thoracic Tumour Experimental Laboratory of Zhengzhou, Zhengzhou, Henan, People's Republic of China
| | - Yang Yang
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, People's Republic of China; Key Thoracic Tumour Experimental Laboratory of Zhengzhou, Zhengzhou, Henan, People's Republic of China
| | - Dengyan Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, People's Republic of China; Key Thoracic Tumour Experimental Laboratory of Zhengzhou, Zhengzhou, Henan, People's Republic of China
| | - Chunyang Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, People's Republic of China; Key Thoracic Tumour Experimental Laboratory of Zhengzhou, Zhengzhou, Henan, People's Republic of China
| | - Donglei Liu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, People's Republic of China; Key Thoracic Tumour Experimental Laboratory of Zhengzhou, Zhengzhou, Henan, People's Republic of China
| | - Kai Wu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, People's Republic of China; Key Thoracic Tumour Experimental Laboratory of Zhengzhou, Zhengzhou, Henan, People's Republic of China
| | - Song Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, People's Republic of China; Key Thoracic Tumour Experimental Laboratory of Zhengzhou, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
38
|
Taucher V, Mangge H, Haybaeck J. Non-coding RNAs in pancreatic cancer: challenges and opportunities for clinical application. Cell Oncol (Dordr) 2016; 39:295-318. [DOI: 10.1007/s13402-016-0275-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2016] [Indexed: 01/17/2023] Open
|
39
|
Jingjing L, Wangyue W, Qiaoqiao X, Jietong Y. MiR-218 increases sensitivity to cisplatin in esophageal cancer cells via targeting survivin expression. Open Med (Wars) 2016; 11:31-35. [PMID: 28352763 PMCID: PMC5329794 DOI: 10.1515/med-2016-0007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 01/07/2016] [Indexed: 02/07/2023] Open
Abstract
Objectives Increasing evidence showed that microRNAs (miRNAs) were implicated in the chemical resistance of human cancers. We intended to investigate the role of miR-218 in cisplatin sensitivity of esophageal cancer cells. Methods Quantitative real-time polymerase chain reaction (qRT-PCR) was carried out to analyze miR-218 expression in human esophageal cancer cell line Eca9706 and a cisplatin-resistant subline (ECa9706-CisR cells). The effects of miR-218 transfection on ECa9706 and ECa9706-CisR cell viability, including cell viability and apoptosis rate were confirmed using MTT assay, or flow cytometry, respectively. qRT-PCR was used to validate survivin as a direct target gene of miR-218 in our system. Results We found that miR-218 was significantly decreased in ECa9706-CisR cells compared with parent Eca9706 cells. Overexpression of miR-218 by mimics transfection would enhance cisplatin sensitivity evaluated by cell viability inhibition and apoptosis promotion. We validated here survivin as a direct target of miR-218 in ECa9706 cells, which might contribute to the chemoresistance of esophageal cancer cells to cisplatin. Conclusions In summary, our data suggest that miR-218 might represent as a promising sensitizer of cisplatin therapy in clinical esophageal cancer patients.
Collapse
Affiliation(s)
- Lin Jingjing
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, 325000 PR China
| | - Wang Wangyue
- Department of Gastroenterology, The People's Hospital of Lishui, 323000 PR China
| | - Xu Qiaoqiao
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, 325000 PR China
| | - Ye Jietong
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, 325000 PR China
| |
Collapse
|
40
|
Winther M, Knudsen S, Dahlgaard J, Jensen T, Hansen A, Jensen PB, Tramm T, Alsner J, Nordsmark M. Clinical Impact of a Novel MicroRNA Chemo-Sensitivity Predictor in Gastrooesophageal Cancer. PLoS One 2016; 11:e0148070. [PMID: 26885979 PMCID: PMC4757421 DOI: 10.1371/journal.pone.0148070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 01/12/2016] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND miRNAs might be potentially useful biomarkers for prediction of response to chemotherapeutic agents, radiotherapy and survival. The aim of this retrospective study was to validate miRNA response predictors in a cohort of patients with gastrooesophageal cancer in order to predict overall survival (OS) and disease-specific survival (DSS). MATERIAL AND METHODS The study population encompassed 53 patients treated with curative intend for loco-regional gastrooesophageal cancer. miRNA expression was quantified from pre-therapeutic and diagnostic, formalin-fixed, paraffin embedded tumour specimens using Affymetrix GeneChip miRNA 1.0 Array. Based on growth inhibition of the NCI60 panel in the presence of cisplatin, epirubicine and capecitabine, a miRNA based response predictor was developed. The Cox proportional hazards model was applied to assess the correlations of the response predictor with OS and DSS. RESULTS A univariate analysis demonstrated a statistical significant improvement of OS for patients who had undergone surgical resection with prediction scores above the median prediction score (HR: 0.41 (95% CI: 0.17-0.96). Adjusting for surgery and stage, this predictor was identified to be independently associated with both OS (HR: 0.37 (95% CI: 0.16-0.87)) and DSS (HR: 0.32 (0.12-0.87)). CONCLUSION The miRNA profile predictive for sensitivity to cisplatin, epirubicine and capecitabine was shown to be independently associated with OS and DSS in patients with gastrooesophageal cancer.
Collapse
Affiliation(s)
- Mette Winther
- Dept. of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
- * E-mail:
| | | | - Jesper Dahlgaard
- Centre for Health Promotion and Rehabilitation, Faculty of Health Sciences, VIA University College, Aarhus, Denmark
| | | | | | | | - Trine Tramm
- Dept. of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Jan Alsner
- Dept. of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
41
|
Lv JF, Hu L, Zhuo W, Zhang CM, Zhou HH, Fan L. Epigenetic alternations and cancer chemotherapy response. Cancer Chemother Pharmacol 2015; 77:673-84. [DOI: 10.1007/s00280-015-2951-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 12/17/2015] [Indexed: 12/29/2022]
|
42
|
Menschikowski M, Hagelgans A, Nacke B, Jandeck C, Sukocheva O, Siegert G. Epigenetic control of phospholipase A2 receptor expression in mammary cancer cells. BMC Cancer 2015; 15:971. [PMID: 26672991 PMCID: PMC4682251 DOI: 10.1186/s12885-015-1937-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 11/16/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND It has recently been proposed that the M-type phospholipase A2 receptor (PLA2R1) acts as a tumour suppressor in certain malignancies including mammary cancer. Considering that DNA methylation is an important regulator of gene transcription during carcinogenesis, in the current study we analyzed the PLA2R1 expression, PLA2R1 promoter methylation, and selected micro RNA (miRNA) levels in normal human mammary epithelial cells (HMEC) and cancer cell lines. METHODS Levels of PLA2R1 and DNA methyltransferases (DNMT) specific mRNA were determined using real-time RT-PCR. Methylation specific-high resolution melting (MS-HRM) analysis was utilized to quantify the methylation degree of selected CpG sites localized in the promoter region of the PLA2R1 gene. Expression of miRNA was tested using miScript Primer Assay system. RESULTS Nearly complete methylation of the analyzed PLA2R1 promoter region along with PLA2R1 gene silencing was identified in MDA-MB-453 mammary cancer cells. In MCF-7 and BT-474 mammary cancer cell lines, a higher DNA methylation degree and reduced PLA2R1 expression were found in comparison with those in normal HMEC. Synergistic effects of demethylating agent (5-aza-2'-deoxycytidine) and histone deacetylase inhibitor (trichostatin A) on PLA2R1 transcription in MDA-MB-453 cells confirmed the importance of DNA methylation and histone modification in the regulation of the PLA2R1 gene expression in mammary cells. Furthermore, significant positive correlation between the expression of DNMT1 and PLA2R1 gene methylation and negative correlation between the cellular levels of hsa-mir-141, -181b, and -181d-1 and the expression of PLA2R1 were identified in the analyzed cells. Analysis of combined z-score of miR-23b, -154 and -302d demonstrated a strong and significant positive correlation with PLA2R1 expression. CONCLUSIONS Our data indicate that (i) PLA2R1 expression in breast cancer cells is controlled by DNA methylation and histone modifications, (ii) hypermethylation of the PLA2R1 promoter region is associated with up-regulation of DNMT1, and (iii) hsa-miR-23b, -154, and -302d, as well as hsa-miR-141, -181b, and -181d-1 are potential candidates for post-transcriptional regulation of PLA2R1 expression in mammary cancer cells.
Collapse
Affiliation(s)
- Mario Menschikowski
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty "Carl Gustav Carus", Technical University of Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| | - Albert Hagelgans
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty "Carl Gustav Carus", Technical University of Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| | - Brit Nacke
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty "Carl Gustav Carus", Technical University of Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| | - Carsten Jandeck
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty "Carl Gustav Carus", Technical University of Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| | - Olga Sukocheva
- School of Health Sciences, Flinders University of South Australia, Bedford Park, SA, 5042, Australia.
| | - Gabriele Siegert
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty "Carl Gustav Carus", Technical University of Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| |
Collapse
|
43
|
14-3-3σ confers cisplatin resistance in esophageal squamous cell carcinoma cells via regulating DNA repair molecules. Tumour Biol 2015; 37:2127-36. [PMID: 26346170 DOI: 10.1007/s13277-015-4018-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 08/28/2015] [Indexed: 02/07/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the predominant type of esophageal cancer in Asia. Cisplatin is commonly used in chemoradiation for unresectable ESCC patients. However, the treatment efficacy is diminished in patients with established cisplatin resistance. To understand the mechanism leading to the development of cisplatin resistance in ESCC, we compared the proteomes from a cisplatin-resistant HKESC-2R cell line with its parental-sensitive counterpart HKESC-2 to identify key molecule involved in this process. Mass spectrometry analysis detected 14-3-3σ as the most abundant molecule expressed exclusively in HKESC-2R cells, while western blot result further validated it to be highly expressed in HKESC-2R cells when compared to HKESC-2 cells. Ectopic expression of 14-3-3σ increased cisplatin resistance in HKESC-2 cells, while its suppression sensitized SLMT-1 cells to cisplatin. Among the molecules involved in drug detoxification, drug transportation, and DNA repair, the examined DNA repair molecules HMGB1 and XPA were found to be highly expressed in HKESC-2R cells with high 14-3-3σ expression. Subsequent manipulation of 14-3-3σ by both overexpression and knockdown approaches concurrently altered the expression of HMGB1 and XPA. 14-3-3σ, HMGB1, and XPA were preferentially expressed in cisplatin-resistant SLMT-1 cells when compared to those more sensitive to cisplatin. In ESCC patients with poor response to cisplatin-based chemoradiation, their pre-treatment tumors expressed higher expression of HMGB1 than those with response to such treatment. In summary, our results demonstrate that 14-3-3σ induces cisplatin resistance in ESCC cells and that 14-3-3σ-mediated cisplatin resistance involves DNA repair molecules HMGB1 and XPA. Results from this study provide evidences for further work in researching the potential use of 14-3-3σ and DNA repair molecules HMGB1 and XPA as biomarkers and therapeutic targets for ESCC.
Collapse
|
44
|
Cetuximab plus pemetrexed as second-line therapy for fluorouracil-based pre-treated metastatic esophageal squamous cell carcinoma. Cancer Chemother Pharmacol 2015; 76:829-34. [PMID: 26330330 DOI: 10.1007/s00280-015-2854-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 08/24/2015] [Indexed: 01/06/2023]
Abstract
PURPOSE The aim of this study was to evaluate the efficacy and toxicity of cetuximab and pemetrexed as the second-line treatment for advanced esophageal cancer patients, who had undergone treatment with the standard cisplatin and 5-FU regimens. METHODS A total of 29 patients accepted this treatment. Cetuximab was administered at an initial dose of 400 mg/m(2) IV followed by weekly doses of 250 mg/m(2) IV, whereas pemetrexed 500 mg/m(2) was administered on day 1 every 3 weeks. RESULTS Twelve patients (41.4 %) presented PR, ten (34.5 %) had SD, and seven (24.1 %) had PD. The response rate was 41.4 %. The median PFS was 4.5 months (95 % CI 4.1-4.9), and the median OS was 9.4 months (95 % CI 8.4-10.5). The expression levels of serum miR-335 were determined, and the subsequent analysis suggested that PFS was significantly different between the patients with higher level of serum miR-335 and those with low level of serum miR-335. The former had a PFS of 4.9 months (95 % CI 4.2-5.7) and the latter 4.1 months (95 % CI 3.7-4.5) (log rank = 0.025). None of the patients experienced grade 4 toxicity. Twenty-four patients presented rash. CONCLUSIONS A combination of cetuximab plus pemetrexed was marginally effective and well tolerated in patients with advanced esophageal squamous cell carcinoma as the second-line treatment.
Collapse
|
45
|
Xu F, Ahmed ASI, Kang X, Hu G, Liu F, Zhang W, Zhou J. MicroRNA-15b/16 Attenuates Vascular Neointima Formation by Promoting the Contractile Phenotype of Vascular Smooth Muscle Through Targeting YAP. Arterioscler Thromb Vasc Biol 2015; 35:2145-52. [PMID: 26293467 DOI: 10.1161/atvbaha.115.305748] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/10/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To investigate the functional role of the microRNA (miR)-15b/16 in vascular smooth muscle (SM) phenotypic modulation. APPROACH AND RESULTS We found that miR-15b/16 is one of the most abundant mRs expressed in contractile vascular smooth muscle cells (VSMCs). However, when contractile VSMCs get converted to a synthetic phenotype, miR-15b/16 expression is significantly reduced. Knocking down endogenous miR-15b/16 in VSMCs attenuates SM-specific gene expression but promotes VSMC proliferation and migration. Conversely, overexpression of miR-15b/16 promotes SM contractile gene expression while attenuating VSMC migration and proliferation. Consistent with this, overexpression of miR-15b/16 in a rat carotid balloon injury model markedly attenuates injury-induced SM dedifferentiation and neointima formation. Mechanistically, we identified the potent oncoprotein yes-associated protein (YAP) as a downstream target of miR-15b/16 in VSMCs. Reporter assays validated that miR-15b/16 targets YAP's 3' untranslated region. Moreover, overexpression of miR-15b/16 significantly represses YAP expression, whereas conversely, depletion of endogenous miR-15b/16 results in upregulation of YAP expression. CONCLUSIONS These results indicate that miR-15b/16 plays a critical role in SM phenotypic modulation at least partly through targeting YAP. Restoring expression of miR-15b/16 would be a potential therapeutic approach for treatment of proliferative vascular diseases.
Collapse
Affiliation(s)
- Fei Xu
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Abu Shufian Ishtiaq Ahmed
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Xiuhua Kang
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Guoqing Hu
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Fang Liu
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Wei Zhang
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Jiliang Zhou
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.).
| |
Collapse
|
46
|
Sharma P, Sharma R. miRNA-mRNA crosstalk in esophageal cancer: From diagnosis to therapy. Crit Rev Oncol Hematol 2015; 96:449-62. [PMID: 26257289 DOI: 10.1016/j.critrevonc.2015.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 04/11/2015] [Accepted: 07/07/2015] [Indexed: 12/11/2022] Open
Abstract
The asymptomatic nature of esophageal cancer (EC) at early stages results in late clinical presentation leading to poor prognosis and limited success of therapeutic modalities. Efforts to identify diagnostic/prognostic markers have proven to be unsuccessful for translation into clinics. Hence, there is a pressing need for establishment of novel non-invasive biomarker for early diagnosis/better prognosis of EC. Recently, alteration in microRNA (miRNA) expression has emerged as an important hallmark of cancer. This review summarizes the differential expression of miRNAs in EC and addresses how their aberrant expression influences crucial biological processes such as apoptosis, cell proliferation, invasion and metastasis. Additionally, this review highlights the current status of circulating miRNA based diagnostic/prognostic markers. An effort has been made to find a connection between different miRNAs involved in EC and a detailed analysis has been done to screen out micoRNAs involved in prognosis and multidrug resistance. Further, investigation of these miRNAs would not only provide a gene therapy based strategy to prevent/treat cancer but also to reverse multidrug resistance leading to decreased requirement of harmful chemotherapeutic drugs.
Collapse
Affiliation(s)
- Priyanka Sharma
- Research Scholar, University School of Biotechnology, Guru Gobind Singh Indraprastha University, New Delhi 110078, India.
| | - Rinu Sharma
- Assistant Professor, University School of Biotechnology, Guru Gobind Singh Indraprastha University, Sector 16C Dwarka, New Delhi 110078, India.
| |
Collapse
|
47
|
Ciamporcero E, Shen H, Ramakrishnan S, Yu Ku S, Chintala S, Shen L, Adelaiye R, Miles KM, Ullio C, Pizzimenti S, Daga M, Azabdaftari G, Attwood K, Johnson C, Zhang J, Barrera G, Pili R. YAP activation protects urothelial cell carcinoma from treatment-induced DNA damage. Oncogene 2015; 35:1541-53. [PMID: 26119935 PMCID: PMC4695331 DOI: 10.1038/onc.2015.219] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 04/09/2015] [Accepted: 04/28/2015] [Indexed: 12/31/2022]
Abstract
Current standard of care for muscle-invasive urothelial cell carcinoma (UCC) is surgery along with perioperative platinum-based chemotherapy. UCC is sensitive to cisplatin-based regimens, but acquired resistance eventually occurs, and a subset of tumors is intrinsically resistant. Thus, there is an unmet need for new therapeutic approaches to target chemotherapy-resistant UCC. Yes-associated protein (YAP) is a transcriptional co-activator that has been associated with bladder cancer progression and cisplatin resistance in ovarian cancer. In contrast, YAP has been shown to induce DNA damage associated apoptosis in non-small cell lung carcinoma. However, no data have been reported on the YAP role in UCC chemo-resistance. Thus, we have investigated the potential dichotomous role of YAP in UCC response to chemotherapy utilizing two patient-derived xenograft models recently established. Constitutive expression and activation of YAP inversely correlated with in vitro and in vivo cisplatin sensitivity. YAP overexpression protected while YAP knock-down sensitized UCC cells to chemotherapy and radiation effects via increased accumulation of DNA damage and apoptosis. Furthermore, pharmacological YAP inhibition with verteporfin inhibited tumor cell proliferation and restored sensitivity to cisplatin. In addition, nuclear YAP expression was associated with poor outcome in UCC patients who received perioperative chemotherapy. In conclusion, these results suggest that YAP activation exerts a protective role and represents a pharmacological target to enhance the anti-tumor effects of DNA damaging modalities in the treatment of UCC.
Collapse
Affiliation(s)
- E Ciamporcero
- Department of Medicine, Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - H Shen
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - S Ramakrishnan
- Department of Medicine, Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Cancer Pathology & Prevention, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - S Yu Ku
- Department of Medicine, Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Cancer Pathology & Prevention, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - S Chintala
- Department of Medicine, Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Pharmacology & Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - L Shen
- Department of Medicine, Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - R Adelaiye
- Department of Medicine, Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Cancer Pathology & Prevention, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - K M Miles
- Department of Medicine, Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - C Ullio
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - S Pizzimenti
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - M Daga
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - G Azabdaftari
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - K Attwood
- Department of Biostatistics & Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - C Johnson
- Department of Medicine, Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Pharmacology & Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - J Zhang
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - G Barrera
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - R Pili
- Department of Medicine, Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Medicine, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
48
|
Zheng Y, Li S, Boohaker RJ, Liu X, Zhu Y, Zhai L, Li H, Gu F, Fan Y, Lang R, Liu F, Qian X, Xu B, Fu L. A MicroRNA Expression Signature In Taxane-anthracycline-Based Neoadjuvant Chemotherapy Response. J Cancer 2015; 6:671-7. [PMID: 26078798 PMCID: PMC4466417 DOI: 10.7150/jca.11616] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 05/06/2015] [Indexed: 02/06/2023] Open
Abstract
There is an unmet clinical need to identify biomarkers for breast cancer neoadjuvant chemotherapy. Here, using miRNA TaqMan Low-Density Arrays (TLDA), we analyzed the miRNA expression profile in pre-treatment needle aspiration tumor samples from patients who received taxane-anthracycline-based neoadjuvant chemotherapy. Although, in an unsupervised hierarchical cluster analysis, the total miRNA expression profile could not generate a tree with clear distinction between pathologic complete response (pCR) and non-pCR classes, we found that elevated expression of miR-125b and miR-141 was associated with non-pCR. In vitro experiments indicated that inhibition of miR-125b and miR-141 expression reduced cellular survival in response to taxane-anthracycline treatment. Furthermore, co-transfection with miR-125b and miR-141 mimics increased resistance of MCF7 and BT549 cells to taxane-anthracycline induced cytotoxicity. Pathway analyses indicated that many of the target proteins of miR-125b are involved in apoptotic pathways and cell cycle control. Together, we provide evidence that elevated miR-125b and 141 expression predicts a poor clinical responsiveness of taxane-anthracycline-based neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Yi Zheng
- 1. Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; State Key Laboratory of Breast Cancer Research, Hexi District, Tianjin, 300060, China
| | - Shuai Li
- 1. Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; State Key Laboratory of Breast Cancer Research, Hexi District, Tianjin, 300060, China
| | - Rebecca J Boohaker
- 3. Department of Oncology, Southern Research Institute, Birmingham, AL 35205, USA
| | - Xinli Liu
- 1. Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; State Key Laboratory of Breast Cancer Research, Hexi District, Tianjin, 300060, China
| | - Yufen Zhu
- 1. Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; State Key Laboratory of Breast Cancer Research, Hexi District, Tianjin, 300060, China
| | - Lili Zhai
- 1. Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; State Key Laboratory of Breast Cancer Research, Hexi District, Tianjin, 300060, China
| | - Huilan Li
- 1. Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; State Key Laboratory of Breast Cancer Research, Hexi District, Tianjin, 300060, China
| | - Feng Gu
- 1. Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; State Key Laboratory of Breast Cancer Research, Hexi District, Tianjin, 300060, China
| | - Yu Fan
- 1. Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; State Key Laboratory of Breast Cancer Research, Hexi District, Tianjin, 300060, China
| | - Ronggang Lang
- 1. Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; State Key Laboratory of Breast Cancer Research, Hexi District, Tianjin, 300060, China
| | - Fangfang Liu
- 1. Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; State Key Laboratory of Breast Cancer Research, Hexi District, Tianjin, 300060, China
| | - Xiaolong Qian
- 1. Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; State Key Laboratory of Breast Cancer Research, Hexi District, Tianjin, 300060, China
| | - Bo Xu
- 3. Department of Oncology, Southern Research Institute, Birmingham, AL 35205, USA
| | - Li Fu
- 1. Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; State Key Laboratory of Breast Cancer Research, Hexi District, Tianjin, 300060, China ; 2. 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics
| |
Collapse
|
49
|
Tanaka K, Miyata H, Sugimura K, Fukuda S, Kanemura T, Yamashita K, Miyazaki Y, Takahashi T, Kurokawa Y, Yamasaki M, Wada H, Nakajima K, Takiguchi S, Mori M, Doki Y. miR-27 is associated with chemoresistance in esophageal cancer through transformation of normal fibroblasts to cancer-associated fibroblasts. Carcinogenesis 2015; 36:894-903. [PMID: 26026166 DOI: 10.1093/carcin/bgv067] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 04/20/2015] [Indexed: 02/07/2023] Open
Abstract
There is increasing evidence that the expression of microRNA (miRNA) in cancer is associated with chemosensitivity but the mechanism of miRNA-induced chemoresistance has not been fully elucidated. The aim of this study was to examine the role of extracellular miRNA in the response to chemotherapy in esophageal cancer. First, serum expression of miRNAs selected by miRNA array was measured by quantitative reverse transcription-polymerase chain reaction in 68 patients with esophageal cancer who received cisplatin-based chemotherapy to examine the relationship between miRNA expression and response to chemotherapy. The serum expression levels of 18 miRNAs were different between responders and non-responders by miRNA array. Of these, high expression levels of miR-27a/b correlated with poor response to chemotherapy in patients with esophageal cancer. Next, in vitro assays were conducted to investigate the mechanism of miRNA-induced chemoresistance. Although transfection of miR-27a/b to cancer cells had no significant impact on chemosensitivity, esophageal cancer cells cultured in supernatant of miR-27a/b-transfected normal fibroblast showed reduced chemosensitivity to cisplatin, compared with cancer cells cultured in supernatant of normal fibroblast. MiR-27a/b-transfected normal fibroblast showed α-smooth muscle actin (α-SMA) expression, a marker of cancer-associated fibroblasts (CAF) and increased production of transforming growth factor-β (TGF-β). Chemosensitivity recovered after administration of neutralizing antibody of TGF-β to the supernatant transfer experiments. Our results indicated that miR-27a/b is involved in resistance to chemotherapy in esophageal cancer, through miR-27a/b-induced transformation of normal fibroblast into CAF.
Collapse
Affiliation(s)
- Koji Tanaka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Hiroshi Miyata
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Keijiro Sugimura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Shuichi Fukuda
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Takashi Kanemura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Kotaro Yamashita
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Yasuhiro Miyazaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Makoto Yamasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Hisashi Wada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Kiyokazu Nakajima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| |
Collapse
|
50
|
Lin G, Xu K. [Advances in tumor chemo-resistance regulated by MicroRNA]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2015; 17:741-9. [PMID: 25342041 PMCID: PMC6000396 DOI: 10.3779/j.issn.1009-3419.2014.10.06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chemotherapy is one of the primary treatment for malignant tumors. Tumor multidrug resistance (MDR) is a major cause of clinical failure of chemotherapy; however the mechanisms of chemo-resistance have not been fully elucidated. Recently, microRNA is one of the new hotspots in life science. MicroRNA regulates the expression of genes and plays roles a series of life events by post-transcriptional regulations, including cell proliferation, apoptosis, fat metabolism, nervous development, hormone secretion, tumor vessels generation, stem cell differentiation, tumor cell invasion and metastasis, and other physiological and pathological processes. Recent studies show that microRNA regulates the expression of multiple genes with high efficiency and specificity. The abnormal regulation of target genes by microRNA is responsible for tumor chemo-resistance, this may be an important component of the complexity of the regulation of chemo-resistance. Therefore, the study of microRNA and tumor drug resistance has profound practical significance. In this review, recent studies of tumor drug resistance, regulation of tumor drug resistance by microRNA, and microRNA as a potential target for tumor drug resistance therapy are reviewed.
Collapse
Affiliation(s)
- Gaoyang Lin
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute,
Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ke Xu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute,
Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|