1
|
Hedayati N, Safaei Naeini M, Ale Sahebfosoul MM, Mafi A, Eshaghi Milasi Y, Rizaneh A, Nabavi N, Farahani N, Alimohammadi M, Ghezelbash B. MicroRNA dysregulation and its impact on apoptosis-related signaling pathways in myelodysplastic syndrome. Pathol Res Pract 2024; 261:155478. [PMID: 39079383 DOI: 10.1016/j.prp.2024.155478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 08/18/2024]
Abstract
Myelodysplastic syndrome (MDS) holds a unique position among blood cancers, encompassing a spectrum of blood-related disorders marked by impaired maturation of blood cell precursors, bone marrow abnormalities, genetic instability, and a higher likelihood of progressing to acute myeloid leukemia. MicroRNAs (miRNAs), short non-coding RNA molecules typically 18-24 nucleotides in length, are known to regulate gene expression and contribute to various biological processes, including cellular differentiation and programmed cell death. Additionally, miRNAs are involved in many aspects of cancer development, influencing cell growth, transformation, and apoptosis. In this study, we explore the impact of microRNAs on cellular apoptosis in MDS.
Collapse
Affiliation(s)
- Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Mobina Safaei Naeini
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran; Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yaser Eshaghi Milasi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Anahita Rizaneh
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, Canada.
| | - Najma Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Behrooz Ghezelbash
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
2
|
Zhang Y, Wu Y, Shi X, Ding H, Zhou Y, Chen H, Shen F, Chen Y, Zhou J, Zhou D, Wang J. M6A-mediated hsa_circ_0061179 inhibits DNA damage in ovarian cancer cells via miR-143-3p/TIMELESS. Mol Carcinog 2024; 63:1542-1558. [PMID: 38751015 DOI: 10.1002/mc.23744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 02/29/2024] [Accepted: 05/02/2024] [Indexed: 07/10/2024]
Abstract
Ovarian cancer (OC) is among the most common and deadly solid malignancies in women. Despite many advances in OC research, the incidence of OC continues to rise, and its pathogenesis remains largely unknown. Herein, we elucidated the function of hsa_circ_0061179 in OC. The levels of hsa_circ_0061179, miR-143-3p, TIMELESS, and DNA damage repair-related proteins in OC or normal ovarian tissues and cells were measured using real-time quantitative polymerase chain reaction and immunoblotting. The biological effects of hsa_circ_0061179 and miR-143-3p on proliferation, clone formation, DNA damage, and apoptosis of OC cells were detected by the cell counting kit-8 assay, 5-methylethyl-2'-deoxyuridine, flow cytometry, the comet assay, and immunofluorescence staining combined with the confocal microscopy. The interaction among hsa_circ_0061179, miR-143-3p, and TIMELESS was validated by the luciferase reporter assay. Mice tumor xenograft models were used to evaluate the influence of hsa_circ_0061179 on OC growth in vivo. We found that human OC biospecimens expressed higher levels of hsa_circ_0061179 and lower levels of miR-143-3p. Hsa_circ_0061179 was found to bind with miR-143-3p, which directly targets TIMELESS. Hsa_circ_0061179 knockdown or miR-143-3p overexpression suppressed the proliferation and clone formation of OC cells and increased DNA damage and apoptosis of OC cells via the miR-143-3p/TIMELESS axis. Furthermore, we demonstrated that METTL3 could direct the formation of has_circ_0061179 through a specific m6A modification site. YTHDC1 facilitated the cytoplasmic transfer of has_circ_0061179 by directly binding to the modified m6A site. Our findings suggest that hsa_circ_0061179 acts as the sponge of miR-143-3p to activate TIMELESS signaling and inhibits DNA damage and apoptosis in OC cells.
Collapse
Affiliation(s)
- Yuhong Zhang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yuhong Wu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiu Shi
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hongmei Ding
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hanqing Chen
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of hematology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fangrong Shen
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Youguo Chen
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jinhua Zhou
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Dingjie Zhou
- NHC Contraceptive Adverse Reaction Surveillance Center, Jiangsu Health Development Research Center, Nanjing, Jiangsu, China
- Jiangsu Provincial Medical Key Laboratory of Fertility Protection and Health Technology Assessment, Nanjing, Jiangsu, China
| | - Juan Wang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
3
|
Zhang R, Gao Y, Wang S, Pan J, Geng S, Li Z, Zhang K, Meng W. Detection of miRNA-378based on a catalytic hairpin self-assembly reaction combined with gold nanoparticle colorimetry. NANOTECHNOLOGY 2024; 35:355602. [PMID: 38821044 DOI: 10.1088/1361-6528/ad5297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/31/2024] [Indexed: 06/02/2024]
Abstract
Recent studies have shown that abnormalmiRNA-378expression is a rule, rather than an exception, in cervical cancer and can be used as a diagnostic and prognostic biomarker to assess tumor initiation. In this study, we developed a general, sensitive strategy for detectingmiRNA-378using catalytic hairpin self-assembly (CHA) combined with gold nanoparticles (AuNP) colorimetry. The presence ofmiRNA-378triggers the repeated self-assembly of two designed hairpin DNAs (H1 and H2) into dsDNA polymers, which leads to changes in the surface plasmon resonance absorption band and the macroscopic color of the AuNP colloids due to the formation of nanoparticle-DNA conjugates. This experimental phenomenon can be observed by ultraviolet-visible spectrometry or even with the naked eye. Using this method,miRNA-378could be quantitatively detected at the picomolar level (as low as 20.7 pM). Compared with traditional methods, such as quantitative polymerase chain reaction and RNA blotting, this strategy has a simple operation, low cost, and high sensitivity and selectivity, and thus, exhibits significant potential for miRNA detection.
Collapse
Affiliation(s)
- Run Zhang
- Key Laboratory of Biomedical Functional Materials, School of Sciences, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yahui Gao
- Key Laboratory of Biomedical Functional Materials, School of Sciences, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Shan Wang
- Key Laboratory of Biomedical Functional Materials, School of Sciences, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Jinru Pan
- Key Laboratory of Biomedical Functional Materials, School of Sciences, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Shuang Geng
- NO. 3 Middle School of Cangzhou, No. 126, Gongnong Road, Xinhua District, Cangzhou 061000, People's Republic of China
| | - Zhen Li
- Tongji Hospital Tongji Medical College of Hust, No. 501, Gaoxin Avenue, East Lake New Technology Development Zone, Wuhan 430030, People's Republic of China
| | - Kejie Zhang
- School of Materials Science and Engineering, Nanjing Institute of Technology, No. 1, Hongjing Road, Jiangning District, Nanjing 211167, People's Republic of China
| | - Wei Meng
- Key Laboratory of Biomedical Functional Materials, School of Sciences, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| |
Collapse
|
4
|
Shen L, Li A, Cui J, Liu H, Zhang S. Integration of single-cell RNA-seq and bulk RNA-seq data to construct and validate a cancer-associated fibroblast-related prognostic signature for patients with ovarian cancer. J Ovarian Res 2024; 17:82. [PMID: 38627854 PMCID: PMC11020192 DOI: 10.1186/s13048-024-01399-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 03/21/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND To establish a prognostic risk profile for ovarian cancer (OC) patients based on cancer-associated fibroblasts (CAFs) and gain a comprehensive understanding of their role in OC progression, prognosis, and therapeutic efficacy. METHODS Data on OC single-cell RNA sequencing (scRNA-seq) and total RNA-seq were collected from the GEO and TCGA databases. Seurat R program was used to analyze scRNA-seq data and identify CAFs clusters corresponding to CAFs markers. Differential expression analysis was performed on the TCGA dataset to identify prognostic genes. A CAF-associated risk signature was designed using Lasso regression and combined with clinicopathological variables to develop a nomogram. Functional enrichment and the immune landscape were also analyzed. RESULTS Five CAFs clusters were identified in OC using scRNA-seq data, and 2 were significantly associated with OC prognosis. Seven genes were selected to develop a CAF-based risk signature, primarily associated with 28 pathways. The signature was a key independent predictor of OC prognosis and relevant in predicting the results of immunotherapy interventions. A novel nomogram combining CAF-based risk and disease stage was developed to predict OC prognosis. CONCLUSION The study highlights the importance of CAFs in OC progression and suggests potential for innovative treatment strategies. A CAF-based risk signature provides a highly accurate prediction of the prognosis of OC patients, and the developed nomogram shows promising results in predicting the OC prognosis.
Collapse
Affiliation(s)
- Liang Shen
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, 67 Dongchang West Road, Liaocheng, Shandong, 252000, P.R. China
- Shandong University, Jinan, P.R. China
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, Shandong, 250021, P.R. China
| | - Aihua Li
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, 67 Dongchang West Road, Liaocheng, Shandong, 252000, P.R. China.
| | - Jing Cui
- Department of Oral and Maxillofacial Surgery, Jinan Stomatology Hospital, 101 Jingliu Road, Jinan, Shandong, 250001, P.R. China
- Central Laboratory of Jinan Stamotological Hospital, Jinan Key Laboratory of Oral Tissue Regeneration, 101 Jingliu Road, Jinan, Shandong, 250001, P.R. China
| | - Haixia Liu
- Department of Obstetrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, Shandong, 250021, P.R. China
| | - Shiqian Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, P.R. China.
| |
Collapse
|
5
|
Dharshini LCP, Mandal AKA. Regulation of gene expression by modulating microRNAs through Epigallocatechin-3-gallate in cancer. Mol Biol Rep 2024; 51:230. [PMID: 38281210 DOI: 10.1007/s11033-023-09145-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/12/2023] [Indexed: 01/30/2024]
Abstract
Cancer is an intricate ailment that has a higher death rate globally and is characterized by aberrant cell proliferation and metastasis in nature. Since the beginning of healthcare, natural products, especially those derived from plants, have been utilized to support human health. Green tea contains an essential catechin called epigallocatechin gallate, which has anti-proliferative, anti-mutagenic, anti-inflammatory, and antioxidative properties. The anticancer properties of EGCG have been extensively studied using pre-clinical cell culture and animal model systems. Dysregulated miRNA may be a biomarker since it influences the different characteristics of cancer like upholding proliferative signaling, cell death, invasiveness, metastasis, and angiogenesis. EGCG either elevates or lowers the expression of dysregulated miRNAs in cancer. Nonetheless, due to its anticancer properties, greater attention has been paid towards the development of efficient strategies for utilizing EGCG in cancer chemotherapy. This review summarizes the modifying effect of EGCG on miRNAs in cancer after briefly discussing the anticancer mechanisms of EGCG and the function of miRNAs in cancer.
Collapse
Affiliation(s)
| | - Abul Kalam Azad Mandal
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| |
Collapse
|
6
|
Han Z, Wang Y, Han L, Yang C. RPN2 in cancer: An overview. Gene 2023; 857:147168. [PMID: 36621657 DOI: 10.1016/j.gene.2023.147168] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/07/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
Oncogenes together with tumor suppresser genes are confirmed to regulate tumor phenotype in human cancers. RPN2, widely verified as an oncogene, encodes a protein that is part of an N-oligosaccharyl transferase, and is observed to be aberrantly expressed in human malignancies. Accumulating evidence unveils the vital functions of RPN2, contributing to tumorigenicity, metastasis, progression, and multi-drug resistance. Furthermore, previous studies partly indicated that RPN2 was involved in tumor progression via contributing to N-glycosylation and regulating multiple signaling pathways. In addition, RPN2 was also confirmed as a downstream target involved in tumor progression. Moreover, with demonstrated prognosis value and therapeutic target, RPN2 was also determined as a promising biomarker for forecasting patients' prognostic and therapy efficacy. In the present review, we aimed to summarize the present studies of RPN2 in cancer, and enhance the understanding of RPN2's extensive functions and clinical significances.
Collapse
Affiliation(s)
- Zhengxuan Han
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Tumour Biological Behaviours, Wuhan, China; Hubei Cancer Clinical Study Center, Wuhan, China; The Clinical Medical Research Center of Peritoneal Cancer of Wuhan, Wuhan, China
| | - You Wang
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lei Han
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Tumour Biological Behaviours, Wuhan, China; Hubei Cancer Clinical Study Center, Wuhan, China; The Clinical Medical Research Center of Peritoneal Cancer of Wuhan, Wuhan, China
| | - Chaogang Yang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Tumour Biological Behaviours, Wuhan, China; Hubei Cancer Clinical Study Center, Wuhan, China; The Clinical Medical Research Center of Peritoneal Cancer of Wuhan, Wuhan, China.
| |
Collapse
|
7
|
Qin Y, Liang R, Lu P, Lai L, Zhu X. Depicting the Implication of miR-378a in Cancers. Technol Cancer Res Treat 2022; 21:15330338221134385. [PMID: 36285472 PMCID: PMC9608056 DOI: 10.1177/15330338221134385] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
MicroRNA-378a (miR-378a), including miR-378a-3p and miR-378a-5p, are encoded in PPARGC1B gene. miR-378a is essential for tumorigenesis and is an independent prognostic biomarker for various malignant tumors. Aberrant expression of miR-378a affects several physiological and pathological processes, including proliferation, apoptosis, tumorigenesis, cancer invasion, metastasis, and therapeutic resistance. Interestingly, miR-378a has a dual functional role in either promoting or inhibiting tumorigenesis, independent of the cancer type. In this review, we comprehensively summarized the role and regulatory mechanisms of miR-378a in cancer development, hoping to provide a direction for its potential use in cancer therapy.
Collapse
Affiliation(s)
- Yuelan Qin
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Renba Liang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Pingan Lu
- Faculty of Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Lin Lai
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Xiaodong Zhu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China,Affiliated Wuming Hospital of Guangxi Medical University, Nanning, People's Republic of China,Key Laboratory of Early Prevention and Treatment for Regional High-Incidence-Tumor, Guangxi Medical University, Ministry of Education, Nanning, People's Republic of China,Xiaodong Zhu, Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, 22 Shuang Yong Road, Nanning 530021, People's Republic of China.
| |
Collapse
|
8
|
Piechowska A, Kruszniewska-Rajs C, Kimsa-Dudek M, Kołomańska M, Strzałka-Mrozik B, Gola J, Głuszek S. The role of miR-370 and miR-138 in the regulation of BMP2 suppressor gene expression in colorectal cancer: preliminary studies. J Cancer Res Clin Oncol 2022; 148:1569-1582. [PMID: 35292840 DOI: 10.1007/s00432-022-03977-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/06/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE Colorectal cancer (CRC) is the fourth-most common cancer worldwide and the second most common cancer cause of death in the world. The components of the TGFβ-signalling pathway, which are often affected by miRNAs, are involved in the regulation of apoptosis and cell cycle. Therefore, in the current study, the expression of BMP2 gene in CRC tissues at different clinical stages compared to the non-tumour tissues has been assessed. Moreover, the plasma BMP2 protein concentration in the same group of CRC patients has been validated. Due to the constant necessity to conduct further research of the correlation between specific miRNAs and mRNAs in CRC, in silico analysis has been performed to select miRNAs that regulate BMP2 mRNA. METHODS The cDNA samples from tumor and non-tumor tissue were used in a qPCR reaction to determine the mRNA expression of the BMP2 gene and the expression of selected miRNAs. The concentration of BMP2 protein in plasma samples was also measured. RESULTS It was indicated that BMP2 was downregulated in CRC tissue. Moreover, miR-370 and miR-138 expression showed an upward trend. Decreased BMP2 with accompanied increasing miR-370 and miR-138 expression was relevant to the malignant clinicopathological features of CRC and consequently poor patient prognosis. CONCLUSION Our data suggest that miR-370 with its clear expression in plasma samples may be a potential diagnostic marker to determine the severity of the disease in patients at a later stage of colorectal cancer.
Collapse
Affiliation(s)
- Agnieszka Piechowska
- Department of Surgical Medicine With the Laboratory of Medical Genetics, Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University, Kielce, Poland
| | - Celina Kruszniewska-Rajs
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jednosci 8, 41-200, Sosnowiec, Poland
| | - Magdalena Kimsa-Dudek
- Department of Nutrigenomics and Bromatology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jednosci 8, 41-200, Sosnowiec, Poland
| | - Magdalena Kołomańska
- Department of Anatomy, Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University, Kielce, Poland
| | - Barbara Strzałka-Mrozik
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jednosci 8, 41-200, Sosnowiec, Poland.
| | - Joanna Gola
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jednosci 8, 41-200, Sosnowiec, Poland
| | - Stanisław Głuszek
- Department of Surgical Medicine With the Laboratory of Medical Genetics, Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University, Kielce, Poland.,Department of Clinic General Oncological and Endocrinological Surgery, Regional Hospital, Kielce, Poland
| |
Collapse
|
9
|
Gao P, Zeng X, Zhang L, Wang L, Shen LL, Hou YY, Zhou F, Zhang X. Overexpression of miR-378 Alleviates Chronic Sciatic Nerve Injury by Targeting EZH2. Neurochem Res 2021; 46:3213-3221. [PMID: 34406548 DOI: 10.1007/s11064-021-03424-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 07/12/2021] [Accepted: 08/06/2021] [Indexed: 11/25/2022]
Abstract
In numerous studies, microRNAs (miRNAs) have been authenticated to play vital roles in the pathophysiology of neuropathic pain and other neurological diseases. In our study, we focused on evaluating miR-378 and its potential effects in neuropathic pain development, as well as the underlying molecular mechanisms. Primarily, a chronic sciatic nerve injury (CCI) rat model was established. Next, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was employed to measure the expression levels of miR-378 and EZH2 mRNA; the EZH2 protein expression levels were detected by western blot. A luciferase activity assay monitored the interaction of miR-378 and EZH2. Mechanical and thermal hyperalgesia was also performed to quantitate the effects of overexpression of miR-378 or EZH2 on the CCI rats. We found that miR-378 was down-regulated in the CCI rats, and the overexpression of miR-378 produced significant relief in their pain management. EZH2 was the downstream gene of miR-378 and was negatively regulated by miR-378. The up-regulation of EZH2 reduced the inhibitory effects of miR-378 on the development of neuropathic pain in the CCI rats. miR-378 acts as an inhibitor in the progression of neuropathic pain via targeting EZH2; the miR-378/EZH2 axis may be a novel target for the diagnosis and therapy of neuropathic pain in clinical treatment.
Collapse
Affiliation(s)
- Pengfei Gao
- Department of Anesthesiology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xin Zeng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Hubei Province, Wuhan, China
| | - Lin Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Hubei Province, Wuhan, China
| | - Long Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Hubei Province, Wuhan, China
| | - Lu-Lu Shen
- Department of Anesthesiology, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, 66 Huaihai South Road, Huai'an, Jiangsu, China
| | - Ya-Yun Hou
- Department of Anesthesiology, Huai'an Hospital of Traditional Chinese Medicine, 3 Heping Road, Huai'an, Jiangsu, China
| | - Fang Zhou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Hubei Province, Wuhan, China
| | - Xianlong Zhang
- Department of Anesthesiology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China.
| |
Collapse
|
10
|
Wen N, Lv Q, Du ZG. MicroRNAs involved in drug resistance of breast cancer by regulating autophagy. J Zhejiang Univ Sci B 2021; 21:690-702. [PMID: 32893526 DOI: 10.1631/jzus.b2000076] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Autophagy is a conserved catabolic process characterized by degradation and recycling of cytosolic components or organelles through a lysosome-dependent pathway. It has a complex and close relationship to drug resistance in breast cancer. MicroRNAs (miRNAs) are small noncoding molecules that can influence numerous cellular processes including autophagy, through the posttranscriptional regulation of gene expression. Autophagy is regulated by many proteins and pathways, some of which in turn have been found to be regulated by miRNAs. These miRNAs may affect the drug resistance of breast cancer. Drug resistance is the main cause of distant recurrence, metastasis and death in breast cancer patients. In this review, we summarize the causative relationship between autophagy and drug resistance of breast cancer. The roles of autophagy-related proteins and pathways and their associated miRNAs in drug resistance of breast cancer are also discussed.
Collapse
Affiliation(s)
- Nan Wen
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qing Lv
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zheng-Gui Du
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
11
|
Huang J, Tian F, Song Y, Cao M, Yan S, Lan X, Cui Y, Cui Y, Cui Y, Jia D, Cai L, Xing Y, Wang X. A feedback circuit comprising EHD1 and 14-3-3ζ sustains β-catenin/c-Myc-mediated aerobic glycolysis and proliferation in non-small cell lung cancer. Cancer Lett 2021; 520:12-25. [PMID: 34217785 DOI: 10.1016/j.canlet.2021.06.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/24/2021] [Accepted: 06/27/2021] [Indexed: 12/17/2022]
Abstract
Mammalian Eps15 homology domain 1 (EHD1) participates in the development of non-small cell lung cancer (NSCLC). However, its role in mediating aerobic glycolysis remains unclear. Herein, microarray analysis revealed that EHD1 expression was significantly correlated with the glycolysis/gluconeogenesis pathway. Clinically, EHD1 expression was positively correlated with the maximum standard uptake value (SUVmax) in 18F-FDG PET/CT scans. Additionally, EHD1 knockdown inhibited aerobic glycolysis and proliferation in vitro and in vivo. Furthermore, Wnt/β-catenin signaling was identified as a critical EHD1-regulated pathway. Co-IP, native gel electrophoresis, and immunoblotting showed that EHD1 contributed to 14-3-3 dimerization via 14-3-3ζ and subsequent activation of β-catenin/c-Myc signaling. Analysis of the EHD1 regulatory region via ENCODE revealed the potential for c-Myc recruitment, leading to transcriptional activation of EHD1 and formation of an EHD1/14-3-3ζ/β-catenin/c-Myc positive feedback circuit. Notably, blocking this circuit with a Wnt/β-catenin inhibitor dramatically inhibited tumor growth in vivo. The positive correlations among EHD1, 14-3-3ζ, c-Myc, and LDHA were further confirmed in NSCLC tissues. Collectively, our study demonstrated that EHD1 activates a 14-3-3ζ/β-catenin/c-Myc regulatory circuit that synergistically promotes aerobic glycolysis and may constitute a promising therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Jian Huang
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Fanglin Tian
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Ying Song
- Department of Dermatology, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, China
| | - Mengru Cao
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Shi Yan
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Xiuwen Lan
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Yimeng Cui
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Yaowen Cui
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Yue Cui
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Dexin Jia
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Li Cai
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China.
| | - Ying Xing
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China.
| | - Xin Wang
- PET/CT-MRI Centre, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China.
| |
Collapse
|
12
|
Rahimian N, Razavi ZS, Aslanbeigi F, Mirkhabbaz AM, Piroozmand H, Shahrzad MK, Hamblin MR, Mirzaei H. Non-coding RNAs related to angiogenesis in gynecological cancer. Gynecol Oncol 2021; 161:896-912. [PMID: 33781555 DOI: 10.1016/j.ygyno.2021.03.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 03/19/2021] [Indexed: 02/07/2023]
Abstract
Gynecological cancer affects the female reproductive system, including ovarian, uterine, endometrial, cervical, vulvar, and vaginal tumors. Non-coding RNAs (ncRNAs), and in particular microRNAs, function as regulatory molecules, which can control gene expression in a post-transcriptional manner. Normal physiological processes like cellular proliferation, differentiation, and apoptosis, and pathological processes such as oncogenesis and metastasis are regulated by microRNAs. Numerous reports have shown a direct role of microRNAs in the modulation of angiogenesis in gynecological cancer, via targeting pro-angiogenic factors and signaling pathways. Understanding the molecular mechanism involved in the regulation of angiogenesis by microRNAs may lead to new treatment options. Recently the regulatory role of some long non-coding RNAs in gynecological cancer has also been explored, but the information on this function is more limited. The aim of this article is to explore the pathways responsible for angiogenesis, and to what extent ncRNAs may be employed as biomarkers or therapeutic targets in gynecological cancer.
Collapse
Affiliation(s)
- Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | | | | | | | - Haleh Piroozmand
- Faculty of Veterinary Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Karim Shahrzad
- Department of Internal Medicine and endocrinology, Shohadae Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
13
|
Chanjiao Y, Chunyan C, Xiaoxin Q, Youjian H. MicroRNA-378a-3p contributes to ovarian cancer progression through downregulating PDIA4. Immun Inflamm Dis 2021; 9:108-119. [PMID: 33159506 PMCID: PMC7860521 DOI: 10.1002/iid3.350] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/18/2020] [Accepted: 09/02/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE MicroRNAs, as essential players in tumorigenesis, have been demonstrated to have a revolutionary effect on human cancer research. Ovarian cancer is the primary reason of death among gynecologic malignancies. In view of this, it is significant to identify prognostic and predictive markers for treatment of ovarian cancer. The aim of this study was to probe into the effects of miR-378a-3p and protein disulfide-isomerase A4 (PDIA4) on the biological functions of ovarian cancer cells. METHODS miR-378a-3p expression and PDIA4 messenger RNA expression in human ovarian cancer cells, normal human ovarian epithelial cells, and serum of both ovarian cancer patients and healthy people were detected by reverse transcription-quantitative polymerase chain reaction, and the PDIA4 protein expression was tested by Western blot analysis. Ovarian cancer OVCAR3 and SKOV3 cells were transfected or cotransfected with miR-378a-3p mimic or pcDNA3.1-PDIA4 or their negative control plasmids to explore their roles in biological functions in ovarian cancer cells. Luciferase activity and RIPA assays were implemented to validate the interaction between miR-378a-3p and PDIA4. Western blot analysis was utilized to detect phosphatidylinositol-3 kinase/serine/threonine kinase (PI3K/AKT) signaling pathway-related protein expression and their phosphate expression levels. RESULTS miR-378a-3p was elevated and PDIA4 was decreased in ovarian cancer cells and serum. In addition, miR-378a-3p mimic induced ovarian cancer cell growth, while miR-378a-3p inhibitor and pcDNA3.1-PDIA4 presented an inverse trend. pcDNA3.1-PDIA4 partially eliminated the capabilities of miR-378a-3p mimic on ovarian cancer progression. Meanwhile, miR-378a-3p was found to negatively regulate PDIA4, and miR-378a-3p mimic increased the phosphorylation levels of AKT and PI3K, while pcDNA3.1-PDIA4 exhibited an opposite tendency. Furthermore, pcDNA3.1-PDIA4 largely eliminated the functions of miR-378a-3p mimic on phosphorylation levels of AKT and PI3K. CONCLUSION This study provides evidences that miR-378a-3p activates PI3K/AKT signaling pathway by modulating PDIA4 expression, thereby playing a role in promoting the growth of ovarian cancer cells. This study provides novel directions for targeted therapy of ovarian cancer.
Collapse
Affiliation(s)
- Yao Chanjiao
- No. 3 Department of Obstetrics and GynecologyHunan Provincial People's HospitalChangshaChina
| | - Chen Chunyan
- No. 3 Department of Obstetrics and GynecologyHunan Provincial People's HospitalChangshaChina
| | - Qiu Xiaoxin
- No. 3 Department of Obstetrics and GynecologyHunan Provincial People's HospitalChangshaChina
| | - Han Youjian
- Department of cardiologyHunan Provincial People's HospitalChangshaChina
| |
Collapse
|
14
|
Wang J, Li Y, Zhou JH, Shen FR, Shi X, Chen YG. CircATRNL1 activates Smad4 signaling to inhibit angiogenesis and ovarian cancer metastasis via miR-378. Mol Oncol 2021; 15:1217-1233. [PMID: 33372356 PMCID: PMC8024733 DOI: 10.1002/1878-0261.12893] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 11/20/2020] [Accepted: 12/24/2020] [Indexed: 01/10/2023] Open
Abstract
Ovarian cancer is one of the most frequent carcinomas in females, and the occurrence rate is still rising despite many advances made. The pathogenesis of ovarian cancer remains greatly unclear. Here, we investigated the mechanisms of ovarian cancer, with the focus on circATRNL1. Human ovarian cancer tissues and cell lines were used to examine levels of circATRNL1, miR‐378, Smad4, AKT, and other proliferation‐related and migration‐related proteins. Cellular assays were used to determine cancer cell proliferation, invasion, migration, apoptosis, and angiogenesis. We validated the interactions of circATRNL1/miR‐378 and miR‐378/Smad4, and a mouse tumor xenograft model was employed to assess the effect of circATRNL1 on tumor growth and metastasis in vivo. We found that circATRNL1 was decreased while miR‐378 was increased in human ovarian cancer tissues and cells. circATRNL1 bound to miR‐378 while miR‐378 directly targeted Smad4. Overexpression of circATRNL1 or knockdown of miR‐378 suppressed angiogenesis and ovarian cancer cell proliferation, invasion, and migration via decreasing proliferation‐ and migration‐related proteins via miR‐378 or Smad4, respectively. Overexpression of circATRNL1 restrained ovarian cancer growth and abdominal metastasis in vivo. Our findings indicate that circATRNL1 acts as a miR‐378 sponge to active Smad4 signaling and suppresses angiogenesis and ovarian cancer metastasis.
Collapse
Affiliation(s)
- Juan Wang
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan Li
- Department of Gynecology and Obstetrics, the First People's Hospital of Yancheng, China
| | - Jin-Hua Zhou
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fang-Rong Shen
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiu Shi
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - You-Guo Chen
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
15
|
Annese T, Tamma R, De Giorgis M, Ribatti D. microRNAs Biogenesis, Functions and Role in Tumor Angiogenesis. Front Oncol 2020; 10:581007. [PMID: 33330058 PMCID: PMC7729128 DOI: 10.3389/fonc.2020.581007] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/27/2020] [Indexed: 12/19/2022] Open
Abstract
microRNAs (miRNAs) are small non-coding RNA molecules, evolutionary conserved. They target more than one mRNAs, thus influencing multiple molecular pathways, but also mRNAs may bind to a variety of miRNAs, either simultaneously or in a context-dependent manner. miRNAs biogenesis, including miRNA transcription, processing by Drosha and Dicer, transportation, RISC biding, and miRNA decay, are finely controlled in space and time. miRNAs are critical regulators in various biological processes, such as differentiation, proliferation, apoptosis, and development in both health and disease. Their dysregulation is involved in tumor initiation and progression. In tumors, they can act as onco-miRNAs or oncosuppressor-miRNA participating in distinct cellular pathways, and the same miRNA can perform both activities depending on the context. In tumor progression, the angiogenic switch is fundamental. miRNAs derived from tumor cells, endothelial cells, and cells of the surrounding microenvironment regulate tumor angiogenesis, acting as pro-angiomiR or anti-angiomiR. In this review, we described miRNA biogenesis and function, and we update the non-classical aspects of them. The most recent role in the nucleus, as transcriptional gene regulators and the different mechanisms by which they could be dysregulated, in tumor initiation and progression, are treated. In particular, we describe the role of miRNAs in sprouting angiogenesis, vessel co-option, and vasculogenic mimicry. The role of miRNAs in lymphoma angiogenesis is also discussed despite the scarcity of data. The information presented in this review reveals the need to do much more to discover the complete miRNA network regulating angiogenesis, not only using high-throughput computational analysis approaches but also morphological ones.
Collapse
Affiliation(s)
- Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Michelina De Giorgis
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| |
Collapse
|
16
|
Liu S, Wang C, Bai J, Li X, Yuan J, Shi Z, Mao N. Involvement of circRNA_0007059 in the regulation of postmenopausal osteoporosis by promoting the microRNA-378/BMP-2 axis. Cell Biol Int 2020; 45:447-455. [PMID: 33200464 DOI: 10.1002/cbin.11502] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/14/2020] [Accepted: 11/11/2020] [Indexed: 01/03/2023]
Abstract
Increasing evidence suggests that postmenopausal osteoporosis (PMO), a severe disturbance, imposes heavy physical, psychosocial, and financial burdens and dramatically influences the quality of life of postmenopausal women. Circular RNAs (circRNAs) and microRNAs (miRs) play important roles in the occurrence and development of PMO. However, the roles of circRNAs and miRs in osteoporosis regulation still need to be further investigated. circRNAs with different expression levels in patients with PMO were screened via RNA-seq and bioinformatics analysis. We found that circ_0007059 was upregulated in patients with PMO and during osteoclastogenesis of human bone marrow stromal cells (hBMSCs). Next, we investigated the effect of circ_0007059 overexpression during osteoclastogenesis of hBMSCs. circ_0007059 overexpression attenuated hBMSC differentiation into osteoclasts in vitro. This was demonstrated by downregulated bone morphogenetic protein 2 (BMP-2) expression, upregulated osteoclast-specific gene expression, and TRAP staining. circ_0007059 was demonstrated to directly target miR-378, which in turn targeted BMP-2 via bioinformatics analysis and the dual-luciferase reporter assay. Transfection of the miR-378 mimic reversed the effect of circ_0007059 on the osteoclastogenesis of hBMSCs. These results suggest that circ_0007059 plays an important role in osteoclastogenesis via the miR-378/BMP-2 signaling pathway. Targeting the circ_0007059/miR-378/BMP-2 axis is possibly a novel idea in osteoporosis treatment.
Collapse
Affiliation(s)
- Shu Liu
- Department of Spinal Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Chao Wang
- Department of Spinal Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jinyi Bai
- Department of Spinal Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiaoming Li
- Department of Spinal Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jiabin Yuan
- Department of Spinal Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhicai Shi
- Department of Spinal Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Ningfang Mao
- Department of Spinal Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
17
|
Barth DA, Drula R, Ott L, Fabris L, Slaby O, Calin GA, Pichler M. Circulating Non-coding RNAs in Renal Cell Carcinoma-Pathogenesis and Potential Implications as Clinical Biomarkers. Front Cell Dev Biol 2020; 8:828. [PMID: 33042985 PMCID: PMC7523432 DOI: 10.3389/fcell.2020.00828] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
Liquid biopsy-the determination of circulating cells, proteins, DNA or RNA from biofluids through a "less invasive" approach-has emerged as a novel approach in all cancer entities. Circulating non-(protein) coding RNAs including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and YRNAs can be passively released by tissue or cell damage or actively secreted as cell-free circulating RNAs, bound to lipoproteins or carried by exosomes. In renal cell carcinoma (RCC), a growing body of evidence suggests circulating non-coding RNAs (ncRNAs) such as miRNAs, lncRNAs, and YRNAs as promising and easily accessible blood-based biomarkers for the early diagnosis of RCC as well as for the prediction of prognosis and treatment response. In addition, circulating ncRNAs could also play a role in RCC pathogenesis and progression. This review gives an overview over the current study landscape of circulating ncRNAs and their involvement in RCC pathogenesis as well as their potential utility as future biomarkers in RCC diagnosis and treatment.
Collapse
Affiliation(s)
- Dominik A Barth
- Research Unit of Non-Coding RNAs and Genome Editing, Division of Clinical Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, Graz, Austria.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rares Drula
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Research Centre for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Leonie Ott
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Linda Fabris
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, Brno, Czechia.,Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Martin Pichler
- Research Unit of Non-Coding RNAs and Genome Editing, Division of Clinical Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, Graz, Austria.,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
18
|
Abstract
Glycosylation is a sophisticated informational system that controls specific biological functions at the cellular and organismal level. Dysregulation of glycosylation may underlie some of the most complex and common diseases of the modern era. In the past 5 years, microRNAs have come to the forefront as a critical regulator of the glycome. Herein, we review the current literature on miRNA regulation of glycosylation and how this work may point to a new way to identify the biological importance of glycosylation enzymes.
Collapse
Affiliation(s)
- Chu T Thu
- Biomedical Chemistry Institute, Department of Chemistry, New York University, New York, New York 10003, United States
| | - Lara K Mahal
- Biomedical Chemistry Institute, Department of Chemistry, New York University, New York, New York 10003, United States
| |
Collapse
|
19
|
Yoshida K, Yokoi A, Kato T, Ochiya T, Yamamoto Y. The clinical impact of intra- and extracellular miRNAs in ovarian cancer. Cancer Sci 2020; 111:3435-3444. [PMID: 32750177 PMCID: PMC7541008 DOI: 10.1111/cas.14599] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/17/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological cancer due to lack of early screening methods and acquired drug resistance. MicroRNAs (miRNAs) are effective post‐transcriptional regulators that are transferred by extracellular vesicles, such as exosomes. Numerous studies have revealed that miRNAs are differentially expressed in epithelial ovarian cancer and act either as oncogenes or tumor suppressor genes. Cancer cells secrete exosomes containing miRNAs, which exert various effects on the components of the tumor microenvironment, including cancer‐associated fibroblasts, macrophages, and adipocytes. Conversely, cancer cells also receive exosomes from these cells. As a result of cell‐to‐cell communication, epithelial ovarian cancer acquires a more aggressive phenotype and resistance to multiple drugs. In addition, some circulating miRNAs are protected from RNase degradation in the peripheral blood and can be potential non‐invasive biomarkers. In particular, the combination of several circulating miRNAs enhances the accuracy of cancer screening. Likewise, comprehensive analyses revealed specific miRNA signatures in non‐epithelial ovarian tumors and several miRNAs contributing to alterations of carcinogenic pathways. Overall, miRNAs play a crucial role in ovarian cancer progression. In this review, we discuss the emerging roles of intra‐ and extracellular miRNAs in ovarian cancers. In the near future, miRNAs will be practical biomarkers and computational deep learning will help in the clinical application of miRNAs. Moreover, miRNAs are potential therapeutic targets and agents, and there are ongoing clinical trials of miRNA replacement therapy. Therefore, accelerating research on miRNA might improve the prognosis of patients with ovarian cancer.
Collapse
Affiliation(s)
- Kosuke Yoshida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Akira Yokoi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoyasu Kato
- Department of Gynecology, National Cancer Center Hospital, Tokyo, Japan
| | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Yusuke Yamamoto
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
20
|
Yoshida K, Yokoi A, Kagawa T, Oda S, Hattori S, Tamauchi S, Ikeda Y, Yoshikawa N, Nishino K, Utsumi F, Niimi K, Suzuki S, Shibata K, Kajiyama H, Yokoi T, Kikkawa F. Unique miRNA profiling of squamous cell carcinoma arising from ovarian mature teratoma: comprehensive miRNA sequence analysis of its molecular background. Carcinogenesis 2020; 40:1435-1444. [PMID: 31353396 DOI: 10.1093/carcin/bgz135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/23/2019] [Accepted: 07/27/2019] [Indexed: 12/18/2022] Open
Abstract
Owing to its rarity, the carcinogenesis and molecular biological characteristics of squamous cell carcinoma arising from mature teratoma remain unclear. This study aims to elucidate the molecular background of malignant transformation from the aspects of microRNA (miRNA) profiling. We examined 7 patients with squamous cell carcinoma and 20 patients with mature teratoma and extracted their total RNA from formalin-fixed paraffin-embedded tissues. Then we prepared small RNA libraries and performed comprehensive miRNA sequencing. Heatmap and principal component analysis revealed markedly different miRNA profiling in cancer, normal ovarian and mature teratoma tissues. Then we narrowed down cancer-related miRNAs, comparing paired-cancer and normal ovaries. Comparisons of cancer and mature teratoma identified two markedly upregulated miRNAs (miR-151a-3p and miR-378a-3p) and two markedly downregulated miRNAs (miR-26a-5p and miR-99a-5p). In addition, these findings were validated in fresh cancer tissues of patient-derived xenograft (PDX) models. Moreover, several miRNAs, including miR-151a-3p and miR-378a-3p, were elevated in the murine plasma when tumor tissues were enlarged although miR-26a-5p and miR-99a-5p were not elucidated in the murine plasma. Finally, we performed target prediction and functional annotation analysis in silico and indicated that targets genes of these miRNAs markedly correlated with cancer-related pathways, including 'pathway in cancer' and 'cell cycle'. In conclusion, this is the first study on miRNA sequencing for squamous cell carcinoma arising from mature teratoma. The study identified four cancer-related miRNAs that were considered to be related to the feature of malignant transformation. Moreover, miRNAs circulating in the murine plasma of the PDX model could be novel diagnostic biomarkers.
Collapse
Affiliation(s)
- Kosuke Yoshida
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akira Yokoi
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Takumi Kagawa
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shingo Oda
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satomi Hattori
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoshi Tamauchi
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiki Ikeda
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobuhisa Yoshikawa
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kimihiro Nishino
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumi Utsumi
- Department of Obstetrics and Gynecology, Fujita Health University Bantane Hospital, Nagoya, Japan
| | - Kaoru Niimi
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shiro Suzuki
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kiyosumi Shibata
- Department of Obstetrics and Gynecology, Fujita Health University Bantane Hospital, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tsuyoshi Yokoi
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumitaka Kikkawa
- Department of Obstetrics and Gynecology, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
21
|
The role of activated leukocyte cell adhesion molecule (ALCAM) in cancer progression, invasion, metastasis and recurrence: A novel cancer stem cell marker and tumor-specific prognostic marker. Exp Mol Pathol 2020; 115:104443. [PMID: 32380056 DOI: 10.1016/j.yexmp.2020.104443] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 03/17/2020] [Accepted: 04/17/2020] [Indexed: 12/21/2022]
Abstract
Activated leukocyte cell adhesion molecule (ALCAM) or CD166 is a 100 to 105 KDa transmembrane immunoglobulin which is involved in activation of T-cells, hematopoiesis, neutrophils trans-endothelial migration, angiogenesis, inflammation and tumor propagation and invasiveness through formation of homophilic and heterophilic interactions. Recently, many studies have proposed that the expression pattern of ALCAM is highly associated with the grade, stage and invasiveness of tumors. Although ALCAM is a valuable prognostic marker in different carcinomas, similar expression patterns in different tumor types may be associated with completely different prognostic states, making it to be a tumor-type-dependent prognostic marker. In addition, ALCAM isoforms provide ways for primary detection of tumor cells with metastatic potential. More importantly, this prognostic marker has shown to be considerably dependent on the cytoplasmic and membranous expression, indirect and direct regulation of post-transcriptional molecules, pro-apoptotic proteins functionalities and several other oncogenic proteins or signalling pathways. This review mainly focuses on the pathways involved in expression of ALCAM and its prognostic value of in different types of cancers and the way in which it is regulated.
Collapse
|
22
|
Wang Y, Wang X, Jiang Y, Liu R, Cao D, Pan J, Luo Y. Identification of key miRNAs and genes for mouse retinal development using a linear model. Mol Med Rep 2020; 22:494-506. [PMID: 32319662 PMCID: PMC7248464 DOI: 10.3892/mmr.2020.11082] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 04/01/2020] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs) are upstream regulators of gene expression and are involved in several biological processes. The purpose of the present study was to obtain a detailed spatiotemporal miRNA expression profile in mouse retina, to identify one or more miRNAs that are key to mouse retinal development and to investigate the roles and mechanisms of these miRNAs. The miRNA expression pattern of the developing mouse retina was acquired from Locked Nucleic Acid microarrays. Data were processed to identify differentially expressed miRNAs (DE‑miRNAs) using the linear model in Python 3.6. Following bioinformatics analysis and reverse transcription‑quantitative polymerase chain reaction validation, 8 miRNAs (miR‑9‑5p, miR‑130a‑3p, miR‑92a‑3p, miR‑20a‑5p, miR‑93‑5p, miR‑9‑3p, miR‑709 and miR‑124) were identified as key DE‑miRNAs with low variability during mouse retinal development. Gene Ontology analysis revealed that the target genes of the DE‑miRNAs were enriched in cellular metabolic processes. Kyoto Encyclopedia of Genes and Genomes analysis demonstrated that the target genes of the DE‑miRNAs were significantly enriched in PI3K/AKT/mTOR, class O of forkhead box transcription factors, mitogen‑activated protein kinase (MAPK), neurotrophin and transforming growth factor (TGF)‑β signaling, as well as focal adhesion and the axon guidance pathway. PI3K, AKT, PTEN, MAPK1, Son of Sevenless, sphingosine‑1‑phosphate receptor 1, BCL‑2L11, TGF‑β receptor type 1/2 and integrin α (ITGA)/ITGAB, which are key components of the aforementioned pathways and were revealed to be target genes of several of the DE‑miRNAs. The present study used a linear model to identify several DE‑miRNAs, as well as their target genes and associated pathways, which may serve crucial roles in mouse retinal development. Therefore, the results obtained in the present study may provide the groundwork for further experiments.
Collapse
Affiliation(s)
- Yishen Wang
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Xiao Wang
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Yukang Jiang
- Department of Statistical Science, School of Mathematics, Southern China Research Center of Statistical Science, Sun Yat‑Sen University, Guangzhou, Guangdong 51027, P.R. China
| | - Ruyuan Liu
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Di Cao
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Jianying Pan
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Yan Luo
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
23
|
Haunschild CE, Tewari KS. Bevacizumab use in the frontline, maintenance and recurrent settings for ovarian cancer. Future Oncol 2020; 16:225-246. [PMID: 31746224 PMCID: PMC7036749 DOI: 10.2217/fon-2019-0042] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/15/2019] [Indexed: 12/16/2022] Open
Abstract
On 13 June 2018, Genentech, Inc. issued a press release announcing that the US FDA had approved the antiangiogenesis drug, bevacizumab, in combination with chemotherapy for frontline and maintenance therapy for women with newly diagnosed ovarian cancer. Regulatory approval was based on the National Cancer Institute-sponsored Gynecologic Oncology Group (GOG) protocol 0218, the Phase III, randomized, placebo-controlled, double-blind, multi-center and multi-national clinical trial that met its primary end point, progression-free survival. Bevacizumab is now approved in the frontline, platinum-sensitive recurrent and platinum-resistant recurrent settings for epithelial ovarian cancer. This review will address the broad range of clinical trials addressing the efficacy of bevacizumab use in ovarian cancer.
Collapse
Affiliation(s)
- Carolyn E Haunschild
- Clinical Instructor, Department of Obstetrics & Gynecology, Research Fellow, Division of Gynecologic Oncology, University of California, 333 City Blvd West, Suite 1400, Orange, CA 92868, USA
| | - Krishnansu S Tewari
- Professor & Division Director, Director, Division of Gynecologic Oncology, University of California, 333 City Blvd, Orange, CA 92868, USA
- Department of Obstetrics & Gynecology, University of California, The City Tower, 333 City Blvd, West – Suite 1400, Orange, CA 92868, USA
| |
Collapse
|
24
|
Leone P, Buonavoglia A, Fasano R, Solimando AG, De Re V, Cicco S, Vacca A, Racanelli V. Insights into the Regulation of Tumor Angiogenesis by Micro-RNAs. J Clin Med 2019; 8:jcm8122030. [PMID: 31757094 PMCID: PMC6947031 DOI: 10.3390/jcm8122030] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/07/2019] [Accepted: 11/14/2019] [Indexed: 12/26/2022] Open
Abstract
One of the hallmarks of cancer is angiogenesis, a series of events leading to the formation of the abnormal vascular network required for tumor growth, development, progression, and metastasis. MicroRNAs (miRNAs) are short, single-stranded, non-coding RNAs whose functions include modulation of the expression of pro- and anti-angiogenic factors and regulation of the function of vascular endothelial cells. Vascular-associated microRNAs can be either pro- or anti-angiogenic. In cancer, miRNA expression levels are deregulated and typically vary during tumor progression. Experimental data indicate that the tumor phenotype can be modified by targeting miRNA expression. Based on these observations, miRNAs may be promising targets for the development of novel anti-angiogenic therapies. This review discusses the role of various miRNAs and their targets in tumor angiogenesis, describes the strategies and challenges of miRNA-based anti-angiogenic therapies and explores the potential use of miRNAs as biomarkers for anti-angiogenic therapy response.
Collapse
Affiliation(s)
- Patrizia Leone
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.B.); (R.F.); (A.G.S.); (S.C.); (A.V.); (V.R.)
- Correspondence: ; Tel.: +39-080-5478050; Fax: +39-080-5478-045
| | - Alessio Buonavoglia
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.B.); (R.F.); (A.G.S.); (S.C.); (A.V.); (V.R.)
| | - Rossella Fasano
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.B.); (R.F.); (A.G.S.); (S.C.); (A.V.); (V.R.)
| | - Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.B.); (R.F.); (A.G.S.); (S.C.); (A.V.); (V.R.)
- Medical Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, Viale Orazio Flacco, 65, 70124 Bari, Italy
| | - Valli De Re
- Bio-Proteomics Facility, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano (PN), Italy;
| | - Sebastiano Cicco
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.B.); (R.F.); (A.G.S.); (S.C.); (A.V.); (V.R.)
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.B.); (R.F.); (A.G.S.); (S.C.); (A.V.); (V.R.)
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.B.); (R.F.); (A.G.S.); (S.C.); (A.V.); (V.R.)
| |
Collapse
|
25
|
Targeting the IL-1β/EHD1/TUBB3 axis overcomes resistance to EGFR-TKI in NSCLC. Oncogene 2019; 39:1739-1755. [DOI: 10.1038/s41388-019-1099-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 12/24/2022]
|
26
|
Lindholm EM, Ragle Aure M, Haugen MH, Kleivi Sahlberg K, Kristensen VN, Nebdal D, Børresen‐Dale A, Lingjærde OC, Engebraaten O. miRNA expression changes during the course of neoadjuvant bevacizumab and chemotherapy treatment in breast cancer. Mol Oncol 2019; 13:2278-2296. [PMID: 31402562 PMCID: PMC6763780 DOI: 10.1002/1878-0261.12561] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/26/2019] [Accepted: 08/09/2019] [Indexed: 12/24/2022] Open
Abstract
One of the hallmarks of cancer is sustained angiogenesis. Favorable results have been reported in some breast cancer (BC) patients receiving antiangiogenic therapy with bevacizumab (Bev) in combination with chemotherapy, and further knowledge on how Bev can be optimally combined with conventional treatment to increase efficacy is strongly needed. In this randomized, neoadjuvant phase II clinical trial, 132 patients with HER2-negative, nonmetastatic BC were treated with Bev in combination with sequential chemotherapy. Biopsies were sampled before treatment, after 12 weeks with anthracycline and after taxane therapy at week 25. MicroRNA (miRNA) expression profiling was performed on biopsies from each time point. Altogether, 241 biopsies were analyzed with the aim of identifying miRNA-based biomarkers of response to therapy. Results from the miRNA analyses were reported for the ER-positive cohort, which were previously demonstrated to benefit from antiangiogenic therapy in this study. For both treatment arms of this cohort, significantly different expression was observed for 217 miRNAs between objective responding and nonresponding patients before treatment initiation. These miRNAs have been linked to regulation of epithelial-mesenchymal transition, metastasis, and tumor growth, among other processes. Bev in combination with chemotherapy resulted in similar miRNA changes to chemotherapy alone. However, the deregulation of miRNA expression occurred earlier in the Bev arm. In both arms, tumor suppressor miRNAs were found upregulated after treatment, while oncogenic miRNAs were downregulated in the Bev arm. Patients responding to Bev showed a strong correlation between deregulated miRNAs and decreased proliferation score during the course of treatment, with downregulation of miR-4465 as the strongest indicator of reduced proliferation. Integrative analyses at miRNA-, gene-, and protein expression further indicated a longitudinal decrease in proliferation. Altogether, the results indicate that proliferation might represent a predictive factor for increased Bev sensitivity, which may aid in the identification of patients who could potentially benefit from Bev.
Collapse
Affiliation(s)
- Evita Maria Lindholm
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
| | - Miriam Ragle Aure
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
| | - Mads Haugland Haugen
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
- Department of Tumor biology, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
| | - Kristine Kleivi Sahlberg
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
- Department of Research and InnovationVestre Viken Hospital TrustDrammenNorway
| | - Vessela N. Kristensen
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
- Department of Clinical Molecular Biology (EpiGen), Division of MedicineAkershus University HospitalLørenskogNorway
| | - Daniel Nebdal
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
| | - Anne‐Lise Børresen‐Dale
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
- Insitute for Clinical MedicineUniversity of OsloNorway
| | - Ole Christian Lingjærde
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
- Department of Computer Science, Faculty of Mathematics and Natural SciencesUniversity of OsloNorway
| | - Olav Engebraaten
- Department of Tumor biology, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
- Insitute for Clinical MedicineUniversity of OsloNorway
- Department of OncologyOslo University HospitalNorway
| |
Collapse
|
27
|
Zhou Z, Ma J. miR-378 serves as a prognostic biomarker in cholangiocarcinoma and promotes tumor proliferation, migration, and invasion. Cancer Biomark 2019; 24:173-181. [PMID: 30594918 DOI: 10.3233/cbm-181980] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
MicroRNAs (miRNAs) have been demonstrated that play a critical role in tumorigenesis. The aim of this study is to identify the functional role of miR-378 in cholangiocarcinoma (CCA). Quantitative real-time polymerase chain reaction (qRT-PCR) was used to measure the expression levels of miR-378 in human CCA tissue samples and CCA cell lines. The receiver operating characteristic (ROC) curve was established, and the area under the ROC curve (AUC) was calculated to estimate the capacity of miR-378 in distinguishing CCA patients with different TNM stages. Kaplan-Meier survival analysis and Cox regression assay were performed to explore the prognostic value of miR-378. Cell proliferation capacity was assessed by MTT assay. Cell migration and invasion were identified by Transwell assays. miR-378 was significantly elevated in CCA tissues when compared with adjacent normal tissues, and in CCA cell lines compared to HIBEC cells. And we found that the expression of miR-378 was significantly associated with TNM stage (P= 0.030) and lymph node metastasis (P= 0.018). ROC curve analysis result showed miR-378 could distinguish CCA patients with TNM stages III and IV from those with stages I and II, with the AUC was 0.816. Patients with high expression of miR-378 had a shorter overall survival rate (Log-rank P= 0.030). The miR-378 was proven to be an independent prognostic predictor for the CCA patients (HR = 1.735, 95% CI = 1.007-2.988, P= 0.041). Downregulation of miR-378 could inhibit cell proliferation, migration, and invasion. These results indicated that miR-378 function as an oncogene and promote CCA cells proliferation, migration, and invasion. The miR-378 could be a novel prognostic marker for CCA.
Collapse
|
28
|
Wang T, Xing Y, Meng Q, Lu H, Liu W, Yan S, Song Y, Xu X, Huang J, Cui Y, Jia D, Cai L. Mammalian Eps15 homology domain 1 potentiates angiogenesis of non-small cell lung cancer by regulating β2AR signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:174. [PMID: 31023336 PMCID: PMC6482525 DOI: 10.1186/s13046-019-1162-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 04/01/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is a devastating disease with a heterogeneous prognosis, and the molecular mechanisms underlying tumor progression remain elusive. Mammalian Eps15 homology domain 1 (EHD1) plays a promotive role in tumor progression, but its role in cancer angiogenesis remains unknown. This study thus explored the role of EHD1 in angiogenesis in NSCLC. METHODS The changes in angiogenesis were evaluated through human umbilical vein endothelial cell (HUVEC) proliferation, migration and tube formation assays. The impact of EHD1 on β2-adrenoceptor (β2AR) signaling was evaluated by Western blotting, quantitative real-time polymerase chain reaction (qRT-PCR) analysis, and enzyme-linked immunosorbent assay (ELISA). The interaction between EHD1 and β2AR was confirmed by immunofluorescence (IF) and coimmunoprecipitation (Co-IP) experiments, and confocal microscopy immunofluorescence studies revealed that β2AR colocalized with the recycling endosome marker Rab11, which indicated β2AR endocytosis. Xenograft tumor models were used to investigate the role of EHD1 in NSCLC tumor growth. RESULTS The microarray analysis revealed that EHD1 was significantly correlated with tumor angiogenesis, and loss- and gain-of-function experiments demonstrated that EHD1 potentiates HUVEC proliferation, migration and tube formation. EHD1 knockdown inhibited β2AR signaling activity, and EHD1 upregulation promoted vascular endothelial growth factor A (VEGFA) and β2AR expression. Interestingly, EHD1 interacted with β2AR and played a novel and critical role in β2AR endocytic recycling to prevent receptor degradation. Aberrant VEGFA or β2AR expression significantly affected EHD1-mediated tumor angiogenesis. The proangiogenic role of EHD1 was confirmed in xenograft tumor models, and immunohistochemistry (IHC) analysis confirmed that EHD1 expression was positively correlated with VEGFA expression, microvessel density (MVD) and β2AR expression in patient specimens. CONCLUSION Collectively, the data obtained in this study suggest that EHD1 plays a critical role in NSCLC angiogenesis via β2AR signaling and highlight a potential target for antiangiogenic therapy.
Collapse
Affiliation(s)
- Ting Wang
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Ying Xing
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Qingwei Meng
- The Sixth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Hailing Lu
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Wei Liu
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Shi Yan
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Yang Song
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Harbin Medical University, Xuefu Road 246, Harbin, 150081, China
| | - Xinyuan Xu
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Jian Huang
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Yue Cui
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Dexin Jia
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Li Cai
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China.
| |
Collapse
|
29
|
Salinas-Vera YM, Marchat LA, Gallardo-Rincón D, Ruiz-García E, Astudillo-De La Vega H, Echavarría-Zepeda R, López-Camarillo C. AngiomiRs: MicroRNAs driving angiogenesis in cancer (Review). Int J Mol Med 2018; 43:657-670. [PMID: 30483765 DOI: 10.3892/ijmm.2018.4003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/22/2018] [Indexed: 01/13/2023] Open
Abstract
Angiogenesis is an important hallmark of cancer serving a key role in tumor growth and metastasis. Therefore, tumor angiogenesis has become an attractive target for development of novel drug therapies. An increased amount of anti‑angiogenic compounds is currently in preclinical and clinical development for personalized therapies. However, resistance to current angiogenesis inhibitors is emerging, indicating that there is a need to identify novel anti‑angiogenic agents. In the last decade, the field of microRNA biology has exploded revealing unsuspected functions in tumor angiogenesis. These small non‑coding RNAs, which have been dubbed as angiomiRs, may target regulatory molecules driving angiogenesis, such as cytokines, metalloproteinases and growth factors, including vascular endothelial growth factor, platelet‑derived growth factor, fibroblast growth factor, epidermal growth factor, hypoxia inducible factor‑1, as well as mitogen‑activated protein kinase, phosphoinositide 3‑kinase and transforming growth factor signaling pathways. The present review discusses the current progress towards understanding the functions of miRNAs in tumor angiogenesis regulation in diverse types of human cancer. Furthermore, the potential clinical application of angiomiRs towards anti‑angiogenic tumor therapy was explored.
Collapse
Affiliation(s)
- Yarely M Salinas-Vera
- Posgrado en Ciencias Genomicas, Universidad Autonoma de la Ciudad de Mexico, Ciudad de Mexico 03100, Mexico
| | - Laurence A Marchat
- Programa en Biomedicina Molecular y Red de Biotecnologia, Instituto Politecnico Nacional, Ciudad de Mexico 07320, Mexico
| | - Dolores Gallardo-Rincón
- Laboratorio de Medicina Translacional, Instituto Nacional de Cancerología, Ciudad de Mexico 14080, Mexico
| | - Erika Ruiz-García
- Laboratorio de Medicina Translacional, Instituto Nacional de Cancerología, Ciudad de Mexico 14080, Mexico
| | - Horacio Astudillo-De La Vega
- Laboratorio de Investigacion Translacional en Cáncer y Terapia Celular, Hospital de Oncologia, Centro Médico Nacional Siglo XXI, Ciudad de Mexico 06720, Mexico
| | | | - César López-Camarillo
- Posgrado en Ciencias Genomicas, Universidad Autonoma de la Ciudad de Mexico, Ciudad de Mexico 03100, Mexico
| |
Collapse
|
30
|
Xu ZH, Yao TZ, Liu W. miR-378a-3p sensitizes ovarian cancer cells to cisplatin through targeting MAPK1/GRB2. Biomed Pharmacother 2018; 107:1410-1417. [DOI: 10.1016/j.biopha.2018.08.132] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/16/2018] [Accepted: 08/25/2018] [Indexed: 12/24/2022] Open
|
31
|
Panoutsopoulou K, Avgeris M, Scorilas A. miRNA and long non-coding RNA: molecular function and clinical value in breast and ovarian cancers. Expert Rev Mol Diagn 2018; 18:963-979. [DOI: 10.1080/14737159.2018.1538794] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Affiliation(s)
- Konstantina Panoutsopoulou
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Margaritis Avgeris
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
32
|
Jiang Y, Sun X, Kong B, Jiang J. Antiangiogenesis therapy in ovarian cancer patients: An updated meta-analysis for 15 randomized controlled trials. Medicine (Baltimore) 2018; 97:e11920. [PMID: 30142803 PMCID: PMC6112884 DOI: 10.1097/md.0000000000011920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Antiangiogenesis therapy has been demonstrated to prolong the free survival with tolerable toxicity. However the efficacy of these drugs in overall survival (OS) remains controversial. This study was designed to assess the overall performance of antiangiogenesis therapy in improving the survival of ovary cancer (OC) patients. METHODS Electronic database of PubMed, Embase, MEDLINE, and the Cochrane Central Register of Controlled Trials were searched to identify relevant clinical randomized control trial (RCTs) assessing the therapeutic value of antiangiogenesis therapy in OC patients during 2011 to 2017. Additionally, abstracts of annual meetings were also conducted. Only English articles were considered. Progression free survival (PFS), OS, and objective response rate (ORR) were obtained from eligible RCTs. The HRs for time-to-event variables and ORs for dichotomous outcomes with their 95% CIs were used for this meta-analysis. All the statistical analyses were carried out by Stata 11.0 software using a fixed or random-models according to heterogeneity. RESULTS A total of 15 RCTs including 9359 patients were recruited into this meta-analysis. Addition of antiangiogenic agents improved PFS (HR = 0.71, 95% CI 0.62-0.81, P < .001), OS (HR = 0.92, 95% CI 0.86-0.98, P = .008) and ORR (OR = 1.74, 95% CI 1.27-2.39, P = .001) compared to placebo or chemotherapy alone in overall analysis. Antiangiogenic agents prolonged both PFS (HR = 0.58, 95% CI 0.52-0.65, P = .000) and OS (HR=0.84, 95% CI 0.76-0.92, P = .000) in recurrent settings but only PFS in primary settings (HR = 0.88, 95% CI 0.79-0.98, P = .020), longer PFS and OS in both platinum-sensitive recurrent patients (HR = 0.56, 95% CI 0.48-0.64, P = .000, PFS; HR = 0.86, 95% CI 0.76-0.98, P = .027, OS) as well as platinum-resistant recurrent cases (HR = 0.54, 95% CI 0.41-0.71, P = .000, PFS; HR = 0.84, 95% CI 0.71-0.98, P = .029, OS). Throughout therapy improved PFS (HR = 0.66, 95% CI 0.57-0.76, P < .001) and OS (HR = 0.89, 95% CI 0.83-0.96, P = .001). However the maintenance therapy of antiangiogenic agents was irrelevant to a longer PFS or OS. CONCLUSION Based on the available studies, antiangiogenic agents play an important role in the survival of OC patients. More randomized controlled trials are needed to reach more convinced conclusion.
Collapse
Affiliation(s)
- Yanyan Jiang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University
| | - Xiaomei Sun
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University
- Key Laboratory of Gynecologic Oncology of Shandong Province, PR China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University
- Key Laboratory of Gynecologic Oncology of Shandong Province, PR China
| | - Jie Jiang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University
| |
Collapse
|
33
|
hsa-miRNA-154-5p expression in plasma of endometriosis patients is a potential diagnostic marker for the disease. Reprod Biomed Online 2018; 37:449-466. [PMID: 29857988 DOI: 10.1016/j.rbmo.2018.05.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 05/02/2018] [Accepted: 05/04/2018] [Indexed: 02/06/2023]
Abstract
RESEARCH QUESTION As microRNA (miRNA) are stable in circulation, this study tested whether they could serve as putative non-invasive biomarkers for endometriosis, and their expression differences between endometriosis patients and controls. It also addressed whether the combination of differently expressed miRNA together with clinical parameters in a statistical model could distinguish between endometriosis patients and controls. DESIGN This prospective cohort study explored the possibility of using changes in extracellular miRNA spectra in plasma of 51 patients with endometriosis compared with 41 controls combined with clinical data as non-invasive biomarkers for the disease. The project was divided into three different phases for biomarker screening, discovery and validation. The differences in expression levels of plasma miRNA obtained from women with and without endometriosis were analysed with quantitative PCR-based microarrays. The diagnostic performance of the selected individual and/or combined differentially expressed miRNA candidates and clinical parameters was assessed using in silico bioinformatics modelling and receiver operating characteristic curve analysis. RESULTS Data showed that a specific plasma miRNA signature is associated with endometriosis and that hsa-miR-154-5p, which alone or in combination with hsa-miR-196b-5p, hsa-miR-378a-3p, and hsa-miR-33a-5p and the clinical parameters of body mass index and age, are potentially applicable for non-invasive diagnosis of the disease. Changes in the levels of expression of certain circulating plasma miRNA also occurred within the phases of the menstrual cycle. CONCLUSIONS miRNA seem to be promising candidates for the non-invasive diagnosis of endometriosis. Further, other clinical parameters may help in distinguishing women suffering from endometriosis from healthy individuals.
Collapse
|
34
|
Thanikachalam PV, Ramamurthy S, Wong ZW, Koo BJ, Wong JY, Abdullah MF, Chin YH, Chia CH, Tan JY, Neo WT, Tan BS, Khan WF, Kesharwani P. Current attempts to implement microRNA-based diagnostics and therapy in cardiovascular and metabolic disease: a promising future. Drug Discov Today 2018; 23:460-480. [DOI: 10.1016/j.drudis.2017.10.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 10/09/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022]
|
35
|
Peng N, Miao Z, Wang L, Liu B, Wang G, Guo X. MiR-378 promotes the cell proliferation of osteosarcoma through down-regulating the expression of Kruppel-like factor 9. Biochem Cell Biol 2018; 96:515-521. [PMID: 29490146 DOI: 10.1139/bcb-2017-0186] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that play important roles in a variety of biological processes. Dysregulation of miRNAs is tightly associated with the malignancy of cancers. Aberrant expression of miR-378 has been observed in human cancers; however, the function of miR-378 in osteosarcoma (OS) remains largely unknown. Here, we showed that miR-378 was highly expressed in human OS tissues and cell lines. Overexpression of miR-378 significantly promoted the cell proliferation of OS cells. Molecular studies identified Kruppel-like factor-9 (KLF9) as a functional downstream target of miR-378. MiR-378 directly bound to the mRNA 3'-UTR region of KLF9 and suppressed the expression of KLF9. Highly expressed KLF9 reversed the promoting effect of miR-378 on the proliferation of OS cells. The expression level of miR-378 was negatively correlated with that of KLF9 in OS tissues. Collectively, our results demonstrated the molecular interaction between miR-378 and KLF9, indicating the therapeutic potential of miR-378 for OS.
Collapse
Affiliation(s)
- Ningning Peng
- Department of Orthopedics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, 061000, Hebei, China.,Department of Orthopedics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, 061000, Hebei, China
| | - Zhigang Miao
- Department of Orthopedics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, 061000, Hebei, China.,Department of Orthopedics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, 061000, Hebei, China
| | - Liguo Wang
- Department of Orthopedics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, 061000, Hebei, China.,Department of Orthopedics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, 061000, Hebei, China
| | - Binbin Liu
- Department of Orthopedics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, 061000, Hebei, China.,Department of Orthopedics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, 061000, Hebei, China
| | - Guijiang Wang
- Department of Orthopedics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, 061000, Hebei, China.,Department of Orthopedics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, 061000, Hebei, China
| | - Xu Guo
- Department of Orthopedics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, 061000, Hebei, China.,Department of Orthopedics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, 061000, Hebei, China
| |
Collapse
|
36
|
Li J, Li S, Chen R, Lu X. Increased risk of poor survival in ovarian cancer patients with high expression of SNAI2 and lymphovascular space invasion. Oncotarget 2018; 8:9672-9685. [PMID: 28039463 PMCID: PMC5354762 DOI: 10.18632/oncotarget.14192] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 11/24/2016] [Indexed: 12/27/2022] Open
Abstract
This study is aimed to conduct a meta-analysis to evaluate the prognostic value of lymphovascular space invasion(LVSI) and to explore the potential association of SNAI1 and SNAI2 with LVSI in ovarian cancer. A systematic literature search in PubMed, ISI Web of Science, and Medline was conducted to identify relevant studies assessing the prognostic value of LVSI in ovarian cancer. The main outcomes analyzed were progression free survival/disease free survival and overall survival. TCGA database was used to explore the potential link of SNAI1 and SNAI2 with LVSI status. A total of 11 eligible studies enrolling 1817 patients were included for the meta-analysis. The overall analysis indicated that LVSI presence was associated with shorter duration of survival in ovarian cancer patients. Multivariate analysis indicated that both advanced stage and SNAI2 expression were associated with increased risk of LVSI presence. Survival analysis indicated that tumors with LVSI presence and high SNAI2 expression were significantly correlated with poorer survival when compared to tumors with both LVSI absence and low SNAI2 expression. In conclusion, LVSI presence was associated with worse clinical outcomes in ovarian cancer. Increased expression of SNAI2 and advanced stage were independent risk factors for LVSI presence. Our findings also emphasizes the potential of SNAI2 in promoting lymphovascular spread of ovarian cancer.
Collapse
Affiliation(s)
- Jun Li
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Shufen Li
- State Key Laboratory of Medical Genomics and Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ruifang Chen
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Xin Lu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| |
Collapse
|
37
|
Wang Y, Wang L, Chen C, Chu X. New insights into the regulatory role of microRNA in tumor angiogenesis and clinical implications. Mol Cancer 2018; 17:22. [PMID: 29415727 PMCID: PMC5804051 DOI: 10.1186/s12943-018-0766-4] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/12/2018] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is essential for tumor growth and metastasis. Understanding the regulation of tumor angiogenesis has become increasingly important. MicroRNAs (miRNAs) are small noncoding RNAs that function in diverse biological processes via post-transcriptional regulation. Extensive studies have revealed two important regulatory roles of miRNAs in tumor angiogenesis: miRNAs in tumor cells affect the activity of endothelial cells via non-cell-autonomous mechanisms, and miRNAs in endothelial cells regulate the cell-autonomous behavior. Recent advances have further highlighted the role of tumor-derived extracellular vesicles in the regulation of tumor angiogenesis via transferring miRNAs to endothelial cells. In this review, we summarize the regulatory role of miRNA in tumor angiogenesis, with a highlight on clinical implications of miRNAs as biomarkers for anti-angiogenic therapy response, and as therapeutic interventions against tumor angiogenesis in vivo.
Collapse
Affiliation(s)
- Ye Wang
- Department of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Liya Wang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, Jiangsu, 210002, China
| | - Cheng Chen
- Department of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China. .,Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, Jiangsu, 210002, China.
| | - Xiaoyuan Chu
- Department of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China. .,Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, Jiangsu, 210002, China.
| |
Collapse
|
38
|
Ho CS, Noor SM, Nagoor NH. MiR-378 and MiR-1827 Regulate Tumor Invasion, Migration and Angiogenesis in Human Lung Adenocarcinoma by Targeting RBX1 and CRKL, Respectively. J Cancer 2018; 9:331-345. [PMID: 29344280 PMCID: PMC5771341 DOI: 10.7150/jca.18188] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 05/03/2017] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs) have been extensively studied over the decades and have been proposed as potential molecular targets for cancer treatment. Studies have shown that miR-378 participates in numerous biological processes in various cancers; whereas miR-1827 has only been reported in pediatric glioma. The mechanism of how miRNAs modulate lung cancer metastasis remains unclear. Our previous study demonstrated that miR-378 is up-regulated while miR-1827 is down-regulated in high invasive lung cancer sub-cell lines, and their biological functions have been described. Here, we report that miR-378 and miR-1827 modulate lung cancer cell invasion and migration via epithelial-mesenchymal transition (EMT). We also demonstrated that cells treated with miR-378 inhibitors or miR-1827 mimics had reduced number of metastases and ectopic vessels in the zebrafish embryo model. We then showed that miR-378 promoted invasion and miR-1827 suppressed migration by targeting RBX1 and CRKL, respectively. Restored protein expression in miRNA-overexpressed/ miRNA-suppressed cells attenuated the inhibitory/ inducing effect of the miRNA on lung cancer cells. Collectively, our findings highlight that miR-378 and miR-1827 could serve as novel therapeutic targets in lung cancer.
Collapse
Affiliation(s)
- Chai San Ho
- Institute of Biological Sciences, Division of Genetics and Molecular Biology, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Suzita Mohd Noor
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Noor Hasima Nagoor
- Institute of Biological Sciences, Division of Genetics and Molecular Biology, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia; Centre for Research in Biotechnology for Agriculture (CEBAR), University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
39
|
Tan W, Liu B, Qu S, Liang G, Luo W, Gong C. MicroRNAs and cancer: Key paradigms in molecular therapy. Oncol Lett 2017; 15:2735-2742. [PMID: 29434998 DOI: 10.3892/ol.2017.7638] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 02/07/2017] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are a type of small non-coding RNA molecule that performs an important role in post-transcriptional gene regulation. Since miRNAs were first identified in 1993, a number of studies have demonstrated that they act as tumor suppressors or oncogenes in human cancer, including colorectal, lung, brain, breast and liver cancer, and leukemia. Large high-throughput studies have previously revealed that miRNA profiling is critical for the diagnosis and prognosis of patients with cancer, while certain miRNAs possess the potential to be used as diagnostic and prognostic biomarkers or therapeutic targets in cancer. The present study reviews the studies and examines the roles of miRNAs in cancer diagnosis, prognosis and treatment, and discusses the potential therapeutic modality of exploiting miRNAs.
Collapse
Affiliation(s)
- Weige Tan
- Breast Tumor Center and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China.,Department of Breast Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 51000, P.R. China
| | - Bodu Liu
- Breast Tumor Center and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Shaohua Qu
- Breast Tumor Center and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Gehao Liang
- Breast Tumor Center and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Wei Luo
- Breast Tumor Center and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Chang Gong
- Breast Tumor Center and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
40
|
Ofek P, Tiram G, Satchi-Fainaro R. Angiogenesis regulation by nanocarriers bearing RNA interference. Adv Drug Deliv Rev 2017; 119:3-19. [PMID: 28163106 DOI: 10.1016/j.addr.2017.01.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 01/25/2017] [Accepted: 01/31/2017] [Indexed: 12/22/2022]
Abstract
Since the approval of bevacizumab as anti-angiogenic therapy in 2004 by the FDA, an array of angiogenesis inhibitors have been developed and approved. However, results were disappointing with regard to their therapeutic efficacy. RNA interference approaches offer the possibility of rational design with high specificity, lacking in many current drug treatments for various diseases including cancer. However, in vivo delivery issues still represent a significant obstacle for widespread clinical applications. In the current review, we summarize the advances in the last decade in the field of angiogenesis-targeted RNA interference approaches, with special emphasis on oncology applications. We present pro-angiogenic and anti-angiogenic factors as potential targets, experimental evidence and clinical trials data on angiogenesis regulation by RNA interference. Consequent challenges and opportunities are discussed.
Collapse
Affiliation(s)
- Paula Ofek
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Galia Tiram
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
41
|
Li S, Yang F, Wang M, Cao W, Yang Z. miR-378 functions as an onco-miRNA by targeting the ST7L/Wnt/β-catenin pathway in cervical cancer. Int J Mol Med 2017; 40:1047-1056. [PMID: 28902356 PMCID: PMC5593456 DOI: 10.3892/ijmm.2017.3116] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/25/2017] [Indexed: 01/08/2023] Open
Abstract
Upregulation or downregulation of microRNAs (miRNAs) has been identified in human cervical cancer (CC). However, the character and function of miR-378 in CC remains unknown. In the present study, the authors demonstrated that miR-378 was upregulated in CC used the reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) assay, and promoted cell proliferation by accelerating the progress of cell cycle and repressing cell apoptosis in CC cells. The predicted target genes of miR-378 were determined by enhanced green fluorescent protein (EGFP) reporter assays, RT-qPCR assay and western blot analysis. miR-378 suppressed the expression of suppression of tumorigenicity 7-like (ST7L) by targeting the 3′-untranslated region (3′-UTR) of ST7L mRNA in HeLa and SiHa cells. ST7L was downregulated in CC using the RT-qPCR assay, and the malignant phenotype of HeLa and SiHa cells were inhibited by ST7L overexpression. In addition, miR-378 activated the Wnt/β-catenin pathway by targeting ST7L in CC cells. In short, miR-378 functions as an onco-miRNA by directly downregulating ST7L mRNA and protein level in HeLa and SiHa cells, and serves important roles in the malignancy of CC.
Collapse
Affiliation(s)
- Shengjie Li
- Department of Clinical Laboratory, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, P.R. China
| | - Fengxia Yang
- Department of Clinical Laboratory, Xintai Affiliated Hospital of Taishan Medical University, Xintai, Shandong 271200, P.R. China
| | - Meiyan Wang
- Department of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Wenjun Cao
- Department of Clinical Laboratory, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, P.R. China
| | - Zhen Yang
- Department of Pathogen Biology, Basic Medical School, Tianjin Medical University, Tianjin 300070, P.R. China
| |
Collapse
|
42
|
El Bairi K, Amrani M, Kandhro AH, Afqir S. Prediction of therapy response in ovarian cancer: Where are we now? Crit Rev Clin Lab Sci 2017; 54:233-266. [PMID: 28443762 DOI: 10.1080/10408363.2017.1313190] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Therapy resistance is a major challenge in the management of ovarian cancer (OC). Advances in detection and new technology validation have led to the emergence of biomarkers that can predict responses to available therapies. It is important to identify predictive biomarkers to select resistant and sensitive patients in order to reduce important toxicities, to reduce costs and to increase survival. The discovery of predictive and prognostic biomarkers for monitoring therapy is a developing field and provides promising perspectives in the era of personalized medicine. This review article will discuss the biology of OC with a focus on targetable pathways; current therapies; mechanisms of resistance; predictive biomarkers for chemotherapy, antiangiogenic and DNA-targeted therapies, and optimal cytoreductive surgery; and the emergence of liquid biopsy using recent studies from the Medline database and ClinicalTrials.gov.
Collapse
Affiliation(s)
- Khalid El Bairi
- a Faculty of Medicine and Pharmacy , Mohamed Ist University , Oujda , Morocco
| | - Mariam Amrani
- b Equipe de Recherche ONCOGYMA, Faculty of Medicine, Pathology Department , National Institute of Oncology, Université Mohamed V , Rabat , Morocco
| | - Abdul Hafeez Kandhro
- c Department of Biochemistry , Healthcare Molecular and Diagnostic Laboratory , Hyderabad , Pakistan
| | - Said Afqir
- d Department of Medical Oncology , Mohamed VI University Hospital , Oujda , Morocco
| |
Collapse
|
43
|
Evaluation of Prognostic and Predictive Significance of Circulating MicroRNAs in Ovarian Cancer Patients. DISEASE MARKERS 2017. [PMID: 28293063 DOI: 10.1155/2017/3098542] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Ovarian cancer patients are recognized with poor prognosis. This study aimed to identify microRNAs in plasma for predicting response to treatment and outcome. We have investigated microRNAs in plasma from ovarian cancer patients enrolled in a large multicenter study (ICON7), investigating the effect of adding bevacizumab to standard chemotherapy in patients diagnosed with epithelial ovarian cancer. Patients with different histology, grade, and FIGO stages were included (n = 207) in this study. Screening of 754 unique microRNAs was performed in the discovery phase (n = 91) using TaqMan Low Density Arrays. The results were validated using single assays and RT-qPCR. Low levels of miR-200b, miR-1274A (tRNALys5), and miR-141 were significantly associated with better survival, confirmed with log-rank test in the validation set. The level of miR-1274A (tRNALys5) correlated with outcome was especially pronounced in the high-grade serous tumors. Interestingly, low level of miR-200c was associated with 5-month prolongation of PFS when treated with bevacizumab compared to standard chemotherapy. We found prognostic significance of miR-200b, miR-141, and miR-1274A (tRNALys5) in all histological types, where miR-1274A (tRNALys5) may be a specific marker in high-grade serous tumors. The level of miR-200c may be predictive of effect of treatment with bevacizumab. However, this needs further validation.
Collapse
|
44
|
Yi S, Zeng L, Kuang Y, Cao Z, Zheng C, Zhang Y, Liao M, Yang L. Antiangiogenic drugs used with chemotherapy for patients with recurrent ovarian cancer: a meta-analysis. Onco Targets Ther 2017; 10:973-984. [PMID: 28255243 PMCID: PMC5322854 DOI: 10.2147/ott.s119879] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Objective The value of antiangiogenic inhibitors for patients with recurrent ovarian cancer has not been completely affirmed. Therefore, we aimed to assess the effectiveness and toxicities of various antiangiogenic drugs for the treatment of recurrent ovarian cancer. Methods In this meta-analysis, we searched PubMed, EMBASE, and the Cochrane Central Register of Controlled Trials databases for complete randomized controlled trials. The searches were extended to May 15, 2016. The risk of bias of the included studies was evaluated via a Cochrane systematic evaluation, and the statistical analyses were performed using RevMan 5.2 software. Results In total, we included 8 randomized controlled trials involving 3,211 patients and divided them into 3 groups, vascular endothelial growth factor receptor inhibitors (VEGFRIs), vascular endothelial growth factor (VEGF) inhibitors (bevacizumab), and angiopoietin inhibitors (trebananib). The progression-free survival improved significantly in all the groups being given antiangiogenic drugs (hazard ratio [HR]: 0.55, 95% confidence interval [CI]: 0.45–0.67, I2=0%, P<0.00001 for the VEGFRI group; HR: 0.53, 95% CI: 0.45–0.63, I2=51%, P<0.00001 for the VEGF inhibitor group; HR: 0.67, 95% CI: 0.58–0.77, I2=0%, P<0.00001 for the trebananib group). Overall survival was obviously prolonged in the VEGFRI (HR: 0.76, 95% CI: 0.59–0.97, I2=0%, P=0.03), the VEGF inhibitor (HR: 0.87, 95% CI: 0.77–0.99, I2=0%, P=0.03), and trebananib groups (HR: 0.81, 95% CI: 0.67–0.99, I2=0%, P=0.04). The incidence of grade 3/4 side effects was different among the 3 groups, for example, proteinuria, hypertension, gastrointestinal perforation, and arterial thromboembolism were presented in the VEGF inhibitor group. Increased incidences of fatigue, diarrhea, and hypertension were seen in the VEGFRI group, and the trebananib group had a higher incidence of hypokalemia. Conclusion This meta-analysis showed that antiangiogenic drugs improved the progression-free survival. The VEGFRI, bevacizumab, and trebananib groups showed increased overall survival. Adding antiangiogenic drugs to chemotherapy treatment resulted in a higher incidence of grade 3/4 side effects, but these were manageable.
Collapse
Affiliation(s)
- SuYi Yi
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - LongJia Zeng
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Yan Kuang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - ZhiJuan Cao
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - ChengJun Zheng
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Yue Zhang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Meng Liao
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Lu Yang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| |
Collapse
|
45
|
Evaluation of Prognostic and Predictive Significance of Circulating MicroRNAs in Ovarian Cancer Patients. DISEASE MARKERS 2017; 2017:3098542. [PMID: 28293063 PMCID: PMC5331307 DOI: 10.1155/2017/3098542] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 01/11/2017] [Indexed: 12/01/2022]
Abstract
Ovarian cancer patients are recognized with poor prognosis. This study aimed to identify microRNAs in plasma for predicting response to treatment and outcome. We have investigated microRNAs in plasma from ovarian cancer patients enrolled in a large multicenter study (ICON7), investigating the effect of adding bevacizumab to standard chemotherapy in patients diagnosed with epithelial ovarian cancer. Patients with different histology, grade, and FIGO stages were included (n = 207) in this study. Screening of 754 unique microRNAs was performed in the discovery phase (n = 91) using TaqMan Low Density Arrays. The results were validated using single assays and RT-qPCR. Low levels of miR-200b, miR-1274A (tRNALys5), and miR-141 were significantly associated with better survival, confirmed with log-rank test in the validation set. The level of miR-1274A (tRNALys5) correlated with outcome was especially pronounced in the high-grade serous tumors. Interestingly, low level of miR-200c was associated with 5-month prolongation of PFS when treated with bevacizumab compared to standard chemotherapy. We found prognostic significance of miR-200b, miR-141, and miR-1274A (tRNALys5) in all histological types, where miR-1274A (tRNALys5) may be a specific marker in high-grade serous tumors. The level of miR-200c may be predictive of effect of treatment with bevacizumab. However, this needs further validation.
Collapse
|
46
|
Chen MJ, Cheng YM, Chen CC, Chen YC, Shen CJ. MiR-148a and miR-152 reduce tamoxifen resistance in ER+ breast cancer via downregulating ALCAM. Biochem Biophys Res Commun 2017; 483:840-846. [PMID: 28063929 DOI: 10.1016/j.bbrc.2017.01.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 01/03/2017] [Indexed: 10/20/2022]
Abstract
Activated leukocyte cell adhesion molecule (ALCAM), also called CD166 is a 105-kDa transmembrane glycoprotein of the immunoglobin superfamily. In this study, we studied the association between ALCAM expression and tamoxifen resistance in ER + breast cancer and further investigated how ALCAM is regulated in the cancer cells. IHC staining data showed that the tumor tissues from non-responders (N = 20) generally had significantly stronger ALCAM staining than that from tamoxifen responders (N = 16). In vitro cell assay also confirmed ALCAM upregulation in tamoxifen resistant (TamR) MCF-7 cells than in tamoxifen sensitive (TamS) MCF-7 cells. ALCAM overexpression significantly alleviated 4-Hydroxytestosterone (4-OHT) induced cell viability inhibition and cell apoptosis in TamS MCF-7 cells, while ALCAM knockdown remarkably enhanced 4-OHT induced cell viability inhibition and cell apoptosis in TamR MCF-7 cells. Demethylation reagent treatment significantly restored miR-148a and miR-152 expression in TamR MCF-7 cells. MiR-148a and miR-152 can directly target ALCAM 3'UTR and decrease ALCAM expression. MiR-148a overexpression had similar effect as ALCAM siRNA on enhancing 4-OHT induced cell viability inhibition and cell apoptosis in TamR MCF-7 cells. MiR-152 overexpression alone caused growth inhibition and increased cell apoptosis in TamR MCF-7 cells. It also enhanced the effect of 4-OHT. Simultaneous inhibition of miR-148a and miR-152 significantly protected TamS MCF-7 cells from 4-OHT induced cell viability inhibition and cell apoptosis. Based on these findings, we infer that MiR-148a and miR-152 can sensitize TamR MCF-7 cells to tamoxifen at least via downregulating ALCAM.
Collapse
Affiliation(s)
- Ming-Jenn Chen
- Department of Surgery, Chi-Mei Medical Center, Tainan, Taiwan; Department of Sports Management, College of Leisure and Recreation Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Ya-Min Cheng
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Chung Chen
- Department of Plastic and Reconstruction Surgery, E-Da Hospital, Kaohsiung, Taiwan
| | - Yu-Chieh Chen
- Department of Gynecology and Obstetrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Ju Shen
- Department of Gynecology and Obstetrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
47
|
Kim R, Park SI, Lee CY, Lee J, Kim P, Oh S, Lee H, Lee MY, Kim J, Chung YA, Hwang KC, Maeng LS, Chang W. Alternative new mesenchymal stem cell source exerts tumor tropism through ALCAM and N-cadherin via regulation of microRNA-192 and -218. Mol Cell Biochem 2016; 427:177-185. [PMID: 28039611 PMCID: PMC5306073 DOI: 10.1007/s11010-016-2909-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 12/03/2016] [Indexed: 12/26/2022]
Abstract
Gliomas are the most common type of malignant primary brain tumors. Some treatments of gliomas exist, but they are rarely curative. Mesenchymal stem cells (MSCs) are emerging as potential modes of targeted cancer therapy owing to their capacity for homing toward tumor sites. It has been proposed that MSCs derived from various sources, such as bone marrow, adipose tissue and umbilical cord blood, can be used as cell-based therapy for brain tumors. Here, MSCs obtained from the synovial fluid of osteoarthritis or rheumatoid arthritis patients were investigated as therapeutic candidates. Specifically, we compared migratory and adhesive abilities, as well as expression levels of related genes and microRNA in bone marrow derived-MSCs (BMMSCs), adipose derived-MSCs (ADMSCs), and synovial fluid derived-MSCs (SFMSCs) after treatment with conditioned medium from gliomas. Migration and adhesion of SFMSCs increased through upregulation of the activated lymphocyte cell adhesion molecule (ALCAM) and N-cadherin by microRNA-192 and -218 downregulation, similar to BMMSCs and ADMSCs. Migratory capacities of all types of MSCs were evaluated in vivo, and SFMSCs migrated intensively toward gliomas. These results suggest that SFMSCs have potential for use in cell-based antitumor therapies.
Collapse
Affiliation(s)
- Ran Kim
- Department of Biology Education, College of Education, Pusan National University, Pusan, 609-735, South Korea
| | - Sang In Park
- Institute of Catholic Integrative Medicine, Incheon St. Mary's Hospital, The Catholic University of Korea, College of Medicine, Incheon, 403-720, South Korea
| | - Chang Youn Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 120-752, South Korea
| | - Jihyun Lee
- Department of Biology Education, College of Education, Pusan National University, Pusan, 609-735, South Korea
| | - Pilseog Kim
- Department of Biology Education, College of Education, Pusan National University, Pusan, 609-735, South Korea
| | - Sekyung Oh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Hojin Lee
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Min Young Lee
- Department of Molecular Physiology, College of Pharmacy, Kyungpook National University, Daegu, 702-701, South Korea
| | - Jongmin Kim
- Department of Life Systems, Sookmyung Women's University, 52 Hyochangwon-gil, Seoul, 140-742, South Korea
| | - Yong-An Chung
- Institute of Catholic Integrative Medicine, Incheon St. Mary's Hospital, The Catholic University of Korea, College of Medicine, Incheon, 403-720, South Korea
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangwon-do, 210-701, South Korea.,Catholic Kwandong University International, St. Mary's Hospital, Incheon, 404-834, South Korea
| | - Lee-So Maeng
- Institute of Catholic Integrative Medicine, Incheon St. Mary's Hospital, The Catholic University of Korea, College of Medicine, Incheon, 403-720, South Korea.
| | - Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, Pusan, 609-735, South Korea.
| |
Collapse
|
48
|
Kuang X, Wei C, Zhang T, Yang Z, Chi J, Wang L. miR-378 inhibits cell growth and enhances apoptosis in human myelodysplastic syndromes. Int J Oncol 2016; 49:1921-1930. [DOI: 10.3892/ijo.2016.3689] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 07/13/2016] [Indexed: 11/06/2022] Open
|
49
|
Xiao M, Wang X, Yan M, Chen W. A systematic evaluation for the potential translation of CD166-related expression as a cancer biomarker. Expert Rev Mol Diagn 2016; 16:925-32. [PMID: 27398729 DOI: 10.1080/14737159.2016.1211932] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Meng Xiao
- Department of Oral and Maxillofacial-Head & Neck Oncology and Faculty of Oral and Maxillofacial Surgery, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Xu Wang
- Department of Oral and Maxillofacial-Head & Neck Oncology and Faculty of Oral and Maxillofacial Surgery, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Ming Yan
- Department of Oral and Maxillofacial-Head & Neck Oncology and Faculty of Oral and Maxillofacial Surgery, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Wantao Chen
- Department of Oral and Maxillofacial-Head & Neck Oncology and Faculty of Oral and Maxillofacial Surgery, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Shanghai, China
| |
Collapse
|
50
|
Li X, Zhu S, Hong C, Cai H. Angiogenesis inhibitors for patients with ovarian cancer: a meta-analysis of 12 randomized controlled trials. Curr Med Res Opin 2016; 32:555-62. [PMID: 26652645 DOI: 10.1185/03007995.2015.1131152] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/09/2015] [Indexed: 02/05/2023]
Abstract
OBJECTIVES To investigate the effects of angiogenesis inhibitors in the treatment of patients with advanced or recurrent ovarian cancer, a meta-analysis was performed and overall survival (OS), progression-free survival (PFS), and toxicity were assessed. PATIENTS AND METHODS The PubMed and Embase databases, and the Cochrane Central Register of Controlled Trials were searched for publications between January 2000 and June 2015. Hazard ratios (HRs) with their 95% confidence intervals (CIs), or data for calculating HRs with 95% CIs were derived. RESULTS The 12 trials in this meta-analysis were divided into three groups: four trials with a VEGF inhibitor (the bevacizumab group), six trials with VEGFR inhibitors (the VEGFRIs group), and two trials with an angiopoietin inhibitor (the trebananib group). PFS improvement was seen in all groups (HR = 0.61, 95% CI 0.48 to 0.79, P < 0.001 for bevacizumab; HR = 0.71, 95% CI 0.59 to 0.87, P = 0.001 for VEGFRIs; and HR = 0.67, 95% CI 0.62 to 0.72, P < 0.001 for trebananib). Regarding OS, bevacizumab showed a trend of improvement (HR = 0.90, 95% CI 0.80 to 1.01, P = 0.079), VEGFRIs showed no improvement (HR = 0.92, 95% CI 0.75 to 1.11, P = 0.368), and trebananib demonstrated a significant prolongation (HR = 0.81, 95% CI 0.67 to 0.99, P = 0.036). Bevacizumab was associated with more class-specific adverse events (RR = 4.05, 95% CI 1.99 to 8.27, P < 0.001). Although the toxicity profiles differed, VEGFRIs developed common higher incidences of hypertension, diarrhea, and fatigue. A higher incidence of edema was reported in the trebananib group (RR = 2.60, 95% CI 0.84 to 8.00, P = 0.097). CONCLUSIONS Anti-angiogenic therapy showed clear PFS benefit with increased toxicity, but its role in OS was undefined for ovarian cancer which emphasized the need for patient selection.
Collapse
Affiliation(s)
- Xuyuan Li
- a a Department of Medical Oncology , Affiliated Shantou Hospital of Sun Yat-sen University , Shantou , Guangdong , China
| | - Sujuan Zhu
- b b Department of Good Clinical Practice , Cancer Hospital of Shantou University Medical College , Shantou , Guangdong , China
| | - Chaoqun Hong
- c c Cancer Research Lab, Cancer Hospital of Shantou University Medical College , Shantou , Guangdong , China
| | - Haoquan Cai
- d d Department of Medical Oncology , First affiliated Hospital of Shantou University Medical College , Shantou , Guangdong , China
| |
Collapse
|