1
|
Wang S, Ma C, Ren Z, Zhang Y, Hao K, Liu C, Xu L, He S, Zhang J. Diagnostic Value of Glycosylated Extracellular Vesicle microRNAs in Gastric Cancer. Cancer Manag Res 2025; 17:145-160. [PMID: 39881946 PMCID: PMC11776422 DOI: 10.2147/cmar.s494747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/03/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction Early diagnosis is crucial for improving the prognosis of patients with gastric cancer (GC). However, the currently used biomarkers for diagnosing GC have limited sensitivity and specificity. This study aimed to develop a novel diagnostic model based on miRNAs from glycosylated extracellular vesicles and evaluate its effectiveness in diagnosing gastric cancer. Methods GlyExo-capture technology was used to isolate glycosylated extracellular vesicles from serum samples. The signatures were screened in a discovery cohort of GC patients (n=55) and non-disease controls (n=46) using an integrated process, including high-throughput sequencing technology, screening using a complete bioinformatics algorithm, validation using RT-qPCR, and evaluation by constructing a diagnostic model. The diagnostic model was evaluated in an independent validation cohort (n=139). Results We developed a diagnostic model for GC based on five miRNA pairs. This diagnostic model demonstrated high sensitivity, specificity, and stable performance in distinguishing GC patients from non-cancer controls with AUC of 0.930 in the independent validation cohort, particularly in differentiating early-stage GC from benign patients. The markers also showed excellent performance in indicating perineural invasion status and lymph node metastasis in the testing cohort. Discussion The model demonstrated high sensitivity and specificity in diagnosing patients with GC, especially in differentiating early-stage GC from benign patients. The five miRNA pairs could also aid in making treatment decisions. Thus, miRNAs derived from glycosylated exosomes are promising biomarkers for cancer diagnosis.
Collapse
Affiliation(s)
- Shunda Wang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Cuidie Ma
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People’s Republic of China
| | - Zhihua Ren
- Department of General Surgery, Qilu Hospital Fo Shandong University, Jinan, 250012, People’s Republic of China
| | - Yufei Zhang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People’s Republic of China
| | - Kun Hao
- Beijing Hotgen Biotech Co., Ltd, Beijing, 102600, People’s Republic of China
| | - Chengxiu Liu
- Beijing Hotgen Biotech Co., Ltd, Beijing, 102600, People’s Republic of China
| | - Lida Xu
- Beijing Hotgen Biotech Co., Ltd, Beijing, 102600, People’s Republic of China
| | - Shun He
- Department of Endoscopy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Jianwei Zhang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| |
Collapse
|
2
|
Lucarini V, Nardozi D, Angiolini V, Benvenuto M, Focaccetti C, Carrano R, Besharat ZM, Bei R, Masuelli L. Tumor Microenvironment Remodeling in Gastrointestinal Cancer: Role of miRNAs as Biomarkers of Tumor Invasion. Biomedicines 2023; 11:1761. [PMID: 37371856 DOI: 10.3390/biomedicines11061761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Gastrointestinal (GI) cancers are the most frequent neoplasm, responsible for half of all cancer-related deaths. Metastasis is the leading cause of death from GI cancer; thus, studying the processes that regulate cancer cell migration is of paramount importance for the development of new therapeutic strategies. In this review, we summarize the mechanisms adopted by cancer cells to promote cell migration and the subsequent metastasis formation by highlighting the key role that tumor microenvironment components play in deregulating cellular pathways involved in these processes. We, therefore, provide an overview of the role of different microRNAs in promoting tumor metastasis and their role as potential biomarkers for the prognosis, monitoring, and diagnosis of GI cancer patients. Finally, we relate the possible use of nutraceuticals as a new strategy for targeting numerous microRNAs and different pathways involved in GI tumor invasiveness.
Collapse
Affiliation(s)
- Valeria Lucarini
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy
| | - Daniela Nardozi
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Valentina Angiolini
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy
| | - Monica Benvenuto
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
- Departmental Faculty of Medicine and Surgery, Saint Camillus International University of Health and Medical Sciences, via di Sant'Alessandro 8, 00131 Rome, Italy
| | - Chiara Focaccetti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Raffaele Carrano
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Zein Mersini Besharat
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Laura Masuelli
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy
| |
Collapse
|
3
|
Ferragut Cardoso AP, Nail AN, Banerjee M, Wise SS, States JC. miR-186 induces tetraploidy in arsenic exposed human keratinocytes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 256:114823. [PMID: 36989553 DOI: 10.1016/j.ecoenv.2023.114823] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 02/21/2023] [Accepted: 03/21/2023] [Indexed: 05/25/2023]
Abstract
Chronic inorganic arsenic (iAs) exposure in drinking water is a global issue affecting >225 million people. Skin is a major target organ for iAs. miRNA dysregulation and chromosomal instability (CIN) are proposed mechanisms of iAs-induced carcinogenesis. CIN is a cancer hallmark and tetraploid cells can better tolerate increase in chromosome number and aberration, contributing to the evolution of CIN. miR-186 is overexpressed in iAs-induced squamous cell carcinoma relative to iAs-induced hyperkeratosis. Bioinformatic analysis indicated that miR-186 targets mRNAs of important cell cycle regulators including mitotic checkpoint serine/threonine kinase B (BUB1) and cell division cycle 27 (CDC27). We hypothesized that miR-186 overexpression contributes to iAs-induced transformation of keratinocytes by targeting mitotic regulators leading to induction of CIN. Ker-CT cells, a near diploid human keratinocyte cell line, were transduced with miR-186 overexpressing or scrambled control lentivirus. Stable clones were isolated after puromycin selection. Clones transduced with lentivirus expressing either a scrambled control miRNA or miR-186 were maintained with 0 or 100 nM iAs for 4 weeks. Unexposed scrambled control clones were considered as passage matched controls. Chronic iAs exposure increased miR-186 expression in miR-186 clones. miR-186 overexpression significantly reduced CDC27 levels irrespective of iAs exposure. The percentage of tetraploid or aneuploid cells was increased in iAs exposed miR-186 clones. Aneuploidy can arise from a tetraploid intermediate. Suppression of CDC27 by miR-186 may lead to impairment of mitotic checkpoint complex formation and its ability to maintain cell cycle arrest leading to chromosome misalignment. As a result, cells overexpressing miR-186 and chronically exposed to iAs may have incorrect chromosome segregation and CIN. These data suggest that dysregulation of miRNA by iAs mediates tetraploidy, aneuploidy and chromosomal instability contributing to iAs-induced carcinogenesis.
Collapse
Affiliation(s)
- Ana P Ferragut Cardoso
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
| | - Alexandra N Nail
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
| | - Mayukh Banerjee
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
| | - Sandra S Wise
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
| | - J Christopher States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
4
|
Zhang F, Wang G, Yan W, Jiang H. MiR-4268 suppresses gastric cancer genesis through inhibiting keratin 80. Cell Cycle 2022; 21:2051-2064. [PMID: 35748914 DOI: 10.1080/15384101.2022.2085351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Gastric cancer (GC) affects a large proportion of cancer patients worldwide, and the prediction of potential biomarkers can greatly improve its diagnosis and treatment. Here, miR-4268 and keratin 80 (KRT80) expression in GC tissues and cell lines was determined. The effect of downregulating miR-4268 and interfering with KRT80 expression on the viability, proliferation, apoptosis, and migration of GC cells were evaluated. The interaction between miR-4268 and KRT80 was studied using luciferase reporter and RNA pull-down assays. The western blot, CCK-8, BrdU, caspase-3 activity, Transwell assays were performed for the functional characterization. In GC tissues and cells, KRT80 expression was found to be significantly higher, while that of miR-4268 was significantly lower than the respective expressions in normal tissues and cells. Interference with KRT80 expression inhibited the viability, proliferation, and migration of GC cells and facilitated cell apoptosis in vitro. We further demonstrated that miR-4268 targeted KRT80 and negatively regulated its expression, and miR-4268 inhibitor alleviated the inhibitory effects of KRT80 downregulation on GC cell growth. Finally, miR-4268 may function as tumor suppressor through inhibiting PI3K/AKT/JNK pathways by targeting KRT80 in GC. Collectively, our present results indicate that the miR-4268/KRT80 axis acts as a potential therapeutic target for patients with GC.AbbreviationsGastric cancer (GC); MicroRNAs (miRNAs); Keratin 80 (KRT80); differentially expressed genes (DEGs); chemoradiotherapy (CRT); negative nonsense sequence (NC); radioimmunoprecipitation assay (RIPA); polyvinylidene fluoride (PVDF).
Collapse
Affiliation(s)
- Fan Zhang
- Department of Gastroenterology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Guoxian Wang
- Department of Radiology, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wenjuan Yan
- Department of Gastroenterology, The Third People's Hospital of Hubei Province, Wuhan, Hubei, China
| | - Hongmei Jiang
- Department of Gastroenterology, Wuhan Third Hospital, Tongren Hospital of Wuhan University (Optics Valley Area), Wuhan, Hubei, China
| |
Collapse
|
5
|
Lee HW, Jose CC, Cuddapah S. Epithelial-mesenchymal transition: Insights into nickel-induced lung diseases. Semin Cancer Biol 2021; 76:99-109. [PMID: 34058338 PMCID: PMC8627926 DOI: 10.1016/j.semcancer.2021.05.020] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023]
Abstract
Nickel compounds are environmental toxicants, prevalent in the atmosphere due to their widespread use in several industrial processes, extensive consumption of nickel containing products, as well as burning of fossil fuels. Exposure to nickel is associated with a multitude of chronic inflammatory lung diseases including asthma, chronic obstructive pulmonary disease (COPD) and pulmonary fibrosis. In addition, nickel exposure is implicated in the development of nasal and lung cancers. Interestingly, a common pathogenic mechanism underlying the development of diseases associated with nickel exposure is epithelial-mesenchymal transition (EMT). EMT is a process by which the epithelial cells lose their junctions and polarity and acquire mesenchymal traits, including increased ability to migrate and invade. EMT is a normal and essential physiological process involved in differentiation, development and wound healing. However, EMT also contributes to a number of pathological conditions, including fibrosis, cancer and metastasis. Growing evidence suggest that EMT induction could be an important outcome of nickel exposure. In this review, we discuss the role of EMT in nickel-induced lung diseases and the mechanisms associated with EMT induction by nickel exposure.
Collapse
Affiliation(s)
- Hyun-Wook Lee
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, 10010, USA
| | - Cynthia C Jose
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, 10010, USA
| | - Suresh Cuddapah
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, 10010, USA.
| |
Collapse
|
6
|
Wang J, Wang T, Zhang Y, Liu J, Song J, Han Y, Wang L, Yang S, Zhu L, Geng R, Li W, Yu X. CPEB1 enhances erastin-induced ferroptosis in gastric cancer cells by suppressing twist1 expression. IUBMB Life 2021; 73:1180-1190. [PMID: 34184391 DOI: 10.1002/iub.2525] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 12/18/2022]
Abstract
The induction of ferroptosis is considered a new strategy for cancer treatment. Cytoplasmic polyadenylation element binding protein 1 (CPEB1) is a post-transcriptional regulatory factor, whose low expression has been reported to link to the enhanced metastasis and angiogenesis of gastric cancer (GC). In this study, to explore the role of CPEB1 in ferroptosis, GC cells with overexpressed or silenced CPEB1 expression were treated with erastin, a classic ferroptosis inducer. The results showed that erastin dose-dependently decreased the viability of four GC cell lines (AGS, SNU-1, Hs-746 T, and HGC-27), suggesting that ferroptosis could be triggered in these GC cells. Interestingly, HGC-27 cells overexpressing CPEB1 were more sensitive to erastin, generated more lipid reactive oxygen species (ROS) and malondialdehyde (MDA), and their glutathione peroxidase 4 (Gpx4) expression and GSH content were reduced. Contrarily, CPEB1-silenced AGS cells were more resistant to erastin. Mechanically, we demonstrated that CPEB1 overexpression reduced the expression of twist1, an inhibitor of activating transcription factor 4 (ATF4), thereby activating the ATF4/ChaC Glutathione Specific Gamma-Glutamylcyclotransferase 1 (CHAC1) pathway (CHAC1, a molecule known to induce GSH degradation). Furthermore, re-expression of twist1 in GC cells impaired the effects of CPEB1 overexpression in presence of erastin. Additionally, similar to the in vitro results, the growth-inhibiting effects of erastin on GC xenografted tumors were also augmented by CPEB1 overexpression in vivo. Collectively, we demonstrate that CPEB1 facilitates erastin-induced ferroptosis by inhibiting twist1.
Collapse
Affiliation(s)
- Jing Wang
- Department of Biology, School of Basic Medical Sciences, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Tao Wang
- Department of Ultrasound, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Yang Zhang
- Department of Laboratory Medicine, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Jiaqi Liu
- Laboratory of Morphology, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Jie Song
- Department of Biology, School of Basic Medical Sciences, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Yanlong Han
- Department of Biology, School of Basic Medical Sciences, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Lihong Wang
- Department of Electrical Diagnosis, Jilin Neuropsychiatric Hospital, Siping, Jilin, China
| | - Shuang Yang
- Office of Academic Affairs, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Lili Zhu
- Department of Foreign Language, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Rui Geng
- Office of Student Affairs, School of Basic Medical Sciences, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Weimin Li
- Department of Imaging, The Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Xiaoguang Yu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
7
|
MiR-142 suppresses progression of gastric carcinoma via directly targeting LRP8. Clin Res Hepatol Gastroenterol 2021; 45:101520. [PMID: 33268037 DOI: 10.1016/j.clinre.2020.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/26/2020] [Accepted: 08/04/2020] [Indexed: 02/04/2023]
Abstract
Recently, the role of miRNA-142 (miR-142) in tumor development has attracted extensive attention. The aim of this study was to investigate the impact of miR-142 and its potential target low-density lipoprotein receptor (LDLR)-related protein 8 (LRP8) on the proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) of gastric carcinoma (GC). Gene and protein expressions were detected using RT-qPCR and Western blotting, respectively. The biological behaviors of GC cell lines were determined by CCK-8, flow cytometry and Transwell assays, respectively. The interaction between miR-142 and LRP8 was confirmed with dual luciferase reporter assay. Xenograft nude mouse model was used to observe tumor growth. Here, miR-142 expression was markedly reduced in GC tissues and cells, and was negatively correlated with lymph node metastasis and poor prognosis in patients with GC. Stable miR-142 overexpression was sufficient to inhibit cell proliferation, migration and invasion in vitro and reduce tumor growth in vivo, accompanied by increased expression of the epithelial marker and reduced levels of mesenchymal markers. Mechanistically, the 3'-untranslated regions (3'-UTR) of LRP8 was a direct target of miR-142. Restoration of LRP8 attenuated the inhibitory effect of miR-142 on GC cells, whereas inhibition of LRP8 caused the opposite outcomes. In conclusion, our findings suggest that miR-142 plays a significant role in suppressing progression of GC by targeting LRP8, and miR-142 may be useful for the development of novel targeted therapies.
Collapse
|
8
|
Lu X, Song X, Hao X, Liu X, Zhang X, Yuan N, Ma H, Zhang Z. MicroRNA-186-3p attenuates tumorigenesis of cervical cancer by targeting MCM2. Oncol Lett 2021; 22:539. [PMID: 34084218 PMCID: PMC8161468 DOI: 10.3892/ol.2021.12800] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/28/2021] [Indexed: 12/22/2022] Open
Abstract
The present study examined the effect of microRNA (miRNA/miR)-186-3p and its target gene, minichromosome maintenance complex component 2 (MCM2), on cervical cancer. Cervical cancer tissues and corresponding normal tissues were collected from 48 patients and bioinformatics analysis was performed to identify the differentially expressed genes in cervical cancer. TargetScan and TarBase were used to identify miRNAs, and reverse transcription-quantitative PCR was conducted to detect and evaluate mRNA expression levels. Additionally, MTT and 5-bromo-2-deoxyuridine assays were performed to examine cell proliferation. Cell adhesion, cell cycle distribution and apoptosis were assessed using cell adhesion, flow cytometry and caspase-3/7 activity assays, respectively. The results revealed that miR-186-3p expression was downregulated in cervical cancer tissues and cells, and it negatively regulated MCM2 expression by directly targeting its 3' untranslated region in cervical cancer. Furthermore, MCM2 facilitated cell proliferation and inhibited cell apoptosis, which were reversed by upregulation of miR-186-3p expression. Collectively, the present study suggested that MCM2 and its negative regulator, miR-186-3p, regulate cervical cancer progression.
Collapse
Affiliation(s)
- Xiurong Lu
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Xiao Song
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Xiaohui Hao
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Xiaoyu Liu
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Xianyu Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Na Yuan
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Huan Ma
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Zhilin Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| |
Collapse
|
9
|
Radiation Can Regulate the Expression of miRNAs Associated with Osteogenesis and Oxidation in Exosomes from Peripheral Blood Plasma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6646323. [PMID: 33628370 PMCID: PMC7899774 DOI: 10.1155/2021/6646323] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/19/2021] [Accepted: 01/28/2021] [Indexed: 02/05/2023]
Abstract
Objectives Radiotherapy is a common therapy in head and neck tumors, which may cause a side effect radiation bone injury (RBI). Furthermore, it has been investigated that microRNA (miRNA) expression levels were altered after radiotherapy. Exosomes play a role in bone formation as miRNA containers, while radiation affects exosomes composition, secretion, and function. So, our objective is to explore changes in miRNA levels during bone formation after radiotherapy and identify the differentially expressed miRNAs (DE-miRs) in plasma exosomes during the process of osteogenesis related to irradiation. Materials and Methods In this study, we analyzed nine samples from three rabbits exposed twice to radiation (15 Gy each) and detected DE-miRs from irradiated plasma exosomes during the process of osteogenesis by RNA sequencing. Further, we identified DE-miRs with significant differences and predicted their target genes via the bioinformatics analysis tools Targetscan v7.2 and miRPathDB v2.0. Finally, we identified radiation-responsive miRNAs and predicted their target genes during osteogenesis. Results Taken together, we have identified some DE-miRs in irradiated plasma exosomes, which were involved in several vital signaling pathways related to bone physiology, such as the Wnt pathway, MAPK cascade, and calcium modulating pathway. Conclusions We have found that plasma exosomes are one of the ways by which radiation can affect bone metabolism and regeneration. However, the specific mechanisms of how these plasma exosomal miRNAs mediate the osteogenesis pathways must be further investigated. Clinical Relevance. Radiotherapy may cause radiation bone injury, and miRNA expression levels in rabbit plasma exosomes are altered after radiotherapy. High-throughput RNA sequencing can identify the differentially expressed miRNAs in irradiated plasma exosomes during the process of osteogenesis. These findings make sense to develop novel therapeutic strategies for treating radiation-induced bone injury disorders.
Collapse
|
10
|
Ghafouri-Fard S, Abak A, Bahroudi Z, Shoorei H, Abbas Raza SH, Taheri M. The interplay between non-coding RNAs and Twist1 signaling contribute to human disorders. Biomed Pharmacother 2021; 135:111220. [PMID: 33433357 DOI: 10.1016/j.biopha.2021.111220] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/23/2020] [Accepted: 12/31/2020] [Indexed: 12/14/2022] Open
Abstract
Twist-related protein 1 (Twist1) is a basic helix-loop-helix (bHLH) transcription factor (TF) being coded by the TWIST1 gene. This TF has a fundamental effect on the normal development and in the pathogenesis of various diseases especially cancer. Twist1 has interactions with some long non-coding RNAs and miRNAs. The interactions between this TF and various miRNAs such as miR-16, miR-26b-5p, miR-1271, miR-539, miR-214, miR-200b/c, miR-335, miR-10b, and miR-381 are implicated in the carcinogenic processes. TP73-AS1, LINC01638, ATB, NONHSAT101069, CASC15, H19, PVT1, LINC00339, LINC01385, TANAR, SNHG5, DANCR, CHRF, and TUG1 are among long non-coding RNAs which interact with Twist1 and participate in the carcinogenesis. This review aims at depicting the interaction between these non-coding transcripts and Twist1 and the consequence of these interactions in human neoplasms.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefe Abak
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Bahroudi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Biranjd University of Medical Sciences, Birjand, Iran
| | - Sayed Haidar Abbas Raza
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, China
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Huang J, Hou S, Xu J, Wu J, Yin J. Long non-coding RNA OIP5-AS1 promotes cell proliferation and aerobic glycolysis in gastric cancer through sponging miR-186. Arch Med Sci 2021; 17:1742-1751. [PMID: 34900056 PMCID: PMC8641525 DOI: 10.5114/aoms.2019.87213] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 11/09/2018] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Long non-coding RNAs (lncRNAs) play vital roles in tumour initiation and progression. LncRNA OIP5-AS1 is a potential oncogene in many types of human malignancies, but its biological functions in gastric cancer (GC) remain to be further elucidated. MATERIAL AND METHODS The expression levels of OIP5-AS1 and miR-186 in GC tissues and cell lines were detected by RT-qPCR analysis. CCK-8 assay and colony formation assay were performed to investigate the proliferation of GC cells in vitro, and a nude mouse tumour model was established to validate the role of OIP5-AS1 in GC tumorigenesis in vivo. The glucose consumption and lactate production of GC cells were detected by ELISA assay. Interaction between OIP5-AS1 and miR-186 was determined using dual luciferase reporter assay. RESULTS The results demonstrated that OIP5-AS1 was upregulated in GC tissues and cell lines and that its high expression was notably correlated with aggressive clinicopathological features of GC patients. Functionally, knockdown of OIP5-AS1 inhibited GC cell proliferation and enhanced cell apoptosis in vitro, and inhibited GC xenograft growth in vivo. In addition, knockdown of OIP5-AS1 reduced the glucose consumption and lactate production in GC cells. In particular, OIP5-AS1 may function as a ceRNA for miR-186, and inhibition of miR-186 blocks the effects of OIP5-AS1 knockdown on aerobic glycolysis in GC cells. CONCLUSIONS Accordingly, our findings suggested that the OIP5-AS1/miR-186 axis might be considered as a potential therapeutic target for GC patients.
Collapse
Affiliation(s)
- Jiaobao Huang
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Shuangshuang Hou
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Jian Xu
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Ju Wu
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Jiajun Yin
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| |
Collapse
|
12
|
Landeros N, Santoro PM, Carrasco-Avino G, Corvalan AH. Competing Endogenous RNA Networks in the Epithelial to Mesenchymal Transition in Diffuse-Type of Gastric Cancer. Cancers (Basel) 2020; 12:cancers12102741. [PMID: 32987716 PMCID: PMC7598708 DOI: 10.3390/cancers12102741] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary The diffuse-type of gastric cancer is associated with epithelial to mesenchymal transition. Loss of E-cadherin expression is the hallmark of this process and is largely due to the upregulation of the transcription factors ZEB1/2, Snail, Slug, and Twist1/2. However, miRNA and lncRNAs can also participate through these transcription factors which directly target E-cadherin. The competing endogenous RNA (ceRNA) network hypothesis state that lncRNA can sponge the miRNA pool that targets these transcripts. Based on the lack of said networks in the epithelial to mesenchymal transition, we performed a prediction analysis that resulted in novel ceRNA networks which will expand our knowledge of the molecular basis of the diffuse-type of gastric cancer. Abstract The diffuse-type of gastric cancer (DGC), molecularly associated with epithelial to mesenchymal transition (EMT), is increasing in incidence. Loss of E-cadherin expression is the hallmark of the EMT process and is largely due to the upregulation of the EMT-inducing transcription factors ZEB1/2, Snail, Slug, and Twist1/2. However, ncRNA, such as miRNA and lncRNAs, can also participate in the EMT process through the direct targeting of E-cadherin and other EMT-inducing transcription factors. Additionally, lncRNA can sponge the miRNA pool that targets these transcripts through competing endogenous RNA (ceRNA) networks. In this review, we focus on the role of ncRNA in the direct deregulation of E-cadherin, as well as EMT-inducing transcription factors. Based on the relevance of the ceRNA network hypothesis, and the lack of said networks in EMT, we performed a prediction analysis for all miRNAs and lncRNAs that target E-cadherin, as well as EMT-inducing transcription factors. This analysis resulted in novel predicted ceRNA networks for E-cadherin and EMT-inducing transcription factors (EMT-TFs), as well as the expansion of the molecular basis of the DGC.
Collapse
Affiliation(s)
- Natalia Landeros
- Advanced Center for Chronic Diseases, Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (N.L.); (P.M.S.)
- Advanced Center for Chronic Diseases, Universidad de Chile, Santiago 8380000, Chile
| | - Pablo M. Santoro
- Advanced Center for Chronic Diseases, Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (N.L.); (P.M.S.)
- Advanced Center for Chronic Diseases, Universidad de Chile, Santiago 8380000, Chile
| | - Gonzalo Carrasco-Avino
- Department of Pathology, Hospital Clinico Universidad de Chile and Clinica Las Condes, Santiago 7550000, Chile;
| | - Alejandro H. Corvalan
- Advanced Center for Chronic Diseases, Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (N.L.); (P.M.S.)
- Advanced Center for Chronic Diseases, Universidad de Chile, Santiago 8380000, Chile
- Correspondence: ; Tel.: +56-2235-48289
| |
Collapse
|
13
|
Lu J, Zhao Z, Ma Y. miR-186 Represses Proliferation, Migration, Invasion, and EMT of Hepatocellular Carcinoma via Directly Targeting CDK6. Oncol Res 2020; 28:509-518. [PMID: 32698940 PMCID: PMC7751224 DOI: 10.3727/096504020x15954139263808] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The present study aimed to investigate the effect of miR-186 on proliferation, migration, invasion, and epithelialmesenchymal transition (EMT) of hepatocellular carcinoma (HCC). In this work, miR-186 was downregulated in HCC tissues and cells, and low miR-186 level helped predict the occurrence of vascular invasion and poor prognosis in patients with HCC. miR-186 overexpression inhibited cell proliferation and tumor growth in nude mice, repressed migration and invasion abilities, and enhanced apoptosis in HCC cells. miR-186 also retarded progression of EMT. miR-186 directly bound to the 3-untranslated regions of cyclin-dependent kinase 6 (CDK6) to inhibit its expression. Overexpression of CDK6 markedly reversed inhibitory effects of miR-186 on proliferation, apoptosis, migration, and invasion of HCC cells. Conversely, inhibition of CDK6 exerted synergic effect on the biological functions of miR-186. In conclusion, miR-186 represses proliferation, migration, invasion, and EMT, and induces apoptosis through targeting CDK6 in HCC, which may provide a new therapeutic target for HCC.
Collapse
Affiliation(s)
- Junfeng Lu
- Department of Vascular Surgery, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong UniversityJinanP.R. China
| | - Zhongsong Zhao
- Department of Gastroenterology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong UniversityJinanP.R. China
| | - Yanhong Ma
- Department of Ultrasound, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong UniversityJinanP.R. China
| |
Collapse
|
14
|
Guo Z, Lv X, Jia H. MiR-186 represses progression of renal cell cancer by directly targeting CDK6. Hum Cell 2020; 33:759-767. [PMID: 32266659 DOI: 10.1007/s13577-020-00357-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/02/2020] [Indexed: 12/29/2022]
Abstract
The function of miR-186 in the progression of renal cell carcinoma (RCC) remains poorly investigated. Our study aims to identify the molecular mechanism underlying miR-186-regulated proliferation, migration and invasion of RCC. Firstly, our data confirmed that miR-186 was significantly reduced and CDK6 was obviously increased in RCC tissues and cells. MiR-186 or CDK6 was associated with advanced TNM stage, lymph node metastasis and poor prognosis. MiR-186 significantly inhibited cell proliferation, migration, invasion and in vivo tumor growth, induced apoptosis, and blocked cell cycle progression in G0/G1 phase. MiR-186 also induced Bax expression and inhibited the expressions of Bcl-2, cyclin D1 and epithelial-mesenchymal transition (EMT)-related genes. Additionally, CDK6 expression was downregulated by miR-186 via binding to its 3'-untranslated region (3'-UTR). Moreover, ectopic expression of CDK6 could partially abrogate the inhibitory effect of miR-186. In conclusion, miR-186 suppresses proliferation, migration and invasion of RCC by inhibiting CDK6 expression.
Collapse
Affiliation(s)
- Zhen Guo
- Department of Urology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, 250033, Shandong, China
| | - Xianbao Lv
- Department of Urology, Chengwu People's Hospital, Heze, 274200, Shandong, China
| | - Haiyan Jia
- Department of Intensive Care Unit, Shandong Provincial Third Hospital, Shandong University, No.11, Central Wuying Hill Road, Jinan, 250031, Shandong, China.
| |
Collapse
|
15
|
Anauate AC, Leal MF, Calcagno DQ, Gigek CO, Karia BTR, Wisnieski F, dos Santos LC, Chen ES, Burbano RR, Smith MAC. The Complex Network between MYC Oncogene and microRNAs in Gastric Cancer: An Overview. Int J Mol Sci 2020; 21:ijms21051782. [PMID: 32150871 PMCID: PMC7084225 DOI: 10.3390/ijms21051782] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/20/2020] [Accepted: 02/25/2020] [Indexed: 12/24/2022] Open
Abstract
Despite the advancements in cancer treatments, gastric cancer is still one of the leading causes of death worldwide. In this context, it is of great interest to discover new and more effective ways of treating this disease. Accumulated evidences have demonstrated the amplification of 8q24.21 region in gastric tumors. Furthermore, this is the region where the widely known MYC oncogene and different microRNAs are located. MYC deregulation is key in tumorigenesis in various types of tissues, once it is associated with cell proliferation, survival, and drug resistance. microRNAs are a class of noncoding RNAs that negatively regulate the protein translation, and which deregulation is related with gastric cancer development. However, little is understood about the interactions between microRNAs and MYC. Here, we overview the MYC role and its relationship with the microRNAs network in gastric cancer aiming to identify potential targets useful to be used in clinic, not only as biomarkers, but also as molecules for development of promising therapies.
Collapse
Affiliation(s)
- Ana Carolina Anauate
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil
| | - Mariana Ferreira Leal
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
| | - Danielle Queiroz Calcagno
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém PA 66075-110, Brazil; (D.Q.C.); (R.R.B.)
| | - Carolina Oliveira Gigek
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
- Departamento de Patologia, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil
| | - Bruno Takao Real Karia
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
| | - Fernanda Wisnieski
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
- Disciplina de Gastroenterologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil
| | - Leonardo Caires dos Santos
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
| | - Elizabeth Suchi Chen
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
| | - Rommel Rodríguez Burbano
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém PA 66075-110, Brazil; (D.Q.C.); (R.R.B.)
- Laboratório de Citogenética Humana, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém PA 66075-110, Brazil
- Laboratório de Biologia Molecular, Hospital Ophir Loyola, Belém PA 66063-240, Brazil
| | - Marília Arruda Cardoso Smith
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
- Correspondence: ; Tel.: +55-11-5576-4848
| |
Collapse
|
16
|
Xiang Y, Tian Q, Guan L, Niu SS. The Dual Role of miR-186 in Cancers: Oncomir Battling With Tumor Suppressor miRNA. Front Oncol 2020; 10:233. [PMID: 32195180 PMCID: PMC7066114 DOI: 10.3389/fonc.2020.00233] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/11/2020] [Indexed: 12/18/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs which regulate gene expression at post-transcriptional level. Alterations of miR-186 expression were demonstrated in numerous cancers, shown to play a vital role in oncogenesis, invasion, metastasis, apoptosis, and drug resistance. MiR-186 was documented as a tumor suppressor miRNA in the majority of studies, while conflicting reports verified miR-186 as an oncomir. The contradictory role in cancers may impede the application of miR-186, as well as other dual-functional miRNAs, as a diagnostic and therapeutic target. This review emphasizes the alterations and functions of miR-186 in cancers and discusses the mechanisms behind the contradictory findings. Among these, target abundance and dose-dependent effects of miR-186 are highlighted. The paper aims to review the challenges involved in developing diagnostic and therapeutic strategies for cancer treatment based on dual-functional miRNAs.
Collapse
Affiliation(s)
- Ying Xiang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Hubei, China.,Department of Cell Biology and Genetics, School of Basic Medicine, Health Science Center, Yangtze University, Hubei, China
| | - Qing Tian
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Hubei, China
| | - Li Guan
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Hubei, China
| | - Shuai-Shuai Niu
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Hubei, China.,The First School of Clinical Medicine, Health Science Center, Yangtze University, Hubei, China
| |
Collapse
|
17
|
Martínez-Barriocanal Á, Arango D, Dopeso H. PVT1 Long Non-coding RNA in Gastrointestinal Cancer. Front Oncol 2020; 10:38. [PMID: 32083000 PMCID: PMC7005105 DOI: 10.3389/fonc.2020.00038] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
Whole genome and transcriptome sequencing technologies have led to the identification of many long non-coding RNAs (lncRNAs) and stimulated the research of their role in health and disease. LncRNAs participate in the regulation of critical signaling pathways including cell growth, motility, apoptosis, and differentiation; and their expression has been found dysregulated in human tumors. Thus, lncRNAs have emerged as new players in the initiation, maintenance and progression of tumorigenesis. PVT1 (plasmacytoma variant translocation 1) lncRNA is located on chromosomal 8q24.21, a large locus frequently amplified in human cancers and predictive of increased cancer risk in genome-wide association studies (GWAS). Combined, colorectal and gastric adenocarcinomas are the most frequent tumor malignancies and also the leading cause of cancer-related deaths worldwide. PVT1 expression is elevated in gastrointestinal tumors and correlates with poor patient prognosis. In this review, we discuss the mechanisms of action underlying PVT1 oncogenic role in colorectal and gastric cancer such as MYC upregulation, miRNA production, competitive endogenous RNA (ceRNA) function, protein stabilization, and epigenetic regulation. We also illustrate the potential role of PVT1 as prognostic biomarker and its relationship with resistance to current chemotherapeutic treatments.
Collapse
Affiliation(s)
- Águeda Martínez-Barriocanal
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.,Group of Molecular Oncology, IRB Lleida, Lleida, Spain
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.,Group of Molecular Oncology, IRB Lleida, Lleida, Spain
| | - Higinio Dopeso
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
18
|
Shen X, Jiang H, Chen Z, Lu B, Zhu Y, Mao J, Chai K, Chen W. MicroRNA-145 Inhibits Cell Migration and Invasion in Colorectal Cancer by Targeting TWIST. Onco Targets Ther 2019; 12:10799-10809. [PMID: 31849487 PMCID: PMC6911328 DOI: 10.2147/ott.s216147] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 10/31/2019] [Indexed: 12/27/2022] Open
Abstract
Introduction MicroRNAs function as oncogenes or tumor suppressors in the development of various human cancers. We investigated the effect of microRNA-145 (miR-145) on colorectal cancer (CRC) cell invasion and migration. Methods The levels of miR-145 in CRC cells were examined by quantitative PCR; Western blotting was used to detect TWIST1 (twist family bHLH transcription factor 1) protein and the epithelial–mesenchymal transition (EMT)-related proteins (E-cadherin, vimentin). Then, we transfected miR-145 mimics or inhibitor into CRC cells and used the wound healing and Transwell invasion assays to investigate their migration and invasive capability, respectively. Results The miR-145 mimics suppressed CRC cell invasion and migration significantly; in contrast, miR-145 downregulation had the opposite effect. Furthermore, miR-145 regulated TWIST1 levels negatively at transcriptional level. TWIST1 knockdown significantly inhibited the CRC cell migration ability and the number of CRC cells that crossed the Transwell membrane. There was no significant difference in terms of migration and invasive capability after the cells had been transfected with miR-145 mimics or inhibitor plus TWIST1 small interfering RNA (siRNA) as compared to the TWIST1 siRNA–only group. Furthermore, we demonstrate that the inhibition of miR-145 could enhance the capability for lung metastasis in vivo. Conclusion Taken together, these findings indicate that miR-145 acts as a new tumor suppressor by regulating TWIST1 and plays a vital role in the invasive and migration ability of CRC cells.
Collapse
Affiliation(s)
- Xuning Shen
- Department of Gastroenterological Surgery, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
| | - Honggang Jiang
- Department of Gastroenterological Surgery, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
| | - Zhiheng Chen
- Department of Gastroenterological Surgery, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
| | - Bohao Lu
- Department of Gastroenterological Surgery, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
| | - Yi Zhu
- Department of Gastroenterological Surgery, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
| | - Jiayan Mao
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang 310012, People's Republic of China
| | - Kequn Chai
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang 310012, People's Republic of China.,Department of Medical Oncology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, People's Republic of China
| | - Wei Chen
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang 310012, People's Republic of China.,Department of Medical Oncology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, People's Republic of China
| |
Collapse
|
19
|
Mercatelli N, Fortini D, Palombo R, Paronetto MP. Small molecule inhibition of Ewing sarcoma cell growth via targeting the long non coding RNA HULC. Cancer Lett 2019; 469:111-123. [PMID: 31639426 DOI: 10.1016/j.canlet.2019.10.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 01/02/2023]
Abstract
Ewing sarcomas (ES) are aggressive pediatric cancers of bone and soft tissues characterized by in frame chromosomal translocations giving rise to chimeric transcription factors, such as EWS-FLI1. An emerging strategy to block EWS-FLI1 activity is represented by the small molecule YK-4-279, which binds to EWS-FLI1 and alters its transcriptional activity. The specific effectors of the anti-oncogenic activity of YK-4-279 are still largely unknown. Herein, by performing a high-throughput screening we identify the lncRNA HULC (Highly Upregulated in Liver Cancer) as a prominent target of YK-4-279 activity in ES cells. High levels of HULC correlate with ES aggressiveness, whereas HULC depletion reduces ES cell growth. Mechanistically, we find that HULC promotes the expression of TWIST1 oncogene by sponging miR-186. Downregulation of HULC upon treatment with YK-4-279 reduces the expression of TWIST1 by unleashing miR-186 and favoring its binding to TWIST1 transcripts. Notably, high levels of miR-186 and low levels of TWIST1 correlate with better prognosis in ES patients. Our results disclose a novel oncogenic regulatory circuit mediated by HULC lncRNA that is disrupted by the small molecule YK-4-279, with promising therapeutic implications for ES treatment.
Collapse
Affiliation(s)
- Neri Mercatelli
- Laboratory of Molecular and Cellular Neurobiology, IRCCS Santa Lucia Foundation, Rome, 00143, Italy.
| | - Diana Fortini
- Laboratory of Molecular and Cellular Neurobiology, IRCCS Santa Lucia Foundation, Rome, 00143, Italy
| | - Ramona Palombo
- Laboratory of Molecular and Cellular Neurobiology, IRCCS Santa Lucia Foundation, Rome, 00143, Italy
| | - Maria Paola Paronetto
- Laboratory of Molecular and Cellular Neurobiology, IRCCS Santa Lucia Foundation, Rome, 00143, Italy; Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Piazza Lauro de Bosis 6, 00135, Rome, Italy.
| |
Collapse
|
20
|
Ayala-Domínguez L, Olmedo-Nieva L, Muñoz-Bello JO, Contreras-Paredes A, Manzo-Merino J, Martínez-Ramírez I, Lizano M. Mechanisms of Vasculogenic Mimicry in Ovarian Cancer. Front Oncol 2019; 9:998. [PMID: 31612116 PMCID: PMC6776917 DOI: 10.3389/fonc.2019.00998] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 09/17/2019] [Indexed: 12/30/2022] Open
Abstract
Solid tumors carry out the formation of new vessels providing blood supply for growth, tumor maintenance, and metastasis. Several processes take place during tumor vascularization. In angiogenesis, new vessels are derived from endothelial cells of pre-existing vessels; while in vasculogenesis, new vessels are formed de novo from endothelial progenitor cells, creating an abnormal, immature, and disorganized vascular network. Moreover, highly aggressive tumor cells form structures similar to vessels, providing a pathway for perfusion; this process is named vasculogenic mimicry (VM), where vessel-like channels mimic the function of vessels and transport plasma and blood cells. VM is developed by numerous types of aggressive tumors, including ovarian carcinoma which is the second most common cause of death among gynecological cancers. VM has been associated with poor patient outcome and survival in ovarian cancer, although the involved mechanisms are still under investigation. Several signaling molecules have an important role in VM in ovarian cancer, by regulating the expression of genes related to vascular, embryogenic, and hypoxic signaling pathways. In this review, we provide an overview of the current knowledge of the signaling molecules involved in the promotion and regulation of VM in ovarian cancer. The clinical implications and the potential benefit of identification and targeting of VM related molecules for ovarian cancer treatment are also discussed.
Collapse
Affiliation(s)
- Lízbeth Ayala-Domínguez
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Leslie Olmedo-Nieva
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Programa de Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - J Omar Muñoz-Bello
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Adriana Contreras-Paredes
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Imelda Martínez-Ramírez
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Marcela Lizano
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
21
|
Wang Z, Sha HH, Li HJ. Functions and mechanisms of miR-186 in human cancer. Biomed Pharmacother 2019; 119:109428. [PMID: 31525641 DOI: 10.1016/j.biopha.2019.109428] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/27/2019] [Accepted: 09/02/2019] [Indexed: 12/30/2022] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression at the post-transcriptional level. Mounting evidence suggests the involvement of miRNAs in carcinogenesis and the development of human cancer. Among the miRNAs, miR-186 has been extensively studied in various cancers. The expression of miR-186 in tissues varies depending on the type of cancer and miR-186 in tissues and body fluids may serve as a marker for the diagnosis and prognosis of cancers. Various biological processes in human cancer are affected by miR-186. Additionally, miR-186 itself is regulated by several factors. Thus, this evidence highlights the potential value of miR-186 in the diagnosis, prognosis and treatment of human cancer.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Orthopedics, Taizhou Clinical Medical School of Nanjing Medical University (Taizhou People's Hospital), Taizhou, Jiangsu, China
| | - Huan-Huan Sha
- Department of Chemotherapy, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, China
| | - Hai-Jun Li
- Department of Orthopedics, Taizhou Clinical Medical School of Nanjing Medical University (Taizhou People's Hospital), Taizhou, Jiangsu, China.
| |
Collapse
|
22
|
Zare A, Alipoor B, Omrani MD, Zali MR, Malekpour Alamdari N, Ghaedi H. Decreased miR-155-5p, miR-15a, and miR-186 Expression in Gastric Cancer Is Associated with Advanced Tumor Grade and Metastasis. IRANIAN BIOMEDICAL JOURNAL 2019; 23. [PMID: 31103022 PMCID: PMC6661124 DOI: 10.29252/.23.5.338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Background Gastric cancer (GC) is one of the most prevalent cancers with a high rate of mortality in the world. In recent years, microRNAs (miRNAs) have been proposed to be involved in GC development. In this study, we aimed at investigating differential expression level of miR-155-5p, miR-15a, miR-15b, and miR-186 in GC. Methods For this research, we used qPCR to investigate miR-15b, miR-155, miR-15a, and miR-186 expression levels in a total of 29 normal gastric tissue, 45 gastric dysplasia, and 39 GC samples. Results We showed significant down-regulation of miR-155-5p (p = 0.0018), miR-15a (p = 0.0159), and miR-186 (p = 0.0005) expression in GC tissue. Conclusion This study provides evidence for deregulated expression of miR155-5p, miR-186, and miR-15a in GC and is providing new insights into the potential implication of these miRNAs in the pathogenesis of GC.
Collapse
Affiliation(s)
- Ali Zare
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnam Alipoor
- Department of Laboratory Sciences, Faculty of Paramedicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; ,Urogenital Stem Cell Research, Shahid Beheshti University of Medical Sciences, Tehran, Iran; ,Corresponding Authors: Mir Davood Omrani ,Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Velenjak st., Shahid Chamran Highway, Tehran, Iran. Tel.: (+98-21) 22439982, E-mail: , Hamid Ghaedi , Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Velenjak st., Shahid Chamran Highway, Tehran, Iran. Tel.: (+98-21) 22439982, E-mail:
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasser Malekpour Alamdari
- Department of General Surgery, Clinical Research and Development Unit at Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Ghaedi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; ,Corresponding Authors: Mir Davood Omrani ,Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Velenjak st., Shahid Chamran Highway, Tehran, Iran. Tel.: (+98-21) 22439982, E-mail: , Hamid Ghaedi , Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Velenjak st., Shahid Chamran Highway, Tehran, Iran. Tel.: (+98-21) 22439982, E-mail:
| |
Collapse
|
23
|
Billa PA, Faulconnier Y, Ye T, Chervet M, Le Provost F, Pires JAA, Leroux C. Deep RNA-Seq reveals miRNome differences in mammary tissue of lactating Holstein and Montbéliarde cows. BMC Genomics 2019; 20:621. [PMID: 31362707 PMCID: PMC6668132 DOI: 10.1186/s12864-019-5987-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
Background Genetic polymorphisms are known to influence milk production and composition. However, the genomic mechanisms involved in the genetic regulation of milk component synthesis are not completely understood. MicroRNAs (miRNAs) regulate gene expression. Previous research suggests that the high developmental potential of the mammary gland may depend in part on a specific miRNA expression pattern. The objective of the present study was to compare the mammary gland miRNomes of two dairy cow breeds, Holstein and Montbéliarde, which have different mammogenic potentials that are related to differences in dairy performance. Results Milk, fat, protein, and lactose yields were lower in Montbéliarde cows than in Holstein cows. We detected 754 distinct miRNAs in the mammary glands of Holstein (n = 5) and Montbéliarde (n = 6) midlactating cows using RNA-Seq technology, among which 738 were known and 16 were predicted miRNAs. The 25 most abundant miRNAs accounted for 90.6% of the total reads. The comparison of their abundances in the mammary glands of Holstein versus Montbéliarde cows identified 22 differentially expressed miRNAs (Padj ≤ 0.05). Among them, 11 presented a fold change ≥2, and 2 (miR-100 and miR-146b) were highly expressed. Among the most abundant miRNAs, miR-186 is known to inhibit cell proliferation and epithelial-to-mesenchymal transition. Data mining showed that 17 differentially expressed miRNAs with more than 20 reads were involved in the regulation of mammary gland plasticity. Several of them may potentially target mRNAs involved in signaling pathways (such as mTOR) and lipid metabolism, thereby indicating that they could influence milk composition. Conclusion We found differences in the mammary gland miRNomes of two dairy cattle breeds. These differences suggest a potential role for miRNAs in mammary gland plasticity and milk component synthesis, both of which are related to milk production and composition. Further research is warranted on the genetic regulation of miRNAs and their role in milk synthesis. Electronic supplementary material The online version of this article (10.1186/s12864-019-5987-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- P A Billa
- Institut National de la Recherche Agronomique (INRA), Université Clermont Auvergne, VetAgro Sup, UMR Herbivores, UMR1213 Herbivores, F-63122, Saint-Genès-Champanelle, France
| | - Y Faulconnier
- Institut National de la Recherche Agronomique (INRA), Université Clermont Auvergne, VetAgro Sup, UMR Herbivores, UMR1213 Herbivores, F-63122, Saint-Genès-Champanelle, France
| | - T Ye
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique, UMR7104, Institut National de la Santé et de la Recherche Médicale, U964, Université de Strasbourg, 67404, Illkirch, France
| | - M Chervet
- Department of Food Science & Technology, University of California Davis, Davis, CA, USA
| | - F Le Provost
- GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78352, France
| | - J A A Pires
- Institut National de la Recherche Agronomique (INRA), Université Clermont Auvergne, VetAgro Sup, UMR Herbivores, UMR1213 Herbivores, F-63122, Saint-Genès-Champanelle, France
| | - C Leroux
- Institut National de la Recherche Agronomique (INRA), Université Clermont Auvergne, VetAgro Sup, UMR Herbivores, UMR1213 Herbivores, F-63122, Saint-Genès-Champanelle, France. .,Department of Food Science & Technology, University of California Davis, Davis, CA, USA.
| |
Collapse
|
24
|
Chen H, Li M, Huang P. LncRNA SNHG16 Promotes Hepatocellular Carcinoma Proliferation, Migration and Invasion by Regulating miR-186 Expression. J Cancer 2019; 10:3571-3581. [PMID: 31293662 PMCID: PMC6603422 DOI: 10.7150/jca.28428] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 04/30/2019] [Indexed: 12/11/2022] Open
Abstract
Long non-coding RNA (lncRNA) and microRNA (miRNA) play an important role in genesis and progression of tumors. The aim of this study was to explore the expression, biological function and molecular mechanism of small nucleolar RNA host gene 16 (SNHG16) in HCC. RT-qPCR was conducted to evaluate the expression level of SNHG16 in HCC tissues and cell lines. Our findings showed for the first time that SNHG16 was up-regulated in HCC tissues and cell lines. The expression of SNHG16 in cancer tissues was highly correlated with tumor size, TNM stage, ALT expression level and HBV DNA level. Moreover, cell proliferation, migration and invasion were detected by CCK-8 assay, transwell migration assay and transwell invasion assay, respectively. Xenograft tumor experiment was used to determine the biological function of SNHG16 in vivo. As revealed by our data, SNHG16 accelerated the proliferation, migration and invasion of HCC cell. SNHG16 facilitated tumor formation in vivo. Next, the relationship between SNHG16, miR-186 and ROCK1 were analyzed using bioinformatics analysis, qRT-PCR, luciferase reporter assay and western blot. Further molecular mechanism studies reported that the expression of SNHG16 was negatively correlated with the level of miR-186 and SNHG16 directly bound to miR-186. SNHG16 and miR-186 repressed each other. Notably, rescue experiments were conducted and showed that miR-186 reversed the effect of SNHG16 on cell. Taken together, SNHG16 promoted HCC cell proliferation, migration and invasion by functioning as a competitive endogenous RNA (ceRNA) to negatively regulate miR-186 expression. Our data suggested that SNHG16 might be a potential biomarker and a new therapeutic target for HCC.
Collapse
Affiliation(s)
- Hang Chen
- National Key Clinical Department, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 400000, P.R. China
| | - Molin Li
- National Key Clinical Department, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 400000, P.R. China
| | - Ping Huang
- National Key Clinical Department, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 400000, P.R. China
| |
Collapse
|
25
|
Li L, Zhang Z, Yang Q, Ning M. Lycorine inhibited the cell growth of non-small cell lung cancer by modulating the miR-186/CDK1 axis. Life Sci 2019; 231:116528. [PMID: 31176784 DOI: 10.1016/j.lfs.2019.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/30/2019] [Accepted: 06/01/2019] [Indexed: 12/24/2022]
Abstract
AIMS Lycorine is a kind of natural alkaloid with anti-cancer potential. It has been demonstrated that lycorine processes high activity and specificity against the progression of cancers. However, the underlying molecular mechanisms by which lycorine regulates the formation and development of non-small cell lung cancer (NSCLC) remain largely unknown. MAIN METHODS The effects of lycorine on the growth of NSCLC cells were determined by the cell counting kit-8 (CCK-8) assay, colony formation and flow cytometry analysis. RT-qPCR was performed to detect the expression of microRNA with lycorine treatment. The binding of miRNA and target genes was confirmed by luciferase reporter assay. KEY FINDINGS Lycorine significantly inhibited the proliferation and induced apoptosis of NSCLC cells. Mechanistically, lycorine up-regulated the expression of microRNA-186 in NSCLC cells. Depletion of miR-186 significantly reversed the suppressive effect of lycorine on the proliferation of NSCLC cells. Furthermore, the cyclin dependent kinase 1 (CDK1) was identified as one of the binding candidates of miR-186. Experimental analysis showed that miR-186 bound the 3'-untranslated region (3'-UTR) of CDK1 and suppressed the level of CDK1 in NSCLC cells. Consistently, exposure of lycorine significantly decreased the expression of CDK1. Restoration of CDK1 remarkably attenuated the inhibition of lycorine on the proliferation of NSCLC cells. SIGNIFICANCE Our results uncovered the novel molecular mechanism of lycorine in suppressing the progression of NSCLC partially via regulating the miR-186/CDK1 axis.
Collapse
Affiliation(s)
- Li Li
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei 061001, China
| | - Zao Zhang
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei 061001, China
| | - Qian Yang
- Traditional Chinese Medicine Department, Cangzhou Central Hospital, Cangzhou, Hebei, China, 061001
| | - Meiying Ning
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei 061001, China.
| |
Collapse
|
26
|
Zhu K, Su Y, Xu B, Wang Z, Sun H, Wang L, Sun C, He X. MicroRNA-186-5p represses neuroblastoma cell growth via downregulation of Eg5. Am J Transl Res 2019; 11:2245-2256. [PMID: 31105832 PMCID: PMC6511750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 01/29/2019] [Indexed: 06/09/2023]
Abstract
Micro RNA (miRNAs) is a kind of non coding small RNAs with negative regulation function, which plays an important role in regulating the occurrence and development of tumors. In this study, we analyzed the expression level and role of miRNA-186-5p and Eg5 in neuroblastoma and neuroblastoma cell lines SHSY-5Y, Kelly, NBL-S and SK-N-AS. Results of Real-time PCR and immunohistochemistry showed that the expression level of Eg5 in tumor tissues was higher than that in tumor adjacent tissues, while miRNA-186-5p expression level in tumor tissues was lower than that in tumor adjacent tissues. miRNA-186-5p mimics or Eg5 siRNA was transfected into SHSY-5Y and Kelly cells, CCK-8 and soft agar clone formation tests' results showed that the cell proliferation was inhibited. Flow cytometry analysis of cell apoptosis and cell cycle showed that overexpression of Mi-186-5p or down-regulation of Eg5 could promote cell apoptosis and lead to arrest cell cycle at G1 phase. Bioinformatics predicts that miRNA-186-5p can bind to the 3'UTR of Eg5. Luciferase reporter gene analysis and Western blot assay also confirmed that microRNA335-5p could target ICAM-1 to inhibit its expression. The tumor growth in nude mice inoculated SHSY-5Y cells with overexpression of miRNA-186-5p was inhibited. In a word, our study found that miR-186-5p could inhibit tumor proliferation by targeting Eg5 in neuroblastoma. This finding will help to better understand the pathogenesis of neuroblastoma and provide new insights into the treatment of tumors.
Collapse
Affiliation(s)
- Kai Zhu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China Hefei 230001, Anhui, P. R. China
| | - Yilin Su
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China Hefei 230001, Anhui, P. R. China
| | - Bing Xu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China Hefei 230001, Anhui, P. R. China
| | - Zhongrong Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China Hefei 230001, Anhui, P. R. China
| | - Hua Sun
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China Hefei 230001, Anhui, P. R. China
| | - Liang Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China Hefei 230001, Anhui, P. R. China
| | - Chuancheng Sun
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China Hefei 230001, Anhui, P. R. China
| | - Xiaorui He
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China Hefei 230001, Anhui, P. R. China
| |
Collapse
|
27
|
Liu C, Jian M, Qi H, Mao WZ. MicroRNA 495 Inhibits Proliferation and Metastasis and Promotes Apoptosis by Targeting Twist1 in Gastric Cancer Cells. Oncol Res 2019; 27:389-397. [PMID: 29615148 PMCID: PMC7848466 DOI: 10.3727/096504018x15223159811838] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Recently, microRNAs (miRNAs) have been reported to participate in multiple biological processes. However, the effects of miR-495 on gastric cancer (GC) remain unclear. The purpose of this study was to explore the functions of miR-495 in GC cell proliferation, metastasis, and apoptosis. SGC-7901 and BGC-823 cell lines were transfected with miR-495 mimic, miR-495 inhibitor, and negative controls (mimic control and inhibitor control). The expressions of miR-495, cell viability, migration, apoptosis, and apoptosis-related factors were examined by qRT-PCR, trypan blue staining, Transwell, flow cytometry, and Western blot, respectively. Simultaneously, key factor expression levels of EMT were detected by qRT-PCR and Western blot. The direct target of miR-495 was confirmed by dual-luciferase assay. Additionally, sh-Twist1, pc-Twist1, and corresponding controls were transfected into SGC-7901 and BGC-823 cells, and the protein levels of EMT-associated factors were detected by Western blot. miR-495 was downregulated in GC cells. miR-495 expression level was effectively overexpressed or suppressed in SGC-7901 and BGC-823 cells. Overexpression of miR-495 significantly decreased cell viability and migration, increased apoptosis, and inhibited the EMT process. Suppression of miR-495 showed contrary results. Twist1 was clarified as a target gene of miR-495, and Twist1 silencing obviously reduced the promoting effect of miR-495 suppression on these biological processes. Twist1 silencing significantly blocked the EMT process in both SGC-7901 and BGC-823 cells. miR-495 inhibited proliferation and metastasis and promoted apoptosis by targeting Twist1 in GC cells. These data indicated that miR-495 might be a novel antitumor factor of GC and provide a new method for the treatment of GC.
Collapse
Affiliation(s)
- Chao Liu
- *Qingdao University, Qingdao, P.R. China
- †Department of General Surgery, Qingdao Municipal Hospital, Qingdao, P.R. China
| | - Min Jian
- ‡Department of Laboratory Medicine, Qingdao Women and Children’s Hospital, Qingdao, P.R. China
| | - Hong Qi
- †Department of General Surgery, Qingdao Municipal Hospital, Qingdao, P.R. China
| | - Wei-Zheng Mao
- †Department of General Surgery, Qingdao Municipal Hospital, Qingdao, P.R. China
| |
Collapse
|
28
|
Norouzinia M, Zamanian Azodi M, Najafgholizadeh Seyfi D, Kardan A, Naseh A, Akbari Z. Predication of hub target genes of differentially expressed microRNAs contributing to Helicobacter pylori infection in gastric non-cancerous tissue. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2019; 12:S44-S50. [PMID: 32099601 PMCID: PMC7011053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AIM The main goal of this investigation was to provide an overview on H.pylori effect on gastric tissue via bioinformatics analysis of microarray-identified miRNAs and its target genes. BACKGROUND MicroRNAs which control about 30 to 60% of gene expression in human body play a critical role in different cell growth stages. Expression modification of non-coding (NC) RNAs in H.pylori infections requires further investigations to provide better understanding of their roles in the body. METHODS GSE54397, the microRNA microarray dataset, was analyzed by GEO2R, the online GEO database for detection of differentially expressed microRNAs and lastly the potential target genes as well as their associated pathways. RESULTS A total of 244 miRNAs were detected as differentially expressed (p<0.05 and FC>2) in non-cancerous tissue of gastric with H.pylori infection in comparison with tissues without H.pylori infection. The findings indicated that hub microRNAs and target genes of up-regulated network are KIF9, DCTN3, and CA5BP1 along with hsa-miR-519d, hsa-miR-573, hsa-miR-646, hsa-miR-92a-1, hsa-miR-186, and hsa-miR-892a, respectively. For the down-regulated network, genes of RABGAP1, HSPB11 and microRNAs of hsa-miR-620, hsa-miR-19b-2, hsa-miR-555, and hsa-let-7f-2 were hubs. Most of the up-regulated microRNAs are involved in gastric cancer development while there is no evidence for the down-regulated ones. Yet, all of the hub down-regulated miRNAs are reported to have associations with different kinds of cancer. CONCLUSION The introduced hub miRNAs and genes may serve as feasible markers in the mechanisms of H.pylori infection for different kinds of gastric diseases, in particular gastric cancer. However, their role requires further investigations.
Collapse
Affiliation(s)
- Mohsen Norouzinia
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Zamanian Azodi
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Diba Najafgholizadeh Seyfi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Kardan
- Tehran Medical Branch, Islamic Azad University, Tehran, Iran
| | - Ali Naseh
- Pediatric and Neonatal Ward, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Akbari
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Dai J, Wang K, Liu T, Wang Q, Pang Y. Retracted Article: Long non-coding RNA KCNQ1OT1 regulates cell proliferation, apoptosis and chemo-sensitivity through modulating the miR-186-5p/NCAM1 axis in acute myeloid leukemia cells. RSC Adv 2019; 9:36256-36265. [PMID: 35540579 PMCID: PMC9074956 DOI: 10.1039/c9ra06378a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/25/2019] [Indexed: 11/21/2022] Open
Abstract
Recent studies show that lncRNA KCNQ1OT1 and microRNA-186-5p (miR-186-5p) are involved in various human cancers. Moreover, it is reported that KCNQ1OT1 expression is upregulated in acute myeloid leukemia (AML). However, their roles in AML remain unknown. This study aimed to reveal the functional mechanism of KCNQ1OT1 and miR-186-5p in AML development. Quantitative real time polymerase chain reaction (qRT-PCR) was performed to detect the levels of genes. Cell proliferation and apoptosis were assessed by a 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay and flow cytometry analysis respectively. A western blot assay was carried out to examine the protein levels. In addition, the interaction between miR-186-5p and KCNQ1OT1 or neural cell adhesion molecule 1 (NCAM1) was predicted by bioinformatics analysis tool starbase2.0 and confirmed by the dual luciferase reporter assay. KCNQ1OT1 and NCAM1 expressions were increased and miR-186-5p expression was decreased in AML samples and cells. The depletion of KCNQ1OT1 inhibited cell proliferation, and promoted apoptosis and chemo-sensitivity in AML. In addition, the upregulation of miR-186-5p suppressed AML cell proliferation, and induced apoptosis and chemo-sensitivity. Interestingly, KCNQ1OT1 directly downregulated miR-186-5p expression and miR-186-5p decreased NCAM1 expression by binding to the 3′ untranslated region (UTR) of NCAM1 mRNA. Furthermore, miR-186-5p knockdown or NCAM1 overexpression reversed the effects of KCNQ1OT1 depletion on AML cell progression. Our results firstly revealed a linear relationship between KCNQ1OT1, miR-186-5p, and NCAM1, and demonstrated that KCNQ1OT1 mediated AML cell progression via regulating the miR-186-5p/NCAM1 axis, revealing functional mechanisms of KCNQ1OT1 and miR-186-5p in AML development. Recent studies show that lncRNA KCNQ1OT1 and microRNA-186-5p (miR-186-5p) are involved in various human cancers.![]()
Collapse
Affiliation(s)
- Jing Dai
- Department of Hematology
- Zhoukou Central Hospital
- Zhengzhou
- China
| | - Kai Wang
- Department of Hematology
- Zhoukou Central Hospital
- Zhengzhou
- China
| | - Tao Liu
- Department of Hematology
- Zhoukou Central Hospital
- Zhengzhou
- China
| | - Qiong Wang
- Department of Hematology
- Zhoukou Central Hospital
- Zhengzhou
- China
| | - Yingxu Pang
- Department of Hematology
- Zhoukou Central Hospital
- Zhengzhou
- China
| |
Collapse
|
30
|
Sun WJ, Zhang YN, Xue P. miR-186 inhibits proliferation, migration, and epithelial-mesenchymal transition in breast cancer cells by targeting Twist1. J Cell Biochem 2018; 120:10001-10009. [PMID: 30552711 DOI: 10.1002/jcb.28283] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 10/24/2018] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Breast cancer (BC) is the most prevalent malignancy in women worldwide. Our study aimed to investigate the expression and biological effect of miR-186 in BC. METHODS Expression of miR-186 was determined by quantitative reverse transcription PCR. Kaplan-Meier curves were calculated for the survival data analysis. Functional assays were performed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and wound healing assay. Protein expression was analyzed by Western blot. RESULTS miR-186 was downregulated in BC tissues and cells. Downregulation of miR-186 was associated with tumor metastasis and a poor overall survival in patients with BC. Overexpression of miR-186 inhibited BC cells proliferation, migration, and epithelial-mesenchymal transition process; while suppression of miR-186 exhibited an opposite effects on BC cells. In addition, Twist1 was identified as a direct target of miR-186 in BC and restoration of Twist1 attenuated the biological effect of miR-186 on BC cells. CONCLUSION Our findings suggest that miR-186 functions as a tumor suppressor by targeting Twist1 in BC. miR-186 may serve as a novel biomarker in BC diagnosis or a new therapeutic target in BC treatment.
Collapse
Affiliation(s)
- Wen-Juan Sun
- Department of General Surgery, Qingdao West Coast New Area Central Hospital, Qingdao, Shandong, China
| | - Ya-Na Zhang
- Department of Anesthesiology, Qingdao West Coast New Area Central Hospital, Qingdao, Shandong, China
| | - Peng Xue
- Department of Anesthesiology, Qingdao West Coast New Area Central Hospital, Qingdao, Shandong, China
| |
Collapse
|
31
|
Neviani P, Wise PM, Murtadha M, Liu CW, Wu CH, Jong AY, Seeger RC, Fabbri M. Natural Killer-Derived Exosomal miR-186 Inhibits Neuroblastoma Growth and Immune Escape Mechanisms. Cancer Res 2018; 79:1151-1164. [PMID: 30541743 DOI: 10.1158/0008-5472.can-18-0779] [Citation(s) in RCA: 235] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 10/24/2018] [Accepted: 12/06/2018] [Indexed: 12/18/2022]
Abstract
In neuroblastoma, the interplay between immune cells of the tumor microenvironment and cancer cells contributes to immune escape mechanisms and drug resistance. In this study, we show that natural killer (NK) cell-derived exosomes carrying the tumor suppressor microRNA (miR)-186 exhibit cytotoxicity against MYCN-amplified neuroblastoma cell lines. The cytotoxic potential of these exosomes was partly dependent upon expression of miR-186. miR-186 was downregulated in high-risk neuroblastoma patients, and its low expression represented a poor prognostic factor that directly correlated with NK activation markers (i.e., NKG2D and DNAM-1). Expression of MYCN, AURKA, TGFBR1, and TGFBR2 was directly inhibited by miR-186. Targeted delivery of miR-186 to MYCN-amplified neuroblastoma or NK cells resulted in inhibition of neuroblastoma tumorigenic potential and prevented the TGFβ1-dependent inhibition of NK cells. Altogether, these data support the investigation of a miR-186-containing nanoparticle formulation to prevent tumor growth and TGFβ1-dependent immune escape in high-risk neuroblastoma patients as well as the inclusion of ex vivo-derived NK exosomes as a potential therapeutic option alongside NK cell-based immunotherapy.Significance: These findings highlight the therapeutic potential of NK cell-derived exosomes containing the tumor suppressor miR-186 that inhibits growth, spreading, and TGFβ-dependent immune escape mechanisms in neuroblastoma.
Collapse
Affiliation(s)
- Paolo Neviani
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Petra M Wise
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Mariam Murtadha
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Cathy W Liu
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Chun-Hua Wu
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Ambrose Y Jong
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Robert C Seeger
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Muller Fabbri
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California.
| |
Collapse
|
32
|
Xie ZC, Huang JC, Zhang LJ, Gan BL, Wen DY, Chen G, Li SH, Yan HB. Exploration of the diagnostic value and molecular mechanism of miR‑1 in prostate cancer: A study based on meta‑analyses and bioinformatics. Mol Med Rep 2018; 18:5630-5646. [PMID: 30365107 PMCID: PMC6236292 DOI: 10.3892/mmr.2018.9598] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 09/24/2018] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (PCa) remains a principal issue to be addressed in male cancer-associated mortality. Therefore, the present study aimed to examine the clinical value and associated molecular mechanism of microRNA (miR)-1 in PCa. A meta-analysis was conducted to evaluate the diagnosis of miR-1 in PCa via Gene Expression Omnibus and ArrayExpress datasets, The Cancer Genome Atlas miR-1 expression data and published literature. It was identified that expression of miR-1 was significantly downregulated in PCa. Decreased miR-1 expression possessed moderate diagnostic value, with area under the curve, sensitivity, specificity and odds ratio values at 0.73, 0.77, 0.57 and 4.60, respectively. Using bioinformatics methods, it was revealed that a number of pathways, including the ‘androgen receptor signaling pathway’, ‘androgen receptor activity’, ‘transcription factor binding’ and ‘protein processing in the endoplasmic reticulum’, were important in PCa. A total of seven hub genes, including phosphoribosylaminoimidazole carboxylase and phosphoribosylaminoimidazolesuccin ocarboxamide synthase (PAICS), cadherin 1 (CDH1), SRC proto-oncogene, non-receptor tyrosine kinase, twist family bHLH transcription factor 1 (TWIST1), ZW10 interacting kinetochore protein (ZWINT), PCNA clamp associated factor (KIAA0101) and androgen receptor, among which, five (PAICS, CDH1, TWIST1, ZWINT and KIAA0101) were significantly upregulated and negatively correlated with miR-1, were identified as key miR-1 target genes in PCa. Additionally, it was investigated whether miR-1 and its hub genes were associated with clinical features, including age, tumor status, residual tumor, lymph node metastasis, pathological T stage and prostate specific antigen level. Collectively the results suggest that miR-1 may be involved in the progression of PCa, and consequently be a promising diagnostic marker. The ‘androgen receptor signaling pathway’, ‘androgen receptor activity’, ‘transcription factor binding’ and ‘protein processing in the endoplasmic reticulum’ may be crucial interactive pathways in PCa. Furthermore, PAICS, CDH1, TWIST1, ZWINT and KIAA0101 may serve as crucial miR-1 target genes in PCa.
Collapse
Affiliation(s)
- Zu-Cheng Xie
- Department of Urological Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jia-Cheng Huang
- Department of Urological Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Li-Jie Zhang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Bin-Liang Gan
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Dong-Yue Wen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Sheng-Hua Li
- Department of Urological Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Hai-Biao Yan
- Department of Urological Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
33
|
Downregulation of MiR-1297 predicts poor prognosis and enhances gastric cancer cell growth by targeting CREB1. Biomed Pharmacother 2018; 105:413-419. [DOI: 10.1016/j.biopha.2018.05.094] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/19/2018] [Accepted: 05/20/2018] [Indexed: 11/24/2022] Open
|
34
|
Huang T, Wang G, Yang L, Peng B, Wen Y, Ding G, Wang Z. MiR-186 inhibits proliferation, migration, and invasion of non-small cell lung cancer cells by downregulating Yin Yang 1. Cancer Biomark 2018; 21:221-228. [PMID: 29060934 DOI: 10.3233/cbm-170670] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the main type of lung cancer. While miR-186 is significantly reduced in lung cancer tissues and cells, its role in NSCLC has not been completely elucidated. MATERIAL AND METHODS We used qRT-PCR and western blot methods to investigate the levels of miR-186 and YY1 in 21 pairs of NSCLC tissues. Dual luciferase reporter gene assays were performed to detect whether miR-186 directly targets YY1. Next, the roles of miR-186 and its target gene (YY1) in determining the proliferation, apoptosis and migration capabilities of selected cell lines (A549 and HCC827) were investigated by using miR-186 mimics or YY1 siRNA. RESULTS Our results showed that miR-186 was downregulated and YYI was upregulated in NSCLC tissue, and miR-186 expression was negatively associated with YY1. Similarly, miR-186 was also downregulated and YY1 expression also was upregulated in both A549 and HCC827 cells; furthermore, miR-186 was found to directly target YY1. Cell proliferation, invasion, and migration, as well as apoptosis induction were more strongly inhibited by YY1 siRNA than by miR-186. CONCLUSION Our results suggest that miR-186 and its target gene (YY1) could possibly serve as new prognostic biomarkers and therapeutic targets for treating NSCLC in humans.
Collapse
|
35
|
Liu Q, Jiang J, Fu Y, Liu T, Yu Y, Zhang X. MiR-129-5p functions as a tumor suppressor in gastric cancer progression through targeting ADAM9. Biomed Pharmacother 2018; 105:420-427. [PMID: 29879625 DOI: 10.1016/j.biopha.2018.05.105] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 05/07/2018] [Accepted: 05/21/2018] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are identified as key regulators in cancer initiation, progression and metastasis including gastric cancer (GC). The aim of the study is to explore clinical significance and potential mechanism of miR-129-5p in GC development. In the study, our results found that miR-129-5p expression was significantly downregulated in GC tissues, compared with adjacent normal tissues using qRT-PCR analyses. Furthermore, lower miR-129-5p expression closely associated with tumor size and lymph node invasion and poor prognosis of GC patients. Using CCK8 assay, cell colony formation, transwell invasion assay, we demonstrated that miR-129-5p overexpression reduced cell proliferation, cell colony formation and cell invasion capacity in MKN45 (higher miR-129-5p expression) and SGC-7901 (lower miR-129-5p expression). However, downregulation of miR-129-5p had reverse effects on cell proliferation and invasion. Targeting association analysis, dual luciferase assay, qRT-PCR and western blot analysis results verified that miR-129-5p could target the 3'UTR of ADAM9 mRNA and regulated its protein expression. Furthermore, we confirmed that miR-129-5p suppressed cell proliferation and invasion ability through regulating ADAM9. In vivo, upregulation of miR-129-5p also inhibited tumor growth. Therefore, these results indicated that miR-129-5p functioned as a tumor suppressor in GC and may be a potential target of GC treatment.
Collapse
Affiliation(s)
- Qi Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, 450000, China
| | - Jianwu Jiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, 450000, China
| | - Yang Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, 450000, China
| | - Tao Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, 450000, China
| | - Yang Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, 450000, China
| | - Xiefu Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, 450000, China.
| |
Collapse
|
36
|
Zhang J, Gao D, Zhang H. Upregulation of miR-614 promotes proliferation and inhibits apoptosis in ovarian cancer by suppressing PPP2R2A expression. Mol Med Rep 2018; 17:6285-6292. [PMID: 29532877 PMCID: PMC5928608 DOI: 10.3892/mmr.2018.8714] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 11/16/2017] [Indexed: 12/22/2022] Open
Abstract
It has previously been demonstrated that microRNAs (miRNAs) have essential roles and participate in various biological processes by regulating their specific target genes. However, the precise role of miRNAs in ovarian cancer (OC) has not yet been elucidated. The present study demonstrated that miR‑614 expression levels were significantly upregulated in OC tissues and cell lines, whereas decreased miR‑614 demonstrated opposite effects. Furthermore, gain‑of‑function and loss‑of‑function experiments indicated that miR‑614 overexpression promoted cell proliferation and suppressed cell apoptosis. Protein phosphatase 2 regulatory subunit B α, (PPP2R2A) was identified as a direct target of miR‑614 using western blotting and luciferase reporter assays. Notably, silencing of PPP2R2A counter‑acted the effect of miR‑614 inhibitor in OC cell proliferation and cell apoptosis. Overall, the data suggested that miR‑614 promoted cell proliferation and inhibited cell apoptosis of OC cells by targeting PPP2R2A, and may therefore act as a potential target for OC therapy in the future.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Traditional Chinese Medicine Gynecology, Central Hospital of Zhumadian, Huang Huai University, Zhumadian, Henan 463000, P.R. China
| | - Dongdong Gao
- Department of Oncology, Central Hospital of Zhumadian, Huang Huai University, Zhumadian, Henan 463000, P.R. China
| | - Hui Zhang
- Department of Traditional Chinese Medicine Gynecology, Central Hospital of Zhumadian, Huang Huai University, Zhumadian, Henan 463000, P.R. China
| |
Collapse
|
37
|
MicroRNA-186 serves as a tumor suppressor in oral squamous cell carcinoma by negatively regulating the protein tyrosine phosphatase SHP2 expression. Arch Oral Biol 2018; 89:20-25. [DOI: 10.1016/j.archoralbio.2018.01.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 12/13/2022]
|
38
|
Jones DZ, Schmidt ML, Suman S, Hobbing KR, Barve SS, Gobejishvili L, Brock G, Klinge CM, Rai SN, Park J, Clark GJ, Agarwal R, Kidd LR. Micro-RNA-186-5p inhibition attenuates proliferation, anchorage independent growth and invasion in metastatic prostate cancer cells. BMC Cancer 2018; 18:421. [PMID: 29653561 PMCID: PMC5899400 DOI: 10.1186/s12885-018-4258-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 03/20/2018] [Indexed: 12/17/2022] Open
Abstract
Background Dysregulation of microRNA (miRNA) expression is associated with hallmarks of aggressive tumor phenotypes, e.g., enhanced cell growth, proliferation, invasion, and anchorage independent growth in prostate cancer (PCa). Methods Serum-based miRNA profiling involved 15 men diagnosed with non-metastatic (stage I, III) and metastatic (stage IV) PCa and five age-matched disease-free men using miRNA arrays with select targets confirmed by quantitative real-time PCR (qRT-PCR). The effect of miR-186-5p inhibition or ectopic expression on cellular behavior of PCa cells (i.e., PC-3, MDA-PCa-2b, and LNCaP) involved the use bromodeoxyuridine (BrdU) incorporation, invasion, and colony formation assays. Assessment of the impact of miR-186-5p inhibition or overexpression on selected targets entailed microarray analysis, qRT-PCR, and/or western blots. Statistical evaluation used the modified t-test and ANOVA analysis. Results MiR-186-5p was upregulated in serum from PCa patients and metastatic PCa cell lines (i.e., PC-3, MDA-PCa-2b, LNCaP) compared to serum from disease-free individuals or a normal prostate epithelial cell line (RWPE1), respectively. Inhibition of miR-186-5p reduced cell proliferation, invasion, and anchorage-independent growth of PC-3 and/or MDA-PCa-2b PCa cells. AKAP12, a tumor suppressor target of miR-186-5p, was upregulated in PC-3 and MDA-PCa-2b cells transfected with a miR-186-5p inhibitor. Conversely, ectopic miR-186-5p expression in HEK 293 T cells decreased AKAP12 expression by 30%. Both pAKT and β-catenin levels were down-regulated in miR-186-5p inhibited PCa cells. Conclusions Our findings suggest miR-186-5p plays an oncogenic role in PCa. Inhibition of miR-186-5p reduced PCa cell proliferation and invasion as well as increased AKAP12 expression. Future studies should explore whether miR-186-5p may serve as a candidate prognostic indicator and a therapeutic target for the treatment of aggressive prostate cancer. Electronic supplementary material The online version of this article (10.1186/s12885-018-4258-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dominique Z Jones
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.,James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA.,Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Denver, USA
| | - M Lee Schmidt
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.,James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Suman Suman
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.,James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Katharine R Hobbing
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.,James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Shirish S Barve
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.,Division of Gastroenterology and Hepatology, University of Louisville School of Medicine, Louisville, USA
| | - Leila Gobejishvili
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.,Division of Gastroenterology and Hepatology, University of Louisville School of Medicine, Louisville, USA
| | - Guy Brock
- Department of Biomedical Informatics, The Ohio State University, Columbus, USA
| | - Carolyn M Klinge
- James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA.,Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, USA
| | - Shesh N Rai
- James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA.,Department of Bioinformatics and Biostatistics, University of Louisville School of Public Health and Information Science, Louisville, USA
| | - Jong Park
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, USA
| | - Geoffrey J Clark
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.,James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Denver, USA
| | - LaCreis R Kidd
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA. .,James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA.
| |
Collapse
|
39
|
Zhang J, Jin M, Chen X, Zhang R, Huang Y, Liu H, Zhu J. Loss of PPM1F expression predicts tumour recurrence and is negatively regulated by miR-590-3p in gastric cancer. Cell Prolif 2018; 51:e12444. [PMID: 29473240 DOI: 10.1111/cpr.12444] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/15/2018] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVES MicroRNAs (miRNAs) as small non-coding RNA molecules act by negatively regulating their target genes. Recent studies have shown that protein phosphatase Mg2+/Mn2+-dependent 1F (PPM1F) plays a critical role in cancer metastasis. But, the regulation mechanisms of PPM1F by miRNAs in gastric cancer (GC) remain undefined. METHODS The correlation of PPM1F or miR-590-3p (miR-590) expression with clinicopathological features and prognosis of the patients with GC was analysed by TCGA RNA-sequencing data. The miRNAs that target PPM1F gene were identified by bioinformatics and Spearman correlation analysis, and the binding site between miR-590 and PPM1F 3'UTR was confirmed by dual luciferase assay. MTT and Transwell assays were conducted to evaluate the effects of miR-590 or (and) PPM1F on cell proliferation and invasion. RESULTS We found that PPM1F expression was downregulated in GC tissues and cell lines and was correlated with tumour recurrence in patients with GC. The decreased expression of PPM1F was attributed to the dysregulation of miR-590 expression rather than its genetic or epigenetic alterations. Overexpression of miR-590 promoted cell proliferation and invasion capability of GC cells, while knockdown of miR-590 reversed these effects. Moreover, PPM1F was validated as a direct target of miR-590 and counteracted the tumour-promoting effects caused by miR-590. The expression of miR-590 presented the negative correlation with PPM1F expression and acted as an independent prognostic factor for tumour recurrence in patients with GC. CONCLUSION PPM1F may function as a suppressive factor and is negatively regulated by miR-590 in GC.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Ming Jin
- Department of Clinical Medicine, Ningbo University School of Medicine, Ningbo, China
| | - Xiaoyu Chen
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Rui Zhang
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yanxia Huang
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Hui Liu
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jinshui Zhu
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
40
|
Wu R, Shen D, Sohun H, Ge D, Chen X, Wang X, Chen R, Wu Y, Zeng J, Rong X, Su X, Chu M. miR‑186, a serum microRNA, induces endothelial cell apoptosis by targeting SMAD6 in Kawasaki disease. Int J Mol Med 2018; 41:1899-1908. [PMID: 29344637 PMCID: PMC5810213 DOI: 10.3892/ijmm.2018.3397] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 01/16/2018] [Indexed: 12/11/2022] Open
Abstract
Kawasaki disease (KD) is an acute, self‑limited vasculitis that predominantly affects medium‑sized arteries, particularly the coronary arteries. Recent studies have indicated that microRNAs are involved in many diseases, including KD. However, the detailed mechanism remains unclear. The aim of the present study was to explore the role of miR‑186 in KD and potentially discover a new target for KD treatment. The results demonstrated that miR‑186 was upregulated in serum from patients with KD and KD serum could increase miR‑186 transcript levels in endothelial cells (HUVECs). Overexpression of miR‑186 mimic induced HUVEC apoptosis through mitogen‑activated protein kinase (MAPK) activation by targeting and inhibiting SMAD family member 6 (SMAD6). Furthermore, KD serum induced HUVEC apoptosis through miR‑186. In conclusion, the present results suggested that KD serum‑associated miR‑186 has an essential role in endothelial cell apoptosis by activating the MAPK pathway through targeting the SMAD6 gene.
Collapse
Affiliation(s)
- Rongzhou Wu
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Danping Shen
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Hareshwaree Sohun
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Donghui Ge
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Xianda Chen
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Xuliang Wang
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Ruiyao Chen
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Yuqing Wu
- Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Jingjing Zeng
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Xing Rong
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Xiaoping Su
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Maoping Chu
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| |
Collapse
|
41
|
Wang Y, Chen F, Zhao M, Yang Z, Li J, Zhang S, Zhang W, Ye L, Zhang X. The long noncoding RNA HULC promotes liver cancer by increasing the expression of the HMGA2 oncogene via sequestration of the microRNA-186. J Biol Chem 2017; 292:15395-15407. [PMID: 28765279 DOI: 10.1074/jbc.m117.783738] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/13/2017] [Indexed: 01/13/2023] Open
Abstract
The long noncoding RNA highly up-regulated in liver cancer (HULC) is aberrantly elevated in hepatocellular carcinoma (HCC), and this up-regulation is crucial for HCC pathogenesis. However, the underlying mechanism in HULC up-regulation is poorly understood. We hypothesized that HULC might modulate the oncogene high mobility group A2 (HMGA2) to promote hepatocarcinogenesis. Quantitative real-time PCR analysis showed that the expression levels of HULC were positively correlated with those of HMGA2 in clinical HCC tissues. Interestingly, we also observed that HULC could up-regulate HMGA2 in HCC cells. Mechanistically, we found that the microRNA-186 inhibited HMGA2 expression by targeting the 3'-untranslated region (3'-UTR) of HMGA2 mRNA. Strikingly, HULC acted as a competing noncoding RNA to sequester miR-186 and thereby relieved miR-186-mediated HMGA2 repression. Functionally, HMGA2 knockdown decreased the HULC-enhanced growth of HCC cells both in vitro and in vivo We conclude that the long noncoding RNA HULC increases HMGA2 expression by sequestering miR-186 post-transcriptionally and thereby promotes liver cancer growth, providing new insights into the mechanism by which HULC enhances hepatocarcinogenesis.
Collapse
Affiliation(s)
- Yuan Wang
- From the State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China and
| | - Fuquan Chen
- From the State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China and
| | - Man Zhao
- From the State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China and
| | - Zhe Yang
- From the State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China and
| | - Jiong Li
- From the State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China and
| | - Shuqin Zhang
- From the State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China and
| | - Weiying Zhang
- From the State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China and
| | - Lihong Ye
- the State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiaodong Zhang
- From the State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China and
| |
Collapse
|
42
|
Lu Y, Tang L, Zhang Q, Zhang Z, Wei W. MicroRNA-613 inhibits the progression of gastric cancer by targeting CDK9. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:980-984. [PMID: 28701053 DOI: 10.1080/21691401.2017.1351983] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
MicroRNAs (miRNAs) play an important role in the development and progression of human malignancy. miR-613, as a tumour suppressor, was reported to decrease in several tumours. However, the expression levels and role of miR-613 in gastric cancer remain unknown. In this study, we found that miR-613 was evidently downregulated in gastric cancer tissue and cell. The functional analysis showed that miR-613 suppressed cell proliferation and migration in gastric cancer. Next, the dual-luciferase reporter system supported CDK9 as a direct target gene of miR-613. miR-613 mimics evidently repressed CDK9 expression in gastric cancer cells. Furthermore, we found that CDK9 in upregulated in gastric cancer and the CDK9 expression levels were inversely correlated with that of miR-613 in gastric cancer tissues. Overall, the results revealed that miR-613, as a tumour suppressor, involves in gastric cancer progression and metastasis by targeting CDK9, implying a novel potential therapeutic target for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Yebin Lu
- a Department of General Surgery , Xiangya Hospital, Central South University , Changsha , China
| | - Ling Tang
- b Department of Pharmacy , Xiangya Hospital, Central South University , Changsha , China
| | - Qi Zhang
- a Department of General Surgery , Xiangya Hospital, Central South University , Changsha , China
| | - Zhipeng Zhang
- a Department of General Surgery , Xiangya Hospital, Central South University , Changsha , China
| | - Wei Wei
- a Department of General Surgery , Xiangya Hospital, Central South University , Changsha , China
| |
Collapse
|
43
|
Jiao D, Wu M, Ji L, Liu F, Liu Y. MicroRNA-186 Suppresses Cell Proliferation and Metastasis Through Targeting Sentrin-Specific Protease 1 in Renal Cell Carcinoma. Oncol Res 2017; 26:249-259. [PMID: 28550686 PMCID: PMC7844750 DOI: 10.3727/096504017x14953948675430] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Recent evidence suggests that dysregulation of microRNAs is associated with the development of multiple malignancies. miR-186 has been reported as a critical cancer regulator in several types of cancers. However, its functional significance and molecular mechanism underlying renal cell carcinoma (RCC) remain unknown. In this study, our results showed that miR-186 expression was dramatically downregulated in RCC tissues and cell lines compared to that in adjacent normal tissues and cell lines. Overexpression of miR-186 significantly inhibited cell growth, colony formation, and cell invasion; caused cell cycle arrest at the G0/G1 phase; and induced cell apoptosis as detected by MTT, colony formation, Transwell assay, and flow cytometry assays in RCC cells. In addition, inhibition of miR-186 expression promoted RCC cell proliferation, invasion, and cell cycle progression and reduced apoptosis. Bioinformatics analysis and luciferase reporter assay confirmed that the 3′-UTR of sentrin-specific protease 1 (SENP1) was a direct target of miR-186. A remarkably reverse correlation was observed between miR-186 and SENP1 mRNA in RCC tissues. Furthermore, immunohistochemical staining revealed that SENP1 was positively expressed in RCC specimens. Restoration of SENP1 expression could partially abrogate the inhibitory effect of miR-186 overexpression on RCC cell proliferation through activating NF-κB signaling and its downstream proteins. These data demonstrated that miR-186 acted as a novel tumor suppressor and potential therapeutic biomarker in the progression of RCC by directly targeting SENP1.
Collapse
Affiliation(s)
- Dan Jiao
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Man Wu
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Lei Ji
- Department of Cardiology, Changchun Central Hospital, Changchun, Jilin, P.R. China
| | - Feng Liu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Yingying Liu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, P.R. China
| |
Collapse
|