1
|
Zhu C, Lin L, Huang C, Li Z. Circ-NMNAT1 Drives Tumor Progression in Bladder Cancer by Modulating the miR-370-3p/ATXN2L Axis. Appl Biochem Biotechnol 2025; 197:3008-3025. [PMID: 39820928 DOI: 10.1007/s12010-024-05162-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2024] [Indexed: 01/19/2025]
Abstract
The relationship between circular RNAs (circRNAs) and tumor growth and metastasis is increasingly well-established. In this study, we sought to shed light on circ-NMNAT1's potential molecular mechanisms in bladder cancer (BCa). circ-NMNAT1, miR-370-3p, and ATXN2L expression profiles were explored using RT-qPCR and/or Western blot techniques. Cell proliferation was detected by MTT and colony formation assay. Transwell assay was used to detect the migration and invasion ability of cells. Western Blot was used to detect the protein expression level of ATXN2L. The targeting relationship between miR-370-3p and circ-NMNAT1 or ATXN2L was confirmed by dual luciferase reporter gene and RIP assay. A xenograft tumor model was created to investigate circ-NMNAT1's function in BCa in vivo. The high expression of circ-NMNAT1 was measured in BCa. circ-NMNAT1 bound competitively to miR-370-3p and downregulated miR-370-3p expression. After knocking down circ-NMNAT1, the proliferation ability of EJ cells was significantly inhibited, and the number of cell colonies was (80.00 ± 7.10). The number of migrated and invaded cells was significantly reduced by (35.49 ± 0.05)% and (59.00 ± 0.04)%, respectively, after silencing circ-NMNAT1. In addition, downregulation of circ-NMNAT1 also significantly increased the apoptosis rate of EJ cells by (23.55 ± 2.95)%. Knockdown of miR-370-3p or overexpression of ATXN2L reduced the effect of circ-NMNAT1 silencing on BCa cells. The promoting effect of circ-NMNAT1 on BCa progression was further validated in vivo tumor models. The weight and volume of the tumor were significantly inhibited after circ-NMNAT1 knockdown, which were (87.50 ± 20.40) mg and (238.90 ± 21.38) mm3, respectively. Circ-NMNAT1 is highly expressed in BCa and promotes the proliferation, migration, and invasion of BCa cells by regulating the miR-370-3p/ATXN2L axis, thereby accelerating the progression of BCa. Our results suggest that circ-NMNAT1 may be a new therapeutic target for BCa.
Collapse
Affiliation(s)
- ChenHui Zhu
- Department of Urology, Central People's Hospital of Zhanjiang, No.236, Yuanzhu Road, Chikan District, Zhanjiang City, 524037, Guangdong Province, China
| | - LiJuan Lin
- Department of Anaesthesia, Central People's Hospital of Zhanjiang, Zhanjiang City, 524037, Guangdong Province, China
| | - ChangQing Huang
- Department of Urology, Central People's Hospital of Zhanjiang, No.236, Yuanzhu Road, Chikan District, Zhanjiang City, 524037, Guangdong Province, China
| | - ZhaoGuan Li
- Department of Urology, Central People's Hospital of Zhanjiang, No.236, Yuanzhu Road, Chikan District, Zhanjiang City, 524037, Guangdong Province, China.
| |
Collapse
|
2
|
Shahani A, Slika H, Elbeltagy A, Lee A, Peters C, Dotson T, Raj D, Tyler B. The epigenetic mechanisms involved in the treatment resistance of glioblastoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:12. [PMID: 40201311 PMCID: PMC11977385 DOI: 10.20517/cdr.2024.157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/21/2024] [Accepted: 12/03/2024] [Indexed: 04/10/2025]
Abstract
Glioblastoma (GBM) is an aggressive malignant brain tumor with almost inevitable recurrence despite multimodal management with surgical resection and radio-chemotherapy. While several genetic, proteomic, cellular, and anatomic factors interplay to drive recurrence and promote treatment resistance, the epigenetic component remains among the most versatile and heterogeneous of these factors. Herein, the epigenetic landscape of GBM refers to a myriad of modifications and processes that can alter gene expression without altering the genetic code of cancer cells. These processes encompass DNA methylation, histone modification, chromatin remodeling, and non-coding RNA molecules, all of which have been found to be implicated in augmenting the tumor's aggressive behavior and driving its resistance to therapeutics. This review aims to delve into the underlying interactions that mediate this role for each of these epigenetic components. Further, it discusses the two-way relationship between epigenetic modifications and tumor heterogeneity and plasticity, which are crucial to effectively treat GBM. Finally, we build on the previous characterization of epigenetic modifications and interactions to explore specific targets that have been investigated for the development of promising therapeutic agents.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Betty Tyler
- Hunterian Neurosurgical Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
3
|
Leili FR, Shali N, Sheibani M, Jafarian MJ, Pashizeh F, Gerami R, Iraj F, Lashkarshekan AA. Detailed pathological role of non-coding RNAs (ncRNAs) in regulating drug resistance of glioblastoma, and update. Pathol Res Pract 2024; 263:155590. [PMID: 39326365 DOI: 10.1016/j.prp.2024.155590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024]
Abstract
Glioma is a kind of brain tumor that develops in the central nervous system and is classified based on its histology and molecular genetic features. The lifespan of patients does not exceed 22 months. One of the motives for the low effectiveness of glioma treatment is its radioresistance and chemoresistance. Noncoding RNAs (ncRNAs) are a diverse set of transcripts that do not undergo translation to become proteins in glioma. The ncRNAs have been identified as significant regulators of several biological processes in different cell types and tissues, and their abnormal function has been linked to glioma. They are known to impact important occurrences, including carcinogenesis, progression, and enhanced treatment resistance in glioma cells. The ncRNAs control cell proliferation, migration, epithelial-to-mesenchymal transition (EMT), invasion, and drug resistance in glioma cells. The main focus of this study is to inspect the involvement of ncRNAs in the drug resistance of glioma.
Collapse
Affiliation(s)
- Foad Rahmanpour Leili
- Department of Neurology Faculty of Medicine, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Niloofar Shali
- Department of Clinical Biochemistry, School of Medicine, Shahrood Branch, Islamic Azad University, Shahrood, Iran
| | - Mehrnaz Sheibani
- Division of Pediatric Neurology, University of Tabriz, Tabriz, Iran
| | | | - Fatemeh Pashizeh
- Department of Immunology, School of Medicine, Shahid Sadoughi University of Medical Science, Yazd 8916188635, Iran
| | - Reza Gerami
- Department of Radiology, Faculty of Medicine, AJA University of Medical Science, Tehran, Iran.
| | | | | |
Collapse
|
4
|
Schuster M, Braun FK, Chiang DML, Ludwig C, Meng C, Grätz C, Kirchner B, Proescholdt M, Hau P, Steinlein OK, Pfaffl MW, Riemenschneider MJ, Reithmair M. Extracellular vesicles secreted by 3D tumor organoids are enriched for immune regulatory signaling biomolecules compared to conventional 2D glioblastoma cell systems. Front Immunol 2024; 15:1388769. [PMID: 38726003 PMCID: PMC11079215 DOI: 10.3389/fimmu.2024.1388769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/01/2024] [Indexed: 05/12/2024] Open
Abstract
Background Newer 3D culturing approaches are a promising way to better mimic the in vivo tumor microenvironment and to study the interactions between the heterogeneous cell populations of glioblastoma multiforme. Like many other tumors, glioblastoma uses extracellular vesicles as an intercellular communication system to prepare surrounding tissue for invasive tumor growth. However, little is known about the effects of 3D culture on extracellular vesicles. The aim of this study was to comprehensively characterize extracellular vesicles in 3D organoid models and compare them to conventional 2D cell culture systems. Methods Primary glioblastoma cells were cultured as 2D and 3D organoid models. Extracellular vesicles were obtained by precipitation and immunoaffinity, with the latter allowing targeted isolation of the CD9/CD63/CD81 vesicle subpopulation. Comprehensive vesicle characterization was performed and miRNA expression profiles were generated by smallRNA-sequencing. In silico analysis of differentially regulated miRNAs was performed to identify mRNA targets and corresponding signaling pathways. The tumor cell media and extracellular vesicle proteome were analyzed by high-resolution mass spectrometry. Results We observed an increased concentration of extracellular vesicles in 3D organoid cultures. Differential gene expression analysis further revealed the regulation of twelve miRNAs in 3D tumor organoid cultures (with nine miRNAs down and three miRNAs upregulated). MiR-23a-3p, known to be involved in glioblastoma invasion, was significantly increased in 3D. MiR-7-5p, which counteracts glioblastoma malignancy, was significantly decreased. Moreover, we identified four miRNAs (miR-323a-3p, miR-382-5p, miR-370-3p, miR-134-5p) located within the DLK1-DIO3 domain, a cancer-associated genomic region, suggesting a possible importance of this region in glioblastoma progression. Overrepresentation analysis identified alterations of extracellular vesicle cargo in 3D organoids, including representation of several miRNA targets and proteins primarily implicated in the immune response. Conclusion Our results show that 3D glioblastoma organoid models secrete extracellular vesicles with an altered cargo compared to corresponding conventional 2D cultures. Extracellular vesicles from 3D cultures were found to contain signaling molecules associated with the immune regulatory signaling pathways and as such could potentially change the surrounding microenvironment towards tumor progression and immunosuppressive conditions. These findings suggest the use of 3D glioblastoma models for further clinical biomarker studies as well as investigation of new therapeutic options.
Collapse
Affiliation(s)
- Martina Schuster
- Institute of Human Genetics, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Frank K. Braun
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany
| | - Dapi Meng-Lin Chiang
- Institute of Human Genetics, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Christina Ludwig
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Chen Meng
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Christian Grätz
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Benedikt Kirchner
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Martin Proescholdt
- Department of Neurosurgery, Regensburg University Hospital, Regensburg, Germany
| | - Peter Hau
- Department of Neurology and Wilhelm Sander Neuro-Oncology Unit, Regensburg University Hospital, Regensburg, Germany
| | - Ortrud K. Steinlein
- Institute of Human Genetics, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Michael W. Pfaffl
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | | | - Marlene Reithmair
- Institute of Human Genetics, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
5
|
Rahmani F, Hashemian P, Tabrizi AT, Ghorbani Z, Ziaeemehr A, Alijannejad S, Ferns GA, Avan A, Shahidsales S. Regulatory role of miRNAs on Wnt/β-catenin signaling in tumorigenesis of glioblastoma. Indian J Cancer 2023; 60:295-302. [PMID: 37787188 DOI: 10.4103/ijc.ijc_251_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Glioblastoma (GBM) is one of the most aggressive tumors in the brain with high mortality worldwide. Despite recent advances in therapeutic strategies, the survival rate remains low in patients with GBM. The pathogenesis of GBM is a very complicated process involving various genetic mutations affecting several oncogenic signaling pathways like Wnt/β-catenin axis. Overactivation of the Wnt/β-catenin signaling pathway is associated with decreased survival and poor prognosis in patients with GBM. MicroRNAs (miRNAs) were shown to play important roles in the regulation of cell proliferation, differentiation, apoptosis, and tumorigenesis by modulating the expression of their target genes. Aberrant expression of miRNAs were reported in various human malignancies including GBM, breast, colorectal, liver, and prostate cancers, but little is known about their cellular mechanisms. Therefore, recognition of the expression profile and regulatory effects of miRNAs on the Wnt/β-catenin pathway may offer a novel approach for the classification, diagnosis, prognosis, and treatment of patients with GBM. This review summarizes previous data on the modulatory role of miRNAs on the Wnt/β-catenin pathway implicated in tumorigenesis of GBM.
Collapse
Affiliation(s)
- Farzad Rahmani
- Metabolic Syndrome Research Center; Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pedram Hashemian
- Department of Pathology, Jahad Daneshgahi Institute, Mashhad Branch, Mashhad, Iran
| | | | - Zeynab Ghorbani
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Aghigh Ziaeemehr
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sajede Alijannejad
- Department of Biology, Faculty of Sciences, Mashhad Branch, Islamic Azad University of Mashhad, Mashhad, Iran
| | - Gordon A Ferns
- Brighton and Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex, UK
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
6
|
Hashemi M, Rashidi M, Hushmandi K, Ten Hagen TLM, Salimimoghadam S, Taheriazam A, Entezari M, Falahati M. HMGA2 regulation by miRNAs in cancer: affecting cancer hallmarks and therapy response. Pharmacol Res 2023; 190:106732. [PMID: 36931542 DOI: 10.1016/j.phrs.2023.106732] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
High mobility group A 2 (HMGA2) is a protein that modulates the structure of chromatin in the nucleus. Importantly, aberrant expression of HMGA2 occurs during carcinogenesis, and this protein is an upstream mediator of cancer hallmarks including evasion of apoptosis, proliferation, invasion, metastasis, and therapy resistance. HMGA2 targets critical signaling pathways such as Wnt/β-catenin and mTOR in cancer cells. Therefore, suppression of HMGA2 function notably decreases cancer progression and improves outcome in patients. As HMGA2 is mainly oncogenic, targeting expression by non-coding RNAs (ncRNAs) is crucial to take into consideration since it affects HMGA2 function. MicroRNAs (miRNAs) belong to ncRNAs and are master regulators of vital cell processes, which affect all aspects of cancer hallmarks. Long ncRNAs (lncRNAs) and circular RNAs (circRNAs), other members of ncRNAs, are upstream mediators of miRNAs. The current review intends to discuss the importance of the miRNA/HMGA2 axis in modulation of various types of cancer, and mentions lncRNAs and circRNAs, which regulate this axis as upstream mediators. Finally, we discuss the effect of miRNAs and HMGA2 interactions on the response of cancer cells to therapy. Regarding the critical role of HMGA2 in regulation of critical signaling pathways in cancer cells, and considering the confirmed interaction between HMGA2 and one of the master regulators of cancer, miRNAs, targeting miRNA/HMGA2 axis in cancer therapy is promising and this could be the subject of future clinical trial experiments.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, 4815733971, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, 4815733971, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Timo L M Ten Hagen
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands.
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mojtaba Falahati
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands.
| |
Collapse
|
7
|
Ma H, Qu S, Zhai Y, Yang X. circ_0025033 promotes ovarian cancer development via regulating the hsa_miR-370-3p/SLC1A5 axis. Cell Mol Biol Lett 2022; 27:94. [PMID: 36273140 PMCID: PMC9588225 DOI: 10.1186/s11658-022-00364-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 07/06/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) appear to be important modulators in ovarian cancer. We aimed to explore the role and mechanism of circ_0025033 in ovarian cancer. METHODS qRT-PCR was conducted to determine circ_0025033, hsa_miR-370-3p, and SLC1A5 mRNA expression. Functional experiments were conducted, including Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, transwell, tube formation, xenograft tumor model assay, western blot analysis of protein levels, and analysis of glutamine metabolism using commercial kits. Their predicted interaction was confirmed using dual-luciferase reporter and RNA pull-down. RESULTS circ_0025033 was upregulated in ovarian cancer; its knockdown induced proliferation, invasion, angiogenesis, glutamine metabolism, and apoptosis in vitro, and blocked tumor growth in vivo. circ_0025033 regulated ovarian cancer cellular behaviors via sponging hsa_miR-370-3p. In parallel, SLC1A5 might abolish the anti-ovarian cancer role of hsa_miR-370-3p. Furthermore, circ_0025033 affected SLC1A5 via regulating hsa_miR-370-3p. CONCLUSION circ_0025033 might promote ovarian cancer progression via hsa_miR-370-3p/SLC1A5, providing an interesting insight into ovarian cancer tumorigenesis.
Collapse
Affiliation(s)
- Huiping Ma
- Department of Gynecology, First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710000, Shaanxi, China
| | - Shuyun Qu
- Department of Gynecology, Gynaecology Hospital of Shaanxi Nuclear Industry, Xi'an, Shaanxi, China
| | - Yao Zhai
- Department of Gynecology, Gynaecology Hospital of Shaanxi Nuclear Industry, Xi'an, Shaanxi, China
| | - Xiaofeng Yang
- Department of Gynecology, First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710000, Shaanxi, China.
| |
Collapse
|
8
|
Zheng X, Zhong T, Yu F, Duan J, Tang Y, Liu Y, Li M, Sun D, Yin D. Deficiency of a novel lncRNA-HRAT protects against myocardial ischemia reperfusion injury by targeting miR-370-3p/RNF41 pathway. Front Cardiovasc Med 2022; 9:951463. [PMID: 36172578 PMCID: PMC9510651 DOI: 10.3389/fcvm.2022.951463] [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: 05/24/2022] [Accepted: 08/22/2022] [Indexed: 01/17/2023] Open
Abstract
Accumulating evidence indicates that long non-coding RNAs (lncRNAs) contribute to myocardial ischemia/reperfusion (I/R) injury. However, the underlying mechanisms by which lncRNAs modulate myocardial I/R injury have not been thoroughly examined and require further investigation. A novel lncRNA named lncRNA-hypoxia/reoxygenation (H/R)-associated transcript (lncRNA-HRAT) was identified by RNA sequencing analysis. The expression of lncRNA-HRAT exhibited a significant increase in the I/R mice hearts and cardiomyocytes treated with H/R. LncRNA-HRAT overexpression facilitates H/R-induced cardiomyocyte apoptosis. Furthermore, cardiomyocyte-specific deficiency of lncRNA-HRAT in vivo after I/R decreased creatine kinase (CK) release in the serum, reduced myocardial infarct area, and improved cardiac dysfunction. Molecular mechanistic investigations revealed that lncRNA-HRAT serves as a competing endogenous RNA (ceRNA) of miR-370-3p, thus upregulating the expression of ring finger protein 41 (RNF41), thereby aggravating apoptosis in cardiomyocytes induced by H/R. This study revealed that the lncRNA-HRAT/miR-370-3p/RNF41 pathway regulates cardiomyocyte apoptosis and myocardial injury. These findings suggest that targeted inhibition of lncRNA-HRAT may offer a novel therapeutic method to prevent myocardial I/R injury.
Collapse
Affiliation(s)
- Xinbin Zheng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, China
| | - Ting Zhong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Fan Yu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Jingsi Duan
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Yao Tang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Yaxiu Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Mingrui Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Deqiang Sun
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Deling Yin
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
- *Correspondence: Deling Yin,
| |
Collapse
|
9
|
Chen Q, Zhou L, Ma D, Hou J, Lin Y, Wu J, Tao M. LncRNA GAS6-AS1 facilitates tumorigenesis and metastasis of colorectal cancer by regulating TRIM14 through miR-370-3p/miR-1296-5p and FUS. J Transl Med 2022; 20:356. [PMID: 35962353 PMCID: PMC9373365 DOI: 10.1186/s12967-022-03550-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/19/2022] [Indexed: 12/24/2022] Open
Abstract
Background Long non-coding RNAs (lncRNAs) are essential regulators of tumorigenesis and the development of colorectal cancer (CRC). Here, we aimed to investigate the role of lncRNA GAS6-AS1 in CRC and its potential mechanisms. Methods Bioinformatics analyses evaluated the level of GAS6-AS1 in colon cancer, its correlation with clinicopathological factors, survival curve and diagnostic value. qRT-PCR were performed to detect the GAS6-AS1 level in CRC samples and cell lines. The CCK8, EdU, scratch healing, transwell assays and animal experiments were conducted to investigate the function of GAS6-AS1 in CRC. RNA immunoprecipitation (RIP) and dual-luciferase reporter gene analyses were carried out to reveal interaction between GAS6-AS1, TRIM14, FUS, and miR-370-3p/miR-1296-5p. Results GAS6-AS1 was greatly elevated in CRC and positively associated with unfavorable prognosis of CRC patients. Functionally, GAS6-AS1 positively regulates CRC proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) in vitro and induces CRC growth and metastasis in vivo. Moreover, GAS6-AS1 exerted oncogenic function by competitively binding to miR-370-3p and miR-1296-5p, thereby upregulating TRIM14. Furthermore, we verified that GAS6-AS1 and TRIM14 both interact with FUS and that GAS6-AS1 stabilized TRIM14 mRNA by recruiting FUS. Besides, rescue experiments furtherly demonstrated that GAS6-AS1 facilitate progression of CRC by regulating TRIM14. Conclusion Collectively, these findings demonstrate that GAS6-AS1 promotes TRIM14-mediated cell proliferation, migration, invasion, and EMT of CRC via ceRNA network and FUS-dependent manner, suggesting that GAS6-AS1 could be utilized as a novel biomarker and therapeutic target for CRC. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03550-0.
Collapse
Affiliation(s)
- Qing Chen
- Department of Oncology, Jingjiang People's Hospital, The Seventh Affiliated Hospital of Yangzhou University, Jingjiang, Jiangsu, China.,Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lin Zhou
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - De Ma
- Department of Oncology, Jingjiang People's Hospital, The Seventh Affiliated Hospital of Yangzhou University, Jingjiang, Jiangsu, China
| | - Juan Hou
- Department of Oncology, Jingjiang People's Hospital, The Seventh Affiliated Hospital of Yangzhou University, Jingjiang, Jiangsu, China
| | - Yuxin Lin
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jie Wu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Min Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China. .,Department of Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
10
|
Li J, Ma J, Zhou L, Huang S, Sun J, Chen L, Lu Z. Green Synthesized Bismuth Oxide Nanoparticles Using Aqueous Rhizome Extract of Curcuma longa Mitigate the Proliferation of Human U87 Glioblastoma Cells by Regulation of the Wnt/ β-Catenin Signaling Pathway. J Biomed Nanotechnol 2022. [DOI: 10.1166/jbn.2022.3345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The low biocompatibility of inorganic nanoparticles (NPs) is a main concern in their wide applications in the biomedical field. Therefore, the green synthesis of NPs from plant extracts can provide safe NPs for biomedical applications. The present study was aimed to assess the anticancer
activity of bismuth oxide (Bi2O3) NPs fabricated using aqueous plant extracts from the rhizome of Curcuma longa (C. longa). Characterization of green Bi2O3 NPs was done using TEM, DLS, and X-ray diffraction analyses. Selective anticancer
activity of green Bi2O3 NPs against human glioblastoma (U87) cells was assessed using MTT, GSH, MDA, ROS, apoptosis, and caspase-3 assays. Also, qPCR analysis was done to explore the expression of β-catenin, cyclin D1, and c-myc at mRNA level as the important
genes of the Wnt/β-catenin signaling pathway. The results showed that the green Bi2O3 NPs have a crystalline nature with a size of around 30 nm with good colloidal stability attributed to potential bio-fabrication of Bi2O3 NPs. Cellular
study indicated that green Bi2O3 NPs triggered selective anticancer activity against U87 cells through reduction of GSH level and increase of MDA level, ROS level, Annexin+ cells, and caspase-3 activity. Also, it was found that IC50 concentration
of biosynthesized Bi2O3 NPs (20 μg/mL) resulted in a significant downregulation in the expression of β-catenin, cyclin D1, and c-myc genes involved in the Wnt/β-catenin signaling pathway. This study concludes that green Bi2O3
NPs bio-fabricated from rhizome of C. longa show potential selective anticancer activity.
Collapse
Affiliation(s)
- Jun Li
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 20040, China
| | - Junfeng Ma
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 20040, China
| | - Liang Zhou
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 20040, China
| | - Shan Huang
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 20040, China
| | - Jiahua Sun
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 20040, China
| | - Lin Chen
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 20040, China
| | - Zhengrong Lu
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 20040, China
| |
Collapse
|
11
|
A diagnostic and prognostic value of blood-based circulating long non-coding RNAs in Thyroid, Pancreatic and Ovarian Cancer. Crit Rev Oncol Hematol 2022; 171:103598. [PMID: 35033662 DOI: 10.1016/j.critrevonc.2022.103598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Several studies have demonstrated the potential of circulating long non-coding RNAs (lncRNAs) as promising cancer biomarkers. Herein, we addressed the regulatory role of circulating lncRNAs and their potential value as diagnostic/prognostic markers for thyroid, pancreatic and ovarian cancers. Furthermore, we analyzed and measured the clinical implications and association of lncRNAs with sensitivity, specificity, and area under the ROC curve (AUC). Based on our meta-analysis, we found that GAS8-AS1 could discriminate thyroid cancer from non-cancer and other cancers with higher accuracy (AUC = 0.746; sensitivity = 61.70%, and specificity = 90.00%). Similarly, for ovarian cancer, lncRNA RP5-837J1.2 was found to have ideal diagnostic potential with critical clinical specifications of AUC = 0.996; sensitivity = 97.30% and specificity = 94.60%. Whereas we could not find any lncRNA having high diagnostic/prognostic efficiency in pancreatic cancer. We believe that lncRNAs mentioned above may explore clinical settings for the diagnosis and prognosis of cancer patients.
Collapse
|
12
|
Pan X, Chen G, Hu W. lncRNA HLA Complex Group 18 (HCG18) Facilitated Cell Proliferation, Invasion, and Migration of Prostate Cancer Through Modulating miR-370-3p/DDX3X Axis. Reprod Sci 2021; 28:3406-3416. [PMID: 34708395 DOI: 10.1007/s43032-021-00614-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 05/10/2021] [Indexed: 11/26/2022]
Abstract
Long non-coding RNAs (lncRNAs) have been reported to exert critical functions in the malignant development of many cancers. lncRNA HLA complex group 18 (HCG18) has been confirmed to have a promoting effect on various cancers. However, whether HCG18 functions in PC is still unclear. Therefore, the current study aimed at unveiling the role of HCG18 in PC progression and its regulatory mechanism on the biological behaviors of PC. Here, RT-qPCR was utilized to detect HCG18 expression, and then, functional experiments were conducted to verify the effects of HCG18 on PC cell proliferation, migration, invasion, and apoptosis. According to the results, HCG18 was significantly up-regulated in PC cells and it facilitated cell proliferation, migration, and invasion in PC. Furthermore, a series of mechanism experiments were carried out to verify the relationship among HCG18, miR-370-3p, and DEAD-box helicase 3 X-linked(DDX3X) in PC cells. Final rescue assays showed that DDX3X overexpression could reverse the inhibitory function of silencing HCG18 on PC progression. In summary, our study showed that lncRNA HCG18 accelerated cell proliferation, invasion, and migration of PC via up-regulating DDX3X through sponging miR-370-3p, providing a novel finding about PC-related regulatory mechanism.
Collapse
Affiliation(s)
- Xiaobo Pan
- Department of Urology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, 315040, Zhejiang, China.
| | - Guangyao Chen
- Department of Urology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, 315040, Zhejiang, China
| | - Wenhao Hu
- Department of Urology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, 315040, Zhejiang, China
| |
Collapse
|
13
|
Regulatory interplay between microRNAs and WNT pathway in glioma. Biomed Pharmacother 2021; 143:112187. [PMID: 34560532 DOI: 10.1016/j.biopha.2021.112187] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/01/2021] [Accepted: 09/10/2021] [Indexed: 12/17/2022] Open
Abstract
Glioma is one of the most common neoplasms of the central nervous system with a poor survival. Due to the obstacles in treating this disease, a part of recent studies mainly focuses on identifying the underlying molecular mechanisms that contribute to its malignancy. Altering microRNAs (miRNAs) expression pattern has been identified obviously in many cancers. Through regulating various targets and signaling pathways, miRNAs play a pivotal role in cancer progression. As one of the essential signaling pathways, WNT pathway is dysregulated in many cancers, and a growing body of evidence emphasis its dysregulation in glioma. Herein, we provide a comprehensive review of miRNAs involved in WNT pathway in glioma. Moreover, we show the interplay between miRNAs and WNT pathway in regulating different processes such as proliferation, invasion, migration, radio/chemotherapy resistance, and epithelial-mesenchymal-transition. Then, we introduce several drugs and treatments against glioma, which their effects are mediated through the interplay of WNT pathway and miRNAs.
Collapse
|
14
|
Emerging roles of circUBAP2 targeting miR-370-3p in proliferation, apoptosis, and invasion of papillary thyroid cancer cells. Hum Cell 2021; 34:1866-1877. [PMID: 34346032 DOI: 10.1007/s13577-021-00585-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 07/23/2021] [Indexed: 02/06/2023]
Abstract
Circular RNAs (circRNAs) have been documented to be aberrantly expressed in many types of malignancies and involved in cancer progression. However, their role in thyroid cancer (TC) remains largely unknown. Our study aimed to explore the role and mechanism of circUBAP2 in TC. The differentially expressed circRNAs in TC tissues were identified using GSE18105 from gene expression omnibus (GEO) database. CircUBAP2 and miR-370-3p expression was analyzed using qRT-PCR. The stability of circUBAP2 was confirmed by actinomycin D and RNase R. The subcellular localization of circUBAP2 was detected using cell fractionation assay. Cell proliferation, apoptosis, and invasion were evaluated using MTT, flow cytometry analysis, and Transwell invasion assay, respectively. The interaction between circUBAP2 and miR-370-3p was predicted using bioinformatics analysis and validated by luciferase reporter assay, RNA pull-down assay, and RNA immunoprecipitation. CircUBAP2 was upregulated and miR-370-3p was downregulated in TC tissues and cells. CircUBAP2 was highly stable, resistant to RNase R digestion, and predominantly localized in the cytoplasm. CircUBAP2 knockdown inhibited cell proliferation and invasion and triggered apoptosis in TC cells. Bioinformatics analysis showed that circUBAP2 contained putative binding sites of miR-370-3p. CircUBAP2 acted as a sponge to inhibit miR-370-3p expression. Mechanistically, miR-370-3p inhibition abolished the effects of circUBAP2 on proliferation, apoptosis, and invasion in TC cells. Taken together, CircUBAP2 knockdown impeded the proliferation and invasion and induced apoptosis in TC cells via sponging miR-370-3p.
Collapse
|
15
|
circRNA derived from CLSPN (circCLSPN) is an oncogene in human glioblastoma multiforme by regulating cell growth, migration and invasion via ceRNA pathway. J Biosci 2021. [DOI: 10.1007/s12038-021-00185-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
16
|
Lei W, Lin J, Liu F, Chen N. Long noncoding RNA GAS6 antisense RNA1 silencing attenuates the tumorigenesis of acute myeloid leukemia cells through targeting microRNA-370-3p/Tetraspanin3 axis. Clin Hemorheol Microcirc 2021; 78:69-81. [PMID: 33523043 DOI: 10.3233/ch-201039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Acute myeloid leukemia (AML) is a type of hematologic malignancy. This study was attempt to explore the effect of long noncoding RNA GAS6 antisense RNA1 (GAS6-AS1) on pediatric AML and the regulation mechanisms. METHODS GAS6-AS1, microRNA-370-3p (miR-370-3p), and Tetraspanin3 (TSPAN3) expression in bone marrow (BM) tissues and cells was determined by qRT-PCR. The correlation between GAS6-AS1 and clinicopathological features of pediatric patients with AML was assessed. In vitro, viability and migration and invasion of AML cells were evaluated via MTT and transwell assays, respectively. Interactions among GAS6-AS1, miR-370-3p, and TSPAN3 were revealed by dual-luciferase reporter assays. Western blot was applied to confirm the protein expression of TSPAN3. RESULTS GAS6-AS1 and TSPAN3 expression was elevated in BM tissues of pediatric patients with AML and AML cells, but miR-370-3p expression was reduced. GAS6-AS1 expression was positively related to French-American-British (FAB) classification in pediatric patients with AML. In vitro, GAS6-AS1 deficiency restrained the viability, migration, and invasion of AML cells. Additionally, GAS6-AS1 mediated miR-370-3p expression indeed and TSPAN3 was identified as a target of miR-370-3p. Furthermore, miR-370-3p overexpression repressed the protein expression of TSPAN3. The feedback experiments demonstrated that miR-370-3p inhibition or TSPAN3 overexpression mitigated the suppressive effect of sh-GAS6-AS1 on the tumorigenesis of AML cells. CONCLUSION GAS6-AS1 silencing restrained AML cell viability, migration, and invasion by targeting miR-370-3p/TSPAN3 axis, affording a novel therapeutic target for pediatric AML.
Collapse
Affiliation(s)
- Weijuan Lei
- Department of Pediatrics, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Juanjuan Lin
- Department of Pediatrics, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Fang Liu
- Department of Pediatrics, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Nina Chen
- Department of Pediatrics, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| |
Collapse
|
17
|
An Insight into the microRNAs Associated with Arteriovenous and Cavernous Malformations of the Brain. Cells 2021; 10:cells10061373. [PMID: 34199498 PMCID: PMC8227573 DOI: 10.3390/cells10061373] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Brain arteriovenous malformations (BAVMs) and cerebral cavernous malformations (CCMs) are rare developmental anomalies of the intracranial vasculature, with an irregular tendency to rupture, and as of yet incompletely deciphered pathophysiology. Because of their variety in location, morphology, and size, as well as unpredictable natural history, they represent a management challenge. MicroRNAs (miRNAs) are strands of non-coding RNA of around 20 nucleotides that are able to modulate the expression of target genes by binding completely or partially to their respective complementary sequences. Recent breakthroughs have been made on elucidating their contribution to BAVM and CCM occurrence, growth, and evolution; however, there are still countless gaps in our understanding of the mechanisms involved. Methods: We have searched the Medline (PubMed; PubMed Central) database for pertinent articles on miRNAs and their putative implications in BAVMs and CCMs. To this purpose, we employed various permutations of the terms and idioms: ‘arteriovenous malformation’, ‘AVM’, and ‘BAVM’, or ‘cavernous malformation’, ‘cavernoma’, and ‘cavernous angioma’ on the one hand; and ‘microRNA’, ‘miRNA’, and ‘miR’ on the other. Using cross-reference search; we then investigated additional articles concerning the individual miRNAs identified in other cerebral diseases. Results: Seven miRNAs were discovered to play a role in BAVMs, three of which were downregulated (miR-18a, miR-137, and miR-195*) and four upregulated (miR-7-5p, miR-199a-5p, miR-200b-3p, and let-7b-3p). Similarly, eight miRNAs were identified in CCM in humans and experimental animal models, two being upregulated (miR-27a and mmu-miR-3472a), and six downregulated (miR-125a, miR-361-5p, miR-370-3p, miR-181a-2-3p, miR-95-3p, and let-7b-3p). Conclusions: The following literature review endeavored to address the recent discoveries related to the various implications of miRNAs in the formation and growth of BAVMs and CCMs. Additionally, by presenting other cerebral pathologies correlated with these miRNAs, it aimed to emphasize the potential directions of upcoming research and biological therapies.
Collapse
|
18
|
Li X, Sun C, Chen J, Ma JF, Pan YH. ERK-CREB pathway is involved in HSPB8-mediated glioma cell growth and metastatic properties. Exp Mol Pathol 2021:104653. [PMID: 34043982 DOI: 10.1016/j.yexmp.2021.104653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/14/2021] [Accepted: 05/21/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To investigate the mechanism of HSPB8 (heat shock protein beta-8) in the growth and metastatic properties of glioma cells. METHODS HSPB8 expression in glioma tissue and cell was detected via Western blotting. Then, glioma U87 and U251 cell lines were divided into Mock group, Control siRNA group, HSPB8 siRNA-1 group and HSPB8 siRNA-2 group. Cell proliferation was detected using MTT assay, while its invasion, migration and apoptosis were determined by Transwell, wound-healing and flow cytometry, respectively. The expression of HSPB8 and ERK-CREB pathway-related molecules were also measured by Western blotting. Xenograft models were constructed on nude mice, and accordingly, the growth curve of subcutaneous xenograft was prepared. RESULTS In glioma tissues, HSPB8 expression was upregulated with the increasing grade of glioma. Besides, glioma cells in the HSPB8 siRNA-1 group and HSPB8 siRNA-2 group manifested the significant enhancement in apoptotic rates and reductions in its proliferation, migration and invasion compared to those in the Mock group, meanwhile, the expression of HSPB8, p-ERK1/2/ERK1/2 and p-CREB/CREB were downregulated. On the other hand, the tumor growth in the nude mice of Ad-HSPB8 shRNA-1 group and Ad-HSPB8 shRNA-2 group was retarded significantly, with an acute decrease in the tumor weight. CONCLUSION Silencing HSPB8 can inhibit the malignant features, while facilitate the apoptosis of glioma cells, with inactivation of ERK-CREB pathway.
Collapse
Affiliation(s)
- Xia Li
- Center for Diagnosis and Treatment of Neuro-oncology Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Cui Sun
- Center for Diagnosis and Treatment of Neuro-oncology Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Jing Chen
- Center for Diagnosis and Treatment of Neuro-oncology Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Ji-Fen Ma
- Center for Diagnosis and Treatment of Neuro-oncology Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Yi-Heng Pan
- Center for Diagnosis and Treatment of Neuro-oncology Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China.
| |
Collapse
|
19
|
Liu Y, Jiang K, Zhi T, Xu X. miR-720 is a key regulator of glioma migration and invasion by controlling TARSL2 expression. Hum Cell 2021; 34:1504-1516. [PMID: 34024034 DOI: 10.1007/s13577-021-00551-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 05/12/2021] [Indexed: 12/28/2022]
Abstract
Glioblastoma (GBM) is the most lethal type of primary brain tumor and is characterized by diffuse infiltrative growth. However, the mechanisms that control this phenotype remain largely unknown. Emerging evidence has demonstrated that the abnormal expression of microRNAs and their target genes are involved in the migration and invasion of glioma cells. In this study, we demonstrated that microRNA-720 (miR-720) was significantly upregulated in glioma tissues and cells. Functional experiments showed that overexpression of miR-720 promotes glioma migration and invasion, while downregulation of miR-720 inhibits glioma migration and invasion. Meanwhile, we found that threonyl-tRNA synthetase like-2 (TARSL2) was a direct and functional target of miR-720 in glioma. Reintroduction of TARSL2 into glioma cells repressed the invasion promoting function of miR-720, whereas downregulation of TARSL2 reversed the anti-invasion function of anti-miR-720. Furthermore, quantitative real-time polymerase chain reaction results showed that miR-720 was inversely correlated with TARSL2 expression in 40 GBM tissues. Finally, in vivo experiments showed that miR-720 promotes glioma growth and upregulates invasion-related genes in nude mice. Overall, our findings suggest increasing miR-720 enhances glioma migration and invasion through downregulation of TARSL2, which may provide novel insight into the treatment of glioma.
Collapse
Affiliation(s)
- Yinlong Liu
- Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215008, Jiangsu Province, China
| | - Kuan Jiang
- Department of Neurosurgery, Yixing People's Hospital, Yixing, 214200, Jiangsu Province, China
| | - Tongle Zhi
- Department of Neurosurgery, The First People's Hospital of Yancheng, The Fourth Affiliated Hospital of Nantong University, Yancheng, 224006, Jiangsu Province, China
| | - Xiupeng Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
20
|
Zhou S, Huang C, Wang W, Liu J. MiR-370-3p inhibits the development of human endometriosis by downregulating EDN1 expression in endometrial stromal cells. Cell Biol Int 2021; 45:1183-1190. [PMID: 33470475 DOI: 10.1002/cbin.11552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 01/06/2021] [Accepted: 01/17/2021] [Indexed: 12/24/2022]
Abstract
MiR-370-3p has been demonstrated to be downregulated in patients with endometriosis (EM). However, its role and molecular mechanisms in the progression of EM remain unclear. Real-time polymerase chain reaction was used to measure the expression of miR-370-3p and endothelin-1 (EDN1) in patients with or without EM. After miR-370-3p overexpression or knockdown in ectopic endometrial hEM15A cells, the changes in the proliferation, apoptosis, and migration and invasion capacities were detected by using cell counting kit-8, flow cytometry, and transwell methods. The interplay between miR-370-3p and EDN1 was confirmed by a luciferase reporter assay. Patients with EM showed adverse expression of EDN1 and miR-370-3p, especially in eutopic endometrium and ectopic endometrium. MiR-370-3p inhibited the proliferation, metastasis, and invasion capacities of hEM15A cells and promoted apoptosis. Investigation of its molecular mechanism revealed that miR-370-3p targeted EDN1 to influence the biological functions of hEM15A cells. MiR-370-3p represented as a therapeutic target for EM treatment.
Collapse
Affiliation(s)
- Shun Zhou
- Department of Interventional Radiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Chengyi Huang
- Department of Interventional Radiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Wei Wang
- Department of Pathology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Juan Liu
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
21
|
Cui X, Yu H, Yu T, Xiao D, Wang X. LncRNA MNX1-AS1 drives aggressive laryngeal squamous cell carcinoma progression and serves as a ceRNA to target FoxM1 by sponging microRNA-370. Aging (Albany NY) 2021; 13:9900-9910. [PMID: 33882027 PMCID: PMC8064170 DOI: 10.18632/aging.202746] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/09/2021] [Indexed: 12/21/2022]
Abstract
Long non-coding RNA (LncRNA) MNX1 antisense RNA 1(MNX1-AS1) is associated with the pathology of numerous cancers. But, the role and underlying pathways of MNX1-AS1 in the regulation of laryngeal squamous cell carcinoma (LSCC) is not known. We demonstrated remarkably elevated levels of MNX1-AS1 in the LSCC tissues, which was correlated with poor disease prognosis. Moreover, MNX1-AS1-silencing strongly suppressed LSCC cell proliferation, migration, and invasion. We also demonstrated that MNX1-AS1 sequesters that activity of miR-370, thereby releasing Forkhead Box ml (FoxM1) from the inhibitory actions of MNX1-AS1. Furthermore, the positive correlation of MNX1-AS1 and FoxM1 as well as the converse correlation between miR-370 and MNX1-AS1 (or FoxM1) were revealed in LSCC tissues using experiments. Based on rescue assays, FoxM1 overexpression or miR-370 downregulation partially recovered the inhibitory effect of MNX1-AS1 silencing on LSCC cells. Moreover, knockdown of MNX1-AS1 retarded tumor growth in nude mice model. In summary, these findings verified that MNX1-AS1 modulated LSCC progression by competitively binding with miR-370 to regulate FoxM1.
Collapse
Affiliation(s)
- Xiangyan Cui
- Department of Otolaryngology-Head and Neck Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Hong Yu
- Department of Otolaryngology-Head and Neck Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Tingting Yu
- Department of Otolaryngology-Head and Neck Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Dong Xiao
- Department of Otolaryngology-Head and Neck Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Xin Wang
- Department of Otolaryngology-Head and Neck Surgery, The First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
22
|
Li X, Qian Y, Tang K, Li Y, Tao R, Gong C, Huang L, Zou K, Liu L. Inhibition of lncRNA H19/miR-370-3p pathway mitigates neuronal apoptosis in an in vitro model of spinal cord injury (SCI). Transl Neurosci 2021; 12:103-113. [PMID: 33708438 PMCID: PMC7925972 DOI: 10.1515/tnsci-2021-0013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/05/2021] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is the most serious complication of spinal injury, often leading to severe dysfunction of the limbs below the injured segment. Conventional therapy approaches are becoming less and less effective, and gene therapy is a new research direction by now. METHODS The Sprague-Dawley rats were haphazardly assigned to two groups, namely sham group and SCI model group, and lncRNA H19 and miR-370-3p levels were investigated using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Correlation between lncRNA H19 and miR-370-3p was ascertained by luciferase report assay and RT-qPCR. After transfection with si-H19, miR-370-3p inhibitor, negative controls (NC), or both, primary spinal neurons were subjected to the simulation of lipopolysaccharide (LPS) for inducing in vitro model of SCI. Cell viability, apoptotic rate, caspase-3 activity, Bax and Bcl-2 protein, ROS generation, TNF-α, IL-1β, and IL-6 protein, as well as IκBα and p65 phosphorylation ratio were evaluated adopting 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), apoptosis, caspase-3 activity, ROS generation, and western blot assays, thereby searching for the specific action mechanism on LPS-induced spinal never injury. RESULTS SCI resulted in lncRNA H19 higher expression and miR-370-3p lower expression. LPS simulation raised a series of cellular biological changes, such as decreased viability, promoted apoptosis, generated ROS, and released inflammatory factors. lncRNA H19 inhibition reversed above LPS-induced changes. Besides, as the downstream target of lncRNA H19, miR-370-3p was oppositely regulated by lncRNA H19. The above biological changes induced by lncRNA H19 inhibition were reversed by miR-370-3p upregulation. Moreover, lncRNA H19 inhibition could block NF-κB pathway through miR-370-3p upregulation. CONCLUSION Inhibition of lncRNA H19/miR-370-3p mitigated spinal neuron apoptosis in an in vitro model of SCI. This provided the possibility for clinical use of gene therapy.
Collapse
Affiliation(s)
- Xin Li
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Yan Qian
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Kaihua Tang
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Yang Li
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Rui Tao
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Chunyan Gong
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Li Huang
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Kaiwen Zou
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Lindong Liu
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| |
Collapse
|
23
|
Abstract
The regulation of brain cytochrome P450 enzymes (CYPs) is different compared with respective hepatic enzymes. This may result from anatomical bases and physiological functions of the two organs. The brain is composed of a variety of functional structures built of different interconnected cell types endowed with specific receptors that receive various neuronal signals from other brain regions. Those signals activate transcription factors or alter functioning of enzyme proteins. Moreover, the blood-brain barrier (BBB) does not allow free penetration of all substances from the periphery into the brain. Differences in neurotransmitter signaling, availability to endogenous and exogenous active substances, and levels of transcription factors between neuronal and hepatic cells lead to differentiated expression and susceptibility to the regulation of CYP genes in the brain and liver. Herein, we briefly describe the CYP enzymes of CYP1-3 families, their distribution in the brain, and discuss brain-specific regulation of CYP genes. In parallel, a comparison to liver CYP regulation is presented. CYP enzymes play an essential role in maintaining the levels of bioactive molecules within normal ranges. These enzymes modulate the metabolism of endogenous neurochemicals, such as neurosteroids, dopamine, serotonin, melatonin, anandamide, and exogenous substances, including psychotropics, drugs of abuse, neurotoxins, and carcinogens. The role of these enzymes is not restricted to xenobiotic-induced neurotoxicity, but they are also involved in brain physiology. Therefore, it is crucial to recognize the function and regulation of CYP enzymes in the brain to build a foundation for future medicine and neuroprotection and for personalized treatment of brain diseases.
Collapse
Affiliation(s)
- Wojciech Kuban
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Władysława Anna Daniel
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
24
|
Guo M, Li S, Zhao X, Yuan Y, Zhang B, Guan Y. Knockdown of Circular RNA Hsa_circ_0000714 Can Regulate RAB17 by Sponging miR-370-3p to Reduce Paclitaxel Resistance of Ovarian Cancer Through CDK6/RB Pathway. Onco Targets Ther 2020; 13:13211-13224. [PMID: 33380810 PMCID: PMC7769200 DOI: 10.2147/ott.s285153] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Purpose Paclitaxel resistance in ovarian cancer has become an urgent clinical problem. This study investigated the regulatory effects of RAB17 on the non-coding RNA network of the paclitaxel-resistant ovarian cancer cell A2780/PTX. Methods Microarray analysis was used to identify differentially expressed genes in paclitaxel-resistant cell A2780/PTX compared to the parent paclitaxel-sensitive cell A2780. Quantitative real-time PCR and Western blot were used to measure the expression of related mRNAs and proteins. The CCK8 assay was used to determine cell survival ratios and drug resistance indices in ovarian cancer cells. The clone forming assay was used to analyze the cell clone proliferation. Flow cytometry was used to analyze the cell cycle. Dual-luciferase reporter gene assays evaluated the relationship between the genes. Results RAB17 is highly expressed in A2780/PTX cells. RAB17 knockdown increased the cell sensitivity to paclitaxel, inhibited proliferation, and caused cell cycle arrest in the G1 phase in A2780/PTX. Western blot confirmed that RAB17 influenced cell behavior by activating the CDK6/RB signaling pathway. Bioinformatics analyses identified RAB17 as a new target by the microRNA miR-370-3p, and the latter was predicted to interact with circular RNA hsa_circ_0000714. Hsa_circ_0000714 indeed acted as a miRNA sponge for miR-370-3p allowing its regulation of RAB17 expression. This regulation was accomplished through the CDK6/RB signaling pathway. Conclusion Hsa_circ_0000714 acts as a sponge for miR-370-3p, and regulates RAB17 expression through the CDK6/RB signaling pathway, which plays a role in the malignant progression of the paclitaxel-resistant ovarian cancer cell A2780/PTX.
Collapse
Affiliation(s)
- Min Guo
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, Liaoning 110122, People's Republic of China
| | - Shuo Li
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, Liaoning 110122, People's Republic of China
| | - Xiaoyun Zhao
- Department of Microbiology and Cell Biology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, People's Republic of China
| | - Ying Yuan
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, Liaoning 110122, People's Republic of China
| | - Bin Zhang
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, Liaoning 110122, People's Republic of China
| | - Yifu Guan
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, Liaoning 110122, People's Republic of China
| |
Collapse
|
25
|
Li Y, Zong J, Zhao C. lncRNA CTBP1-AS2 promotes proliferation and migration of glioma by modulating miR-370-3p-Wnt7a-mediated epithelial-mesenchymal transition. Biochem Cell Biol 2020; 98:661-668. [PMID: 33150795 DOI: 10.1139/bcb-2020-0065] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Glioma is one of the most common and aggressive malignant primary brain tumors, with a poor 5-year survival rate. The long noncoding RNA (lncRNA) CTBP1-AS2 has been shown to be correlated with the prognosis of cancer, but the role of CTBP1-AS2 in glioma and its concrete mechanism is fully unknown. The clinical data and tissues of glioma patients were analyzed. Cell viability and migration assays were performed. Western blotting and qRT-PCR were adopted for investigation of target protein expressions. Double luciferase assay was used to investigate the interaction between different elements. The lncRNA CTBP1-AS2 had increased expression profiles in tumor tissues, which is associated with poor prognosis. In detail, CTBP1-AS2 knockdown decreased proliferation and migration phenotypes in both U87-MG and LN229 cells. Moreover, CTBP1-AS2 knockdown suppressed the key epithelial-mesenchymal transition (EMT) markers by downregulating Wnt7a-mediated signaling. Furthermore, miR-370-3p functioned as a link that could be absorbed by CTBP1-AS2, thus regulating Wnt7a expression. Lastly, the CTBP1-AS2-miR-370-3p-Wnt7a axis modulated EMT in glioma cells in vitro and in vivo. This study provides new insights that a novel lncRNA, CTBP1-AS2, regulates EMT of glioma by modulating the miR-370-3p-Wnt7a axis.
Collapse
Affiliation(s)
- Yongfeng Li
- Department of Neurology, Sishui County People's Hospital, Jining, Shandong Province 273200, People's Republic of China
| | - Jin Zong
- Department of Neurosurgery, Liaocheng Hospital of Traditional Chinese Medicine, Liaocheng, Shandong Province 252004, People's Republic of China
| | - Cong Zhao
- Department of Oncology, Jining No. 1 People's Hospital, Jining, Shandong Province 272000, People's Republic of China
| |
Collapse
|
26
|
Li J, Chen J, Hu Z, Xu W. MicroRNA-1236-3p inhibits human osteosarcoma growth. Oncol Lett 2020; 20:367. [PMID: 33133267 PMCID: PMC7590435 DOI: 10.3892/ol.2020.12229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 09/18/2020] [Indexed: 12/16/2022] Open
Abstract
Osteosarcoma (OS) is a common bone tumor with high mortality worldwide. The long-term survival rate of patients with metastatic or recurrent disease is <20%. The present study explored the biological role of microRNA (miRNA/miR)-1236-3p in OS. miRNA and mRNA expression levels were measured via reverse transcription-quantitative PCR. Fluorescence in situ hybridization was performed to determine miR-1236-3p expression levels in clinical specimens. Protein expression was measured via western blotting. Immunohistochemical analysis was used to detect Wnt target gene expression in tumor tissues. The interaction between the Wnt3a 3′untranslated region and miR-1236-3p was assessed via dual-luciferase reporter assays. Cell cycle, Transwell, Cell Counting Kit-8 and wound healing assays were conducted to evaluate the function of the miR-1236-3p/Wnt3a axis. Human OS (HOS) cells stably transfected with vector or miR-1236-3p sponge were injected subcutaneously into nude mice to assess the role of miR-1236-3p in vivo. miR-1236-3p expression was downregulated in OS tissues compared with chondroma tissues, and miR-1236-3p overexpression inhibited OS cell migration and proliferation compared with the negative control group. Furthermore, in vivo xenograft assays displayed enhanced tumour growth rates in the miR-1236-3p sponge group compared with the vector control group. In the present study, the results indicated that miR-1236-3p inhibited OS progression and Wnt3a was identified as a target of miR-1236-3p.
Collapse
Affiliation(s)
- Jiarui Li
- Department of Urology Surgery, The First Affiliated Hospital of Nanchang University, Medical College of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Junxin Chen
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang 310016, P.R. China
| | - Zhijun Hu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang 310016, P.R. China
| | - Wenbin Xu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang 310016, P.R. China
| |
Collapse
|
27
|
Jin Y, Zhang M, Duan R, Yang J, Yang Y, Wang J, Jiang C, Yao B, Li L, Yuan H, Zha X, Ma C. Long noncoding RNA FGF14-AS2 inhibits breast cancer metastasis by regulating the miR-370-3p/FGF14 axis. Cell Death Discov 2020; 6:103. [PMID: 33083023 PMCID: PMC7548970 DOI: 10.1038/s41420-020-00334-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/20/2020] [Accepted: 09/03/2020] [Indexed: 12/18/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) have emerged as important regulators in cancers, including breast cancer. However, the overall biological roles and clinical significance of most lncRNAs are not fully understood. This study aimed to elucidate the potential role of a novel lncRNA FGF14-AS2 and the mechanisms underlying metastasis in breast cancer. The lncRNA FGF14-AS2 was significantly downregulated in breast cancer tissues; patients with lower FGF14-AS2 expression had advanced clinical stage. In vitro and in vivo assays of FGF14-AS2 alterations revealed a complex integrated phenotype affecting breast cancer cell migration, invasion, and tumor metastasis. Mechanistically, FGF14-AS2 functioned as a competing endogenous RNA of miR-370-3p, thereby leading to the activation of its coding counterpart, FGF14. Clinically, we observed increased miR-370-3p expression in breast cancer tissues, whereas FGF14 expression was decreased in breast cancer tissues compared to the adjacent normal breast tissues. FGF14-AS2 expression was significantly negatively correlated with miR-370-3p expression, and correlated positively to FGF14 expression. Collectively, our findings support a model in which the FGF14-AS2/miR-370-3p/FGF14 axis is a critical regulator in breast cancer metastasis, suggesting a new therapeutic direction in breast cancer.
Collapse
Affiliation(s)
- Yucui Jin
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Longmian Road 101, Nanjing, People's Republic of China.,Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, People's Republic of China
| | - Ming Zhang
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, People's Republic of China
| | - Rui Duan
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, People's Republic of China
| | - Jiashu Yang
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, People's Republic of China
| | - Ying Yang
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, People's Republic of China
| | - Jue Wang
- Division of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Chaojun Jiang
- Division of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Bing Yao
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, People's Republic of China
| | - Lingyun Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Longmian Road 101, Nanjing, People's Republic of China.,Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, People's Republic of China
| | - Hongyan Yuan
- Department of Oncology and Lombardi Comprehensive Cancer Center, Lombardi Comprehensive Cancer Center, Washington, DC USA
| | - Xiaoming Zha
- Division of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Changyan Ma
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Longmian Road 101, Nanjing, People's Republic of China.,Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, People's Republic of China
| |
Collapse
|
28
|
Liu L, Yan C, Tao S, Wang H. Circ_0058124 Aggravates the Progression of Papillary Thyroid Carcinoma by Activating LMO4 Expression via Targeting miR-370-3p. Cancer Manag Res 2020; 12:9459-9470. [PMID: 33061633 PMCID: PMC7534870 DOI: 10.2147/cmar.s271778] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 08/26/2020] [Indexed: 12/14/2022] Open
Abstract
Background Thyroid cancer is the most common malignant tumor in the endocrine system. Papillary thyroid carcinoma (PTC) accounts for the vast majority of cases in this cancer. Recently, the vital role of circular RNA (circRNA) has been acknowledged in various cancers, and this study aimed to investigate the role of circ_0058124 and related mechanism of its action in PTC. Materials and Methods The expression of circ_0058124, miR-370-3p and LIM domain only (LMO4) was detected by qRT-PCR in tissue samples (PTC tissues or normal tissues, n=20) and cell lines (non-cancer cell line, Nthy-ori 3–1, and PTC cell lines, IHH-4 and TPC-1). For functional analysis, cell proliferation was investigated using CCK-8 assay and colony formation assay. Cell migration and invasion were determined using transwell assay, and cell migration was also assessed by wound healing assay. Cell apoptosis was monitored by flow cytometry assay. For mechanism analysis, the interaction between miR-370-3p and circ_0058124 or LMO4 predicted by the bioinformatics analysis was validated by dual-luciferase reporter assay or RIP assay. The effect of circ_0058124 on tumor growth in vivo was identified by establishing the Xenograft model. Results The expression of circ_0058124 was enhanced in PTC tissues and cells. Circ_0058124 knockdown inhibited viability, colony formation, migration and invasion and promoted apoptosis of PTC cells. Besides, circ_0058124 knockdown also blocked tumor growth in vivo. miR-370-3p was a target of circ_0058124, and circ_0058124 regulated the expression of LMO4, a target of miR-370-3p, by targeting miR-370-3p. Rescue experiments presented that miR-370-3p inhibition reversed the inhibitory effects of circ_0058124 knockdown on PTC development, and LMO4 overexpression reversed the effect of miR-370-3p restoration on PTC development. Conclusion Circ_0058124 promoted the development of PTC by mediating the miR-370-3p/LMO4 axis, and circ_0058124, functioned as an oncogene in PTC, might be used as a promising biomarker for PTC diagnosis and treatment.
Collapse
Affiliation(s)
- Lei Liu
- Department of Otorhinolaryngology & Head and Neck Surgery, Tianjin Third Central Hospital, Tianjin, People's Republic of China
| | - Chaohui Yan
- Department of Otorhinolaryngology & Head and Neck Surgery, Tianjin Third Central Hospital, Tianjin, People's Republic of China
| | - Shudong Tao
- Department of Otorhinolaryngology & Head and Neck Surgery, Tianjin Third Central Hospital, Tianjin, People's Republic of China
| | - Hailing Wang
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, People's Republic of China
| |
Collapse
|
29
|
Chen W, Hong L, Hou C, Wang Y, Wang F, Zhang J. MicroRNA-585 inhibits human glioma cell proliferation by directly targeting MDM2. Cancer Cell Int 2020; 20:469. [PMID: 33005102 PMCID: PMC7523344 DOI: 10.1186/s12935-020-01528-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023] Open
Abstract
Background MicroRNAs (miRNAs) are important regulators for cancer cell proliferation. miR-585 has been shown to inhibit the proliferation of several types of cancer, however, little is known about its role in human glioma cells. Methods miR-585 levels in human glioma clinical samples and cell lines were examined by quantitative real-time PCR (qRT-PCR) analysis. Cell proliferation was measured by Cell Counting Kit-8 (CCK-8) and EdU incorporation assays in vitro. For in vivo investigations, U251 cells were intracranially inoculated in BALB/c nude mice and xenografted tumors were visualized by magnetic resonance imaging (MRI). Results miR-585 expression is downregulated in human glioma tissues and cell lines compared with non-cancerous counterparts. Additionally, miR-585 overexpression inhibits and its knockdown promotes human glioma cell proliferation in vitro. Moreover, miR-585 overexpression also inhibits the growth of glioma xenografts in vivo, suggesting that miR-585 may act as a tumor suppressor to inhibit the proliferation of human glioma. Furthermore, miR-585 directly targets and decreases the expression of oncoprotein murine double minute 2 (MDM2). More importantly, the restoration of MDM2 via enforced overexpression markedly rescues miR-585 inhibitory effect on human glioma cell proliferation, thus demonstrating that targeting MDM2 is a critical mechanism by which miR-585 inhibits human glioma cell proliferation. Conclusions Our study unveils the anti-proliferative role of miR-585 in human glioma cells, and also implicates its potential application in clinical therapy.
Collapse
Affiliation(s)
- Wangsheng Chen
- Department of Radiology, Hainan General Hospital/Hainan Hospital of Hainan Medical University, Haikou, 570311 China
| | - Lan Hong
- Department of Gynecology, Hainan General Hospital/Hainan Hospital of Hainan Medical University, Haikou, 570311 China
| | - Changlong Hou
- Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, No 150, JiMo Road, Pudong New Area, Shanghai, 200120 China
| | - Yibin Wang
- Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, No 150, JiMo Road, Pudong New Area, Shanghai, 200120 China
| | - Fei Wang
- Department of Radiology, Hainan General Hospital/Hainan Hospital of Hainan Medical University, Haikou, 570311 China
| | - Jianhua Zhang
- Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, No 150, JiMo Road, Pudong New Area, Shanghai, 200120 China
| |
Collapse
|
30
|
Wang Q, Jiang S, Wang W, Jiang H. Effects of baohuoside-I on epithelial-mesenchymal transition and metastasis in nasopharyngeal carcinoma. Hum Exp Toxicol 2020; 40:566-576. [PMID: 32945196 DOI: 10.1177/0960327120960765] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
To investigate the effect of baohuoside-I against nasopharyngeal carcinoma (NPC) and its underlying mechanism, baohuoside-I was employed to treat NPC cell lines CNE1 and CNE2 in vitro, followed by attachment and detachment assays to evalute the epithelial-mesenchymal transition (EMT) phenotype markers. Baohuoside-I was also administered to experimental mice to assess its effect on xenograft tumor growth and NPC cell metastasis. A microRNA (miRNA, miR) microarray was performed to screen for miRNA altered by baohuoside-I in NPC cells. Bioinformatic tools and luciferase activity assay was conducted to identify the downstream molecules mediating the anti-tumor property of baohuoside-I. Baohuoside-I inhibited EMT and metastasis and upregulated miR-370-3p in NPC cells, which was shown to directly recognize and inhibit expression of Hedgehog pathway component Smoothened (SMO). Baohuoside-I suppresses metastasis as well as EMT of NPC cells through targeting the Hedgehog pathway component SMO, and may serve as a potent anti-tumor agent in the clinical management of NPC.
Collapse
Affiliation(s)
- Q Wang
- Department of Otorhinolaryngology Head and Neck, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - S Jiang
- Department of Otorhinolaryngology Head and Neck, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - W Wang
- Department of Ophthalmology and Otolaryngology, Shandong Province Wendeng Orthopic and Traumatic Hospital, Weihai, Shandong, China
| | - H Jiang
- Department of Pathology, Shandong Province Wendeng Orthopic and Traumatic Hospital, Weihai, Shandong, China
| |
Collapse
|
31
|
Lulli V, Buccarelli M, Ilari R, Castellani G, De Dominicis C, Di Giamberardino A, D′Alessandris QG, Giannetti S, Martini M, Stumpo V, Boe A, De Luca G, Biffoni M, Marziali G, Pallini R, Ricci-Vitiani L. Mir-370-3p Impairs Glioblastoma Stem-Like Cell Malignancy Regulating a Complex Interplay between HMGA2/HIF1A and the Oncogenic Long Non-Coding RNA (lncRNA) NEAT1. Int J Mol Sci 2020; 21:ijms21103610. [PMID: 32443824 PMCID: PMC7279259 DOI: 10.3390/ijms21103610] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive and prevalent form of a human brain tumor in adults. Several data have demonstrated the implication of microRNAs (miRNAs) in tumorigenicity of GBM stem-like cells (GSCs). The regulatory functions of miRNAs in GSCs have emerged as potential therapeutic candidates for glioma treatment. The current study aimed at investigating the function of miR-370-3p in glioma progression, as aberrant expression of miR-370-3p, is involved in various human cancers, including glioma. Analyzing our collection of GBM samples and patient-derived GSC lines, we found the expression of miR-370-3p significantly downregulated compared to normal brain tissues and normal neural stem cells. Restoration of miR-370-3p expression in GSCs significantly decreased proliferation, migration, and clonogenic abilities of GSCs, in vitro, and tumor growth in vivo. Gene expression analysis performed on miR-370-3p transduced GSCs, identified several transcripts involved in Epithelial to Mesenchymal Transition (EMT), and Hypoxia signaling pathways. Among the genes downregulated by the restored expression of miR-370-3p, we found the EMT-inducer high-mobility group AT-hook 2 (HMGA2), the master transcriptional regulator of the adaptive response to hypoxia, Hypoxia-inducible factor (HIF)1A, and the long non-coding RNAs (lncRNAs) Nuclear Enriched Abundant Transcript (NEAT)1. NEAT1 acts as an oncogene in a series of human cancers including gliomas, where it is regulated by the Epidermal Growth Factor Receptor (EGFR) pathways, and contributes to tumor growth and invasion. Noteworthy, the expression levels of miR-370-3p and NEAT1 were inversely related in both GBM tumor specimens and GSCs, and a dual-luciferase reporter assay proved the direct binding between miR-370-3p and the lncRNAs NEAT1. Our results identify a critical role of miR-370-3p in the regulation of GBM development, indicating that miR-370-3p acts as a tumor-suppressor factor inhibiting glioma cell growth, migration and invasion by targeting the lncRNAs NEAT1, HMGA2, and HIF1A, thus, providing a potential candidate for GBM patient treatment.
Collapse
Affiliation(s)
- Valentina Lulli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.L.); (M.B.); (R.I.); (G.C.); (C.D.D.); (A.D.G.); (G.D.L.); (M.B.)
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.L.); (M.B.); (R.I.); (G.C.); (C.D.D.); (A.D.G.); (G.D.L.); (M.B.)
| | - Ramona Ilari
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.L.); (M.B.); (R.I.); (G.C.); (C.D.D.); (A.D.G.); (G.D.L.); (M.B.)
| | - Giorgia Castellani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.L.); (M.B.); (R.I.); (G.C.); (C.D.D.); (A.D.G.); (G.D.L.); (M.B.)
| | - Chiara De Dominicis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.L.); (M.B.); (R.I.); (G.C.); (C.D.D.); (A.D.G.); (G.D.L.); (M.B.)
| | - Alessandra Di Giamberardino
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.L.); (M.B.); (R.I.); (G.C.); (C.D.D.); (A.D.G.); (G.D.L.); (M.B.)
| | - Quintino Giorgio D′Alessandris
- Department of Neuroscience, Institute of Neurosurgery, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS; 00168 Rome, Italy; (Q.G.D.); (V.S.); (R.P.)
| | - Stefano Giannetti
- Department of Neuroscience, Institute of Anatomy, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Maurizio Martini
- Department of Health Science and Public Health, Institute of Pathology, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Vittorio Stumpo
- Department of Neuroscience, Institute of Neurosurgery, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS; 00168 Rome, Italy; (Q.G.D.); (V.S.); (R.P.)
| | - Alessandra Boe
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Gabriele De Luca
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.L.); (M.B.); (R.I.); (G.C.); (C.D.D.); (A.D.G.); (G.D.L.); (M.B.)
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.L.); (M.B.); (R.I.); (G.C.); (C.D.D.); (A.D.G.); (G.D.L.); (M.B.)
| | - Giovanna Marziali
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.L.); (M.B.); (R.I.); (G.C.); (C.D.D.); (A.D.G.); (G.D.L.); (M.B.)
- Correspondence: (G.M.); (L.R.-V.)
| | - Roberto Pallini
- Department of Neuroscience, Institute of Neurosurgery, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS; 00168 Rome, Italy; (Q.G.D.); (V.S.); (R.P.)
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.L.); (M.B.); (R.I.); (G.C.); (C.D.D.); (A.D.G.); (G.D.L.); (M.B.)
- Correspondence: (G.M.); (L.R.-V.)
| |
Collapse
|
32
|
Kirstein A, Schmid TE, Combs SE. The Role of miRNA for the Treatment of MGMT Unmethylated Glioblastoma Multiforme. Cancers (Basel) 2020; 12:cancers12051099. [PMID: 32354046 PMCID: PMC7281574 DOI: 10.3390/cancers12051099] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/23/2020] [Accepted: 04/26/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common high-grade intracranial tumor in adults. It is characterized by uncontrolled proliferation, diffuse infiltration due to high invasive and migratory capacities, as well as intense resistance to chemo- and radiotherapy. With a five-year survival of less than 3% and an average survival rate of 12 months after diagnosis, GBM has become a focus of current research to urgently develop new therapeutic approaches in order to prolong survival of GBM patients. The methylation status of the promoter region of the O6-methylguanine–DNA methyltransferase (MGMT) is nowadays routinely analyzed since a methylated promoter region is beneficial for an effective response to temozolomide-based chemotherapy. Furthermore, several miRNAs were identified regulating MGMT expression, apart from promoter methylation, by degrading MGMT mRNA before protein translation. These miRNAs could be a promising innovative treatment approach to enhance Temozolomide (TMZ) sensitivity in MGMT unmethylated patients and to increase progression-free survival as well as long-term survival. In this review, the relevant miRNAs are systematically reviewed.
Collapse
Affiliation(s)
- Anna Kirstein
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Thomas E. Schmid
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Stephanie E. Combs
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 81675 Munich, Germany
- Correspondence: ; Tel.: +49-89-4140-4501
| |
Collapse
|
33
|
Zou FW, Yang SZ, Li WY, Liu CY, Liu XH, Hu CH, Liu ZH, Xu S. circRNA_001275 upregulates Wnt7a expression by competitively sponging miR‑370‑3p to promote cisplatin resistance in esophageal cancer. Int J Oncol 2020; 57:151-160. [PMID: 32319613 PMCID: PMC7252462 DOI: 10.3892/ijo.2020.5050] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 02/05/2020] [Indexed: 12/16/2022] Open
Abstract
Circular RNAs (circRNAs) are aberrantly expressed in various tumors and are associated with tumorigenesis. The present study aimed to determine the role of circRNA_001275 in cisplatin‑resistant esophageal cancer. Three pairs of cisplatin‑resistant tissues and corresponding adjacent tissues were collected and subjected to circRNA chip analysis. Additionally, the effect of circRNA_001275 on cisplatin‑resistant cells was investigated. The relationship between circRNA_001275, microRNAs (miRs) and target genes were analyzed using luciferase assays, and validated via reverse transcription‑quantitative PCR (RT‑qPCR) and western blotting. The results showed that circRNA_001275 was significantly upregulated in cisplatin‑resistant esophageal cancer tissues and cells (P<0.05). Overexpression of circRNA_001275 promoted the proliferation and invasion, and decreased the apoptosis of cisplatin‑resistant cells. On the other hand, circRNA_001275 silencing inhibited cell proliferation and invasion, and promoted cell apoptosis (P<0.05). Dual‑luciferase reporter assays revealed that circRNA_001275 directly binds to miR‑370‑3p, and that Wnt family member 7A (Wnt7a) is targeted by miR‑370‑3p. RT‑qPCR and western blotting further demonstrated that circRNA_001275 serves as an miR‑370‑3p sponge to upregulate Wnt7a expression. In conclusion, the present study revealed that circRNA_001275 was upregulated in cisplatin‑resistant esophageal cancer and promoted cisplatin resistance by sponging miR‑370‑3p to upregulate Wnt7a expression. Therefore, circRNA_001275 may serve as a potential diagnostic biomarker and therapeutic target for patients with cisplatin‑resistant esophageal cancer.
Collapse
Affiliation(s)
- Fang-Wen Zou
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410000, P.R. China
| | - Shi-Ze Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 11000, P.R. China
| | - Wen-Ya Li
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 11000, P.R. China
| | - Chao-Yuan Liu
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410000, P.R. China
| | - Xu-Hong Liu
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410000, P.R. China
| | - Chun-Hong Hu
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410000, P.R. China
| | - Zheng-Hua Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 11000, P.R. China
| | - Shun Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 11000, P.R. China
| |
Collapse
|
34
|
Lin L, Wang D, Qu S, Zhao H, Lin Y. miR-370-3p Alleviates Ulcerative Colitis-Related Colorectal Cancer in Mice Through Inhibiting the Inflammatory Response and Epithelial-Mesenchymal Transition. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1127-1141. [PMID: 32214798 PMCID: PMC7078899 DOI: 10.2147/dddt.s238124] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/27/2020] [Indexed: 12/12/2022]
Abstract
Introduction Ulcerative colitis (UC) is a chronic and inflammatory bowel disease. UC-associated colorectal cancer (UC-CRC) is one of the most severe complications of long-standing UC. In the present study, we explored the effects of miR-370-3p on UC-CRC in vivo and investigated its underlying mechanisms in vivo and in vitro. Methods Azoxymethane (AOM) and dextran sodium sulfate (DSS) were used to induce UC-CRC in C57BL/6 mice. AOM/DSS-induced mice were treated with 5×108 pfu miR-370-3p overexpressing-adenovirus via tail-vein injection every two weeks. Results We found that miR-370-3p significantly improved the body weights and survival rates and inhibited the tumorigenesis of UC-CRC in AOM/DSS mice. Mechanically, miR-370-3p inhibited AOM/DSS-induced inflammatory response by decreasing tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) through targeting toll-like receptor 4 (TLR4), as demonstrated by down-regulation of TLR4, cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), and phosphorylated epidermal growth factor receptor (pEGFR). miR-370-3p decreased the expression of tumor-associated proteins, including p53, β-catenin, and ki67 in AOM/DSS-treated mice. Additionally, miR-370-3p remarkably inhibited epithelial-mesenchymal transition (EMT) via increasing E-cadherin expression and reducing N-cadherin and Vimentin expression in vivo. Further studies showed that miR-370-3p repressed proliferation and EMT of colon cancer cells in vitro. Moreover, we proved that miR-370-3p decreased the expression of tumor-associated proteins and reversed EMT by regulating β-catenin in colon cancer cells. Conclusion Taken together, miR-370-3p alleviated UC-CRC by inhibiting the inflammatory response and EMT in mice, which suggested miR-370-3p as a novel potential target for UC-CRC therapy.
Collapse
Affiliation(s)
- Lianjie Lin
- Department of Gastroenterology and Hepatology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Dongxu Wang
- Department of Gastroenterology and Hepatology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Suxuan Qu
- Department of Gastroenterology and Hepatology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Hong Zhao
- Department of Gastroenterology and Hepatology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.,Department of Gastroenterology, The Second Affiliated Hospital of Shenyang Medical College, Shenyang 110035, People's Republic of China
| | - Yan Lin
- Department of Gastroenterology and Hepatology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| |
Collapse
|
35
|
Zhang J, Mao F, Zhao G, Wang H, Yan X, Zhang Q. Long non-coding RNA SNHG16 promotes lipopolysaccharides-induced acute pneumonia in A549 cells via targeting miR-370-3p/IGF2 axis. Int Immunopharmacol 2019; 78:106065. [PMID: 31841752 DOI: 10.1016/j.intimp.2019.106065] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/23/2019] [Accepted: 11/17/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Pneumonia is an infectious lung inflammation in children with high mortality and morbidity rates. Small nucleolar RNA host gene 16 (SNHG16) has been verified to accelerate the progression of acute pneumonia. However, the role of SNHG16 in acute pneumonia has not yet been fully elucidated. The study was aimed to explore the regulatory mechanism of SNHG16 in LPS-induced acute pneumonia in A549 cells. METHODS The levels of SNHG16, miR-370-3p and IGF2 in serum samples and LPS-induced A549 cells were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The cell viability and apoptosis of A549 cells were examined by Cell Counting Kit-8 (CCK-8) assay and flow cytometer, respectively. The levels of interleukin 1β (IL-1β), interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) were determined by enzyme-linked immunosorbent assay (ELISA). The binding relationships among SNHG16, miR-370-3p and IGF2 were predicted by online database and verified by Dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. The protein levels of IGF2 were tested by Western blot. RESULTS SNHG16 and IGF2 were upregulated while miR-370-3p was downregulated in serum of acute pneumonia patients and LPS-induced A549 cells. SNHG16 regulated proliferation, apoptosis and inflammatory cytokines by inhibiting miR-370-3p in LPS-induced A549 cells. MiR-370-3p targeted IGF2 and inhibited LPS-induced inflammatory injury via IGF2 in A549 cells. Furthermore, SNHG16 was verified to promote IGF2 expression by sponging miR-370-3p in A549 cells. CONCLUSION SNHG16 impeded cell viability and promoted apoptosis, inflammatory injury by targeting IGF2 mediated by miR-370-3p in LPS-induced A549 cells.
Collapse
Affiliation(s)
- Ju Zhang
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Fengxia Mao
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Gai Zhao
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Haixia Wang
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Xiaomin Yan
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Qian Zhang
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China.
| |
Collapse
|
36
|
Li G, Zheng P, Wang H, Ai Y, Mao X. Long Non-Coding RNA TUG1 Modulates Proliferation, Migration, And Invasion Of Acute Myeloid Leukemia Cells Via Regulating miR-370-3p/MAPK1/ERK. Onco Targets Ther 2019; 12:10375-10388. [PMID: 31819520 PMCID: PMC6890183 DOI: 10.2147/ott.s217795] [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: 05/30/2019] [Accepted: 10/04/2019] [Indexed: 12/20/2022] Open
Abstract
Background Acute myeloid leukemia (AML) is the most common form of acute leukemia in adults. Long non-coding RNA taurine-upregulated gene 1 (lncRNA TUG1) has been discovered to participate in multiple cancers including AML. However, the detailed mechanism of TUG1 in AML remains obscure. Materials and methods AML cell lines HL-60 and Kasumi-1 were taken as cell models. TUG1 knockdown or overexpression cell lines were generated. Then, the biological influence of TUG1 on cancer cells was studied using CCK-8 assay, transwell assay and Western blot in vitro. Interaction between TUG1 and miR-370-3p was determined by bioinformatics analysis, RT-PCR, and luciferase assay. Western blot, RT-PCR, and luciferase assay were carried out to validate the interaction between miR-370-3p and its target gene Mitogen-Activated Protein Kinase 1 (MAPK1). Results Knockdown of TUG1 markedly reduced viability and metastasis of AML cells, while its overexpression had the opposite effect. MAPK1 was verified as a target gene of miR-370-3p. TUG1 could reduce the level of functional miR-370-3p, facilitate MAPK1 expression, and in turn activate ERK1/2 signaling. Conclusion TUG1 could modulate malignant phenotypes of AML cells via miR-370-3p/MAPK1/ERK signaling. Our study would help to clarify the mechanism of AML tumorigenesis and progression.
Collapse
Affiliation(s)
- Gang Li
- Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, People's Republic of China
| | - Peiming Zheng
- Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, People's Republic of China
| | - Huiling Wang
- Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, People's Republic of China
| | - Yushu Ai
- Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, People's Republic of China
| | - Xiaohuan Mao
- Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, People's Republic of China
| |
Collapse
|
37
|
Wu F, Zhou J. CircAGFG1 promotes cervical cancer progression via miR-370-3p/RAF1 signaling. BMC Cancer 2019; 19:1067. [PMID: 31703640 PMCID: PMC6842182 DOI: 10.1186/s12885-019-6269-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/15/2019] [Indexed: 01/10/2023] Open
Abstract
Background In past decades, circular RNAs (circRNAs) have achieved increasing attention because of its regulatory role in different kinds of cancers. However, how circAGFG1 regulates cervical cancer (CC) is still largely undiscovered. This study aims to evaluate the role of a novel circRNAs and related molecular mechanism in CC cells. Methods High or low level of circAGFG1 was detected in CC cells or normal cell line with qRT-PCR. The proliferative and migratory abilities of CC cells were assessed with loss-of function assays. The downstream miRNA and mRNA of circAGFG1 were searched out and proved by using bioinformatics analysis and mechanism experiments. Recue assays were designed to confirm the role of circAGFG1/miR-370-3p/RAF1 axis in CC cell activities. Results The levels of circAGFG1 was abundant in CC cells in comparison with normal cervical cell End1/E6E7. The inhibitory effect of decreased circAGFG1 level on the proliferative and migratory abilities of CC cells was assessed. CircAGFG1 and miR-370-3p were localized in the cytoplasm and they can interact with each other. Moreover, miR-370-3p was downregulated in CC cells. We also determined the negative effect of miR-370-3p on RAF1. CircAGFG1 could promote RAF1 expression by absorbing miR-370-3p, thereby activating RAF/MEK/ERK pathway. circAGFG1 promoted proliferation and migration of CC cells via enhancing the activity of RAF/MEK/ERK pathway by sponging miR-370-3p and further regulating RAF1. Conclusion The results of this study provided new evidence that circAGFG1 acted as a vital regulator in cervical cancer proliferation and migration, giving great promise to apply it as a potential biomarker for diagnosis and therapy in CC treatment.
Collapse
Affiliation(s)
- Fengqin Wu
- Department of Gynecology, Shangluo Central Hospital, Shangluo City, 726000, Shaanxi Province, China
| | - Jingjing Zhou
- Department of Gynaecology, Ankang Hospital of Traditional Chinese Medicine, No.47, Bashan Road(east), Hanbin District, Ankang City, 725000, Shaanxi Province, China.
| |
Collapse
|
38
|
Chen J, Liu G, Wu Y, Ma J, Wu H, Xie Z, Chen S, Yang Y, Wang S, Shen P, Fang Y, Fan S, Shen S, Fang X. CircMYO10 promotes osteosarcoma progression by regulating miR-370-3p/RUVBL1 axis to enhance the transcriptional activity of β-catenin/LEF1 complex via effects on chromatin remodeling. Mol Cancer 2019; 18:150. [PMID: 31665067 PMCID: PMC6819556 DOI: 10.1186/s12943-019-1076-1] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022] Open
Abstract
Background CircMYO10 is a circular RNA generated by back-splicing of gene MYO10 and is upregulated in osteosarcoma cell lines, but its functional role in osteosarcoma is still unknown. This study aimed to clarify the mechanism of circMYO10 in osteosarcoma. Methods CircMYO10 expression in 10 paired osteosarcoma and chondroma tissues was assessed by quantitative reverse transcription polymerase chain reaction (PCR). The function of circMYO10/miR-370-3p/RUVBL1 axis was assessed regarding two key characteristics: proliferation and endothelial–mesenchymal transition (EMT). Bioinformatics analysis, western blotting, real-time PCR, fluorescence in situ hybridization, immunoprecipitation, RNA pull-down assays, luciferase reporter assays, chromatin immunoprecipitation, and rescue experiments were used to evaluate the mechanism. Stably transfected MG63 cells were injected via tail vein or subcutaneously into nude mice to assess the role of circMYO10 in vivo. Results CircMYO10 was significantly upregulated, while miR-370-3p was downregulated, in osteosarcoma cell lines and human osteosarcoma samples. Silencing circMYO10 inhibited cell proliferation and EMT in vivo and in vitro. Mechanistic investigations revealed that miR-370-3p targets RUVBL1 directly, and inhibits the interaction between RUVBL1 and β-catenin/LEF1 complex while circMYO10 showed a contrary effect via the inhibition of miR-370-3p. RUVBL1 was found to be complexed with chromatin remodeling and histone-modifying factor TIP60, and lymphoid enhancer factor-1 (LEF1) to promote histone H4K16 acetylation (H4K16Ac) in the vicinity of the promoter region of gene C-myc. Chromatin immunoprecipitation methods showed that miR-370-3p sponge promotes H4K16Ac in the indicated region, which is partially abrogated by RUVBL1 small hairpin RNA (shRNA) while circMYO10 showed a contrary result via the inhibition of miR-370-3p. Either miR-370-3p sponge or ShRUVBL1 attenuated circMYO10-induced phenotypes in osteosarcoma cell lines. MiR-370-3p inhibition abrogated the inhibition of proliferation, EMT of osteosarcoma cells in vitro and in vivo seen upon circMYO10 suppression via Wnt/β-catenin signaling. Conclusions CircMYO10 promotes osteosarcoma progression by regulating miR-370-3p/RUVBL1 axis to promote chromatin remodeling and thus enhances the transcriptional activity of β-catenin/LEF1 complex, which indicates that circMYO10 may be a potential therapeutic target for osteosarcoma treatment.
Collapse
Affiliation(s)
- Junxin Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Gang Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Yizheng Wu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Jianjun Ma
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Hongfei Wu
- Department of Spinal Surgery, Orthopaedic Medical Center, Hospital of Zhejiang Armed Police Corps, Jiaxing, Zhejiang Province, China
| | - Ziang Xie
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Shuai Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Yute Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Shengyu Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Panyang Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Yifan Fang
- Hangzhou Foreign Language School, Hangzhou, Zhejiang Province, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Shuying Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China.
| | - Xiangqian Fang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China.
| |
Collapse
|
39
|
Cai F, Dai C, Chen S, Wu Q, Liu X, Hong Y, Wang Z, Li L, Yan W, Wang R, Zhang J. CXCL12-regulated miR-370-3p functions as a tumor suppressor gene by targeting HMGA2 in nonfunctional pituitary adenomas. Mol Cell Endocrinol 2019; 488:25-35. [PMID: 30853598 DOI: 10.1016/j.mce.2019.02.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/26/2019] [Accepted: 02/22/2019] [Indexed: 12/27/2022]
Abstract
Silencing of noncoding genes within the imprinted DLK1-MEG3 locus is exclusive to human nonfunctional pituitary adenomas (NFPAs), but the exact mechanism is still unclear. This study was designed to demonstrate the impact of CXCL12 on the expression of miRNAs within this locus and phenotypic alterations of NFPAs. Human NFPA samples were collected for screening differentially expressed miRNAs by CXCL12. Target mRNAs of the miRNAs were predicted and verified in vitro. Tumor phenotypic alterations were also tested. Another 51 NFPA samples were enrolled to examine the correlation and clinical features. The expression of miR-370 was decreased by CXCL12 treatment in NFPAs. miR-370-3p was predicted and verified to target HMGA2 as a tumor suppressor gene. Overexpression of HMGA2 inhibited its antitumor function. miR-370-3p was downregulated and HMGA2 was upregulated significantly in High grade NFPAs. In conclusion, the CXCL12/miR-370-3p/HMGA2 signaling pathway is involved in tumor growth and invasiveness of NFPAs.
Collapse
Affiliation(s)
- Feng Cai
- The Dept. of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, The City of Hangzhou, Zhejiang Province, PR China
| | - Congxin Dai
- The Dept. of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Shasha Chen
- Zhejiang Provincial Key Lab of Geriatrics, Dept. of Geriatrics, Zhejiang Hospital, Hangzhou, Zhejiang Province, PR China
| | - Qun Wu
- The Dept. of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, The City of Hangzhou, Zhejiang Province, PR China
| | - Xiaohai Liu
- The Dept. of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Yuan Hong
- The Dept. of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, The City of Hangzhou, Zhejiang Province, PR China
| | - Zhen Wang
- The Dept. of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, The City of Hangzhou, Zhejiang Province, PR China
| | - Li Li
- The Dept. of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, The City of Hangzhou, Zhejiang Province, PR China
| | - Wei Yan
- The Dept. of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, The City of Hangzhou, Zhejiang Province, PR China
| | - Renzhi Wang
- The Dept. of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| | - Jianmin Zhang
- The Dept. of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, The City of Hangzhou, Zhejiang Province, PR China.
| |
Collapse
|
40
|
Hu Z, Mamillapalli R, Taylor HS. Increased circulating miR-370-3p regulates steroidogenic factor 1 in endometriosis. Am J Physiol Endocrinol Metab 2019; 316:E373-E382. [PMID: 30576245 PMCID: PMC6459299 DOI: 10.1152/ajpendo.00244.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Endometriosis is a gynecologic disease common among reproductive-aged women caused by the growth of endometrial tissue outside the uterus. Altered expression of numerous genes and microRNAs has been reported in endometriosis. Steroidogenic factor 1 (SF-1), an essential transcriptional regulator of multiple genes involved in estrogen biosynthesis, is aberrantly increased and plays an important role in the pathogenesis of endometriosis. Here, we show the expression of SF-1 in endometriosis is regulated by miR-370-3p. Sera and tissue were collected from 20 women surgically diagnosed with endometriosis and 26 women without endometriosis. We found that miR-370-3p levels were decreased in the serum of patients with endometriosis while SF-1 mRNA levels were inversely upregulated in endometriotic lesions compared with respective controls. Transfection of primary endometriotic cells with miR-370-3p mimic or inhibitor resulted in the altered expression of SF-1 and SF-1 downstream target genes steroidogenic acute regulatory protein (StAR) and CYP19A1. Overexpression of miR-370-3p inhibited cell proliferation and induced apoptosis in endometriotic cells. This study reveals that miR-370-3p functions as a negative regulator of SF-1 and cell proliferation in endometriotic cells. We suggest a novel therapeutic strategy for controlling SF-1 in endometriosis.
Collapse
Affiliation(s)
- Zhuoying Hu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine , New Haven, Connecticut
| | - Ramanaiah Mamillapalli
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine , New Haven, Connecticut
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine , New Haven, Connecticut
| |
Collapse
|
41
|
Radke DI, Ling Q, Häsler R, Alp G, Ungefroren H, Trauzold A. Downregulation of TRAIL-Receptor 1 Increases TGFβ Type II Receptor Expression and TGFβ Signalling Via MicroRNA-370-3p in Pancreatic Cancer Cells. Cancers (Basel) 2018; 10:399. [PMID: 30366420 PMCID: PMC6267290 DOI: 10.3390/cancers10110399] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/09/2018] [Indexed: 12/28/2022] Open
Abstract
The accumulation of perturbations in signalling pathways resulting in an apoptosis-insensitive phenotype is largely responsible for the desperate prognosis of patients with pancreatic ductal adenocarcinoma (PDAC). Accumulating evidence suggests that the death receptors TRAIL-R1 and TRAIL-R2 play important roles in PDAC biology by acting as either tumour suppressors through induction of cell death or tumour promoters through induction of pro-inflammatory signalling, invasion and metastasis. TRAIL-R2 can also associate with nuclear proteins and alter the maturation of micro RNAs (miRs). By genome-wide miR profiling and quantitative PCR analyses we now demonstrate that knockdown of TRAIL-R1 in PDAC cells decreased the level of mature miR-370 and led to an increased abundance of the type II receptor for transforming growth factor β (TGFβ). Transfection of cells with an artificial miR-370-3p decreased the levels of TGFβ-RII. We further show that transient expression of the miR-370 mimic decreased TGFβ1-induced expression of SERPINE1 encoding plasminogen activator-inhibitor 1 and partially relieved TGFβ1-induced growth inhibition. Moreover, stable TRAIL-R1 knockdown in Colo357 cells increased TGFβ1-induced SERPINE1 expression and this effect was partially reversed by transient expression of the miR-370 mimic. Finally, after transient knockdown of TRAIL-R1 in Panc1 cells there was a tendency towards enhanced activation of Smad2 and JNK1/2 signalling by exogenous TGFβ1. Taken together, our study reveals that TRAIL-R1 through regulation of miR-370 can decrease the sensitivity of PDAC cells to TGFβ and therefore represents a potential tumour suppressor in late-stage PDAC.
Collapse
Affiliation(s)
- David I Radke
- Institute for Experimental Cancer Research, University of Kiel, D-24105 Kiel, Germany.
| | - Qi Ling
- Institute for Experimental Cancer Research, University of Kiel, D-24105 Kiel, Germany.
- Department of Surgery, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 31000, China.
| | - Robert Häsler
- Institute of Clinical Molecular Biology, University of Kiel, D-24105 Kiel, Germany.
| | - Gökhan Alp
- Institute for Experimental Cancer Research, University of Kiel, D-24105 Kiel, Germany.
| | - Hendrik Ungefroren
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany.
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany.
| | - Anna Trauzold
- Institute for Experimental Cancer Research, University of Kiel, D-24105 Kiel, Germany.
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany.
| |
Collapse
|
42
|
Zhang J, Zhang J, Qiu W, Zhang J, Li Y, Kong E, Lu A, Xu J, Lu X. MicroRNA-1231 exerts a tumor suppressor role through regulating the EGFR/PI3K/AKT axis in glioma. J Neurooncol 2018; 139:547-562. [PMID: 29774498 PMCID: PMC6132976 DOI: 10.1007/s11060-018-2903-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/09/2018] [Indexed: 11/26/2022]
Abstract
PURPOSE MicroRNAs (miRNAs) have been shown to be involved in the initiation and progression of glioma. However, the underlying molecular mechanisms are still unclear. METHODS We performed microarray analysis to evaluate miRNA expression levels in 158 glioma tissue samples, and examined miR-1231 levels in glioma samples and healthy brain tissues using qRT-PCR. In vitro analyses were performed using miR-1231 mimics, inhibitors, and siRNA targeting EGFR. We used flow cytometry, CCK-8 assays, and colony formation assays to examine glioma proliferation and cell cycle analysis. A dual luciferase reporter assay was performed to examine miR-1231 regulation of EGFR, and the effect of upregulated miR-1231 was investigated in a subcutaneous GBM model. RESULTS We found that miR-1231 expression was decreased in human glioma tissues and negatively correlated with EGFR levels. Moreover, the downregulation of miR-1231 negatively correlated with the clinical stage of human glioma patients. miR-1231 overexpression dramatically downregulated glioma cell proliferation, and suppressed tumor growth in a nude mouse model. Bioinformatics prediction and a luciferase assay confirmed EGFR as a direct target of miR-1231. EGFR overexpression abrogated the suppressive effect of miR-1231 on the PI3K/AKT pathway and G1 arrest. CONCLUSIONS Taken together, these results demonstrated that EGFR is a direct target of miR-1231. Our findings suggest that the miR-1231/EGFR axis may be a helpful future diagnostic target for malignant glioma.
Collapse
Affiliation(s)
- Jiale Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Jie Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Wenjin Qiu
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, 28 Guiyi Street Road, Guiyang, 550004, Guizhou Province, People's Republic of China
| | - Jian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Yangyang Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Enjun Kong
- Department of Emergency, Danyang People's Hospital of Jiangsu Province, 2 Xinmin West Road, Danyang, 212300, Jiangsu Province, People's Republic of China
| | - Ailin Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Jia Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Xiaoming Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China.
| |
Collapse
|
43
|
Huang X, Zhu H, Gao Z, Li J, Zhuang J, Dong Y, Shen B, Li M, Zhou H, Guo H, Huang R, Yan J. Wnt7a activates canonical Wnt signaling, promotes bladder cancer cell invasion, and is suppressed by miR-370-3p. J Biol Chem 2018; 293:6693-6706. [PMID: 29549123 DOI: 10.1074/jbc.ra118.001689] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/12/2018] [Indexed: 12/13/2022] Open
Abstract
Once urinary bladder cancer (UBC) develops into muscle-invasive bladder cancer, its mortality rate increases dramatically. However, the molecular mechanisms of UBC invasion and metastasis remain largely unknown. Herein, using 5637 UBC cells, we generated two sublines with low (5637 NMI) and high (5637 HMI) invasive capabilities. Mass spectrum analyses revealed that the Wnt family protein Wnt7a is more highly expressed in 5637 HMI cells than in 5637 NMI cells. We also found that increased Wnt7a expression is associated with UBC metastasis and predicted worse clinical outcome in UBC patients. Wnt7a depletion in 5637 HMI and T24 cells reduced UBC cell invasion and decreased levels of active β-catenin and its downstream target genes involved in the epithelial-to-mesenchymal transition (EMT) and extracellular matrix (ECM) degradation. Consistently, treating 5637 NMI and J82 cells with recombinant Wnt7a induced cell invasion, EMT, and expression of ECM degradation-associated genes. Moreover, TOP/FOPflash luciferase assays indicated that Wnt7a activated canonical β-catenin signaling in UBC cells, and increased Wnt7a expression was associated with nuclear β-catenin in UBC samples. Wnt7a ablation suppressed matrix metalloproteinase 10 (MMP10) expression, and Wnt7a overexpression increased MMP10 promoter activity through two TCF/LEF promoter sites, confirming that Wnt7a-mediated MMP10 activation is mediated by the canonical Wnt/β-catenin pathway. Of note, the microRNA miR-370-3p directly repressed Wnt7a expression and thereby suppressed UBC cell invasion, which was partially restored by Wnt7a overexpression. Our results have identified an miR-370-3p/Wnt7a axis that controls UBC invasion through canonical Wnt/β-catenin signaling, which may offer prognostic and therapeutic opportunities.
Collapse
Affiliation(s)
- Xiaojing Huang
- From the MOE Key Laboratory of Model Animals for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Nanjing University, Nanjing, Jiangsu 210061
| | - Hongwen Zhu
- the Shanghai Institute of Materia Medica and
| | - Zemin Gao
- From the MOE Key Laboratory of Model Animals for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Nanjing University, Nanjing, Jiangsu 210061
| | - Junzun Li
- From the MOE Key Laboratory of Model Animals for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Nanjing University, Nanjing, Jiangsu 210061
| | - Junlong Zhuang
- the Department of Urology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu 210008
| | - Yu Dong
- the Shanghai Institute of Materia Medica and.,the Shanghai University, Shanghai 200444
| | - Bing Shen
- the Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | - Meiqian Li
- From the MOE Key Laboratory of Model Animals for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Nanjing University, Nanjing, Jiangsu 210061
| | - Hu Zhou
- the Shanghai Institute of Materia Medica and
| | - Hongqian Guo
- the Department of Urology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu 210008,
| | - Ruimin Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203,
| | - Jun Yan
- From the MOE Key Laboratory of Model Animals for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Nanjing University, Nanjing, Jiangsu 210061, .,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203
| |
Collapse
|
44
|
Chen M, Liu YY, Zheng MQ, Wang XL, Gao XH, Chen L, Zhang GM. microRNA-544 promoted human osteosarcoma cell proliferation by downregulating AXIN2 expression. Oncol Lett 2018; 15:7076-7082. [PMID: 29725431 PMCID: PMC5920246 DOI: 10.3892/ol.2018.8218] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 01/12/2018] [Indexed: 12/15/2022] Open
Abstract
microRNAs (miRNAs) perform various oncogenic or tumor suppressor functions in carcinogenesis. Currently, the underlying mechanisms of miRNAs in osteosarcoma (OS) are poorly understood. In the present study, it is demonstrated that expression of miR-544 was markedly upregulated in OS cells and clinical tissues. Furthermore, overexpression of miR-544 enhanced OS cell proliferation in vitro. Bioinformatics analysis indicated that miR-544 may target the 3′-untranslated region of axis formation inhibitor 2, which was validated using luciferase reporter gene assays. The present study demonstrated a vital role for miR-544 in promoting OS cell proliferation, indicating that it may represent a novel prognostic factor or therapeutic target for OS.
Collapse
Affiliation(s)
- Ming Chen
- Department of Orthopedics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Yong-Yi Liu
- Department of Orthopedics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Min-Qing Zheng
- Department of Orthopedics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Xin-Liang Wang
- Department of Orthopedics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Xing-Hua Gao
- Department of Orthopedics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Lin Chen
- Department of Orthopedics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Guang-Ming Zhang
- Department of Orthopedics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| |
Collapse
|
45
|
Chen S, Zhang Y, Wang H, Zeng YY, Li Z, Li ML, Li FF, You J, Zhang ZM, Tzeng CM. WW domain-binding protein 2 acts as an oncogene by modulating the activity of the glycolytic enzyme ENO1 in glioma. Cell Death Dis 2018; 9:347. [PMID: 29497031 PMCID: PMC5832848 DOI: 10.1038/s41419-018-0376-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 01/09/2023]
Abstract
WW domain-binding protein 2 (WBP2) has been demonstrated as oncogenic in breast cancer. Many studies have revealed the WBP2 gene as a high-risk gene for leukoariaosis and cerebral white matter lesions is important in the pathologic stage of glioma development. This study aimed to illustrate the underlying mechanism by which WBP2 regulates the process of glioma development. The expression pattern of WBP2 in several tumor cells was determined, clarifying the carcinogenic action of WBP2 in glioma cells. Overexpression of WBP2 in glioma cells promoted cell proliferation and migration, and the number of S-phase cells, whereas the depletion of WBP2 by RNAi-mediated knockdown restrained cell growth and cell cycle progression. Upregulation of WBP2 significantly enhanced the tumorigenic ability of U251 cells in vivo. MS/GST pulldown assay identified α-enolase (ENO1) and Homer protein homolog 3 (Homer3) as novel potent interaction partners of WBP2. Knockdown of ENO1 or Homer3 allowed cell growth and migration to return to normal levels. Furthermore, in vitro and in vivo experiments indicated that the oncogenic role of WBP2 in glioma was through modulating ENO1 and glycolysis activity via the ENO1-PI3K/Akt signaling pathway. Collectively, these results reveal that WBP2 plays a vital role in the occurrence and development of glioma, indicating a target gene for glioblastoma treatment.
Collapse
Affiliation(s)
- Shuai Chen
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, 361005, P.R. China.,Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361005, P.R. China.,Key Laboratory for Cancer T-Cell Therapeutics and Clinical Translation (CTCTCT), Xiamen, Fujian, 361005, P.R. China
| | - Ya Zhang
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, 361005, P.R. China.,Key Laboratory for Cancer T-Cell Therapeutics and Clinical Translation (CTCTCT), Xiamen, Fujian, 361005, P.R. China
| | - Han Wang
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, 361005, P.R. China.,Key Laboratory for Cancer T-Cell Therapeutics and Clinical Translation (CTCTCT), Xiamen, Fujian, 361005, P.R. China
| | - Yu-Ying Zeng
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361005, P.R. China
| | - Zhi Li
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, 361005, P.R. China.,Key Laboratory for Cancer T-Cell Therapeutics and Clinical Translation (CTCTCT), Xiamen, Fujian, 361005, P.R. China
| | - Ming-Li Li
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, 361005, P.R. China.,Key Laboratory for Cancer T-Cell Therapeutics and Clinical Translation (CTCTCT), Xiamen, Fujian, 361005, P.R. China
| | - Fang-Fang Li
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, 361005, P.R. China.,INNOVA Cell Theranostics/Clinics and TRANSLA Health Group, Yangzhou, Jiangsu, P.R. China.,Key Laboratory for Cancer T-Cell Therapeutics and Clinical Translation (CTCTCT), Xiamen, Fujian, 361005, P.R. China
| | - Jun You
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361005, P.R. China
| | - Zhi-Ming Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361005, P.R. China. .,Teaching Hospital of Fujian Medical University, Fuzhou, Fujian, 350004, P.R. China.
| | - Chi-Meng Tzeng
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, 361005, P.R. China. .,INNOVA Cell Theranostics/Clinics and TRANSLA Health Group, Yangzhou, Jiangsu, P.R. China. .,Key Laboratory for Cancer T-Cell Therapeutics and Clinical Translation (CTCTCT), Xiamen, Fujian, 361005, P.R. China. .,College of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China. .,Jiansu Provincial Institute of Translation Medicine and Women-Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
46
|
Li J, Huang Y, Deng X, Luo M, Wang X, Hu H, Liu C, Zhong M. Long noncoding RNA H19 promotes transforming growth factor-β-induced epithelial-mesenchymal transition by acting as a competing endogenous RNA of miR-370-3p in ovarian cancer cells. Onco Targets Ther 2018; 11:427-440. [PMID: 29403287 PMCID: PMC5783024 DOI: 10.2147/ott.s149908] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ovarian cancer is a gynecological malignant tumor with a high mortality rate among women, owing to metastatic progression and recurrence. Acquisition of invasiveness is accompanied by the loss of epithelial features and a gain of a mesenchymal phenotype, a process known as epithelial–mesenchymal transition (EMT). Transforming growth factor-β (TGF-β) has been implicated in the regulation of EMT. In the present study, we aimed to investigate the role of long noncoding RNA H19 and microRNA-370 (miR-370-3p) in TGF-β-induced EMT. Ovarian cancer cell lines SKOV-3 and OVCAR3 were incubated with different concentrations of TGF-β, and the results showed that TGF-β treatment upregulated H19 and downregulated miR-370-3p. In addition, an H19 knockdown or miR-370-3p overexpression suppressed TGF-β-induced EMT, while H19 overexpression or a miR-370-3p knockdown promoted TGF-β-induced EMT. Mechanistically, H19 could directly bind to miR-370-3p and effectively act as its competing endogenous RNA. Furthermore, we demonstrated that this activity of H19 was involved in its promotion of TGF-β-induced EMT. Thus, our results may provide novel insights into the process of TGF-β-induced EMT.
Collapse
Affiliation(s)
- Jing Li
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - YingYing Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - XiaoJun Deng
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - ManLing Luo
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - XueFei Wang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - HaiYan Hu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - CiDi Liu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Mei Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
47
|
Molecular Determinants of Malignant Brain Cancers: From Intracellular Alterations to Invasion Mediated by Extracellular Vesicles. Int J Mol Sci 2017; 18:ijms18122774. [PMID: 29261132 PMCID: PMC5751372 DOI: 10.3390/ijms18122774] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/29/2017] [Accepted: 12/19/2017] [Indexed: 12/15/2022] Open
Abstract
Malignant glioma cells invade the surrounding brain parenchyma, by migrating along the blood vessels, thus promoting cancer growth. The biological bases of these activities are grounded in profound alterations of the metabolism and the structural organization of the cells, which consequently acquire the ability to modify the surrounding microenvironment, by altering the extracellular matrix and affecting the properties of the other cells present in the brain, such as normal glial-, endothelial- and immune-cells. Most of the effects on the surrounding environment are probably exerted through the release of a variety of extracellular vesicles (EVs), which contain many different classes of molecules, from genetic material to defined species of lipids and enzymes. EV-associated molecules can be either released into the extracellular matrix (ECM) and/or transferred to neighboring cells: as a consequence, both deep modifications of the recipient cell phenotype and digestion of ECM components are obtained, thus causing cancer propagation, as well as a general brain dysfunction. In this review, we first analyze the main intracellular and extracellular transformations required for glioma cell invasion into the brain parenchyma; then we discuss how these events may be attributed, at least in part, to EVs that, like the pawns of a dramatic chess game with cancer, open the way to the tumor cells themselves.
Collapse
|
48
|
Abstract
The canonical Wnt/β-catenin signaling pathway, an important modulator of progenitor cell proliferation and differentiation, is highly regulated for the maintenance of critical biological homeostasis. Decades of studies in cancer genetics and genomics have demonstrated that multiple genes encoding key proteins in this signaling pathway serve as targets for recurrent mutational alterations. Among these proteins, β-catenin and adenomatosis polyposis coli (APC) are two key nodes. β-catenin contributes in transporting extracellular signals for nuclear programming. Mutations of the CTNNB1 gene that encodes β-catenin occur in a wide spectrum of cancers. These mutations alter the spatial characteristics of the β-catenin protein, leading to drastic reprogramming of the nuclear transcriptional network. Among the outcomes of this reprogramming are increased cell proliferation, enhanced immunosuppression, and disruption of metabolic regulation. Herein we review the current understanding of CTNNB1 mutations, their roles in tumorigenesis and discuss their possible therapeutic implications for cancer.
Collapse
|
49
|
Lu M, Wang Y, Zhou S, Xu J, Li J, Tao R, Zhu Y. MicroRNA-370 suppresses the progression and proliferation of human astrocytoma and glioblastoma by negatively regulating β-catenin and causing activation of FOXO3a. Exp Ther Med 2017; 15:1093-1098. [PMID: 29399110 DOI: 10.3892/etm.2017.5494] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 06/29/2017] [Indexed: 02/07/2023] Open
Abstract
Certain microRNAs (miRs) regulate the progression and metastasis of various cancer types. In the present study, the role of miR-370 in the progression and proliferation of human astrocytoma and glioblastoma cells was assessed and the underlying molecular mechanism was investigated. miR-370 levels in clinical specimens of human glioma and peritumoral tissues were determined by reverse-transcription quantitative PCR. Oligonucleotide mimics and inhibitors were transfected into the U-251MG human astrocytoma cell line and the and U-87MG glioblastoma cell line and the cell viability of was determined by an MTT assay. The expression of β-catenin and forkhead box protein (FOX)O3a was determined by western blot analysis. The results revealed that the expression of miR-370 in human glioma tissues was significantly decreased compared with that in peritumoral tissues. The miR-370 levels in patients with grade III/IV gliomas were significantly decreased compared with those in grade I/II. Transfection with miR-370 mimics inhibited the proliferation of U-251MG and U-87MG cells. Furthermore, the miR-370 levels were negatively correlated with β-catenin and positively correlated with nuclear FOXO3a. In conclusion, miR-370 inhibited the proliferation of human glioma cells by regulating the levels of β-catenin and the activation of FOXO3a, suggesting that miR-370 was a tumor suppressor in the progression of human astrocytoma and glioblastoma cells.
Collapse
Affiliation(s)
- Ming Lu
- Department of Radiotherapy, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Yong Wang
- Department of Neurosurgery, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Shizhen Zhou
- Department of Neurosurgery, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Jun Xu
- Department of Neurosurgery, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Jing Li
- Department of Neurosurgery, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Rongjie Tao
- Department of Neurosurgery, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Yufang Zhu
- Department of Neurosurgery, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
50
|
MicroRNA-370 inhibits the growth and metastasis of lung cancer by down-regulating epidermal growth factor receptor expression. Oncotarget 2017; 8:88139-88151. [PMID: 29152147 PMCID: PMC5675699 DOI: 10.18632/oncotarget.21537] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 07/25/2017] [Indexed: 12/13/2022] Open
Abstract
Abnormal microRNA-370 (miR-370) expression has been frequently reported in several types of cancers, including lung cancer. However, the role and molecular mechanisms of miR-370 in regulating the growth and metastasis of lung cancer have not been clarified. Here, we show higher levels of epidermal growth factor receptor (EGFR), but lower levels of miR-370 expression in most human lung cancer cells and non-tumor cells. Induction of miR-370 over-expression significantly reduced the levels of EGFR expression and the EGFR 3′untranslated region (UTR)-regulated luciferase activity in XWLC-05 and H157 cells, suggesting that miR-370 may bind to the 3′UTR of EGFR mRNA. Compared with the control cells, induction of miR370 overexpression significantly inhibited the proliferation, clone formation capacity, migration and invasion of XWLC-05 and H157 cells while miR-370 inhibitor over-expression enhanced their tumor behaviors in vitro. Furthermore, miR-370 over-expression down-regulated the EGFR and hypoxia-inducible factor (HIF)-1α expression, and attenuated the extracellular single-regulated kinase (ERK)1/2 and AKT phosphorylation in XWLC-05 and H157 cells. In contrast, miR370 inhibitor over-expression increased the EGFR and HIF-1α expression as well as the ERK1/2 and AKT phosphorylation in XWLC-05 and H157 cells. Moreover, miR-370 over-expression significantly reduced the levels of EGFR and CD31 expression and inhibited the growth and lung metastasis of xenograft NSCLC tumors in mice. Our study indicates that miR-370 may bind to the 3′UTR of EGFR to inhibit EGFR expression and the growth, angiogenesis and metastasis of non-small cell lung cancer by down-regulating the ERK1/2 and AKT signaling.
Collapse
|