1
|
Peng M, Meng H, Wang J, Guo M, Li T, Qian X, Chen R, Jin H, Huang C. p27 specifically decreases in squamous carcinoma, and mediates NNK-induced transformation of human bronchial epithelial cells. J Cell Mol Med 2024; 28:e18577. [PMID: 39099000 PMCID: PMC11298314 DOI: 10.1111/jcmm.18577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 07/05/2024] [Accepted: 07/17/2024] [Indexed: 08/06/2024] Open
Abstract
Lung cancer remains the leading cause of cancer-related deaths, with cigarette smoking being the most critical factor, linked to nearly 90% of lung cancer cases. NNK, a highly carcinogenic nitrosamine found in tobacco, is implicated in the lung cancer-causing effects of cigarette smoke. Although NNK is known to mutate or activate certain oncogenes, its potential interaction with p27 in modulating these carcinogenic effects is currently unexplored. Recent studies have identified specific downregulation of p27 in human squamous cell carcinoma, in contrast to adenocarcinoma. Additionally, exposure to NNK significantly suppresses p27 expression in human bronchial epithelial cells. Subsequent studies indicates that the downregulation of p27 is pivotal in NNK-induced cell transformation. Mechanistic investigations have shown that reduced p27 expression leads to increased level of ITCH, which facilitates the degradation of Jun B protein. This degradation in turn, augments miR-494 expression and its direct regulation of JAK1 mRNA stability and protein expression, ultimately activating STAT3 and driving cell transformation. In summary, our findings reveal that: (1) the downregulation of p27 increases Jun B expression by upregulating Jun B E3 ligase ITCH, which then boosts miR-494 transcription; (2) Elevated miR-494 directly binds to 3'-UTR of JAK1 mRNA, enhancing its stability and protein expression; and (3) The JAK1/STAT3 pathway is a downstream effector of p27, mediating the oncogenic effect of NNK in lung cancer. These findings provide significant insight into understanding the participation of mechanisms underlying p27 inhibition of NNK induced lung squamous cell carcinogenic effect.
Collapse
Affiliation(s)
- Minggang Peng
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouZhejiangChina
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Hao Meng
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouZhejiangChina
| | - Jingjing Wang
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouZhejiangChina
| | - Mengxin Guo
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouZhejiangChina
| | - Tengda Li
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouZhejiangChina
| | - Xiaohui Qian
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouZhejiangChina
| | - Ruifan Chen
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouZhejiangChina
| | - Honglei Jin
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouZhejiangChina
| | - Chuanshu Huang
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouZhejiangChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouZhejiangChina
| |
Collapse
|
2
|
Patil N, Abdelrahim OG, Leupold JH, Allgayer H. JAK1 Is a Novel Target of Tumor- and Invasion-Suppressive microRNA 494-5p in Colorectal Cancer. Cancers (Basel) 2023; 16:24. [PMID: 38201452 PMCID: PMC10778350 DOI: 10.3390/cancers16010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
MiR-494-5p expression has been suggested to be associated with colorectal cancer (CRC) and its metastases in our previous studies. However, functional investigations on the molecule-mediating actions of this miR in CRC are lacking. In silico analysis in the present study revealed a putative binding sequence within the 3'UTR of JAK1. Overexpression of miR-494-5p in cultured CRC significantly reduced the luciferase activity of a reporter plasmid containing the wild-type JAK1-3'UTR, which was abolished by seed sequence mutation. Furthermore, the overexpression of miR-494-5p in CRC cell lines led to a significant reduction in JAK1 expression, proliferation, in vitro migration, and invasion. These effects were abolished by co-transfection with a specific double-stranded RNA that inhibits endogenous miR-494-5p. Moreover, IL-4-induced migration, invasion, and phosphorylation of JAK1, STAT6, and AKT proteins were reduced after an overexpression of this miR, suggesting that this miR affects one of the most essential pathways in CRC. A Kaplan-Meier plotter analysis revealed that patients with high JAK1 expression show reduced survival. Together, these data suggest that miR-494-5p physically inhibits the expression of JAK1 at the translational level as well as in migration and invasion, supporting the hypothesis of miR-494-5p as an early tumor suppressor and inhibitor of early steps of metastasis in CRC.
Collapse
Affiliation(s)
| | | | | | - Heike Allgayer
- Correspondence: ; Tel.: +49-(0)621-383-71630 or +49-(0)621-383-71635; Fax: +49-(0)621-383-71631
| |
Collapse
|
3
|
Kandettu A, Adiga D, Devi V, Suresh PS, Chakrabarty S, Radhakrishnan R, Kabekkodu SP. Deregulated miRNA clusters in ovarian cancer: Imperative implications in personalized medicine. Genes Dis 2022; 9:1443-1465. [PMID: 36157483 PMCID: PMC9485269 DOI: 10.1016/j.gendis.2021.12.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/04/2021] [Accepted: 12/31/2021] [Indexed: 11/25/2022] Open
Abstract
Ovarian cancer (OC) is one of the most common and fatal types of gynecological cancer. OC is usually detected at the advanced stages of the disease, making it highly lethal. miRNAs are single-stranded, small non-coding RNAs with an approximate size ranging around 22 nt. Interestingly, a considerable proportion of miRNAs are organized in clusters with miRNA genes placed adjacent to one another, getting transcribed together to result in miRNA clusters (MCs). MCs comprise two or more miRNAs that follow the same orientation during transcription. Abnormal expression of the miRNA cluster has been identified as one of the key drivers in OC. MC exists both as tumor-suppressive and oncogenic clusters and has a significant role in OC pathogenesis by facilitating cancer cells to acquire various hallmarks. The present review summarizes the regulation and biological function of MCs in OC. The review also highlights the utility of abnormally expressed MCs in the clinical management of OC.
Collapse
Affiliation(s)
- Amoolya Kandettu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Vasudha Devi
- Department of Pharmacology, Centre for Cardiovascular Pharmacology, Melaka Manipal Medical College, Manipal Academy of Higher Education, Manipal Campus, Manipal, Karnataka 576104, India
| | - Padmanaban S. Suresh
- School of Biotechnology, National Institute of Technology, Calicut, Kerala 673601, India
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| |
Collapse
|
4
|
Identification of Candidate MicroRNA-mRNA Subnetwork for Predicting the Osteosarcoma Progression by Bioinformatics Analysis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:1821233. [PMID: 36238488 PMCID: PMC9553349 DOI: 10.1155/2022/1821233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 08/24/2022] [Indexed: 12/04/2022]
Abstract
Osteosarcoma (OS) is the pretty common primary cancer of the bone among the malignancies in adolescents. A single molecular component or a limited number of molecules is insufficient as a predictive biomarker of OS progression. Hence, it is necessary to find novel network biomarkers to improve the prediction and therapeutic effect for OS. Here, we identified 230 DE-miRNAs and 821 DE-mRNAs through two miRNA expression-profiling datasets and three mRNA expression-profiling datasets. We found that hsa-miR-494 is closely linked with the survival of OS patients. In addition, we analyzed GO and KEGG enrichment for targets of hsa-miR-494-5p and hsa-miR-494-3p through R programming. And five mRNAs were predicted as common targets of hsa-miR-494-5p and hsa-miR-494-3p. We further revealed that upregulated TRPS1 was strongly correlated with poor outcomes in OS patients through the survival analysis based on the TARGET database. The qRT-PCR study verified that the expression of hsa-miR-494-5p and hsa-miR-494-3p was declined considerably, while TRPS1 was notably raised in OS cells when compared to the osteoblasts. Thus, we generated a new regulatory subnetwork of key miRNAs and target mRNAs using Cytoscape software. These results indicate that the novel miRNA-mRNA subnetwork composed of hsa-miR-494-5p, hsa-miR-494-3p, and TRPS1 might be a characteristic molecule for assessing the prognostic value of OS patients.
Collapse
|
5
|
Wang H, Zhang X, Qiao L, Wang H. CircRNA circ_0000554 promotes ovarian cancer invasion and proliferation by regulating miR-567. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:19072-19080. [PMID: 34709546 DOI: 10.1007/s11356-021-13710-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 03/25/2021] [Indexed: 06/13/2023]
Abstract
Circular RNAs (circRNAs) indicated potential modulating effects in tumor development. However, the specific role of circ_0000554 in ovarian tumor remains unknown. We found that circ_0000554 was overexpressed in ovarian tumor specimens and cells. Forced expression of circ_0000554 promoted cell growth, invasion, and epithelial to mesenchymal transition (EMT). We illustrated that miR-567 was downregulated in ovarian tumor specimens and cells. circ_0000554 was negatively correlated with miR-567 in ovarian tumor specimens. circ_0000554 sponged miR-567 expression in ovarian tumor. RIP assay showed that elevated expression of miR-567 could be enriched with circ_0000554. Luciferase reporter assay indicated that luciferase intensity was inhibited after treated with miR-567 mimic; however, the luciferase value of mut type was not decreased. Elevated expression of circ_0000554 suppressed miR-567 expression in HO8910 cell. circ_0000554 promoted ovarian tumor cell growth, invasion, and EMT via sponging miR-567. It suggested that circ_0000554 represent a potential therapy target for ovarian tumor.
Collapse
Affiliation(s)
- Hui Wang
- Department of Gynaecology and Obstetrics, Shengli Oilfield Central Hospital, Dongying, 57000, Shandong, China
| | - Xuezhong Zhang
- Department of Laboratory Medicine, Zibo Central Hospital, Zibo, 255036, Shandong, China
| | - Lujun Qiao
- Intensive Care Unit, Shengli Oilfield Central Hospital, Dongying, 257000, Shandong, China
| | - Heng Wang
- Intensive Care Unit, Shengli Oilfield Central Hospital, Dongying, 257000, Shandong, China.
| |
Collapse
|
6
|
Liu S, Duan K, Zhang X, Cao X, Wang X, Meng F, Liu H, Xu B, Wang X. Circ_0081001 down-regulates miR-494-3p to enhance BACH1 expression and promotes osteosarcoma progression. Aging (Albany NY) 2021; 13:17274-17284. [PMID: 34191748 PMCID: PMC8312427 DOI: 10.18632/aging.203207] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 05/24/2021] [Indexed: 12/13/2022]
Abstract
The study was aimed at deciphering the function and mechanism of circ_0081001 in osteosarcoma (OS). In this study, quantitative real-time polymerase chain reaction (qRT-PCR) was utilized for quantifying circ_0081001, miR-494-3p, and BTB domain and CNC homolog 1 (BACH1) mRNA expressions in OS tissues and cells. Cell counting kit-8 (CCK-8) assay, together with 5-Ethynyl-2'-deoxyuridine (EdU) assay, was performed for evaluating cell proliferation; the alterations in apoptosis were analyzed utilizing flow cytometry; Transwell assay was conducted for examining cell migration and invasion; moreover, Western blot was utilized for the quantification of BACH1 protein expression; bioinformatics, dual-luciferase reporter gene, and RNA-binding protein immunoprecipitation assays were executed for validating the binding relationships between circ_0081001 and miR-494-3p, and between miR-494-3p and BACH1. As shown, circ_0081001, whose expression was elevated in OS tissues, had a negative association with miR-494-3p expression and a positive correlation with BACH1 expression. After circ_0081001 was overexpressed, the proliferation, migration, and invasion of OS cells were boosted but the apoptosis was reduced, whereas miR-494-3p exhibited opposite effects. The binding sites between circ_0081001 and miR-494-3p, and between miR-494-3p and the 3'UTR of BACH1 were experimentally verified. In conclusion, circ_0081001/miR-494-3p/BACH1 axis promoted the malignant biological behaviors of OS cells.
Collapse
Affiliation(s)
- Shizhang Liu
- Department of Orthopedics, Shaanxi Provincial People’s Hospital, Xian 710068, Shanxi, China
| | - Keke Duan
- Department of Orthopedics, Shaanxi Provincial People’s Hospital, Xian 710068, Shanxi, China
| | - Xiaoxia Zhang
- Department of Orthopedics, Shaanxi Provincial People’s Hospital, Xian 710068, Shanxi, China
| | - Xiane Cao
- Department of Orthopedics, Linyi People's Hospital, Linyi 276100, Shandong, China
| | - Xixia Wang
- Department of Central Sterile Supply, Linyi People's Hospital, Linyi 276100, Shandong, China
| | - Fanbin Meng
- Department of Orthopedics, Linyi People's Hospital, Linyi 276100, Shandong, China
| | - Huitong Liu
- Department of Orthopedics, Shaanxi Provincial People’s Hospital, Xian 710068, Shanxi, China
| | - Bingqiang Xu
- Department of Orthopedics, Shaanxi Provincial People’s Hospital, Xian 710068, Shanxi, China
| | - Xi Wang
- Department of Orthopedics, Shaanxi Provincial People’s Hospital, Xian 710068, Shanxi, China
| |
Collapse
|
7
|
Dwivedi SKD, Rao G, Dey A, Mukherjee P, Wren JD, Bhattacharya R. Small Non-Coding-RNA in Gynecological Malignancies. Cancers (Basel) 2021; 13:1085. [PMID: 33802524 PMCID: PMC7961667 DOI: 10.3390/cancers13051085] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/18/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
Gynecologic malignancies, which include cancers of the cervix, ovary, uterus, vulva, vagina, and fallopian tube, are among the leading causes of female mortality worldwide, with the most prevalent being endometrial, ovarian, and cervical cancer. Gynecologic malignancies are complex, heterogeneous diseases, and despite extensive research efforts, the molecular mechanisms underlying their development and pathology remain largely unclear. Currently, mechanistic and therapeutic research in cancer is largely focused on protein targets that are encoded by about 1% of the human genome. Our current understanding of 99% of the genome, which includes noncoding RNA, is limited. The discovery of tens of thousands of noncoding RNAs (ncRNAs), possessing either structural or regulatory functions, has fundamentally altered our understanding of genetics, physiology, pathophysiology, and disease treatment as they relate to gynecologic malignancies. In recent years, it has become clear that ncRNAs are relatively stable, and can serve as biomarkers for cancer diagnosis and prognosis, as well as guide therapy choices. Here we discuss the role of small non-coding RNAs, i.e., microRNAs (miRs), P-Element induced wimpy testis interacting (PIWI) RNAs (piRNAs), and tRNA-derived small RNAs in gynecological malignancies, specifically focusing on ovarian, endometrial, and cervical cancer.
Collapse
Affiliation(s)
- Shailendra Kumar Dhar Dwivedi
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.K.D.D.); (A.D.)
| | - Geeta Rao
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (G.R.); (P.M.)
| | - Anindya Dey
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.K.D.D.); (A.D.)
| | - Priyabrata Mukherjee
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (G.R.); (P.M.)
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jonathan D. Wren
- Biochemistry and Molecular Biology Department, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.K.D.D.); (A.D.)
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
8
|
Ferreira P, Roela RA, Lopez RVM, Del Pilar Estevez-Diz M. The prognostic role of microRNA in epithelial ovarian cancer: a systematic review of literature with an overall survival meta-analysis. Oncotarget 2020; 11:1085-1095. [PMID: 32256980 PMCID: PMC7105164 DOI: 10.18632/oncotarget.27246] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/16/2019] [Indexed: 01/22/2023] Open
Abstract
Objective: To accomplish a systematic review of literature with overall survival meta-analysis about the role of microRNA in epithelial ovarian cancer as prognostic and predictive factor to chemotherapy response.
Methods: A search was conducted in the PubMed database, using the keywords “microRNA” and “ovarian cancer” or “miRNA” and “ovarian cancer”. Original articles published before 02/02/2019 that had as main subject microRNA (miRNA) and ovarian cancer were included. We considered for inclusion only studies that associated microRNA to chemotherapy-related diagnosis, prognosis, or response in ovarian cancer.
Results: The literature search returned 1,482 articles, 497 of which fulfilled inclusion criteria, yielding 350 miRNAs. The status of each miRNA was assessed in serum and tissue of ovarian cancer, benign tumors, and healthy tissue. The status of up-/downregulation of miRNAs was related to prognostic features (overall survival and disease-free survival) and response predictive features such as platinum and paclitaxel sensitivity/resistance. The miRNAs that had been cited three or more times were selected for prognostic and response predictive features analysis. Twelve miRNAs fulfilled all these criteria and were included in the overall survival meta-analysis.
Conclusions: miRNAs affect virtually all mechanisms of carcinogenesis, working as either oncogenes or tumor suppressor genes. In this systematic review we identified miRNAs that may be related to prognosis, diagnosis, and chemotherapy sensitivity. The 12 miRNAs identified here should be included in future studies for validation.
Collapse
Affiliation(s)
- Patricia Ferreira
- Instituto do Cancer do Estado de Sao Paulo, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Rosimeire Aparecida Roela
- Instituto do Cancer do Estado de Sao Paulo, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | - Maria Del Pilar Estevez-Diz
- Instituto do Cancer do Estado de Sao Paulo, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
9
|
Ou L, He X, Liu N, Song Y, Li J, Gao L, Huang X, Deng Z, Wang X, Lin S. Sialylation of FGFR1 by ST6Gal‑I overexpression contributes to ovarian cancer cell migration and chemoresistance. Mol Med Rep 2020; 21:1449-1460. [PMID: 32016470 PMCID: PMC7003046 DOI: 10.3892/mmr.2020.10951] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 08/05/2019] [Indexed: 01/01/2023] Open
Abstract
Fibroblast growth factor receptors (FGFRs) have been implicated in the malignant transformation and chemoresistance of epithelial ovarian cancer; however, the underlying molecular mechanisms are poorly understood. Increased sialyltransferase activity that enhances protein sialylation is an important post‑translational process promoting cancer progression and malignancy. In the present study, α2,6‑sialyltransferase (ST6Gal‑I) overexpression or knockdown cell lines were developed, and FGFR1 was examined to understand the effect of sialylation on migration and drug resistance, and the underlying mechanisms. It was identified that cells with ST6Gal‑I overexpression had increased cell viability and migratory ability upon serum deprivation. Moreover, ST6Gal‑I overexpression cells had strong resistance to paclitaxel, as demonstrated by low growth inhibition rate and cell apoptosis level. A mechanistic study showed that ST6Gal‑I overexpression induced high α2,6‑sialylation of FGFR1 and increased the expression of phospho‑ERK1/2 and phospho‑focal adhesion kinase. Further study demonstrated that the FGFR1 inhibitor PD173047 reduced cell viability and induced apoptosis; however, ST6Gal‑I overexpression decreased the anticancer effect of PD173047. In addition, ST6Gal‑I overexpression attenuated the effect of Adriamycin on cancer cells. Collectively, these results suggested that FGFR1 sialylation plays an important role in cell migration and drug chemoresistance in ovarian cancer cells.
Collapse
Affiliation(s)
- Lingling Ou
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Xiuzhen He
- Chongqing Three Gorges Medical College, Chongqing 404120, P.R. China
| | - Naihua Liu
- Integrated Traditional and Western Medicine Research Center of The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Yuwei Song
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Jinyuan Li
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Lvfen Gao
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Xinke Huang
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Zhendong Deng
- Integrated Traditional and Western Medicine Research Center of The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Xiaoyu Wang
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
- Dr Xiaoyu Wang, Department of Stomatology, The First Affiliated Hospital of Jinan University, 613 West Huangpu Avenue, Guangzhou, Guangdong 510632, P.R. China, E-mail:
| | - Shaoqiang Lin
- Integrated Traditional and Western Medicine Research Center of The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
- School of Pharmaceutical Sciences of Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
- Correspondence to: Dr Shaoqiang Lin, Integrated Traditional and Western Medicine Research Center of The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nonglinxia Road, Guangzhou, Guangdong 510000, P.R. China, E-mail:
| |
Collapse
|
10
|
Ou L, He X, Liu N, Song Y, Li J, Gao L, Huang X, Deng Z, Wang X, Lin S. Sialylation of FGFR1 by ST6Gal‑I overexpression contributes to ovarian cancer cell migration and chemoresistance. Mol Med Rep 2020. [PMID: 32016470 DOI: 10.3892/mmr.2020.10951/html] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
Fibroblast growth factor receptors (FGFRs) have been implicated in the malignant transformation and chemoresistance of epithelial ovarian cancer; however, the underlying molecular mechanisms are poorly understood. Increased sialyltransferase activity that enhances protein sialylation is an important post‑translational process promoting cancer progression and malignancy. In the present study, α2,6‑sialyltransferase (ST6Gal‑I) overexpression or knockdown cell lines were developed, and FGFR1 was examined to understand the effect of sialylation on migration and drug resistance, and the underlying mechanisms. It was identified that cells with ST6Gal‑I overexpression had increased cell viability and migratory ability upon serum deprivation. Moreover, ST6Gal‑I overexpression cells had strong resistance to paclitaxel, as demonstrated by low growth inhibition rate and cell apoptosis level. A mechanistic study showed that ST6Gal‑I overexpression induced high α2,6‑sialylation of FGFR1 and increased the expression of phospho‑ERK1/2 and phospho‑focal adhesion kinase. Further study demonstrated that the FGFR1 inhibitor PD173047 reduced cell viability and induced apoptosis; however, ST6Gal‑I overexpression decreased the anticancer effect of PD173047. In addition, ST6Gal‑I overexpression attenuated the effect of Adriamycin on cancer cells. Collectively, these results suggested that FGFR1 sialylation plays an important role in cell migration and drug chemoresistance in ovarian cancer cells.
Collapse
Affiliation(s)
- Lingling Ou
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Xiuzhen He
- Chongqing Three Gorges Medical College, Chongqing 404120, P.R. China
| | - Naihua Liu
- Integrated Traditional and Western Medicine Research Center of The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Yuwei Song
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Jinyuan Li
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Lvfen Gao
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Xinke Huang
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Zhendong Deng
- Integrated Traditional and Western Medicine Research Center of The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Xiaoyu Wang
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Shaoqiang Lin
- Integrated Traditional and Western Medicine Research Center of The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
11
|
Hsieh PL, Liao YW, Pichler M, Yu CC. MicroRNAs as Theranostics Targets in Oral Carcinoma Stem Cells. Cancers (Basel) 2020; 12:cancers12020340. [PMID: 32028645 PMCID: PMC7072536 DOI: 10.3390/cancers12020340] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 12/25/2022] Open
Abstract
Oral cancer belongs to head and neck squamous cell carcinoma and has been recognized as one of the most prevalent malignancies worldwide. Recent studies have suggested that cancer stem cells (CSCs) may participate in tumor initiation, metastasis and even recurrence, so the regulation of CSCs has drawn significant attention over the past decade. Among various molecules that are associated with CSCs, non-coding RNAs (ncRNAs) have been indicated as key players in the acquisition and maintenance of cancer stemness. In addition, accumulating studies have shown that the aberrant expression of these ncRNAs may serve as surrogate diagnostic markers or even therapeutic targets for cancer treatment. The current study reviews the previous work by us and others to summarize how these ncRNAs affect oral cancer stemness and their potential theranostic applications. A better understanding of the implication of these ncRNAs in oral tumorigenesis will facilitate the translation of basic ncRNA research into clinical application in the future.
Collapse
Affiliation(s)
- Pei-Ling Hsieh
- Department of Anatomy, School of Medicine, China Medical University, Taichung 404, Taiwan;
| | - Yi-Wen Liao
- School of Dentistry, Chung Shan Medical University, Taichung 40201, Taiwan;
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Martin Pichler
- Research Unit of Non-Coding RNAs and Genome Editing, Division of Clinical Oncology, Department of Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, 8036 Graz, Austria;
| | - Cheng-Chia Yu
- School of Dentistry, Chung Shan Medical University, Taichung 40201, Taiwan;
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Institute of Oral Sciences, Chung Shan Medical University, Taichung 40201, Taiwan
- Correspondence: ; Tel.: +886-4-24718668
| |
Collapse
|
12
|
Su Q, Lv XW, Sun YH, Ye ZL, Kong BH, Qin ZB. MicroRNA-494 Inhibits the LRG1 Expression to Induce Proliferation and Migration of VECs in Rats following Myocardial Infarction. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:110-122. [PMID: 31541797 PMCID: PMC6796686 DOI: 10.1016/j.omtn.2019.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 07/15/2019] [Accepted: 08/07/2019] [Indexed: 12/16/2022]
Abstract
Myocardial infarction (MI) is a life-threatening cardiac event that results in extreme damage to the heart muscle. The Wnt signaling pathway has been implicated in the development of heart diseases. Hence, the current study aimed to investigate the role of microRNA (miRNA) in association with the Wnt signaling pathway to identify potential candidates for MI therapy. Differentially expressed miRNAs associated with MI occurrence were screened, and miR-494 was selected for subsequent experiments. Sprague-Dawley rats were included to establish a MI model via intraperitoneal injection of 0.1 mg/kg atropine sulfate and 40 mg/kg pentobarbital sodium. Then, the interaction between miR-494 and LRG1 was identified. The effect of miR-494 on expression of the Wnt signaling pathway-related genes, proliferation, migration, and invasion ability of fibroblasts and vascular endothelial cells (VECs) was subsequently evaluated through a series of gain- and loss-of-function experiments. The results revealed that miR-494 was poorly expressed and LRG1 was highly expressed in MI rats. miR-494 targets and downregulates LRG1, which resulted in the inactivation of the Wnt signaling pathway and promoted proliferation, migration, and invasion ability of fibroblasts and VECs. In conclusion, this study provided evidence suggesting that overexpressed miR-494 could potentially promote the proliferation, migration, and invasion of fibroblasts and VECs in MI through the inactivation of the Wnt signaling pathway by binding to LRG1.
Collapse
Affiliation(s)
- Qiang Su
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, Guilin 541001, P.R. China.
| | - Xiang-Wei Lv
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, Guilin 541001, P.R. China
| | - Yu-Han Sun
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Zi-Liang Ye
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Bing-Hui Kong
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Zhen-Bai Qin
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| |
Collapse
|
13
|
Wang W, Yin Y, Shan X, Zhou X, Liu P, Cao Q, Zhu D, Zhang J, Zhu W. The Value of Plasma-Based MicroRNAs as Diagnostic Biomarkers for Ovarian Cancer. Am J Med Sci 2019; 358:256-267. [DOI: 10.1016/j.amjms.2019.07.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 07/04/2019] [Accepted: 07/16/2019] [Indexed: 02/07/2023]
|
14
|
Li J, Lv H, Che YQ. Upregulated microRNA-31 inhibits oxidative stress-induced neuronal injury through the JAK/STAT3 pathway by binding to PKD1 in mice with ischemic stroke. J Cell Physiol 2019; 235:2414-2428. [PMID: 31517390 DOI: 10.1002/jcp.29146] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/26/2019] [Indexed: 12/21/2022]
Abstract
Ischemic stroke (IS), which is characterized by high morbidity, disability, and mortality, is recognized as a major cerebrovascular disease. MicroRNA-31 (miR-31) was reported to participate in the progression of brain disease. The present study was conducted in order to investigate the effect of miR-31 on oxidative stress-induced neuronal injury in IS mice with the involvement of protein kinase D1 (PKD1) and the JAK/STAT3 pathway. C57BL/6J mice were used to establish the middle cerebral artery occlusion (MCAO) model. Astrocytes were transfected with miR-31 mimic, miR-31 inhibitor, si-PKD1, or JAK-STAT3 pathway inhibitor. Following the establishment of an oxygen-glucose deprivation (OGD) model, the astrocytes were cocultured with neuronal OGD. Lower miR-31, higher PKD1 expressions, and activated JAK/STAT3 pathway were found in both the MCAO and OGD models. miR-31 could negatively target PKD1. In an MCAO model, overexpressing miR-31 and silencing PKD1 reduced neuronal injury, cerebral infarct volume, neuron loss, and oxidative stress injury, inhibited the activation of JAK/STAT3 pathway and the expressions of PKD1, interleukin (IL)-1β, IL-6, tumor necrosis factor-α, malondialdehyde, 4-HNE, 8-HOdG, caspase-3, and Bax, but increased the superoxide dismutase content. In the OGD model, overexpression of miR-31 and silencing of PKD1 attenuated oxidative stress-induced neuronal injury, and diminished the lactate dehydrogenase leakage and reactive oxygen species level, accompanied by elevated neuronal viability. These results indicate that miR-31 alleviates inflammatory response as well as an oxidative stress-induced neuronal injury in IS mice by downregulating PKD1 and JAK/STAT3 pathway.
Collapse
Affiliation(s)
- Jie Li
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Hui Lv
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yu-Qin Che
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
15
|
Li X, Nie C, Tian B, Tan X, Han W, Wang J, Jin Y, Li Y, Guan X, Hong A, Chen X. miR-671-5p Blocks The Progression Of Human Esophageal Squamous Cell Carcinoma By Suppressing FGFR2. Int J Biol Sci 2019; 15:1892-1904. [PMID: 31523191 PMCID: PMC6743296 DOI: 10.7150/ijbs.32429] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/02/2019] [Indexed: 12/31/2022] Open
Abstract
Esophageal cancer is the eighth most common malignant tumor worldwide, of which esophageal squamous cell carcinoma (ESCC) is the dominant histological subtype. A drug shortage for ESCC therapy triggered us to explore the roles of fibroblast growth factor receptor 2 (FGFR2) and its upstream regulator miR-671-5p in ESCC progression. We compared the levels of FGFR2 and miR-671-5p between human ESCC tissues and their matched normal esophageal tissues and found an association between higher levels of FGFR2 and lower levels of miR-671-5p in ESCC tissues. High levels of FGFR2 resulted in the activation of the ERK and AKT pathways and a promotion of ESCC progression. High levels of miR-671-5p specifically reduced the expression of FGFR2 and suppressed ESCC progression in both in vitro and in vivo models. Therefore, suppressing FGFR2 and enhancing miR-671-5p expression may be the right approaches for ESCC therapy.
Collapse
Affiliation(s)
- Xiaoyan Li
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Changjun Nie
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Baoqing Tian
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Xuan Tan
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Wei Han
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Jiakang Wang
- Cancer Center of Guangzhou Medical University, Guangzhou, Guangdong, 510090, P. R. China
| | - Yuan Jin
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Yadan Li
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Xinyuan Guan
- Department of Clinical Oncology, University of Hong Kong, Hong Kong, China
| | - An Hong
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Xiaojia Chen
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| |
Collapse
|
16
|
Wang W, Wu LR, Li C, Zhou X, Liu P, Jia X, Chen Y, Zhu W. Five serum microRNAs for detection and predicting of ovarian cancer. Eur J Obstet Gynecol Reprod Biol X 2019; 3:100017. [PMID: 31404211 PMCID: PMC6687444 DOI: 10.1016/j.eurox.2019.100017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/24/2019] [Accepted: 03/31/2019] [Indexed: 01/26/2023] Open
Abstract
OBJECTIVE Ovarian cancer (OC) was one of the deadliest gynecological malignancy among women in global. Serum microRNAs (miRNAs) could serve as promising diagnostic biomarkers for patients with OC. STUDY DESIGN Using quantitative reverse transcription polymerase chain reaction (qRT-PCR) based Exiqon panel, we identified 27 differentially expressed miRNAs from one normal control (NC) pool and two OC pool samples in the initial screening stage. We further verified the miRNAs in the training (30 OC VS. 36 NCs) and validation stages (80 OC VS. 80 NCs) based on qRT-PCR. Later, the expression levels of the identified miRNAs were also evaluated in exosomes and tissues. RESULTS We found a serum microRNA signature including five overexpressed miRNAs (miR-200c-3p, miR-346, miR-127-3p, miR-143-3p and miR-205-5p) in OC in comparison with NCs. The areas under the receiver operating characteristic (ROC) curve (AUC) of the five-miRNA panel were 0.783 for the training stage and 0.745 for the validation stage. The diagnostic sensitivity and specificity of the combined five-miRNA panel was 0.818 and 0.609 when the cut-off value was 0.636. The levels of miR-200c-3p, miR-346 and miR-127-3p in serum were related to tumor grade and distant metastasis of OC. The expression levels of the five miRNAs were also significantly up-regulated in serum exosomes (32 OC VS. 32 NCs). Furthermore, miR-200c-3p was significantly elevated in OC tissues (22 OC VS. 22 NCs). But the levels of the miR-346 and miR-143-3p were significantly lower in OC tissues. CONCLUSION Our findings showed a five-miRNA panel in serum for the detection of OC. Moreover, serum expression levels of miR-200c-3p, miR-346 and miR-127-3p were concerned with tumor grade and distant metastasis of OC.
Collapse
Affiliation(s)
- Weiwei Wang
- Intensive Care Unit, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Li-rong Wu
- Department of Radiation Oncology, The Nanjing Medical University Affiliated Cancer Hospital, 42 Baiziting Road, Nanjing 210009, China
| | - Chunyu Li
- Emergency Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Xin Zhou
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Ping Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Xuemei Jia
- Department of Obstetrics and Gynecology, Nanjing Maternity and Child Health Care hospital, 123 Mochou Road, Nanjing 210004, China
| | - Yan Chen
- Emergency Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
- Medical Department, Kizilsu Kirghiz Autonomous Prefecture People’s Hospital, Artux 845350, China
| | - Wei Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
- Department of Oncology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, No.1399 West Road, Shengze Town, Wujiang District, Suzhou 215000, China
| |
Collapse
|
17
|
Mirza-Aghazadeh-Attari M, Ostadian C, Saei AA, Mihanfar A, Darband SG, Sadighparvar S, Kaviani M, Samadi Kafil H, Yousefi B, Majidinia M. DNA damage response and repair in ovarian cancer: Potential targets for therapeutic strategies. DNA Repair (Amst) 2019; 80:59-84. [PMID: 31279973 DOI: 10.1016/j.dnarep.2019.06.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 06/01/2019] [Accepted: 06/15/2019] [Indexed: 12/24/2022]
Abstract
Ovarian cancer is among the most lethal gynecologic malignancies with a poor survival prognosis. The current therapeutic strategies involve surgery and chemotherapy. Research is now focused on novel agents especially those targeting DNA damage response (DDR) pathways. Understanding the DDR process in ovarian cancer necessitates having a detailed knowledge on a series of signaling mediators at the cellular and molecular levels. The complexity of the DDR process in ovarian cancer and how this process works in metastatic conditions is comprehensively reviewed. For evaluating the efficacy of therapeutic agents targeting DNA damage in ovarian cancer, we will discuss the components of this system including DDR sensors, DDR transducers, DDR mediators, and DDR effectors. The constituent pathways include DNA repair machinery, cell cycle checkpoints, and apoptotic pathways. We also will assess the potential of active mediators involved in the DDR process such as therapeutic and prognostic candidates that may facilitate future studies.
Collapse
Affiliation(s)
- Mohammad Mirza-Aghazadeh-Attari
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Caspian Ostadian
- Department of Biology, Faculty of Science, Urmia University, Urmia, Iran
| | - Amir Ata Saei
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Ainaz Mihanfar
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Saber Ghazizadeh Darband
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden; Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Shirin Sadighparvar
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Mojtaba Kaviani
- School of Nutrition and Dietetics, Acadia University, Wolfville, Nova Scotia, Canada
| | | | - Bahman Yousefi
- Molecular MedicineResearch Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
18
|
Zhao X, Wang Y, Yang J, Liu H, Wang L. MicroRNA‐326 suppresses iNOS expression and promotes autophagy of dopaminergic neurons through the JNK signaling by targeting XBP1 in a mouse model of Parkinson's disease. J Cell Biochem 2019; 120:14995-15006. [PMID: 31135066 DOI: 10.1002/jcb.28761] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/26/2018] [Accepted: 01/10/2019] [Indexed: 02/03/2023]
Affiliation(s)
- Xiao‐Hui Zhao
- Department of Neurology Shanghai University of Medicine & Health Sciences Affiliated Pudong New District People's Hospital Shanghai P. R. China
| | - Yong‐Bing Wang
- Department of General Surgery Shanghai University of Medicine & Health Sciences Affiliated Pudong New District People's Hospital Shanghai P. R. China
| | - Juan Yang
- Department of Neurology Shanghai University of Medicine & Health Sciences Affiliated Pudong New District People's Hospital Shanghai P. R. China
| | - Hui‐Qin Liu
- Department of Neurology Shanghai University of Medicine & Health Sciences Affiliated Pudong New District People's Hospital Shanghai P. R. China
| | - Ling‐Ling Wang
- Department of Neurology Shanghai University of Medicine & Health Sciences Affiliated Pudong New District People's Hospital Shanghai P. R. China
| |
Collapse
|
19
|
Pu L, Su L, Kang X. The efficacy of cisplatin on nasopharyngeal carcinoma cells may be increased via the downregulation of fibroblast growth factor receptor 2. Int J Mol Med 2019; 44:57-66. [PMID: 31115494 PMCID: PMC6559331 DOI: 10.3892/ijmm.2019.4193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 04/12/2019] [Indexed: 11/06/2022] Open
Abstract
Cisplatin is one of the primary compounds used in the treatment of nasopharyngeal carcinoma (NPC), and fibroblast growth factor receptor 2 (FGFR2) has emerged to be a promising target for treatment in various tumors. Therefore, the present study aimed to explore whether the expression levels of FGFR2 in NPC tissues and cell lines were altered, and whether the efficiency of cisplatin was increased following the downregulation of FGFR2. The downregulation of FGFR2 was achieved by transfection with a small interfering RNA against FGFR2. Tissues of patients with NPC were analyzed by immunohistochemistry. Cell viability was examined using a Cell Counting Kit‑8 assay. Cell cycle analysis was performed using flow cytometry. mRNA and protein levels were measured by reverse transcription quantitative polymerase chain reaction and western blot analysis, respectively. FGFR2 was observed to be overexpressed in cancer tissues of patients with NPC and in the NPC SUNE1, C666‑1, 6‑10B and HNE‑3 cell lines, and resulted in an unfavorable prognosis. Cisplatin treatment decreased cell viability and increased FGFR2 expression. The silencing of FGFR2 was demonstrated to augment the effects of cisplatin treatment, including decreasing the cell viability and inducing cell cycle arrest, which involved the increase and decrease of the durations of G1 and S phases, respectively, and a decrease in the expression levels of cyclin D1 and CDC25A, and increasing the rate of apoptosis via the intrinsic apoptosis pathway, as demonstrated by the upregulation of cleaved caspase‑3 and B‑cell lymphoma 2 (Bcl‑2)‑associated X protein and downregulation of Bcl‑2, in SUNE1 and C666‑1 cell lines. FGFR2 was overexpressed in the cancer tissues of patients with NPC and in NPC cell lines, resulting in an unfavorable prognosis. The downregulation of FGFR2 decreased cell viability via cell cycle arrest at G1 phase, and increased the efficacy of the cisplatin‑based induction of apoptosis through the intrinsic apoptosis pathway.
Collapse
Affiliation(s)
- Li Pu
- Department of Otolaryngology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Lizhong Su
- Department of Otolaryngology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Xixun Kang
- Department of Otolaryngology, Head and Neck Surgery, Shenzhen Hospital, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518106, P.R. China
| |
Collapse
|
20
|
Yu HM, Yao XD, Zhang RM, Zhuang HF, Wang PW, Li YZ. Repression of let-7b-5p prevents the development of multifidus muscle dysfunction by promoting vitamin D accumulation via upregulation of electron transfer flavoprotein alpha subunit in a rat model of multifidus muscle injury. J Cell Biochem 2019; 120:7458-7473. [PMID: 30387180 DOI: 10.1002/jcb.28020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/15/2018] [Indexed: 01/24/2023]
Abstract
Multifidus muscle dysfunction is associated with the multifidus muscle injury (MMI), which ultimately result in the low-back pain. Increasing evidence shows that microRNAs (miRs) may be involved in multifidus muscle dysfunction. In this study, we tested the hypothesis that downregulation of let-7b-5p may inhibit the multifidus muscle dysfunction development and progression. The target prediction program and luciferase activity determination confirmed electron transfer flavoprotein alpha subunit (ETFA) as a direct target gene of let-7b-5p. To study the mechanisms and functions of let-7b-5p in relation to ETFA in MMI progression, we prepared rats with experimental MMI, and a lentivirus-based packaging system was designed to upregulate expressions of let-7b-5p, and downregulate the expression of ETFA. ETFA was identified as a target gene of let-7b-5p. Older age, a longer duration of pain, and higher visual analog scale and Oswestry disability index scores for the patients with chronic low-back pain were linked to a more severe degree of degenerative muscle atrophy and fatty infiltration. Increased expression of let-7b-5p and decreased expression of ETFA and vitamin D receptor (VDR) were positively correlated with multifidus muscle dysfunction. Downregulated let-7b-5p could inhibit infiltration of collagen fibers, reverse the ultrastructural changes of multifidus muscle, and induce the VDR expression, thereby repair the MMI. The results provided a potential basis for let-7b-5p that could support targeted intervention in multifidus muscle dysfunction. Collectively, this study confirmed that downregulation of let-7b-5p has a potential inhibitory effect on the development of the function of the musculus myocytes by upregulating ETFA.
Collapse
Affiliation(s)
- Hai-Ming Yu
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xue-Dong Yao
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Rong-Mou Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Hua-Feng Zhuang
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Pei-Wen Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yi-Zhong Li
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
21
|
Wang X, Zhou Z, Zhang T, Wang M, Xu R, Qin S, Zhang S. Overexpression of miR-664 is associated with poor overall survival and accelerates cell proliferation, migration and invasion in hepatocellular carcinoma. Onco Targets Ther 2019; 12:2373-2381. [PMID: 30992673 PMCID: PMC6445241 DOI: 10.2147/ott.s188658] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Introduction Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide. This study aimed to investigate the expression patterns of microRNA-664 (miR-664) in HCC tissues and cells, and assess its clinical significance and functional role in HCC. Patients and methods One hundred and thirty-four paired HCC and non-cancerous tissues were collected from patients who underwent surgery in Qianfoshan Hospital affiliated to Shandong University (Shandong, China) between 2009 and 2012. Expression of miR-664 was measured by quantitative real-time polymerase chain reaction (qRT-PCR). Prognostic value of miR-664 in HCC was evaluated using Kaplan–Meier survival analysis and Cox regression analysis. Cell proliferation was analyzed using the CCK-8 assay, and cell migration and invasion of HCC cells was evaluated by the Transwell assay. Results Expression of miR-664 was significantly upregulated in HCC tissues and cells when compared with the normal controls (all P<0.05). MiR-664 expression was associated with lymph node metastasis, TNM stage and differentiation (all P<0.05) in the HCC patients. High miR-664 expression predicted poor overall survival (log-rank P=0.004) and acted as an independent prognostic factor (HR =1.945, 95% CI=1.078–3.508, P=0.027). According to cell experiments, the upregulation of miR-664 could promote, whereas the downregulation of miR-664 could inhibit proliferation, migration and invasion of HCC cells (all P<0.05). SIVA1 was predicted as a direct target gene of miR-664 in HCC. Conclusion All data indicated that overexpression of miR-664 is associated with poor prognosis of HCC patients, and may enhance tumor progression of HCC by targeting SIVA1. MiR-664 may be a candidate therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Xianming Wang
- Department of General Surgery, Qianfoshan Hospital Affiliated to Shandong University, Shandong 250014, China, ;
| | - Zhengtong Zhou
- Department of General Surgery, Qianfoshan Hospital Affiliated to Shandong University, Shandong 250014, China, ;
| | - Tao Zhang
- Department of General Surgery, Qianfoshan Hospital Affiliated to Shandong University, Shandong 250014, China, ;
| | - Minghai Wang
- Department of General Surgery, Qianfoshan Hospital Affiliated to Shandong University, Shandong 250014, China, ;
| | - Rongwei Xu
- Department of General Surgery, Qianfoshan Hospital Affiliated to Shandong University, Shandong 250014, China, ;
| | - Shiyong Qin
- Department of General Surgery, Qianfoshan Hospital Affiliated to Shandong University, Shandong 250014, China, ;
| | - Shuguang Zhang
- Department of General Surgery, Qianfoshan Hospital Affiliated to Shandong University, Shandong 250014, China, ;
| |
Collapse
|
22
|
MiR-494 acts as a tumor promoter by targeting CASP2 in non-small cell lung cancer. Sci Rep 2019; 9:3008. [PMID: 30816202 PMCID: PMC6395740 DOI: 10.1038/s41598-019-39453-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 01/24/2019] [Indexed: 11/23/2022] Open
Abstract
MiR-494 plays an important role in several types of human cancers, including non-small cell lung cancer (NSCLC). Although the role of miR-494 has been investigated in several studies, the expression profile and underlying mechanism are still poorly understood. In this study, we found that overexpression of miR-494 promoted the proliferation and colony formation of NSCLC cells and reduced their sensitivity to cisplatin-induced apoptosis. By using microarray and Dual luciferase reporter assays, we further showed that caspase-2 (CASP2) is a functional target of miR-494, and the expression of CASP2 is inversely associated with miR-494 in vitro. In addition, miR-494 promoted the proliferation and colony formation of NSCLC cells and reduced their sensitivity to cisplatin-induced apoptosis by targeting CASP2. Therefore, our results suggest that miR-494 plays an oncomiR role in NSCLC cells and may be a candidate biomarker for malignant transformation and a therapeutic target of NSCLC.
Collapse
|
23
|
Xu XH, Zhang SJ, Hu QB, Song XY, Pan W. Retracted: Effects of microRNA-494 on proliferation, migration, invasion, and apoptosis of medulloblastoma cells by mediating c-myc through the p38 MAPK signaling pathway. J Cell Biochem 2019; 120:2594-2606. [PMID: 30304554 DOI: 10.1002/jcb.27559] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 08/06/2018] [Indexed: 02/02/2023]
Abstract
Medulloblastoma (MB) is the most prevalent brain tumor that occurs during childhood and originates from cerebellar granule cell precursors. Based on recent studies, the differential expression of several microRNAs is involved in MB, while the role of microRNA-494 (miR-494) in MB remains unclear. Therefore, we conducted this study to investigate the regulative role of miR-494 in MB cells via the p38 mitogen-activated protein kinase (MAPK) signaling pathway by mediating c-myc. In the current study, MB cells were collected and transfected with miR-494 mimic, miR-494 inhibitor, siRNA- c-myc, and miR-494 inhibitor + siRNA-c-myc. The expressions of miR-494, c-myc, p38 MAPK, B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X protein (Bax), interleukin-6 (IL-6), metadherin (MTDH), phosphatase and tensin homolog (PTEN) and survivin were determined. Cell proliferation, cell-cycle distribution, apoptosis, migration, and invasion were evaluated. The results revealed that there was a poor expression of miR-494 and high expression of c-myc in MB tissues. C-myc was determined as the target gene of miR-494. In response to miR-494 mimic, MB cells were found to have increased Bax and PTEN expressions, as well as cell number in G1 phase and cell apoptosis and decreased c-myc, p38 MAPK, Bcl-2, MTDH, IL-6, and survivin expression and cell number count in the S phase, cell proliferation, migration, and invasion. In conclusion, the results demonstrated that the upregulation of miR-494 results in the suppression of cell proliferation, migration, and invasion, while it promotes apoptosis of MB cells through the negative mediation of c-myc, which in turn inactivates the p38 MAPK pathway.
Collapse
Affiliation(s)
- Xiao-Heng Xu
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, China
| | - Si-Jin Zhang
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, China
| | - Qi-Bo Hu
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, China
| | - Xing-Yu Song
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, China
| | - Wei Pan
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
24
|
Zheng XM, Zhang P, Liu MH, Chen P, Zhang WB. MicroRNA-30e inhibits adhesion, migration, invasion and cell cycle progression of prostate cancer cells via inhibition of the activation of the MAPK signaling pathway by downregulating CHRM3. Int J Oncol 2019; 54:443-454. [PMID: 30483762 PMCID: PMC6317654 DOI: 10.3892/ijo.2018.4647] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/21/2018] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer (PCa) testing is currently based on measurement of serum prostate‑specific antigen levels and digital rectal examination, which are limited by a low predictive value and the adverse effects associated with overdiagnosis and overtreatment. Recent studies have reported that the abnormal expression of microRNAs (miRNAs) is associated with the mechanism underlying the development of PCa. Thus, the aim of the present study was to investigate the effects of miR‑30e and its target gene, M3 muscarinic acetylcholine receptor (CHRM3), on the adhesion, migration, invasion and cell cycle distribution of PCa cells via the mitogen‑activated protein kinase (MAPK) signaling pathway. The differentially expressed genes were screened in the Gene Expression Omnibus database from a gene expression microarray (GSE55945) of PCa. PCa tissues and adjacent tissues were collected from patients with PCa. The PC‑3 and DU145 human PCa cell lines were treated with activator, inhibitor and siRNAs. The effects of miR‑30e on cell adhesion, migration, invasion and cell cycle distribution with the involvement of CHRM3 and the MAPK signaling pathway were investigated. The bioinformatics results demonstrated that the CHRM3 gene and the MAKP signaling pathway were involved in the progression of PCa, and has‑miR‑30e was selected for further study. The levels of miR‑30e were significantly downregulated, while the levels of CHRM3 were obviously upregulated in PCa. CHRM3 was verified as a target gene of miR‑30e. Upregulation of miR‑30e and downregulation of CHRM3 decreased the levels of p‑P38, p‑extracellular signal‑regulated kinase, p‑c‑Jun N‑terminal kinase, p‑c‑fos and p‑c‑JUN, cell adhesion, migration and invasion ability, and the number of cells in the S phase, while they increased the number of cells in the G0 and G1 phases. The findings of the present study suggest that miR‑30e inhibited the adhesion, migration, invasion and cell cycle entry of PCa cells by suppressing the activation of the MAPK signaling pathway and inhibiting CHRM3 expression. Thus, miR‑30e may serve as a candidate target for the treatment of PCa.
Collapse
Affiliation(s)
- Xin-Min Zheng
- Department of Urology, Zhongnan Hospital of WuHan University, Wuhan, Hubei 430071
| | - Peng Zhang
- Department of Urology, Zhongnan Hospital of WuHan University, Wuhan, Hubei 430071
| | - Man-Hua Liu
- The Second Department of Surgery, Jingan People’s Hospital, Jingan, Jiangxi 330600, P.R. China
| | - Ping Chen
- Department of Urology, Zhongnan Hospital of WuHan University, Wuhan, Hubei 430071
| | - Wei-Bing Zhang
- Department of Urology, Zhongnan Hospital of WuHan University, Wuhan, Hubei 430071
| |
Collapse
|
25
|
Kashyap D, Tuli HS, Garg VK, Goel N, Bishayee A. Oncogenic and Tumor-Suppressive Roles of MicroRNAs with Special Reference to Apoptosis: Molecular Mechanisms and Therapeutic Potential. Mol Diagn Ther 2018; 22:179-201. [PMID: 29388067 DOI: 10.1007/s40291-018-0316-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNAs) are the non-coding class of minute RNA molecules that negatively control post-transcriptional regulation of various functional genes. These miRNAs are transcribed from the loci present in the introns of functional or protein-coding genes, exons of non-coding genes, or even in the 3'-untranslated region (3'-UTR). They have potential to modulate the stability or translational efficiency of a variety of target RNA [messenger RNA (mRNA)]. The regulatory function of miRNAs has been elucidated in several pathological conditions, including neurological (Alzheimer's disease and Parkinson's disease) and cardiovascular conditions, along with cancer. Importantly, miRNA identification in cancer progression and invasion has evolved as an incipient era in cancer treatment. Several studies have shown the influence of miRNAs on various cancer processes, including apoptosis, invasion, metastasis and angiogenesis. In particular, apoptosis induction in tumor cells through miRNA has been extensively studied. The biphasic mode (up- and down-regulation) of miRNA expression in apoptosis and other cancer processes has already been determined. The findings of these studies could be utilized to develop potential therapeutic strategies for the management of various cancers. The present review critically describes the oncogenic and tumor suppressor role of miRNAs in apoptosis and other cancer processes, therapy resistance, and use of their presence in the body fluids as biomarkers.
Collapse
Affiliation(s)
- Dharambir Kashyap
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, Punjab, India
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar University, Mullana-Ambala, 133207, Haryana, India.
| | - Vivek Kumar Garg
- Department of Biochemistry, Government Medical College and Hospital, Chandigarh, 160030, Punjab, India
| | - Neelam Goel
- Department of Information Technology, University Institute of Engineering and Technology, Panjab University, Chandigarh, 160014, Punjab, India
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL, 33169, USA.
| |
Collapse
|
26
|
Liu L, Pang XL, Shang WJ, Xie HC, Wang JX, Feng GW. Over-expressed microRNA-181a reduces glomerular sclerosis and renal tubular epithelial injury in rats with chronic kidney disease via down-regulation of the TLR/NF-κB pathway by binding to CRY1. Mol Med 2018; 24:49. [PMID: 30241461 PMCID: PMC6145098 DOI: 10.1186/s10020-018-0045-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 07/30/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) contribute to the progression of chronic kidney disease (CKD) by regulating renal homeostasis. This study explored the effects of miR-181a on CKD through the Toll-like receptor (TLR)/nuclear factor-kappa B (NF-κB) pathway by binding to CRY1. METHODS Seventy male rats were selected and assigned into specific groups: miR-181a mimic, miR-181a inhibitor, and siRNA against CRY1, with each group undergoing different treatments to investigate many different outcomes. First, 24-h urinary protein was measured. ELISA was used to determine the serum levels of SOD, ROS, MDA, IL-1β, IL-6, and TNF-α. Biochemical tests for renal function were performed to measure albumin, uric acid, and urea in urine and urea nitrogen and creatinine in serum. The glomerulosclerosis index (GSI) and renal tubular epithelial (RTE) cell apoptosis were detected using PASM staining and TUNEL staining, respectively. Finally, RT-qPCR and western blot were done to determine miR-181a, CRY1, TLR2, TLR4, and NF-κB expression. RESULTS CRY1 is the target gene of miR-181a, according to a target prediction program and luciferase assay. Rats diagnosed with CKD presented increases in 24-h urinary protein; GSI; RTE cell apoptosis rate; serum ROS, MDA, IL-1β, IL-6, and TNF-α; and CRY1, TLR2, TLR4, and NF-κB expression, as well as decreases in SOD level and miR-181a expression. Following transfection with either the miR-181a mimic or si-CRY1, 24-h urinary protein, renal damage, GSI, and cell apoptosis rate were all decreased. In addition, the overexpression of miR-181a or inhibition of CRY1 alleviated the degree of kidney injury through suppression of the TLR/NF-κB pathway. CONCLUSION miR-181a alleviates both GS and RTE injury in CKD via the down-regulation of the CRY1 gene and the TLR/NF-κB pathway.
Collapse
Affiliation(s)
- Lei Liu
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe Road, Erqi District, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Xin-Lu Pang
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe Road, Erqi District, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Wen-Jun Shang
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe Road, Erqi District, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Hong-Chang Xie
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe Road, Erqi District, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Jun-Xiang Wang
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe Road, Erqi District, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Gui-Wen Feng
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe Road, Erqi District, Zhengzhou, 450052, Henan Province, People's Republic of China.
| |
Collapse
|
27
|
Zhang LN, Tian H, Zhou XL, Tian SC, Zhang XH, Wu TJ. Upregulation of microRNA-351 exerts protective effects during sepsis by ameliorating skeletal muscle wasting through the Tead-4-mediated blockade of the Hippo signaling pathway. FASEB J 2018; 32:fj201800151RR. [PMID: 30040486 DOI: 10.1096/fj.201800151rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Sepsis-induced skeletal muscle wasting may lead to various severe clinical consequences. Understanding molecular mechanisms of the regulation of the loss of skeletal muscle mass in septic patients remains a significant clinical challenge. The current study was conducted to establish septic mice models to explore the relationship between microRNA (miR)-351 and the transcription element apical (TEA) domain transcription factor (Tead)-4 gene and to investigate its effects on the skeletal muscle through mediating the Hippo signaling pathway in mice with acute sepsis. A total of 60 mice were collected to establish mouse models of acute sepsis. The positive expression rate of Tead-4 and the apoptotic index (AI) were measured. A dual-luciferase reporter gene assay was conducted to verify the targeting relationship between miR-351 and Tead-4. Furthermore, the muscle fiber diameter (MFD) and area (MFA) and the content of 3-methylhistidine (3-MH) and tyrosine (Tyr) were assessed. The expression levels of miR-351, p38-MAPK, Yes-associated protein, Tead-4, B-cell lymphoma X protein (Bax), and Caspase-3 were determined with quantitative RT-PCR and Western blot analysis. Finally, cell viability, apoptosis, and levels of inflammatory factors, including IL-1β, IL-6, IGF-1, TNF-α, and monocyte chemoattractant protein-1 were detected by 3-(4,5-dimethylthiazol-2- yl)-2,5-diphenyltetrazolium bromide assay, flow cytometry, and ELISA. Initially, Tead-4 protein expression was higher in skeletal muscle tissues of mice with acute sepsis. Tead-4 was identified to negatively regulate miR-351. Upregulation of miR-351 increased MFA and MFD, muscle weight water content, Bcl-2 expression levels, and cell viability. Up-regulation of miR-351 reduced AI; 3-MH and Tyr content; positive expression of Tead-4 protein; the expression levels of p38-MAPK, Yap, Tead-4, Bax, and Caspase-3; apoptosis; and inflammatory responses. The current study demonstrated that up-regulation of miR-351 inhibits the degradation of skeletal muscle protein and the atrophy of skeletal muscle in mice with acute sepsis by targeting Tead-4 through suppression of the Hippo signaling pathway. Thus, miR-351 overexpression may be a future therapeutic strategy for acute sepsis.-Zhang, L.-N., Tian, H., Zhou, X.-L., Tian, S.-C., Zhang, X.-H., Wu, T.-J. Upregulation of microRNA-351 exerts protective effects during sepsis by ameliorating skeletal muscle wasting through the Tead-4-mediated blockade of the Hippo signaling pathway.
Collapse
Affiliation(s)
- Li-Na Zhang
- Intensive Care Unit, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, China
| | - Hui Tian
- Intensive Care Unit, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, China
| | - Xiu-Li Zhou
- Intensive Care Unit, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, China
| | - Suo-Chen Tian
- Intensive Care Unit, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, China
| | - Xi-Hong Zhang
- Intensive Care Unit, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, China
| | - Tie-Jun Wu
- Intensive Care Unit, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, China
| |
Collapse
|
28
|
Liu J, Jiang Y, Wan Y, Zhou S, Thapa S, Cheng W. MicroRNA‑665 suppresses the growth and migration of ovarian cancer cells by targeting HOXA10. Mol Med Rep 2018; 18:2661-2668. [PMID: 30015865 PMCID: PMC6102655 DOI: 10.3892/mmr.2018.9252] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 06/07/2018] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological cancer and its metastasis leads to a poor prognosis. The present study was designed to elucidate how microRNA (miR)-665 regulates the proliferation and migration of ovarian tumor cells. Reverse transcription-polymerase chain reaction (RT-PCR) demonstrated that miR-665 expression was decreased in ovarian cancer tissues. Increased expression of miR-665 suppressed the growth and migration of ovarian cancer cells, whereas the downregulated expression of miR-665 led to the opposite results. Bioinformatics tools identified homeobox A10 (HOXA10) as a target of miR-665. Following miR-665 overexpression, HOXA10 protein expression was significantly reduced. A dual luciferase assay revealed that miR-665 bound to the 3′-untranslated region of HOXA10. Immunohistochemistry and RT-PCR revealed that the expression of HOXA10 was negatively correlated with the expression of miR-665. It was concluded that miR-665 targets HOXA10 and may act as a tumor-suppressing gene in ovarian cancer. This pathway may be involved in the development and metastasis of ovarian cancer.
Collapse
Affiliation(s)
- Jinhui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yi Jiang
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yicong Wan
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Shulin Zhou
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Sunita Thapa
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wenjun Cheng
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
29
|
Ma Y, Li G, Hu J, Liu X, Shi B. MicroRNA-494 regulates Gli3 expression and inhibits pancreatic cancer cells growth and migration. J Cell Biochem 2018; 119:5324-5331. [PMID: 29315756 DOI: 10.1002/jcb.26658] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/04/2018] [Indexed: 02/04/2023]
Abstract
MicroRNA (miR)-494 has been identified as a predictor and inhibitor in pancreatic cancer. This study aimed to explore the role of miR-494 in pancreatic cancer cells, and the regulation of glioma-associated oncogene 3 (Gli3) by miR-494. The mRNA level of Gli3 in 99 pairs of pancreatic cancer and correspondingly adjacent tissues was monitored by qRT-PCR. Correlation of Gli3 expression with miR-494 level was assessed by Pearson χ2 test. Dual-luciferase reporter assay was used to detect whether Gli3 was a target of miR-494. Following miR-494 mimics and miR-494 inhibitor transfection, the changes in cell viability and migration were detected by using CCK-8 and Transwell chamber. Furthermore, Gli3 siRNA was co-transfected with miR-494 inhibitor, and then cell viability and migration were redetected. Result showed that, the mRNA level of Gli3 in tumor tissues was higher than in the adjacent tissues (P < 0.01). There were 45 in 99 patients with pancreatic cancer expressed Gli3, and significant correlations were observed between the Gli3 level and vascular invasion (P = 0.04), distant metastasis (P = 0.001), and histologic grade (P = 0.03). Gli3 was a direct target of miR-494 (P < 0.01) and it was negatively related by miR-494 (P < 0.01). Overexpression of miR-494 suppressed PANC-1 cells viability (P < 0.05, P < 0.01, or P < 0.001) and migration (P < 0.01). Additionally, Gli3 silence suppresses miR-494 suppression-induced cell viability and migration (P < 0.01). In conclusion, these data demonstrate miR-494 exhibits tumor-suppressive effects on pancreatic cancer, possibly via targeting Gli3.
Collapse
Affiliation(s)
- Yongbiao Ma
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
- Department of Hepatobiliary Surgery, Weifang People's Hospital, Weifang, China
| | - Gaoxue Li
- Department of Gastroenterology, Shouguang People's Hospital, Shouguang, China
| | - Jingxia Hu
- Department of General Surgery, Shouguang People's Hospital, Shouguang, China
| | - Xin Liu
- Department of Medical Oncology, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Baomin Shi
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
- Department of General Surgery, Tongji Hospital of Tongji University, Shanghai, China
| |
Collapse
|
30
|
Yun S, Kim WK, Kwon Y, Jang M, Bauer S, Kim H. Survivin is a novel transcription regulator of KIT and is downregulated by miRNA-494 in gastrointestinal stromal tumors. Int J Cancer 2018; 142:2080-2093. [PMID: 29277888 PMCID: PMC5900938 DOI: 10.1002/ijc.31235] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 11/02/2017] [Accepted: 12/21/2017] [Indexed: 12/30/2022]
Abstract
Gain-of-function mutations of KIT are pathognomonic in sporadic gastrointestinal stromal tumors (GISTs). Several microRNAs have been shown to be dysregulated in GISTs and impact KIT expression. Little is known though on KIT-independent targets of KIT-regulating mRNAs. We sought to investigate how miR-494 inhibits GIST proliferation and to identify novel target gene. We used microarray-based gene expression analyses to identify pathways and target genes affected by miR-494. The expressional relationship between survivin and miR-494 was determined in 35 GIST tissues. Cell proliferation assay, FACS analysis, colony formation assay, promoter assays and chromatin immunoprecipitation (ChiP) were performed to clarify the roles of survivin in GIST progression. Gene expression microarray analysis revealed that miR-494 inhibited GISTs by affecting multiple genes in the cell cycle pathway. Survivin (BIRC5) was a key target of miR-494, and its expression showed an inverse correlation with miR-494 expression in 35 GIST tissues (Pearson's correlation coefficient, r = -0.418, p = 0.012). Downregulation of survivin inhibited proliferation and colony formation, and resulted in cell cycle alteration. Induced survivin overexpression relieved miR-494-mediated inhibition of GIST progression. Targeting PI3K effectively suppressed proliferation of GISTs with downregulation of survivin. Survivin also regulated KIT expression at the transcription level. Immunohistochemical analysis using 113 GISTs revealed that survivin expression was significantly correlated with overall survival of GIST patients (p = 0.004). Our findings indicated that miR-494 synergistically suppressed GISTs by concomitantly targeting survivin and KIT.
Collapse
Affiliation(s)
- SeongJu Yun
- Department of Pathology and Brain Korea 21 PLUS Projects for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Won Kyu Kim
- Department of Pathology and Brain Korea 21 PLUS Projects for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yujin Kwon
- Department of Pathology and Brain Korea 21 PLUS Projects for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mi Jang
- Department of Pathology and Brain Korea 21 PLUS Projects for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sebastian Bauer
- Germany and German Cancer Consortium (DKTK), Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Heidelberg, Germany
| | - Hoguen Kim
- Department of Pathology and Brain Korea 21 PLUS Projects for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
31
|
Diagnostic and Prognostic Biomarkers in ovarian cancer and the potential roles of cancer stem cells – An updated review. Exp Cell Res 2018; 362:1-10. [DOI: 10.1016/j.yexcr.2017.10.018] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 10/20/2017] [Indexed: 01/06/2023]
|
32
|
Infantile hemangioma: factors causing recurrence after propranolol treatment. Pediatr Res 2018; 83:175-182. [PMID: 29019471 DOI: 10.1038/pr.2017.220] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 08/31/2017] [Indexed: 12/21/2022]
Abstract
BackgroundPropranolol is the first-choice treatment for severe infantile hemangioma (IH). However, 10- 30% of lesions relapse after propranolol treatment. The mechanisms underlying IH recurrence after propranolol treatment have not been completely elucidated.MethodsThis study combined an examination of hemodynamic changes with research regarding hemangioma stem cells (hscs) with differentially expressed microRNAs (miRNAs) to identify the factors affecting IH recurrence after propranolol treatment. Hemodynamic changes were monitored in 21 recurrent cases using high-frequency color Doppler ultrasound, and hscs were treated with different concentrations of propranolol. The levels of differentially expressed miRNAs and the activity of related pathways were then compared between 18 recurrent and 20 non-recurrent IH cases.ResultsDuring treatment, lesion depth and vessel density decreased, and the lesion resistance index increased. Obvious lesions and vessel signals were observed in recurrent cases compared with non-recurrent cases. Propranolol effectively inhibited hscs proliferation. Twenty-two differentially expressed miRNAs were found in the recurrent group compared with the non-recurrent group.ConclusionRecurrence may be attributed to a combination of events. Serum biomarkers and drug treatments for IH recurrence must be studied further.
Collapse
|
33
|
Srivastava SK, Ahmad A, Zubair H, Miree O, Singh S, Rocconi RP, Scalici J, Singh AP. MicroRNAs in gynecological cancers: Small molecules with big implications. Cancer Lett 2017; 407:123-138. [PMID: 28549791 PMCID: PMC5601032 DOI: 10.1016/j.canlet.2017.05.011] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/10/2017] [Accepted: 05/15/2017] [Indexed: 12/14/2022]
Abstract
Gynecological cancers (GCs) are often diagnosed at advanced stages, limiting the efficacy of available therapeutic options. Thus, there remains an urgent and unmet need for innovative research for the efficient clinical management of GC patients. Research over past several years has revealed the enormous promise of miRNAs. These small non-coding RNAs can aid in the diagnosis, prognosis and therapy of all major GCs, viz., ovarian cancers, cervical cancers and endometrial cancers. Mechanistic details of the miRNAs-mediated regulation of multiple biological functions are under constant investigation, and a number of miRNAs are now believed to influence growth, proliferation, invasion, metastasis, chemoresistance and the relapse of different GCs. Modulation of tumor microenvironment by miRNAs can possibly explain some of their reported biological effects. miRNA signatures have been proposed as biomarkers for the early detection of GCs, even the various subtypes of individual GCs. miRNA signatures are also being pursued as predictors of response to therapies. This review catalogs the knowledge gained from collective studies, so as to assess the progress made so far. It is time to ponder over the knowledge gained, so that more meaningful pre-clinical and translational studies can be designed to better realize the potential that miRNAs have to offer.
Collapse
Affiliation(s)
- Sanjeev K Srivastava
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| | - Aamir Ahmad
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Haseeb Zubair
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Orlandric Miree
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Seema Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Rodney P Rocconi
- Division of Gynecologic Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Jennifer Scalici
- Division of Gynecologic Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Ajay P Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA.
| |
Collapse
|
34
|
Xu L, Li H, Su L, Lu Q, Liu Z. MicroRNA-455 inhibits cell proliferation and invasion of epithelial ovarian cancer by directly targeting Notch1. Mol Med Rep 2017; 16:9777-9785. [DOI: 10.3892/mmr.2017.7790] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 09/05/2017] [Indexed: 11/05/2022] Open
|
35
|
Yuan W, Wang D, Liu Y, Tian D, Wang Y, Zhang R, Yin L, Deng Z. miR‑494 inhibits cell proliferation and metastasis via targeting of CDK6 in osteosarcoma. Mol Med Rep 2017; 16:8627-8634. [PMID: 28990071 PMCID: PMC5779916 DOI: 10.3892/mmr.2017.7709] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 06/01/2017] [Indexed: 12/25/2022] Open
Abstract
Tumorigenesis is a multistep process involving various cell growth-associated factors. Accumulated evidence indicates that the disordered regulation of microRNAs (miRNAs) contributes to tumorigenesis. However, the detailed mechanism underlying the involvement of miRNAs in oncogenesis remains to be fully elucidated. In the present study, the repressed expression of microRNA (miR)-494 was identified in 18 patients with osteosarcoma (OS) and OS cell lines, compared with corresponding controls. To determine whether deregulated miR-494 exerts tumor-suppressive effects in the development of OS, the effects of miR-494 on cell proliferation and metastasis were evaluated. It was found that the restoration of miR-494 in MG-63 and U2OS cells led to inhibited cell proliferation and attenuated migratory propensity in vitro, determined through analysis using MTT, colony formation and Transwell assays. In addition, overexpression of miR-494 markedly suppressed the tumor volume and weight in vivo. In accordance, the ectopic expression of miR-494 induced cell cycle arrest at the G1/S phase in OS cells. Bioinformatics analysis and luciferase reporter assays were performed to investigate the potential regulatory role of miR-494, the results of which indicated that miR-494 directly targeted cyclin-dependent kinase 6 (CDK6). Of note, the data obtained through reverse transcription-quantitative polymerase chain reaction and western blot analyses suggested that the elevated expression of miR-494 resulted in reduced mRNA and protein expression levels of CDK6. Taken together, these findings indicated that the miR-494/CDK6 axis has a significant tumor-suppressive effect on OS, and maybe a diagnostic and therapeutic target for the treatment of OS.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Du Wang
- Department of Orthopedics, Wuhan Hospital No. 3 and Tongren Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yang Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Dongdong Tian
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yang Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Ranxi Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Liangjun Yin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Zhongliang Deng
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
36
|
Sun X, Xu M, Liu H, Ming K. MicroRNA-219 is downregulated in non-small cell lung cancer and inhibits cell growth and metastasis by targeting HMGA2. Mol Med Rep 2017; 16:3557-3564. [PMID: 28714014 DOI: 10.3892/mmr.2017.7000] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 04/21/2017] [Indexed: 11/05/2022] Open
Abstract
Lung cancer is the most commonly diagnosed cancer and the leading cause of cancer-associated mortality worldwide. Non-small cell lung cancer (NSCLC) is the predominant type of lung cancer, and accounts for ~85% of all lung cancer cases. An increasing number of studies suggest that microRNAs (miRs) may be involved in the regulation of NSCLC carcinogenesis and progression. However, the expression and function of miRNA-219 in NSCLC, and its underlying mechanisms of action, remain unknown. In the present study, miR-219 expression in NSCLC tissues and cell lines was determined using reverse transcription-quantitative polymerase chain reaction. Following transfection with miR-219 mimics, the effects of miR-219 overexpression on NSCLC cell proliferation, migration and invasion were examined. Furthermore, the miR-219 target in NSCLC was investigated. miR-219 was observed to be downregulated in NSCLC tissues and NSCLC cell lines. In addition, miR-219 was demonstrated to function as a tumor suppressor in NSCLC, through inhibiting cell proliferation, migration and invasion in vitro. Furthermore, high mobility group AT-hook 2 (HMGA2) was identified to be a direct target of miR-219 in NSCLC, and downregulation of HMGA2 suppressed NSCLC cell proliferation, migration and invasion in vitro. HMGA2 expression was upregulated in NSCLC tissues, and was inversely correlated with miR-219 expression. In conclusion, miR-219 functions as a tumor suppressor and may be important in inhibiting the growth and metastasis of NSCLC cells via directly targeting HMGA2. Therefore, miR-219 may present a potential novel therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Xiaoping Sun
- Department of Emergency, Yidu Central Hospital of Weifang, Weifang Medical University, Weifang, Shandong 262500, P.R. China
| | - Min Xu
- Department of Emergency, Yidu Central Hospital of Weifang, Weifang Medical University, Weifang, Shandong 262500, P.R. China
| | - Haiyan Liu
- Department of Emergency, Yidu Central Hospital of Weifang, Weifang Medical University, Weifang, Shandong 262500, P.R. China
| | - Kunxiu Ming
- Department of Emergency Medicine, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
| |
Collapse
|
37
|
Yang X, Ruan H, Hu X, Cao A, Song L. miR-381-3p suppresses the proliferation of oral squamous cell carcinoma cells by directly targeting FGFR2. Am J Cancer Res 2017; 7:913-922. [PMID: 28469963 PMCID: PMC5411798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 03/02/2017] [Indexed: 06/07/2023] Open
Abstract
Mutiple microRNAs are implicated in oral squamous cell carcinoma (OSCC), which is characterized by a high rate of proliferation and nodal metastasis. Data from the present study showed that miR-381-3p is significantly underexpressed in both OSCC tissues and cell lines. Overexpression of miR-381-3p led to marked suppression of proliferation and cell cycle progression of OSCC cells and promotion of apoptosis. Notably, fibroblast growth factor receptor 2 (FGFR2) was downregulated by miR-381-3p through direct interactions with its 3' untranslated region. Knockdown of FGFR2 recapitulated the growth suppressive effect of miR-381-3p. Conversely, restoring FGFR2 expression attenuated miR-381-3p-induced effects in OSCC cells. Expression patterns of miR-381-3p and FGFR2 were inversely correlated in OSCC tissues. Our collective results provide novel evidence that miR-381-3p acts as a tumor suppressor in OSCC by directly targeting FGFR2, thereby presenting a promising therapeutic target.
Collapse
Affiliation(s)
- Xiao Yang
- Department of Stomatology, The Second Affiliated Hospital of Nanchang UniversityNanchang, Jiangxi, China
| | - Huibing Ruan
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang, Jiangxi, China
| | - Xi Hu
- Department of Stomatology, The Second Affiliated Hospital of Nanchang UniversityNanchang, Jiangxi, China
| | - Anyi Cao
- Department of Orthodontics, The Affiliated Huizhou Stomatological Hospital of Jinan University & Huizhou Stomatological HospitalHuizhou, Guangdong, China
| | - Li Song
- Department of Stomatology, The Second Affiliated Hospital of Nanchang UniversityNanchang, Jiangxi, China
| |
Collapse
|
38
|
Yang Y, Pan Q, Sun B, Yang R, Fang X, Liu X, Yu X, Zhao Z. miR-29b Targets LPL and TDG Genes and Regulates Apoptosis and Triglyceride Production in MECs. DNA Cell Biol 2016; 35:758-765. [DOI: 10.1089/dna.2016.3443] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Yuwei Yang
- College of Animal Science, Jilin University, Changchun, People's Republic of China
| | - Qiqi Pan
- College of Animal Science, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Boxing Sun
- College of Animal Science, Jilin University, Changchun, People's Republic of China
| | - Runjun Yang
- College of Animal Science, Jilin University, Changchun, People's Republic of China
| | - Xibi Fang
- College of Animal Science, Jilin University, Changchun, People's Republic of China
| | - Xin Liu
- College of Animal Science, Jilin University, Changchun, People's Republic of China
| | - Xianzhong Yu
- Department of Biological Sciences, Clemson University, Clemson, South Carolina
| | - Zhihui Zhao
- College of Animal Science, Jilin University, Changchun, People's Republic of China
| |
Collapse
|
39
|
Han X, Fang Z, Wang H, Jiao R, Zhou J, Fang N. CUL4A functions as an oncogene in ovarian cancer and is directly regulated by miR-494. Biochem Biophys Res Commun 2016; 480:675-681. [PMID: 27983981 DOI: 10.1016/j.bbrc.2016.10.114] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 10/26/2016] [Indexed: 01/22/2023]
Abstract
Cullin 4A (CUL4A), as a well-defined oncogene, has been reported to be upregulated in ovarian cancer clinically. However, the biological functions of CUL4A and the molecular mechanism underlying its upregulation in ovarian cancer remains unknown throughly. Here, we show that expression of CUL4A is significantly higher in ovarian cancer tissues compared to corresponding non-cancerous tissues. Moreover, silencing of CUL4A by siRNA markedly inhibits cell proliferation, invasion and epithelial-mesenchymal transition (EMT). We identified CUL4A as a novel target gene of miR-494. Further investigations showed that miR-494 was remarkably downregulated and correlated with poor prognosis in ovarian cancer. Overexpression of miR-494 inhibited proliferation, migration, invasion and EMT of ovarian cancer cells by directly suppressing CUL4A expression. Therefore, our findings indicate that miR-494/CUL4A axis is important in the control of ovarian cancer tumorigenesis.
Collapse
Affiliation(s)
- Xiaoni Han
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, 330003, Jiangxi Province, China
| | - Ziling Fang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Heng Wang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Rongfang Jiao
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, 330003, Jiangxi Province, China
| | - Jing Zhou
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, 330003, Jiangxi Province, China
| | - Nian Fang
- Department of Gastroenterology, The Third Affiliated Hospital of Nanchang University, Nanchang, 330003, Jiangxi Province, China.
| |
Collapse
|