1
|
Ahram M, Abu Alragheb B, Abushukair H, Bawadi R, Al-Hussaini M. MicroRNAs Associated with Androgen Receptor and Metastasis in Triple-Negative Breast Cancer. Cancers (Basel) 2024; 16:665. [PMID: 38339416 PMCID: PMC10854913 DOI: 10.3390/cancers16030665] [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: 12/11/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
It is crucial to identify novel molecular biomarkers and therapeutic targets for triple-negative breast cancer (TNBC). The androgen receptor (AR) is a regulator of TNBC, acting partially via microRNA molecules (miRNAs). In this study, we used PCR arrays to profile the expression of 84 miRNAs in 24 TNBC tissue samples, which were equally classified according to AR expression and/or metastasis. Several bioinformatics tools were then utilized to determine the potentially affected protein targets and signaling pathways. Seven miRNAs were found to be significantly more highly expressed in association with AR expression, including miR-328-3p and miR-489-3p. Increased expression of miR-205-3p was found to be significantly associated with metastasis. Certain miRNAs were specifically found to be differentially expressed in either metastatic or non-metastatic AR-positive tumors. A gene ontology (GO) analysis indicated biological roles in the regulation of transcription, cellular response to DNA damage, and the transforming growth factor-beta (TGF-beta) signaling pathway. The GO analysis also showed enrichment in kinase and transcription factor activities. The TGF-beta and a number of kinase-dependent pathways were also retrieved using the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. This study offers an understanding of the role of AR in TNBC and further implicates miRNAs in mediating the effects of AR on TNBC.
Collapse
Affiliation(s)
- Mamoun Ahram
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | | | - Hassan Abushukair
- School of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan;
| | - Randa Bawadi
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | - Maysa Al-Hussaini
- Department of Pathology and Laboratory Medicine, King Hussein Cancer Center, Amman 11941, Jordan;
| |
Collapse
|
2
|
MicroRNA-489 Promotes the Apoptosis of Cardiac Muscle Cells in Myocardial Ischemia-Reperfusion Based on Smart Healthcare. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:2538769. [PMID: 35035817 PMCID: PMC8759872 DOI: 10.1155/2022/2538769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/01/2021] [Accepted: 12/09/2021] [Indexed: 01/10/2023]
Abstract
With the development of information technology, the concept of smart healthcare has gradually come to the fore. Smart healthcare uses a new generation of information technologies, such as the Internet of Things (loT), big data, cloud computing, and artificial intelligence, to transform the traditional medical system in an all-around way, making healthcare more efficient, more convenient, and more personalized. miRNAs can regulate the proliferation, differentiation, and apoptosis of human cells. Relevant studies have also shown that miRNAs may play a key role in the occurrence and development of myocardial ischemia-reperfusion injury (MIRI). This study aims to explore the effects of miR-489 in MIRI. In this study, miR-489 expression in a myocardial ischemia-reperfusion animal model and H9C2 cells induced by H/R was detected by qRT-PCR. The release of lactate dehydrogenase (LDH) and the activity of creatine kinase (CK) was detected after miR-489 knockdown in H9C2 cells induced by H/R. The apoptosis of H9C2 cells and animal models were determined by ELISA. The relationship between miR-489 and SPIN1 was verified by a double fluorescence reporter enzyme assay. The expression of the PI3K/AKT pathway-related proteins was detected by Western blot. Experimental results showed that miR-489 was highly expressed in cardiac muscle cells of the animal model and in H9C2 cells induced by H/R of the myocardial infarction group, which was positively associated with the apoptosis of cardiac muscle cells with ischemia-reperfusion. miR-489 knockdown can reduce the apoptosis of cardiac muscle cells caused by ischemia-reperfusion. In downstream targeting studies, it was found that miR-489 promotes the apoptosis of cardiac muscle cells after ischemia-reperfusion by targeting the inhibition of the SPIN1-mediated PI3K/AKT pathway. In conclusion, high expression of miR-489 is associated with increased apoptosis of cardiac muscle cells after ischemia-reperfusion, which can promote the apoptosis after ischemia-reperfusion by targeting the inhibition of the SPIN1-mediated PI3K/AKT pathway. Therefore, miR-489 can be one of the potential therapeutic targets for reducing the apoptosis of cardiac muscle cells after ischemia-reperfusion.
Collapse
|
3
|
Jia T, Wang M, Yan W, Wu W, Shen R. Upregulation of miR-489-3p attenuates cerebral ischemia/reperfusion injury by targeting histone deacetylase 2 (HDAC2). Neuroscience 2021; 484:16-25. [PMID: 34914969 DOI: 10.1016/j.neuroscience.2021.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 11/25/2021] [Accepted: 12/07/2021] [Indexed: 12/31/2022]
Abstract
Cerebral ischemia/reperfusion (I/R) injury is the continuation and deterioration of ischemic injury, and there are no effective treatment strategies for this condition. It has been reported that microRNAs (miRNAs) are considered as potential targets to protect the brain against I/R injury. Previous studies have shown that miR-489-3p plays a vital role in regulating apoptosis of neurons. miR-489-3p is considered as a potential target to protect the brain against I/R injury-induced neuron apoptosis. This study aimed to explore the molecular mechanism of miR-489-3p in protection against cerebral I/R injury. A rat model with cerebral I/R injury was established using the MCAO method. The cell model was constructed using the oxygen‑glucose deprivation (OGD) method. The expression of miR-489-3p was detected by qRT-PCR. The expression of HDAC2 was detected by western blot assay and immunofluorescence assay. Cell apoptosis was evaluated by flow cytometry and TUNEL staining assay. The relationship between miR-489-3p and HDAC2 was determined by bioinformatics analysis and luciferase reporter assay. Rescue experiments were performed to investigate the mechanism of the miR-489-3p/HDAC2 axis. miR-489-39 was significantly downregulated, while HDAC2 was upregulated during cerebral I/R injury both in vitro and in vivo. Upregulation of miR-489-3p obviously attenuated cerebral I/R injury by increasing PC12 cell viability, reducing LDH release, and inhibiting cell apoptosis. HDAC2 was identified as a direct target of miR-489-3p. Silencing of HDAC2 showed a neuroprotective effect against OGD/R injury in vitro. Overexpression of HDAC2 significantly attenuated the protective effects of miR-489-3p mimics on cell injury in vitro. Our results revealed that the upregulation of miR-489-3p attenuated cerebral I/R injury by negatively regulating HDAC2.
Collapse
Affiliation(s)
- Tianxia Jia
- Department of neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of science and Technology, Luoyang City, Henan Province,471003, PR. China
| | - Mengjie Wang
- Department of neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of science and Technology, Luoyang City, Henan Province,471003, PR. China
| | - Wenjun Yan
- Department of neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of science and Technology, Luoyang City, Henan Province,471003, PR. China
| | - Wenjuan Wu
- Department of neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of science and Technology, Luoyang City, Henan Province,471003, PR. China
| | - Ruile Shen
- Department of neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of science and Technology, Luoyang City, Henan Province,471003, PR. China.
| |
Collapse
|
4
|
Zhang D, He Z, Shen Y, Wang J, Liu T, Jiang J. MiR-489-3p Reduced Pancreatic Cancer Proliferation and Metastasis By Targeting PKM2 and LDHA Involving Glycolysis. Front Oncol 2021; 11:651535. [PMID: 34868902 PMCID: PMC8632778 DOI: 10.3389/fonc.2021.651535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 10/14/2021] [Indexed: 01/03/2023] Open
Abstract
Introduction Malignant proliferation and metastasis are some of the causes of high mortality in pancreatic cancer. MicroRNAs have been a hot spot in cancer research and are involved in tumor formation and metabolic stress responses. However, the biology function and underlying mechanism of miRNA regulating pancreatic cancer progress is remained uncleared. Methods RNA-seq analysis the glycolysis associated miRNAs and verified miRNA-489-3p was involving in glycolysis. We used RNA in situ hybridization (ISH) and qRT-PCR to analyze the differential expression of miR-489-3p in pancreatic cancer tissues and adjacent tissues and cell lines. Then the function assay of in vivo and in vitro were used to evaluated the role of miR-489-3p in the proliferation, metastasis and glucose metabolism of pancreatic cancer. Furthermore, dual luciferase reporter and rescue experiments were performed to explore the mechanism underlying in the role of miRNA-489-3p. Results We determined that glycolysis associated miRNA miR-489-3p was downregulated in pancreatic cancer tissues and cell lines. The gain and loos of function experiments confirmed that miR-489-3p could inhibit the proliferation, metastasis and glucose metabolism of pancreatic cancer. Further, we found that miR-489-3p could target regulating LDHA and PKM through the luciferase report experiment. Finally, in vivo experiment confirmed that highly expressed miR-489-3p inhibited the growth of pancreatic cancer. Conclusion In short, this study identified miR-489-3p as a novel therapy target for pancreatic cancer which was involving in the proliferation, metastasis and glycolysis, but its diagnostic value deserves further study.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhiwei He
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yiyi Shen
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jie Wang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao Liu
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jianxin Jiang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
5
|
Tzeng HE, Tang CH, Tsai CH, Chiu CH, Wu MH, Yen Y. ET-1 Promotes Epithelial-Mesenchymal Transition in Oral Squamous Cell Carcinoma Cells via the microRNA-489-3p /TWIST Axis. Onco Targets Ther 2021; 14:5005-5018. [PMID: 34675545 PMCID: PMC8502871 DOI: 10.2147/ott.s294312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Oral squamous cell carcinoma (OSCC) constitutes almost 90% of head and neck malignancies and has a poor prognosis. To improve the efficacy of OSCC therapy, it is of great significance to explore other therapy for OSCC. Endothelin-1 (ET-1), a potent vasoconstrictor peptide, is implicated in cancer pathogenesis. Moreover, ET-1 promotes epithelial-mesenchymal transition (EMT) during the development of human cancers. We further to found that ET-1 exposure induced EMT in human squamous cell carcinoma cell lines SCC4 and SAS, by enhancing the expression of EMT biomarkers N-cadherin and vimentin and reducing E-cadherin expression via upregulation of the transcription factor TWIST. MATERIALS AND METHODS Cell motility was examined by migration, invasion and wound-healing assays. Quantitative real time polymerase chain reaction (q-PCR), and promoter assays confirmed the inhibitory effects of ET-1 on miRNAs expression in oral cancer cells. We demonstrate an intravenous injection model of lung metastasis followed by an advanced method for quantifying metastatic tumor using image analysis software. RESULTS In addition, ET-1/ETAR reduced levels of microRNA-489-3p (miR-489-3p), a transcriptional repressor of TWIST. We have identified a novel bypass mechanism through which ET-1/ETAR are involved in TWIST signaling and downregulate miR-489-3p expression, enabling OSCC cells to acquire the EMT phenotype. Notably, ET-1 knockdown dramatically decreased levels of EMT markers and cell migration potential. CONCLUSION The role of ET-1 in OSCC progression is supported by our findings from an in vivo murine model of OSCC. ET-1 may therefore represent a novel molecular therapeutic target in OSCC metastasis.
Collapse
Affiliation(s)
- Huey-En Tzeng
- Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan
- PhD Program & Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Division of Hematology/Oncology, Department of Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Hui Chiu
- Graduate Program in Department of Exercise Health Science, National Taiwan University of Sport, Taichung, Taiwan
| | - Min-Huan Wu
- Sports Recreation and Health Management Continuing Studies, Tunghai University, Taichung, Taiwan
- Bachelor of Science in Senior Wellness and Sport Science, Tunghai University, Taichung, Taiwan
| | - Yun Yen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Informatics, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
6
|
Metformin Inhibits the Development of Hypopharyngeal Squamous Cell Carcinoma through Circ_0003214-Mediated MiR-489-3p-ADAM10 Pathway. JOURNAL OF ONCOLOGY 2021; 2021:2265475. [PMID: 34335751 PMCID: PMC8295003 DOI: 10.1155/2021/2265475] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/24/2021] [Indexed: 12/15/2022]
Abstract
Purpose This study aims to explore the function of metformin in hypopharyngeal squamous cell carcinoma (HSCC) and the underlying mechanism. Methods Cell viability, colony formation, cell apoptosis, and cell cycle were investigated using cell counting kit-8 assay, colony formation, and flow cytometry assay. Gene expression was detected by quantitative real-time polymerase chain reaction and western blot. The target relationship was validated by dual-luciferase reporter assay or RNA immunoprecipitation assay. An animal study was implemented to clarify the effect of metformin in vivo. Results Metformin suppressed HSCC cell viability and colony formation ability and induced cell cycle arrest and apoptosis, and circ_0003214 overexpression weakened these effects. Circ_0003214 regulated A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) expression via targeting miR-489-3p. Besides, miR-489-3p restoration reversed the role of circ_0003214, and ADAM10 knockdown reversed miR-489-3p inhibition-mediated effect. Moreover, metformin blocked tumor growth via the circ_0003214-miR-489-3p-ADAM10 axis in vivo. Conclusion Metformin inhibits HSCC progression through the circ_0003214/miR-489-3p/ADAM10 pathway.
Collapse
|
7
|
Mao SH, Zhu CH, Nie Y, Yu J, Wang L. Levobupivacaine Induces Ferroptosis by miR-489-3p/SLC7A11 Signaling in Gastric Cancer. Front Pharmacol 2021; 12:681338. [PMID: 34177591 PMCID: PMC8220201 DOI: 10.3389/fphar.2021.681338] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer is one of the most the prevalent malignancies and the therapeutic strategies for patients with gastric cancer remains limited. Local anesthetic levobupivacaine has demonstrated potential anti-cancer property, but its correlation with gastric cancer and ferroptosis is poor understood. Here, we identified the novel function of levobupivacaine in regulating ferroptosis of gastric cancer cells. The treatment of levobupivacaine suppressed gastric cancer cell viabilities and Edu-positive cell proportions. The gastric cancer cell growth was reduced by levobupivacaine in vivo. Moreover, the treatment of levobupivacaine enhanced erastin-induced inhibitory impact on gastric cancer cell viabilities. The levels of Fe2+/iron and lipid ROS were induced by levobupivacaine in erastin and RSL3-stimulated gastric cancer cells. levobupivacaine-upregulated miR-489-3p enhanced ferroptosis of gastric cancer cells by targeting SLC7A11. MiR-489-3p was involved in levobupivacaine-induced ferroptosis of gastric cancer cells. Levobupivacaine/miR-489-3p/SLC7A11 axis attenuates gastric cancer cell proliferation in vitro. Therefore, we concluded that the local anesthetic levobupivacaine induced ferroptosis of gastric cancer cells to repress gastric cancer cell growth by miR-489-3p/SLC7A11 axis.
Collapse
Affiliation(s)
- Shun-Hong Mao
- Department of Anesthesia, Cangzhou Central Hospital, Cangzhou, China
| | - Chun-Hua Zhu
- Department of Anesthesia, Cangzhou Central Hospital, Cangzhou, China
| | - Yu Nie
- Department of Anesthesia, Cangzhou Central Hospital, Cangzhou, China
| | - Jian Yu
- Department of Anesthesia, Cangzhou Central Hospital, Cangzhou, China
| | - Lei Wang
- Department of Anesthesia, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|
8
|
Gao W, Zheng W, Sun Y, Xu T. microRNA-489 negatively modulates RIG-I signaling pathway via targeting TRAF6 in miiuy croaker after poly(I:C) stimulation. FISH & SHELLFISH IMMUNOLOGY 2021; 113:61-68. [PMID: 33785469 DOI: 10.1016/j.fsi.2021.03.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/21/2021] [Accepted: 03/21/2021] [Indexed: 06/12/2023]
Abstract
The innate immune response is first line of host defense against pathogen invasion. However, excessive activation of immune responses may cause autoimmune diseases and excessive inflammation. Retinoic acid-inducible gene I (RIG-I) is an important cytoplasmic pathogen recognition receptor that is activated on virus infection. TNF-receptor-associated factor 6 (TRAF6) plays an essential role in the RIG-I-mediated signaling pathway. MicroRNAs (miRNAs) are noncoding RNAs that are emerging as important regulators of immune responses. In this study, we found that the overexpression of miR-489 mimics and pre-miR-489 significantly suppressed the luciferase activity of the wild-type TRAF6 3'UTR, whereas mutant-type led to a complete abrogation of the negative effect. In addition, we also observed that miR-489 can negatively regulate TRAF6 at the level of translation. More importantly, we demonstrated that miR-489 is a negative regulator of TRAF6 involved in the immune response to poly(I:C) stimulation. These common findings indicated that miR-489 plays a regulatory role in host-virus interactions by targeting TRAF6. Overall, all of the present results provide direct evidence that miR-489 is involved in the regulation of TRAF6 expression in miiuy croaker, which will help to better understand the complex regulatory networks of teleost fish.
Collapse
Affiliation(s)
- Wenya Gao
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Weiwei Zheng
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Yuena Sun
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China; Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, China.
| | - Tianjun Xu
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China; Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, China.
| |
Collapse
|
9
|
Ye Y, Wang P, Zhou F. miR-489-3p inhibits TLR4/NF-κB signaling to prevent inflammation in psoriasis. Exp Ther Med 2021; 22:744. [PMID: 34055060 PMCID: PMC8138277 DOI: 10.3892/etm.2021.10176] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 03/08/2021] [Indexed: 02/07/2023] Open
Abstract
Psoriasis is a chronic inflammatory skin disease whose etiology has not yet been determined. MicroRNAs (miRs) regulate the early stages of psoriasis and are targets for therapeutic intervention. The present study aimed to investigate the functional role of miR-489-3p in psoriasis. The present study first assessed the expression levels of miR-489-3p and Toll-like receptor (TLR)4 mRNA using reverse transcription-quantitative PCR, and also detected the protein expression levels of TLR4 and NF-κB via western blot analysis. TargetScan and miRDB target gene prediction tools were used to confirm the regulation of Toll-like receptor (TLR)4 by miR-489-3p. Moreover, a Cell Counting Kit (CCK)-8 assay was conducted to evaluate cell viability, while cell cycle and colony formation assays were performed to evaluate cell proliferation. Human keratinocytes (HaCaT) were co-transfected with TLR4-small interfering RNA and miR-489-3p-inhibitor plasmids, and analysis of cell proliferation and inflammatory cytokine secretion was performed using CCK-8 assay and ELISA. It was found that miR-489-3p expression was downregulated in patients with psoriasis. Bioinformatics analysis identified that TLR4 was a direct target of miR-489-3p. This was confirmed via luciferase reporter assays in HaCaT cells. The overexpression of miR-489-3p inhibited the TLR4/NF-κB signaling pathway and reduced cell proliferation. TLR4 silencing alleviated the effects of miR-489-3p, and enhanced cell proliferation and inflammatory cytokine secretion. Taken together, these data suggested that miR-489-3p may be a key effector of psoriasis, which promotes inflammatory responses by direct targeting of TLR4. miR-489-3p therefore represents a promising prognostic biomarker and therapeutic target for psoriasis treatment.
Collapse
Affiliation(s)
- Yujian Ye
- Department of Dermatology, The Third People's Hospital of Hangzhou, Hangzhou, Zhejiang 310009, P.R. China
| | - Ping Wang
- Department of Dermatology, The Third People's Hospital of Hangzhou, Hangzhou, Zhejiang 310009, P.R. China
| | - Fangmei Zhou
- College of Medical Technology, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
10
|
Zhang GF, Zhou BS, An XC, An FM, Li SH. LINC01278 is Highly Expressed in Osteosarcoma and Participates in the Development of Tumors by Mediating the miR-134-5p/KRAS Axis. Onco Targets Ther 2021; 14:683-695. [PMID: 33531816 PMCID: PMC7847385 DOI: 10.2147/ott.s265591] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 12/08/2020] [Indexed: 12/18/2022] Open
Abstract
Purpose There is increasing evidence that non-coding RNAs (ncRNAs), including long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), produce a critical regulatory effect on osteosarcoma (OS). LINC01278, as a newly discovered lncRNA, is found to be highly expressed in OS, but its related mechanism remains unclear. This research, therefore, is designed to study the mechanism of LINC01278 in OS and to find potential targets for clinical use. Methods qRT-PCR was applied to determine the relative expression of LINC01278 and analyze its diagnostic value in OS. CCK-8, Transwell and flow cytometry were utilized for the determination of cell proliferation, migration/invasion, and apoptosis. RIP and RNA pull-down experiments were used to verify the targeted binding effect of miR-134-5p and LINC01278. The relationship between miR-134-5p and LINC01278 or KRAS was analyzed using dual luciferase reporter gene. The effects of LINC01278 on tumor growth in nude mice was analyzed by in vivo experiment. Results qRT-PCR showed that LINC01278 increased in OS tissues and serum, indicating poor prognosis. In addition, LINC01278 was also of high value for OS diagnosis. Functional experiments showed that LINC01278 inhibited KRAS-mediated OS cell proliferation and metastasis through miR-134-5p. Finally, the results of an in vivo animal model indicated that LINC01278 promoted OS growth. Conclusion LINC01278 is expressed highly in OS, and patients with high LINC01278 expression have poor prognosis. Moreover, LINC01278 can suppress the proliferation and apoptosis of OS cells through mediating miR-134-5p/KRAS axis, which is expected to become a potential therapeutic target for OS.
Collapse
Affiliation(s)
- Guo-Feng Zhang
- Department of Orthopedics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai 261400, People's Republic of China
| | - Bai-Sui Zhou
- Department of Orthopedics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai 261400, People's Republic of China
| | - Xiao-Chun An
- Department of Orthopedics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai 261400, People's Republic of China
| | - Feng-Min An
- Department of Orthopedics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai 261400, People's Republic of China
| | - Shan-Hui Li
- Department of Orthopedics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai 261400, People's Republic of China
| |
Collapse
|
11
|
Feng W, Li B, Wang J, Zhang H, Liu Y, Xu D, Cheng K, Zhuang J. Long Non-coding RNA LINC00115 Contributes to the Progression of Colorectal Cancer by Targeting miR-489-3p via the PI3K/AKT/mTOR Pathway. Front Genet 2020; 11:567630. [PMID: 33193658 PMCID: PMC7525183 DOI: 10.3389/fgene.2020.567630] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are tumor-related regulators and have been found to be involved in the underlying molecular mechanisms of colorectal cancer (CRC). However, the role of lncRNA LINC00115 during CRC progression is not entirely elucidated. In this study, we discovered that LINC00115 was significantly overexpressed in CRC, and its overexpression predicted poor patient outcomes. Downregulation of LINC00115 markedly inhibited CRC cell proliferation, increased cell apoptosis, and suppressed cell migration and invasion. Moreover, downregulation of LINC00115 led to the inactivation of PI3K/AKT/mTOR signaling. Bioinformatics analysis identified miR-489-3p as a candidate target of LINC00115. Furthermore, we revealed an inverse correlation between LINC00115 and miR-489-3p in CRC tissues. Importantly, by luciferase reporter assay, we found that miR-489-3p might directly target LINC00115, and downregulation of miR-489-3p could rescue the biological effects induced by the absence of LINC0015. In conclusion, our findings demonstrated that LINC00115 serves as an oncogene in CRC metastasis. Deeper understanding of the LINC00115/miR-489-3p axis might provide potential therapeutic targets against CRC metastasis.
Collapse
Affiliation(s)
- Weiyu Feng
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Baodong Li
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Jinbang Wang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Huiliang Zhang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yonggang Liu
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Dongli Xu
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Ke Cheng
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Jing Zhuang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
12
|
Cheng Z, Wang G, Zhu W, Luo C, Guo Z. LEF1-AS1 accelerates tumorigenesis in glioma by sponging miR-489-3p to enhance HIGD1A. Cell Death Dis 2020; 11:690. [PMID: 32826866 PMCID: PMC7442828 DOI: 10.1038/s41419-020-02823-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 11/25/2022]
Abstract
Long non-coding (lncRNA) lymphoid enhancer-binding factor 1 antisense RNA 1 (LEF1-AS1) has been validated to be implicated in manifold cancers, whereas its function in glioma has not been understood thoroughly. Hence, in this study, we tested that LEF1-AS1 expression was significantly upregulated in glioma tissues and cell lines. Besides, knockdown of LEF1-AS1 repressed cell proliferation while activated apoptosis in glioma cells in vitro, and also suppressed tumor growth in vivo. RNA pull-down and luciferase reporter assays affirmed that LEF1-AS1 could bind with miR-489-3p. In addition, miR-489-3p expression was downregulated in glioma cells. Moreover, miR-489-3p depletion partly offset LEF1-AS1 knockdown-mediated function on proliferation and apoptosis. Further, HIGD1A identified as the target gene of miR-489-3p was upregulated in glioma cells. HIGD1A silence could restrict the process of glioma. In rescue assays, upregulation of HIGD1A remedied the inhibitory impacts of LEF1-AS1 silence on glioma cell growth. In summary, our studies corroborated the regulatory mechanism of LEF1-AS1/miR-489-3p/HIGD1A axis in glioma, suggesting that targeting LEF1-AS1 might be a promising method for glioma therapy in the future.
Collapse
Affiliation(s)
- Zhihua Cheng
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639 Zhizaoju Road, 200011, Shanghai, China
| | - Guangyu Wang
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639 Zhizaoju Road, 200011, Shanghai, China
| | - Weiyi Zhu
- Department of Neurosurgery, Shanghai Jing'an District Central Hospital, No. 259 Xikang Road, 200040, Shanghai, China
| | - Cong Luo
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639 Zhizaoju Road, 200011, Shanghai, China
| | - Zhilin Guo
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639 Zhizaoju Road, 200011, Shanghai, China.
| |
Collapse
|
13
|
Czarnecka AM, Synoradzki K, Firlej W, Bartnik E, Sobczuk P, Fiedorowicz M, Grieb P, Rutkowski P. Molecular Biology of Osteosarcoma. Cancers (Basel) 2020; 12:E2130. [PMID: 32751922 PMCID: PMC7463657 DOI: 10.3390/cancers12082130] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/19/2022] Open
Abstract
Osteosarcoma (OS) is the most frequent primary bone cancer in children and adolescents and the third most frequent in adults. Many inherited germline mutations are responsible for syndromes that predispose to osteosarcomas including Li Fraumeni syndrome, retinoblastoma syndrome, Werner syndrome, Bloom syndrome or Diamond-Blackfan anemia. TP53 is the most frequently altered gene in osteosarcoma. Among other genes mutated in more than 10% of OS cases, c-Myc plays a role in OS development and promotes cell invasion by activating MEK-ERK pathways. Several genomic studies showed frequent alterations in the RB gene in pediatric OS patients. Osteosarcoma driver mutations have been reported in NOTCH1, FOS, NF2, WIF1, BRCA2, APC, PTCH1 and PRKAR1A genes. Some miRNAs such as miR-21, -34a, -143, -148a, -195a, -199a-3p and -382 regulate the pathogenic activity of MAPK and PI3K/Akt-signaling pathways in osteosarcoma. CD133+ osteosarcoma cells have been shown to exhibit stem-like gene expression and can be tumor-initiating cells and play a role in metastasis and development of drug resistance. Although currently osteosarcoma treatment is based on adriamycin chemoregimens and surgery, there are several potential targeted therapies in development. First of all, activity and safety of cabozantinib in osteosarcoma were studied, as well as sorafenib and pazopanib. Finally, novel bifunctional molecules, of potential imaging and osteosarcoma targeting applications may be used in the future.
Collapse
Affiliation(s)
- Anna M Czarnecka
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute-Oncology Centre, 02-781 Warsaw, Poland
| | - Kamil Synoradzki
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Wiktoria Firlej
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute-Oncology Centre, 02-781 Warsaw, Poland
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Ewa Bartnik
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, 02-106 Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Pawel Sobczuk
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute-Oncology Centre, 02-781 Warsaw, Poland
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Michal Fiedorowicz
- Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Interinstitute Laboratory of New Diagnostic Applications of MRI, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Pawel Grieb
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute-Oncology Centre, 02-781 Warsaw, Poland
| |
Collapse
|
14
|
Wang J, Zhang JQ, Zhao XL, Lu JY, Weng ZM, Ding ZM, Yang FQ. Circular RNA DHX33 promotes malignant behavior in ccRCC by targeting miR-489-3p/MEK1 axis. Aging (Albany NY) 2020; 12:14885-14896. [PMID: 32717723 PMCID: PMC7425503 DOI: 10.18632/aging.103550] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022]
Abstract
Mounting evidence indicates that circular RNAs modulate the initiation of clear cell renal cell carcinoma (ccRCC). However, their specific roles in the malignancy of ccRCC is understudied. Here, we present a novel circular RNA, circDHX33, that is up-regulated in ccRCC cell lines and tissues. Upregulated circDHX33 in ccRCC patients significantly correlates with advanced TNM stage and metastasis. Suppressing circDHX33 expression inhibits the proliferation and invasion of cultured cells, and suppresses tumor growth in vivo. Mechanistically, we show that circDHX33 promotes ccRCC progression by sponging miR-489-3p and modulating MEK1 expression. In conclusion, our findings suggest that circDHX33 plays a role in promoting ccRCC via the miR-489-3p/MEK1 axis and may serve as a novel therapeutic target for the treatment of ccRCC patients
Collapse
Affiliation(s)
- Jie Wang
- Department of Urology, Ninghai First Hospital, Zhejiang 315600, China.,Department of Urology, Ninghai Hospital, Branch of Shanghai Tenth People's Hospital, Zhejiang 315600, China
| | - Jian-Qiu Zhang
- Department of Urology, Ninghai First Hospital, Zhejiang 315600, China.,Department of Urology, Ninghai Hospital, Branch of Shanghai Tenth People's Hospital, Zhejiang 315600, China
| | - Xiao-Lei Zhao
- Department of Urology, Huaihe Hospital of Henan University, Kaifeng 475000, China
| | - Jing-Yu Lu
- Department of Anesthesia, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Ze-Ming Weng
- Department of Urology, Ninghai First Hospital, Zhejiang 315600, China.,Department of Urology, Ninghai Hospital, Branch of Shanghai Tenth People's Hospital, Zhejiang 315600, China
| | - Zhen-Min Ding
- Department of Anesthesia, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Feng-Qiang Yang
- Department of Urology, Ninghai First Hospital, Zhejiang 315600, China.,Department of Urology, Ninghai Hospital, Branch of Shanghai Tenth People's Hospital, Zhejiang 315600, China.,Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| |
Collapse
|
15
|
Sun D, Li T, Xin H, An J, Yang J, Lin J, Meng X, Wang B, Ozaki T, Yu M, Zhu Y. miR-489-3p inhibits proliferation and migration of bladder cancer cells through downregulation of histone deacetylase 2. Oncol Lett 2020; 20:8. [PMID: 32774482 PMCID: PMC7405606 DOI: 10.3892/ol.2020.11869] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/18/2020] [Indexed: 12/24/2022] Open
Abstract
Since human bladder cancer (BC) is a common malignancy of the urinary system with poor prognosis, it is crucial to clarify the molecular mechanisms of BC development and progression. To the best of our knowledge, the current study demonstrated for the first time that miR-489-3p suppressed BC cell-derived tumor growth in vivo via the downregulation of histone deacetylase 2 (HDAC2). According to the results, expression levels of miR-489-3p were lower in BC tissues compared with corresponding normal tissues. Expression of miR-489-3p mimics in BC-derived T24 and 5637 cells resulted in a significant reduction in proliferation and migration rates. Furthermore, bioinformatics analyses indicated that HDAC2 may be a potential downstream target of miR-489-3p. In contrast to miR-489-3p, HDAC2 was expressed at higher levels in BC tissues compared with corresponding normal tissues. Additionally, small interfering RNA-mediated knockdown of HDAC2 caused a marked decrease in the proliferation and migration rates of T24 and 5637 cells. Consistent with these observations, expression of miR-489-3p mimics attenuated the growth of xenograft tumors arising from T24 cells and resulted in HDAC2 downregulation. In conclusion, the results of the current study indicated that the miR-489-3p/HDAC2 axis serves a role in the development and/or the progression of BC and may be a potential molecular target for the development of a novel strategy to treat patients with BC.
Collapse
Affiliation(s)
- Dan Sun
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Tianren Li
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Haotian Xin
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jun An
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jieping Yang
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jiaxing Lin
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xin Meng
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Biao Wang
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Toshinori Ozaki
- Department of DNA Damage Signaling, Research Center, The 5th Hospital of Xiamen, Xiamen, Fujian 361101, P.R. China
| | - Meng Yu
- Key Laboratory of Transgenetic Animal Research, Department of Laboratory Animal Science, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Yuyan Zhu
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
16
|
Li C, Gao Q, Wang M, Xin H. LncRNA SNHG1 contributes to the regulation of acute myeloid leukemia cell growth by modulating miR-489-3p/SOX12/Wnt/β-catenin signaling. J Cell Physiol 2020; 236:653-663. [PMID: 32592199 DOI: 10.1002/jcp.29892] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 04/28/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022]
Abstract
The long noncoding RNA (lncRNA) small nucleolar RNA host gene 1 (SNHG1) is a critical regulator for the development and progression of multiple tumors. Yet, the role of SNHG1 in acute myeloid leukemia (AML) is unknown. The present study demonstrated that SNHG1 expression was upregulated in AML. SNHG1 silencing markedly repressed AML cell growth, whereas SNHG1 overexpression had the opposite effect. MicroRNA-489-3p (miR-489-3p) was identified as a SNHG1-targeting miRNA. SNHG1 knockdown increased miR-489-3p expression. Low expression of miR-489-3p was correlated with high expression of SNHG1 in AML tissues. miR-489-3p overexpression restricted AML cell growth, and SRY-related high-mobility-group box 12 (SOX12) was identified as a miR-489-3p-targeting gene. SNHG1 inhibition or miR-489-3p overexpression inactivated Wnt/β-catenin signaling through downregulation of SOX12. SOX12 overexpression partially reversed the SNHG1 knockdown- or miR-489-3p overexpression-mediated effects. Taken together, these data indicate that suppression of SNHG1 downregulates AML cell growth by inactivating SOX12/Wnt/β-catenin signaling via upregulating miR-489-3p.
Collapse
Affiliation(s)
- Chengliang Li
- Department of Hematology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Qiuying Gao
- Department of Haematology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Minjuan Wang
- Department of General Practice, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Hong Xin
- Department of Cardiovasology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| |
Collapse
|
17
|
Parafioriti A, Cifola I, Gissi C, Pinatel E, Vilardo L, Armiraglio E, Di Bernardo A, Daolio PA, Felsani A, D’Agnano I, Berardi AC. Expression profiling of microRNAs and isomiRs in conventional central chondrosarcoma. Cell Death Discov 2020; 6:46. [PMID: 32566253 PMCID: PMC7287106 DOI: 10.1038/s41420-020-0282-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 05/05/2020] [Accepted: 05/16/2020] [Indexed: 12/24/2022] Open
Abstract
Conventional central chondrosarcoma (CCC) is a malignant bone tumor that is characterized by the production of chondroid tissue. Since radiation therapy and chemotherapy have limited effects on CCC, treatment of most patients depends on surgical resection. This study aimed to identify the expression profiles of microRNAs (miRNAs) and isomiRs in CCC tissues to highlight their possible participation to the regulation of pathways critical for the formation and growth of this type of tumor. Our study analyzed miRNAs and isomiRs from Grade I (GI), Grade II (GII), and Grade III (GIII) histologically validated CCC tissue samples. While the different histological grades shared a similar expression profile for the top abundant miRNAs, we found several microRNAs and isomiRs showing a strong different modulation in GII + GIII vs GI grade samples and their involvement in tumor biology could be consistently hypothesized. We then in silico validated these differently expressed miRNAs in a larger chondrosarcoma public dataset and confirmed the expression trend for 17 out of 34 miRNAs. Our results clearly suggests that the contribution of miRNA deregulation, and their targeted pathways, to the progression of CCC could be relevant and strongly indicates that when studying miRNA deregulation in tumors, not only the canonical miRNAs, but the whole set of corresponding isomiRs should be taken in account. Improving understanding of the precise roles of miRNAs and isomiRs over the course of central chondrosarcoma progression could help identifying possible targets for precision medicine therapeutic intervention.
Collapse
Affiliation(s)
- Antonina Parafioriti
- Pathology Department, Azienda Socio Sanitaria Territoriale Gaetano Pini, Milan, Italy
| | - Ingrid Cifola
- Institute for Biomedical Technologies (ITB), CNR, Segrate, Italy
| | - Clarissa Gissi
- U.O.C. of Immunohaematology and Transfusion Medicine, Laboratory of Stem Cells, Spirito Santo Hospital, Pescara, Italy
| | - Eva Pinatel
- Institute for Biomedical Technologies (ITB), CNR, Segrate, Italy
| | - Laura Vilardo
- Institute for Biomedical Technologies (ITB), CNR, Segrate, Italy
| | - Elisabetta Armiraglio
- Pathology Department, Azienda Socio Sanitaria Territoriale Gaetano Pini, Milan, Italy
| | - Andrea Di Bernardo
- Pathology Department, Azienda Socio Sanitaria Territoriale Gaetano Pini, Milan, Italy
| | | | - Armando Felsani
- Institute of Biochemistry and Cell Biology (IBBC), CNR, Monterotondo, Italy
- Genomnia Srl, Bresso, Italy
| | - Igea D’Agnano
- Institute for Biomedical Technologies (ITB), CNR, Segrate, Italy
| | - Anna Concetta Berardi
- U.O.C. of Immunohaematology and Transfusion Medicine, Laboratory of Stem Cells, Spirito Santo Hospital, Pescara, Italy
| |
Collapse
|
18
|
Zheng B, Chen T. MiR-489-3p inhibits cell proliferation, migration, and invasion, and induces apoptosis, by targeting the BDNF-mediated PI3K/AKT pathway in glioblastoma. Open Life Sci 2020; 15:274-283. [PMID: 33817216 PMCID: PMC7874546 DOI: 10.1515/biol-2020-0024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/09/2019] [Accepted: 01/03/2020] [Indexed: 12/15/2022] Open
Abstract
Among astrocyte tumors, glioblastoma (GBM) is the most malignant glioma, highly aggressive and invasive, with extremely poor prognosis. Previous research has reported that microRNAs (miRNAs) participate in the progression of many cancers. Thus, this study aimed to explore the role and the underlying mechanisms of microRNA (miR)-489-3p in GBM progression. The expression of miR-489-3p and brain-derived neurotrophic factor (BDNF) mRNA was measured by quantitative real-time polymerase chain reaction. Western blot analysis was used to detect BDNF protein and the PI3K/AKT pathway-related protein. Cell proliferation, apoptosis, migration, and invasion were analyzed using CKK-8 assay, flow cytometry, and transwell assay, respectively. The interaction between BDNF and miR-489-3p was explored by luciferase reporter assay and RNA immunoprecipitation (RIP) assay. MiR-489-3p was down-regulated and BDNF was up-regulated in GBM tissues and cells. MiR-489-3p re-expression or BDNF knockdown inhibited GBM cell proliferation, migration, and invasion, and promoted apoptosis. BDNF was a target of miR-489-3p, and BDNF up-regulation reversed the effects of miR-489-3p on GBM cells. The protein levels of p-AKT and p-PI3K were notably reduced in GBM cells by overexpression of miR-489-3p, but were rescued following BDNF up-regulation. Therefore, miR-489-3p inhibited proliferation, migration, and invasion, and induced apoptosis, by targeting the BDNF-mediated PI3K/AKT pathway in GBM, providing new strategies for clinical treatment of GBM.
Collapse
Affiliation(s)
- Bo Zheng
- Department of Neurosurgery, Jingzhou Central Hospital, Hubei Province, Jingzhou, 434020, China
| | - Tao Chen
- Department of Neurosurgery, Jingzhou Central Hospital, Hubei Province, Jingzhou, 434020, China
| |
Collapse
|
19
|
Boresowicz J, Kober P, Rusetska N, Maksymowicz M, Paziewska A, Dąbrowska M, Zeber-Lubecka N, Kunicki J, Bonicki W, Ostrowski J, Siedlecki JA, Bujko M. DNA Methylation Influences miRNA Expression in Gonadotroph Pituitary Tumors. Life (Basel) 2020; 10:E59. [PMID: 32413978 PMCID: PMC7281098 DOI: 10.3390/life10050059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/30/2022] Open
Abstract
microRNAs are involved in pathogenesis of cancer. DNA methylation plays a role in transcription of miRNA-encoding genes and may contribute to changed miRNA expression in tumors. This issue was not investigated in pituitary neuroendocrine tumors (PitNETs) previously. DNA methylation patterns, assessed with HumanMethylation450K arrays in 34 PitNETs and five normal pituitaries, were used to determine differentially methylated CpGs located at miRNA genes. It showed aberrant methylation in regions encoding for 131 miRNAs. DNA methylation data and matched miRNA expression profiles, determined with next-generation sequencing (NGS) of small RNAs, were correlated in 15 PitNETs. This showed relationship between methylation and expression levels for 12 miRNAs. DNA methylation and expression levels of three of them (MIR145, MIR21, and MIR184) were determined in the independent group of 80 tumors with pyrosequencing and qRT-PCR and results confirmed both aberrant methylation in PitNETs and correlation between methylation and expression. Additionally, in silico target prediction was combined with analysis of established miRNA profiles and matched mRNA expression pattern, assessed with amplicon-based NGS to indicate putative target genes of epigenetically deregulated miRNAs. This study reveals aberrant DNA methylation in miRNA-encoding genes in gonadotroph PitNETs. Methylation changes affect expression level of miRNAs that regulate putative target genes with tumorigenesis-relevant functions.
Collapse
Affiliation(s)
- Joanna Boresowicz
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (J.B.); (P.K.); (N.R.); (J.A.S.)
| | - Paulina Kober
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (J.B.); (P.K.); (N.R.); (J.A.S.)
| | - Natalia Rusetska
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (J.B.); (P.K.); (N.R.); (J.A.S.)
| | - Maria Maksymowicz
- Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| | - Agnieszka Paziewska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.P.); (M.D.); (J.O.)
- Department of Gastroenterology, Hepatology and Clinical Oncology, Medical Center for Postgraduate Education, 01-813 Warsaw, Poland;
| | - Michalina Dąbrowska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.P.); (M.D.); (J.O.)
| | - Natalia Zeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Medical Center for Postgraduate Education, 01-813 Warsaw, Poland;
| | - Jacek Kunicki
- Department of Neurosurgery, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (J.K.); (W.B.)
| | - Wiesław Bonicki
- Department of Neurosurgery, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (J.K.); (W.B.)
| | - Jerzy Ostrowski
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.P.); (M.D.); (J.O.)
- Department of Gastroenterology, Hepatology and Clinical Oncology, Medical Center for Postgraduate Education, 01-813 Warsaw, Poland;
| | - Janusz A. Siedlecki
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (J.B.); (P.K.); (N.R.); (J.A.S.)
| | - Mateusz Bujko
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (J.B.); (P.K.); (N.R.); (J.A.S.)
| |
Collapse
|
20
|
Bai P, Li W, Wan Z, Xiao Y, Xiao W, Wang X, Wu Z, Zhang K, Wang Y, Chen B, Xing J, Wang T. miR-489-3p Inhibits Prostate Cancer Progression by Targeting DLX1. Cancer Manag Res 2020; 12:2719-2729. [PMID: 32368149 PMCID: PMC7185642 DOI: 10.2147/cmar.s239796] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/31/2020] [Indexed: 01/26/2023] Open
Abstract
Purpose Prostate cancer (PCa) is the third most common cancer in men and the second leading cause of cancer-related death in men. DLX1 belongs to the DLX homeobox family and exhibits antitumor activity in many kinds of tumors. MicroRNAs (miRNAs) play important roles in the progression of cancer. However, whether miRNAs affect the development of PCa by targeting DLX1 has not been determined. In this study, we aimed to investigate the role of miR-489-3p in the regulation of DLX1 expression and PCa progression and to provide a potential therapeutic target for PCa treatment. Methods and Materials The Cancer Genome Atlas database was used to analyze the divergent expression of DLX1 in carcinomas and adjacent normal tissues. The expression level of DLX1 in malignant and normal prostate cells was also measured using RT-qPCR and Western blotting. A dual-luciferase reporter assay was performed to determine whether miR-489-3p directly targets DLX1. We transfected 22Rv1 and DU145 cells with miR-489-3p mimics to overexpress miR-489-3p and then evaluated its effect on cellular function. MTT, EdU, colony formation and cell cycle assays were used to evaluate cell growth. JC-1 and ROS assays with flow cytometry were performed to indirectly analyze apoptosis. Transwell assays were conducted to investigate metastasis. Results The expression level of DLX1 was upregulated in both PCa tissues and cell lines. MiR-489-3p directly targeted DLX1 and downregulated its expression. Overexpression of miR-489-3p significantly suppressed cell growth. MiR-489-3p induced apoptosis through mitochondrial function impairment. Overexpression of miR-489-3p also inhibited cell migration and invasion. DLX1 overexpression reversed the above effects induced by miR-489-3p. Conclusion We identified the involvement of the miR-489-3p/DLX1 pathway in PCa for the first time. In this pathway, miR-489-3p acts as a tumor suppressor by negatively regulating the expression of DLX1. MiR-489-3p may be a potential therapeutic target for PCa treatment.
Collapse
Affiliation(s)
- Peide Bai
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361003, People's Republic of China
| | - Wei Li
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361003, People's Republic of China
| | - Zhenghua Wan
- Xiang'an Branch, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361101, People's Republic of China
| | - Yujuan Xiao
- Department of Pediatrics, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361003, People's Republic of China
| | - Wen Xiao
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361003, People's Republic of China
| | - Xuegang Wang
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361003, People's Republic of China
| | - Zhun Wu
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361003, People's Republic of China
| | - Kaiyan Zhang
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361003, People's Republic of China
| | - Yongfeng Wang
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361003, People's Republic of China
| | - Bin Chen
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361003, People's Republic of China
| | - Jinchun Xing
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361003, People's Republic of China
| | - Tao Wang
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361003, People's Republic of China
| |
Collapse
|
21
|
Ying X, Zhang W, Fang M, Wang C, Han L, Yang C. LncRNA SNHG5 regulates SOX4 expression through competitive binding to miR-489-3p in acute myeloid leukemia. Inflamm Res 2020; 69:607-618. [PMID: 32266420 DOI: 10.1007/s00011-020-01345-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 03/01/2020] [Accepted: 03/30/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES Currently, lncRNA plays an important role in the occurrence and development of acute myeloid leukemia (AML), including SNHG5. However, the role and mechanism of SNHG5 in AML remains unclear. In this study, we explored the regulatory mechanism of SNHG5 in the development of AML. METHODS AND RESULTS QRT-PCR was used to investigate the expression of SNHG5, miR-489-3p, and SOX. The proliferation and apoptosis of AML cells were analyzed by cell transfection, cell counting kit-8 (CCK8), and flow cytometric analysis. Moreover, the expression analysis of marker proteins was detected by western blot. Through luciferase activity assay, RNA pull-down, and RNA-binding protein immunoprecipitation (RIP), we proved that SNHG5 could bind miR-489-3p and SOX4 which might be the target gene of miR-489-3p. RESULTS We first found that SNHG5 was up-regulated in both AML patient bone marrow samples and various AML cell lines. Second, we found that knockdown of SNHG5 inhibited proliferation of AML cells and promoted apoptosis. It was found that SNHG5 could bind miR-489-3p, and the relative expression of SNHG5 was negatively correlated with miR-489-3p. Further results suggested that SOX4 might be the target gene of miR-489-3p. Finally, our experimental data indicated that knockdown of SNHG5 could reduce the tumor volume and down-regulated SOX4 levels in vivo. CONCLUSIONS Our results demonstrated that SNHG5 affected the expression of SOX4 through binding miR-489-3p to regulate proliferation and apoptosis of AML, which might act as a prospective prognostic biological marker and a promising therapeutic target for AML.
Collapse
Affiliation(s)
- Xiaoyang Ying
- Department of Clinical Hematology, Affiliated No. 2 Hospital School of Medicine, Xi'an Jiaotong University, Xi'an Jiaotong University West Five Road, No 157, Xi'an, 710004, People's Republic of China.,Department of Hematology, Affiliated Zhongshan Hospital of Dalian University, Liaoning, Dalian, 116001, China
| | - Wanggang Zhang
- Department of Clinical Hematology, Affiliated No. 2 Hospital School of Medicine, Xi'an Jiaotong University, Xi'an Jiaotong University West Five Road, No 157, Xi'an, 710004, People's Republic of China.
| | - Meiyun Fang
- Department of Hematology, Affiliated Zhongshan Hospital of Dalian University, Liaoning, Dalian, 116001, China
| | - Chenchen Wang
- Department of Hematology, Affiliated Zhongshan Hospital of Dalian University, Liaoning, Dalian, 116001, China
| | - Li Han
- Department of Hematology, Affiliated Zhongshan Hospital of Dalian University, Liaoning, Dalian, 116001, China
| | - Chenmeng Yang
- Department of Hematology, Affiliated Zhongshan Hospital of Dalian University, Liaoning, Dalian, 116001, China
| |
Collapse
|
22
|
He P, Ding J. EWS promotes cell proliferation and inhibits cell apoptosis by regulating miR-199a-5p/Sox2 axis in osteosarcoma. Biotechnol Lett 2020; 42:1263-1274. [PMID: 32236759 DOI: 10.1007/s10529-020-02859-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/06/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Osteosarcoma is one of the most common malignant bone tumors which mainly occurs in children and adolescents. It is characterized by high malignancy and high metastasis rate, resulting in high mortality and disability. Accumulating studies have validated that long noncoding RNAs (lncRNAs) exerted vital roles in multiple cancer progression by regulating the expression of specific genes. This work aimed to explore the potential molecular mechanism of EWS in osteosarcoma. RESULTS In this study, we discovered that both EWS and Sox2 were highly expressed in osteosarcoma tissue samples. In addition, the expression of EWS was positively associated with Sox2 level. We conducted a series of functional assays and observed that Sox2 overexpression could significantly overturned the enhancement of cell proliferation and the decline of cell apoptosis induced by EWS knockdown in osteosarcoma. Moreover, we found a key upstream regulatory gene of Sox2: miR-199a-5p. CONCLUSIONS Through molecular biology studies and rescue assays, we further demonstrated that EWS promotes tumor growth through the miR-199a-5p/Sox2 signaling axis in osteosarcoma. These findings may provide an important theoretical basis for the clinical diagnosis and treatment of osteosarcoma.
Collapse
Affiliation(s)
- Peng He
- Department of Orthopedics, XD Group Hospital, Xi'an, 710077, Shaanxi, China
| | - Junjie Ding
- Department of Orthopedics, Yan'an People's Hospital, No. 57 Qilipu Street, Baota District, Yan'an, 716000, Shaanxi, China.
| |
Collapse
|
23
|
Zhan H, Tu S, Zhang F, Shao A, Lin J. MicroRNAs and Long Non-coding RNAs in c-Met-Regulated Cancers. Front Cell Dev Biol 2020; 8:145. [PMID: 32219093 PMCID: PMC7078111 DOI: 10.3389/fcell.2020.00145] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/21/2020] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are components of many signaling pathways associated with tumor aggressiveness and cancer metastasis. Some lncRNAs are classified as competitive endogenous RNAs (ceRNAs) that bind to specific miRNAs to prevent interaction with target mRNAs. Studies have shown that the hepatocyte growth factor/mesenchymal-epithelial transition factor (HGF/c-Met) pathway is involved in physiological and pathological processes such as cell growth, angiogenesis, and embryogenesis. Overexpression of c-Met can lead to sustained activation of downstream signals, resulting in carcinogenesis, metastasis, and resistance to targeted therapies. In this review, we evaluated the effects of anti-oncogenic and oncogenic non-coding RNAs (ncRNAs) on c-Met, and the interactions among lncRNAs, miRNAs, and c-Met in cancer using clinical and tissue chromatin immunoprecipition (ChIP) analysis data. We summarized current knowledge of the mechanisms and effects of the lncRNAs/miR-34a/c-Met axis in various tumor types, and evaluated the potential therapeutic value of lncRNAs and/or miRNAs targeted to c-Met on drug-resistance. Furthermore, we discussed the functions of lncRNAs and miRNAs in c-Met-related carcinogenesis and potential therapeutic strategies.
Collapse
Affiliation(s)
- Hong Zhan
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Tu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Zhang
- School of Medicine, Zhejiang University Hangzhou, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Lin
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
24
|
Liu X, Sun R, Chen J, Liu L, Cui X, Shen S, Cui G, Ren Z, Yu Z. Crosstalk Mechanisms Between HGF/c-Met Axis and ncRNAs in Malignancy. Front Cell Dev Biol 2020; 8:23. [PMID: 32083078 PMCID: PMC7004951 DOI: 10.3389/fcell.2020.00023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/13/2020] [Indexed: 12/24/2022] Open
Abstract
Several lines of evidence have confirmed the magnitude of crosstalk between HGF/c-Met axis (hepatocyte growth factor and its high-affinity receptor c-mesenchymal-epithelial transition factor) and non-coding RNAs (ncRNAs) in tumorigenesis. Through activating canonical or non-canonical signaling pathways, the HGF/c-Met axis mediates a range of oncogenic processes such as cell proliferation, invasion, apoptosis, and angiogenesis and is increasingly becoming a promising target for cancer therapy. Meanwhile, ncRNAs are a cluster of functional RNA molecules that perform their biological roles at the RNA level and are essential regulators of gene expression. The expression of ncRNAs is cell/tissue/tumor-specific, which makes them excellent candidates for cancer research. Many studies have revealed that ncRNAs play a crucial role in cancer initiation and progression by regulating different downstream genes or signal transduction pathways, including HGF/c-Met axis. In this review, we discuss the regulatory association between ncRNAs and the HGF/c-Met axis by providing a comprehensive understanding of their potential mechanisms and roles in cancer development. These findings could reveal their possible clinical applications as biomarkers for therapeutic interventions.
Collapse
Affiliation(s)
- Xin Liu
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ranran Sun
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianan Chen
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liwen Liu
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xichun Cui
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shen Shen
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guangying Cui
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhigang Ren
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zujiang Yu
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
25
|
Dao R, Wudu M, Hui L, Jiang J, Xu Y, Ren H, Qiu X. Knockdown of lncRNA MIR503HG suppresses proliferation and promotes apoptosis of non-small cell lung cancer cells by regulating miR-489-3p and miR-625-5p. Pathol Res Pract 2020; 216:152823. [PMID: 31983569 DOI: 10.1016/j.prp.2020.152823] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/16/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022]
Abstract
The long noncoding RNA (lncRNA) MIR503HG has been shown to play an important role in cancer development. The aim of the present study was to investigate the potential roles of MIR503HG in the proliferation and apoptosis of non-small cell lung cancer cell (NSCLC). We used short hairpin RNA (shRNA) against MIR503HG to knock down and vector containing full length of MIR503HG to overexpress MIR503HG in NSCLC cells. The expression of MIR503HG in NSCLC tissues and cells was detected and the effects of MIR503HG on the cell proliferation and apoptosis were determined. Results showed that the expression of MIR503HG was significantly upregulated in NSCLC tissues compared with adjacent tissues. We found that downregulation of MIR503HG could clearly suppressed cell proliferation and cell cycle progression. Moreover, MIR503HG knockdown also promoted apoptosis of NSCLC cells. As expected, overexpression of MIR503HG significantly promoted cell proliferation and inhibited cell apoptosis in NSCLC NCI-H1975 cells. We predicted and verified miR-489-3p and miR-625-5p as the direct targets of MIR503HG by bioinformatics analysis and luciferase reporter assay. Mechanically, MIR503HG negatively regulated miR-489-3p and miR-625-5p expressions in NSCLC cells. Moreover, downregulation of miR-489-3p and miR-625-5p weaken the decreased cell proliferation and increased apoptosis of A549 cells after MIR503HG knocking down. In conclusion, knockdown of MIR503HG suppressed proliferation and promoted apoptosis of NSCLC cells through regulating miR-489-3p and miR-625-5p. Our findings of this study suggested that MIR503HG could be a potential therapeutic target for NSCLC development.
Collapse
Affiliation(s)
- Runa Dao
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China.
| | - Muli Wudu
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China.
| | - Linping Hui
- Department of Pathology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, People's Republic of China.
| | - Jun Jiang
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China.
| | - Yitong Xu
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China.
| | - Hongjiu Ren
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China.
| | - Xueshan Qiu
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China.
| |
Collapse
|
26
|
Zhang J, Chou X, Zhuang M, Zhu C, Hu Y, Cheng D, Liu Z. LINC00657 activates PD-L1 to promote osteosarcoma metastasis via miR-106a. J Cell Biochem 2020; 121:4188-4195. [PMID: 31898338 DOI: 10.1002/jcb.29574] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/11/2019] [Indexed: 01/14/2023]
Abstract
Osteosarcoma (OS) cells are one of the primary cancer-related causes of death around the world. Long noncoding RNAs are key for OS progression; however, the detailed molecular mechanism remains unknown. LINC00657, miR-106a, and programmed death ligand-1 (PD-L1) genes expression were detected by reverse transcription-quantitative PCR (RT-qPCR) and Western blot approaches. Invasion and lymphangiogenesis were studied using transwell invasion assay and lymphatic vessel formation assay, respectively. We used bioinformatic analyses to identify putative targets of LINC00657 and miR-106a. Luciferase activity was measured by dual-luciferase reporter assay. PD-L1 protein levels were examined by flow cytometry experiments. LINC00657 knockdown attenuates cell invasion and tumor growth of MG63 and lymphatic vessel formation. miR-106a directly binds LINC00657 and they regulate each other. Furthermore, miR-106a inhibitor strikingly enhanced lymphatic vessel formation and invasion of shLINC00657 MG63 cells. miR-106a mimic directly targeted and downregulated PD-L1. PD-L1 overexpression largely rescued miR-106a mimic-modulated OS cell metastasis. LINC00657 and PD-L1 were upregulated in clinical OS tumors compared to normal tissues. Lower expression levels of LINC00657 and PD-L1 were closely associated with higher overall survival of patients with OS. Here, we suggest a novel mechanism for LINC00657-regulated metastasis of OS cells by regulating the miR-106a/PD-L1 axis. Our conclusions facilitate the development of therapeutical approaches by targeting LINC00657.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Spine Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xubin Chou
- Department of Spine Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ming Zhuang
- Department of Spine Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Chenlei Zhu
- Department of Spine Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yong Hu
- Department of Spine Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Dong Cheng
- Department of Spine Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zhiwei Liu
- Department of Spine Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
27
|
Viera GM, Salomao KB, de Sousa GR, Baroni M, Delsin LEA, Pezuk JA, Brassesco MS. miRNA signatures in childhood sarcomas and their clinical implications. Clin Transl Oncol 2019; 21:1583-1623. [PMID: 30949930 DOI: 10.1007/s12094-019-02104-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/27/2019] [Indexed: 02/06/2023]
Abstract
Progresses in multimodal treatments have significantly improved the outcomes for childhood cancer. Nonetheless, for about one-third of patients with Ewing sarcoma, rhabdomyosarcoma, or osteosarcoma steady remission has remained intangible. Thus, new biomarkers to improve early diagnosis and the development of precision-targeted medicine remain imperative. Over the last decade, remarkable progress has been made in the basic understanding of miRNAs function and in interpreting the contribution of their dysregulation to cancer development and progression. On this basis, this review focuses on what has been learned about the pivotal roles of miRNAs in the regulation of key genes implicated in childhood sarcomas.
Collapse
Affiliation(s)
- G M Viera
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - K B Salomao
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - G R de Sousa
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - M Baroni
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - L E A Delsin
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - J A Pezuk
- Anhanguera University of Sao Paulo, UNIAN/SP, Sao Paulo, Brasil
| | - M S Brassesco
- Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brasil.
- Departamento de Biologia, FFCLRP-USP, Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirao Preto, SP, CEP 14040-901, Brazil.
| |
Collapse
|
28
|
Wang Y, Lin S, Chen L, Qiu H, Wang J. MicroRNA-489 suppresses osteosarcoma invasion, migration and epithelial-to-mesenchymal transition by directly targeting NAA10. MINERVA ENDOCRINOL 2019; 45:150-153. [PMID: 31738038 DOI: 10.23736/s0391-1977.19.03075-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Yuanji Wang
- Department of Orthopedics, The People's Hospital of Rizhao, Rizhao, China -
| | - Shunhua Lin
- Department of Orthopedics, The People's Hospital of Rizhao, Rizhao, China
| | - Li Chen
- Department of Orthopedics, The People's Hospital of Rizhao, Rizhao, China
| | - Hongwei Qiu
- Department of Orthopedics, The People's Hospital of Rizhao, Rizhao, China
| | - Junxiang Wang
- Department of Orthopedics, The People's Hospital of Rizhao, Rizhao, China
| |
Collapse
|
29
|
Chen L, Chen L, Qin Z, Lei J, Ye S, Zeng K, Wang H, Ying M, Gao J, Zeng S, Yu L. Upregulation of miR-489-3p and miR-630 inhibits oxaliplatin uptake in renal cell carcinoma by targeting OCT2. Acta Pharm Sin B 2019; 9:1008-1020. [PMID: 31649850 PMCID: PMC6804444 DOI: 10.1016/j.apsb.2019.01.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 11/04/2018] [Accepted: 11/28/2018] [Indexed: 01/20/2023] Open
Abstract
Renal cell carcinoma (RCC) is one of the most common malignant tumors affecting the urogenital system, accounting for 90% of renal malignancies. Traditional chemotherapy options are often the front-line choice of regimen in the treatment of patients with RCC, but responses may be modest or limited due to resistance of the tumor to anticarcinogen. Downregulated expression of organic cation transporter OCT2 is a possible mechanism underlying oxaliplatin resistance in RCC treatment. In this study, we observed that miR-489-3p and miR-630 suppress OCT2 expression by directly binding to the OCT2 3'-UTR. Meanwhile, via 786-O-OCT2-miRNAs stable expression cell models, we found that miRNAs could repress the classic substrate 1-methyl-4-phenylpyridinium (MPP+), fluorogenic substrate N,N-dimethyl-4-(2-pyridin-4-ylethenyl) aniline (ASP+), and oxaliplatin uptake by OCT2 both in vitro and in xenografts. In 33 clinical samples, miR-489-3p and miR-630 were significantly upregulated in RCC, negatively correlating with the OCT2 expression level compared to that in adjacent normal tissues, using tissue microarray analysis and qPCR validation. The increased binding of c-Myc to the promoter of pri-miR-630, responsible for the upregulation of miR-630 in RCC, was further evidenced by chromatin immunoprecipitation and dual-luciferase reporter assay. Overall, this study indicated that miR-489-3p and miR-630 function as oncotherapy-obstructing microRNAs by directly targeting OCT2 in RCC.
Collapse
|
30
|
Zhu Y, Yang L, Chong QY, Yan H, Zhang W, Qian W, Tan S, Wu Z, Lobie PE, Zhu T. Long noncoding RNA Linc00460 promotes breast cancer progression by regulating the miR-489-5p/FGF7/AKT axis. Cancer Manag Res 2019; 11:5983-6001. [PMID: 31308741 PMCID: PMC6612969 DOI: 10.2147/cmar.s207084] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/27/2019] [Indexed: 01/14/2023] Open
Abstract
Purpose: Evidence indicates that long noncoding RNAs (lncRNA) possess important roles in various cellular processes and that dysregulation of lncRNAs promotes tumor progression. However, the expression patterns and biological functions of many specific lncRNAs in breast cancer remain to be determined. Methods: Quantitative real-time polymerase chain reaction was performed to detect Linc00460, miR-489-5p and FGF7 expression. Protein levels were determined using Western blot. MTT and colony formation assay were used to measure cell proliferation. Transwell assays were conducted to determine cell migration and invasion. Luciferase reporter assays were carried out to assess the interaction between miR-489-5p and Linc00460 or FGF7. Biotin pull-down assay was used to detect the direct interaction between miR-489-5p and Linc00460. In vivo experiments were performed to measure tumor formation and lung metastasis. Results: We demonstrated that lncRNA Linc00460 was upregulated in breast cancer, and its expression level was positively associated with lymphatic metastasis and poor overall survival. Forced expression of Linc00460 increased, whereas Linc00460 silencing decreased, breast cancer cell viability, migration and invasion both in vitro and in vivo. Linc00460 was identified as a direct target of miR-489-5p, which further targeted FGF7 and exerted oncogenic functions in breast cancer. Mechanistically, Linc00460 served as a competing endogenous RNA of FGF-7 mRNA by sponging miR-489-5p, resulting in upregulated FGF7 expression and AKT activity. Notably, forced expression of miR-489-5p abrogated Linc00460-mediated oncogenic behavior and activation of the FGF7-AKT pathway in breast cancer cells. Conclusion: We have demonstrated that Linc00460 promotes breast cancer progression partly through the miR-489-5p/FGF7/AKT axis.
Collapse
Affiliation(s)
- Yong Zhu
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Leiyan Yang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Qing-Yun Chong
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore, Singapore
| | - Hong Yan
- Department of Pathology, Anhui Provincial Cancer Hospital, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Weijie Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Wenchang Qian
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Sheng Tan
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Zhengsheng Wu
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, People's Republic of China
| | - Peter E Lobie
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, Guangdong, People's Republic of China
| | - Tao Zhu
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| |
Collapse
|
31
|
Fujii R, Osaka E, Sato K, Tokuhashi Y. MiR-1 Suppresses Proliferation of Osteosarcoma Cells by Up-regulating p21 via PAX3. Cancer Genomics Proteomics 2019; 16:71-79. [PMID: 30587501 DOI: 10.21873/cgp.20113] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND/AIM miRNA-1(miR-1) is down-regulated in various cancer cells including osteosarcoma cells. This study was conducted to analyze the function of miR-1 in osteosarcoma cells. MATERIALS AND METHODS miR-1 expression in osteosarcoma cells was evaluated by qRT-PCR. Cell proliferation was evaluated after transfecting miR-1 by WST8 assay and FACS analysis, both in vitro and in vivo. RESULTS Overexpression of miR-1 suppressed cell proliferation and induced cell-cycle arrest in the G0-G1 phase by increasing p21 levels via a p53-independent pathway. Overexpression of miR-1 down-regulated PAX3, a potential p21-regulating gene. Moreover, knockdown of PAX3 suppressed cell proliferation by increasing p21 levels, and induced arrest at the G0/G1 phase. Administration of miR-1 showed an in vivo antitumor effect. CONCLUSION Overexpression of miR-1 suppressed cell proliferation and induced arrest in the G0/G1 phase by increasing p21 levels via a p53-independent pathway through PAX3 suppression. These results indicate that miR-1 could be a therapeutic target for osteosarcoma.
Collapse
Affiliation(s)
- Ryota Fujii
- Department of Orthopaedic Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Eiji Osaka
- Department of Orthopaedic Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Kentaro Sato
- Department of Orthopaedic Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Yasuaki Tokuhashi
- Department of Orthopaedic Surgery, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
32
|
Zhan FB, Zhang XW, Feng SL, Cheng J, Zhang Y, Li B, Xie LZ, Deng QR. MicroRNA-206 Reduces Osteosarcoma Cell Malignancy In Vitro by Targeting the PAX3-MET Axis. Yonsei Med J 2019; 60:163-173. [PMID: 30666838 PMCID: PMC6342722 DOI: 10.3349/ymj.2019.60.2.163] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/14/2018] [Accepted: 12/18/2018] [Indexed: 12/16/2022] Open
Abstract
PURPOSE This study was undertaken to explore how miR-206 represses osteosarcoma (OS) development. MATERIALS AND METHODS Expression levels of miR-206, PAX3, and MET mRNA were explored in paired OS and adjacent tissue specimens. A patient-derived OS cell line was established. miR-206 overexpression and knockdown were achieved by lentiviral transduction. PAX3 and MET overexpression were achieved by plasmid transfection. Treatment with hepatocyte growth factor (HGF) was utilized to activate c-Met receptor. Associations between miR-206 and PAX3 or MET mRNA in OS cells were verified by AGO2-RNA immunoprecipitation assay and miRNA pulldown assay. OS cell malignancy was evaluated in vitro by cell proliferation, metastasis, and apoptosis assays. PAX3 and MET gene expression in OS cells was assayed by RT-qPCR and Western blot. Activation of PI3K-AKT and MAPK-ERK in OS cells were assayed by evaluating Akt1 Ser473 phosphorylation and total threonine phosphorylation of Erk1/2, respectively. RESULTS Expression levels of miR-206 were significantly decreased in OS tissue specimens, compared to adjacent counterparts, and were inversely correlated with expression of PAX3 and MET mRNA. miR-206 directly interacted with PAX3 and MET mRNA in OS cells. miR-206 overexpression significantly reduced PAX3 and MET gene expression in OS cells in vitro, resulting in significant decreases in Akt1 and Erk1/2 activation, cell proliferation, and metastasis, as well as increases in cell apoptosis, while miR-206 knockdown showed the opposite effects. The effects of miR-206 overexpression on OS cells were reversed by PAX3 or MET overexpression, but only partially attenuated by HGF treatment. CONCLUSION miR-206 reduces OS cell malignancy in vitro by targeting PAX3 and MET gene expression.
Collapse
Affiliation(s)
- Fang Biao Zhan
- Department of Spine Surgery, Chongqing Three Gorges Central Hospital, Chongqing, China
| | - Xian Wei Zhang
- Department of Neurology, Chongqing Three Gorges Central Hospital, Chongqing, China
| | - Shi Long Feng
- Department of Spine Surgery, Chongqing Three Gorges Central Hospital, Chongqing, China
| | - Jun Cheng
- Department of Spine Surgery, Chongqing Three Gorges Central Hospital, Chongqing, China
| | - You Zhang
- Department of Spine Surgery, Chongqing Three Gorges Central Hospital, Chongqing, China
| | - Bo Li
- Department of Spine Surgery, Chongqing Three Gorges Central Hospital, Chongqing, China
| | - Li Zhong Xie
- Department of Spine Surgery, Chongqing Three Gorges Central Hospital, Chongqing, China
| | - Qian Rong Deng
- Department of Internal Medicine, Chongqing Wanzhou District Traditional Chinese Hospital, Chongqing, China.
| |
Collapse
|
33
|
Paired box 8 suppresses tumor angiogenesis and metastasis in gastric cancer through repression of FOXM1 via induction of microRNA-612. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:159. [PMID: 30021604 PMCID: PMC6052629 DOI: 10.1186/s13046-018-0830-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 07/05/2018] [Indexed: 12/31/2022]
Abstract
Background Paired box 8 (PAX8) has been documented to be downregulated in gastric cancer. However, its biological function in this malignancy is poorly understood. Methods In the present work, we investigated the effects of PAX8 overexpression and knockdown on the aggressive phenotype of gastric cancer cells. We further checked the involvement of forkhead box M1 (FOXM1), a ubiquitously expressed oncogene that can facilitate gastric cancer progression, in the action of PAX8. Results Ectopic expression of PAX8 blocked the migration and invasion of both AGS and SGC-7901 cells, but had no effect on cell proliferation. Conversely, knockdown of PAX8 enhanced gastric cancer cell migration and invasion. PAX8 overexpression inhibited epithelial-mesenchymal transition (EMT) and pro-angiogenic activity of gastric cancer cells. Mechanistically, PAX8 overexpression downregulated FOXM1 by stimulating microRNA (miR)-612 expression. Ectopic expression of miR-612 recapitulated the effect of PAX8 overexpression on gastric cancer cells, causing an inhibition of migration, invasion, EMT, and angiogenesis. Knockdown of miR-612 or overexpression of FOXM1 significantly reversed the tumor-suppressive activity of PAX8. In vivo studies further demonstrated that PAX8 overexpression restrained tumor angiogenesis and metastasis in nude mice, which was accompanied by increased expression of miR-612 and decreased expression of FOXM1. Conclusions PAX8 exerts a tumor-suppressive effect against gastric cancer cells, largely through induction of miR-612 and repression of FOXM1. Therefore, restoration of PAX8 expression may offer therapeutic benefits in the treatment of gastric cancer.
Collapse
|
34
|
Wang H, Fang ZL, Zhang GH, Ma X. TRIM44, a crucial target of miR-410, functions as a potential oncogene in osteosarcoma. Onco Targets Ther 2018; 11:3637-3647. [PMID: 29950867 PMCID: PMC6016597 DOI: 10.2147/ott.s163163] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Purpose Mounting evidence highlights the essential role of TRIM44 in tumor initiation and malignant progression in several cancers; however, the function of TRIM44 in osteosarcoma (OS) remains unknown. In this study, we aim to investigate the role of TRIM44 and reveal its regulation by deregulated miRNAs in OS. Materials and methods The expression profiles of TRIM44 were examined by immunohistochemistry, Western blotting, and qRT-PCR. The biological functions of TRIM44 were investigated through siRNA-mediated knockdown experiments. The regulation of TRIM44 by miR-410 was confirmed by Western blotting, dual luciferase reporter assays, and rescue experiments. Results TRIM44 was upregulated in OS tissues and cell lines, and its overexpression was positively correlated with TNM stage, metastasis, and recurrence. Knockdown of TRIM44 in OS cells suppressed cell proliferation, migration, invasion, and epithelial–mesenchymal transition. In addition, we identified TRIM44 as a novel target gene of miR-410 and miR-410 was remarkably downregulated in OS. Moreover, overexpression of miR-410 suppressed proliferation, migration, invasion, and epithelial–mesenchymal transition of OS cells by directly targeting TRIM44 expression. Furthermore, reintroduction of TRIM44 partially reversed miR-410-induced inhibitory effects on OS cells. Conclusion Collectively, our findings indicate that the miR-410/TRIM44 link is critical in the control of OS progression.
Collapse
Affiliation(s)
- Heng Wang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Zi-Ling Fang
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Gong-Hao Zhang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xin Ma
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
35
|
Soni M, Patel Y, Markoutsa E, Jie C, Liu S, Xu P, Chen H. Autophagy, Cell Viability, and Chemoresistance Are Regulated By miR-489 in Breast Cancer. Mol Cancer Res 2018; 16:1348-1360. [PMID: 29784669 DOI: 10.1158/1541-7786.mcr-17-0634] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 02/13/2018] [Accepted: 05/10/2018] [Indexed: 12/13/2022]
Abstract
It is postulated that the complexity and heterogeneity in cancer may hinder most efforts that target a single pathway. Thus, discovery of novel therapeutic agents targeting multiple pathways, such as miRNAs, holds promise for future cancer therapy. One such miRNA, miR-489, is downregulated in a majority of breast cancer cells and several drug-resistant breast cancer cell lines, but its role and underlying mechanism for tumor suppression and drug resistance needs further investigation. The current study identifies autophagy as a novel pathway targeted by miR-489 and reports Unc-51 like autophagy activating kinase 1 (ULK1) and lysosomal protein transmembrane 4 beta (LAPTM4B) to be direct targets of miR-489. Furthermore, the data demonstrate autophagy inhibition and LAPTM4B downregulation as a major mechanism responsible for miR-489-mediated doxorubicin sensitization. Finally, miR-489 and LAPTM4B levels were inversely correlated in human tumor clinical specimens, and more importantly, miR-489 expression levels predict overall survival in patients with 8q22 amplification (the region in which LAPTM4B resides).Implications: These findings expand the understanding of miR-489-mediated tumor suppression and chemosensitization in and suggest a strategy for using miR-489 as a therapeutic sensitizer in a defined subgroup of resistant breast cancer patients. Mol Cancer Res; 16(9); 1348-60. ©2018 AACR.
Collapse
Affiliation(s)
- Mithil Soni
- Department of Biological Science, University of South Carolina, Columbia, South Carolina.,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| | - Yogin Patel
- Department of Biological Science, University of South Carolina, Columbia, South Carolina.,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| | - Eleni Markoutsa
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina
| | - Chunfa Jie
- Master of Science in Biomedical Sciences Program, Des Moines University, Des Moines, Iowa
| | - Shou Liu
- Department of Biological Science, University of South Carolina, Columbia, South Carolina.,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| | - Peisheng Xu
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina
| | - Hexin Chen
- Department of Biological Science, University of South Carolina, Columbia, South Carolina. .,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
36
|
Boudjadi S, Chatterjee B, Sun W, Vemu P, Barr FG. The expression and function of PAX3 in development and disease. Gene 2018; 666:145-157. [PMID: 29730428 DOI: 10.1016/j.gene.2018.04.087] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 04/26/2018] [Accepted: 04/27/2018] [Indexed: 12/27/2022]
Abstract
The PAX3 gene encodes a member of the PAX family of transcription factors that is characterized by a highly conserved paired box motif. The PAX3 protein is a transcription factor consisting of an N-terminal DNA binding domain (containing a paired box and homeodomain) and a C-terminal transcriptional activation domain. This protein is expressed during development of skeletal muscle, central nervous system and neural crest derivatives, and regulates expression of target genes that impact on proliferation, survival, differentiation and motility in these lineages. Germline mutations of the murine Pax3 and human PAX3 genes cause deficiencies in these developmental lineages and result in the Splotch phenotype and Waardenburg syndrome, respectively. Somatic genetic rearrangements that juxtapose the PAX3 DNA binding domain to the transcriptional activation domain of other transcription factors deregulate PAX3 function and contribute to the pathogenesis of the soft tissue cancers alveolar rhabdomyosarcoma and biphenotypic sinonasal sarcoma. The wild-type PAX3 protein is also expressed in other cancers related to developmental lineages that normally express this protein and exerts phenotypic effects related to its normal developmental role.
Collapse
Affiliation(s)
- Salah Boudjadi
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | | | - Wenyue Sun
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Prasantha Vemu
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Frederic G Barr
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
37
|
Yang Z, Wa QD, Lu C, Pan W, Lu ZΜ, Ao J. miR‑328‑3p enhances the radiosensitivity of osteosarcoma and regulates apoptosis and cell viability via H2AX. Oncol Rep 2017; 39:545-553. [PMID: 29207178 PMCID: PMC5783622 DOI: 10.3892/or.2017.6112] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 09/26/2017] [Indexed: 11/11/2022] Open
Abstract
Osteosarcoma is a kind of high-risk sarcoma of the skeleton typically observed in people under 25 years old. Currently, radiotherapy is widely applied in cancer treatment. However, osteosarcoma is radioresistant and accordingly new, more effective radiosensitizers are needed. miRNAs have been reported to play an important role in osteosarcoma radiosensitivity. We examined the modulating effect of miR-328-3p in vivo and in vitro. miR-328-3p was downregulated in HOS-2R cells. The overexpression of miR-328-3p enhanced the radiosensitivity of osteosarcoma cells. miR-328-3p inhibited proliferation and promoted apoptosis in osteosarcoma cells under radiation conditions. In cells overexpressing miR-328-3p, H2AX expression was downregulated. We found that miR-328-3p targets H2AX and inhibits its expression. It was concluded, that miR-328-3p enhances the radiosensitization of osteosarcoma following X-ray irradiation, and determined that it directly targets H2AX to regulate radiosensitization.
Collapse
Affiliation(s)
- Zhen Yang
- Guizhou Provincial People's Hospital, Guiyang, Guizhou, P.R. China
| | - Qing-De Wa
- Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, P.R. China
| | - Chao Lu
- Guizhou Provincial People's Hospital, Guiyang, Guizhou, P.R. China
| | - Wei Pan
- Guizhou Provincial People's Hospital, Guiyang, Guizhou, P.R. China
| | - Zi-Μo Lu
- Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, P.R. China
| | - Jun Ao
- Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, P.R. China
| |
Collapse
|
38
|
Biological analysis of cancer specific microRNAs on function modeling in osteosarcoma. Sci Rep 2017; 7:5382. [PMID: 28710380 PMCID: PMC5511279 DOI: 10.1038/s41598-017-05819-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/02/2017] [Indexed: 12/24/2022] Open
Abstract
Osteosarcoma (OS) is the most common bone tumor characterized with a high risk of amputation and malignant morbidity among teenagers and adolescents. However, relevant pathogenic/biological mechanisms underlying OS-genesis remains to be ambiguous. The aim of this study was to elucidate functional relationship about microRNAs-mRNAs networks and to identify potential molecular markers via a computational method. Gene expression profile (GSE70415) was recruited from Gene Expression Omnibus. 3856 differentially expressed genes and 250 significantly expressed microRNAs were identified by using GCBI. The results of GO and KEGG pathways associated proteomics analysis indicated that extracellular matrix organization, small molecule metabolic process, cell adhesion (GO IDs: 0030198, 0044281, 0007155) and pathways in cancer, PI3K-Akt signaling pathway, metabolic pathways (pathway IDs: 5200, 4151, 1100) were significantly enriched. In addition, CKMT2, miR-93b-5p, miR-29b-3p were found to be positively/negatively correlated with TP53, EGFR, and MMP members mediated OS development, including angiogenesis, migration and invasion. Further visualization of collective effect of 1181 microRNAs-mRNAs pairs and protein-protein interactions was realized by applying with cytosacpe. In summary, our work provided a better understanding of non-coding regulatory mechanisms of transcriptomics and unraveled essential molecular biomarkers in osteosarcoma.
Collapse
|