1
|
Ding Y, Feng X, Liu Z, Liao Y, Pu L, Liu J, Wang H, Zhai Z, Xiong S. HDAC2-miR183-5p epigenetic circuit contributes to the growth of Philadelphia chromosome-positive B cell acute lymphoblastic leukemia via PTEN/AKT and c-MYC signaling pathway. J Leukoc Biol 2025; 117:qiae200. [PMID: 39258325 DOI: 10.1093/jleuko/qiae200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/08/2024] [Accepted: 09/10/2024] [Indexed: 09/12/2024] Open
Abstract
Philadelphia chromosome-positive B cell acute lymphoblastic leukemia [Ph(+) B-ALL] is a hematological malignancy with a poor prognosis. Epigenetic abnormalities, especially abnormal histone acetylation and microRNA (miRNA) dysregulation, are a group of epigenetic patterns that contribute to leukemia progression. However, their regulatory mechanisms in Ph(+) B-ALL have not been fully elucidated. In this study, we identified that miR-183-5p is significantly downregulated in Ph(+) B-ALL and associated with poor prognosis. Moreover, we found that the BCR-ABL fusion gene is a key target gene of miR-183-5p. MiR-183-5p directly targets the BCR-ABL gene and induces cell apoptosis via PTEN/AKT and c-MYC signaling pathways. In addition, a histone deacetylase inhibitor could mitigate the suppressive effects of HDAC2 on miR-183-5p by promoting promoter acetylation, thereby enhancing cell apoptosis. In conclusion, our results indicate that miR-183-5p is a potential biomarker and suggest that a novel HDAC2-miR-183-5p epigenetic circuitry regulation may be involved in the pathogenesis of Ph(+) B-ALL. Taken together, These findings provide new insights into the design of promising molecular-targeted drugs for Ph(+) B-ALL.
Collapse
Affiliation(s)
- Yangyang Ding
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, Anhui 230031, China
| | - Xiangjiang Feng
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, Anhui 230031, China
| | - Zelin Liu
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, Anhui 230031, China
| | - Ya Liao
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, Anhui 230031, China
| | - Lianfang Pu
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, Anhui 230031, China
| | - Jun Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230031, China
| | - Huiping Wang
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, Anhui 230031, China
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, Anhui 230031, China
| | - Zhimin Zhai
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, Anhui 230031, China
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, Anhui 230031, China
| | - Shudao Xiong
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, Anhui 230031, China
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, Anhui 230031, China
| |
Collapse
|
2
|
Cui X, Chen Y, Zhao L, Ding X. Extracellular vesicles derived from paclitaxel-sensitive nasopharyngeal carcinoma cells deliver miR-183-5p and impart paclitaxel sensitivity through a mechanism involving P-gp. Cell Biol Toxicol 2023; 39:2953-2970. [PMID: 37296288 DOI: 10.1007/s10565-023-09812-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 05/23/2023] [Indexed: 06/12/2023]
Abstract
Paclitaxel treatment has been applied for late-stage nasopharyngeal carcinoma (NPC), but therapy failure usually occurs due to paclitaxel resistance. Besides, microRNAs (miRs) delivered by extracellular vesicles (EVs) have been demonstrated as promising biomarkers affecting cancer development. Our work clarified the role of bioinformatically predicted miR-183-5p, which could be delivered by EVs, in the paclitaxel resistance of NPC. Downstream targets of miR-183-5p were predicted in publicly available databases, followed by GO enrichment analysis. A confirmatory dual-luciferase reporter assay determined the targeting relationship between miR-183-5p and P-glycoprotein (P-gp). The shuttling of extracellular miR-183-5p was identified by immunofluorescence. EVs transferred miR-183-5p from paclitaxel-sensitive NPC cells to paclitaxel-resistant NPC cells. Furthermore, overexpression of miR-183-5p and under-expression of P-gp occurred in clinical samples and cells of NPC. High expression of miR-183-5p corresponded to better survival of paclitaxel-treated patients. The effects of manipulated expression of miR-183-5p on NPC cell activities, tumor growth, and paclitaxel resistance were investigated in vitro and in vivo. Its effect was achieved through negatively regulating drug transporters P-gp. Ectopically expressed miR-183-5p enhanced the cancer-suppressive effects of paclitaxel by targeting P-gp, corresponding to diminished cell viability and tumor growth. Taken together, this work goes to elucidate the mechanical actions of miR-183-5p delivered by EVs and its significant contribution towards paclitaxel sensitivity to NPC. 1. This study provides mechanistic insight into the role of miR-183-5p-containing EVs in NPC. 2. The intercellular transportation of miR-183-5p is mediated by EVs in NPC. 3. Overexpressing miR-183-5p facilitates the anti-tumor effects of paclitaxel in NPC. 4. miR-183-5p suppresses paclitaxel resistance of NPC cells by inhibiting P-gp.
Collapse
Affiliation(s)
- Xiangguo Cui
- Department of Otorhinolaryngology Head and Neck, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang, 110000, China
| | - Yu Chen
- Department of Otorhinolaryngology Head and Neck, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang, 110000, China
| | - Lanqing Zhao
- Department of Sleep Medical Center, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Xiaoxu Ding
- Department of Otorhinolaryngology Head and Neck, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang, 110000, China.
| |
Collapse
|
3
|
Tolue Ghasaban F, Maharati A, Zangouei AS, Zangooie A, Moghbeli M. MicroRNAs as the pivotal regulators of cisplatin resistance in head and neck cancers. Cancer Cell Int 2023; 23:170. [PMID: 37587481 PMCID: PMC10428558 DOI: 10.1186/s12935-023-03010-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 07/28/2023] [Indexed: 08/18/2023] Open
Abstract
Although, there is a high rate of good prognosis in early stage head and neck tumors, about half of these tumors are detected in advanced stages with poor prognosis. A combination of chemotherapy, radiotherapy, and surgery is the treatment option in head and neck cancer (HNC) patients. Although, cisplatin (CDDP) as the first-line drug has a significant role in the treatment of HNC patients, CDDP resistance can be observed in a large number of these patients. Therefore, identification of the molecular mechanisms involved in CDDP resistance can help to reduce the side effects and also provides a better therapeutic management. MicroRNAs (miRNAs) as the post-transcriptional regulators play an important role in drug resistance. Therefore, in the present review we investigated the role of miRNAs in CDDP response of head and neck tumors. It has been reported that the miRNAs exerted their roles in CDDP response by regulation of signaling pathways such as WNT, NOTCH, PI3K/AKT, TGF-β, and NF-kB as well as apoptosis, autophagy, and EMT process. The present review paves the way to suggest a non-invasive miRNA based panel marker for the prediction of CDDP response among HNC patients. Therefore, such diagnostic miRNA based panel marker reduces the CDDP side effects and improves the clinical outcomes of these patients following an efficient therapeutic management.
Collapse
Affiliation(s)
- Faezeh Tolue Ghasaban
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sadra Zangouei
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Zangooie
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
- Student research committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Schneider B, William D, Lamp N, Zimpfer A, Henker C, Classen CF, Erbersdobler A. The miR-183/96/182 cluster is upregulated in glioblastoma carrying EGFR amplification. Mol Cell Biochem 2022; 477:2297-2307. [PMID: 35486213 PMCID: PMC9395473 DOI: 10.1007/s11010-022-04435-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 04/08/2022] [Indexed: 11/29/2022]
Abstract
Glioblastoma (GBM) is one of the most frequent primary brain tumors. Limited therapeutic options and high recurrency rates lead to a dismal prognosis. One frequent, putative driver mutation is the genomic amplification of the oncogenic receptor tyrosine kinase EGFR. Often accompanied by variants like EGFRvIII, heterogenous expression and ligand independent signaling render this tumor subtype even more difficult to treat, as EGFR-directed therapeutics show only weak effects at best. So EGFR-amplified GBM is considered to have an even worse prognosis, and therefore, deeper understanding of molecular mechanisms and detection of potential targets for novel therapeutic strategies is urgently needed. In this study, we looked at the level of microRNAs (miRs), small non-coding RNAs frequently deregulated in cancer, both acting as oncogenes and tumor suppressors. Comparative analysis of GBM with and without EGFR amplification should give insight into the expression profiles of miRs, which are considered both as potential targets for directed therapies or as therapeutic reagents. Comparison of miR profiles of EGFR-amplified and EGFR-normal GBM revealed an upregulation of the miR-183/96/182 cluster, which is associated with oncogenic properties in several tumor entities. One prominent target of this miR cluster is FOXO1, a pro-apoptotic factor. By observing FOXO1 downregulation in EGFR-amplified tumors, we can see a significant correlation of EGFR amplification, miR-183/96/182 cluster upregulation, and repression of FOXO1. Although no significant difference in overall survival is shown, these data may contribute to the molecular understanding of this tumor subtype and offer potential targets for miR-based therapies.
Collapse
Affiliation(s)
- Björn Schneider
- Institute of Pathology, University Medicine Rostock, Strempelstr. 14, 18057 Rostock, Germany
| | - Doreen William
- Children and Adolescents Hospital, University Medicine Rostock, Ernst-Heydemann-Str. 8, 18057 Rostock, Germany
- Present Address: ERN-GENTURIS, Hereditary Cancer Syndrome Center Dresden, Institute for Clinical Genetics, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Nora Lamp
- Institute of Pathology, University Medicine Rostock, Strempelstr. 14, 18057 Rostock, Germany
| | - Annette Zimpfer
- Institute of Pathology, University Medicine Rostock, Strempelstr. 14, 18057 Rostock, Germany
| | - Christian Henker
- Department of Neurosurgery, University Medicine Rostock, Schillingallee 35, 18057 Rostock, Germany
| | - Carl Friedrich Classen
- Children and Adolescents Hospital, University Medicine Rostock, Ernst-Heydemann-Str. 8, 18057 Rostock, Germany
| | - Andreas Erbersdobler
- Institute of Pathology, University Medicine Rostock, Strempelstr. 14, 18057 Rostock, Germany
| |
Collapse
|
5
|
Xiao J, He X. Involvement of Non-Coding RNAs in Chemo- and Radioresistance of Nasopharyngeal Carcinoma. Cancer Manag Res 2021; 13:8781-8794. [PMID: 34849030 PMCID: PMC8627240 DOI: 10.2147/cmar.s336265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/04/2021] [Indexed: 12/16/2022] Open
Abstract
The crucial treatment for nasopharyngeal carcinoma (NPC) is radiation therapy supplemented by chemotherapy. However, long-term radiation therapy can cause some genetic and proteomic changes to produce radiation resistance, leading to tumour recurrence and poor prognosis. Therefore, the search for new markers that can overcome the resistance of tumor cells to drugs and radiotherapy and improve the sensitivity of tumor cells to drugs and radiotherapy is one of the most important goals of pharmacogenomics and cancer research, which is important for predicting treatment response and prognosis. Non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), may play important roles in regulating chemo- and radiation resistance in nasopharyngeal carcinoma by controlling the cell cycle, proliferation, apoptosis, and DNA damage repair, as well as other signalling pathways. Recent research has suggested that selective modulation of ncRNA activity can improve the response to chemotherapy and radiotherapy, providing an innovative antitumour approach based on ncRNA-related gene therapy. Therefore, ncRNAs can serve as biomarkers for tumour prediction and prognosis, play a role in overcoming drug resistance and radiation resistance in NPC, and can also serve as targets for developing new therapeutic strategies. In this review, we discuss the involvement of ncRNAs in chemotherapy and radiation resistance in NPC. The effects of these molecules on predicting therapeutic cancer are highlighted.
Collapse
Affiliation(s)
- Jiaxin Xiao
- Hunan Province Key Laboratory of Tumour Cellular & Molecular Pathology Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang, 421001, Hunan Province, People’s Republic of China
| | - Xiusheng He
- Hunan Province Key Laboratory of Tumour Cellular & Molecular Pathology Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang, 421001, Hunan Province, People’s Republic of China
| |
Collapse
|
6
|
Li M, Wang Y, Zhao Q, Ma W, Liu J. MiR-30a-5p inhibits proliferation, migration and invasion of nasopharyngeal carcinoma cells by targeting NUCB2. Hum Exp Toxicol 2021; 40:1274-1285. [PMID: 33567921 DOI: 10.1177/0960327121991913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a malignant head and neck tumor arising in the nasopharynx. MicroRNAs (miRNAs) are elucidated to exert tumor-suppressing function in human cancers. Numerous studies have manifested that miR-30a-5p serves as an anti-oncogene in various cancers. OBJECTIVE To research the biological function and molecular mechanism of miR-30a-5p in NPC. METHODS The morphology of NPC tissues was revealed by H&E staining. Transwell and wound healing assays were applied to investigate the effects of miR-30a-5p on NPC cell migration. The binding interaction between miR-30a-5p and nucleobindin 2 (NUCB2) was identified by luciferase reporter assay. Xenograft nude mice were used to detect the influence of miR-30a-5p on NPC tumor growth. RESULTS MiR-30a-5p was downregulated in NPC tissues and cells. The overexpression ofmiR-30a-5p inhibited proliferation, migration and invasion abilities of NPC cells. Moreover, NUCB2 was revealed to be a downstream target gene of miR-30a-5p, and knockdown of NUCB2 repressed the malignant behaviors of NPC cells and tumor growth. Additionally, rescue experiments revealed that miR-30a-5p suppressed the proliferation, migration and invasion of NPC cells via targeting NUCB2 in vitro. Meanwhile, in vivo assays depicted that NUCB2 overexpression rescued the effects induced by miR-30a-5p upregulation on tumor growth. CONCLUSION MiR-30a-5p modulates NPC progression by targeting NUCB2. These findings lay a foundation for exploring the clinical treatment of NPC.
Collapse
Affiliation(s)
- M Li
- Department of Otorhinolaryngology, 74566The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- M Li and Y Wang are co-first authors
| | - Y Wang
- Department of Otorhinolaryngology, 74566The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- M Li and Y Wang are co-first authors
| | - Q Zhao
- Department of Radiation Oncology, 74566The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - W Ma
- Department of Quality Management, 74566The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - J Liu
- Department of Otorhinolaryngology, 74566The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
7
|
Hu W, Yao W, Li H, Chen L. MiR-30e-5p inhibits the migration and invasion of nasopharyngeal carcinoma via regulating the expression of MTA1. Biosci Rep 2020; 40:BSR20194309. [PMID: 32458989 PMCID: PMC7253402 DOI: 10.1042/bsr20194309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/14/2020] [Accepted: 04/21/2020] [Indexed: 11/17/2022] Open
Abstract
The study explored the effect of miR-30e-5p on nasopharyngeal carcinoma (NPC). MiR-30e-5p levels in NPC cancer and adjacent normal samples, in metastatic and non-metastatic cancer samples of NPC, and in NP69 cell and five NPC cell lines were determined by quantitative real-time polymerase chain reaction (qRT-PCR). The relationship between miR-30e-5p and MTA1 was confirmed by dual-luciferase reporter assay, Western blot and qRT-PCR. The viability, migration and invasion of 5-8F and 6-10B cells were determined by CCK-8, scratch test and transwell assays, respectively. The levels of migration-related proteins (vimentin and Snail) and invasion-related proteins (MMP2 and MMP3) in NPC cells were detected by Western blot. The results showed that low expression of miR-30e-5p was associated with HNSC cancer, NPC, metastasis of NPC and NPC cell lines. Overexpressed miR-30e-5p in HNSC cancer and NPC was predictive of a better prognosis of patients. In addition, the viability, migration and invasion were reduced by up-regulating miR-30e-5p in 5-8F cells, but promoted by down-regulated miR-30e-5p in 6-10B cells. MiR-30e-5p reversed the migration and invasion of NPC cells regulated by MTA1, and inhibited migration and invasion of NPC cells via regulating MTA1 expression.
Collapse
Affiliation(s)
- Weiqun Hu
- Department of Otorhinolaryngology, The Affiliated Hospital of Putian University, China
| | - Wenfeng Yao
- Department of Otorhinolaryngology, Xinxiang First People’s Hospital, China
| | - Haolin Li
- Department of Otorhinolaryngology, Xinxiang First People’s Hospital, China
| | - Li Chen
- Department of Otorhinolaryngology, Zaozhuang Municipal Hospital, China
| |
Collapse
|
8
|
Wu J, Zhang C, Chen L. MiR-511 mimic transfection inhibits the proliferation, invasion of osteosarcoma cells and reduces metastatic osteosarcoma tumor burden in nude mice via targeting MAPK1. Cancer Biomark 2020; 26:343-351. [PMID: 31524148 PMCID: PMC6918904 DOI: 10.3233/cbm-190534] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Osteosarcoma, a highly aggressive cancer, can rapidly metastasize to distant organs such as lung, liver, brain. Despite much progress in the therapeutic regime has been made, the prognosis of osteosarcoma remains poor. In present study, microRNA-511 (miR-511) is lowly expressed in osteosarcoma cells, including MG63, U-2 OS, Saos-2 cells, while mitogen activated protein kinase1 (MAPK1) is highly expressed in osteosarcoma cells. Interestingly, MAPK1 might be a target of miR-511. We found that overexpression of miR-511 by miR-511 mimic transfection may result to low expression of MAPK1. Further study showed that miR-511 mimic inhibits the development of osteosarcoma MG63 cell, including proliferation and invasion. Moreover, miR-511 mimic transfection reduces metastatic osteosarcoma tumor burden in nude mice. These activities are mediated by targeting MAPK1. Our study provides a new sight for the molecular pathogenesis of osteosarcoma.
Collapse
Affiliation(s)
| | | | - Lu Chen
- Corresponding author: Lu Chen, Orthopedics, North Sichuan Medical College Affiliated Hospital, No. 63 Wenhua Road, Shunqing District, Nanchong, 637000, Sichuan, China. Tel.: +86 138 908 52575; Fax: +86 0817 2262642; E-mail:
| |
Collapse
|
9
|
Cao D, Di M, Liang J, Shi S, Tan Q, Wang Z. MicroRNA-183 in Cancer Progression. J Cancer 2020; 11:1315-1324. [PMID: 32047538 PMCID: PMC6995398 DOI: 10.7150/jca.39044] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/16/2019] [Indexed: 12/15/2022] Open
Abstract
MicroRNA-183(miR-183) is abnormally expressed in many kinds of tumors. It participates in the initiation and development of tumors. There are many pathways regulate the expression of miR-183. The action mechanism of miR-183 in cancer is very extensive, and contradictory conclusions are often drawn. It was upregulated in 18 kinds of cancer, downregulated in 6 kinds of cancer. In addition, there are seven types of cancer, both upregulated and downregulated reports can be found. Evidence showed that miR-183 can not only directly play the role of oncogene or antioncogene, but also regulate the expression of other oncogene or antioncogene in different cancer types. In this review, we discuss the regulator of miR-183 and summarized the expression of miR-183 in different cancers. We also counted the target genes of miR-183 and the functional roles they play. Furthermore, we focused on the roles of miR-183 in cell migration, cell invasion, epithelial-mesenchymal transition (EMT) and microangiogenesis, which play the most important roles in cancer processes. It sheds light on the likely reasons why miR-183 plays different roles in various cancers. In addition, miR-183 and its downstream effectors have the potential to be promising prognostic markers and therapeutic targets in cancer.
Collapse
Affiliation(s)
- Dingren Cao
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Min Di
- Sir Run Shaw Hospital, Zhejiang University College of Medicine, Hangzhou, 310058, P. R. China
| | - Jingjie Liang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Shuang Shi
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Qiang Tan
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Zhengguang Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| |
Collapse
|
10
|
Li X, Zeng X. Shikonin suppresses progression and epithelial-mesenchymal transition in hepatocellular carcinoma (HCC) cells by modulating miR-106b/SMAD7/TGF-β signaling pathway. Cell Biol Int 2019; 44:467-476. [PMID: 31617643 DOI: 10.1002/cbin.11247] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/13/2019] [Indexed: 12/13/2022]
Abstract
Shikonin is a natural naphthoquinone component with antioxidant and anti-tumor function and has been used for hepatocellular carcinoma (HCC) treatment. According to the previous study, many herbs can regulate cancer cell progression by targeting specific microRNA (miRNA) (Liu, 2016). However, the underlying pathological mechanism of shikonin in HCC therapy is still unclear. The detection of cell growth and death rate were performed by hemacytometry and trypan blue staining, respectively. The expression of miR-106b and SMAD7 messenger RNA (mRNA) in HCC cells was evaluated by quantitative real-time polymerase chain reaction. Cell proliferation, apoptosis, and migration ability were measured by cell counting kit-8 (CCK-8), flow cytometry, and transwell assay. The expression of proteins E-cadherin, N-cadherin, vimentin, SMAD7, TGF-β1, p-SMAD3, SMAD3, and GAPDH was examined by western blot. The interaction between SMAD7 and miR-106b was assessed by luciferase reporter system. Shikonin inhibited Huh7 and HepG2 cell growth in a dose-dependent manner while induced cell death in a time-dependent manner. In addition, the expression of miR-106b was reduced after shikonin treatment. Moreover, miR-106b attenuated the suppressive effects of shikonin on HCC cell migration and epithelial-mesenchymal transition (EMT). SMAD7 was predicted as a target of miR-106b and the prediction was confirmed by luciferase reporter system. Additionally, we observed that SMAD7 reversed the promotive effects of miR-106b on HCC cell progression and EMT. The subsequent western blot assay revealed that shikonin could modulate SMAD7/TGF-β signaling pathway by targeting miR-106b. In conclusion, Shikonin suppresses cell progression and EMT and accelerates cell death of HCC cells via modulating miR-106b/SMAD7/TGF-β signaling pathway, suggesting shikonin could be an effective agent for HCC treatment.
Collapse
Affiliation(s)
- Xiaojing Li
- Department of Minimal Invasive Surgery, the Second Xiangya Hospital of Central South University, Changsha, 410000, China
| | - Xianpeng Zeng
- Department of Institute of Hepatobiliary Diseases, the Zhongnan Hospital of Wuhan University, Wuhan, 430000, China
| |
Collapse
|
11
|
Banks SA, Pierce ML, Soukup GA. Sensational MicroRNAs: Neurosensory Roles of the MicroRNA-183 Family. Mol Neurobiol 2019; 57:358-371. [DOI: 10.1007/s12035-019-01717-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/19/2019] [Indexed: 12/20/2022]
|
12
|
The role and mechanisms of action of microRNAs in cancer drug resistance. Clin Epigenetics 2019; 11:25. [PMID: 30744689 PMCID: PMC6371621 DOI: 10.1186/s13148-018-0587-8] [Citation(s) in RCA: 457] [Impact Index Per Article: 76.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 11/19/2018] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs with a length of about 19–25 nt, which can regulate various target genes and are thus involved in the regulation of a variety of biological and pathological processes, including the formation and development of cancer. Drug resistance in cancer chemotherapy is one of the main obstacles to curing this malignant disease. Statistical data indicate that over 90% of the mortality of patients with cancer is related to drug resistance. Drug resistance of cancer chemotherapy can be caused by many mechanisms, such as decreased antitumor drug uptake, modified drug targets, altered cell cycle checkpoints, or increased DNA damage repair, among others. In recent years, many studies have shown that miRNAs are involved in the drug resistance of tumor cells by targeting drug-resistance-related genes or influencing genes related to cell proliferation, cell cycle, and apoptosis. A single miRNA often targets a number of genes, and its regulatory effect is tissue-specific. In this review, we emphasize the miRNAs that are involved in the regulation of drug resistance among different cancers and probe the mechanisms of the deregulated expression of miRNAs. The molecular targets of miRNAs and their underlying signaling pathways are also explored comprehensively. A holistic understanding of the functions of miRNAs in drug resistance will help us develop better strategies to regulate them efficiently and will finally pave the way toward better translation of miRNAs into clinics, developing them into a promising approach in cancer therapy.
Collapse
|
13
|
Zheng Z, Zheng X, Zhu Y, Gu X, Gu W, Xie X, Hu W, Jiang J. miR-183-5p Inhibits Occurrence and Progression of Acute Myeloid Leukemia via Targeting Erbin. Mol Ther 2019; 27:542-558. [PMID: 30799283 PMCID: PMC6401194 DOI: 10.1016/j.ymthe.2019.01.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 01/15/2019] [Accepted: 01/25/2019] [Indexed: 02/07/2023] Open
Abstract
Erbin has been shown to have significant effects on the development of solid tumors. However, little is known about its function and regulatory mechanism in hematological malignancies. The biological function of Erbin on cell proliferation was measured in vitro and in vivo. The predicted target of Erbin was validated by dual-luciferase reporter assay and rescue experiment. We found that overexpression of Erbin could inhibit the cell proliferation and promote the cell differentiation of acute myeloid leukemia (AML) cells, whereas depletion of Erbin could enhance the cell proliferation and block the cell differentiation in AML cells in vitro and in vivo. Besides, miR-183-5p was identified as the upstream regulator that negatively regulated the Erbin expression. The results were confirmed by dual-luciferase reporter and RNA pull-down assay. Furthermore, we found that miR-183-5p negatively regulated Erbin, resulting in enhanced cell proliferation of AML cells via activation of RAS/RAF/MEK/ERK and PI3K/AKT/FoxO3a pathways. The activation of RAS/RAF/MEK/ERK and PI3K/AKT/FoxO3a pathways was mediated by Erbin interacting with Grb2. These results were also validated by rescue experiments in vitro and in vivo. All above-mentioned findings indicated that the miR-183-5p/Erbin signaling pathway might represent a novel prognostic biomarker or therapeutic target for treatment of AML.
Collapse
Affiliation(s)
- Zhuojun Zheng
- Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China; Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China; Cancer Immunotherapy Engineering Research Center of Jiangsu Province, Changzhou, Jiangsu Province, China; Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu Province, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China; Cancer Immunotherapy Engineering Research Center of Jiangsu Province, Changzhou, Jiangsu Province, China; Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu Province, China
| | - Yuandong Zhu
- Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Xiaoyan Gu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China; Cancer Immunotherapy Engineering Research Center of Jiangsu Province, Changzhou, Jiangsu Province, China; Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu Province, China
| | - Weiying Gu
- Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China.
| | - Xiaobao Xie
- Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Wenwei Hu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China; Cancer Immunotherapy Engineering Research Center of Jiangsu Province, Changzhou, Jiangsu Province, China; Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu Province, China.
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China; Cancer Immunotherapy Engineering Research Center of Jiangsu Province, Changzhou, Jiangsu Province, China; Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu Province, China.
| |
Collapse
|
14
|
Meng F, Zhang L. miR-183-5p functions as a tumor suppressor in lung cancer through PIK3CA inhibition. Exp Cell Res 2019; 374:315-322. [DOI: 10.1016/j.yexcr.2018.12.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/05/2018] [Accepted: 12/03/2018] [Indexed: 01/10/2023]
|
15
|
Sun X, Xu Y, Zhang S, Li X, Wang Y, Zhang Y, Zhao X, Li Y, Wang Y. MicroRNA-183 suppresses the vitality, invasion and migration of human osteosarcoma cells by targeting metastasis-associated protein 1. Exp Ther Med 2018; 15:5058-5064. [PMID: 29805531 DOI: 10.3892/etm.2018.6068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 03/01/2018] [Indexed: 12/16/2022] Open
Abstract
The aim of the present study was to investigate the effects of microRNA (miR)-183 on vitality, invasion, metastasis and apoptosis in osteosarcoma (OS) cells, mediated by its binding to metastasis-associated protein 1 (MTA1). A dual luciferase reporter assay was performed to determine whether MTA1 was a direct target of miR-183. Cell Counting Kit-8, Transwell, scratch-wound healing, fluorescence-activated cell sorting andterminal deoxynucleotidyl transferase dUTP nick end labeling assays were also performed to investigate the effects of miR-183 expression on the proliferation, invasion, migration and apoptosis of MG63 cells. It was demonstrated that that MTA1 expression levels were significantly higher in OS tissues and MG63 cells compared with corresponding adjacent noncancerous tissues and normal cells, respectively, while miR-183 expression levels were significantly lower (both P<0.05). Furthermore, miR-183 overexpression downregulated MTA1 levels and inhibited cell proliferation (P<0.05), migration (P<0.05) and invasion (P<0.01), as well as promoting apoptosis (P<0.01) by binding to the 3'-untranslated region of MTA1. These results indicate that miR-183 inhibits the vitality, invasion, migration and apoptosis of the OS cell line MG63 by targeting MTA1. These findings may contribute to the development of novel clinical therapeutic approaches for the treatment of OS.
Collapse
Affiliation(s)
- Xiaoya Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yan Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Shanfeng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Xinjie Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yadong Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yan Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xuefeng Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yuebai Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yisheng Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
16
|
Gao JM, Huang LZ, Huang ZG, He RQ. Clinical value and potential pathways of miR-183-5p in bladder cancer: A study based on miRNA-seq data and bioinformatics analysis. Oncol Lett 2018; 15:5056-5070. [PMID: 29616090 DOI: 10.3892/ol.2018.7967] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/29/2018] [Indexed: 02/06/2023] Open
Abstract
The clinicopathological value and exploration of the potential molecular mechanism of microRNA-183-5p (miR-183-5p) have been investigated in various cancers; however, to the best of the author's knowledge, no similar research has been reported for bladder cancer. In the present study, it was revealed that the expression level of miR-183-5p was notably increased in bladder cancer tissues compared with adjacent non-cancerous tissues (P=0.001) and was markedly increased in the tissue samples of papillary, pathological T stage (T0-T2) and pathological stage (I-II) compared with tissue samples of their counterparts (P=0.05), according to data from The Cancer Genome Atlas. Receiver operating characteristic analysis revealed the robust diagnostic value of miR-183-5p for distinguishing bladder cancer from non-cancerous bladder tissues (area under curve=0.948; 95% confidence interval: 0.919-0.977). Amplification and deep deletion of miR-183-5p were indicated by cBioPortal, accounting for 1% (4/412) of bladder cancer cases. Data from YM500v3 demonstrated that compared with other cancers, bladder cancer exhibited high expression levels of miR-183-5p, and miR-183-5p expression in primary solid tumors was much higher compared with solid normal tissues. A meta-analysis indicated that miR-183-5p was more highly expressed in bladder cancer samples compared with normal counterparts. A total of 88 potential target genes of miR-183-5p were identified, 13 of which were discerned as hub genes by protein-protein interaction. The epithelial-to-mesenchymal transition pathway was the most significantly enriched pathway by FunRich (P=0.0001). In summary, miR-183-5p may participate in the tumorigenesis and development of bladder cancer via certain signaling pathways, particularly the epithelial-to-mesenchymal transition pathway. However, the exact molecular mechanism of miR-183-5p in bladder cancer must be validated by in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Jia-Min Gao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China.,Department of Biochemistry, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Lin-Zhen Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China.,Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Zhi-Guang Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China.,Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Rong-Quan He
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China.,Department of Biochemistry, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
17
|
Kim CW, Han JH, Wu L, Choi JY. microRNA-183 is Essential for Hair Cell Regeneration after Neomycin Injury in Zebrafish. Yonsei Med J 2018; 59:141-147. [PMID: 29214789 PMCID: PMC5725352 DOI: 10.3349/ymj.2018.59.1.141] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 10/11/2017] [Accepted: 10/29/2017] [Indexed: 12/21/2022] Open
Abstract
PURPOSE microRNAs (miRNAs) are non-coding RNAs composed of 20 to 22 nucleotides that regulate development and differentiation in various organs by silencing specific RNAs and regulating gene expression. In the present study, we show that the microRNA (miR)-183 cluster is upregulated during hair cell regeneration and that its inhibition reduces hair cell regeneration following neomycin-induced ototoxicity in zebrafish. MATERIALS AND METHODS miRNA expression patterns after neomycin exposure were analyzed using microarray chips. Quantitative polymerase chain reaction was performed to validate miR-183 cluster expression patterns following neomycin exposure (500 μM for 2 h). After injection of an antisense morpholino (MO) to miR-183 (MO-183) immediately after fertilization, hair cell regeneration after neomycin exposure in neuromast cells was evaluated by fluorescent staining (YO-PRO1). The MO-183 effect also was assessed in transgenic zebrafish larvae expressing green fluorescent protein (GFP) in inner ear hair cells. RESULTS Microarray analysis clearly showed that the miR-183 cluster (miR-96, miR-182, and miR-183) was upregulated after neomycin treatment. We also confirmed upregulated expression of the miR-183 cluster during hair cell regeneration after neomycin-induced ototoxicity. miR-183 inhibition using MO-183 reduced hair cell regeneration in both wild-type and GFP transgenic zebrafish larvae. CONCLUSION Our work demonstrates that the miR-183 cluster is essential for the regeneration of hair cells following ototoxic injury in zebrafish larvae. Therefore, regulation of the miR-183 cluster can be a novel target for stimulation of hair cell regeneration.
Collapse
Affiliation(s)
- Chang Woo Kim
- Department of Otorhinolaryngology, Hallym University College of Medicine, Seoul, Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hyuk Han
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
| | - Ling Wu
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Young Choi
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|