1
|
Xu F, Yang YH, Yang H, Li W, Hao Y, Zhang S, Zhang YZ, Cao WX, Li XX, Du GH, Ji TF, Wang JH. Progress of studies on natural products for glioblastoma therapy. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024; 26:154-176. [PMID: 38321773 DOI: 10.1080/10286020.2023.2300367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 12/25/2023] [Indexed: 02/08/2024]
Abstract
Glioblastoma (GBM) is the most common, malignant, and lethal primary brain tumor in adults. Up to now, the chemotherapy approaches for GBM are limited. Therefore, more studies on identifying and exploring new chemotherapy drugs or strategies overcome the GBM are essential. Natural products are an important source of drugs against various human diseases including cancers. With the better understanding of the molecular etiology of GBM, the development of new anti-GBM drugs has been increasing. Here, we summarized recent researches of natural products for the GBM therapy and their potential mechanisms in details, which will provide new ideas for the research on natural products and promote developing drugs from nature products for GBM therapy.
Collapse
Affiliation(s)
- Fang Xu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yi-Hui Yang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Hong Yang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Wan Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yue Hao
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Sen Zhang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yi-Zhi Zhang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Wan-Xin Cao
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Xiao-Xue Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Guan-Hua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Teng-Fei Ji
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Jin-Hua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
2
|
Yu S, Lv L, Li Y, Ning Q, Liu T, Hu T. PLK3 promotes the proneural-mesenchymal transition in glioblastoma via transcriptional regulation of C5AR1. Mol Biol Rep 2023; 50:8249-8258. [PMID: 37568042 DOI: 10.1007/s11033-023-08716-7] [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: 02/19/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023]
Abstract
BACKGROUND Accumulating evidence suggests that polo-like kinase 3 (PLK3) plays an essential role in tumor cells and induces cell proliferation and may have implications for the prognosis of various cancers. We sought to define the role of PLK3-dependent proneural-mesenchymal transition (PMT) in the glioblastoma (GBM) therapy. METHODS AND RESULTS We analyzed the expression data for PLK3 by using the TCGA database. PLK3 expression in GBM cell lines was determined by qRT-PCR and Western blotting. PLK3 levels were modulated using Lentivirus infection, and the effects on symptoms, tumor volume, and survival in mice intracranial xenograft models were determined. Irradiation (IR) was performed to induce PMT. PLK3 expression was significantly elevated in mesenchymal subtype GBM and promoted tumor proliferation in GBM. Additionally enriched PLK3 expression could be associated with poor prognosis in GBM patients compared with those who have lower PLK3 expression. Mechanically, PLK3-dependent PMT induced radioresistance in GBM cells via transcriptional regulation of complement C5a receptor 1 (C5AR1). In therapeutic experiments conducted in vitro, targeting PLK3 by using small molecule inhibitor decreased tumor growth and radioresistance of GBM cells both in vitro and in vivo. CONCLUSIONS PLK3-C5AR1 axis induced PMT thus enhanced radioresistance in GBM and could become a novel potential therapeutic target for GBM.
Collapse
Affiliation(s)
- Shuo Yu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710000, Shaanxi, China
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, Shaanxi, China
| | - Lin Lv
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710000, Shaanxi, China
| | - Yang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710000, Shaanxi, China
| | - Qian Ning
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710000, Shaanxi, China
| | - Tingting Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710000, Shaanxi, China
| | - Tinghua Hu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710000, Shaanxi, China.
| |
Collapse
|
3
|
A Signature of Three microRNAs Is a Potential Diagnostic Biomarker for Glioblastoma. IRANIAN BIOMEDICAL JOURNAL 2022; 26:301-12. [PMID: 35490305 PMCID: PMC9432466 DOI: 10.52547/ibj.3671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Background: Glioblastoma is the most common primary malignant neoplasm of the central nervous system. Despite progress in diagnosis and treatment, glioblastoma still has a poor prognosis. This study aimed to examine whether a signature of three candidate miRNAs (i.e. hsa-let-7c-5p, hsa-miR-206-5p, and hsa-miR-1909-5p) can be used as a diagnostic biomarker for distinguishing glioblastoma from healthy brain tissues. Methods: In this study, 50 FFPE glioblastoma tissue samples and 50 healthy tissue samples adjacent to tumor were included. The expression of each candidate miRNA (i.e. hsa-let-7c-5p, hsa-miR-206-5p, and hsa-miR-1909-5p) was measured using RT-qPCR. To show the roles of each miRNA and their biological effects on glioblastoma development and clinicopathological characteristics, in silico tools were used. ROC curves were performed to assess the diagnostic accuracy of each miRNA. Results: Based on the results, hsa-let-7c-5p and hsa-miR-206-5p were downregulated, while hsa-miR-1909-5p was upregulated in glioblastoma tumors compared to healthy samples. No association was detected between the expression of each candidate miRNA and sex. Except for hsa-let-7c-5p, other miRNAs did not correlate with age status. ROC curve analysis indicated that the signature of candidate miRNAs is a potential biomarker distinguishing between glioblastoma and healthy samples. Only hsa-miR-206-5p suggested the association with poor prognosis in glioblastoma patients. Conclusion: Our findings revealed that the signature of three miRNAs is capable of distinguishing glioblastoma tumor and healthy tissues. These results are beneficial for the clinical management of glioblastoma patients.
Collapse
|
4
|
Keqi H, Handong L. The Long Non-coding RNA Cytoskeleton Regulator (CYTOR) Sponges microRNA- 206 (miR-206) to Promote Proliferation and Invasion of HP75 Cells. Curr Cancer Drug Targets 2021; 21:526-535. [PMID: 33653250 DOI: 10.2174/1568009621666210302090309] [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: 08/19/2020] [Revised: 01/10/2021] [Accepted: 01/31/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The role and mechanism of long non-coding RNA cytoskeleton regulator (CYTOR) in Invasive Pituitary Adenomas (IPA) have not been elucidated previously. OBJECTIVE This study aimed to investigate the interaction between CYTOR and miR-206 and their roles in IPA using HP75 cells as the model. METHODS The expression levels of CYTOR and miR-206 were detected by quantitative real-time polymerase chain reaction (qRT-PCR) in IPA tissues and cell lines. The Chi-square test was used to analyze the correlation between CYTOR expression and clinical-pathological parameters. HP75 cell proliferation was detected by Cell Counting Kit-8 assay and colony formation assay. Scratch healing experiments and Transwell assay were used to detect migration and invasion of HP75 cells. The relationship between CYTOR and miR-206 was predicted by bioinformatics and verified by qRT-PCR and the dual-luciferase reporter gene method. RESULTS CYTOR is up-regulated in IPA tissues and cell lines. The high expression of CYTOR is associated with adenoma invasiveness and adenoma size of the patients. Down-regulation of CYTOR decreases the proliferation, migration and invasion of HP75 cells, while up-regulation of miR-206 can inhibit proliferation, migration and invasion of HP75 cells. MiR-206 is identified as a target of CYTOR and could be negatively regulated by it in IPA. DISCUSSION CYTOR, as a tumor-promoting factor, facilitates the proliferation, migration and invasion of HP75 cells through sponging miR-206. CONCLUSION The CYTOR-miR-206 axis provides new insights into the diagnosis and treatment of IPA.
Collapse
Affiliation(s)
- Hu Keqi
- Department of Neurosurgery, Xiangyang Center Hospital, Affiliated Hospital of Hubei University of Arts and Science, Jingzhou Street 39, Xiangyang 441021, China
| | - Liu Handong
- Department of Neurosurgery, Xiangyang Center Hospital, Affiliated Hospital of Hubei University of Arts and Science, Jingzhou Street 39, Xiangyang 441021, China
| |
Collapse
|
5
|
Wang C, Chen F, Fan Z, Yao C, Xiao L. lncRNA CCAT1/miR-490-3p/MAPK1/c-Myc positive feedback loop drives progression of acute myeloid leukaemia. J Biochem 2020; 167:379-388. [PMID: 31790145 DOI: 10.1093/jb/mvz107] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 11/11/2019] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukaemia (AML) is a frequently diagnosed malignancy in adults. Long non-coding RNA (lncRNA) colon cancer-associated transcript 1 (CCAT1) has been well known to play vital roles in multiple malignancies including AML. Unfortunately, the detailed mechanism of CCAT1 in AML progression remains obscure. In this study, we demonstrated that CCAT1 was up-regulated in AML samples while its target, miR-490-3p, was relatively down-regulated. CCAT1 markedly increased viability and metastasis of AML cells, while miR-490-3p had opposite effects. CCAT1 could specifically bind to miR-490-3p and reduce its expression and activity, and MAPK1 was a target gene of miR-490-3p. Overexpressed CCAT1 could induce MAPK1 expression and c-Myc reciprocally increased CCAT1 expression. Our data implied that miR-490-3p could be a novel therapeutic target for AML, and highlights the crucial role of CCAT1/miR-490-3p/MAPK1/c-Myc positive feedback loop in AML progression.
Collapse
Affiliation(s)
- Chenghong Wang
- Department of Hematology, The 3rd Xiangya Hospital, Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, Hunan 410013, China
| | - Fangping Chen
- Department of Hematology, The 3rd Xiangya Hospital, Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, Hunan 410013, China
| | - Zili Fan
- Department of Hematology, The 3rd Xiangya Hospital, Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, Hunan 410013, China
| | - Chenjiao Yao
- Department of Hematology, The 3rd Xiangya Hospital, Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, Hunan 410013, China
| | - Lijun Xiao
- Department of Hematology, The 3rd Xiangya Hospital, Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, Hunan 410013, China
| |
Collapse
|
6
|
Ge J, Yao Y, Jia H, Li P, Sun W. Inhibition of miR-21 ameliorates LPS-induced acute lung injury through increasing B cell lymphoma-2 expression. Innate Immun 2020; 26:693-702. [PMID: 32727244 PMCID: PMC7787552 DOI: 10.1177/1753425920942574] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/06/2020] [Accepted: 06/22/2020] [Indexed: 02/02/2023] Open
Abstract
The aberrant expression of microRNAs (miRNAs) is associated with the pathogenesis of inflammation-related diseases. However, the biological functions of miR-21 in acute lung injury (ALI) remain largely unknown. In this study, the level of miR-21 was obviously increased, but B cell lymphoma-2 (Bcl-2) expression was markedly decreased in LPS-treated human pulmonary alveolar epithelial cells (HPAEpiC). Suppression of miR-21 attenuated LPS-induced apoptosis and inflammation in HPAEpiC and promoted the survival of mice with ALI by decreasing the inflammatory cell count, release of cytokines and permeability in lung tissues. Importantly, Bcl-2 was a direct target of miR-21, and its expression was significantly inhibited by miR-21 mimics at a post-transcriptional level. Besides, Bcl-2 over-expression reversed miR-21-induced apoptosis and inflammation status and showed synergic effects with miR-21 inhibitor in LPS-treated HPAEpiC. In conclusion, inhibition of miR-21 could ameliorate apoptosis and inflammation by restoring the expression of Bcl-2 in LPS-induced HPAEpiC and mice, which might provide therapeutic strategies for the treatment of ALI.
Collapse
Affiliation(s)
- Junke Ge
- Department of Intensive Care Unit, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, PR China
| | - Yanfen Yao
- Department of Intensive Care Unit, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, PR China
| | - Haiyan Jia
- Department of Intensive Care Unit, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, PR China
| | - Pibao Li
- Department of Intensive Care Unit, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, PR China
| | - Wei Sun
- Department of Intensive Care Unit, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, PR China
| |
Collapse
|
7
|
Liu H, Hu K. The Long Intergenic Noncoding RNA 00707 Sponges MicroRNA-613 (miR-613) to Promote Proliferation and Invasion of Gliomas. Technol Cancer Res Treat 2020; 19:1533033820962092. [PMID: 33107401 PMCID: PMC7607719 DOI: 10.1177/1533033820962092] [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] [Indexed: 11/17/2022] Open
Abstract
Background: Glioma is one of the most deadly malignant tumors in humans. Long non-coding RNA (lncRNA) plays a key role in the occurrence, development and invasion of tumors by regulating oncogenic and tumor suppressor pathways. However, the role and action mechanism of long intergenic non-coding RNA 00707 (LINC00707) in gliomas have not been elucidated. This study aimed to investigate the interaction between LINC00707 and miR-613 as well as its role in gliomas. Materials and Methods: The expression levels of LINC00707 and miR-613 were detected by qRT-PCR. The chi-square test was used to analyze the correlation between LINC00707 expression and clinicopathological parameters. CCK-8 and colony formation assays were used to detect glioma cell proliferation; and wound healing and transwell assays were used to detect glioma cell migration and invasion. The relationship between LINC00707 and miR-613 was predicted by Starbase, and verified by qRT-PCR and dual luciferase reporter gene assay. Results: LINC00707 was up-regulated in gliomas. Up-regulated LINC00707 increased the proliferation, migration and invasion of glioma cells, and silenced LINC00707 reduced these abilities. The increase of the expression level of LINC00707 down-regulated miR-613 in glioma cells, while the inhibition of the expression level of LINC00707 up-regulated miR-613 in glioma cells. The high expression of LINC00707 was related to the Karnofsky performance status (KPS) score and WHO staging. LINC00707 could offset the ability of miR-613 to inhibit glioma proliferation and invasion. Conclusion: LINC00707 promotes proliferation and invasion of glioma cells by sponging miR-613. The regulatory axis of LINC00707/miR-613 provides new insights into the mechanism and treatment of gliomas.
Collapse
Affiliation(s)
- Handong Liu
- Department of Neurosurgery, Xiangyang Center Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Keqi Hu
- Department of Neurosurgery, Xiangyang Center Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
8
|
Rezaei O, Honarmand K, Nateghinia S, Taheri M, Ghafouri-Fard S. miRNA signature in glioblastoma: Potential biomarkers and therapeutic targets. Exp Mol Pathol 2020; 117:104550. [PMID: 33010295 DOI: 10.1016/j.yexmp.2020.104550] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/19/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) are transcripts with sizes of about 22 nucleotides, which are produced through a multistep process in the nucleus and cytoplasm. These transcripts modulate the expression of their target genes through binding with certain target regions, particularly 3' suntranslated regions. They are involved in the pathogenesis of several kinds of cancers, such as glioblastoma. Several miRNAs, including miR-10b, miR-21, miR-17-92-cluster, and miR-93, have been up-regulated in glioblastoma cell lines and clinical samples. On the other hand, expression of miR-7, miR-29b, miR-32, miR-34, miR-181 family members, and a number of other miRNAs have been decreased in this type of cancer. In the current review, we explain the role of miRNAs in the pathogenesis of glioblastoma through providing a summary of studies that reported dysregulation of these epigenetic effectors in this kind of brain cancer.
Collapse
Affiliation(s)
- Omidvar Rezaei
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kasra Honarmand
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeedeh Nateghinia
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Du G, Zhou J, Cheng L, Ma X, Gui Y, Tan B. High Expression of miR-206 Predicts Adverse Outcomes: A Potential Therapeutic Target for Esophageal Cancer. Comb Chem High Throughput Screen 2020; 22:599-611. [PMID: 31648633 DOI: 10.2174/1386207322666191018145825] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/11/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND MicroRNA-206 (miR-206) inhibits cell proliferation, invasion and migration in a variety of tumors, but the prognostic value of its Esophageal Cancer (EC) remains unclear. OBJECTIVE To study the role of miR-206 in EC. METHODS The datasets of RNA-Seq, miRNA-Seq, methylation, copy number variation (CNV), and clinical follow-up information were download from The Cancer Genome Atlas (TCGA). After integration and standardization, the prognostic value and potential function of miR-206 were analyzed. The important roles of miR-206 expression in EC genetic and epigenetic mechanisms were analyzed by RNA-Seq, miRNA-Seq, and methylation data. The potential mechanism of CNV in different miR-206 expression groups was analyzed using GISTIC. RESULTS High expression of miR-206 was associated with poor outcome of EC (OS: p=0.005, AUC=0.69, N=178). Transforming growth factor β (TGF-β) signaling pathway, Wnt signaling pathway, mitogen-activated protein kinases (MAPK) signaling pathway, mammalian target of rapamycin (mTOR) signaling pathway were inhibited in high expression group. the aberrant methylation sites in the high and low expression groups were mainly distributed in the promoter region containing CpG islands, and there were different copy number patterns in the H and L samples, and the genes in the differential copy number were mainly enriched in cancer-related pathways, such as thyroid cancer, central carbon metabolism. CONCLUSION This study explored the unique genomic and epigenetic landscape associated with the expression of miR-206, provided evidence of mir-206 as a prognostic biomarker or a potential therapeutic target for EC patients.
Collapse
Affiliation(s)
- Guobo Du
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan 637000, China
| | - Jing Zhou
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan 637000, China
| | - Long Cheng
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan 637000, China
| | - Xiaojie Ma
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan 637000, China
| | - Yan Gui
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan 637000, China
| | - Bangxian Tan
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan 637000, China
| |
Collapse
|
10
|
Hu W, Feng H, Xu X, Huang X, Huang X, Chen W, Hao L, Xia W. Long noncoding RNA FOXD2-AS1 aggravates hepatocellular carcinoma tumorigenesis by regulating the miR-206/MAP3K1 axis. Cancer Med 2020; 9:5620-5631. [PMID: 32558350 PMCID: PMC7402827 DOI: 10.1002/cam4.3204] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 05/09/2020] [Accepted: 05/14/2020] [Indexed: 12/14/2022] Open
Abstract
LncRNAs play crucial roles in the development of various cancers including hepatocellular carcinoma (HCC). Nevertheless, the function of the long noncoding RNA (lncRNA) FOXD2‐AS1 in HCC is still poorly understood. In this study, we focused on the role of FOXD2‐AS1 in HCC. We found that FOXD2‐AS1 was significantly upregulated in HCC cells in comparison to normal human liver cells, LO2. In this study, we also demonstrated that miR‐206 expression was greatly reduced in HCC cells. Furthermore, the inhibition of FOXD2‐AS1 repressed HCC cell proliferation, enhanced cell apoptosis, and restrained cell invasion and migration. The knockdown of FOXD2‐AS1 elevated miR‐206 expression, and we validated an interaction between these RNAs. Additionally, miR‐206 mimics inhibited HCC development while miR‐206 mimics had the opposite effect. MAP kinase 1 (MAP3K1) was predicted to be a target of miR‐206. We discovered that FOXD2‐AS1 modulated MAP3K1 expression by sponging miR‐206 in MHCC‐97L and HepG2 cells. Finally, our in vivo experiments validated that the knockdown of FOXD2‐AS1 inhibited HCC progression by modulating the miR‐206/MAP3K1 axis. In conclusion, this work implies FOXD2‐AS1 accelerates HCC progression through sponging miR‐206 and regulating MAP3K1 expression.
Collapse
Affiliation(s)
- Wei Hu
- Department of Gynecology and Obstetrics Ultrasound, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hui Feng
- Department of Administration Office, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoyu Xu
- Department of Obstetrics, East Hospital of Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin Huang
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xingyue Huang
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenwei Chen
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lidan Hao
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenfang Xia
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
11
|
MiR-206 suppresses proliferation and epithelial-mesenchymal transition of renal cell carcinoma by inhibiting CDK6 expression. Hum Cell 2020; 33:750-758. [PMID: 32277426 DOI: 10.1007/s13577-020-00355-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 03/25/2020] [Indexed: 12/11/2022]
Abstract
Increasing evidence indicates that miRNAs are involved in tumorigenesis of human renal cell carcinoma (RCC). However, the role of miR-206 is still unknown. This study aimed to investigate the possible mechanism of miR-206 in progression of RCC. Here, compared with adjacent normal renal tissues and HK-2 cells, miR-206 level was markedly decreased, whereas CDK6 level was obviously increased in RCC tissues and cell lines. MiR-206 was inversely associated with lymph node metastasis and TNM stage, and acted as an independent prognostic factor in RCC. MiR-206 effectively caused apoptosis and cell cycle arrest at G0/G1 phase, and affected the growth of xenograft tumor of nude mice. MiR-206 also inhibited migration and invasion of RCC cells by modulating the expressions of EMT-related genes. Dual-luciferase assay demonstrated CDK6 was a direct target of miR-206. CDK6 silencing aggravated the inhibition effects of miR-206. In conclusion, miR-206 suppresses proliferation and EMT of RCC by inhibiting CDK6 expression. The miR-206/CDK6 axis may provide a novel insight into tumorigenesis of RCC.
Collapse
|
12
|
Zhuang D, Liang L, Zhang H, Feng X. miR-202 Suppresses Hepatocellular Carcinoma Progression via Downregulating BCL2 Expression. Oncol Res 2020; 28:399-408. [PMID: 32272979 PMCID: PMC7851524 DOI: 10.3727/096504020x15864296270581] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
miRNAs play an important role in progression of hepatocellular carcinoma (HCC). In this work, we assessed the function of miR-202 in human HCC and identified BCL2 as its target. We found miR-202 expression was found significantly downregulated, while BCL2 expression was markedly upregulated in HCC tissues and cell lines (HepG2, Hep3B, and HCCLM3). Both miR-202 and BCL2 were closely correlated with major vascular invasion and advanced TNM stage as well as overall survival of HCC patients. Overexpression of miR-202 significantly inhibited cell proliferation, induced apoptosis and cell cycle arrest at the G0/G1 phase, and prevented tumor formation in a xenograft nude mouse model. Further, miR-202 dramatically inhibited migration, invasion, and epithelial–mesenchymal transition. miR-202 bound to the 3′-untranslated region (3′-UTR) of BCL2 mRNA and downregulated the expression level of BCL2 protein. Exogenous BCL2 overexpression weakened the inhibitory effects of miR-202, while inhibition of BCL2 enhanced the inhibitory effects of miR-202. In conclusion, miR-202 serves as a tumor suppressor in HCC progression by downregulating BCL2 expression, indicating miR-202 might be a potential target for HCC.
Collapse
Affiliation(s)
- Donghai Zhuang
- Department of Hepatobiliary Surgery, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong UniversityJinanP.R. China
| | - Li Liang
- Department of Hepatobiliary Surgery, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong UniversityJinanP.R. China
| | - Hongzhan Zhang
- Department of Hepatobiliary Surgery, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong UniversityJinanP.R. China
| | - Xianguang Feng
- Department of Hepatobiliary Surgery, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong UniversityJinanP.R. China
| |
Collapse
|
13
|
Wang D, Yang T, Liu J, Liu Y, Xing N, He J, Yang J, Ai Y. Propofol Inhibits the Migration and Invasion of Glioma Cells by Blocking the PI3K/AKT Pathway Through miR-206/ROCK1 Axis. Onco Targets Ther 2020; 13:361-370. [PMID: 32021281 PMCID: PMC6969687 DOI: 10.2147/ott.s232601] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/19/2019] [Indexed: 12/12/2022] Open
Abstract
Background Propofol has been identified to perform anti-tumor functions in glioma. However, the molecular mechanisms underlying propofol-induced prevention on migration and invasion of glioma cells remain unclear. Methods Cell proliferation, invasion and migration were measured by 3-(4,5)-dimethylthiahiazo(−z-y1)-3,5-di-phenytetrazoliumromide assay and transwell assay, respectively. The expression of microRNA (miR)-206 and Rho-associated coiled coil-containing protein kinase 1 (ROCK1) was detected by quantitative real-time polymerase chain reaction. Western blot was used to measure the activation of the PI3K/AKT pathway. The interaction between miR-206 and ROCK1 was analyzed using the dual-luciferase reporter assay, RNA immunoprecipitation assay, and pull-down assay. Results Propofol treatment inhibited the migration, invasion, and PI3K/AKT pathway activation in glioma cells. MiR-206 was decreased in glioma tissues and cells, while propofol exposure induced the upregulation of miR-206 in glioma cells. Besides that, we also found overexpressed miR-206 enhanced propofol-mediated inhibition on the migration, invasion, and PI3K/AKT pathway activation of glioma cells. Subsequently, ROCK1 was confirmed to be a target of miR-206. ROCK1 was elevated in glioma tissues and cells, but was reduced by propofol exposure in glioma cells. The rescue assay indicated that the miR-206/ROCK1 axis was involved in propofol-induced inhibition on the migration, invasion, and PI3K/AKT pathway activation in glioma cells. Conclusion Propofol inhibited the migration and invasion of glioma cells by blocking the PI3K/AKT pathway through the miR-206/ROCK1 axis, suggesting an effective clinical implication for the anesthetic to prevent the metastasis of glioma.
Collapse
Affiliation(s)
- Dongmei Wang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Tao Yang
- Department of Anesthesiology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Junqi Liu
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Yafei Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Juan He
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Yanqiu Ai
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| |
Collapse
|
14
|
Wang R, Nakshatri H. Systemic Actions of Breast Cancer Facilitate Functional Limitations. Cancers (Basel) 2020; 12:cancers12010194. [PMID: 31941005 PMCID: PMC7016719 DOI: 10.3390/cancers12010194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is a disease of a specific organ, but its effects are felt throughout the body. The systemic effects of breast cancer can lead to functional limitations in patients who suffer from muscle weakness, fatigue, pain, fibromyalgia, or many other dysfunctions, which hasten cancer-associated death. Mechanistic studies have identified quite a few molecular defects in skeletal muscles that are associated with functional limitations in breast cancer. These include circulating cytokines such as TNF-α, IL-1, IL-6, and TGF-β altering the levels or function of myogenic molecules including PAX7, MyoD, and microRNAs through transcriptional regulators such as NF-κB, STAT3, and SMADs. Molecular defects in breast cancer may also include reduced muscle mitochondrial content and increased extracellular matrix deposition leading to energy imbalance and skeletal muscle fibrosis. This review highlights recent evidence that breast cancer-associated molecular defects mechanistically contribute to functional limitations and further provides insights into therapeutic interventions in managing functional limitations, which in turn may help to improve quality of life in breast cancer patients.
Collapse
Affiliation(s)
- Ruizhong Wang
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- VA Roudebush Medical Center, Indianapolis, IN 46202, USA
- Correspondence: ; Tel.: +1-317-278-2238
| |
Collapse
|
15
|
Huang Z, Zhao X, Wu X, Xiang L, Yuan Y, Zhou S, Yu W. LncRNA UCA1 facilitated cell growth and invasion through the miR-206/CLOCK axis in glioma. Cancer Cell Int 2019; 19:316. [PMID: 31798345 PMCID: PMC6883638 DOI: 10.1186/s12935-019-1023-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 11/11/2019] [Indexed: 01/27/2023] Open
Abstract
Background Glioma is a lethal malignant brain tumor, which affects the brain functions and is life-threatening. LncRNA UCA1 was identified as a pivotal regulator for tumorigenesis of glioma. MiR-206 was discovered to promote tumorigenesis and is critical in the regulation of cell proliferation in glioma. This study will discuss the expression of UCA1 regarding miR-206 and CLOCK, and their integrative effects in the proliferation and cell cycle of glioma cells. Methods qRT-PCR was conducted to measure the mRNA expressions of IgG and Ago2 in cells co-transfected with UCA1, and miR-216 in U251. Bioinformation was analyzed for the prediction of association between UCA1 and miR-206. Transwell migrations assays and invasion assays were utilized to observe the cell invasive ability. Western blot and immunofluorescence imaging were used to examine the protein expressions. In vivo comparisons and observations were also performed to investigate the role of UCA1 in glioma growth. Results LncRNA UCA1 was up-regulated in glioma cell lines and tissues. It elevated cell invasion via the inducing of epithelial-mesenchymal transition. We found that UCA1 can modulate miR-206 expression and serve as an endogenous sponge of miR-206. The EMT-inducer CLOCK was validated as a messenger RNA target of miR-206. At last, we demonstrated that UCA1 exerted the biology function through regulating miR-206 and CLOCK in vivo. Conclusions Overall, the results demonstrated that UCA1/miR-206/CLOCK axis participated in the progressing of glioma and could act as a promising therapeutic target.
Collapse
Affiliation(s)
- Zhi Huang
- 1Department of interventional radiology, The Second Affiliated Hospital of Guizhou Medical University, Guiyang, 556000 Guizhou People's Republic of China.,2Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550005 Guizhou People's Republic of China.,3Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, No. 9 Beijing Road, Guiyang, 550002 Guizhou People's Republic of China
| | - Xuya Zhao
- 2Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550005 Guizhou People's Republic of China
| | - Xiaowen Wu
- 4Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, No. 9 Beijing Road, Guiyang, 550002 Guizhou People's Republic of China
| | - Lei Xiang
- 4Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, No. 9 Beijing Road, Guiyang, 550002 Guizhou People's Republic of China
| | - Yingnan Yuan
- 4Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, No. 9 Beijing Road, Guiyang, 550002 Guizhou People's Republic of China
| | - Shi Zhou
- 4Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, No. 9 Beijing Road, Guiyang, 550002 Guizhou People's Republic of China
| | - Wenfeng Yu
- 3Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, No. 9 Beijing Road, Guiyang, 550002 Guizhou People's Republic of China
| |
Collapse
|
16
|
Janaki Ramaiah M, Divyapriya K, Kartik Kumar S, Rajesh YBRD. Drug-induced modifications and modulations of microRNAs and long non-coding RNAs for future therapy against Glioblastoma Multiforme. Gene 2019; 723:144126. [PMID: 31589963 DOI: 10.1016/j.gene.2019.144126] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 02/07/2023]
Abstract
Non-coding RNAs are known to participate in cancer initiation, progression, and metastasis by regulating the status of chromatin epigenetics and gene expression. Although these non-coding RNAs do not possess defined protein-coding potential, they are involved in the expression and stability of messenger RNA (mRNA). The length of microRNAs (miRs) ranges between 20 and 22 nt, whereas, long non-coding RNAs (lncRNAs) length ranges between 200 nt to 1 Kb. In the case of circular RNAs (circRNAs), the size varies depending upon the length of the exon from where they were derived. Epigenetic regulations of miR and lncRNA genes will influence the gene expression by modulating histone acetylation and methylation patterns. Especially, lncRNAs will act as a scaffold for various epigenetic proteins, such as EZH2 and LSD1, and influence the chromatin epigenetic state at various genomic loci involved at silencing. Thus investigations on the expression of lncRNAs and designing drugs to modulate the expression of these genes will have a profound impact on future therapeutics against cancers such as Glioblastoma Multiforme (GBM) and also against various other diseases. With the recent advancements in genome-wide transcriptomic studies, scientists are focused on the non-coding RNAs and their regulations on various cellular processes involved in GBM and on other types of cancer as well as trying to understand possible epigenetic modulations that help in generating promising therapeutics for the future generations. In this review, the involvement of epigenetic proteins, enzymes that change chromatin architecture and epigenetic landscape and new roles of lncRNAs that are involved in GBM progression are elaborately discussed.
Collapse
Affiliation(s)
- M Janaki Ramaiah
- Laboratory of Functional Genomics and Disease Biology, School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613401, Tamil Nadu, India.
| | - Karthikeyan Divyapriya
- Laboratory of Functional Genomics and Disease Biology, School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613401, Tamil Nadu, India
| | - Sarwareddy Kartik Kumar
- Laboratory of Functional Genomics and Disease Biology, School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613401, Tamil Nadu, India
| | - Y B R D Rajesh
- Organic Synthesis and Catalysis Laboratory, School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613401, Tamil Nadu, India
| |
Collapse
|
17
|
Down-regulation of microRNA-34a-5p promotes trophoblast cell migration and invasion via targetting Smad4. Biosci Rep 2019; 39:BSR20181631. [PMID: 30617054 PMCID: PMC6900429 DOI: 10.1042/bsr20181631] [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: 09/15/2018] [Revised: 12/02/2018] [Accepted: 12/18/2018] [Indexed: 02/07/2023] Open
Abstract
Trophoblastic dysfunction, such as insufficient migration and invasion, is well-known to be correlated with preeclampsia (PE). Recently, microRNAs (miRNAs) have been implicated in diverse biological processes and human diseases, including PE. However, the expression and functions of miRNAs in the progression of PE, especially in the regulation of trophoblast cell migration and invasion remain largely unclear. Here, we compared the miRNAs expression profiles of PE patients with healthy controls using microarray assay and chose a significant increased miRNA-miR-34a-5p for further investigation. Overexpression of miR-34a-5p dramatically reduced migration and invasion in trophoblast HTR-8/SVneo cells, whereas enhanced by its inhibitor. Luciferase activity assay showed that miR-34a-5p directly target Smad family member 4 (Smad4), which is associated with cancer cell invasiveness and metastasis. We also found that Smad4 was down-regulated in PE patients, and an inverse relationship between Smad4 and miR-34a-5p expression levels was observed in placental tissues from PE patients. Further study showed that knockdown of Smad4 effectively attenuated the promoting effects of miR-34a-5p inhibition on the migration and invasion of HTR-8/SVneo cells. Taken together, these findings suggest that inhibition of miR-34a-5p improves invasion and migration of trophoblast cells by directly targetting Smad4, which indicated the potential of miR-34a-5p as a therapeutic target against PE.
Collapse
|
18
|
Pourbagheri-Sigaroodi A, Bashash D, Safaroghli-Azar A, Farshi-Paraasghari M, Momeny M, Mansoor FN, Ghaffari SH. Contributory role of microRNAs in anti-cancer effects of small molecule inhibitor of telomerase (BIBR1532) on acute promyelocytic leukemia cell line. Eur J Pharmacol 2019; 846:49-62. [PMID: 30658112 DOI: 10.1016/j.ejphar.2019.01.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 01/01/2019] [Accepted: 01/14/2019] [Indexed: 12/29/2022]
Abstract
Telomerase-mediated immortalization and proliferation of tumor cells is a promising anti-cancer treatment strategy and development of potent telomerase inhibitors is believed to open new window of treatments in human malignancies. In the present study, we found that BIBR1532, a small molecule inhibitor of human telomerase, exerted cytotoxic effects on a panel of human cancer cells spanning from solid tumors to hematologic malignancies; however, as compared with solid tumors, leukemic cells were more sensitive to this inhibitor. This was independent of molecular status of p53 in the leukemic cells. The results of a miRNA PCR array revealed that BIBR1532-induced cytotoxic effects in NB4, the most sensitive cell line, was coupled with alteration in a substantial number of cancer-related miRNAs. Interestingly, most of these miRNAs were found to act as tumor suppressors with validated targets in cell cycle or nuclear factor (NF)-κB-mediated apoptosis. In accordance with a bioinformatics analysis, our experimental studies showed that BIBR1532-induced apoptosis is mediated, at least partly, by inhibition of NF-κB. Moreover, we found that the alteration in the expression of miRNAs was coupled with the alteration in the cell cycle progression. To sum up with, a straightforward interpretation of our results is that telomerase inhibition using BIBR1532 not only induced CDKN1A-mediated G1 arrest in NB4, but also resulted in a caspase-3-dependent apoptotic cell death mostly through suppression of NF-κB axis.
Collapse
Affiliation(s)
- Atieh Pourbagheri-Sigaroodi
- Department of Biotechnology, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ava Safaroghli-Azar
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Farshi-Paraasghari
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Momeny
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Fahimeh Nemati Mansoor
- Department of Biotechnology, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Seyed H Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Cai KT, Feng CX, Zhao JC, He RQ, Ma J, Zhong JC. Upregulated miR‑203a‑3p and its potential molecular mechanism in breast cancer: A study based on bioinformatics analyses and a comprehensive meta‑analysis. Mol Med Rep 2018; 18:4994-5008. [PMID: 30320391 PMCID: PMC6236224 DOI: 10.3892/mmr.2018.9543] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 08/31/2018] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC) has been identified as the leading malignancy in women worldwide. However, the potential molecular mechanism of microRNA (miR)‑203a‑3p in BC remains to be elucidated. The present study evaluated the expression of miR‑203a‑3p in BC and adjacent normal tissue in several publically available datasets. The distinguishability of precursor miR‑203a and miR‑203a‑3p in BC tissue and adjacent breast tissue was assessed using receiver operating characteristic (ROC) and summarized ROC (sROC) approaches. In addition, gene ontology (GO) enrichment, Kyoto Encyclopedia of Genes and Genomes pathway analysis and protein‑protein interaction analysis were performed to determine the potential molecular mechanism of miR‑203a‑3p in BC. It was identified that the expression of precursor miR‑203a was markedly upregulated in 1,077 BC tissue samples compared to 104 adjacent breast tissue samples from The Cancer Genome Atlas. Additionally, an increasing trend in miR‑203a‑3p expression was observed in 756 BC tissue samples compared with 76 adjacent breast tissue samples from the University of California Santa Cruz Xena project. In addition, a comprehensive meta‑analysis suggested that the expression of miR‑203a‑3p was markedly increased in 2,444 BC tissue samples compared with 559 adjacent breast tissue samples. The area under the curve of the ROC and sROC revealed that miR‑203a‑3p expression was able to distinguish between BC tissue and adjacent breast tissue. However, miR‑203a‑3p exhibited no prognostic value in BC. The results of GO enrichment demonstrated that the miR‑203a target genes were associated with 'plasma membrane integrity', 'cell surface receptor linked signal and transduction' and '3',5'‑cyclic nucleotide phosphodiesterase activity'. 'Purine metabolism' was identified as the pathway with the most enrichment of miR‑203a‑3p target genes in BC. The present study also identified insulin‑like growth factor receptor (IGF1) as a hub gene associated with miR‑203a in BC. In summary, miR‑203a‑3p may enhance the development and oncogenesis of BC, and IGF1 was defined as a hub gene of miR‑203a‑3p in BC.
Collapse
Affiliation(s)
- Kai-Teng Cai
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Cai-Xia Feng
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jin-Che Zhao
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Rong-Quan He
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jie Ma
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jin-Cai Zhong
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
20
|
MiRNA-206 suppresses PGE2-induced colorectal cancer cell proliferation, migration, and invasion by targetting TM4SF1. Biosci Rep 2018; 38:BSR20180664. [PMID: 30135139 PMCID: PMC6146292 DOI: 10.1042/bsr20180664] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/10/2018] [Accepted: 08/20/2018] [Indexed: 12/21/2022] Open
Abstract
MiRNA (miR)-206 plays a tumor suppressor role in various cancer types. Here, we investigated whether miR-206 is involved in prostaglandin E2 (PGE2)-induced epithelial–mesenchymal transition (EMT) in colorectal cancer (CRC) cells through the targetting of transmembrane 4 L six family member 1 (TM4SF1). The effect of PGE2 on growth and apoptosis of CRC cells was evaluated using the MTT assay and flow cytometry analysis, respectively. TM4SF1 and miR-206 expression levels were determined with quantitative polymerase chain reaction (qRT-PCR) in CRC tissues and cell lines. The concentration of PGE2 in the serum of CRC patients and healthy controls was measured with an ELISA kit. A miR-206 or TM4SF1 construct was transfected into cells with PGE2. Transwell migration and invasion assays were used to examine cell migration and invasion properties. Additionally, a luciferase assay was performed to determine whether TM4SF1 was directly targetted by miR-206. We found that miR-206 was down-regulated and TM4SF1 was up-regulated in human CRC tissues and cell lines. Moreover, miR-206 was negatively correlated with TM4SF1 expression. Bioinformatics analysis and a luciferase reporter assay revealed that miR-206 directly targetted the 3′-untranslated region (UTR) of TM4SF1, and TM4SF1 expression was reduced by miR-206 overexpression at both the mRNA and protein levels. Additionally, PGE2 significantly suppressed the expression of miR-206 and increased the expression of TM4SF1 in CRC cells. PGE2 induction led to enhanced CRC cell proliferation, migration, and invasion. Moreover, the overexpression of miR-206 decreased CRC cell proliferation, migration, and invasion compared with control group in PGE2-induced cells, and these effects could be recovered by the overexpression of TM4SF1. Overexpression of miR-206 also suppressed the expression of β-catenin, VEGF, MMP-9, Snail, and Vimentin and enhanced E-cadherin expression in PGE2-induced cells. These results could be reversed by the overexpression of TM4SF1. At last, up-regulation of miR-206 suppressed expression of p-AKT and p-ERK by targetting TM4SF1 in PGE2-induced cells. Our results provide further evidence that miR-206 has a protective effect on PGE2-induced colon carcinogenesis.
Collapse
|
21
|
Post-Transcriptional Regulation of Anti-Apoptotic BCL2 Family Members. Int J Mol Sci 2018; 19:ijms19010308. [PMID: 29361709 PMCID: PMC5796252 DOI: 10.3390/ijms19010308] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/05/2018] [Accepted: 01/16/2018] [Indexed: 12/20/2022] Open
Abstract
Anti-apoptotic B cell lymphoma 2 (BCL2) family members (BCL2, MCL1, BCLxL, BCLW, and BFL1) are key players in the regulation of intrinsic apoptosis. Dysregulation of these proteins not only impairs normal development, but also contributes to tumor progression and resistance to various anti-cancer therapies. Therefore, cells maintain strict control over the expression of anti-apoptotic BCL2 family members using multiple mechanisms. Over the past two decades, the importance of post-transcriptional regulation of mRNA in controlling gene expression and its impact on normal homeostasis and disease have begun to be appreciated. In this review, we discuss the RNA binding proteins (RBPs) and microRNAs (miRNAs) that mediate post-transcriptional regulation of the anti-apoptotic BCL2 family members. We describe their roles and impact on alternative splicing, mRNA turnover, and mRNA subcellular localization. We also point out the importance of future studies in characterizing the crosstalk between RBPs and miRNAs in regulating anti-apoptotic BCL2 family member expression and ultimately apoptosis.
Collapse
|
22
|
Pang C, Huang G, Luo K, Dong Y, He F, Du G, Xiao M, Cai W. miR-206 inhibits the growth of hepatocellular carcinoma cells via targeting CDK9. Cancer Med 2017; 6:2398-2409. [PMID: 28940993 PMCID: PMC5633544 DOI: 10.1002/cam4.1188] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 07/22/2017] [Accepted: 08/12/2017] [Indexed: 12/24/2022] Open
Abstract
miR‐206 plays an important role in regulating the growth of multiple cancer cells. Cyclin‐dependent kinase 9 (CDK9) stimulates the production of abundant prosurvival proteins, leading to impaired apoptosis of cancer cells. However, it is unknown whether CDK9 is involved in the miR‐206‐mediated growth suppression of hepatocellular carcinoma (HCC) cells. In this study, we found that the expression level of miR‐206 was significantly lower in HCC cell lines than that in normal hepatic cell line (L02). Meanwhile, CDK9 was upregulated in HCC cell lines. Moreover, miR‐206 downregulated CDK9 in HCC cells via directly binding to its mRNA 3′ UTR, which resulted in a decrease of RNA PolII Ser2 phosphorylation and Mcl‐1 level. Additionally, miR‐206 suppressed the cell proliferation, and induced cell cycle arrest and apoptosis. Similarly, silence or inhibition of CDK9 also repressed the cell proliferation, and induced cell cycle arrest and apoptosis. Taken together, the results demonstrated that miR‐206 inhibited the growth of HCC cells through targeting CDK9, suggesting that the miR‐206‐CDK9 pathway may be a novel target for the treatment of HCC.
Collapse
Affiliation(s)
- Chi Pang
- Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, 570102, China
| | - Gang Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China
| | - Kaili Luo
- Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, 570102, China
| | - Yuying Dong
- Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, 570102, China
| | - Fengtian He
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China
| | - Guankui Du
- Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, 570102, China
| | - Man Xiao
- Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, 570102, China
| | - Wangwei Cai
- Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, 570102, China
| |
Collapse
|