1
|
Anoushirvani AA, Jafarian Yazdi A, Amirabadi S, Asouri SA, Shafabakhsh R, Sheida A, Hosseini Khabr MS, Jafari A, Tamehri Zadeh SS, Hamblin MR, Kalantari L, Talaei Zavareh SA, Mirzaei H. Role of non-coding RNAs in neuroblastoma. Cancer Gene Ther 2023; 30:1190-1208. [PMID: 37217790 DOI: 10.1038/s41417-023-00623-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 03/25/2023] [Accepted: 05/04/2023] [Indexed: 05/24/2023]
Abstract
Neuroblastoma is known as the most prevalent extracranial malignancy in childhood with a neural crest origin. It has been widely accepted that non-coding RNAs (ncRNAs) play important roles in many types of cancer, including glioma and gastrointestinal cancers. They may regulate the cancer gene network. According to recent sequencing and profiling studies, ncRNAs genes are deregulated in human cancers via deletion, amplification, abnormal epigenetic, or transcriptional regulation. Disturbances in the expression of ncRNAs may act either as oncogenes or as anti-tumor suppressor genes, and can lead to the induction of cancer hallmarks. ncRNAs can be secreted from tumor cells inside exosomes, where they can be transferred to other cells to affect their function. However, these topics still need more study to clarify their exact roles, so the present review addresses different roles and functions of ncRNAs in neuroblastoma.
Collapse
Affiliation(s)
- Ali Arash Anoushirvani
- Department of Internal Medicine, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Sanaz Amirabadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sahar Ahmadi Asouri
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University, Kashan, Iran
| | - Rana Shafabakhsh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University, Kashan, Iran
| | - Amirhossein Sheida
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Sadat Hosseini Khabr
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ameneh Jafari
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, P.O. BOX: 15179/64311, Tehran, Iran
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Leila Kalantari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| | | | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University, Kashan, Iran.
| |
Collapse
|
2
|
Corallo D, Dalla Vecchia M, Lazic D, Taschner-Mandl S, Biffi A, Aveic S. The molecular basis of tumor metastasis and current approaches to decode targeted migration-promoting events in pediatric neuroblastoma. Biochem Pharmacol 2023; 215:115696. [PMID: 37481138 DOI: 10.1016/j.bcp.2023.115696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 07/24/2023]
Abstract
Cell motility is a crucial biological process that plays a critical role in the development of multicellular organisms and is essential for tissue formation and regeneration. However, uncontrolled cell motility can lead to the development of various diseases, including neoplasms. In this review, we discuss recent advances in the discovery of regulatory mechanisms underlying the metastatic spread of neuroblastoma, a solid pediatric tumor that originates in the embryonic migratory cells of the neural crest. The highly motile phenotype of metastatic neuroblastoma cells requires targeting of intracellular and extracellular processes, that, if affected, would be helpful for the treatment of high-risk patients with neuroblastoma, for whom current therapies remain inadequate. Development of new potentially migration-inhibiting compounds and standardized preclinical approaches for the selection of anti-metastatic drugs in neuroblastoma will also be discussed.
Collapse
Affiliation(s)
- Diana Corallo
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padova, Italy
| | - Marco Dalla Vecchia
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padova, Italy
| | - Daria Lazic
- St. Anna Children's Cancer Research Institute, CCRI, Zimmermannplatz 10, 1090, Vienna, Austria
| | - Sabine Taschner-Mandl
- St. Anna Children's Cancer Research Institute, CCRI, Zimmermannplatz 10, 1090, Vienna, Austria
| | - Alessandra Biffi
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Woman's and Child Health Department, University of Padova, 35121 Padova, Italy
| | - Sanja Aveic
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padova, Italy.
| |
Collapse
|
3
|
Deciphering the Role of p53 and TAp73 in Neuroblastoma: From Pathogenesis to Treatment. Cancers (Basel) 2022; 14:cancers14246212. [PMID: 36551697 PMCID: PMC9777536 DOI: 10.3390/cancers14246212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Neuroblastoma (NB) is an embryonic cancer that develops from neural crest stem cells, being one of the most common malignancies in children. The clinical manifestation of this disease is highly variable, ranging from spontaneous regression to increased aggressiveness, which makes it a major therapeutic challenge in pediatric oncology. The p53 family proteins p53 and TAp73 play a key role in protecting cells against genomic instability and malignant transformation. However, in NB, their activities are commonly inhibited by interacting proteins such as murine double minute (MDM)2 and MDMX, mutant p53, ΔNp73, Itch, and Aurora kinase A. The interplay between the p53/TAp73 pathway and N-MYC, a known biomarker of poor prognosis and drug resistance in NB, also proves to be decisive in the pathogenesis of this tumor. More recently, a strong crosstalk between microRNAs (miRNAs) and p53/TAp73 has been established, which has been the focused of great attention because of its potential for developing new therapeutic strategies. Collectively, this review provides an updated overview about the critical role of the p53/TAp73 pathway in the pathogenesis of NB, highlighting encouraging clues for the advance of alternative NB targeted therapies.
Collapse
|
4
|
Tang F, Jiang X, Liao S, Liu Y, He M. Construction of a transcription factor-miRNA-mRNA interactive network elucidates underlying pathogenesis for osteosarcoma and validation by qRT-PCR. Medicine (Baltimore) 2022; 101:e31049. [PMID: 36254052 PMCID: PMC9575767 DOI: 10.1097/md.0000000000031049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Osteosarcoma is characterized by features of rapid growth and early metastasis with a poor prognosis. The aim of our research is to investigate the potential transcription factor (TF)-miRNA-mRNA regulatory mechanism in osteosarcoma utilizing bioinformatics methods and validate by qRT-PCR. METHODS The microRNA (miRNA) expression profiling datasets (GSE28423 and GSE65071) and mRNA expression profiling dataset GSE33382 were collected from the Gene Expression Omnibus (GEO) database. Differentially expressed miRNAs (DEMs) and differentially expressed genes (DEGs) were screened using the limma package. Then, the TransmiR v2.0, miRDB, and Targetscan 7.2 database were applied for the acquisition of TF-miRNA and miRNA-mRNA interaction relationships, respectively. Finally, we built a TF-miRNA-mRNA interactive network. Furthermore, survival analysis was performed to identify sub-network with prognostic value and validate through qRT-PCR. RESULTS Eight overlapping DEMs and 682 DEGs were identified. Based on bioinformatics methods, 30 TF-miRNA interaction pairs and 25 miRNA-mRNA interaction pairs were screened. Finally, we constructed a TF-miRNA-mRNA regulatory network. Furthermore, laminin subunit gamma 1 (LAMC1) and thrombospondin-1 (THBS1), which involved in the network, were determined to have prognostic value and the corresponding subnetwork was identified. qRT-PCR results showed that LAMC1 mRNA expression was higher in osteosarcoma cells. CONCLUSION Based on the survival analysis, a TF-miRNA-mRNA sub-network, that is TFs (SPI1, HEY1, and CEBPB)-hsa-miR-338-3p-target genes (LAMC1 and THBS1) was established. In conclusion, the construction of a potential TF-related regulatory network will help elucidate the underlying pathological mechanisms of osteosarcoma, and may provide novel insights for the diagnosis and treatment of osteosarcoma.
Collapse
Affiliation(s)
- Fuxing Tang
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, China
| | - Xiaohong Jiang
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, China
| | - Shijie Liao
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, China
| | - Yun Liu
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, China
| | - Maolin He
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, China
- *Correspondence: Maolin He, Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China (e-mail: )
| |
Collapse
|
5
|
Zhou Y, Tang X, Huang Z, Wen J, Xiang Q, Liu D. KLF5 promotes KIF1A expression through transcriptional repression of microRNA-338 in the development of pediatric neuroblastoma. J Pediatr Surg 2022; 57:192-201. [PMID: 35033353 DOI: 10.1016/j.jpedsurg.2021.12.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/16/2021] [Accepted: 12/20/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Neuroblastoma (NB) comprises about 8-10% of pediatric cancers, and microRNA (miR)-338 downregulation has been implicated in NB. However, the underlying molecular mechanism remains largely unclear. The main goal of this study is to probe the regulatory role of miR-338 and the upstream and downstream biomolecules involved in NB. METHODS The differentially expressed miRNAs were screened by analyzing the NB gene expression microarray GSE121513 from the GEO database, and the differences in expression of the screened miRNAs were verified in clinically collected NB tissues versus dorsal root ganglions. Subsequently, the relationship between the miR-338 expression and NB cell growth was validated in vitro and in vivo, and the upstream and downstream regulatory mechanisms of miR-338 were further analyzed by bioinformatics. Functional rescue experiments were used to verify their effects on NB cell growth. RESULTS miR-338 expressed poorly in NB tissues, and overexpression of miR-338 significantly inhibited NB cell growth in vitro and in vivo. The prediction results showed that miR-338 could target KIF1A, and miR-338 expression was negatively correlated with the expression of KIF1A. We further found that miR-338 was transcriptionally regulated by the transcription factor KLF5. Overexpression of KLF5 or KIF1A significantly attenuated the inhibitory effect of miR-338 mimic on NB cell growth. Finally, miR-338 blocked the Hedgehog signaling pathway by inhibiting the expression of KIF1A. CONCLUSION Overexpression of KLF5 reduced expression of miR-338, which in turn increased the expression of KIF1A and activated the Hedgehog signaling pathway, leading to the progression of NB.
Collapse
Affiliation(s)
- Yuxiang Zhou
- Department of Pediatric Surgery, Hunan Children's Hospital, No. 86, Ziyuan Road, Yuhua District, Changsha, Hunan 410007, PR China
| | - Xianglian Tang
- Department of Pediatric Surgery, Hunan Children's Hospital, No. 86, Ziyuan Road, Yuhua District, Changsha, Hunan 410007, PR China
| | - Zhao Huang
- Department of Pediatric Surgery, Hunan Children's Hospital, No. 86, Ziyuan Road, Yuhua District, Changsha, Hunan 410007, PR China
| | - Jiabing Wen
- Department of Pediatric Surgery, Hunan Children's Hospital, No. 86, Ziyuan Road, Yuhua District, Changsha, Hunan 410007, PR China
| | - Qiangxing Xiang
- Department of Pediatric Surgery, Hunan Children's Hospital, No. 86, Ziyuan Road, Yuhua District, Changsha, Hunan 410007, PR China
| | - Denghui Liu
- Department of Pediatric Surgery, Hunan Children's Hospital, No. 86, Ziyuan Road, Yuhua District, Changsha, Hunan 410007, PR China.
| |
Collapse
|
6
|
miR-338-3p Plays a Significant Role in Casticin-Induced Suppression of Acute Myeloid Leukemia via Targeting PI3K/Akt Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9214130. [PMID: 35765408 PMCID: PMC9233736 DOI: 10.1155/2022/9214130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 11/29/2022]
Abstract
Objective Casticin is generally used in traditional herbal medicine for its anti-inflammatory and anticarcinogenic pharmacological properties. Also, microRNAs are indispensable oncogenes or cancer suppressors being dysregulated in various diseases. In this study, we aimed to elucidate the mechanisms underlying effects of casticin on the progression of acute myeloid leukemia (AML). Methods CCK-8 and flow cytometry were utilized to measure the proliferation and apoptosis of AML cell lines, respectively, after treatment with different concentrations of casticin. The alteration of several microRNA expressions in response to casticin treatment was detected by performing qRT-PCR, and the activity of PI3K/Akt pathways was evaluated through immunoblotting. Afterwards, the potential target gene of miR-338-3p was investigated by dual-luciferase reporter assay. In order to evaluate the role of miR-338-3p in the casticin-induced cellular phenotype changes, AML cells were transfected with miR-338-3p mimics or inhibitor and then subjected to proliferation and apoptosis analysis. Finally, a mouse xenograft model system was employed to investigate the role of casticin in AML progression in vivo. Results Suppressed cellular proliferation and enhanced apoptosis were observed in HL-60 and THP-1 cells after exposure to casticin, accompanied by remarkable upregulation of the miR-338-3p expression as well as a decline in the phosphorylation of PI3K and Akt proteins. RUNX2 was identified as a direct target molecular of miR-338-3p, which might account for the findings that miR-338-3p knockdown enhanced the PI3K/Akt pathway activity, whereas the miR-338-3p overexpression inactivated this signaling pathway. In addition, the inhibition of the miR-338-3p expression attenuated severe cell apoptosis and suppressions of PI3K/Akt pathway induced by casticin. Furthermore, casticin treatment retarded tumor growth rate in mouse models, whilst elevating miR-338 expression and repressing the activity of PI3K/Akt pathway in vivo. However, miR-338-3p depletion could also abolish the phenotypic alterations caused by casticin treatment. Conclusion Casticin promotes AML cell apoptosis but inhibits AML cell proliferation in vitro and tumor growth in vivo by upregulating miR-338-3p, which targets RUNX2 and thereafter inactivates PI3K-Akt signaling pathway. Our results provide insights into the mechanisms underlying the action of casticin in the control of AML progression.
Collapse
|
7
|
Veeraraghavan VP, Jayaraman S, Rengasamy G, Mony U, Ganapathy DM, Geetha RV, Sekar D. Deciphering the Role of MicroRNAs in Neuroblastoma. MOLECULES (BASEL, SWITZERLAND) 2021; 27:molecules27010099. [PMID: 35011335 PMCID: PMC8746473 DOI: 10.3390/molecules27010099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/19/2021] [Accepted: 12/21/2021] [Indexed: 01/31/2023]
Abstract
Neuroblastoma (NB) is a type of peripheral sympathetic nervous system cancer that most commonly affects children. It is caused by the improper differentiation of primitive neural crest cells during embryonic development. Although NB occurs for 8% of paediatric cancers, it accounts for 15% of cancer-related deaths. Despite a considerable increase in cytotoxic chemo- and radiotherapy, patients in advanced stages remain virtually incurable. Therefore, there is a desperate necessity for new treatment strategies to be investigated. Accumulating evidence suggested that microRNAs (miRNAs) are a class of non-coding RNAs with 19–25 nucleotides lengths and play a central role in the development of NB carcinogenesis. Fascinatingly, miRNA inhibitors have an antisense property that can inhibit miRNA function and suppress the activity of mature miRNA. However, many studies have addressed miRNA inhibition in the treatment of NB, but their molecular mechanisms and signalling pathways are yet to be analysed. In this study, we impart the current state of knowledge about the role of miRNA inhibition in the aetiology of NB.
Collapse
Affiliation(s)
- Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600 077, India; (V.P.V.); (G.R.); (U.M.)
| | - Selvaraj Jayaraman
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600 077, India; (V.P.V.); (G.R.); (U.M.)
- Correspondence: (S.J.); (D.S.); Tel.: +91-7397331154 (S.J.); +91-9361216583 (D.S.)
| | - Gayathri Rengasamy
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600 077, India; (V.P.V.); (G.R.); (U.M.)
| | - Ullas Mony
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600 077, India; (V.P.V.); (G.R.); (U.M.)
| | - Dhanraj M Ganapathy
- Department of Prosthodontics, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600 077, India;
| | - Royapuram Veeraragavan Geetha
- Department of Microbiology, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600 077, India;
| | - Durairaj Sekar
- Cellular and Molecular Research Centre, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600 077, India
- Correspondence: (S.J.); (D.S.); Tel.: +91-7397331154 (S.J.); +91-9361216583 (D.S.)
| |
Collapse
|
8
|
Kuai J, Zheng L, Yi X, Liu Z, Qiu B, Lu Z, Jiang Y. ST8SIA6-AS1 promotes the development of hepatocellular carcinoma cells through miR-338-3p/NONO Axis. Dig Liver Dis 2021; 53:1192-1200. [PMID: 33722502 DOI: 10.1016/j.dld.2021.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 01/19/2021] [Accepted: 02/09/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Increasing studies have shown a vital fact that long non-coding RNAs (lncRNAs) play a considerable regulatory role in hepatocellular carcinoma (HCC) progression. However, whether ST8 alpha-N-acetyl-neuraminide alpha-2, 8-sialyltransferase 6 antisense RNA 1 (ST8SIA6-AS1) affects the development of HCC is unclear. METHODS The target genes in HCC cell lines were quantified via utilzing quantitative real-time polymerase chain reaction (RT-qPCR) analysis and western blot. Effects of ST8SIA6-AS1 on proliferative, apoptosis and migratory ability of HCC cells were proved by a series of function experiments. The cellular distribution of ST8SIA6-AS1 was examined through fluorescent in situ hybridization (FISH) assay and subcellular fractionation experiments. RNA pulldown assay was implemented to explore the target of ST8SIA6-AS1. RNA Binding Protein Immunoprecipitation (RIP) and luciferase reporter assays were performed to identify the specific relationships between miR-338-3p and ST8SIA6-AS1/ non-POU domain containing octamer binding (NONO). RESULTS The expression of ST8SIA6-AS1 was apparently elevated in HCC cell. Silenced ST8SIA6-AS1 reduced proliferative, migratory and invasive ability of HCC cells. Moreover, ST8SIA6-AS1 targeted miR-338-3p to modulate the expression of NONO in HCC cells. CONCLUSIONS ST8SIA6-AS1 enhances the progression of HCC via miR-338-3p/NONO axis in vitro.
Collapse
Affiliation(s)
- Jinghua Kuai
- Department of Gastroenterology, Qilu Hospital of Shandong University, Qingdao 266000, Shandong, China
| | - Lijie Zheng
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China.
| | - Xin Yi
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China
| | - Zengli Liu
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China
| | - Bo Qiu
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China
| | - Zhihua Lu
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China
| | - Yuanhui Jiang
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China
| |
Collapse
|
9
|
Phosphatidylinositol 3,4,5-Trisphosphate-Dependent Rac Exchanger 2 Protein Facilitates Glioma Progression via Akt and Stat3 Signaling. J Mol Neurosci 2021; 71:1674-1682. [PMID: 34322848 DOI: 10.1007/s12031-021-01883-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/29/2021] [Indexed: 10/20/2022]
Abstract
Glioblastoma multiforme (GBM) is the recognized as the most aggressive brain tumor with poor prognosis and low 1-year and 5-year survival rate. The treatment methods for GBM are limited and inefficient, and novel strategies for GBM treatment are urgently warranted. MiR-338-3p is described as a tumor suppressor in a variety of malignancies, including GBM. However, its role in GBM is not fully understood. The mRNA or protein levels of targets in cells or tissues were determined by quantitative reverse transcription PCR (RT-qPCR) or Western blot, respectively. The GBM cell growth rate in vitro or in vivo was measured by Cell Counting Kit-8 or bioluminescence imaging, respectively. Upregulation of hsa-miR-338-3p and downregulation of phosphatidylinositol 3,4,5-trisphosphate-dependent Rac exchanger 2 protein (Prex2) were observed in GBM tissues compared to normal brain tissues. We further confirmed that murine Prex2 was a target of mmu-miR-338-3p in GBM. Mmu-miR-338-3p exerted profound inhibition effects on GBM cell growth in vitro or in vivo through targeting Prex2, leading to attenuation of (Protein kinase B) AKT/Signal transducer and activator of transcription 3 (STAT3) signaling activation. Restoration of mmu-miR-338-3p or inhibition of Prex2 may facilitate the development of innovative therapies for GBM treatment.
Collapse
|
10
|
Hu F, Jiang J, Yu G, Zang H, Sun H. Propofol Pretreatment Prevents Oxygen-Glucose Deprivation/Reoxygenation (OGD/R)-induced Inflammation Through Nuclear Transcription Factor κB (NF-κB) Pathway in Neuroblastoma Cells. Curr Neurovasc Res 2021; 17:27-34. [PMID: 31880261 DOI: 10.2174/1567202617666191227110158] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 11/17/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Inflammation is one of the causes of neuroblastoma progression. Propofol attenuates inflammation by repressing nuclear transcription factor κB (NF-κB) in different diseases. But its effect on oxygen-glucose deprivation/reoxygenation (OGD/R)-induced inflammation is not known. OBJECTIVE This study investigated the role and mechanism of action of propofol on OGD/Rinduced inflammation in mouse N2A neuroblastoma cells. METHODS MTT was performed on mouse neuroblastoma cells N2A to assess and select the maximum safe dose of propofol. Next, N2A cells were pretreated with propofol and then, exposed to the OGD condition for 3 h and reoxygenated for 6 h. The content of the inflammatory factors, interleukin-1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α), in the medium was measured by ELISA, while their protein expression was detected by western blot and immunofluorescence. The protein expression of P65, p-P65, IKBα and p-IKBα belonging to the NF-κB pathway was also determined by western blot in N2A cells. To further confirm the mechanism of propofol on OGD/R-induced inflammation in mouse N2A cells, P65 was over-expressed and the above experiments were repeated. RESULTS Propofol did not affect cell viability of N2A cells even at the maximum concentration used (30 µM), thus, 30 µM of propofol was selected to perform our experiments. Besides, OGD/R induced inflammation and activation of NF-κB pathway with increased p-P65 and p-IKBα expression, and propofol pretreatment inhibited OGD/R induced inflammation and activation of NF-κB pathway in N2A cells. Over-expression of P56 abolished the effects of propofol on OGD/Rinduced inflammation and activation of NF-κB pathway in N2A cells. CONCLUSION Our work demonstrated for the first time that propofol pretreatment ameliorated OGD/R induced inflammation via NF-κB pathway modulation in mouse neuroblastoma N2A cells, indicating that propofol might be considered as a potential therapeutic approach to reduce inflammation in neuroblastoma.
Collapse
Affiliation(s)
- Fang Hu
- Department of Anesthesiology, The First People's Hospital of Fuyang Hangzhou, Hangzhou 311400, China
| | - Jianhua Jiang
- Endoscopy Center, The First People's Hospital of Fuyang Hangzhou, Hangzhou 311400, China
| | - Guocan Yu
- Department of Anesthesiology, The First People's Hospital of Fuyang Hangzhou, Hangzhou 311400, China
| | - Hongcheng Zang
- Department of Anesthesiology, The First People's Hospital of Fuyang Hangzhou, Hangzhou 311400, China
| | - Haoliang Sun
- Department of Anesthesiology, The First People's Hospital of Fuyang Hangzhou, Hangzhou 311400, China
| |
Collapse
|
11
|
MiR-338-3p inhibits cell migration and invasion in human hypopharyngeal cancer via downregulation of ADAM17. Anticancer Drugs 2021; 31:925-931. [PMID: 32889897 DOI: 10.1097/cad.0000000000000919] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Studies have confirmed that microRNAs play important roles in the development and progression of cancer. Therefore, to identify the differentially expressed microRNAs between the cancer and the normal tissues, microRNAs will provide new clues for exploring the molecular mechanisms of cancer development and potential targeted therapies. In the present study, we found that miR-338-3p was downregulated in hypopharyngeal carcinoma and inversely correlated with the pathological grade. When the miR-338-3p was further downregulated, the migration and invasion ability of the FaDu hypopharyngeal carcinoma cells were enhanced, and these functions were inhibited when the miR-338-3p was upregulated. In addition, we demonstrated that ADAM17 was a target of miR-338-3p, and that ADAM17 directly activated the wnt/β-catenin signaling pathway and promoted the expression of its target gene MMP2, Nanog and SOX2, which affected the growth, migration and invasion of hypopharyngeal carcinoma cells. In conclusion, our results demonstrate for the first time that miR-338-3p targets ADAM17 and blocks the development of hypopharyngeal carcinoma involving the wnt/β-catenin signaling pathway, which may be a new target for clinical intervention in human cancer.
Collapse
|
12
|
Yang G, Cui M, Jiang W, Sheng J, Yang Y, Zhang X. Molecular switch in human diseases-disintegrin and metalloproteinases, ADAM17. Aging (Albany NY) 2021; 13:16859-16872. [PMID: 34182543 PMCID: PMC8266367 DOI: 10.18632/aging.203200] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023]
Abstract
The ADAMs (a disintegrin and metalloproteinase) are a family of cell surface proteins with crucial roles in the regulation of cell adhesion, cell proliferation to migration, proteolysis and cell signaling transduction pathways. Among these enzymes, the ADAM17 shows significant effects in the “ectodomain shedding” of its substrates such as cytokines (e.g., tumor necrosis factor α, TNFα), growth factors (e.g., epidermal growth factor, EGF), adhesion proteins (e.g., L-selectin), and their receptors (e.g., IL-6R and TNFα). Several studies focus on the underlying molecular mechanisms of ADAM17 in diseased conditions. Here, we took several different approaches to elucidate the function of ADAM17, the participation of ADAM17 in several human diseases, and the potential as targeted therapy reagents. As more and more studies verify the miRNA-mediated expression variation of ADAM17, the specific regulation network of miRNAs and ADAM17 was exploited in this review as well.
Collapse
Affiliation(s)
- Guang Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Mengying Cui
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Weibo Jiang
- Department of Orthopaedic, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Jiyao Sheng
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Yongsheng Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Xuewen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| |
Collapse
|
13
|
Yan S, Chen J, Zhang T, Zhou J, Wang G, Li Y. Micro-RNA-338-3p Promotes the Development of Atherosclerosis by Targeting Desmin and Promoting Proliferation. Mol Biotechnol 2021; 63:840-848. [PMID: 34100182 PMCID: PMC8316222 DOI: 10.1007/s12033-021-00341-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/15/2021] [Indexed: 12/21/2022]
Abstract
Atherosclerosis (AS) is a dynamic and multi-stage process that involves various cells types, such as vascular smooth muscle cells (VSMCs) and molecules such as microRNAs. In this study, we investigated how miR-338-3p works in the process of AS. To determine how miR-338-3p was expressed in AS, an AS rat model was established and primary rat VSMCs were cultured. Real-time polymerase chain reaction was performed to detect miR-338-3p expression. Markers of different VSMC phenotypes were tested by Western blot. Immunofluorescent staining was employed to observe the morphologic changes of VSMCs transfected with miR-338-3p mimics. A dual luciferase reporter assay system was used to verify that desmin was a target of miR-338-3p. To further identify the role of miR-338-3p in the development of AS, VSMC proliferation and migration were evaluated by EdU incorporation assay, MTT assay, and wound healing assay. miR-338-3p expression was upregulated in the aortic tissues of an AS rat model and in primary rat VSMCs from a later passage. The transfection of miR-338-3p mimics in VSMCs promoted the synthetic cell phenotype. Bioinformatics analysis proposed desmin as a candidate target for miR-338-3p and the dual luciferase reporter assay confirmed in vivo that desmin was a direct target of miR-338-3p. The MTT and EdU incorporation assay revealed increased cell viability when miR-338-3p mimics were transfected. The increased expression of PCNA was a consistent observation, although a positive result was not obtained with respect to VSMC mobility. In AS, miR-338-3p expression was elevated. Elevated miR-338-3p inhibited the expression of desmin, thus promoting the contractile-to-synthetic VSMC phenotypic transition. In addition to morphologic changes, miR-338-3p enhanced the proliferative but not mobile ability of VSMCs. In summary, miR-338-3p promotes the development of AS.
Collapse
Affiliation(s)
- Shiran Yan
- Department of Cardiology, Heze Municipal Hospital, No. 2888, Caozhou West Road, Heze, 274000, China
| | - Jing Chen
- Department of Cardiology, Heze Municipal Hospital, No. 2888, Caozhou West Road, Heze, 274000, China
| | - Teng Zhang
- Department of Internal Medicine, Licun Township Health Center, Heze, 274038, China
| | - Jian Zhou
- Gaozhuang Town Central Health Center, Heze, 274000, China
| | - Ge Wang
- Department of Central Laboratory, Affiliated Beijing Chaoyang Hospital of Capital Medical University, Beijing, 100043, China
| | - Yanfen Li
- Department of Cardiology, Heze Municipal Hospital, No. 2888, Caozhou West Road, Heze, 274000, China.
| |
Collapse
|
14
|
Long noncoding RNA differentiation antagonizing nonprotein coding RNA promotes the proliferation, invasion and migration of neuroblastoma cells via targeting β-1, 4-galactosyltransferase III by sponging miR-338-3p. Neuroreport 2021; 32:965-974. [PMID: 34050113 DOI: 10.1097/wnr.0000000000001664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Neuroblastoma is a common malignant tumor in children, and patients often have a poor prognosis. Long noncoding RNAs (lncRNAs) are involved in the regulation of neuroblastoma progression. However, the regulatory effect of lncRNA differentiation antagonizing nonprotein coding RNA (DANCR) on neuroblastoma is still not clear. The expression levels of DANCR, miR-338-3p and β-1, 4-galactosyltransferase III (B4GALT3) were determined by quantitative real-time PCR. 3-(4, 5-dimethyl-2 thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide, flow cytometry and transwell assays were used to evaluate the proliferation, apoptosis, migration and invasion abilities of neuroblastoma cells. Moreover, western blot analysis was performed to assess the levels of B4GALT3 and the proliferation, apoptosis and migration-related proteins. Besides, a dual-luciferase reporter assay was used to verify the interactions among DANCR, miR-338-3p and B4GALT3. Mice xenograft models were used to ascertain the effect of DANCR on neuroblastoma tumor growth in vivo. Our results revealed that DANCR was highly expressed in neuroblastoma tissues and cells, and its silencing impeded the progression of neuroblastoma cells. DANCR could interact with miR-338-3p. Knockdown of miR-338-3p recovered the inhibitory effect of DANCR knockdown on neuroblastoma progression. B4GALT3 was a target of miR-338-3p. B4GALT3 overexpression reversed the suppression effect of DANCR silencing on neuroblastoma progression. In-vivo experiments further confirmed that DANCR silencing inhibited neuroblastoma tumor growth. Our results indicated that DANCR promoted B4GALT3 expression to increase the proliferation, migration and invasion of neuroblastoma cells via sponging miR-338-3p, which provided a theoretical basis for the targeted therapy of neuroblastoma.
Collapse
|
15
|
Bulfoni M, Pravisani R, Dalla E, Cesselli D, Hidaka M, Di Loreto C, Eguchi S, Baccarani U. miRNA expression profiles in liver grafts of HCV and HIV/HCV-infected recipients, 6 months after liver transplantation. J Med Virol 2021; 93:4992-5000. [PMID: 33818800 PMCID: PMC8360178 DOI: 10.1002/jmv.26999] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 03/03/2021] [Accepted: 03/23/2021] [Indexed: 12/14/2022]
Abstract
In hepatitis C virus (HCV)/human immunodeficiency virus (HIV) co‐infected patients, HIV enhances HCV replication and liver damage. Several microRNAs (miRNAs), active in pro‐fibrotic and inflammatory pathways, have been implicated in the pathogenesis of this phenomenon. However, these miRNAs have been tested only in explanted cirrhotic livers, when the liver damage has become chronic and irreversible. No data are available on the early phase of viral infection, such as early after liver transplantation (LT). In the present study, the expression of miR‐101, miR‐122, miR‐155, miR‐192, miR‐200c, miR‐338, and miR‐532 was determined by quantitative real‐time polymerase chain reaction in liver biopsies of HCV (n = 19) and HCV/HIV‐infected (n = 20) LT recipients, as well as in a control group (n = 18) of noninfected patients, transplanted for alcoholic cirrhosis. The timing of liver biopsy was 6 months post‐LT. None of the patients was treated with direct‐acting anti‐HCV drugs. All co‐infected recipients had suppressed HIV viral load. Grading and staging were assessed according to the Ishak Classification. HCV and HIV viral load were measured in the sera. miR‐101 (p = .03), miR‐122 (p = .012), and miR‐192 (p = .038) were significantly downregulated in HCV/HIV co‐infected and HCV mono‐infected recipients when compared with noninfected recipients, and such downregulation was more pronounced in co‐infected ones. Moreover, in co‐infected recipients but not in mono‐infected ones, miR‐101 inversely correlated with the peripheral HCV‐RNA levels (r = .41, p = .04) and miR‐122 inversely correlated with peripheral HCV‐RNA levels (r = .49, p = .03) and with the histological grading (r = .51, p = .02). In conclusion, as early as 6 months after LT, the presence of HIV‐HCV co‐infection enhanced a significant downregulation of certain miRNAs that showed a direct correlation with HCV viral load and liver inflammation.
Collapse
Affiliation(s)
| | - Riccardo Pravisani
- Department of Medicine, Liver-Kidney Transplant Unit, University of Udine, Udine, Italy
| | - Emiliano Dalla
- Department of Medicine, University of Udine, Udine, Italy
| | - Daniela Cesselli
- Department of Medicine, University of Udine, Udine, Italy.,Institute of Pathology, ASU FC, Udine, Italy
| | - Masaaki Hidaka
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Carla Di Loreto
- Department of Medicine, University of Udine, Udine, Italy.,Institute of Pathology, ASU FC, Udine, Italy
| | - Susumu Eguchi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Umberto Baccarani
- Department of Medicine, Liver-Kidney Transplant Unit, University of Udine, Udine, Italy
| |
Collapse
|
16
|
Moghbeli M. Molecular interactions of miR-338 during tumor progression and metastasis. Cell Mol Biol Lett 2021; 26:13. [PMID: 33827418 PMCID: PMC8028791 DOI: 10.1186/s11658-021-00257-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/25/2021] [Indexed: 02/08/2023] Open
Abstract
Background Cancer, as one of the main causes of human deaths, is currently a significant global health challenge. Since the majority of cancer-related deaths are associated with late diagnosis, it is necessary to develop minimally invasive early detection markers to manage and reduce mortality rates. MicroRNAs (miRNAs), as highly conserved non-coding RNAs, target the specific mRNAs which are involved in regulation of various fundamental cellular processes such as cell proliferation, death, and signaling pathways. MiRNAs can also be regulated by long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs). They are highly stable in body fluids and have tumor-specific expression profiles, which suggest their suitability as efficient non-invasive diagnostic and prognostic tumor markers. Aberrant expression of miR-338 has been widely reported in different cancers. It regulates cell proliferation, migration, angiogenesis, and apoptosis in tumor cells. Main body In the present review, we have summarized all miR-338 interactions with other non-coding RNAs (ncRNAs) and associated signaling pathways to clarify the role of miR-338 during tumor progression. Conclusions It was concluded that miR-338 mainly functions as a tumor suppressor in different cancers. There were also significant associations between miR-338 and other ncRNAs in tumor cells. Moreover, miR-338 has a pivotal role during tumor progression using the regulation of WNT, MAPK, and PI3K/AKT signaling pathways. This review highlights miR-338 as a pivotal ncRNA in biology of tumor cells.
Collapse
Affiliation(s)
- Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
17
|
Yuan H, Liu F, Ma T, Zeng Z, Zhang N. miR-338-3p inhibits cell growth, invasion, and EMT process in neuroblastoma through targeting MMP-2. Open Life Sci 2021; 16:198-209. [PMID: 33817311 PMCID: PMC7968531 DOI: 10.1515/biol-2021-0013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022] Open
Abstract
This study aimed to explore the regulatory mechanisms of miR-338-3p and matrix metalloproteinase-2 (MMP-2) in neuroblastoma. Putative target interaction regions of miR-338-3p on MMP-2 were predicted by miRcode and miRbase bioinformatics tools. Relative expression of miRNA-338-3p and MMP-2 in neuroblastoma tissues and GI-LI-N and SK-N-SH cells was determined by reverse transcription polymerase chain reaction experiment. Furthermore, the cell proliferation was determined by Cell Counting Kit-8 assay, the cell apoptosis rate was analyzed by flow cytometry assay, and the cell invasion was evaluated by transwell assay. miR-338-3p expression was downregulated, whereas MMP-2 expression was upregulated in metastasis tissue site compared to that in primary tissue site in total. Furthermore, miR-338-3p overexpression suppressed proliferation, invasion, and epithelial-mesenchymal transition (EMT) of neuroblastoma cells but promoted apoptosis, and the knockdown of MMP-2 triggered similar effects. Furthermore, MMP-2 was directly targeted by miR-338-3p, and overexpression of MMP-2 rescued the inhibitory effects of miR-338-3p on human neuroblastoma cell progression. Collectively, these data demonstrated that miR-338-3p could suppress cell growth, invasion, and EMT pathway and induce apoptosis in neuroblastoma cells by targeting MMP-2. MiR-338-3p sponged MMP-2 to regulate the PI3K/AKT pathway in human neuroblastoma cells.
Collapse
Affiliation(s)
- Haibin Yuan
- Department of Neonatal Surgery, Xuzhou Children's Hospital, No.18 Sudi North Road, Quanshan District, 221001, Xuzhou, China
| | - Fengli Liu
- Department of Neonatal Surgery, Xuzhou Children's Hospital, No.18 Sudi North Road, Quanshan District, 221001, Xuzhou, China
| | - Tongsheng Ma
- Department of Neonatal Surgery, Xuzhou Children's Hospital, No.18 Sudi North Road, Quanshan District, 221001, Xuzhou, China
| | - Zhandong Zeng
- Department of Neonatal Surgery, Xuzhou Children's Hospital, No.18 Sudi North Road, Quanshan District, 221001, Xuzhou, China
| | - Ning Zhang
- Department of Neonatal Surgery, Xuzhou Children's Hospital, No.18 Sudi North Road, Quanshan District, 221001, Xuzhou, China
| |
Collapse
|
18
|
Luo Q, Guo F, Fu Q, Sui G. hsa_circ_0001018 promotes papillary thyroid cancer by facilitating cell survival, invasion, G 1/S cell cycle progression, and repressing cell apoptosis via crosstalk with miR-338-3p and SOX4. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 24:591-609. [PMID: 33898108 PMCID: PMC8054110 DOI: 10.1016/j.omtn.2021.02.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 02/19/2021] [Indexed: 11/26/2022]
Abstract
We identified a novel interactome, circ_0001018/miR-338-3p/SOX4, in papillary thyroid cancer (PTC), and we intended to confirm the regulatory relationship between the three and to study the effects of the three in PTC. The bioinformatics method was used to screen out the circular RNA and mRNA of interest. A cellular fractionation assay and fluorescence in situ hybridization (FISH) assay were conducted to prove that circ_0001018 and CCT4 (the host gene of circ_0001018) mRNA primarily localized in the cytoplasm of PTC cell lines. By qRT-PCR analysis, the expression of circ_0001018 and SOX4 mRNA was found upregulated while the expression of miR-338-3p was found downregulated in PTC tissues and cells. circ_0001018 silence significantly inhibited the tumor growth in xenografted nude mice. A series of cytological experiments such as a Cell Counting Kit-8 (CCK-8) assay, a 5-ethynyl-2′-deoxyuridine (EdU) assay, cell cycle profiling, wound healing, a transwell assay, and cell apoptosis were conducted and showed that circ_0001018 and SOX4 promoted cell proliferation, migration, and invasion, inhibited cell apoptosis, and reduced the cell cycle arrest at the G1 phase in PTC cells. Compared with circ_0001018 and SOX4, miR-338-3p held the opposite function. The regulatory relationship between circ_0001018 and miR-338-3p, and between miR-338-3p and SOX4 mRNA, was validated using a luciferase reporter gene assay and/or RNA immunoprecipitation (RIP assay). Our findings showed that circ_0001018 acted as the tumor promoter via sponging miR-338-3p to elevate SOX4 expression level in PTC. Importantly, this novel circ_0001018/miR-338-3p/SOX4 axis has the potential to be considered as a therapy target for PTC.
Collapse
Affiliation(s)
- Qiang Luo
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun 130033, Jilin, China
| | - Feng Guo
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun 130033, Jilin, China
| | - Qingfeng Fu
- Jilin Provincial Key Laboratory of Surgical Translational Medicine, Division of Thyroid Surgery, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun 130033, Jilin, China
| | - Guoqing Sui
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun 130033, Jilin, China
| |
Collapse
|
19
|
Marengo B, Pulliero A, Corrias MV, Leardi R, Farinini E, Fronza G, Menichini P, Monti P, Monteleone L, Valenti GE, Speciale A, Perri P, Madia F, Izzotti A, Domenicotti C. Potential Role of miRNAs in the Acquisition of Chemoresistance in Neuroblastoma. J Pers Med 2021; 11:jpm11020107. [PMID: 33562297 PMCID: PMC7916079 DOI: 10.3390/jpm11020107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/20/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023] Open
Abstract
Neuroblastoma (NB) accounts for about 8–10% of pediatric cancers, and the main causes of death are the presence of metastases and the acquisition of chemoresistance. Metastatic NB is characterized by MYCN amplification that correlates with changes in the expression of miRNAs, which are small non-coding RNA sequences, playing a crucial role in NB development and chemoresistance. In the present study, miRNA expression was analyzed in two human MYCN-amplified NB cell lines, one sensitive (HTLA-230) and one resistant to Etoposide (ER-HTLA), by microarray and RT-qPCR techniques. These analyses showed that miRNA-15a, -16-1, -19b, -218, and -338 were down-regulated in ER-HTLA cells. In order to validate the presence of this down-regulation in vivo, the expression of these miRNAs was analyzed in primary tumors, metastases, and bone marrow of therapy responder and non-responder pediatric patients. Principal component analysis data showed that the expression of miRNA-19b, -218, and -338 influenced metastases, and that the expression levels of all miRNAs analyzed were higher in therapy responders in respect to non-responders. Collectively, these findings suggest that these miRNAs might be involved in the regulation of the drug response, and could be employed for therapeutic purposes.
Collapse
Affiliation(s)
- Barbara Marengo
- Department of Experimental Medicine, University of Genova, 16100 Genova, Italy; (L.M.); (G.E.V.); (A.I.); (C.D.)
- Correspondence: ; Tel.: +39-010-3538831
| | | | - Maria Valeria Corrias
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16100 Genova, Italy; (M.V.C.); (P.P.)
| | - Riccardo Leardi
- Department of Pharmacy, University of Genova, 16100 Genova, Italy; (R.L.); (E.F.)
| | - Emanuele Farinini
- Department of Pharmacy, University of Genova, 16100 Genova, Italy; (R.L.); (E.F.)
| | - Gilberto Fronza
- UOC Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, 16100 Genova, Italy; (G.F.); (P.M.); (P.M.); (A.S.)
| | - Paola Menichini
- UOC Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, 16100 Genova, Italy; (G.F.); (P.M.); (P.M.); (A.S.)
| | - Paola Monti
- UOC Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, 16100 Genova, Italy; (G.F.); (P.M.); (P.M.); (A.S.)
| | - Lorenzo Monteleone
- Department of Experimental Medicine, University of Genova, 16100 Genova, Italy; (L.M.); (G.E.V.); (A.I.); (C.D.)
| | - Giulia Elda Valenti
- Department of Experimental Medicine, University of Genova, 16100 Genova, Italy; (L.M.); (G.E.V.); (A.I.); (C.D.)
| | - Andrea Speciale
- UOC Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, 16100 Genova, Italy; (G.F.); (P.M.); (P.M.); (A.S.)
| | - Patrizia Perri
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16100 Genova, Italy; (M.V.C.); (P.P.)
| | - Francesca Madia
- Medical Genetics Unit, IRCCS Giannina Gaslini Institute, 16100 Genova, Italy;
| | - Alberto Izzotti
- Department of Experimental Medicine, University of Genova, 16100 Genova, Italy; (L.M.); (G.E.V.); (A.I.); (C.D.)
- UOC Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, 16100 Genova, Italy; (G.F.); (P.M.); (P.M.); (A.S.)
| | - Cinzia Domenicotti
- Department of Experimental Medicine, University of Genova, 16100 Genova, Italy; (L.M.); (G.E.V.); (A.I.); (C.D.)
| |
Collapse
|
20
|
The role of microRNA-338-3p in cancer: growth, invasion, chemoresistance, and mediators. Life Sci 2021; 268:119005. [PMID: 33421526 DOI: 10.1016/j.lfs.2020.119005] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/25/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022]
Abstract
Cancer still remains as one of the leading causes of death worldwide. Metastasis and proliferation are abnormally increased in cancer cells that subsequently, mediate resistance of cancer cells to different therapies such as radio-, chemo- and immune-therapy. MicroRNAs (miRNAs) are endogenous short non-coding RNAs that can regulate expression of target genes at post-transcriptional level and capable of interaction with mRNA-coding genes. Vital biological mechanisms including apoptosis, migration and differentiation are modulated by these small molecules. MiRNAs are key players in regulating cancer proliferation and metastasis as well as cancer therapy response. MiRNAs can function as both tumor-suppressing and tumor-promoting factors. In the present review, regulatory impact of miRNA-338-3p on cancer growth and migration is discussed. This new emerging miRNA can regulate response of cancer cells to chemotherapy and radiotherapy. It seems that miRNA-338-3p has dual role in cancer chemotherapy, acting as tumor-promoting or tumor-suppressor factor. Experiments reveal anti-tumor activity of miRNA-338-3p in cancer. Hence, increasing miRNA-338-3p expression is of importance in effective cancer therapy. Long non-coding RNAs, circular RNAs and hypoxia are potential upstream mediators of miRNA-338-3p in cancer. Anti-tumor agents including baicalin and arbutin can promote expression of miRNA-338-3p in suppressing cancer progression. These topics are discussed to shed some light on function of miRNA-338-3p in cancer cells.
Collapse
|
21
|
Yang L, Pei L, Yi J. LINC00839 Regulates Proliferation, Migration, Invasion, Apoptosis and Glycolysis in Neuroblastoma Cells Through miR-338-3p/GLUT1 Axis. Neuropsychiatr Dis Treat 2021; 17:2027-2040. [PMID: 34188473 PMCID: PMC8232867 DOI: 10.2147/ndt.s309467] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/27/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are related to the development and treatment of neuroblastoma. The lncRNA LINC00839 is dysregulated in neuroblastoma, while its function and mechanism in neuroblastoma development remain largely unclear. METHODS The tumor and adjacent noncancerous tissues were collected from 48 neuroblastoma patients. LINC00839, glucose transporter 1 (GLUT1) and microRNA-338-3p (miR-338-3p) abundances were examined via quantitative reverse transcription polymerase chain reaction or Western blot. Cell proliferation, apoptosis, migration, invasion and glycolysis were assessed via Cell Counting Kit-8, colony formation, flow cytometry, wound healing, transwell, glucose uptake and lactate production. The target relationship of miR-338-3p and LINC00839 or GLUT1 was tested via dual-luciferase reporter analysis and RNA immunoprecipitation. The function of LINC00839 on neuroblastoma cell growth in vivo was tested via a xenograft model. RESULTS LINC00839 and GLUT1 abundances were increased in neuroblastoma tissues and cell lines. The high expression of LINC00839 and GLUT1 indicated the lower overall survival. LINC00839 interference constrained neuroblastoma cell proliferation, migration, invasion and glycolysis, and facilitated apoptosis. GLUT1 overexpression or miR-338-3p knockdown could mitigate the influence of LINC00839 silence on neuroblastoma cell processes. LINC00839 could regulate GLUT1 expression via miR-338-3p. LINC00839 knockdown reduced neuroblastoma cell growth in xenograft model. CONCLUSION LINC00839 silence repressed neuroblastoma cell proliferation, migration, invasion and glycolysis and promoted apoptosis via regulating miR-338-3p/GLUT1 axis.
Collapse
Affiliation(s)
- Lixia Yang
- Department of Neurology, The First People's Hospital of Jingmen Affiliated to Hubei Minzu University, Jingmen, 434000, People's Republic of China
| | - Liangyan Pei
- Department of Neurology, The First People's Hospital of Jingmen Affiliated to Hubei Minzu University, Jingmen, 434000, People's Republic of China
| | - Jilong Yi
- Department of Neurology, The First People's Hospital of Jingmen Affiliated to Hubei Minzu University, Jingmen, 434000, People's Republic of China
| |
Collapse
|
22
|
Wang WY, Lu WC. Reduced Expression of hsa-miR-338-3p Contributes to the Development of Glioma Cells by Targeting Mitochondrial 3-Oxoacyl-ACP Synthase (OXSM) in Glioblastoma (GBM). Onco Targets Ther 2020; 13:9513-9523. [PMID: 33061435 PMCID: PMC7522303 DOI: 10.2147/ott.s262873] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/22/2020] [Indexed: 12/19/2022] Open
Abstract
Background MicroRNAs have been identified as major regulators and therapeutic targets of glioblastoma (GBM). It is thus meaningful to study the miRNAs differentially expressed (DE-miRNAs) in GBM. Materials and Methods We performed a meta-analysis of previously published microarray data using the R-based “metaMA” package to identify DE-miRNAs.The biological processes of the DE-miRNAs were then analyzed using FunRich. KEGG pathways of the DE-miRNAs gene targets were analyzed by mirPath V.3. Luciferase activity assay was performed to validate that OXSM is a direct target of hsa-miR338-3p. Flow cytometry was used to detect the effects of miR-338-3p on GBM cell proliferation, apoptosis and cell cycle. Results DE-miRNAs in blood and brain tissue from GBM were identified. “Type I interferon signaling pathway” and “VEGF and VEGFR signaling network” were the most significantly enriched biological processes shared by all GBM types. In KEGG pathway analysis, DE-miRNAs both in blood and tissue show altered fatty acid biosynthesis. Further validation shows hsa-miR-338-3p regulates fatty acid metabolism by directly targeting OXSM gene. In addition, our data revealed an accelerated cell cycle and an anti-apoptotic role for OXSM in glioma cells, which has not been reported. Finally, we confirmed that hsa-miR-338-3p inhibitor antagonized the effect of downregulation of OXSM on cell cycle and apoptosis of GBM cells. Conclusion We revealed that hsa-miR-338-3p, down-regulated in GBM, may affect the biogenesis and rapid proliferation of glioma cells by regulating the level of OXSM, providing new insights into understanding the pathogenesis of GBM and developing strategies to improve GBM prognosis.
Collapse
Affiliation(s)
- Wen-Yi Wang
- Department of Neurosurgery, Dafeng People's Hospital of Yancheng City, Yancheng City, Jiangsu Province, People's Republic of China
| | - Wei-Cheng Lu
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
23
|
Xiang D, Li Y, Lin Y. Circular RNA circCCDC66 Contributes to Malignant Phenotype of Osteosarcoma by Sponging miR-338-3p to Upregulate the Expression of PTP1B. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4637109. [PMID: 32851074 PMCID: PMC7439191 DOI: 10.1155/2020/4637109] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/21/2020] [Indexed: 12/31/2022]
Abstract
In recent years, the mechanism of cancer research has become hotspots of life science and medicine, especially due to the rapid development of molecular medicine and bioinformatics research. Similarly, the molecular mechanism also has received increasing attention in osteosarcoma (OS) research. Also, a considerable amount of research confirmed that circular RNAs (circRNAs) could regulate cancer cell growth and metastasis. This study aimed to explore the effect of a circRNA, circCCDC66, on OS and reveal its potential molecular mechanism. High circCCDC66 expression level was found in OS patient-derived tissue samples and OS cell lines by qRT-PCR. The abilities cell proliferation and metastatic of U2OS and SW1353 cells were then assessed by Cell Counting Kit-8 and transwell assay, respectively. The interaction between circCCDC66 and its target miRNAs were verified by the dual-luciferase reporter assay. Through functional experiments, we found that circCCDC66 knockdown promoted the inhibition of cell proliferation and metastatic of OS cell lines. From mechanistic perspective, circCCDC66 upregulated PTP1B by sponging miR-338-3p. Collectively, our findings demonstrated that circCCDC66 contributed to malignant behaviors of OS cells by miR-338-3p/PTP1B pathway, which suggested circCCDC66/miR-338-3p/PTP1B axis might be a potential therapeutic target.
Collapse
Affiliation(s)
- Deng Xiang
- Department of Orthopaedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Yugang Li
- Department of Orthopaedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Yanshui Lin
- Department of Orthopaedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, China
| |
Collapse
|
24
|
miR-142-3p Expression Is Predictive for Severe Traumatic Brain Injury (TBI) in Trauma Patients. Int J Mol Sci 2020; 21:ijms21155381. [PMID: 32751105 PMCID: PMC7432828 DOI: 10.3390/ijms21155381] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Predictive biomarkers in biofluids are the most commonly used diagnostic method, but established markers in trauma diagnostics lack accuracy. This study investigates promising microRNAs (miRNA) released from affected tissue after severe trauma that have predictive values for the effects of the injury. METHODS A retrospective analysis of prospectively collected data and blood samples of n = 33 trauma patients (ISS ≥ 16) is provided. Levels of miR-9-5p, -124-3p, -142-3p, -219a-5p, -338-3p and -423-3p in severely injured patients (PT) without traumatic brain injury (TBI) or with severe TBI (PT + TBI) and patients with isolated TBI (isTBI) were measured within 6 h after trauma. RESULTS The highest miR-423-3p expression was detected in patients with severe isTBI, followed by patients with PT + TBI, and lowest levels were found in PT patients without TBI (2-∆∆Ct, p = 0.009). A positive correlation between miR-423-3p level and increasing AIShead (p = 0.001) and risk of mortality (RISC II, p = 0.062) in trauma patients (n = 33) was found. ROC analysis of miR-423-3p levels revealed them as statistically significant to predict the severity of brain injury in trauma patients (p = 0.006). miR-124-3p was only found in patients with severe TBI, miR-338-3p was shown in all trauma groups. miR-9-5p, miR-142-3p and miR-219a-5p could not be detected in any of the four groups. CONCLUSION miR-423-3p expression is significantly elevated after isolated traumatic brain injury and predictable for severe TBI in the first hours after trauma. miR-423-3p could represent a promising new biomarker to identify severe isolated TBI.
Collapse
|
25
|
SNHG16 promotes tumorigenesis and cisplatin resistance by regulating miR-338-3p/PLK4 pathway in neuroblastoma cells. Cancer Cell Int 2020; 20:236. [PMID: 32536824 PMCID: PMC7291484 DOI: 10.1186/s12935-020-01291-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/23/2020] [Indexed: 02/07/2023] Open
Abstract
Background Long noncoding RNA small nucleolar RNA host gene 16 (lncRNA SNHG16) has been revealed to be involved in the tumorigenesis of neuroblastoma. However, the role of SNHG16 in regulating cisplatin sensitivity in neuroblastoma remains largely unknown. Methods The expression of SNHG16, microRNA (miR)-338-3p and polo-like kinase 4 (PLK4) mRNA was measured using quantitative real-time polymerase chain reaction. The protein levels of PLK4, multidrug resistance protein 1 (MRP1), multidrug-resistance gene 1-type p-glycoprotein (P-gp) and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway-related proteins were detected by Western blot. The half maximal inhibitory concentration (IC50) value, cell proliferation, migration and invasion were analyzed using Cell Counting Kit-8 assays or Transwell assay. Apoptotic cells were measured by Flow cytometry. The interaction between miR-338-3p and SNHG16 or PLK4 was confirmed by dual-luciferase reporter and RNA immunoprecipitation assay. In vivo experiments were conducted through the murine xenograft model. Results SNHG16 was up-regulated, while miR-338-3p was down-regulated in cisplatin-resistant neuroblastoma tissues and cells. SNHG16 silencing weakened cisplatin resistance, reflected by the reduction of IC50 value, down-regulation of MRP-1 and P-gp protein expression, suppression of proliferation, migration and invasion, as well as enhancement of apoptosis in SNHG16 deletion cisplatin-resistant neuroblastoma cells. Besides that, SNHG16 could regulate PLK4 expression by sponging miR-338-3p and SNHG16/miR-338-3p/PLK4 axis could affect the activation of PI3K/AKT pathway in cisplatin-resistant neuroblastoma cells. MiR-338-3p inhibition attenuated SNHG16 deletion-mediated impairment on cisplatin resistance and PLK4 overexpression reversed the decrease of cisplatin-resistance induced by miR-338-3p re-expression. Furthermore, SNHG16 knockdown contributed to the anti-tumor effect of cisplatin in neuroblastoma in vivo. Conclusion SNHG16 contributed to the tumorigenesis and cisplatin resistance in neuroblastoma possibly through miR-338-3p/PLK4 pathway, indicating a novel insight for overcoming chemoresistance in neuroblastoma patients.
Collapse
|
26
|
Humphries BA, Wang Z, Yang C. MicroRNA Regulation of the Small Rho GTPase Regulators-Complexities and Opportunities in Targeting Cancer Metastasis. Cancers (Basel) 2020; 12:E1092. [PMID: 32353968 PMCID: PMC7281527 DOI: 10.3390/cancers12051092] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/24/2020] [Accepted: 04/25/2020] [Indexed: 02/07/2023] Open
Abstract
The small Rho GTPases regulate important cellular processes that affect cancer metastasis, such as cell survival and proliferation, actin dynamics, adhesion, migration, invasion and transcriptional activation. The Rho GTPases function as molecular switches cycling between an active GTP-bound and inactive guanosine diphosphate (GDP)-bound conformation. It is known that Rho GTPase activities are mainly regulated by guanine nucleotide exchange factors (RhoGEFs), GTPase-activating proteins (RhoGAPs), GDP dissociation inhibitors (RhoGDIs) and guanine nucleotide exchange modifiers (GEMs). These Rho GTPase regulators are often dysregulated in cancer; however, the underlying mechanisms are not well understood. MicroRNAs (miRNAs), a large family of small non-coding RNAs that negatively regulate protein-coding gene expression, have been shown to play important roles in cancer metastasis. Recent studies showed that miRNAs are capable of directly targeting RhoGAPs, RhoGEFs, and RhoGDIs, and regulate the activities of Rho GTPases. This not only provides new evidence for the critical role of miRNA dysregulation in cancer metastasis, it also reveals novel mechanisms for Rho GTPase regulation. This review summarizes recent exciting findings showing that miRNAs play important roles in regulating Rho GTPase regulators (RhoGEFs, RhoGAPs, RhoGDIs), thus affecting Rho GTPase activities and cancer metastasis. The potential opportunities and challenges for targeting miRNAs and Rho GTPase regulators in treating cancer metastasis are also discussed. A comprehensive list of the currently validated miRNA-targeting of small Rho GTPase regulators is presented as a reference resource.
Collapse
Affiliation(s)
- Brock A. Humphries
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | - Zhishan Wang
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, 1095 V A Drive, Lexington, KY 40536, USA;
| | - Chengfeng Yang
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, 1095 V A Drive, Lexington, KY 40536, USA;
| |
Collapse
|
27
|
Abstract
Abdominal tumors (AT) in children account for approximately 17% of all pediatric solid tumor cases, and frequently exhibit embryonal histological features that differentiate them from adult cancers. Current molecular approaches have greatly improved the understanding of the distinctive pathology of each tumor type and enabled the characterization of novel tumor biomarkers. As seen in abdominal adult tumors, microRNAs (miRNAs) have been increasingly implicated in either the initiation or progression of childhood cancer. Moreover, besides predicting patient prognosis, they represent valuable diagnostic tools that may also assist the surveillance of tumor behavior and treatment response, as well as the identification of the primary metastatic sites. Thus, the present study was undertaken to compile up-to-date information regarding the role of dysregulated miRNAs in the most common histological variants of AT, including neuroblastoma, nephroblastoma, hepatoblastoma, hepatocarcinoma, and adrenal tumors. Additionally, the clinical implications of dysregulated miRNAs as potential diagnostic tools or indicators of prognosis were evaluated.
Collapse
|
28
|
Aravindan N, Subramanian K, Somasundaram DB, Herman TS, Aravindan S. MicroRNAs in neuroblastoma tumorigenesis, therapy resistance, and disease evolution. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:1086-1105. [PMID: 31867575 PMCID: PMC6924638 DOI: 10.20517/cdr.2019.68] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neuroblastoma (NB) deriving from neural crest cells is the most common extra-cranial solid cancer at infancy. NB originates within the peripheral sympathetic ganglia in adrenal medulla and along the midline of the body. Clinically, NB exhibits significant heterogeneity stretching from spontaneous regression to rapid progression to therapy resistance. MicroRNAs (miRNAs, miRs) are small (19-22 nt in length) non-coding RNAs that regulate human gene expression at the post-transcriptional level and are known to regulate cellular signaling, growth, differentiation, death, stemness, and maintenance. Consequently, the function of miRs in tumorigenesis, progression and resistance is of utmost importance for the understanding of dysfunctional cellular pathways that lead to disease evolution, therapy resistance, and poor clinical outcomes. Over the last two decades, much attention has been devoted to understanding the functional roles of miRs in NB biology. This review focuses on highlighting the important implications of miRs within the context of NB disease progression, particularly miRs’ influences on NB disease evolution and therapy resistance. In this review, we discuss the functions of both the “oncomiRs” and “tumor suppressor miRs” in NB progression/therapy resistance. These are the critical components to be considered during the development of novel miR-based therapeutic strategies to counter therapy resistance.
Collapse
Affiliation(s)
- Natarajan Aravindan
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Karthikeyan Subramanian
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Dinesh Babu Somasundaram
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Terence S Herman
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | |
Collapse
|
29
|
Zhang R, Shi H, Ren F, Feng W, Cao Y, Li G, Liu Z, Ji P, Zhang M. MicroRNA-338-3p suppresses ovarian cancer cells growth and metastasis: implication of Wnt/catenin beta and MEK/ERK signaling pathways. J Exp Clin Cancer Res 2019; 38:494. [PMID: 31842953 PMCID: PMC6916056 DOI: 10.1186/s13046-019-1494-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 11/27/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Downregulation of microRNA-338-3p (miR-338-3p) was detected in many malignant tumors, which indicated miR-338-3p might serve as a role of antioncogene in those cancers. The present study aimed to explore the roles of miR-338-3p in the growth and metastasis of ovarian cancer cells and elaborate the underlying possible molecular mechanism. METHODS Multiply biomedical databases query and KEGG pathway enrichment assay were used to infilter possible target genes and downstream pathways regulated by miR-338-3p. Overexpression miR-338-3p lentiviral vectors were transfected into ovarian cancer OVCAR-3 and OVCAR-8 cells, cell proliferation, migration and invasion were analyzed by MTT, colony formation, transwell, Matrigel assay and xenograft mouse model. One 3'-untranslated regions (UTRs) binding target gene of miR-338-3p, MACC1 (MET transcriptional regulator MACC1), and its regulated gene MET and downstream signaling pathway activities were examined by western blot. RESULTS Biomedical databases query indicated that miR-338-3p could target MACC1 gene and regulate Met, downstream Wnt/Catenin beta and MEK/ERK pathways. Rescue of miR-338-3p could inhibit the proliferation, migration and invasion of ovarian cancer cells, and suppress the growth and metastasis of xenograft tumor. Restoration of miR-338-3p could attenuate MACC1 and Met overexpression induced growth, epithelial to mesenchymal transition (EMT) and activities of Wnt/Catenin beta and MEK/ERK signaling in vitro and in vivo. CONCLUSIONS The present data indicated that restoration of miR-338-3p could suppress the growth and metastasis of ovarian cancer cells, which might due to the inhibition of proliferation and EMT induced by MACC1, Met and its downstream Wnt/Catenin beta and MEK/ERK signaling pathways.
Collapse
Affiliation(s)
- Ruitao Zhang
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, NO.1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450052 People’s Republic of China
| | - Huirong Shi
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, NO.1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450052 People’s Republic of China
| | - Fang Ren
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, NO.1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450052 People’s Republic of China
| | - Wei Feng
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, NO.1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450052 People’s Republic of China
| | - Yuan Cao
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, NO.1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450052 People’s Republic of China
| | - Gailing Li
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, NO.1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450052 People’s Republic of China
| | - Zheying Liu
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, NO.1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450052 People’s Republic of China
| | - Pengcheng Ji
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, NO.1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450052 People’s Republic of China
| | - Minghui Zhang
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, NO.1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450052 People’s Republic of China
| |
Collapse
|
30
|
Zhang J, Li WY, Yang Y, Yan LZ, Zhang SY, He J, Wang JX. LncRNA XIST facilitates cell growth, migration and invasion via modulating H3 histone methylation of DKK1 in neuroblastoma. Cell Cycle 2019; 18:1882-1892. [PMID: 31208278 PMCID: PMC6681787 DOI: 10.1080/15384101.2019.1632134] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) have been confirmed to be aberrantly expressed and involved in the progression of neuroblastoma. This study aimed to explore the expression profile of lncRNA X-inactive specific transcript (XIST) and its functional involvement in neuroblastoma. In this study, the relative level of XIST in neuroblastoma tissues and cell lines was detected by qPCR, and DKK1 protein expression was determined using western blot. The effect of XIST on cell growth, invasion and migration in vitro and in tumorigenesis of neuroblastoma was assessed. The level of H3K27me3 in DKK1 promoter was analyzed with ChIP-qPCR. Interaction between XIST and EZH2 was verified by RNA immunoprecipitation (RIP) and RNA pull-down assay. XIST was significantly upregulated in neuroblastoma tissues (n = 30) and cells lines, and it was statistically associated with the age and International Neuroblastoma Staging System (INSS) staging in neuroblastoma patients. Downregulation of XIST suppressed the growth, migration and invasion of neuroblastoma cells. EZH2 inhibited DKK1 expression through inducing H3 histone methylation in its promoter. XIST increased the level of H3K27me3 in DKK1 promoter via interacting with EZH2. Downregulation of XIST increased DKK1 expression to suppress neuroblastoma cell growth, invasion, and migration, which markedly restrained the tumor progression. In conclusion, XIST downregulated DKK1 by inducing H3 histone methylation via EZH2, thereby facilitating the growth, migration and invasion of neuroblastoma cells and retarding tumor progression.
Collapse
Affiliation(s)
- Jiao Zhang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- CONTACT Jiao Zhang
| | - Wen-Ya Li
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Yang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li-Zhao Yan
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Song-Yang Zhang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jia-Xiang Wang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
31
|
Zhang C, Li H, Wang J, Zhang J, Hou X. MicroRNA-338-3p suppresses cell proliferation, migration and invasion in human malignant melanoma by targeting MACC1. Exp Ther Med 2019; 18:997-1004. [PMID: 31316597 PMCID: PMC6601406 DOI: 10.3892/etm.2019.7644] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 11/07/2018] [Indexed: 12/27/2022] Open
Abstract
Malignant melanoma (MM) is the most aggressive form of skin cancer originating from melanocytes with increased proliferative and metastatic ability. Previous studies have revealed that microRNA-338-3p (miR-338-3p) functions as a tumor suppressor in several types of cancer, including cervical cancer, renal cell carcinoma and thyroid cancer. However, the function and mechanism underlying the action of miR-383-3p in MM remain unclear. In the study, aberrant downregulation of miR-338-3p was observed in 60 pairs of MM and adjacent non-tumor tissue using quantitative polymerase chain reaction assay. Decreased miR-383-3p expression was associated with advanced clinical stage (P<0.05) and lymph node metastasis (P<0.001). The overexpression of miR-338-3p in A375 and G361 cells suppressed cell proliferation and migration using MTT, colony formation, wound healing and transwell assays. Mechanistically, MACC1 was identified as a direct target for miR-338-3p using bioinformatics prediction and dual-luciferase assays. Furthermore, MACC1 expression was significantly increased and inversely correlated with the levels of miR-338-3p in MM tissues. More importantly, restoration of MACC1 resulted in reversed the inhibitory effects of miR-338-3p overexpression on MM cells by altering the expression levels of PCNA and epithelial-mesenchymal transition (EMT)-associated proteins. These results suggest that miR-338-3p functions as a novel tumor suppressor, at least in part, via targeting MACC1 and suggest that miR-338-3p may serve as a potential target for treatment of MM patients.
Collapse
Affiliation(s)
- Chunhua Zhang
- Department of Burn and Plastic Surgery, Cangzhou Center Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Hui Li
- Department of Burn and Plastic Surgery, Cangzhou Center Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Junling Wang
- Department of Burn and Plastic Surgery, Cangzhou Center Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Jibei Zhang
- Department of Burn and Plastic Surgery, Cangzhou Center Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Xiaoqian Hou
- Department of Burn and Plastic Surgery, Cangzhou Center Hospital, Cangzhou, Hebei 061001, P.R. China
| |
Collapse
|
32
|
Yuan XL, Wen FQ, Chen XW, Jiang XP, Liu SX. miR-373 promotes neuroblastoma cell proliferation, migration, and invasion by targeting SRCIN1. Onco Targets Ther 2019; 12:4927-4936. [PMID: 31417287 PMCID: PMC6593744 DOI: 10.2147/ott.s205582] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 05/17/2019] [Indexed: 12/17/2022] Open
Abstract
Introduction Previous studies have shown that miR-373 functions as either a tumor suppressor or an oncogene depending on which type of cancer it’s operating in. However, the functional role of miR-373 in neuroblastoma (NB) remains largely unclear. Methods Expression of miR-373 and SRC kinase signaling inhibitor 1 (SRCIN1) in 20 metastatic and 20 primary NB tissues was detected by quantitative real-time PCR (qRT-PCR) and Western blotting. MTT assay, flow cytometry analysis and transwell migration and invasion assays were performed to evaluate the influence of miR-373 inhibition on the growth, migration and invasion of NB cells, respectively. In vivo experiment was applied to determine the effect of miR-373 inhibition on tumor growth. Dual-luciferase reporter assay was used to confirm the interaction between miR-373 and SRCIN1. Results We observed a significant increase in the expression of miR-373 in metastatic NB samples compared with primary NB samples, and this was inversely correlated with SRCIN1 expression. Functional studies revealed that depletion of miR-373 inhibited in vitro NB cell growth, migration and invasion, and also suppressed tumor growth in an in vivo mouse model. Moreover, we identified that SRCIN1 was a direct and functional target gene of miR-373. Silencing of SRCIN1 partially rescued the antimiR-373-mediated inhibition of cell growth, migration and invasion. Conclusion The data from our study verified a potential oncogenic role of miR-373 in NB cells that occurs through direct targeting SRCIN1. The newly identified miR-373/SRCIN1 axis represents a new potential candidate for therapeutic intervention of malignant NB.
Collapse
Affiliation(s)
- Xiu-Li Yuan
- Department of Hematology/Oncology, Shenzhen Children's Hospital, Shenzhen 518036, People's Republic of China
| | - Fei-Qiu Wen
- Department of Hematology/Oncology, Shenzhen Children's Hospital, Shenzhen 518036, People's Republic of China
| | - Xiao-Wen Chen
- Department of Hematology/Oncology, Shenzhen Children's Hospital, Shenzhen 518036, People's Republic of China
| | - Xian-Ping Jiang
- Department of Hematology/Oncology, Shenzhen Children's Hospital, Shenzhen 518036, People's Republic of China
| | - Si-Xi Liu
- Department of Hematology/Oncology, Shenzhen Children's Hospital, Shenzhen 518036, People's Republic of China
| |
Collapse
|
33
|
Liu J, Cao L, Zhao N, Feng Y, Yu Z, Li Y, Teng C, Hu J, Li T. miR‑338‑3p inhibits A549 lung cancer cell proliferation and invasion by targeting AKT and β‑catenin signaling pathways. Mol Med Rep 2019; 20:33-40. [PMID: 31115502 PMCID: PMC6579971 DOI: 10.3892/mmr.2019.10215] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 01/17/2019] [Indexed: 12/21/2022] Open
Abstract
The aim of the present study was to explore the underlying mechanism of microRNA-338-3p (miR-338-3p) in lung cancer cell (A549) invasion and proliferation. A microarray assay showed that miR-338-3p upregulated 216 and downregulated 147 genes in A549 cells, and the differentially expressed genes were enriched for several signaling pathways, including AKT and β-catenin signaling pathways. Western blotting results showed that phosphorylated (p)-AKT at Ser473 and at Thr308 and p-β-catenin at Ser552 were downregulated. Inhibiting AKT signaling pathway decreased β-catenin signaling pathway. Overexpression of β-catenin promoted the invasion of A549 cells. In addition, miR-338-3p showed inhibitory effect on the primary tumor developed from KrasG12D mice. Together, the data of the present study support that miR-338-3p inhibits lung cancer cell invasion and proliferation by downregulating AKT/β-catenin signaling pathway. The present findings supported the potential use of miR-338-3p in the treatment of lung cancer.
Collapse
Affiliation(s)
- Jia Liu
- Department of Genetics, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Linggai Cao
- Department of Genetics, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Na Zhao
- Department of Genetics, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Yue Feng
- Department of Genetics, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Ze Yu
- Department of Genetics, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Yuhua Li
- Department of Genetics, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Chunbo Teng
- Department of Genetics, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Jifan Hu
- Department of Basic Science, Stanford University Medical School, Palo Alto Veterans Institute for Research, Palo Alto, CA 94305, USA
| | - Tao Li
- Department of Genetics, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| |
Collapse
|
34
|
Zhang R, Shi H, Ren F, Liu Z, Ji P, Zhang W, Wang W. Down-regulation of miR-338-3p and Up-regulation of MACC1 Indicated Poor Prognosis of Epithelial Ovarian Cancer Patients. J Cancer 2019; 10:1385-1392. [PMID: 31031848 PMCID: PMC6485222 DOI: 10.7150/jca.29502] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/16/2019] [Indexed: 12/12/2022] Open
Abstract
Objective To detect the expression of microRNA-338-3p (miR-338-3p) and MET transcriptional regulator MACC1 (MACC1) gene in different ovarian tissues, to analyze their relationships, their correlations to the clinicopathologic characteristics of epithelial ovarian cancer and their significant to the progression of ovarian cancer. Methods The expression of miR-338-3p and MACC1 gene in 20 specimens of normal ovarian tissues, 20 specimens of benign epithelial ovarian tumor and 65 specimens of epithelial ovarian cancer was detected by real-time PCR method. Their interrelationships and their correlations to the clinicopathologic characteristics of epithelial ovarian cancer were analyzed. Risk factors of recurrence and death were discussed by binary Logistic regression analysis. The relations between miR-338-3p and MACC1 expression and the survival of ovarian cancer were measured by Kaplan-Meier analysis. Results The expressions of miR-338-3p and MACC1 gene in epithelial ovarian cancer tissues were (0.331±0.038) and (0.774±0.025), significant differences were noted between epithelial ovarian cancer and normal ovarian tissues, benign ovarian tumors (F=77.916, P=1.205E-18; F=77.945, P=1.187E-18). In different ovarian tissues, miR-338-3p expression was negatively correlated to MACC1 expression (r = -0.968, P<0.0001). In epithelial ovarian cancer, lower expression of miR-338-3p and higher expression of MACC1 were associated with more advanced FIGO stage, higher histological grade and developed lymph node metastasis. Down-regulation of miR-338-3p was related with the recurrence (P=0. 005, OR=12.862, 95%CI: 2.120~78.026) and death (P=0. 007, OR=12.837, 95%CI: 2.205~81.389) of ovarian cancer patients, which was showed by binary Logistic regression analysis. Compared to other patients, the overall survival rate and progression free survival rate of patients with lower miR-338-3p and higher MACC1 expression were obviously poorer (χ2=16.955, P=7.219E-5; χ2=18.929, P=2.828E-5). Conclusions Down-regulation of miR-338-3p and up-regulation of MACC1 gene were closely related with the poor prognosis of epithelial ovarian cancer patients, which could served as bio-markers of the progression and recurrence of ovarian cancer.
Collapse
Affiliation(s)
- Ruitao Zhang
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China.,Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Huirong Shi
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Fang Ren
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Zheying Liu
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Pengcheng Ji
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Weiwei Zhang
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Wenwen Wang
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
35
|
Yang MH, Yen CH, Chen YF, Fang CC, Li CH, Lee KJ, Lin YH, Weng CH, Liu TT, Huang SF, Teh BT, Chen YMA. Somatic mutations of PREX2 gene in patients with hepatocellular carcinoma. Sci Rep 2019; 9:2552. [PMID: 30796242 PMCID: PMC6385191 DOI: 10.1038/s41598-018-36810-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 11/09/2018] [Indexed: 12/12/2022] Open
Abstract
Characterized with a high recurrence rate and low detection rate, prevention is the best approach to reduce mortality in hepatocellular carcinoma (HCC). The overexpression of Phosphatidylinositol-3,4,5-Trisphosphate Dependent Rac Exchange Factor 2 (PREX2) is observed in various tumors, including HCC; and the frequent PREX2 mutations in melanoma are associated with invasiveness. We sought to identify somatic mutations and the functional changes in mutational signatures of PREX2. Genomic DNA sequencing was performed in 68 HCC samples with three types of hepatitis viral infection status: HBs Ag-positive, anti-HCV Ab-positive, and negative for any hepatitis B or C markers. Stabilities and interactions of proteins as well as cell proliferation and migration were evaluated. Fourteen non-silent point mutations in PREX2 were detected, with 16 of 68 HCC patients harboring at least one non-silent mutation. All mutant forms of PREX2, except for K400f, had an extended half-life compared with wild-type PREX2. Moreover, only the half-life of S1113R was twice that of the wild-type. PREX2 mutant-S1113R also promoted migration and activated the AKT pathway as well as impaired HectH9-mediated ubiquitination. Our study identified a gain-of-function mutation of PREX2 – S1113R in HCC. Such mutation enhanced PREX2 protein stability, promoted cell proliferation, and was associated with aggressiveness of HCC.
Collapse
Affiliation(s)
- Ming-Hui Yang
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan.,Master Program in Clinical Pharmacogenomics and Pharmacoproteomics, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Chia-Hung Yen
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yen-Fu Chen
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Cheng-Chieh Fang
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chung-Hsien Li
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Kuo-Jui Lee
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yi-Hsiung Lin
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chien-Hui Weng
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Tze-Tze Liu
- VYM Genome Research Center, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Shiu-Feng Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Bin Tean Teh
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre, Singapore, 169610, Singapore
| | - Yi-Ming Arthur Chen
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung, 80708, Taiwan. .,Master Program in Clinical Pharmacogenomics and Pharmacoproteomics, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan.
| |
Collapse
|
36
|
Nazari B, Soleimanifar F, Kazemi M, Nazari B, Enderami SE, Ai A, Sadroddiny E, Ebrahimi-Barough S, Ai J. Derivation of preoligodendrocytes from human-induced pluripotent stem cells through overexpression of microRNA 338. J Cell Biochem 2018; 120:9700-9708. [PMID: 30582206 DOI: 10.1002/jcb.28248] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 10/24/2018] [Indexed: 01/30/2023]
Abstract
MicroRNAs (miRNAs) control gene expression at the posttranscriptional level and have a critical role in many biological processes such as oligodendrocyte differentiation. Recent studies have shown that microRNA 338 (miR-338) is overexpressed during the oligodendrocyte development process in the central nervous system; this finding indicates a potentially important role for miR-338 in oligodendrocyte development. To evaluate this assumption, we studied the effect of miR-338 overexpression on promoting the differentiation of oligodendrocyte progenitor cells (OPCs), derived from human-induced pluripotent stem cells (hiPSC), into preoligodendrocyte. hiPSCs were differentiated into OPCs after treating for 16 days with basic fibroblast growth factor (BFGF), epidermal growth factor (FGF), and platelet-derived growth factor (PDGF)-AA. Bipolar OPCs appeared and the expression of OPC-related markers, including Nestin, Olig2, Sox10, PDGFRα, and A2B5 was confirmed by real-time polymerase chain reaction (PCR) and immunofluorescence. Then, OPCs were transduced by miR-338 expressing lentivirus or were treated with triiodothyronine (T3) for 6 days. Data obtained from real-time PCR and immunofluorescence experiment indicated that preoligodendrocyte markers such as Sox10, O4, and MBP were expressed at higher levels in transduced cells with miR-338 in comparison with the T3 group. So, the overexpression of miR-338 in iPSC-derived OPCs can promote their differentiation into preoligodendrocyte which can be used in cell therapy of myelin-related diseases.
Collapse
Affiliation(s)
- Bahareh Nazari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Soleimanifar
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Mansure Kazemi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Banafsheh Nazari
- Section of Rheumatology, Boston University School of Medicine, Boston, Massachusetts
| | - Seyed Ehsan Enderami
- Department of Stem Cell Biology, Stem Cell Technology Research Center, Tehran, Iran
| | - Arman Ai
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Esmaeil Sadroddiny
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
MicroRNAs as Biomarkers in Amyotrophic Lateral Sclerosis. Cells 2018; 7:cells7110219. [PMID: 30463376 PMCID: PMC6262636 DOI: 10.3390/cells7110219] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/15/2018] [Accepted: 11/17/2018] [Indexed: 12/30/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable and fatal disorder characterized by the progressive loss of motor neurons in the cerebral cortex, brain stem, and spinal cord. Sporadic ALS form accounts for the majority of patients, but in 1–13.5% of cases the disease is inherited. The diagnosis of ALS is mainly based on clinical assessment and electrophysiological examinations with a history of symptom progression and is then made with a significant delay from symptom onset. Thus, the identification of biomarkers specific for ALS could be of a fundamental importance in the clinical practice. An ideal biomarker should display high specificity and sensitivity for discriminating ALS from control subjects and from ALS-mimics and other neurological diseases, and should then monitor disease progression within individual patients. microRNAs (miRNAs) are considered promising biomarkers for neurodegenerative diseases, since they are remarkably stable in human body fluids and can reflect physiological and pathological processes relevant for ALS. Here, we review the state of the art of miRNA biomarker identification for ALS in cerebrospinal fluid (CSF), blood and muscle tissue; we discuss advantages and disadvantages of different approaches, and underline the limits but also the great potential of this research for future practical applications.
Collapse
|
38
|
Luan X, Wang Y. LncRNA XLOC_006390 facilitates cervical cancer tumorigenesis and metastasis as a ceRNA against miR-331-3p and miR-338-3p. J Gynecol Oncol 2018; 29:e95. [PMID: 30207103 PMCID: PMC6189437 DOI: 10.3802/jgo.2018.29.e95] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/19/2018] [Accepted: 07/31/2018] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Cervical cancer is one of the most common malignant tumors. Our previous results showed that long non-coding RNA (lncRNA) XLOC_006390 plays an important role in cervical cancer. In this study, we have explored the mechanism of action of lncRNA XLOC_006390. METHODS LncRNA XLOC_006390 was proposed to exercise its function as a competing endogenous RNA (ceRNA), and its potential targeted miRNAs was predicted through the database LncBase Predicted v.2. Two miRNAs, miR-331-3p, and miR-338-3p, were chosen for the study. Expression of miRNAs and lncRNA in cervical cancer cells and tissues was detected by reverse transcription polymerase chain reaction. To determine the correlation, silencing of XLOC_006390, over-expression of miR-331-3p, and miR-338-3p was performed in SiHa and Caski cell lines, respectively. RESULTS Based on the interactive effect between miRNA and lncRNA, miR-331-3p and miR-338-3p were significantly downregulated in cervical cancer cells and tissues, and their expression levels were negatively related to that of lncRNA. Our results also showed that the expression of miR-331-3p target gene NRP2, miR-338-3p target genes PKM2, EYA2 was significantly downregulated when the XLOC_006390 was knocked down. Further, XLOC_006390 was found to facilitate cervical cancer tumorigenesis and metastasis by downregulating miR-331-3p and miR-338-3p expression. CONCLUSION Taken together, our study demonstrated that XLOC_006390 may serve as a ceRNA and reversely regulates the expression of miR-331-3p and miR-338-3p, thus facilitating cervical cancer tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Xiaotian Luan
- Department of Obstetrics and Gynecology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yankui Wang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
39
|
He JG, Xie QL, Li BB, Zhou L, Yan D. Exosomes Derived from IDO1-Overexpressing Rat Bone Marrow Mesenchymal Stem Cells Promote Immunotolerance of Cardiac Allografts. Cell Transplant 2018; 27:1657-1683. [PMID: 30311501 PMCID: PMC6299201 DOI: 10.1177/0963689718805375] [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] [Indexed: 12/11/2022] Open
Abstract
Background: The immunosuppressive activity of mesenchymal stem cells (MSCs) has been exploited to induce tolerance after organ transplantation. The indoleamine 2,3-dioxygenase (IDO) may have beneficial effects in the immunoregulatory properties of MSCs. It was recently revealed that exosomes derived from MSCs play important roles in mediating the biological functions of MSCs. This study aimed to explore the roles of exosomes derived from MSCs in the induction of immune tolerance. Methods: Dendritic cells (DCs) and T-cells were cultured with exosomes derived from rat bone marrow MSCs (BMSCs) overexpressing IDO1 or controls. For the in-vivo study, rats received heart transplants and were treated with exosomes from IDO-BMSCs and heart function was evaluated. Flow cytometry was used to detect expression of cell surface markers. Cytokine levels were detected in culture supernatants or serum samples. Protein and microRNA expressions in exosomes were investigated by chips. Results: Exosomes from IDO-BMSCs cultured with DCs and T-cells (1) downregulated CD40, CD86, CD80, MHC-II, CD45RA, CD45RA+CD45RB, OX62, and upregulated CD274 expression, (2) increased the number of regulatory T-cells (Tregs) and decreased the number of CD8+ T-cells, and (3) decreased the levels of pro-inflammatory cytokines, but increased the levels of anti-inflammatory cytokines compared with the other groups. Transplanted rats, which were injected with exosomes from IDO-BMSCs, had reduced allograft-targeting immune responses and improved cardiac allograft function. Exosomes secreted by IDO-BMSCs exhibited significant upregulations of the immunoregulatory protein FHL-1, miR-540-3p, and a downregulation of miR-338-5p. Conclusion: Exosomes derived from IDO-BMSCs can be used to promote immunotolerance and prolong the survival of cardiac allografts.
Collapse
Affiliation(s)
- Ji-Gang He
- Department of Cardiovascular Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Qiao-Li Xie
- Department of Cardiovascular Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Bei-Bei Li
- Department of Cardiovascular Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Liang Zhou
- Department of Cardiology, First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Dan Yan
- Department of Intensive Care Unit, First People's Hospital of Yunnan Province, Yunnan Province, China
| |
Collapse
|
40
|
Association of plasma microRNA expression with age, genetic background and functional traits in dairy cattle. Sci Rep 2018; 8:12955. [PMID: 30154501 DOI: 10.1038/s41598-018-31099-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/13/2018] [Indexed: 02/07/2023] Open
Abstract
A number of blood circulating microRNAs (miRNAs) are proven disease biomarkers and have been associated with ageing and longevity in multiple species. However, the role of circulating miRNAs in livestock species has not been fully studied. We hypothesise that plasma miRNA expression profiles are affected by age and genetic background, and associated with health and production traits in dairy cattle. Using PCR arrays, we assessed 306 plasma miRNAs for effects of age (calves vs mature cows) and genetic background (control vs select lines) in 18 animals. We identified miRNAs which were significantly affected by age (26 miRNAs) and genetic line (5 miRNAs). Using RT-qPCR in a larger cow population (n = 73) we successfully validated array data for 12 age-related miRNAs, one genetic line-related miRNA, and utilised expression data to associate their levels in circulation with functional traits in these animals. Plasma miRNA levels were associated with telomere length (ageing/longevity indicator), milk production and composition, milk somatic cell count (mastitis indicator), fertility, lameness, and blood metabolites linked with body energy balance and metabolic stress. In conclusion, circulating miRNAs could provide useful selection markers for dairy cows to help improve health, welfare and production performance.
Collapse
|
41
|
Lu R, Yang Z, Xu G, Yu S. miR-338 modulates proliferation and autophagy by PI3K/AKT/mTOR signaling pathway in cervical cancer. Biomed Pharmacother 2018; 105:633-644. [PMID: 29898430 DOI: 10.1016/j.biopha.2018.06.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/31/2018] [Accepted: 06/05/2018] [Indexed: 12/11/2022] Open
Abstract
Cervical cancer (CC) is a malignant solid tumor, which is one of the main causes of morbidity and mortality in women. Given that autophagy is an important factor promoting tumor progression, we aim to investigate the functional role of miR-338 in autophagy and proliferation of cervical cancer. In our study, expression of miR-338 was validated by quantitative RT-PCR in 30 paired cervical cancer tissues and normal tissues. We performed MTT, colony formation and cell cycle assay to explore the effect of miR-338 on cell proliferation. The level of autophagy was evaluated by observing the expression of LC3 formation under fluorescence microscope and detected the LC3 expression by western blot. We used luciferase reporter assays to identify the target gene about miR-338. We not only found that the level of miR-338 is decreased in cervical cancer tissues and cells, but also negatively correlated with the protein level of ATF2. In turn, restoring the expression of miR-338 inhibited proliferation in Hela and SiHa cells. Further mechanistic study identified that ATF2 as a direct target of miR-338. Forced lowexpression of miR-338 directly led to increased the level of autophagy in cervical cancer cells, which was similar to the mTOR signaling inhibitor rapamycin. The western blot analysis show that inhibited miR-338 expression could decrease the p-mTOR and p-p70S6 expression. Thus, we infer that miR-338 decreases autophagy level in cervical cancer cells by activating mTOR signaling pathway. In summary, our study demonstrate that miR-338 could inhibites proliferation and autophagy by targeting ATF2 via mTOR signaling pathway on cervical cancer cells. These results suggest a potential application of miR-338 in cervical cancer as a novel mechanism of tumor therapeutic.
Collapse
Affiliation(s)
- Rong Lu
- Department of Gynecology, Huai'an Second People's Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huai'an, 223002, China
| | - Zhanhua Yang
- Department of Nursing, Huaian Maternity and Child Healthcare Hospital Affiliated to Yangzhou University Medical College, Huai'an, 223002, China
| | - Guoying Xu
- Department of gynecology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University. Huai'an, 223300, China.
| | - Shengsheng Yu
- Department of laboratory medicine, Jiangsu college of nursing. Huai'an, 223005, China.
| |
Collapse
|
42
|
Li SH, Li JP, Chen L, Liu JL. miR-146a induces apoptosis in neuroblastoma cells by targeting BCL11A. Med Hypotheses 2018; 117:21-27. [PMID: 30077189 DOI: 10.1016/j.mehy.2018.05.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/13/2018] [Accepted: 05/25/2018] [Indexed: 12/27/2022]
Abstract
Aberrant expression of miR-146a has been reported to be involved in the progression and metastasis of various types of human cancers; however, its potential role in human neuroblastoma is still poorly understood. The purpose of our study was to investigate the molecular mechanism and possible role of miR-146a in human neuroblastoma. In this study, targeted genes were predicted by bioinformatic analysis and confirmed by dual-Luciferase reporter assay. The expression level of miR-146a in the human neuroblastoma SK-N-SH cell line was detected by quantitative RT-PCR. We used flow cytometric analysis to determine apoptosis and necrosis of SK-N-SH cells after transfection with miR-146a inhibitor, miR-146a mimic, and negative controls. The expression level of target genes was detected by RT-PCR and Western blotting. We identified BCL11A as a target of miR-146a. Thus, miR-146a targets the 3'UTR of BCL11A and inhibits its mRNA and protein expression. Overexpression of miR-146a can inhibit the growth and promote the apoptosis of human neuroblastoma SK-N-SH cells through inhibiting the expression of BCL11A. Furthermore, we found that upregulation of BCL11A by miR-146a inhibitor can promote SK-N-SH cells growth and protect SK-N-SH cells against apoptosis. Our results showed that miR-146a is a potential tumor suppressor gene in human neuroblastoma via directly targeting BCL11A. These findings suggest that miR-146a might be a new candidate target for treatment of human neuroblastoma.
Collapse
Affiliation(s)
- Sheng-Hua Li
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University in Nanning, China
| | - Jin-Pin Li
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University in Nanning, China
| | - Lan Chen
- Department of Internal Medicine, The Second Affiliated Hospital of Guangxi Medical University in Nanning, China
| | - Jing-Li Liu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University in Nanning, China.
| |
Collapse
|
43
|
Cao R, Shao J, Hu Y, Wang L, Li Z, Sun G, Gao X. microRNA-338-3p inhibits proliferation, migration, invasion, and EMT in osteosarcoma cells by targeting activator of 90 kDa heat shock protein ATPase homolog 1. Cancer Cell Int 2018; 18:49. [PMID: 29618948 PMCID: PMC5879792 DOI: 10.1186/s12935-018-0551-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 03/26/2018] [Indexed: 01/29/2023] Open
Abstract
Background Osteosarcoma (OS) is a rare, malignant bone tumor that primarily affects adolescents and has a high degree of malignancy and high incidence of recurrence and metastasis. Our study aimed to explore the role of miR-338-3p in OS cells. Methods qRT-qPCR was performed to quantify miR-338-3p expression levels in OS tissue samples and in three common OS cell lines. MG-63 and Saos2 cells were separately transfected with miR-338-3p or NC mimics and miR-338-3p expression levels was determined by qRT-PCR. Cell proliferation was monitored using the Cell Counting Kit-8. Flow cytometer analysis was carried out to determine the distribution of cell cycle stages and apoptosis. Transwell assay was performed to measure the migratory and invasive capacities of MG-63 and Saos2 cells. The expression of Vimentin and E-cadherin was detected by western blot. Luciferase reporter assay, qRT-PCR and western blotting were performed to confirm the target of miR-338-3p. Results Analysis by qRT-PCR revealed that miR-338-3p was downregulated in the tissue samples of 20 OS patients when compared with that in their matched adjacent non-tumor tissues. Furthermore, miR-338-3p was significantly downregulated in three common OS cell lines, namely, MG-63, Saos2, and HOS, when compared with that in the human osteoblast cell line hFOB1.19. Analysis by luciferase reporter assay, qRT-PCR, and western blotting revealed that activator of 90 kDa heat shock protein ATPase homolog 1 (AHSA1) is a direct target of miR-338-3p. miR-338-3p overexpression led to significant reduction in AHSA1 protein levels in MG63 and Saos2 cells. miR-338-3p overexpression reduced cell viability and migration and invasion behavior of MG63 and Saos2 cells. In addition, miR-338-3p overexpression suppressed epithelial–mesenchymal transition (EMT), induced a significant G1-phase arrest and did not affect the apoptosis in both MG-63 and Saos2 cells. Moreover, overexpression of AHSA1 reversed the inhibitory effect of miR-338-3p overexpression on proliferation, cell cycle, apoptosis, EMT, migration, and invasion of MG63 and Saos2 cells, thereby suggesting that miR-338-3p acts as a tumor suppressor in OS cells by targeting AHSA1. Conclusions miR-338-3p/AHSA1 can serve as a potential therapeutic target for OS therapy.
Collapse
Affiliation(s)
- Riliang Cao
- 1Department of Pediatric Surgery, Guangdong Women and Children Hospital, Guangzhou, 511400 China
| | - Jianli Shao
- 2Department of Orthopedic and Traumatology, First Affiliated Hospital, Jinan University, Guangzhou, 510632 China
| | - Yabin Hu
- 3Department of Spinal Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830002 Xinjiang China
| | - Liang Wang
- 4Department of Oncology, First Affiliated Hospital, Jinan University, Guangzhou, 510632 China
| | - Zhizhong Li
- 2Department of Orthopedic and Traumatology, First Affiliated Hospital, Jinan University, Guangzhou, 510632 China
| | - Guodong Sun
- 2Department of Orthopedic and Traumatology, First Affiliated Hospital, Jinan University, Guangzhou, 510632 China
| | - Xiaoliang Gao
- 3Department of Spinal Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830002 Xinjiang China
| |
Collapse
|
44
|
Montero JC, Seoane S, García-Alonso S, Pandiella A. Multisite phosphorylation of P-Rex1 by protein kinase C. Oncotarget 2018; 7:77937-77949. [PMID: 27788493 PMCID: PMC5363633 DOI: 10.18632/oncotarget.12846] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/12/2016] [Indexed: 02/07/2023] Open
Abstract
P-Rex proteins are guanine nucleotide exchange factors (GEFs) that act on the Rho/Rac family of GTP binding proteins. The activity of P-Rex proteins is regulated by several extracellular stimuli. In fact, activation of growth factor receptors has been reported to activate a phosphorylation/dephosphorylation cycle of P-Rex1. Such cycle includes dephosphorylation of serines 313 and 319 which negatively regulate the GEF activity of P-Rex1, together with phosphorylation of serines 605 and 1169 which favour P-Rex1 GEF activity. However, the kinases that regulate phosphorylation at these different regulatory sites are largely unknown. Here we have investigated the potential regulatory action of several kinases on the phosphorylation of P-Rex1 at S313, S319, S605 and S1169. We show that activation of protein kinase C (PKC) caused phosphorylation of S313, S319 and S1169. Activation of growth factor receptors induced phosphorylation of S1169 through a mechanism that was independent of PKC, indicating that distinct kinases and mechanisms control the phosphorylation of P-Rex1 at different regulatory serines. Genetic and biochemical studies confirmed that the PKC isoform PKCδ was able to directly phosphorylate P-Rex1 at S313. Functional studies using cells with very low endogenous P-Rex1 expression, transfected with wild type P-Rex1 or a mutant form in which S313 was substituted by alanine, indicated that phosphorylation at that residue negatively regulated P-Rex1 exchange activity. We suggest that control of P-Rex1 activity depends on a highly dynamic interplay among distinct signalling routes and its multisite phosphorylation is controlled by the action of different kinases.
Collapse
Affiliation(s)
- Juan Carlos Montero
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Spain
| | - Samuel Seoane
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Spain
| | - Sara García-Alonso
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Spain
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Spain
| |
Collapse
|
45
|
Yin G, Yang X, Li Q, Guo Z. GATA1 activated lncRNA (Galont) promotes anoxia/reoxygenation-induced autophagy and cell death in cardiomyocytes by sponging miR-338. J Cell Biochem 2018; 119:4161-4169. [PMID: 29247537 DOI: 10.1002/jcb.26623] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/12/2017] [Indexed: 12/19/2022]
Abstract
The hypernomic autophagy is associated with various cardiovascular diseases. Long noncoding RNAs (lncRNAs) are emerging as important regulators in gene expression, which have been involved in multiple physiological and pathological processes. However, the function of lncRNAs and how they functioned in the autophagy in cardiomyocytes were rarely reported. In this study, we report that a lncRNA, named GATA1 activated lncRNA (Galont), can directly interact with miR-338 and promote ATG5-mediated autophagic cell death in murine cardiomyocytes. First, we found that Galont was upregulated by anoxia/reoxygenation (A/R) stimulus, and it was able to promote autophagy and cell death in cardiomyocytes exposure to A/R. Then, miR-338 was identified as a novel suppressor in autophagy and autophagic cell death. Our results from bioinformatic analysis and luciferase reporter gene assay indicated that ATG5 is a target gene of miR-338. Furthermore, RNA pull-down assays demonstrated that Galont directly interacted with miR-338, and thus promoted ATG5 expression and autophagic cell death. Our findings reveal a novel regulatory circuit in the autophagy in cardiomyocytes, which consists of Galont, miR-338 and ATG5.
Collapse
Affiliation(s)
- Guotian Yin
- Department of Cardiology, Third Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Xiuli Yang
- Department of Cardiology, Third Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Qiong Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Zhikun Guo
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
46
|
Liu S, Suo J, Wang C, Sun X, Wang D, He L, Zhang Y, Li W. Downregulation of tissue miR-338-3p predicts unfavorable prognosis of gastric cancer. Cancer Biomark 2017; 21:117-122. [PMID: 29060930 DOI: 10.3233/cbm-170339] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Suoning Liu
- Department of the Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Jian Suo
- Department of the Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Chunxi Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xuan Sun
- Department of the Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Daguang Wang
- Department of the Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Liang He
- Department of the Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yang Zhang
- Department of the Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Wei Li
- Department of the Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
47
|
Liu DZ, Zhao H, Zou QG, Ma QJ. MiR-338 suppresses cell proliferation and invasion by targeting CTBP2 in glioma. Cancer Biomark 2017; 20:289-297. [PMID: 28826173 DOI: 10.3233/cbm-170128] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- De-Zhi Liu
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, Jilin, China
| | - Hui Zhao
- Department of Thoracic Oncology, Jilin Cancer Hospital, Jilin, China
| | - Qin-Guang Zou
- Department of Thoracic Oncology, Jilin Cancer Hospital, Jilin, China
| | - Qing-Jie Ma
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, Jilin, China
| |
Collapse
|
48
|
Mohamed AA, Ali-Eldin ZA, Elbedewy TA, El-Serafy M, Ali-Eldin FA, AbdelAziz H. MicroRNAs and clinical implications in hepatocellular carcinoma. World J Hepatol 2017; 9:1001-1007. [PMID: 28878865 PMCID: PMC5569275 DOI: 10.4254/wjh.v9.i23.1001] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/13/2017] [Accepted: 06/20/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To assess the role of some circulating miRNAs (miR-23a, miR-203, miR338, miR-34, and miR-16) as tumor markers for diagnosis of hepatocellular carcinoma (HCC).
METHODS One hundred and seventy-one subjects were enrolled, 57 patients with HCC, 57 patients with liver cirrhosis (LC) and 57 healthy subjects as control group. Severity of liver disease was assessed by Child Pugh score. Tumor staging was done using Okuda staging system. Quantification of Micro RNA (miR-23a, miR-203, miR338, miR-34, and miR-16) was performed.
RESULTS All studied miRNA showed significant difference between HCC and cirrhotic patients in comparison to healthy control. miR-23a showed statistically significant difference between HCC and cirrhotic patients being higher in HCC group than cirrhotic. miR-23a is significantly higher in HCC patients with focal lesion size equal or more than 5 cm, patients with multiple focal lesions and Okuda stage III. At cutoff value ≥ 210, miR-23a showed accuracy 79.3% to diagnose HCC patients with sensitivity 89.47% and specificity about 64.91%. At cut off level ≥ 200 ng/mL, serum alpha fetoprotein had 73.68% sensitivity, 52.63% specificity, 43.75% PPV, 80% NPV for diagnosis of HCC.
CONCLUSION MicroRNA 23a can be used as a screening test for early detection of HCC. Also, it is related to larger size of tumour, late Okuda staging and multiple hepatic focal lesions, so it might be a prognostic biomarker.
Collapse
|
49
|
P-Rex1 and P-Rex2 RacGEFs and cancer. Biochem Soc Trans 2017; 45:963-77. [PMID: 28710285 DOI: 10.1042/bst20160269] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 06/01/2017] [Accepted: 06/05/2017] [Indexed: 12/15/2022]
Abstract
Phosphatidylinositol 3,4,5-trisphosphate-dependent Rac exchanger (P-Rex) proteins are RacGEFs that are synergistically activated by phosphatidylinositol 3,4,5-trisphosphate and Gβγ subunits of G-protein-coupled receptors. P-Rex1 and P-Rex2 share similar amino acid sequence homology, domain structure, and catalytic function. Recent evidence suggests that both P-Rex proteins may play oncogenic roles in human cancers. P-Rex1 and P-Rex2 are altered predominantly via overexpression and mutation, respectively, in various cancer types, including breast cancer, prostate cancer, and melanoma. This review compares the similarities and differences between P-Rex1 and P-Rex2 functions in human cancers in terms of cellular effects and signalling mechanisms. Emerging clinical data predict that changes in expression or mutation of P-Rex1 and P-Rex2 may lead to changes in tumour outcome, particularly in breast cancer and melanoma.
Collapse
|
50
|
MiR-338-3p inhibits TNF-α-induced lipogenesis in human sebocytes. Biotechnol Lett 2017; 39:1343-1349. [PMID: 28597147 DOI: 10.1007/s10529-017-2369-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/25/2017] [Indexed: 10/19/2022]
Abstract
OBJECTIVE To suppress TNF-α-induced lipogenesis in sebocytes (associated with acne development) with microRNA-338-3p (miR-338-3p) and to explore the underlying mechanisms. RESULTS TNF-α increased lipid droplet formation in sebocytes which were used as in vitro model of inflammation-induced acne. Flow cytometry and TLC assays validated that miR-338-3p could suppress TNF-α-induced lipid droplet formation, down-regulate the expression of PREX2a, and inactivate AKT signaling in sebocytes. In addition, suppression of AKT activity by the PI3 K and AKT inhibitors diminished TNF-α-induced lipogenesis. PREX2a siRNA mimics the effects of miR-338-3p on AKT phosphorylation and lipogenesis. PREX2a overexpression consistently restored lipogenesis and AKT phosphorylation attenuated by miR-338-3p. CONCLUSIONS MiR-338-3p suppresses the TNF-α-induced lipogenesis in sebocytes by targeting PREX2a and down-regulating PI3K/AKT signaling.
Collapse
|