1
|
William JNG, Dhar R, Gundamaraju R, Sahoo OS, Pethusamy K, Raj AFPAM, Ramasamy S, Alqahtani MS, Abbas M, Karmakar S. SKping cell cycle regulation: role of ubiquitin ligase SKP2 in hematological malignancies. Front Oncol 2024; 14:1288501. [PMID: 38559562 PMCID: PMC10978726 DOI: 10.3389/fonc.2024.1288501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 02/15/2024] [Indexed: 04/04/2024] Open
Abstract
SKP2 (S-phase kinase-associated protein 2) is a member of the F-box family of substrate-recognition subunits in the SCF ubiquitin-protein ligase complexes. It is associated with ubiquitin-mediated degradation in the mammalian cell cycle components and other target proteins involved in cell cycle progression, signal transduction, and transcription. Being an oncogene in solid tumors and hematological malignancies, it is frequently associated with drug resistance and poor disease outcomes. In the current review, we discussed the novel role of SKP2 in different hematological malignancies. Further, we performed a limited in-silico analysis to establish the involvement of SKP2 in a few publicly available cancer datasets. Interestingly, our study identified Skp2 expression to be altered in a cancer-specific manner. While it was found to be overexpressed in several cancer types, few cancer showed a down-regulation in SKP2. Our review provides evidence for developing novel SKP2 inhibitors in hematological malignancies. We also investigated the effect of SKP2 status on survival and disease progression. In addition, the role of miRNA and its associated families in regulating Skp2 expression was explored. Subsequently, we predicted common miRNAs against Skp2 genes by using miRNA-predication tools. Finally, we discussed current approaches and future prospective approaches to target the Skp2 gene by using different drugs and miRNA-based therapeutics applications in translational research.
Collapse
Affiliation(s)
- Jonahunnatha Nesson George William
- Department of Medical, Oral and Biotechnological Sciences (DSMOB), Ageing Research Center and Translational Medicine-CeSI-MeT, “G. d’Annunzio” University Chieti-Pescara, Chieti, Italy
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Rohit Gundamaraju
- ER Stress and Intestinal Mucosal Biology Lab, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Om Saswat Sahoo
- Department of Biotechnology, National Institute of Technology, Durgapur, India
| | - Karthikeyan Pethusamy
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | | | - Subbiah Ramasamy
- Cardiac Metabolic Disease Laboratory, Department Of Biochemistry, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Mohammed S. Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
- BioImaging Unit, Space Research Centre, University of Leicester, Leicester, United Kingdom
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, Saudi Arabia
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
2
|
Zhang C, Pan G, Qin JJ. Role of F-box proteins in human upper gastrointestinal tumors. Biochim Biophys Acta Rev Cancer 2024; 1879:189035. [PMID: 38049014 DOI: 10.1016/j.bbcan.2023.189035] [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: 08/23/2023] [Revised: 11/22/2023] [Accepted: 11/25/2023] [Indexed: 12/06/2023]
Abstract
Protein ubiquitination and degradation is an essential physiological process in almost all organisms. As the key participants in this process, the E3 ubiquitin ligases have been widely studied and recognized. F-box proteins, a crucial component of E3 ubiquitin ligases that regulates diverse biological functions, including cell differentiation, proliferation, migration, and apoptosis by facilitating the degradation of substrate proteins. Currently, there is an increasing focus on studying the role of F-box proteins in cancer. In this review, we present a comprehensive overview of the significant contributions of F-box proteins to the development of upper gastrointestinal tumors, highlighting their dual roles as both carcinogens and tumor suppressors. We delve into the molecular mechanisms underlying the involvement of F-box proteins in upper gastrointestinal tumors, exploring their interactions with specific substrates and their cross-talks with other key signaling pathways. Furthermore, we discuss the implications of F-box proteins in radiotherapy resistance in the upper gastrointestinal tract, emphasizing their potential as clinical therapeutic and prognostic targets. Overall, this review provides an up-to-date understanding of the intricate involvement of F-box proteins in human upper gastrointestinal tumors, offering valuable insights for the identification of prognostic markers and the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Che Zhang
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Guangzhao Pan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Jiang-Jiang Qin
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| |
Collapse
|
3
|
Ryu Y, Hwang JS, Bo Noh K, Park SH, Seo JH, Shin YJ. Adipose Mesenchymal Stem Cell-Derived Exosomes Promote the Regeneration of Corneal Endothelium Through Ameliorating Senescence. Invest Ophthalmol Vis Sci 2023; 64:29. [PMID: 37850944 PMCID: PMC10593138 DOI: 10.1167/iovs.64.13.29] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 09/14/2023] [Indexed: 10/19/2023] Open
Abstract
Purpose Human corneal endothelial cells (hCECs) have been considered unable to regenerate in vivo, resulting in corneal decompensation after significant loss of hCECs. adipose-derived mesenchymal stem cell (ASC)-derived exosomes can regenerate tissues and organs. In this study, we investigated whether ASC-derived exosomes could protect and regenerate CECs. Methods We performed cell viability and cell-cycle analyses to evaluate the effect of ASC-derived exosomes on the regeneration capacity of cultured hCECs. Transforming growth factor-β (TGF-β) and hydrogen peroxide (H2O2) were used to induce biological stress in CECs. The effect of ASC-derived exosomes on CECs was investigated in vivo. ASC-derived exosomes were introduced into rat CECs using electroporation, and rat corneas were injured using cryoinjury. Next-generation sequencing analysis was performed to compare the differentially expressed microRNAs (miRNAs) between ASC-derived and hCEC-derived exosomes. Results ASC-derived exosomes induced CEC proliferation and suppressed TGF-β- or H2O2-induced oxidative stress and senescence. ASC-derived exosomes protect hCECs against TGF-β- or H2O2-induced endothelial-mesenchymal transition and mitophagy. In an in vivo study, ASC-derived exosomes promoted wound healing of rat CECs and protected the corneal endothelium against cryoinjury-induced corneal endothelium damage. Next-generation sequencing analysis revealed differentially expressed miRNAs for ASC-derived and hCEC-derived exosomes. They are involved in lysine degradation, adherens junction, the TGF-β signaling pathway, the p53 signaling pathway, the Hippo signaling pathway, the forkhead box O (FoxO) signaling pathway, regulation of actin cytoskeleton, and RNA degradation based on Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Conclusions ASC-derived exosomes promoted wound healing and regeneration of endothelial cells by inducing a shift in the cell cycle and suppressing senescence and autophagy.
Collapse
Affiliation(s)
- Yunkyoung Ryu
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Jin Sun Hwang
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Kyung Bo Noh
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Se Hie Park
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Je Hyun Seo
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Young Joo Shin
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
4
|
Darvish L, Bahreyni Toossi MT, Azimian H, Shakeri M, Dolat E, Ahmadizad Firouzjaei A, Rezaie S, Amraee A, Aghaee-Bakhtiari SH. The role of microRNA-induced apoptosis in diverse radioresistant cancers. Cell Signal 2023; 104:110580. [PMID: 36581218 DOI: 10.1016/j.cellsig.2022.110580] [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: 09/18/2022] [Revised: 12/07/2022] [Accepted: 12/21/2022] [Indexed: 12/27/2022]
Abstract
Resistance to cancer radiotherapy is one of the biggest concerns for success in treating and preventing recurrent disease. Malignant tumors may develop when they block genetic mutations associated with apoptosis or abnormal expression of apoptosis; Tumor treatment may induce the expression of apoptosis-related genes to promote tumor cell apoptosis. MicroRNAs have been shown to contribute to forecasting prognosis, distinguishing between cancer subtypes, and affecting treatment outcomes in cancer. Constraining these miRNAs may be an attractive treatment strategy to help overcome radiation resistance. The delivery of these future treatments is still challenging due to the excess downstream targets that each miRNA can control. Understanding the role of miRNAs brings us one step closer to attaining patient treatment and improving patient outcomes. This review summarized the current information on the role of microRNA-induced apoptosis in determining the radiosensitivity of various cancers.
Collapse
Affiliation(s)
- Leili Darvish
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hosein Azimian
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahsa Shakeri
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Dolat
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Ahmadizad Firouzjaei
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samaneh Rezaie
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Azadeh Amraee
- Department of Medical Physics, Faculty of Medicine, School of Medicine, Lorestan University of Medical Sciences, khorramabad, Iran
| | - Seyed Hamid Aghaee-Bakhtiari
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Bioinformatics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
5
|
Zhang YY, Li SQ, Song Y, Wang P, Song XG, Zhu WF, Wang DM. Silencing the ADAM9 Gene through CRISPR/Cas9 Protects Mice from Alcohol-Induced Acute Liver Injury. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5110161. [PMID: 35707386 PMCID: PMC9192226 DOI: 10.1155/2022/5110161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 04/30/2022] [Accepted: 05/11/2022] [Indexed: 11/17/2022]
Abstract
Alcoholic liver injury is a major global public health concern at present. The ADAM9 gene plays a crucial role in the occurrence and development of various liver diseases, but its role in acute alcoholic liver injury remains ambiguous. In this study, a chimeric single-guide RNA targeting the genomic regions of mouse ADAM9 was designed using the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) technology. Next, the role of ADAM9 in acute alcoholic liver injury in vitro in cultured mouse cells and in vivo in a hydrodynamic injection-based alcoholic liver injury mouse model was documented. The findings of this study suggest that ADAM9 induces by regulating cell proliferation, apoptosis, and stress metabolism in mice. Thus, inhibiting the expression of ADAM9 gene using CRISPR/Cas9 can attenuate alcohol-induced acute liver injury in mice.
Collapse
Affiliation(s)
- Yong-Yong Zhang
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471003, China
- Orthopedic Institute of Henan Province, Luoyang, 471003 Henan, China
- Henan Center for Engineering and Technology Research on Prevention and Treatment of Liver Diseases, Luoyang 471003, China
| | - San-Qiang Li
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471003, China
- Henan Center for Engineering and Technology Research on Prevention and Treatment of Liver Diseases, Luoyang 471003, China
| | - Ying Song
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471003, China
- Henan Center for Engineering and Technology Research on Prevention and Treatment of Liver Diseases, Luoyang 471003, China
| | - Ping Wang
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471003, China
- Henan Center for Engineering and Technology Research on Prevention and Treatment of Liver Diseases, Luoyang 471003, China
| | - Xiao-Gai Song
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471003, China
- Henan Center for Engineering and Technology Research on Prevention and Treatment of Liver Diseases, Luoyang 471003, China
| | - Wen-Feng Zhu
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471003, China
- Henan Center for Engineering and Technology Research on Prevention and Treatment of Liver Diseases, Luoyang 471003, China
| | - Dong-Mei Wang
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471003, China
- Henan Center for Engineering and Technology Research on Prevention and Treatment of Liver Diseases, Luoyang 471003, China
| |
Collapse
|
6
|
Lu X, Song X, Hao X, Liu X, Zhang X, Yuan N, Ma H, Zhang Z. miR-186-3p attenuates the tumorigenesis of cervical cancer via targeting insulin-like growth factor 1 to suppress PI3K-Akt signaling pathway. Bioengineered 2021; 12:7079-7092. [PMID: 34551673 PMCID: PMC8806770 DOI: 10.1080/21655979.2021.1977053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
miR-186-3p acts as a tumor suppressor in various cancers. This study aimed to explore the expression levels of miR-186-3p and its role in cervical cancer. We analyzed the effects of miR-186-3p and insulin-like growth factor 1 (IGF1) on the proliferation, invasion, and apoptosis of cervical cancer cells in vitro by regulating the PI3K/Akt signaling pathway. In cervical cancer tissues and cells, miR-186-3p was downregulated, and IGF1 was upregulated. In addition, miR-186-3p inhibited cell proliferation and invasion and enhanced apoptosis of cervical cancer cells. Moreover, our results showed that miR-186-3p inversely regulated the mRNA expression of IGF1 through direct contact. Knockdown of IGF1 reversed the results of miR-186-3p inhibitor in cervical cancer cells. In addition, the PI3K/Akt signaling pathway was activated by the miR-186-3p inhibitor, although partially arrested by IGF1 knockdown. The PI3K/Akt signaling pathway inhibitor suppressed miR-186-3p inhibitor-stimulated cell proliferation in cervical cancer. In conclusion, miR-186-3p inhibits tumorigenesis of cervical cancer by repressing IGF1, which inactivates the PI3K/Akt signaling pathway, implicating miR-186-3p as a potential new target for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Xiurong Lu
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Xiao Song
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Xiaohui Hao
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Xiaoyu Liu
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Xianyu Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Na Yuan
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Huan Ma
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Zhilin Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| |
Collapse
|
7
|
Retraction: Long Noncoding RNA RGMB-AS1 Indicates a Poor Prognosis and Modulates Cell Proliferation, Migration and Invasion in Lung Adenocarcinoma. PLoS One 2021; 16:e0257517. [PMID: 34520497 PMCID: PMC8439456 DOI: 10.1371/journal.pone.0257517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
8
|
Li L, Xie R, Wei Q. Network analysis of miRNA targeting m6A-related genes in patients with esophageal cancer. PeerJ 2021; 9:e11893. [PMID: 34395102 PMCID: PMC8325912 DOI: 10.7717/peerj.11893] [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: 01/28/2021] [Accepted: 07/29/2021] [Indexed: 01/07/2023] Open
Abstract
Background We investigated the miRNA-m6A related gene network and identified a miRNA-based prognostic signature in patients with esophageal cancer using integrated genomic analysis. Methods We obtained expression data for m6A-related genes and miRNAs from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets. Survival analysis was conducted to identify potential prognostic biomarkers. LASSO Cox regression was performed to construct the overall survival (OS) associated prediction signature. We used the Kaplan-Meier (K-M) curve and receiver operating characteristic (ROC) curves to explore the signature's efficiency and accuracy. Interactions between the m6A-related genes and miRNAs were identified in starBase3.0 and used to construct the miRNA-m6A related gene network. Results We found that HNRNPC, YTHDF, ZC3H13, YTHDC2, and METTL14 were dysregulated in esophageal cancer tissues. Multivariate Cox regression analysis revealed that HNRNPC may be an independent risk factor for OS. Five hundred twenty-two potential upstream miRNAs were obtained from starBase3.0. Four miRNAs (miR-186, miR-320c, miR-320d, and miR-320b) were used to construct a prognostic signature, which could serve as a prognostic predictor independent from routine clinicopathological features. Finally, we constructed a key miRNA-m6A related gene network and used one m6A-related gene and four miRNAs associated with the prognosis. The results of our bioinformatics analysis were successfully validated in the human esophageal carcinoma cell lines KYSE30 and TE-1. Conclusion Our study identified a 4-miRNA prognostic signature and established a key miRNA-m6A related gene network. These tools may reliably assist with esophageal cancer patient prognosis.
Collapse
Affiliation(s)
- Lili Li
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Medical Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rongrong Xie
- Department of Medical Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qichun Wei
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
9
|
Zhang Y, Zhang W. FOXD1, negatively regulated by miR-186, promotes the proliferation, metastasis and radioresistance of nasopharyngeal carcinoma cells. Cancer Biomark 2021; 28:511-521. [PMID: 32568181 DOI: 10.3233/cbm-191311] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Foxhead box D1 (FOXD1) is validated to be over-expressed in a variety of human malignancies and promotes cancer progression. Nevertheless, the role of FOXD1 and the associated mechanism in nasopharyngeal carcinoma (NPC) remain largely unknown. METHODS A total of seventy-five cases of NPC tissue samples were collected. FOXD1 expression in NPC tissues and cells (SUNE1, CNE1, CNE2, and HONE1) was detected using immunohistochemistry and Western blot, respectively. The relationship between FOXD1 expression and clinicopathological parameters of NPC patients was analyzed. FOXD1 mRNA and miR-186 expression in NPC tissues and cells was detected using quantitative polymerase chain reaction (qPCR). The cell viability of NPC cells was detected using CCK-8 assay. Colony survival of NPC cells exposed to different doses of radiation was detected using colony formation assay. Transwell assay was used to evaluate the migration and invasion of NPC cells. The dual-luciferase reporter gene assay was employed to verify the targeting relationship between miR-186 and FOXD1. RESULTS FOXD1 was over-expressed in NPC tissues (average fold change on mRNA level = 4.72), and its high expression was correlated to NPC positive lymph node metastasis and tissue differentiation. The over-expression of FOXD1 promoted the proliferation, migration, invasion and radio-resistance of NPC cells. On the contrary, the knock-down of FOXD1 inhibited the malignant phenotypes of the above cells. It was verified that FOXD1 was one of the downstream targets of miR-186 and was negatively regulated by it. CONCLUSION FOXD1, which is negatively regulated by miR-186, acts as a novel oncogene in NPC and serves as potential biomarker and therapeutic target for NPC. The research will provide great theoretical basis for further clinical diagnosis and therapy.
Collapse
|
10
|
Ding L, Sun R, Yan Q, Wang C, Han X, Cui Y, Li R, Liu J. MiR-506 exerts antineoplastic effects on osteosarcoma cells via inhibition of the Skp2 oncoprotein. Aging (Albany NY) 2021; 13:6724-6739. [PMID: 33621206 PMCID: PMC7993745 DOI: 10.18632/aging.202530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/11/2020] [Indexed: 12/27/2022]
Abstract
S-phase kinase-associated protein 2 (Skp2) performs oncogenic functions in cancers; however, how Skp2 is regulated post-transcriptionally is elusive in osteosarcoma. Therefore, we determined whether miR-506 could directly target Skp2 in osteosarcoma to perform its tumor suppressive functions. Here, we found that miR-506 mimics suppressed cell viability, induced apoptosis, and attenuated migration and invasion in osteosarcoma cells. Moreover, upregulation of Skp2 accelerated cell viability and motility and rescued the tumor suppressive effect of miR-506 in osteosarcoma cells. Moreover, downregulation of Skp2 inhibited cell viability and decreased cell motility, which enhanced the antitumor activity induced by miR-506 mimic transfection in osteosarcoma cells. Our western blotting results implied that miR-506 inhibited Skp2 expression and subsequently upregulated Foxo1 and p57 in OS cells. In summary, miR-506 performs an anticancer activity via directly targeting Skp2 in osteosarcoma cells, indicating that inactivation of Skp2 by miR-506 might be an alternative strategy for treating osteosarcoma.
Collapse
Affiliation(s)
- Lu Ding
- Postdoctoral Research Center on Public Health and Preventive Medicine, Xinjiang Medical University, Xinjiang, China.,Fifth Affiliated Hospital, Xinjiang Medical University, Xinjiang, China
| | - Rongxin Sun
- Department of Orthopedics, Sixth Affiliated Hospital, Xinjiang Medical University, Xinjiang, China
| | - Qi Yan
- Department of Maternal, Child and Adolescent Health, College of Public Health, Xinjiang Medical University, Xinjiang, China
| | - Chengwei Wang
- Tumor Hospital Affiliated to Xinjiang Medical University, Xinjiang, China
| | - Xiaoping Han
- Fifth Affiliated Hospital, Xinjiang Medical University, Xinjiang, China
| | - Yong Cui
- Fifth Affiliated Hospital, Xinjiang Medical University, Xinjiang, China
| | - Rong Li
- Postdoctoral Research Center on Public Health and Preventive Medicine, Xinjiang Medical University, Xinjiang, China.,Department of Maternal, Child and Adolescent Health, College of Public Health, Xinjiang Medical University, Xinjiang, China.,Postdoctoral Research Center on Clinical Medicine, First Affiliated Hospital, Xinjiang Medical University, Xinjiang, China
| | - Jiwen Liu
- Postdoctoral Research Center on Public Health and Preventive Medicine, Xinjiang Medical University, Xinjiang, China.,Department of Maternal, Child and Adolescent Health, College of Public Health, Xinjiang Medical University, Xinjiang, China
| |
Collapse
|
11
|
Radiation Can Regulate the Expression of miRNAs Associated with Osteogenesis and Oxidation in Exosomes from Peripheral Blood Plasma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6646323. [PMID: 33628370 PMCID: PMC7899774 DOI: 10.1155/2021/6646323] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/19/2021] [Accepted: 01/28/2021] [Indexed: 02/05/2023]
Abstract
Objectives Radiotherapy is a common therapy in head and neck tumors, which may cause a side effect radiation bone injury (RBI). Furthermore, it has been investigated that microRNA (miRNA) expression levels were altered after radiotherapy. Exosomes play a role in bone formation as miRNA containers, while radiation affects exosomes composition, secretion, and function. So, our objective is to explore changes in miRNA levels during bone formation after radiotherapy and identify the differentially expressed miRNAs (DE-miRs) in plasma exosomes during the process of osteogenesis related to irradiation. Materials and Methods In this study, we analyzed nine samples from three rabbits exposed twice to radiation (15 Gy each) and detected DE-miRs from irradiated plasma exosomes during the process of osteogenesis by RNA sequencing. Further, we identified DE-miRs with significant differences and predicted their target genes via the bioinformatics analysis tools Targetscan v7.2 and miRPathDB v2.0. Finally, we identified radiation-responsive miRNAs and predicted their target genes during osteogenesis. Results Taken together, we have identified some DE-miRs in irradiated plasma exosomes, which were involved in several vital signaling pathways related to bone physiology, such as the Wnt pathway, MAPK cascade, and calcium modulating pathway. Conclusions We have found that plasma exosomes are one of the ways by which radiation can affect bone metabolism and regeneration. However, the specific mechanisms of how these plasma exosomal miRNAs mediate the osteogenesis pathways must be further investigated. Clinical Relevance. Radiotherapy may cause radiation bone injury, and miRNA expression levels in rabbit plasma exosomes are altered after radiotherapy. High-throughput RNA sequencing can identify the differentially expressed miRNAs in irradiated plasma exosomes during the process of osteogenesis. These findings make sense to develop novel therapeutic strategies for treating radiation-induced bone injury disorders.
Collapse
|
12
|
Pourhanifeh MH, Vosough M, Mahjoubin-Tehran M, Hashemipour M, Nejati M, Abbasi-Kolli M, Sahebkar A, Mirzaei H. Autophagy-related microRNAs: Possible regulatory roles and therapeutic potential in and gastrointestinal cancers. Pharmacol Res 2020; 161:105133. [DOI: 10.1016/j.phrs.2020.105133] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/23/2020] [Accepted: 08/07/2020] [Indexed: 02/08/2023]
|
13
|
He W, Pang L, Gong S, Wang X, Hou L. Nei Endonuclease VIII-like 2 Gene rs8191670 Polymorphism affects the Sensitivity of Non-small Cell Lung Cancer to Cisplatin by binding with MiR-548a. J Cancer 2020; 11:4801-4809. [PMID: 32626527 PMCID: PMC7330683 DOI: 10.7150/jca.47495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 05/25/2020] [Indexed: 11/05/2022] Open
Abstract
Background: Nei endonuclease VIII-like 2 (NEIL2) is a gene encoding DNA repair enzyme, which is involved in the base excision repair (BER) pathway in mammalian cells. Cisplatin is a common cytotoxic anti-tumor agent in clinic by destroying normal structure of DNA and inducing cell apoptosis. However, how NEIL2 affects the sensitivity of NSCLC to cisplatin is still unclear. Methods: The clinical data from 206 patients diagnosed pathologically were collected. The DNA sequencing of NEIL2 gene 3'UTR and the PFS curve of NSCLC patients receiving cisplatin-based chemotherapy were performed. Western blot analysis and immunohistochemistry were used to detect NEIL2 protein expression. Human NSCLC cell lines A549 and H1299 were cultured and evaluated for cell viability. RT-PCR was performed for quantitative detection of miR-548a. 3'UTR reporter plasmid was constructed and luciferase reporter assay was used to verify the target gene regulated by miR-548a. Results: In this study, we found that the Neil2 gene had the polymorphism (T/C) in rs8191670 and it is associated with the PFS of advanced NSCLC patients. MiR-548a targets NEIL2 3'UTR to suppress its expression. Upregulation of NEIL2 expression or downregulation of miR-548a could reduce the sensitivity of NSCLC cells to cisplatin. Conclusion: Our results demonstrated that NEIL2 gene rs8191670 polymorphism affects the PFS of advanced NSCLC patients, and the underlying molecular mechanisms may be that miR-548a can regulate NEIL2 expression by binding to its 3'UTR seed region containing rs8191670.
Collapse
Affiliation(s)
- Wei He
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Lina Pang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shuai Gong
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xin Wang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Lixia Hou
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
14
|
Xiang Y, Tian Q, Guan L, Niu SS. The Dual Role of miR-186 in Cancers: Oncomir Battling With Tumor Suppressor miRNA. Front Oncol 2020; 10:233. [PMID: 32195180 PMCID: PMC7066114 DOI: 10.3389/fonc.2020.00233] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/11/2020] [Indexed: 12/18/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs which regulate gene expression at post-transcriptional level. Alterations of miR-186 expression were demonstrated in numerous cancers, shown to play a vital role in oncogenesis, invasion, metastasis, apoptosis, and drug resistance. MiR-186 was documented as a tumor suppressor miRNA in the majority of studies, while conflicting reports verified miR-186 as an oncomir. The contradictory role in cancers may impede the application of miR-186, as well as other dual-functional miRNAs, as a diagnostic and therapeutic target. This review emphasizes the alterations and functions of miR-186 in cancers and discusses the mechanisms behind the contradictory findings. Among these, target abundance and dose-dependent effects of miR-186 are highlighted. The paper aims to review the challenges involved in developing diagnostic and therapeutic strategies for cancer treatment based on dual-functional miRNAs.
Collapse
Affiliation(s)
- Ying Xiang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Hubei, China.,Department of Cell Biology and Genetics, School of Basic Medicine, Health Science Center, Yangtze University, Hubei, China
| | - Qing Tian
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Hubei, China
| | - Li Guan
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Hubei, China
| | - Shuai-Shuai Niu
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Hubei, China.,The First School of Clinical Medicine, Health Science Center, Yangtze University, Hubei, China
| |
Collapse
|
15
|
Mu Y, Wang Q, Tan L, Lin L, Zhang B. microRNA-144 inhibits cell proliferation and invasion by directly targeting TIGAR in esophageal carcinoma. Oncol Lett 2020; 19:3079-3088. [PMID: 32256808 PMCID: PMC7074326 DOI: 10.3892/ol.2020.11420] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 10/18/2019] [Indexed: 12/21/2022] Open
Abstract
microRNAs (miRNAs) have been identified to play vital roles in the development and progression of numerous different types of human malignancy, including esophageal squamous cell carcinoma (ESCC). In the present study, the biological function of microRNA-144 (miR-144) was investigated, as well as its underlying molecular mechanism in ESCC. The results revealed that miR-144 expression was significantly decreased, whereas the expression of TP53-inducible glycolysis and apoptosis regulator (TIGAR) was significantly increased in human ESCC tissues when compared with adjacent non-tumor tissues. An increase in TIGAR was significantly associated with tumor size and Tumor-Node-Metastasis staging in patients. Functional analysis revealed that the overexpression of miR-144 using lentivirus particles significantly inhibited cell proliferation and tumor colony formation, and induced cell apoptosis in EC9706 and EC109 cells. The autophagy activity was also enhanced by miR-144 activity. In addition, overexpression of miR-144 significantly inhibited tumor growth in vivo. In the present study, TIGAR was confirmed to be the downstream target of miR-144 in ESCC. siRNA-mediated downregulation of TIGAR inversely regulated the inhibition effect of miR-144 on ESCC cells. To conclude, the present study demonstrated that miR-144 inhibits proliferation and invasion in esophageal cancer by directly targeting TIGAR.
Collapse
Affiliation(s)
- Yushu Mu
- Department of Thoracic Surgery, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, P.R. China
| | - Qifei Wang
- Department of Thoracic Surgery, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, P.R. China
| | - Lei Tan
- Department of Thoracic Surgery, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Lin Lin
- Department of Digestive Medicine, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Benhua Zhang
- Department of Oncology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, P.R. China
| |
Collapse
|
16
|
Xie H, Chen J, Lv X, Zhang L, Wu J, Ge X, Yang Q, Zhang D, Chen J. Clinical Value of Serum and Exhaled Breath Condensate miR-186 and IL-1β Levels in Non-Small Cell Lung Cancer. Technol Cancer Res Treat 2020; 19:1533033820947490. [PMID: 32851926 PMCID: PMC7457640 DOI: 10.1177/1533033820947490] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 06/28/2020] [Accepted: 07/13/2020] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Our study aimed to investigate the expression level and clinical significance of serum and exhaled breath condensate miR-186 and IL-1β in non-small cell lung cancer patients. METHODS The serum and exhaled breath condensate specimens of 62 non-small cell lung cancer patients and 60 healthy controls were collected to detect miR-186 expression levels by real-time fluorescent quantitative PCR. Enzyme linked immunosorbent assay was applied to examine IL-1β concentration. Statistical analyses were used to evaluate the correlation between miR-186 and IL-1β in serum and clinicopathological features, traditional serum tumor markers, and inflammatory markers. The diagnostic efficacy of miR-186 and IL-1β for non-small cell lung cancer was evaluated by receiver operating characteristic curve analysis. The correlation between miR-186 and IL-1β was determined. RESULTS ① The relative expression level of miR-186 was greatly reduced in the serum and EBC of patients with non-small cell lung cancer, and the miR-186 expression level was reduced in different TNM stages of non-small cell lung cancer, from the early to later stages. ② The IL-1β concentration in serum and exhaled breath condensate of patients with non-small cell lung cancer was increased. ③ Serum miR-186 and IL-1β levels were closely related to lymph node metastasis, and the low expression of serum miR-186 and the high concentration of IL-1β were associated with higher serum carcinoembryonic antigen, C-reactive protein, and erythrocyte sedimentation rate levels. ④ ROC curve analysis showed that exhaled breath condensate miR-186 had higher area under the curve than serum miR-186, and the combined detection showed higher diagnostic efficacy than the separate detection. In addition, the combined detection of IL-1β and miR-186 has a larger AUC than the separate detection of both. ⑤ The correlation between serum miR-186 and IL-1β was negative. CONCLUSION miR-186 and IL-1β are expected to be potential diagnostic biomarkers for non-small cell lung cancer.
Collapse
Affiliation(s)
- Haiqin Xie
- Department of Respiratory Medicine, Affiliated Hospital 2 of Nantong
University, Nantong, China
| | - Jinliang Chen
- Department of Respiratory Medicine, Affiliated Hospital 2 of Nantong
University, Nantong, China
| | - Xuedong Lv
- Department of Respiratory Medicine, Affiliated Hospital 2 of Nantong
University, Nantong, China
| | - Lu Zhang
- Department of Respiratory Medicine, Affiliated Hospital 2 of Nantong
University, Nantong, China
| | - Jinnan Wu
- Department of Respiratory Medicine, Affiliated Hospital 2 of Nantong
University, Nantong, China
| | - Xin Ge
- Medical Research Center, Affiliated Hospital 2 of Nantong
University, Nantong, China
| | - Qichang Yang
- Department of Pathology, Affiliated Hospital 2 of Nantong
University, Nantong, China
| | - Dongmei Zhang
- Medical Research Center, Affiliated Hospital 2 of Nantong
University, Nantong, China
| | - Jianrong Chen
- Department of Respiratory Medicine, Affiliated Hospital 2 of Nantong
University, Nantong, China
- Medical Research Center, Affiliated Hospital 2 of Nantong
University, Nantong, China
| |
Collapse
|
17
|
Lin M, Xu Y, Gao Y, Pan C, Zhu X, Wang ZW. Regulation of F-box proteins by noncoding RNAs in human cancers. Cancer Lett 2019; 466:61-70. [DOI: 10.1016/j.canlet.2019.09.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/11/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022]
|
18
|
Wang Z, Sha HH, Li HJ. Functions and mechanisms of miR-186 in human cancer. Biomed Pharmacother 2019; 119:109428. [PMID: 31525641 DOI: 10.1016/j.biopha.2019.109428] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/27/2019] [Accepted: 09/02/2019] [Indexed: 12/30/2022] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression at the post-transcriptional level. Mounting evidence suggests the involvement of miRNAs in carcinogenesis and the development of human cancer. Among the miRNAs, miR-186 has been extensively studied in various cancers. The expression of miR-186 in tissues varies depending on the type of cancer and miR-186 in tissues and body fluids may serve as a marker for the diagnosis and prognosis of cancers. Various biological processes in human cancer are affected by miR-186. Additionally, miR-186 itself is regulated by several factors. Thus, this evidence highlights the potential value of miR-186 in the diagnosis, prognosis and treatment of human cancer.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Orthopedics, Taizhou Clinical Medical School of Nanjing Medical University (Taizhou People's Hospital), Taizhou, Jiangsu, China
| | - Huan-Huan Sha
- Department of Chemotherapy, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, China
| | - Hai-Jun Li
- Department of Orthopedics, Taizhou Clinical Medical School of Nanjing Medical University (Taizhou People's Hospital), Taizhou, Jiangsu, China.
| |
Collapse
|
19
|
Chen H, Li M, Huang P. LncRNA SNHG16 Promotes Hepatocellular Carcinoma Proliferation, Migration and Invasion by Regulating miR-186 Expression. J Cancer 2019; 10:3571-3581. [PMID: 31293662 PMCID: PMC6603422 DOI: 10.7150/jca.28428] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 04/30/2019] [Indexed: 12/11/2022] Open
Abstract
Long non-coding RNA (lncRNA) and microRNA (miRNA) play an important role in genesis and progression of tumors. The aim of this study was to explore the expression, biological function and molecular mechanism of small nucleolar RNA host gene 16 (SNHG16) in HCC. RT-qPCR was conducted to evaluate the expression level of SNHG16 in HCC tissues and cell lines. Our findings showed for the first time that SNHG16 was up-regulated in HCC tissues and cell lines. The expression of SNHG16 in cancer tissues was highly correlated with tumor size, TNM stage, ALT expression level and HBV DNA level. Moreover, cell proliferation, migration and invasion were detected by CCK-8 assay, transwell migration assay and transwell invasion assay, respectively. Xenograft tumor experiment was used to determine the biological function of SNHG16 in vivo. As revealed by our data, SNHG16 accelerated the proliferation, migration and invasion of HCC cell. SNHG16 facilitated tumor formation in vivo. Next, the relationship between SNHG16, miR-186 and ROCK1 were analyzed using bioinformatics analysis, qRT-PCR, luciferase reporter assay and western blot. Further molecular mechanism studies reported that the expression of SNHG16 was negatively correlated with the level of miR-186 and SNHG16 directly bound to miR-186. SNHG16 and miR-186 repressed each other. Notably, rescue experiments were conducted and showed that miR-186 reversed the effect of SNHG16 on cell. Taken together, SNHG16 promoted HCC cell proliferation, migration and invasion by functioning as a competitive endogenous RNA (ceRNA) to negatively regulate miR-186 expression. Our data suggested that SNHG16 might be a potential biomarker and a new therapeutic target for HCC.
Collapse
Affiliation(s)
- Hang Chen
- National Key Clinical Department, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 400000, P.R. China
| | - Molin Li
- National Key Clinical Department, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 400000, P.R. China
| | - Ping Huang
- National Key Clinical Department, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 400000, P.R. China
| |
Collapse
|
20
|
Wang H, Ou J, Jian Z, Ou Y. miR-186 modulates hepatocellular carcinoma cell proliferation and mobility via targeting MCRS1-mediated Wnt/β-catenin signaling. J Cell Physiol 2019; 234:23135-23145. [PMID: 31140612 DOI: 10.1002/jcp.28878] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/03/2019] [Accepted: 05/07/2019] [Indexed: 12/20/2022]
Abstract
Previous studies have revealed that miR-186 is involved in the pathogenesis of many malignancies. However, the role of miR-186 in hepatocellular carcinoma (HCC) carcinogenesis and its detailed mechanism are poorly understood. This study was to investigate the function of miR-186 in modulating HCC cell proliferation, cell cycle, migration, and invasion. We found that miR-186 was decreased in HCC tissues and cell lines. Loss-of-function experiments showed that reduction of miR-186 dramatically enhanced tumor cell proliferation and metastasis. Besides, miR-186 also participated in the modulation of the cell cycle. In addition, luciferase reporter assays and Western blot analysis showed that MCRS1 was a novel target of miR-186 in HCC cells. Notably, upregulation of miR-186 suppressed the nuclear β-catenin accumulation and blocked the activation of Wnt/β-catenin signaling in HCC cells. Forced MCRS1 expression abrogated the inhibitory effect of miR-186 on cell growth, metastasis and Wnt/β-catenin signaling in HCC cells. Our findings may provide new insight into the pathogenesis of HCC and miR-186/ MCRS1 might function as new therapeutic targets for HCC.
Collapse
Affiliation(s)
- Huiling Wang
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jinrui Ou
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhixiang Jian
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yingliang Ou
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
21
|
Neviani P, Wise PM, Murtadha M, Liu CW, Wu CH, Jong AY, Seeger RC, Fabbri M. Natural Killer-Derived Exosomal miR-186 Inhibits Neuroblastoma Growth and Immune Escape Mechanisms. Cancer Res 2018; 79:1151-1164. [PMID: 30541743 DOI: 10.1158/0008-5472.can-18-0779] [Citation(s) in RCA: 215] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 10/24/2018] [Accepted: 12/06/2018] [Indexed: 12/18/2022]
Abstract
In neuroblastoma, the interplay between immune cells of the tumor microenvironment and cancer cells contributes to immune escape mechanisms and drug resistance. In this study, we show that natural killer (NK) cell-derived exosomes carrying the tumor suppressor microRNA (miR)-186 exhibit cytotoxicity against MYCN-amplified neuroblastoma cell lines. The cytotoxic potential of these exosomes was partly dependent upon expression of miR-186. miR-186 was downregulated in high-risk neuroblastoma patients, and its low expression represented a poor prognostic factor that directly correlated with NK activation markers (i.e., NKG2D and DNAM-1). Expression of MYCN, AURKA, TGFBR1, and TGFBR2 was directly inhibited by miR-186. Targeted delivery of miR-186 to MYCN-amplified neuroblastoma or NK cells resulted in inhibition of neuroblastoma tumorigenic potential and prevented the TGFβ1-dependent inhibition of NK cells. Altogether, these data support the investigation of a miR-186-containing nanoparticle formulation to prevent tumor growth and TGFβ1-dependent immune escape in high-risk neuroblastoma patients as well as the inclusion of ex vivo-derived NK exosomes as a potential therapeutic option alongside NK cell-based immunotherapy.Significance: These findings highlight the therapeutic potential of NK cell-derived exosomes containing the tumor suppressor miR-186 that inhibits growth, spreading, and TGFβ-dependent immune escape mechanisms in neuroblastoma.
Collapse
Affiliation(s)
- Paolo Neviani
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Petra M Wise
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Mariam Murtadha
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Cathy W Liu
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Chun-Hua Wu
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Ambrose Y Jong
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Robert C Seeger
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Muller Fabbri
- Children's Center for Cancer and Blood Diseases and Divisions of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, USC-Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California.
| |
Collapse
|
22
|
Wang R, Bao H, Zhang S, Li R, Chen L, Zhu Y. miR-186-5p Promotes Apoptosis by Targeting IGF-1 in SH-SY5Y OGD/R Model. Int J Biol Sci 2018; 14:1791-1799. [PMID: 30443183 PMCID: PMC6231212 DOI: 10.7150/ijbs.25352] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 08/01/2018] [Indexed: 12/20/2022] Open
Abstract
In recent years, accumulating evidence has revealed that microRNAs play critical roles in ischemia stroke. This study was designed to investigate the expression level and effects of microRNA (miR)-186-5p on ischemia stroke, and its underlying molecular mechanism. Firstly, we demonstrated that miR-186-5p were significantly up-regulated and induced apoptosis in oxygen and glucose deprivation/reperfusion (OGD/R) model. Moreover, we found that miR-186-5p reduced the expression of insulin-like growth factor (IGF)-1, an essential factor for the development of the nervous system. Meanwhile, miR-186-5p inhibitor enhanced cell viability and IGF-1 expression. Furthermore, IGF-1 was confirmed as a direct target gene of miR-186-5p by luciferase activity assay. In addition, miR-186-5p was upregulated in ischemia stroke patients' serum compared with healthy donors. These data demonstrated that miR-186-5p was an adverse factor by inducing neuron apoptosis and suppressing IGF-1 in ischemia stroke model, and suggested that miR-186-5p may be a diagnostic marker and potential therapeutic target for ischemia stroke patients.
Collapse
Affiliation(s)
- Rui Wang
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
| | - Hongbo Bao
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, 150001, China
| | - Shihua Zhang
- Department of Neurosurgery, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province, 154002, China
| | - Ruiyan Li
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
| | - Lijie Chen
- Department of Neurology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450000, China
| | - Yulan Zhu
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
| |
Collapse
|
23
|
Li J, Song J, Guo F. miR-186 reverses cisplatin resistance and inhibits the formation of the glioblastoma-initiating cell phenotype by degrading Yin Yang 1 in glioblastoma. Int J Mol Med 2018; 43:517-524. [PMID: 30365062 DOI: 10.3892/ijmm.2018.3940] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 10/08/2018] [Indexed: 11/05/2022] Open
Abstract
Glioblastoma multiforme (GBM) is among the most devastating types of cancer, with a median survival of <1 year. Despite the development of new surgical and radiation techniques, and the use of multiple anti‑neoplastic drugs, effective treatment strategies for malignant gliomas have not yet been developed. The limited efficacy of current treatments reflects the resistance of glioblastoma cells to cytotoxic agents. In this study, using western blot analysis, we found that Yin Yang 1 (YY1) expression was increased in cisplatin‑resistant glioblastoma U87MG cells (U87MG‑CR). We observed that the silencing of YY1 sensitized the U87MG‑CR cells to cisplatin and that the overexpression of YY1 promoted the resistance of LN‑229 glioblastoma cells to cisplatin, as shown by MTT assay. Using sphere formation assay, we also found that the silencing of YY1 inhibited the formation of the glioblastoma‑initiating cell (GIC) phenotype in the U87MG‑CR cells. In addition, the results of RT‑qPCR revealed that miR‑186 expression was decreased in U87MG‑CR cells. Using RT‑PCR and western blot analysis, we observed that overexpression of miR‑186 inhibited YY1 expression in U87MG‑CR cells. The overexpression of miR‑186 also reversed cisplatin resistance and the formation of the GIC phenotype in glioblastoma cells. On the whole, the findings of this study demonstrate that miR‑186 reverses cisplatin resistance and inhibits the formation of the GIC phenotype by degrading YY1 in glioblastoma.
Collapse
Affiliation(s)
- Jian Li
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Jie Song
- Department of Neurosurgery, Yishui Central Hospital, Yishui, Shandong 276400, P.R. China
| | - Feng Guo
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| |
Collapse
|
24
|
Wu D, Wen X, Wang Y, Han X, Wang S, Shen M, Fan S, Zhuang J, Zhang Z, Shan Q, Li M, Hu B, Sun C, Lu J, Chen G, Zheng Y. Retracted
: Effect of microRNA‐186 on oxidative stress injury of neuron by targeting interleukin 2 through the janus kinase‐signal transducer and activator of transcription pathway in a rat model of Alzheimer’s disease. J Cell Physiol 2018; 233:9488-9502. [DOI: 10.1002/jcp.26843] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 05/10/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Dong‐Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Xin Wen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Yong‐Jian Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Xin‐Rui Han
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Shan Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Min Shen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Shao‐Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Juan Zhuang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- School of Environment Science and Spatial Informatics China University of Mining and Technology Xuzhou China
- Jiangsu Key Laboratory for Eco‐Agricultural Biotechnology Around Hongze Lake, School of Life Sciences Huaiyin Normal University Huaian China
| | - Zi‐Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Qun Shan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Meng‐Qiu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Chun‐Hui Sun
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Gui‐Quan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center Nanjing University Nanjing China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center Nanjing University Nanjing China
| | - Yuan‐Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| |
Collapse
|
25
|
Lin YS, Lin YY, Yang YH, Lin CL, Kuan FC, Lu CN, Chang GH, Tsai MS, Hsu CM, Yeh RA, Yang PR, Lee IY, Shu LH, Cheng YC, Liu HT, Lee KD, Chang DC, Wu CY. Antrodia cinnamomea extract inhibits the proliferation of tamoxifen-resistant breast cancer cells through apoptosis and skp2/microRNAs pathway. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:152. [PMID: 29743060 PMCID: PMC5944021 DOI: 10.1186/s12906-018-2204-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 04/16/2018] [Indexed: 01/07/2023]
Abstract
BACKGROUND Breast cancer is the most common cancer in women and affects 1.38 million women worldwide per year. Antiestrogens such as tamoxifen, a selective estrogen receptor (ER) modulator, are widely used in clinics to treat ER-positive breast tumors. However, remissions of breast cancer are often followed by resistance to tamoxifen and disease relapse. Despite the increasing understanding of the resistance mechanisms, effective regimens for treating tamoxifen-resistant breast cancer are limited. Antrodia cinnamomea is a traditional medicinal mushroom native only to Taiwan. In this study, we aimed to examine in vitro effect of antrodia cinnamomea in the tamoxifen-resistant cancer. METHODS Antrodia cinnamomea was studied for its biological activity against proliferation of tamoxifen-resistant breast cancer by XTT assay. Next, the underlying mechanism was studied by flow cytometry, qPCR and Western's blotting assay. RESULTS Our results revealed that the ethanol extract of antrodia cinnamomea (AC) can inhibit the growth of breast cancer cells, including MCF-7 cell and tamoxifen-resistant MCF-7 cell lines. Combination treatment with AC and 10- 6 M tamoxifen have the better inhibitory effect on the proliferation of tamoxifen-resistant MCF-7 cells than only AC did. AC can induce apoptosis in these breast cancer cells. Moreover, it can suppress the mRNA expression of skp2 (S-phase kinase-associated protein 2) by increasing the expressions of miR-21-5p, miR-26-5p, and miR-30-5p in MCF-7 and tamoxifen-resistant MCF-7 cells. CONCLUSIONS These results suggest that the ethanol extract of antrodia cinnamomea could be a novel anticancer agent in the armamentarium of tamoxifen-resistant breast cancer management. Moreover, we hope to identify additional pure compounds that could serve as promising anti-breast cancer candidates for further clinical trials.
Collapse
Affiliation(s)
- Yu-Shih Lin
- Department of Pharmacy, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- Institute of Molecular Biology, National Chung Cheng University, No.168, Sec. 1, University Rd., Minhsiung Chiayi County, 62102, Taiwan, Republic of China
| | - Yin-Yin Lin
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yao-Hsu Yang
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- School of Chinese medicine, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Center of Excellence for Chang Gung Research Datalink, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Chun-Liang Lin
- Departments of Nephrology, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- Kidney and Diabetic Complications Research Team (KDCRT), Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Feng-Che Kuan
- Department of Hematology and oncology, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Cheng-Nan Lu
- Division of Acupuncture and Chinese Traumatology, Department of TCM, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Geng-He Chang
- Department of Otolaryngology, Chang Gung Memorial Hospital, Chiayi, Taiwan
- Center of Excellence for Chang Gung Research Datalink, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Ming-Shao Tsai
- Department of Otolaryngology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Cheng-Ming Hsu
- Department of Otolaryngology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Reming-Albert Yeh
- Department of Otolaryngology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Pei-Rung Yang
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - I-Yun Lee
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Li-Hsin Shu
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yu-Ching Cheng
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Hung-Te Liu
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Kuan-Der Lee
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - De-Ching Chang
- Institute of Molecular Biology, National Chung Cheng University, No.168, Sec. 1, University Rd., Minhsiung Chiayi County, 62102, Taiwan, Republic of China.
| | - Ching-Yuan Wu
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan.
- School of Chinese medicine, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.
| |
Collapse
|
26
|
Jones DZ, Schmidt ML, Suman S, Hobbing KR, Barve SS, Gobejishvili L, Brock G, Klinge CM, Rai SN, Park J, Clark GJ, Agarwal R, Kidd LR. Micro-RNA-186-5p inhibition attenuates proliferation, anchorage independent growth and invasion in metastatic prostate cancer cells. BMC Cancer 2018; 18:421. [PMID: 29653561 PMCID: PMC5899400 DOI: 10.1186/s12885-018-4258-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 03/20/2018] [Indexed: 12/17/2022] Open
Abstract
Background Dysregulation of microRNA (miRNA) expression is associated with hallmarks of aggressive tumor phenotypes, e.g., enhanced cell growth, proliferation, invasion, and anchorage independent growth in prostate cancer (PCa). Methods Serum-based miRNA profiling involved 15 men diagnosed with non-metastatic (stage I, III) and metastatic (stage IV) PCa and five age-matched disease-free men using miRNA arrays with select targets confirmed by quantitative real-time PCR (qRT-PCR). The effect of miR-186-5p inhibition or ectopic expression on cellular behavior of PCa cells (i.e., PC-3, MDA-PCa-2b, and LNCaP) involved the use bromodeoxyuridine (BrdU) incorporation, invasion, and colony formation assays. Assessment of the impact of miR-186-5p inhibition or overexpression on selected targets entailed microarray analysis, qRT-PCR, and/or western blots. Statistical evaluation used the modified t-test and ANOVA analysis. Results MiR-186-5p was upregulated in serum from PCa patients and metastatic PCa cell lines (i.e., PC-3, MDA-PCa-2b, LNCaP) compared to serum from disease-free individuals or a normal prostate epithelial cell line (RWPE1), respectively. Inhibition of miR-186-5p reduced cell proliferation, invasion, and anchorage-independent growth of PC-3 and/or MDA-PCa-2b PCa cells. AKAP12, a tumor suppressor target of miR-186-5p, was upregulated in PC-3 and MDA-PCa-2b cells transfected with a miR-186-5p inhibitor. Conversely, ectopic miR-186-5p expression in HEK 293 T cells decreased AKAP12 expression by 30%. Both pAKT and β-catenin levels were down-regulated in miR-186-5p inhibited PCa cells. Conclusions Our findings suggest miR-186-5p plays an oncogenic role in PCa. Inhibition of miR-186-5p reduced PCa cell proliferation and invasion as well as increased AKAP12 expression. Future studies should explore whether miR-186-5p may serve as a candidate prognostic indicator and a therapeutic target for the treatment of aggressive prostate cancer. Electronic supplementary material The online version of this article (10.1186/s12885-018-4258-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dominique Z Jones
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.,James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA.,Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Denver, USA
| | - M Lee Schmidt
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.,James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Suman Suman
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.,James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Katharine R Hobbing
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.,James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Shirish S Barve
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.,Division of Gastroenterology and Hepatology, University of Louisville School of Medicine, Louisville, USA
| | - Leila Gobejishvili
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.,Division of Gastroenterology and Hepatology, University of Louisville School of Medicine, Louisville, USA
| | - Guy Brock
- Department of Biomedical Informatics, The Ohio State University, Columbus, USA
| | - Carolyn M Klinge
- James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA.,Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, USA
| | - Shesh N Rai
- James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA.,Department of Bioinformatics and Biostatistics, University of Louisville School of Public Health and Information Science, Louisville, USA
| | - Jong Park
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, USA
| | - Geoffrey J Clark
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.,James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Denver, USA
| | - LaCreis R Kidd
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA. .,James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA.
| |
Collapse
|
27
|
Construction of differential mRNA-lncRNA crosstalk networks based on ceRNA hypothesis uncover key roles of lncRNAs implicated in esophageal squamous cell carcinoma. Oncotarget 2018; 7:85728-85740. [PMID: 27966444 PMCID: PMC5349869 DOI: 10.18632/oncotarget.13828] [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: 09/24/2016] [Accepted: 11/18/2016] [Indexed: 12/14/2022] Open
Abstract
Increasing evidence has indicated that lncRNAs acting as competing endogenous RNAs (ceRNAs) play crucial roles in tumorigenesis, metastasis and diagnosis of cancer. However, the function of lncRNAs as ceRNAs involved in esophageal squamous cell carcinoma (ESCC) is still largely unknown. In this study, clinical implications of two intrinsic subtypes of ESCC were identified based on expression profiles of lncRNA and mRNA. ESCC subtype-specific differential co-expression networks between mRNAs and lncRNAs were constructed to reveal dynamic changes of their crosstalks mediated by miRNAs during tumorigenesis. Several well-known cancer-associated lncRNAs as the hubs of the two networks were firstly proposed in ESCC. Based on the ceRNA mechanism, we illustrated that the"loss" of miR-186-mediated PVT1-mRNA and miR-26b-mediated LINC00240-mRNA crosstalks were related to the two ESCC subtypes respectively. In addition, crosstalks between LINC00152 and EGFR, LINC00240 and LOX gene family were identified, which were associated with the function of "response to wounding" and "extracellular matrix-receptor interaction". Furthermore, functional cooperation of multiple lncRNAs was discovered in the two differential mRNA-lncRNA crosstalk networks. These together systematically uncovered the roles of lncRNAs as ceRNAs implicated in ESCC.
Collapse
|
28
|
Suppression of Disheveled–Axin Domain Containing 1 (DIXDC1) by MicroRNA-186 Inhibits the Proliferation and Invasion of Retinoblastoma Cells. J Mol Neurosci 2017; 64:252-261. [DOI: 10.1007/s12031-017-1017-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/12/2017] [Indexed: 12/19/2022]
|
29
|
Jiang J, Mo H, Liu C, Wu B, Wu Z, Li X, Li T, He S, Li S, You Q, Wu K, Guo R. Inhibition of miR-186-5p contributes to high glucose-induced injury in AC16 cardiomyocytes. Exp Ther Med 2017; 15:627-632. [PMID: 29399065 PMCID: PMC5772612 DOI: 10.3892/etm.2017.5445] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/04/2017] [Indexed: 01/12/2023] Open
Abstract
A growing body of evidence has demonstrated that microRNAs (miRs) have pivotal roles in the pathophysiological development mechanisms of diabetic cardiomyopathy (DCM). Previous studies have demonstrated that miR-186-5p was significantly decreased in DCM. In addition, it has recently been reported that an imbalance of miR-186 is associated with a variety of physiological and pathological processes. Therefore, the present study was designed to investigate the role of miR-186-5p in high glucose (HG)-induced cytotoxicity and apoptosis in AC16 cardiomyocytes. Reverse transcription-polymerase chain reaction was used to demonstrate the significant decrease in the level of miR-186-5p in HG-treated AC16 cells (P<0.05). Subsequently, it was clarified that pre-transfection with miR-186-5p mimic significantly ameliorated the effects of high glucose, which induced a significant decrease in the viability of AC16 cells (P<0.05) and increases in apoptosis, as evidenced by the appearance of apoptotic nucleus and the significant upregulation of apoptosis rate in AC16 cells (P<0.05). In addition, the significantly increased expression of caspase-3 induced by HG (P<0.01) was also reversed by miR-186-5p mimic (P<0.01). Conversely, transfection with miR-186-5p inhibitor significantly reduced the viability of AC16 cells (P<0.05) and promoted apoptosis (P<0.05) as well as the expression of caspase-3 in AC16 cells (P<0.01), indicating the beneficial role of miR-186-5p in the physiological process of HG-induced damage. In conclusion, these results suggest that the distribution of miR-186-5p contributes to HG-induced cytotoxicity and apoptosis in AC16 cardiomyocytes.
Collapse
Affiliation(s)
- Jiamei Jiang
- Department of Cardiology, The Affiliated Hospital, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Hailiang Mo
- Department of Cardiology, The Affiliated Hospital, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Chang Liu
- Department of Cardiology, The Affiliated Hospital, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Bin Wu
- Department of Cardiology, The Affiliated Hospital, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Zijun Wu
- Department of Cardiology, The Affiliated Hospital, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Xingyue Li
- Department of Cardiology, The Affiliated Hospital, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Teng Li
- Department of Cardiology, The Affiliated Hospital, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Songjian He
- Department of Cardiology, The Affiliated Hospital, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Shanghai Li
- Department of Cardiology, The Affiliated Hospital, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Qiong You
- Department of Cardiology, The Affiliated Hospital, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Keng Wu
- Department of Cardiology, The Affiliated Hospital, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Runmin Guo
- Department of Cardiology, The Affiliated Hospital, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
30
|
Wang F, Jiang H, Wang S, Chen B. Dual Functional MicroRNA-186-5p Targets both FGF2 and RelA to Suppress Tumorigenesis of Glioblastoma Multiforme. Cell Mol Neurobiol 2017; 37:1433-1442. [PMID: 28213656 DOI: 10.1007/s10571-017-0474-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/12/2017] [Indexed: 01/08/2023]
Abstract
Glioblastoma multiforme (GBM) is one of the most malignant cancers. MicroRNAs (miRs) were reported to play important roles in GBM recently. However, the role of a novel miR-186-5p in GBM tumorigenesis is still elusive. Using bioinformatics, miR-186-5p was identified as potential regulators of both fibroblast growth factor (FGF)-2 and NF-κB subunit RelA. Luciferase reporter assay was used to confirm the direct recognition FGF2 and RelA mRNAs by miR-186-5p. Invasion and migration assays were employed to study the effect of miR-186-5p on GBM cell growth in vitro. Xenograft tumor animal model was established to elucidate the in vivo function of miR-186-5p. MiR-186-5p directly targeted mRNAs of both FGF2 and RelA, and repressed their expressions. Invasive and migratory abilities of GBM cells and growth of xenograft tumors were significantly inhibited by miR-186-5p, which can be restored by re-introduction of FGF2 and RelA expressions. MiR-186-5p is a novel tumor suppressor miR that functions to inhibit tumorigenesis of GBM both in vitro and in vivo, by targeting both FGF2 and RelA. MiR-186-5p/FGF2/RelA pathway may be potentially used as molecular targets of in the clinical treatment of GBM.
Collapse
Affiliation(s)
- Fachen Wang
- Department of Neurosurgery, Yidu Central Hospital of Weifang, No. 4138 Linglongshan Road, Qingzhoushi, Shandong, People's Republic of China
| | - Hui Jiang
- Department of Medicine, Qingzhou Hospital of Traditional Chinese Medicine, No.2727 Haidai Road, Qingzhoushi, Shandong, People's Republic of China
| | - Shanjun Wang
- Department of Neurosurgery, Yidu Central Hospital of Weifang, No. 4138 Linglongshan Road, Qingzhoushi, Shandong, People's Republic of China
| | - Bing Chen
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, No. 1677 Wutaishan Road, Qingdao, Shandong, People's Republic of China.
| |
Collapse
|
31
|
Jiang J, Wang W, Fang D, Jin X, Ding L, Sun X. MicroRNA-186 targets IGF-1R and exerts tumor-suppressing functions in glioma. Mol Med Rep 2017; 16:7821-7828. [DOI: 10.3892/mmr.2017.7586] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 06/26/2017] [Indexed: 11/06/2022] Open
|
32
|
Niinuma T, Kai M, Kitajima H, Yamamoto E, Harada T, Maruyama R, Nobuoka T, Nishida T, Kanda T, Hasegawa T, Tokino T, Sugai T, Shinomura Y, Nakase H, Suzuki H. Downregulation of miR-186 is associated with metastatic recurrence of gastrointestinal stromal tumors. Oncol Lett 2017; 14:5703-5710. [PMID: 29113198 PMCID: PMC5661378 DOI: 10.3892/ol.2017.6911] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 03/03/2017] [Indexed: 01/14/2023] Open
Abstract
Although dysregulation of microRNAs (miRNAs/miRs) is a common feature of human malignancies, its involvement in gastrointestinal stromal tumors (GISTs) is not fully understood. The present study aimed to identify the miRNAs that perform a role in GIST metastasis. miRNA expression profiles from a series of 32 primary GISTs were analyzed using microarrays, and miR-186 was observed to be downregulated in tumors exhibiting metastatic recurrence. Reverse transcription-quantitative polymerase chain reaction analysis of an independent cohort of 100 primary GISTs revealed that low miR-186 expression is associated with metastatic recurrence and a poor prognosis. Inhibition of miR-186 in GIST-T1 cells promoted cell migration. Gene expression microarray analysis demonstrated that miR-186 inhibition upregulated a set of genes implicated in cancer metastasis, including insulin-like growth factor-binding protein 3, AKT serine/threonine kinase 2, hepatocyte growth factor receptor, CXC chemokine receptor 4 and epidermal growth factor-containing fibulin-like extracellular matrix protein 1. These results suggest that the downregulation of miR-186 is involved in the metastatic recurrence of GISTs, and that miR-186 levels could potentially be a predictive biomarker for clinical outcome.
Collapse
Affiliation(s)
- Takeshi Niinuma
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Masahiro Kai
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Hiroshi Kitajima
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Eiichiro Yamamoto
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan.,Department of Gastroenterology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Taku Harada
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Reo Maruyama
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Takayuki Nobuoka
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Toshirou Nishida
- Department of Surgery, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Tatsuo Kanda
- Department of Surgery, Sanjo General Hospital, Sanjo 955-0055, Japan
| | - Tadashi Hasegawa
- Department of Surgical Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Takashi Tokino
- Medical Genome Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Tamotsu Sugai
- Department of Molecular Diagnostic Pathology, Iwate Medical University School of Medicine, Morioka 020-8505, Japan
| | | | - Hiroshi Nakase
- Department of Gastroenterology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Hiromu Suzuki
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| |
Collapse
|
33
|
Ketcham CM, Umezawa A, Zou H, Siegal GP. Laboratory Investigation web focus on China. J Transl Med 2016; 96:1144-1146. [PMID: 27777411 DOI: 10.1038/labinvest.2016.110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The vast growth of China's publishing output is a reflection of the increasing strength of Chinese science. The editors of Laboratory Investigation (LI) present a collection of papers that showcases research by authors from institutions across China, highlighting the significant contributions of Chinese scientists to the journal.
Collapse
Affiliation(s)
| | - Akihiro Umezawa
- Department of Reproductive Biology, National Institute for Child Health and Development, Tokyo, Japan
| | - Hejian Zou
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Gene P Siegal
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|