1
|
Tian Y, Zhou Y, Chen F, Qian S, Hu X, Zhang B, Liu Q. Research progress in MCM family: Focus on the tumor treatment resistance. Biomed Pharmacother 2024; 173:116408. [PMID: 38479176 DOI: 10.1016/j.biopha.2024.116408] [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: 12/12/2023] [Revised: 02/22/2024] [Accepted: 03/06/2024] [Indexed: 03/27/2024] Open
Abstract
Malignant tumors constitute a significant category of diseases posing a severe threat to human survival and health, thereby representing one of the most challenging and pressing issues in the field of biomedical research. Due to their malignant nature, which is characterized by a high potential for metastasis, rapid dissemination, and frequent recurrence, the prevailing approach in clinical oncology involves a comprehensive treatment strategy that combines surgery with radiotherapy, chemotherapy, targeted drug therapies, and other interventions. Treatment resistance remains a major obstacle in the comprehensive management of tumors, serving as a primary cause for the failure of integrated tumor therapies and a critical factor contributing to patient relapse and mortality. The Minichromosome Maintenance (MCM) protein family comprises functional proteins closely associated with the development of resistance in tumor therapy.The influence of MCMs manifests through various pathways, encompassing modulation of DNA replication, cell cycle regulation, and DNA damage repair mechanisms. Consequently, this leads to an enhanced tolerance of tumor cells to chemotherapy, targeted drugs, and radiation. Consequently, this review explores the specific roles of the MCM family in various cancer treatment strategies. Its objective is to enhance our comprehension of resistance mechanisms in tumor therapy, thereby presenting novel targets for clinical research aimed at overcoming resistance in cancer treatment. This bears substantial clinical relevance.
Collapse
Affiliation(s)
- Yuxuan Tian
- Department of Hepatobiliary and Intestinal Surgery of Hunan Cancer Hospital & the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; Department of Histology and Embryology, Basic School of Medicine Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of Medicine Sciences, Central South University, Changsha, Hunan 410078, PR China
| | - Fuxin Chen
- Department of Histology and Embryology, Basic School of Medicine Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Siyi Qian
- Department of Histology and Embryology, Basic School of Medicine Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Xingming Hu
- The 1st Department of Thoracic Surgery of Hunan Cancer Hospital & the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China
| | - Bin Zhang
- Department of Hepatobiliary and Intestinal Surgery of Hunan Cancer Hospital & the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; Department of Histology and Embryology, Basic School of Medicine Sciences, Central South University, Changsha, Hunan 410013, PR China.
| | - Qiang Liu
- Department of Hepatobiliary and Intestinal Surgery of Hunan Cancer Hospital & the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China.
| |
Collapse
|
2
|
Brown JS. Comparison of Oncogenes, Tumor Suppressors, and MicroRNAs Between Schizophrenia and Glioma: The Balance of Power. Neurosci Biobehav Rev 2023; 151:105206. [PMID: 37178944 DOI: 10.1016/j.neubiorev.2023.105206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023]
Abstract
The risk of cancer in schizophrenia has been controversial. Confounders of the issue are cigarette smoking in schizophrenia, and antiproliferative effects of antipsychotic medications. The author has previously suggested comparison of a specific cancer like glioma to schizophrenia might help determine a more accurate relationship between cancer and schizophrenia. To accomplish this goal, the author performed three comparisons of data; the first a comparison of conventional tumor suppressors and oncogenes between schizophrenia and cancer including glioma. This comparison determined schizophrenia has both tumor-suppressive and tumor-promoting characteristics. A second, larger comparison between brain-expressed microRNAs in schizophrenia with their expression in glioma was then performed. This identified a core carcinogenic group of miRNAs in schizophrenia offset by a larger group of tumor-suppressive miRNAs. This proposed "balance of power" between oncogenes and tumor suppressors could cause neuroinflammation. This was assessed by a third comparison between schizophrenia, glioma and inflammation in asbestos-related lung cancer and mesothelioma (ALRCM). This revealed that schizophrenia shares more oncogenic similarity to ALRCM than glioma.
Collapse
|
3
|
MCM6 Promotes Hepatocellular Carcinoma Progression via the Notch Pathway: Clinical, Functional, and Genomic Insights. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:3116303. [PMID: 35720029 PMCID: PMC9203181 DOI: 10.1155/2022/3116303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/21/2022] [Accepted: 04/30/2022] [Indexed: 11/17/2022]
Abstract
Objective To evaluate the expression profile of MCM6 in HCC and the relationship between MCM6 level and clinicopathological parameters through bioinformatics analysis of several databases. Methods MCM expression level, clinical parameters, survival data, and gene set enrichment analysis were analyzed by bioinformatics database, including Oncomine™, UALCAN, HCCDB, TCGA, cBioPortal, and LinkedOmics. Real-time PCR, western blotting, and IHC staining were conducted to identify the expression of MCM6 in HCC compared to normal liver tissues. Results Bioinformatics analysis indicated that the mRNA of MCM6 was obviously increased in multiple cancer types, especially in HCC. MCM6 level was positively associated with multiple clinical parameters (stage 3 and grades 3 and 4) and negatively associated with patient outcomes (overall survival). Moreover, enrichment of functions and signaling pathways analysis of MCM6 suggested that MCM6 might mediate DNA replication and cellular metabolism to promote the development and progression of HCC. Furthermore, IHC staining and western blotting indicated that the MCM6 was enhanced in HCC tissue, and MCM6 could promote HCC proliferation in activating Notch pathway via WB and bioinformatic analysis. Conclusion This study actually revealed the expression and related functions of MCM6 in HCC. Furthermore, MCM6 is a carcinogenic role in activating Notch pathway to promote HCC cell proliferation, which may be a new prognostic biomarker and therapeutic target for HCC patients.
Collapse
|
4
|
Zhang Z, Wang Y, Zeng L, Yu K, Wang Y, Luo Y, Liu F, Yang B, Zou Y, Wang L, Huang O. miR-218-5p in endometrial microenvironment prevents the migration of ectopic endometrial stromal cells by inhibiting LASP1. Reprod Biol Endocrinol 2022; 20:64. [PMID: 35379225 PMCID: PMC8978357 DOI: 10.1186/s12958-022-00928-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 03/13/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Our previous two-dimensional electrophoresis experiment showed that the expression of LASP1 in patients with endometriosis was significantly higher than that of control endometrium. However, the molecular mechanism by which LASP1 is regulated in endometriosis/adenomyosis is unknown. METHODS Herein, qPCR was performed to analyze the expression levels of LASP1 and miR-218-5p between endometriosis (Ems) cells and control cells. Fluorescence in situ hybridization was carried out to measure the expression level of miR-218-5p in ectopic endometrium versus normal endometrium. After miR-218-5p mimic or inhibitor were transfected, the transwell experiment was carried out to see the effect of miR-218-5p on the migration of endometrial stromal cells (ESCs). EdU was used to measure cell proliferation rate. Dual-luciferase reporter assay was used to verify the binding of hsa-miR-218-5p to the 3'UTR of LASP1. Western blot and immunofluorescence analysis were carried out to identify the protein expression pattern of LASP1 and EMT markers in endometrial tissue. RESULTS The miR-218-5p is mainly secreted from blood vessels and expressed in the muscle layer around the endometrium, which inhibits the expression level of LASP1 by binding the 3'UTR region of LASP1 in normal ESCs. Overexpression of miR-218-5p impedes the epithelial-to-mesenchymal transition (EMT) and prevents the migration of ESCs and the expression of Vimentin in Ems. CONCLUSIONS Our findings revealed that miR-218-5p in endometrial microenvironment prevents the migration of ectopic endometrial stromal cells by inhibiting LASP1.
Collapse
Affiliation(s)
- Ziyu Zhang
- Department of Pathology, Jiangxi Maternal & Child Health Hospital, Nanchang, Jiangxi, 330006, PR China
- Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, 330006, Jiangxi, China
| | - Yaoqing Wang
- Department of Reproductive Health, Jiangxi Maternal & Child Health Hospital, Nanchang, Jiangxi, 330006, PR China
| | - Liqin Zeng
- The College of Medicine, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Kaihui Yu
- The College of Medicine, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Yuanqin Wang
- The College of Medicine, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Yong Luo
- Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, 330006, Jiangxi, China
| | - Faying Liu
- Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, 330006, Jiangxi, China
| | - Bicheng Yang
- Jiangxi Provincial Key Laboratory of Birth Defect for Prevention and Control, Jiangxi Maternal & Child Health Hospital, Nanchang, Jiangxi, 330006, PR China
| | - Yang Zou
- Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, 330006, Jiangxi, China.
| | - Liqun Wang
- Department of Reproductive Health, Jiangxi Maternal & Child Health Hospital, Nanchang, Jiangxi, 330006, PR China.
| | - Ouping Huang
- Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
5
|
Spectrum of microRNAs and their target genes in cancer: intervention in diagnosis and therapy. Mol Biol Rep 2022; 49:6827-6846. [PMID: 35031927 DOI: 10.1007/s11033-021-07040-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 11/30/2021] [Indexed: 12/11/2022]
Abstract
Till date, several groups have studied the mechanism of microRNA (miRNA) biogenesis, processing, stability, silencing, and their dysregulation in cancer. The miRNA coding genes recurrently go through abnormal amplification, deletion, transcription, and epigenetic regulation in cancer. Some miRNAs function as tumor promoters while few others are tumor suppressors based on the transcriptional regulation of target genes. A review of miRNAs and their target genes in a wide range of cancers is attempted in this article, which may help in the development of new diagnostic tools and intervention therapies. The contribution of miRNAs for drug sensitivity or resistance in cancer therapy and opportunities of miRNAs in cancer prognosis or diagnosis and therapy is also presented in detail.
Collapse
|
6
|
Sakai H, Kimura H, Otsubo K, Miyazawa T, Marushima H, Kojima K, Chosokabe M, Furuya N, Koike J, Fujii K, Nishimura T, Nakamura H, Saji H. Minichromosome maintenance 2 is an independent predictor of survival in patients with lung adenocarcinoma. Mol Clin Oncol 2022; 16:22. [PMID: 34909200 PMCID: PMC8655736 DOI: 10.3892/mco.2021.2455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 10/01/2021] [Indexed: 12/30/2022] Open
Abstract
Minichromosome maintenance (MCM) protein deregulation is associated with tumor formation, progression and malignant transformation. MCM2 is frequently expressed during premalignant lung cell proliferation and is a sensitive marker for the early detection of pulmonary malignant lesions. The present study was undertaken to investigate whether MCM2 expression is of clinical and prognostic value in patients who have undergone lung adenocarcinoma resection. Between January 2009 and December 2010, 102 consecutive patients underwent complete pulmonary resection (involving lobectomy or more extensive resection) for lung adenocarcinoma at St. Marianna Medical University Hospital (Kanagawa, Japan). Among those, 73 patients, who had a final pathological diagnosis of lung adenocarcinoma measuring ≥10 mm, were enrolled in the present study. High MCM2 expression was found in 35 patients (48.0%). Univariate analysis of the overall survival (OS) revealed that pathological stage and MCM2 expression were significant prognostic factors in lung adenocarcinoma (P<0.001 and P<0.002, respectively). Univariate analysis of the recurrence-free survival (RFS), the significant prognostic factors included pathological stage, EGFR mutation status and MCM2 expression (P<0.001, P<0.034 and P<0.003, respectively). On multivariate survival analysis, high MCM2 expression and pathological stage II-III were identified as independent strong prognostic factors (OS: HR=5.084, 95% CI: 1.715-15.080, P=0.003; RFS: HR=2.761, 95% CI: 1.090-6.998, P=0.032). Therefore, the findings of the present study demonstrated that MCM2 may serve as a potential biomarker and therapeutic target for lung adenocarcinoma.
Collapse
Affiliation(s)
- Hiroki Sakai
- Department of Thoracic Surgery, St. Marianna Medical University Hospital, Kawasaki, Kanagawa 216-8511, Japan
| | - Hiroyuki Kimura
- Department of Thoracic Surgery, St. Marianna Medical University Hospital, Kawasaki, Kanagawa 216-8511, Japan
| | - Kanji Otsubo
- Department of Thoracic Surgery, St. Marianna Medical University Hospital, Kawasaki, Kanagawa 216-8511, Japan
| | - Tomoyuki Miyazawa
- Department of Thoracic Surgery, St. Marianna Medical University Hospital, Kawasaki, Kanagawa 216-8511, Japan
| | - Hideki Marushima
- Department of Thoracic Surgery, St. Marianna Medical University Hospital, Kawasaki, Kanagawa 216-8511, Japan
| | - Koji Kojima
- Department of Thoracic Surgery, St. Marianna Medical University Hospital, Kawasaki, Kanagawa 216-8511, Japan
| | - Motohiro Chosokabe
- Department of Pathology, St. Marianna Medical University Hospital, Kawasaki, Kanagawa 216-8511, Japan
| | - Naoki Furuya
- Division of Respiratory Medicine, Department of Internal Medicine, St. Marianna Medical University Hospital, Kawasaki, Kanagawa 216-8511, Japan
| | - Junki Koike
- Department of Pathology, St. Marianna Medical University Hospital, Kawasaki, Kanagawa 216-8511, Japan
| | - Kiyonaga Fujii
- Department of Translational Medicine Informatics, St. Marianna Medical University Hospital, Kawasaki, Kanagawa 216-8511, Japan
- Laboratory of Analytical Chemistry, Daiichi University of Pharmacy, Fukuoka 815-8511, Japan
| | - Toshihide Nishimura
- Department of Translational Medicine Informatics, St. Marianna Medical University Hospital, Kawasaki, Kanagawa 216-8511, Japan
| | - Haruhiko Nakamura
- Department of Thoracic Surgery, St. Marianna Medical University Hospital, Kawasaki, Kanagawa 216-8511, Japan
- Department of Translational Medicine Informatics, St. Marianna Medical University Hospital, Kawasaki, Kanagawa 216-8511, Japan
| | - Hisashi Saji
- Department of Thoracic Surgery, St. Marianna Medical University Hospital, Kawasaki, Kanagawa 216-8511, Japan
| |
Collapse
|
7
|
Role of MicroRNAs in the Development and Progression of the Four Medulloblastoma Subgroups. Cancers (Basel) 2021; 13:cancers13246323. [PMID: 34944941 PMCID: PMC8699467 DOI: 10.3390/cancers13246323] [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: 12/04/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/21/2022] Open
Abstract
Medulloblastoma is the most frequent malignant brain tumour in children. Medulloblastoma originate during the embryonic stage. They are located in the cerebellum, which is the area of the central nervous system (CNS) responsible for controlling equilibrium and coordination of movements. In 2012, medulloblastoma were divided into four subgroups based on a genome-wide analysis of RNA expression. These subgroups are named Wingless, Sonic Hedgehog, Group 3 and Group 4. Each subgroup has a different cell of origin, prognosis, and response to therapies. Wingless and Sonic Hedgehog medulloblastoma are so named based on the main mutation originating these tumours. Group 3 and Group 4 have generic names because we do not know the key mutation driving these tumours. Gene expression at the post-transcriptional level is regulated by a group of small single-stranded non-coding RNAs. These microRNA (miRNAs or miRs) play a central role in several cellular functions such as cell differentiation and, therefore, any malfunction in this regulatory system leads to a variety of disorders such as cancer. The role of miRNAs in medulloblastoma is still a topic of intense clinical research; previous studies have mostly concentrated on the clinical entity of the single disease rather than in the four molecular subgroups. In this review, we summarize the latest discoveries on miRNAs in the four medulloblastoma subgroups.
Collapse
|
8
|
Chen S, Deng X, Sheng H, Rong Y, Zheng Y, Zhang Y, Lin J. Noncoding RNAs in pediatric brain tumors: Molecular functions and pathological implications. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:417-431. [PMID: 34552822 PMCID: PMC8426460 DOI: 10.1016/j.omtn.2021.07.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Brain tumors are common solid pediatric malignancies and the main reason for cancer-related death in the pediatric setting. Recently, evidence has revealed that noncoding RNAs (ncRNAs), including microRNAs (miRNAs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs), play a critical role in brain tumor development and progression. Therefore, in this review article, we describe the functions and molecular mechanisms of ncRNAs in multiple types of cancer, including medulloblastoma, pilocytic astrocytoma, ependymoma, atypical teratoid/rhabdoid tumor, glioblastoma, diffuse intrinsic pontine glioma, and craniopharyngioma. We also mention the limitations of using ncRNAs as therapeutic targets because of the nonspecificity of ncRNA targets and the delivery methods of ncRNAs. Due to the critical role of ncRNAs in brain oncogenesis, targeting aberrantly expressed ncRNAs might be an effective strategy to improve the outcomes of pediatric patients with brain tumors.
Collapse
Affiliation(s)
- Shaohuai Chen
- Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiangyang Deng
- Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hansong Sheng
- Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuxi Rong
- Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yanhao Zheng
- Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yusong Zhang
- Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Lin
- Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
9
|
Liu Z, Wang H, Jia Y, Wang J, Wang Y, Bian L, Liu B, Lian X, Zhang B, Ren Z, Zhang W, Dai W, Gao Y. Significantly high expression of NUP37 leads to poor prognosis of glioma patients by promoting the proliferation of glioma cells. Cancer Med 2021; 10:5218-5234. [PMID: 34264013 PMCID: PMC8335818 DOI: 10.1002/cam4.3954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 01/02/2023] Open
Abstract
Background The carcinogenic effect of NUP37 has been reported recently in a variety of tumors, but its research in the field of glioma has not been paid attention. The main purpose of this study is to reveal the relationship between NUP37 and prognosis or clinical characteristics of glioma patients. Methods First, as a retrospective study, this study included thousands of tissue samples based on a variety of public databases and clinicopathological tissues. Second, a series of bioinformatics analysis methods were used to analyze the NUP37 and glioma samples from multiple databases such as the CGGA, TCGA, GEO, HPA, and GEPIA. Third, to analyze the relationship between the expression level of NUP37 in tumor tissues and cells and a variety of clinical prognostic molecular characteristics, whether it can be an independent risk factor leading to poor prognosis in glioma and whether it has clinical diagnostic value; GSEA was used to analyze the cancer‐related signaling pathways that may be activated by high expression of NUP37. Fifth, CMap was used to analyze small molecule drugs that may inhibit NUP37 expression. Finally, the meta‐analysis of thousands of tissue samples from seven datasets and cell proliferation and migration experiments confirmed that NUP37 has a malignant effect on glioma. Results NUP37 is highly expressed in glioma patient tissues and glioma cells, significantly correlates with reduced overall survival, and may serve as an independent prognostic factor with some diagnostic value. Silencing NUP37 suppresses malignant biological behaviors of glioma cells. 4 small molecule drugs that had potential targeting inhibitory effects on NUP37 overexpression. Conclusions This study demonstrates for the first time a malignant role of NUP37 in glioma and provides a vision to unravel the complex pathological mechanisms of glioma and a potentially valuable biomarker for implementing individualized diagnosis and treatment of glioma.
Collapse
Affiliation(s)
- Zhendong Liu
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Hongbo Wang
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China.,Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Yulong Jia
- Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Jialin Wang
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China.,Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Yanbiao Wang
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China.,Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Lu Bian
- Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Binfeng Liu
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China.,Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Xiaoyu Lian
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China.,Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Bo Zhang
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China.,Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhishuai Ren
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China.,Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Wang Zhang
- Department of Neurosurgery of the First Affiliate Hospital of Harbin Medical University, Harbin, China
| | - Weiwei Dai
- Xiangya Hospital Central South University, Changsha, China
| | - Yanzheng Gao
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| |
Collapse
|
10
|
Li Y, Zou J, Zhang Q, Quan F, Cao L, Zhang X, Liu J, Wu D. Systemic Analysis of the DNA Replication Regulator MCM Complex in Ovarian Cancer and Its Prognostic Value. Front Oncol 2021; 11:681261. [PMID: 34178669 PMCID: PMC8220296 DOI: 10.3389/fonc.2021.681261] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/18/2021] [Indexed: 01/11/2023] Open
Abstract
Microliposome maintenance (MCM) 2, MCM3, MCM4, MCM5, MCM6, and MCM7 are DNA replication regulators and are involved in the progression of multiple cancer types, but their role in ovarian cancer is still unclear. The purpose of this study is to clarify the biological function and prognostic value of the MCM complex in ovarian cancer (OS) progression. We analyzed DNA alterations, mRNA and protein levels, protein structure, PPI network, functional enrichment, and prognostic value in OC based on the Oncomine, cBioPortal, TCGA, CPTAC, PDB, GeneMANIA, DAVID, KEGG, and GSCALite databases. The results indicated that the protein levels of these DNA replication regulators were increased significantly. Moreover, survival analysis showed a prognostic signature based on the MCM complex, which performed moderately well in terms of OS prognostic prediction. Additionally, protein structure, functional enrichment, and PPI network analyses indicated that the MCM complex synergistically promoted OC progression by accelerating DNA replication and the cell cycle. In conclusion, our study suggested that the MCM complex might be a potential target and prognostic marker for OC patients.
Collapse
Affiliation(s)
- Yukun Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Juan Zou
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Qunfeng Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Feifei Quan
- Department of Obstetrics and Gynecology, Foshan First People's Hospital, Foshan, China
| | - Lu Cao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Xiaodi Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Jue Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Daichao Wu
- Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, University of South China, Hengyang, China.,Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD, United States
| |
Collapse
|
11
|
Zeng T, Guan Y, Li YK, Wu Q, Tang XJ, Zeng X, Ling H, Zou J. The DNA replication regulator MCM6: An emerging cancer biomarker and target. Clin Chim Acta 2021; 517:92-98. [PMID: 33609557 DOI: 10.1016/j.cca.2021.02.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 01/07/2023]
Abstract
MCM6 is a significant DNA replication regulator that plays a crucial role in sustaining the cell cycle. In many cancer cells, MCM6 expression is enhanced. For example, persistently increased expression of MCM6 promotes the formation, development and progression of hepatocellular carcinoma (HCC). Up- and down-regulation studies have indicated that MCM6 regulates cell cycle, proliferation, metastasis, immune response and the maintenance of the DNA replication system. MCM6 can also regulate downstream signaling such as MEK/ERK thus promoting carcinogenesis. Accordingly, MCM6 may represent a sensitive and specific biomarker to predict adverse progression and poor outcome. Furthermore, inhibition of MCM6 may be an effective cancer treatment. The present review summarizes the latest results on the inactivating and activating functions of MCM6, underlining its function in carcinogenesis. Further studies of the carcinogenic functions of MCM6 may provide novel insight into cancer biology and shed light on new approaches for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Tian Zeng
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China
| | - Yang Guan
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330000, PR China
| | - Yu-Kun Li
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China
| | - Qing Wu
- Department of Digestive Medical, The Affiliated Nanhua Hospital, University of South China, Hengyang 421002, PR China
| | - Xiao-Jun Tang
- Department of Spinal Surgery, The Second Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China
| | - Xin Zeng
- Department of Histology and Embryology, Chongqing Three Gorges Medical College, Wanzhou, Chongqing 404000, PR China
| | - Hui Ling
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China.
| | - Juan Zou
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
12
|
Ma H, Cao W, Ding M. MicroRNA-31 weakens cisplatin resistance of medulloblastoma cells via NF-κB and PI3K/AKT pathways. Biofactors 2020; 46:831-838. [PMID: 32027070 DOI: 10.1002/biof.1616] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/10/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Medulloblastoma (MB) is a malignant intracranial tumor. Cisplatin is a broad-spectrum antitumor drug. It is important to study the cisplatin resistance of MB cells for the treatment of MB. In this article, we preliminarily studied the cisplatin resistance of microRNA (miR)-31 and the possible mechanism in DAOY and UW228 cells, laying a theoretical foundation for clinical treatment of MB. METHODS Following anti-miR-31 and pre-miR-31 transfections, cell viability, BrdU, CyclinD1, and apoptosis levels of DAOY and UW228 cell were detected by CCK8, BrdU, and western blot. Meanwhile, migration, invasion, and western blot assay were respectively used to detect the functions of miR-31 migration and invasion. miR-31 levels were changed by cell transfection and detected by RT-qPCR. Furthermore, the related-proteins of pathways were also detected by western blot. RESULTS Anti-miR-31 increased DAOY and UW228 cells viability, BrdU+ numbers, and expression of CyclinD1. The migration/invasion rate and expression levels of MMP-9 and vimentin after anti-miR-31 transfection were increased. Furthermore, anti-miR-31 enhanced cells' cisplatin resistance and triggered PI3K/AKT and NF-κB pathways. Pre-miR-31 played opposite roles and promoted the apoptosis. CONCLUSION miR-31 regulated cell growth, migration, invasion and cisplatin resistance of MB cells via PI3K/AKT and NF-κB pathways.
Collapse
Affiliation(s)
- Hui Ma
- Department of Neurosurgery, Jining No. 1 People's Hospital, Jining, China
| | - Wei Cao
- Department of Neurosurgery, Jining No. 1 People's Hospital, Jining, China
| | - Meili Ding
- Department of Pediatrics, Jining No. 1 People's Hospital, Jining, China
| |
Collapse
|
13
|
Haltom AR, Toll SA, Cheng D, Maegawa S, Gopalakrishnan V, Khatua S. Medulloblastoma epigenetics and the path to clinical innovation. J Neurooncol 2020; 150:35-46. [PMID: 32816225 DOI: 10.1007/s11060-020-03591-9] [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: 07/01/2020] [Accepted: 08/06/2020] [Indexed: 12/30/2022]
Abstract
INTRODUCTION In the last decade, a number of genomic and pharmacological studies have demonstrated the importance of epigenetic dysregulation in medulloblastoma initiation and progression. High throughput approaches including gene expression array, next-generation sequencing (NGS), and methylation profiling have now clearly identified at least four molecular subgroups within medulloblastoma, each with distinct clinical and prognostic characteristics. These studies have clearly shown that despite the overall paucity of mutations, clinically relevant events do occur within the cellular epigenetic machinery. Thus, this review aims to provide an overview of our current understanding of the spectrum of epi-oncogenetic perturbations in medulloblastoma. METHODS Comprehensive review of epigenetic profiles of different subgroups of medulloblastoma in the context of molecular features. Epigenetic regulation is mediated mainly by DNA methylation, histone modifications and microRNAs (miRNA). Importantly, epigenetic mis-events are reversible and have immense therapeutic potential. CONCLUSION The widespread epigenetic alterations present in these tumors has generated intense interest in their use as therapeutic targets. We provide an assessment of the progress that has been made towards the development of molecular subtypes-targeted therapies and the current status of clinical trials that have leveraged these recent advances.
Collapse
Affiliation(s)
- Amanda R Haltom
- Division of Pediatrics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.,Center for Cancer Epigenetics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Stephanie A Toll
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Children's Hospital of Michigan, Detroit, USA
| | - Donghang Cheng
- Division of Pediatrics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.,Center for Cancer Epigenetics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Shinji Maegawa
- Division of Pediatrics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.,Center for Cancer Epigenetics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Vidya Gopalakrishnan
- Division of Pediatrics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA. .,Department of Molecular and Cellular Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA. .,Center for Cancer Epigenetics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA. .,Brain Tumor Center, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| | - Soumen Khatua
- Division of Pediatrics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA. .,Brain Tumor Center, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
14
|
MCM family in gastrointestinal cancer and other malignancies: From functional characterization to clinical implication. Biochim Biophys Acta Rev Cancer 2020; 1874:188415. [PMID: 32822825 DOI: 10.1016/j.bbcan.2020.188415] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/15/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023]
Abstract
Despite the recent advances in cancer research and treatment, gastrointestinal (GI) cancers remain the most common deadly disease worldwide. The aberrant DNA replication serves as a major source of genomic instability and enhances cell proliferation that contributes to tumor initiation and progression. Minichromosome maintenance family (MCMs) is a well-recognized group of proteins responsible for DNA synthesis. Recent studies suggested that dysregulated MCMs lead to tumor initiation, progression, and chemoresistance via modulating cell cycle and DNA replication stress. Their underlying mechanisms in various cancer types have been gradually identified. Furthermore, multiple studies have investigated the association between MCMs expression and clinicopathological features of cancer patients, implying that MCMs might serve as prominent prognostic biomarkers for GI cancers. This review summarizes the current knowledge on the oncogenic role of MCM proteins and highlights their clinical implications in various malignancies, especially in GI cancers. Targeting MCMs might shed light on the potential for identifying novel therapeutic strategies.
Collapse
|
15
|
MiR-30b-5p inhibits proliferation and promotes apoptosis of medulloblastoma cells via targeting MYB proto-oncogene like 2 (MYBL2). J Investig Med 2020; 68:1179-1185. [DOI: 10.1136/jim-2020-001354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2020] [Indexed: 12/15/2022]
Abstract
Medulloblastoma (MB) is the most common malignant brain tumors among children. MiR-30b-5p is a potential tumor suppressor in a variety of human cancers. However, its expression and function in MB remain poorly understood. This study aimed to investigate the expression, role and regulatory mechanism of miR-30b-5p in MB. The expression of miR-30b-5p in MB tissues and cell lines was detected by real-time PCR. The effects of miR-30b-5p on cell proliferation and apoptosis were monitored by CCK-8 (Cell Counting Kit-8) assay, colony formation assay and flow cytometry, respectively. Bioinformatics database TargetScan predicted the target genes of miR-30b-5p. The interaction between miR-30b-5p and MYB proto-oncogene Like 2 (MYBL2) was determined by luciferase reporter gene assay. We demonstrated that the expression of miR-30b-5p was significantly downregulated in MB. Upregulated miR-30b-5p could inhibit the proliferation and induce apoptosis of MB.Moreover, overexpressed miR-30b-5p could increase the expression of BAX but decrease that of Bcl-2. Downregulated miR-30b-5p exerted the opposite effect. MYBL2 was proved to be the target gene of miR-30b-5p and was negatively regulated by miR-30b-5p. These results indicate that miR-30b-5p inhibits the progression of MB via targeting the expression of MYBL2.
Collapse
|
16
|
Laneve P, Caffarelli E. The Non-coding Side of Medulloblastoma. Front Cell Dev Biol 2020; 8:275. [PMID: 32528946 PMCID: PMC7266940 DOI: 10.3389/fcell.2020.00275] [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: 02/14/2020] [Accepted: 03/31/2020] [Indexed: 12/18/2022] Open
Abstract
Medulloblastoma (MB) is the most common pediatric brain tumor and a primary cause of cancer-related death in children. Until a few years ago, only clinical and histological features were exploited for MB pathological classification and outcome prognosis. In the past decade, the advancement of high-throughput molecular analyses that integrate genetic, epigenetic, and expression data, together with the availability of increasing wealth of patient samples, revealed the existence of four molecularly distinct MB subgroups. Their further classification into 12 subtypes not only reduced the well-characterized intertumoral heterogeneity, but also provided new opportunities for the design of targets for precision oncology. Moreover, the identification of tumorigenic and self-renewing subpopulations of cancer stem cells in MB has increased our knowledge of its biology. Despite these advancements, the origin of MB is still debated, and its molecular bases are poorly characterized. A major goal in the field is to identify the key genes that drive tumor growth and the mechanisms through which they are able to promote tumorigenesis. So far, only protein-coding genes acting as oncogenic drivers have been characterized in each MB subgroup. The contribution of the non-coding side of the genome, which produces a plethora of transcripts that control fundamental biological processes, as the cell choice between proliferation and differentiation, is still unappreciated. This review wants to fill this major gap by summarizing the recent findings on the impact of non-coding RNAs in MB initiation and progression. Furthermore, their potential role as specific MB biomarkers and novel therapeutic targets is also highlighted.
Collapse
Affiliation(s)
- Pietro Laneve
- Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy
| | - Elisa Caffarelli
- Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy
| |
Collapse
|
17
|
Liu Z, Zhang R, Sun Z, Yao J, Yao P, Chen X, Wang X, Gao M, Wan J, Du Y, Zhao S. Identification of hub genes and small-molecule compounds in medulloblastoma by integrated bioinformatic analyses. PeerJ 2020; 8:e8670. [PMID: 32328342 PMCID: PMC7164431 DOI: 10.7717/peerj.8670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/30/2020] [Indexed: 01/03/2023] Open
Abstract
Background Medulloblastoma (MB) is the most common intracranial malignant tumor in children. The genes and pathways involved in the pathogenesis of MB are relatively unknown. We aimed to identify potential biomarkers and small-molecule drugs for MB. Methods Gene expression profile data sets were obtained from the Gene Expression Omnibus (GEO) database and the differentially expressed genes (DEGs) were identified using the Limma package in R. Functional annotation, and cell signaling pathway analysis of DEGs was carried out using DAVID and Kobas. A protein-protein interaction network was generated using STRING. Potential small-molecule drugs were identified using CMap. Result We identified 104 DEGs (29 upregulated; 75 downregulated). Gene ontology analysis showed enrichment in the mitotic cell cycle, cell cycle, spindle, and DNA binding. Cell signaling pathway analysis identified cell cycle, HIF-1 signaling pathway, and phospholipase D signaling pathway as key pathways. SYN1, CNTN2, FAIM2, MT3, and SH3GL2 were the prominent hub genes and their expression level were verified by RT-qPCR. Vorinostat, resveratrol, trichostatin A, pyrvinium, and prochlorperazine were identified as potential drugs for MB. The five hub genes may be targets for diagnosis and treatment of MB, and the small-molecule compounds are promising drugs for effective treatment of MB. Conclusion In this study we obtained five hub genes of MB, SYN1, CNTN2, FAIM2, MT3, and SH3GL2 were confirmed as hub genes. Meanwhile, Vorinostat, resveratrol, trichostatin A, pyrvinium, and prochlorperazine were identified as potential drugs for MB.
Collapse
Affiliation(s)
- Zhendong Liu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Ruotian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Zhenying Sun
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Jiawei Yao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Penglei Yao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Xin Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Xinzhuang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Ming Gao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Jinzhao Wan
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Yiming Du
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Shiguang Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| |
Collapse
|
18
|
Abstract
Even though the treatment of childhood cancer has evolved significantly in recent decades, aggressive central nervous system (CNS) tumors are still a leading cause of morbidity and mortality in this population. Consequently, the identification of molecular targets that can be incorporated into diagnostic practice, effectively predict prognosis, follow treatment response, and materialize into potential targeted therapeutic approaches are still warranted. Since the first evidence of the participation of miRNAs in cancer development and progression 20 years ago, notable progress has been made in the basic understanding of the contribution of their dysregulation as epigenetic driver of tumorigenesis. Nevertheless, among the plethora of articles in the literature, microRNA profiling of pediatric tumors are scarce. This article gives an overview of the recent advances in the diagnostic/prognostic potential of miRNAs in a selection of pediatric CNS tumors: medulloblastoma, ependymoma, pilocytic astrocytoma, glioblastoma, diffuse intrinsic pontine glioma, atypical teratoid/rhabdoid tumors, and choroid plexus tumors.
Collapse
|
19
|
Ashrafizadeh M, Ahmadi Z, Mohammadinejad R, Farkhondeh T, Samarghandian S. MicroRNAs mediate the anti-tumor and protective effects of ginsenosides. Nutr Cancer 2019; 72:1264-1275. [PMID: 31608663 DOI: 10.1080/01635581.2019.1675722] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
MicroRNAs (miRs(, as short non-coding RNAs, regulate important biological processes and mainly are associated with regulation of gene expression. The miRs are beneficial targets for diagnosis of various disorders, particularly cancer, since their expression profile undergoes alterations in pathological conditions. The numerous drugs have been designed with the capability of targeting miRs for treating pathological conditions. On the other hand, the application of naturally occurring compounds has been increased due to their minimal side effects and valuable biological and therapeutic activities. Ginsenosides are able to act as anti-tumor agents via either increasing or decreasing the expression level of miRs. Ginsenosides affect the expression profile of miRNAs to induce their protective impacts. Angiogenesis as a key factor in the progression of cancer can be suppressed by ginsenosides which is mediated by miR regulation. The aim of this review is to shed some light on the protective and anti-tumor activities of ginsenosides mediated by miRNAs.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Veterinary Medicine, Department of Basic Science, University of Tabriz, Tabriz, Iran
| | - Zahra Ahmadi
- Department of Basic Science, Shoushtar Branch, Islamic Azad University, Shoushtar, Iran
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Science, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
20
|
Kaur G, Balasubramaniam SD, Lee YJ, Balakrishnan V, Oon CE. Minichromosome Maintenance Complex (MCM) Genes Profiling and MCM2 Protein Expression in Cervical Cancer Development. Asian Pac J Cancer Prev 2019; 20:3043-3049. [PMID: 31653153 PMCID: PMC6982666 DOI: 10.31557/apjcp.2019.20.10.3043] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Minichromosome maintenance complex (MCM) proteins are essential for the process of DNA replication and cell division. This study aimed to evaluate MCM genes expression profiles and MCM2 protein in HPV-associated cervical carcinogenesis. METHODOLOGY MCM2, 4, 5 and 7 genes expression profiles were evaluated in three cervical tissue samples each of normal cervix, human papillomavirus (HPV)-infected low grade squamous intraepithelial lesion (LSIL), high grade squamous intraepithelial lesion (HSIL) and squamous cell carcinoma (SCC), using Human Transcriptome Array 2.0 and validated by nCounter® PanCancer Pathway NanoString Array. Immunohistochemical expression of MCM2 protein was semi-quantitatively assessed by histoscore in tissue microarrays containing 9 cases of normal cervix, 10 LSIL, 10 HSIL and 42 cases of SCC. RESULTS MCM2, 4, 5 and 7 genes expressions were upregulated with increasing fold change during the progression from LSIL to HSIL and the highest in SCC. MCM2 gene had the highest fold change in SCC compared to normal cervix. Immunohistochemically, MCM2 protein was localised in the nuclei of basal cells of normal cervical epithelium and dysplastic-neoplastic cells of CIN and SCC. There was a significant difference in MCM2 protein expression between the histological groups (P = 0.039), and histoscore was the highest in HSIL compared to normal cervix (P = 0.010). CONCLUSION The upregulation of MCM genes expressions in cervical carcinogenesis reaffirms MCM as a proliferative marker in DNA replication pathway, whereby proliferation of dysplastic and cancer cells become increasingly dysregulated and uncontrolled. A strong expression of MCM2 protein in HSIL may aid as a concatenated screening tool in detecting pre-cancerous cervical lesions.
Collapse
Affiliation(s)
- Gurjeet Kaur
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | | | - Yung Jen Lee
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Venugopal Balakrishnan
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| |
Collapse
|
21
|
Li J, Lv H, Che YQ. Upregulated microRNA-31 inhibits oxidative stress-induced neuronal injury through the JAK/STAT3 pathway by binding to PKD1 in mice with ischemic stroke. J Cell Physiol 2019; 235:2414-2428. [PMID: 31517390 DOI: 10.1002/jcp.29146] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/26/2019] [Indexed: 12/21/2022]
Abstract
Ischemic stroke (IS), which is characterized by high morbidity, disability, and mortality, is recognized as a major cerebrovascular disease. MicroRNA-31 (miR-31) was reported to participate in the progression of brain disease. The present study was conducted in order to investigate the effect of miR-31 on oxidative stress-induced neuronal injury in IS mice with the involvement of protein kinase D1 (PKD1) and the JAK/STAT3 pathway. C57BL/6J mice were used to establish the middle cerebral artery occlusion (MCAO) model. Astrocytes were transfected with miR-31 mimic, miR-31 inhibitor, si-PKD1, or JAK-STAT3 pathway inhibitor. Following the establishment of an oxygen-glucose deprivation (OGD) model, the astrocytes were cocultured with neuronal OGD. Lower miR-31, higher PKD1 expressions, and activated JAK/STAT3 pathway were found in both the MCAO and OGD models. miR-31 could negatively target PKD1. In an MCAO model, overexpressing miR-31 and silencing PKD1 reduced neuronal injury, cerebral infarct volume, neuron loss, and oxidative stress injury, inhibited the activation of JAK/STAT3 pathway and the expressions of PKD1, interleukin (IL)-1β, IL-6, tumor necrosis factor-α, malondialdehyde, 4-HNE, 8-HOdG, caspase-3, and Bax, but increased the superoxide dismutase content. In the OGD model, overexpression of miR-31 and silencing of PKD1 attenuated oxidative stress-induced neuronal injury, and diminished the lactate dehydrogenase leakage and reactive oxygen species level, accompanied by elevated neuronal viability. These results indicate that miR-31 alleviates inflammatory response as well as an oxidative stress-induced neuronal injury in IS mice by downregulating PKD1 and JAK/STAT3 pathway.
Collapse
Affiliation(s)
- Jie Li
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Hui Lv
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yu-Qin Che
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
22
|
Involvement of Dual Strands of miR-143 ( miR-143-5p and miR-143-3p) and Their Target Oncogenes in the Molecular Pathogenesis of Lung Adenocarcinoma. Int J Mol Sci 2019; 20:ijms20184482. [PMID: 31514295 PMCID: PMC6770575 DOI: 10.3390/ijms20184482] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/02/2019] [Accepted: 09/09/2019] [Indexed: 12/24/2022] Open
Abstract
Our analyses of tumor-suppressive microRNAs (miRNAs) and their target oncogenes have identified novel molecular networks in lung adenocarcinoma (LUAD). Moreover, our recent studies revealed that some passenger strands of miRNAs contribute to cancer cell malignant transformation. Downregulation of both strands of the miR-143 duplex was observed in LUAD clinical specimens. Ectopic expression of these miRNAs suppressed malignant phenotypes in cancer cells, suggesting that these miRNAs have tumor-suppressive activities in LUAD cells. Here, we evaluated miR-143-5p molecular networks in LUAD using genome-wide gene expression and miRNA database analyses. Twenty-two genes were identified as potential miR-143-5p-controlled genes in LUAD cells. Interestingly, the expression of 11 genes (MCM4, RAD51, FAM111B, CLGN, KRT80, GPC1, MTL5, NETO2, FANCA, MTFR1, and TTLL12) was a prognostic factor for the patients with LUAD. Furthermore, knockdown assays using siRNAs showed that downregulation of MCM4 suppressed cell growth, migration, and invasion in LUAD cells. Aberrant expression of MCM4 was confirmed in the clinical specimens of LUAD. Thus, we showed that miR-143-5p and its target genes were involved in the molecular pathogenesis of LUAD. Identification of tumor-suppressive miRNAs and their target oncogenes may be an effective strategy for elucidation of the molecular oncogenic networks of this disease.
Collapse
|
23
|
Joshi P, Katsushima K, Zhou R, Meoded A, Stapleton S, Jallo G, Raabe E, Eberhart CG, Perera RJ. The therapeutic and diagnostic potential of regulatory noncoding RNAs in medulloblastoma. Neurooncol Adv 2019; 1:vdz023. [PMID: 31763623 PMCID: PMC6859950 DOI: 10.1093/noajnl/vdz023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Medulloblastoma, a central nervous system tumor that predominantly affects children, always requires aggressive therapy. Nevertheless, it frequently recurs as resistant disease and is associated with high morbidity and mortality. While recent efforts to subclassify medulloblastoma based on molecular features have advanced our basic understanding of medulloblastoma pathogenesis, optimal targets to increase therapeutic efficacy and reduce side effects remain largely undefined. Noncoding RNAs (ncRNAs) with known regulatory roles, particularly long noncoding RNAs (lncRNAs) and microRNAs (miRNAs), are now known to participate in medulloblastoma biology, although their functional significance remains obscure in many cases. Here we review the literature on regulatory ncRNAs in medulloblastoma. In providing a comprehensive overview of ncRNA studies, we highlight how different lncRNAs and miRNAs have oncogenic or tumor suppressive roles in medulloblastoma. These ncRNAs possess subgroup specificity that can be exploited to personalize therapy by acting as theranostic targets. Several of the already identified ncRNAs appear specific to medulloblastoma stem cells, the most difficult-to-treat component of the tumor that drives metastasis and acquired resistance, thereby providing opportunities for therapy in relapsing, disseminating, and therapy-resistant disease. Delivering ncRNAs to tumors remains challenging, but this limitation is gradually being overcome through the use of advanced technologies such as nanotechnology and rational biomaterial design.
Collapse
Affiliation(s)
- Piyush Joshi
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland.,Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St. Petersburg, Florida
| | - Keisuke Katsushima
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland.,Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St. Petersburg, Florida
| | - Rui Zhou
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St. Petersburg, Florida
| | - Avner Meoded
- Pediatric Neuroradiology, Johns Hopkins All Children's Hospital, St. Petersburg, Florida
| | - Stacie Stapleton
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St. Petersburg, Florida
| | - George Jallo
- Institute Brain Protection Sciences, Johns Hopkins All Children's Hospital, St. Petersburg, Florida
| | - Eric Raabe
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Charles G Eberhart
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ranjan J Perera
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland.,Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St. Petersburg, Florida.,Sanford Burnham Prebys Medical Discovery Institute, NCI-Designated Cancer Center, La Jolla, California
| |
Collapse
|
24
|
Li Y, Zhang Q, Liu F, Zhang Z, Zou Y, Yang B, Luo Y, Wang L, Huang O. Inhibition of formin like 2 promotes the transition of ectopic endometrial stromal cells to epithelial cells in adenomyosis through a MET-like process. Gene 2019; 710:186-192. [PMID: 31175924 DOI: 10.1016/j.gene.2019.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/10/2019] [Accepted: 06/04/2019] [Indexed: 12/11/2022]
Abstract
EMT (Epithelial-Mesenchymal Transition) is one of the factors in the pathogenesis of adenomyosis. FMNL2 induced invasion of cancer cell through promoting EMT, but it is unclear the role of FMNL2 in the adenomyosis. By IHC staining, we found the expression level of FMNL2 was significantly higher in the ectopic endometrial stromal cells from women with adenomyosis when compared with normal endometrial stromal cells. Knockdown of FMNL2 inhibited the invasion and migration of ectopic endometrial stromal cells and promoted the protein levels of E-cadherin and Vimentin. Meanwhile, inhibition of FMNL2 could induce the cell membrane localization of E-cadherin. Our findings reveal that the aberrant activation of FMNL2 promotes the pathogenesis of adenomyosis through inducing the EMT process. On the contrary, inhibition of FMNL2 promotes the transition of ectopic endometrial stromal cells to epithelial cells in adenomyosis through a MET-like process.
Collapse
Affiliation(s)
- You Li
- The College of Medicine, Nanchang University, Nanchang, Jiangxi 330006, PR China; Reproductive Medicine Center, Jiangxi Maternal & Child Health Hospital, Nanchang, Jiangxi 330006, PR China
| | - Quan Zhang
- The College of Medicine, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Faying Liu
- Key Laboratory of Women's Reproductive Health of Jiangxi, Jiangxi Maternal & Child Health Hospital, Nanchang, Jiangxi 330006, PR China
| | - Ziyu Zhang
- Key Laboratory of Women's Reproductive Health of Jiangxi, Jiangxi Maternal & Child Health Hospital, Nanchang, Jiangxi 330006, PR China
| | - Yang Zou
- Key Laboratory of Women's Reproductive Health of Jiangxi, Jiangxi Maternal & Child Health Hospital, Nanchang, Jiangxi 330006, PR China
| | - Bicheng Yang
- Key Laboratory of Women's Reproductive Health of Jiangxi, Jiangxi Maternal & Child Health Hospital, Nanchang, Jiangxi 330006, PR China
| | - Yong Luo
- Key Laboratory of Women's Reproductive Health of Jiangxi, Jiangxi Maternal & Child Health Hospital, Nanchang, Jiangxi 330006, PR China
| | - Liqun Wang
- Division of Obstetrics, Jiangxi Maternal & Child Health Hospital, Nanchang, Jiangxi 330006, PR China.
| | - Ouping Huang
- Key Laboratory of Women's Reproductive Health of Jiangxi, Jiangxi Maternal & Child Health Hospital, Nanchang, Jiangxi 330006, PR China.
| |
Collapse
|
25
|
Wang X, Holgado BL, Ramaswamy V, Mack S, Zayne K, Remke M, Wu X, Garzia L, Daniels C, Kenney AM, Taylor MD. miR miR on the wall, who's the most malignant medulloblastoma miR of them all? Neuro Oncol 2019; 20:313-323. [PMID: 28575493 DOI: 10.1093/neuonc/nox106] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
microRNAs (miRNAs) have wide-ranging effects on large-scale gene regulation. As such, they play a vital role in dictating normal development, and their aberrant expression has been implicated in cancer. There has been a large body of research on the role of miRNAs in medulloblastoma, the most common malignant brain tumor of childhood. The identification of the 4 molecular subgroups with distinct biological, genetic, and transcriptional features has revolutionized the field of medulloblastoma research over the past 5 years. Despite this, the growing body of research on miRNAs in medulloblastoma has largely focused on the clinical entity of a single disease rather than the molecular subgroups. This review begins by highlighting the role of miRNAs in development and progresses to explore their myriad of implications in cancer. Medulloblastoma is characterized by increased proliferation, inhibition of apoptosis, and maintenance of stemness programs-features that are inadvertently regulated by altered expression patterns in miRNAs. This review aims to contextualize the large body of work on miRNAs within the framework of medulloblastoma subgroups. The goal of this review is to stimulate new areas of research, including potential therapeutics, within a rapidly growing field.
Collapse
Affiliation(s)
- Xin Wang
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Borja L Holgado
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Vijay Ramaswamy
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.,Department of Haematology & Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stephen Mack
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Kory Zayne
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Marc Remke
- German Cancer Consortium, University of Düsseldorf, Düsseldorf, Germany
| | - Xiaochong Wu
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Livia Garzia
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Craig Daniels
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Anna M Kenney
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.,Department of Pediatric Oncology, Emory University, Atlanta, Georgia, USA.,Winship Cancer Institute, Atlanta, Georgia, USA
| | - Michael D Taylor
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Division of Neurosurgery, Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Mahadevappa R, Neves H, Yuen SM, Jameel M, Bai Y, Yuen HF, Zhang SD, Zhu Y, Lin Y, Kwok HF. DNA Replication Licensing Protein MCM10 Promotes Tumor Progression and Is a Novel Prognostic Biomarker and Potential Therapeutic Target in Breast Cancer. Cancers (Basel) 2018; 10:cancers10090282. [PMID: 30135378 PMCID: PMC6162382 DOI: 10.3390/cancers10090282] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/17/2018] [Accepted: 08/20/2018] [Indexed: 12/20/2022] Open
Abstract
Breast cancer is one of the most common malignancies in women worldwide. In breast cancer, the cell proliferation rate is known to influence the cancer malignancy. Recent studies have shown that DNA replication initiation/licensing factors are involved in cancer cell proliferation as well as cancer cell migration and invasion. Licensing factors have also been reported as important prognostic markers in lung, prostrate, and bladder cancers. Here, we studied the role of MCM10, a novel licensing factor, in breast cancer progression. From the public database, NCBI, we investigated six independent breast cancer patient cohorts, totaling 1283 patients. We observed a significant association between high MCM10 mRNA expression with tumor grading and patients’ survival time. Most importantly, using breast cancer cohorts with available treatment information, we also demonstrated that a high level of MCM10 is associated with a better response to conventional treatment. Similarly, in in vitro studies, the expression level of MCM10 in breast cancer cell lines is significantly higher compared to paired normal breast epithelium cells. Knockdown of MCM10 expression in the cancer cell line showed significantly decreased tumorigenic properties such as cell proliferation, migration and anchorage independence. The MCF7 breast cancer cell line, after MCM10 expression knockdown, showed significantly decreased tumorigenic properties such as cell proliferation, migration, and anchorage independent growth. Mechanistically, MCM10 expression is observed to be regulated by an Estrogen Receptor (ER) signaling pathway, where its expression is suppressed by the inhibition of the ER or serum withdrawal. Our results suggest that MCM10 plays an important role in breast cancer progression and is a potential prognostic/predictive biomarker and therapeutic target for breast cancer patients.
Collapse
Affiliation(s)
- Ravikiran Mahadevappa
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau, China.
| | - Henrique Neves
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau, China.
| | - Shun Ming Yuen
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau, China.
| | - Muhammad Jameel
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau, China.
| | - Yuchen Bai
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau, China.
| | - Hiu-Fung Yuen
- Institute of Molecular and Cell Biology, A*STAR, Singapore 138673, Singapore.
| | - Shu-Dong Zhang
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, Ulster University, Londonderry BT47 6SB, UK.
| | - Youzhi Zhu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China.
| | - Yao Lin
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China.
| | - Hang Fai Kwok
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau, China.
| |
Collapse
|
27
|
Wu W, Wang X, Shan C, Li Y, Li F. Minichromosome maintenance protein 2 correlates with the malignant status and regulates proliferation and cell cycle in lung squamous cell carcinoma. Onco Targets Ther 2018; 11:5025-5034. [PMID: 30174440 PMCID: PMC6109654 DOI: 10.2147/ott.s169002] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Minichromosome maintenance protein 2 (MCM2), which is a member of MCM family, has been found to be a relevant marker for progression and prognosis in a variety of human cancers. Due to lack of effective therapeutic target in lung squamous cell carcinoma (LUSC) patients, the aim of our study was to screen and identify biomarkers which are associated to clinicopathological characteristics including prognosis in LUSC patients. Methods The expression status of MCM2 in lung cancer was analyzed using the publicly available Gene Expression Omnibus databases (GSE3268 and GSE10245). The mRNA and protein expression of MCM2 in lung cancer tissues and cell lines was detected by quantitative real-time PCR and Western blot, and the association between MCM2 expression and clinicopathological factors was analyzed by immunohistochemistry. The loss-of-function study of MCM2 was conducted in LUSC cell lines. Results In our study, we found MCM2 expression was increased in LUSC tissues compared with paired adjacent normal lung tissues or lung adenocarcinoma tissues through analyzing microarray data sets (GSE3268 and GSE10245), which confirmed that MCM2 mRNA and protein were overexpressed in LUSC tissues and cell lines. Meanwhile, we analyzed the association between MCM2 protein expression and clinicopathological characteristics of LUSC patients, and found high expression of MCM2 protein was obviously associated with malign differentiated degree, advanced clinical stage, large tumor size, more lymph node metastasis and present distant metastasis. The survival analysis showed MCM2 overexpression was an independent unfavorable prognostic factor for LUSC patients. Conclusion MCM2 is involved in the development and progression of LUSC as an oncogene, and thereby may act as a potential therapeutic target for LUSC patients.
Collapse
Affiliation(s)
- Wei Wu
- Department of Respiratory Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, People's Republic of China,
| | - Xianwei Wang
- Department of Respiratory Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, People's Republic of China,
| | - Changting Shan
- Department of Respiratory Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, People's Republic of China,
| | - Yong Li
- Department of Emergency, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, People's Republic of China
| | - Fengzhu Li
- Department of Paediatric Surgery, Jining No 1 People's Hospital, Jining, Shandong 272011, People's Republic of China
| |
Collapse
|
28
|
Wang F, Zhang L, Bai X, Cao X, Jiao X, Huang Y, Li Y, Qin Y, Wen Y. Stimuli-Responsive Nanocarrier for Co-delivery of MiR-31 and Doxorubicin To Suppress High MtEF4 Cancer. ACS APPLIED MATERIALS & INTERFACES 2018; 10:22767-22775. [PMID: 29897733 DOI: 10.1021/acsami.8b07698] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Gene interference-based therapeutics represent a fascinating challenge and show enormous potential for cancer treatment, in which microRNA is used to correct abnormal gene. On the basis of the above, we introduced microRNA-31 to bind to 3'-untranslated region of mtEF4, resulting in the downregulation of its messenger RNA and protein to trigger cancer cells apoptosis through mitochondria-related pathway. To achieve better therapeutic effect, a mesoporous silica nanoparticle-based controlled nanoplatform had been developed. This system was fabricated by conjugation of microRNA-31 onto doxorubicin-loaded mesoporous silica nanoparticles with a poly(ethyleneimine)/hyaluronic acid coating, and drug release was triggered by acidic environment of tumors. By feat of surface functionalization and tumor-specific conjugation to nanoparticles, our drug delivery system could promote intracellular accumulation of drugs via the active transport at tumor site. More importantly, microRNA-31 not only directly targeted to mtEF4 to promote cell's death, but had synergistic effects when used in combination with doxorubicin, and achieved excellent superadditive effects. As such, our research might provide new insights toward detecting high mtEF4 cancer and exploiting highly effective anticancer drugs.
Collapse
Affiliation(s)
- Fang Wang
- Research Center for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering , University of Science and Technology Beijing , Beijing 100083 , China
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics , Chinese Academy of Sciences , 15 Datun Road , Chaoyang District, Beijing 100101 , China
| | - Lingyun Zhang
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics , Chinese Academy of Sciences , 15 Datun Road , Chaoyang District, Beijing 100101 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Xiufeng Bai
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics , Chinese Academy of Sciences , 15 Datun Road , Chaoyang District, Beijing 100101 , China
| | - Xintao Cao
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics , Chinese Academy of Sciences , 15 Datun Road , Chaoyang District, Beijing 100101 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Xiangyu Jiao
- Research Center for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering , University of Science and Technology Beijing , Beijing 100083 , China
| | - Yan Huang
- Research Center for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering , University of Science and Technology Beijing , Beijing 100083 , China
| | - Yansheng Li
- Research Center for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering , University of Science and Technology Beijing , Beijing 100083 , China
| | - Yan Qin
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics , Chinese Academy of Sciences , 15 Datun Road , Chaoyang District, Beijing 100101 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Yongqiang Wen
- Research Center for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering , University of Science and Technology Beijing , Beijing 100083 , China
| |
Collapse
|
29
|
Chen Y, Shang H, Zhang S, Zhang X. Retracted
: Ginsenoside Rh2 inhibits proliferation and migration of medulloblastoma Daoy by down‐regulation of microRNA‐31. J Cell Biochem 2018; 119:6527-6534. [DOI: 10.1002/jcb.26716] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/24/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Yan Chen
- Department of PediatricsJining No. 1 People's HospitalJiningShandongChina
| | - Hong Shang
- Department of PediatricsJining No. 1 People's HospitalJiningShandongChina
| | - Shunli Zhang
- Department of PediatricsJining No. 1 People's HospitalJiningShandongChina
| | - Xiaohong Zhang
- Department of PediatricsJining No. 1 People's HospitalJiningShandongChina
| |
Collapse
|
30
|
Cai P, Li H, Huo W, Zhu H, Xu C, Zang R, Lv W, Xia Y, Tang W. Aberrant expression of LncRNA-MIR31HG regulates cell migration and proliferation by affecting miR-31 and miR-31* in Hirschsprung's disease. J Cell Biochem 2018; 119:8195-8203. [PMID: 29626357 DOI: 10.1002/jcb.26830] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 02/28/2018] [Indexed: 01/17/2023]
Abstract
Hirschsprung's disease (HSCR) is a birth defect that causes a failure of the enteric nervous system to cover the distal gut during early embryonic development. Evidence shows that long non-coding RNAs (lncRNA) play important roles in HSCR. The MIR31 host gene (MIR31HG), also known as Loc554202, is a long non-coding RNA (lncRNA), which acts as the host gene of (microRNA) miR-31 and miR-31*. There have been no studies regarding its function in early developmental defects during pregnancy, and its downstream genetic receptors. We report that downregulation of MIR31HG inhibited migration and proliferation in 293T and SH-SY5Y cell lines, by suppressing miR-31 and miR-31*. Moreover, the downregulation of miR-31 and miR-31* enhanced inter-α-trypsin inhibitor heavy chain 5 (ITIH5) and the phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic gamma subunit (PIK3CG), respectively with reductions of cell migration and proliferation in 293T and SH-SY5Y cell lines. In addition, synergistic actions were observed between miR-31 and miR-31* in cell migration and proliferation. Our results demonstrated that the MIR31HG-miR-31/31*-ITIH5/PIK3CG pathway plays a role in the pathogenesis of HSCR.
Collapse
Affiliation(s)
- Peng Cai
- Children's Hospital of Soochow University, Suzhou, P.R. China
| | - Hongxing Li
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, P.R. China.,Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, P.R. China
| | - Weiwei Huo
- Department of Gynecology, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, P.R. China
| | - Hairong Zhu
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, Shanghai, P.R. China
| | - Chao Xu
- Huai'an First People's Hospital, Nanjing Medical University, Huai'an, P.R. China
| | - Rujin Zang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, P.R. China.,Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, P.R. China
| | - Wei Lv
- School of Business, Nanjing University, Nanjing, P.R. China
| | - Yankai Xia
- Children's Hospital of Soochow University, Suzhou, P.R. China.,Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, P.R. China
| | - Weibing Tang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, P.R. China.,Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, P.R. China
| |
Collapse
|
31
|
MCM10 overexpression implicates adverse prognosis in urothelial carcinoma. Oncotarget 2018; 7:77777-77792. [PMID: 27780919 PMCID: PMC5363620 DOI: 10.18632/oncotarget.12795] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 10/12/2016] [Indexed: 12/12/2022] Open
Abstract
Urothelial carcinoma (UC) occurs in the upper urinary tract (UTUC) and the urinary bladder (UBUC). The molecular pathogenesis of UC has not been fully elucidated. Through data mining of a published transcriptome of UBUC (GSE31684), we identified Minichromosome Maintenance Complex Component 2 (MCM2) and MCM10 as the two most significantly upregulated genes in UC progression among the MCM gene family, the key factors for the initiation of DNA replication. To validate the clinical significance of MCM2 and MCM10, immunohistochemistry, evaluated by H-score, was used in a pilot study of 50 UTUC and 50 UBUC samples. Only a high expression level of MCM10 predicted worse disease-specific survival (DSS) and inferior metastasis-free survival (MeFS) for both UTUC and UBUC. Correspondingly, evaluation of MCM10 mRNA expression in 36 UTUCs and 30 UBUCs showed significantly upregulated levels in high stage UC, suggesting its role in tumor progression. Evaluation of 340 UTUC and 296 UBUC tissue samples, respectively, demonstrated that high MCM10 immunoexpression was significantly associated with advanced primary tumors, nodal status, and the presence of vascular invasion in both groups of UCs. In multivariate Cox regression analyses, adjusted for standard clinicopathological features, MCM10 overexpression was independently associated with DSS (UTUC hazard ratio [HR]=2.401, P = 0.013; UBUC HR=4.323, P=0.001) and with MeFS (UTUC HR=3.294, P<0.001; UBUC HR=1.972, P=0.015). In vitro, knockdown of MCM10 gene significantly suppressed cell proliferation in both J82 and TCCSUP cells. In conclusion, MCM10 overexpression was associated with unfavorable clinicopathological characteristics and independent negative prognostic effects, justifying its potential theranostic value in UC.
Collapse
|
32
|
Liu Z, Li J, Chen J, Shan Q, Dai H, Xie H, Zhou L, Xu X, Zheng S. MCM family in HCC: MCM6 indicates adverse tumor features and poor outcomes and promotes S/G2 cell cycle progression. BMC Cancer 2018; 18:200. [PMID: 29463213 PMCID: PMC5819696 DOI: 10.1186/s12885-018-4056-8] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 01/25/2018] [Indexed: 11/23/2022] Open
Abstract
Background Minichromosome Maintenance family (MCMs), as replication licensing factors, is involved in the pathogenesis of tumors. Here, we investigated the expression of MCMs and their values in hepatocellular carcinoma (HCC). Methods MCMs were analyzed in 105 samples including normal livers (n = 15), cirrhotic livers (n = 40), HCC (n = 50) using quantitative polymerase chain reaction (qPCR) (Cohort 1). Significantly up-regulated MCMs were verified in 102 HCC and matched peritumoral livers using PCR (Cohort 2), and the correlations with clinical features and outcomes were determined. In addition, the focused MCMs were analyzed in parallel immunohistochemistry of 345 samples on spectrum of hepatocarcinogenesis (Cohort 3) and queried for the potential specific role in cell cycle. Results MCM2–7, MCM8 and MCM10 was significantly up-regulated in HCC in Cohort 1. In Cohort 2, overexpression of MCM2–7, MCM8 and MCM10 was verified and significantly correlated with each other. Elevated MCM2, MCM6 and MCM7 were associated with adverse tumor features and poorer outcomes. In Cohort 3, MCM6 exhibited superior HCC diagnostic performance compared with MCM2 and MCM7 (AUC: 0.896 vs. 0.675 and 0.771, P < 0.01). Additionally, MCM6 other than MCM2 and MCM7 independently predicted poorer survival in 175 HCC patients. Furthermore, knockdown of MCM6 caused a delay in S/G2-phase progression as evidenced by down-regulation of CDK2, CDK4, CyclinA, CyclinB1, CyclinD1, and CyclinE in HCC cells. Conclusions We analyze MCMs mRNA and protein levels in tissue samples during hepatocarcinogenesis. MCM6 is identified as a driver of S/G2 cell cycle progression and a potential diagnostic and prognostic marker in HCC. Electronic supplementary material The online version of this article (10.1186/s12885-018-4056-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhikun Liu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Beijing, China
| | - Jie Li
- Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Beijing, China
| | - Jun Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Beijing, China
| | - Qiaonan Shan
- Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Beijing, China
| | - Haojiang Dai
- Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Beijing, China
| | - Haiyang Xie
- Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Beijing, China
| | - Lin Zhou
- Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Beijing, China
| | - Xiao Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Beijing, China. .,Collaborative innovation center for diagnosis and treatment of infectious diseases, Hangzhou, China.
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Beijing, China. .,Collaborative innovation center for diagnosis and treatment of infectious diseases, Hangzhou, China.
| |
Collapse
|
33
|
MicroRNA-31 functions as a tumor suppressor and increases sensitivity to mitomycin-C in urothelial bladder cancer by targeting integrin α5. Oncotarget 2018; 7:27445-57. [PMID: 27050274 PMCID: PMC5053662 DOI: 10.18632/oncotarget.8479] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 03/16/2016] [Indexed: 11/25/2022] Open
Abstract
Urothelial bladder cancer (UBC) is a common genitourinary malignancy. MiR-31, a well-identified miRNA, exhibits diverse properties in different cancers. However, the specific functions and mechanisms of miR-31 in UBC have not been investigated. In this study, tumor samples, especially invasive UBC, showed significantly reduced level of miR-31, as compared with normal urothelium. Prognostic analysis using the EORTC model showed that down-regulation of miR-31 correlated with higher risks of recurrence and progression in noninvasive UBC cases. Remarkably, overexpression of miR-31 mimics in UBC cell lines inhibited cell proliferation, migration and invasion. Integrin α5 (ITGA5), an integrin family member, was subsequently identified as a direct target of miR-31 in UBC cells. When treated with mitomycin-C (MMC), miR-31-expressing UBC cells displayed lower survival and higher apoptotic rates, and deactivated Akt and ERK. These effects arising from miR-31 overexpression were abrogated by ITGA5 restoration. Furthermore, miR-31 markedly inhibited tumor growth and increased the effectiveness of MMC in UBC xenografts. In summary, our data suggest that miR-31 is a prognostic predictor and can serve as a potential therapeutic target of UBC.
Collapse
|
34
|
Pan X, Wang Z, Wan B, Zheng Z. MicroRNA-206 inhibits the viability and migration of medulloblastoma cells by targeting LIM and SH3 protein 1. Exp Ther Med 2017; 14:3894-3900. [PMID: 29042998 DOI: 10.3892/etm.2017.5016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 04/07/2017] [Indexed: 12/30/2022] Open
Abstract
MicroRNA (miR)-206 has been found to be deregulated in various types of human cancer, including medulloblastoma. However, the regulatory mechanism of miR-206 in medulloblastoma growth and metastasis remains largely unclear. In the present study, reverse transcription-quantitative polymerase chain reaction data indicated that miR-206 was significantly downregulated in medulloblastoma tissues compared with adjacent non-tumor tissues (P<0.01). Furthermore, low expression of miR-206 was significantly associated with seeding at presentation and anaplastic histology (P<0.01), but not with sex, age, or residual tumors. Overexpression of miR-206 significantly reduced the viability and migration of medulloblastoma D341 cells (P<0.01). LIM and SH3 protein 1 (LASP1) was further identified as a novel target of miR-206 in D341 cells. mRNA levels of LASP1 were significantly higher in medulloblastoma tissues compared to adjacent non-tumor tissues (P<0.01), with an inverse correlation to the miR-206 levels in medulloblastoma tissues. In addition, protein expression levels of LASP1 ere negatively regulated by miR-206 in D341 cells. Further investigation showed that overexpression of LASP1 significantly eliminated the inhibitory effects of miR-206 on the migration and invasion of D341 cells (P<0.01). In conclusion, our study demonstrates that miR-206 has a suppressive role in medulloblastoma cell viability and invasion, partly at least, via the targeting of LASP1. Our study highlights the importance of the miR-206/LASP1 in medulloblastoma.
Collapse
Affiliation(s)
- Xuexia Pan
- Third Department of Pediatrics, People's Hospital of Binzhou, Binzhou, Shandong 256610, P.R. China
| | - Zhimin Wang
- Department of Hematology, People's Hospital of Binzhou, Binzhou, Shandong 256610, P.R. China
| | - Bin Wan
- Department of Neonatology, Affiliated Hospital of Binzhou Medical College, Binzhou, Shandong 256603, P.R. China
| | - Zhenwen Zheng
- Third Department of Pediatrics, People's Hospital of Binzhou, Binzhou, Shandong 256610, P.R. China
| |
Collapse
|
35
|
Neves H, Kwok HF. In sickness and in health: The many roles of the minichromosome maintenance proteins. Biochim Biophys Acta Rev Cancer 2017; 1868:295-308. [DOI: 10.1016/j.bbcan.2017.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/29/2017] [Accepted: 06/01/2017] [Indexed: 01/09/2023]
|
36
|
Chen L, Zhang YH, Lu G, Huang T, Cai YD. Analysis of cancer-related lncRNAs using gene ontology and KEGG pathways. Artif Intell Med 2017; 76:27-36. [PMID: 28363286 DOI: 10.1016/j.artmed.2017.02.001] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/31/2017] [Accepted: 02/05/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Cancer is a disease that involves abnormal cell growth and can invade or metastasize to other tissues. It is known that several factors are related to its initiation, proliferation, and invasiveness. Recently, it has been reported that long non-coding RNAs (lncRNAs) can participate in specific functional pathways and further regulate the biological function of cancer cells. Studies on lncRNAs are therefore helpful for uncovering the underlying mechanisms of cancer biological processes. METHODS We investigated cancer-related lncRNAs using gene ontology (GO) terms and KEGG pathway enrichment scores of neighboring genes that are co-expressed with the lncRNAs by extracting important GO terms and KEGG pathways that can help us identify cancer-related lncRNAs. The enrichment theory of GO terms and KEGG pathways was adopted to encode each lncRNA. Then, feature selection methods were employed to analyze these features and obtain the key GO terms and KEGG pathways. RESULTS The analysis indicated that the extracted GO terms and KEGG pathways are closely related to several cancer associated processes, such as hormone associated pathways, energy associated pathways, and ribosome associated pathways. And they can accurately predict cancer-related lncRNAs. CONCLUSIONS This study provided novel insight of how lncRNAs may affect tumorigenesis and which pathways may play important roles during it. These results could help understanding the biological mechanisms of lncRNAs and treating cancer.
Collapse
Affiliation(s)
- Lei Chen
- School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China; College of Information Engineering, Shanghai Maritime University, Shanghai 201306, People's Republic of China.
| | - Yu-Hang Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, People's Republic of China.
| | - Guohui Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China.
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, People's Republic of China.
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China.
| |
Collapse
|
37
|
STEPICHEVA NADEZDAA, SONG JIAL. Function and regulation of microRNA-31 in development and disease. Mol Reprod Dev 2016; 83:654-74. [PMID: 27405090 PMCID: PMC6040227 DOI: 10.1002/mrd.22678] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 06/29/2016] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that orchestrate numerous cellular processes both under normal physiological conditions as well as in diseases. This review summarizes the functional roles and transcriptional regulation of the highly evolutionarily conserved miRNA, microRNA-31 (miR-31). miR-31 is an important regulator of embryonic implantation, development, bone and muscle homeostasis, and immune system function. Its own regulation is disrupted during the onset and progression of cancer and autoimmune disorders such as psoriasis and systemic lupus erythematosus. Limited studies suggest that miR-31 is transcriptionally regulated by epigenetics, such as methylation and acetylation, as well as by a number of transcription factors. Overall, miR-31 regulates diverse cellular and developmental processes by targeting genes involved in cell proliferation, apoptosis, cell differentiation, and cell motility. Mol. Reprod. Dev. 83: 654-674, 2016 © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - JIA L. SONG
- Department of Biological Sciences, University of Delaware, Newark, Delaware
| |
Collapse
|
38
|
MicroRNA Biogenesis and Hedgehog-Patched Signaling Cooperate to Regulate an Important Developmental Transition in Granule Cell Development. Genetics 2016; 202:1105-18. [PMID: 26773048 DOI: 10.1534/genetics.115.184176] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/10/2016] [Indexed: 12/20/2022] Open
Abstract
The Dicer1, Dcr-1 homolog (Drosophila) gene encodes a type III ribonuclease required for the canonical maturation and functioning of microRNAs (miRNAs). Subsets of miRNAs are known to regulate normal cerebellar granule cell development, in addition to the growth and progression of medulloblastoma, a neoplasm that often originates from granule cell precursors. Multiple independent studies have also demonstrated that deregulation of Sonic Hedgehog (Shh)-Patched (Ptch) signaling, through miRNAs, is causative of granule cell pathologies. In the present study, we investigated the genetic interplay between miRNA biogenesis and Shh-Ptch signaling in granule cells of the cerebellum by way of the Cre/lox recombination system in genetically engineered models of Mus musculus (mouse). We demonstrate that, although the miRNA biogenesis and Shh-Ptch-signaling pathways, respectively, regulate the opposing growth processes of cerebellar hypoplasia and hyperplasia leading to medulloblastoma, their concurrent deregulation was nonadditive and did not bring the growth phenotypes toward an expected equilibrium. Instead, mice developed either hypoplasia or medulloblastoma, but of a greater severity. Furthermore, some genotypes were bistable, whereby subsets of mice developed hypoplasia or medulloblastoma. This implies that miRNAs and Shh-Ptch signaling regulate an important developmental transition in granule cells of the cerebellum. We also conclusively show that the Dicer1 gene encodes a haploinsufficient tumor suppressor gene for Ptch1-induced medulloblastoma, with the monoallielic loss of Dicer1 more severe than biallelic loss. These findings exemplify how genetic interplay between pathways may produce nonadditive effects with a substantial and unpredictable impact on biology. Furthermore, these findings suggest that the functional dosage of Dicer1 may nonadditively influence a wide range of Shh-Ptch-dependent pathologies.
Collapse
|
39
|
Jing J, Wang C, Liang Q, Zhao Y, Zhao Q, Wang S, Ma J. Lentivirus-Mediated knockdown of tectonic family member 1 inhibits medulloblastoma cell proliferation. Int J Clin Exp Med 2015; 8:13127-13135. [PMID: 26550235 PMCID: PMC4612920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 08/03/2015] [Indexed: 06/05/2023]
Abstract
Tectonic family member 1 (TCTN1) encodes a member of the tectonic family which are evolutionarily conserved secreted and transmembrane proteins, involving in a diverse variety of developmental processes. It has been demonstrated that tectonics expressed in regions that participate in Hedgehog (Hh) signaling during mouse embryonic development and was imperative for Hh-mediated patterning of the ventral neural tube. However, the expression and regulation of tectonics in human tumor is still not clear. In this study, shRNA-expressing lentivirus was constructed to knockdown TCTN1 in medulloblastoma cell line Daoy. The results showed that knockdown of TCTN1 inhibited cell proliferation and colony formation in Daoy cell line, also caused cell cycle arrest at the G2/M boundary. Taken all together, our data suggest that TCTN1 might play an important role in the progression of medulloblastoma.
Collapse
Affiliation(s)
- Junjie Jing
- Neurosurgery, Fuzhou General Hospital of Second Military Medical UniversityFuzhou 350025
| | - Chengfeng Wang
- Department of Pediatrics, Fuzhou General Hospital of Second Military Medical UniversityFuzhou 350025
| | - Qinchuan Liang
- Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University, School of MedicineShanghai 200092
| | - Yang Zhao
- Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University, School of MedicineShanghai 200092
| | - Qingshuang Zhao
- Neurosurgery, Fuzhou General Hospital of Second Military Medical UniversityFuzhou 350025
| | - Shousen Wang
- Neurosurgery, Fuzhou General Hospital of Second Military Medical UniversityFuzhou 350025
| | - Jie Ma
- Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University, School of MedicineShanghai 200092
| |
Collapse
|
40
|
Aleassa EM, Xing M, Keijzer R. Nanomedicine as an innovative therapeutic strategy for pediatric cancer. Pediatr Surg Int 2015; 31:611-6. [PMID: 25690563 DOI: 10.1007/s00383-015-3683-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/10/2015] [Indexed: 11/29/2022]
Abstract
Childhood cancer is the leading cause of mortality in children between 1 and 14 years of age. Malignancy accounts for 18 % of overall childhood mortality. Therapeutic advances in the field of pediatric oncology have helped to increase survival. Nanotechnology is the modification of materials at a nanoscale and can be used to deliver therapeutic agents. Examples of nanotechnology applications are organic self-assembled amphiphilic polymers, non-organic nanocarriers such as nanotubes and quantum dots. Each of these has their own utility in different settings. Application of nanotechnology in medicine has been extensively studied. Examples of pediatric tumors that received special attention are: neuroblastoma, retinoblastoma, central nervous system tumors and musculoskeletal tumors. This review will summarize the application of nanomedicine as an innovative management strategy in pediatric oncology.
Collapse
|
41
|
Braoudaki M, Lambrou GI. MicroRNAs in pediatric central nervous system embryonal neoplasms: the known unknown. J Hematol Oncol 2015; 8:6. [PMID: 25652781 PMCID: PMC4333163 DOI: 10.1186/s13045-014-0101-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 12/27/2014] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs (miRNAs) are endogenous short non-coding RNAs that repress post-transcriptional regulation of gene expression, while embryonal central nervous system tumors are the foremost cause of mortality in children suffering from a neoplasm. MiRNAs and their regulatory mechanisms are new to understand, while pediatric CNS tumors are difficult to comprehend. Therefore, identification of the link between them composes a major scientific challenge. The present study, reviewed the current knowledge on the role of miRNA in pediatric CNS embryonal tumors, attempting to collect the existing information in one piece of work that could ideally be used as a guide for future reference and research.
Collapse
Affiliation(s)
- Maria Braoudaki
- First Department of Pediatrics, University of Athens, Choremeio Research Laboratory, Athens, Greece. .,University Research Institute for the Study and Treatment of Childhood Genetic and Malignant Diseases, University of Athens, Aghia Sophia Children's Hospital, Athens, Greece.
| | - George I Lambrou
- First Department of Pediatrics, University of Athens, Choremeio Research Laboratory, Athens, Greece.
| |
Collapse
|