1
|
Han SB, Lee SS. Isolation and Characterization of Exosomes from Cancer Cells Using Antibody-Functionalized Paddle Screw-Type Devices and Detection of Exosomal miRNA Using Piezoelectric Biosensor. SENSORS (BASEL, SWITZERLAND) 2024; 24:5399. [PMID: 39205093 PMCID: PMC11359151 DOI: 10.3390/s24165399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/08/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Exosomes are small extracellular vesicles produced by almost all cell types in the human body, and exosomal microRNAs (miRNAs) are small non-coding RNA molecules that are known to serve as important biomarkers for diseases such as cancer. Given that the upregulation of miR-106b is closely associated with several types of malignancies, the sensitive and accurate detection of miR-106b is important but difficult. In this study, a surface acoustic wave (SAW) biosensor was developed to detect miR-106b isolated from cancer cells based on immunoaffinity separation technique using our unique paddle screw device. Our novel SAW biosensor could detect a miR-106b concentration as low as 0.0034 pM in a linear range from 0.1 pM to 1.0 μM with a correlation coefficient of 0.997. Additionally, we were able to successfully detect miR-106b in total RNA extracted from the exosomes isolated from the MCF-7 cancer cell line, a model system for human breast cancer, with performance comparable to commercial RT-qPCR methods. Therefore, the exosome isolation by the paddle screw method and the miRNA detection using the SAW biosensor has the potential to be used in basic biological research and clinical diagnosis as an alternative to RT-qPCR.
Collapse
Affiliation(s)
| | - Soo Suk Lee
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea;
| |
Collapse
|
2
|
Panina SB, Schweer JV, Zhang Q, Raina G, Hardtke HA, Kim S, Yang W, Siegel D, Zhang YJ. Targeting of REST with rationally-designed small molecule compounds exhibits synergetic therapeutic potential in human glioblastoma cells. BMC Biol 2024; 22:83. [PMID: 38609948 PMCID: PMC11015551 DOI: 10.1186/s12915-024-01879-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 04/04/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is an aggressive brain cancer associated with poor prognosis, intrinsic heterogeneity, plasticity, and therapy resistance. In some GBMs, cell proliferation is fueled by a transcriptional regulator, repressor element-1 silencing transcription factor (REST). RESULTS Using CRISPR/Cas9, we identified GBM cell lines dependent on REST activity. We developed new small molecule inhibitory compounds targeting small C-terminal domain phosphatase 1 (SCP1) to reduce REST protein level and transcriptional activity in glioblastoma cells. Top leads of the series like GR-28 exhibit potent cytotoxicity, reduce REST protein level, and suppress its transcriptional activity. Upon the loss of REST protein, GBM cells can potentially compensate by rewiring fatty acid metabolism, enabling continued proliferation. Combining REST inhibition with the blockade of this compensatory adaptation using long-chain acyl-CoA synthetase inhibitor Triacsin C demonstrated substantial synergetic potential without inducing hepatotoxicity. CONCLUSIONS Our results highlight the efficacy and selectivity of targeting REST alone or in combination as a therapeutic strategy to combat high-REST GBM.
Collapse
Affiliation(s)
- Svetlana B Panina
- Department of Molecular Biosciences, The University of Texas at Austin, 2500 Speedway, Austin, TX, USA
| | - Joshua V Schweer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of California San Diego, 9500 Gilman Drive 0741, La Jolla, CA, USA
| | - Qian Zhang
- Department of Molecular Biosciences, The University of Texas at Austin, 2500 Speedway, Austin, TX, USA
| | - Gaurav Raina
- Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of California San Diego, 9500 Gilman Drive 0741, La Jolla, CA, USA
| | - Haley A Hardtke
- Department of Molecular Biosciences, The University of Texas at Austin, 2500 Speedway, Austin, TX, USA
| | - Seungjin Kim
- Department of Molecular Biosciences, The University of Texas at Austin, 2500 Speedway, Austin, TX, USA
| | - Wanjie Yang
- Department of Molecular Biosciences, The University of Texas at Austin, 2500 Speedway, Austin, TX, USA
| | - Dionicio Siegel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of California San Diego, 9500 Gilman Drive 0741, La Jolla, CA, USA
| | - Y Jessie Zhang
- Department of Molecular Biosciences, The University of Texas at Austin, 2500 Speedway, Austin, TX, USA.
| |
Collapse
|
3
|
Jodeiry Zaer S, Aghamaali M, Amini M, Doustvandi MA, Hosseini SS, Baradaran B, Najafi S, Baghay Esfandyari Y, Mokhtarzadeh A. Cooperatively inhibition effect of miR-143-5p and miR-145-5p in tumorigenesis of glioblastoma cells through modulating AKT signaling pathway. BIOIMPACTS : BI 2023; 14:29913. [PMID: 38938754 PMCID: PMC11199930 DOI: 10.34172/bi.2023.29913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/30/2023] [Accepted: 10/07/2023] [Indexed: 06/29/2024]
Abstract
Introduction As the most common aggressive primary brain tumor, glioblastoma is inevitably a recurrent malignancy whose patients' prognosis is poor. miR-143 and miR-145, as tumor suppressor miRNAs, are downregulated through tumorigenesis of multiple human cancers, including glioblastoma. These two miRNAs regulate numerous cellular processes, such as proliferation and migration. This research was intended to explore the simultaneous replacement effect of miR-143, and miR-145 on in vitro tumorgenicity of U87 glioblastoma cells. Methods U87 cells were cultured, and transfected with miR-143-5p and miR-145-5p. Afterward, the changes in cell viability, and apoptosis induction were determined by MTT assay and Annexin V/PI staining. The accumulation of cells at the cell cycle phases was assessed using the flow cytometry. Wound healing and colony formation assays were performed to study cell migration. qRT-PCR and western blot techniques were utilized to quantify gene expression levels. Results Our results showed that miR-143-5p and 145-5p exogenous upregulation cooperatively diminished cell viability, and enhanced U-87 cell apoptosis by modulating Caspase-3/8/9, Bax, and Bcl-2 protein expression. The combination therapy increased accumulation of cells at the sub-G1 phase by modulating CDK1, Cyclin D1, and P53 protein expression. miR-143/145-5p significantly decreased cell migration, and reduced colony formation ability by the downregulation of c-Myc and CD44 gene expression. Furthermore, the results showed the combination therapy of these miRNAs could remarkably downregulate phosphorylated-AKT expression levels. Conclusion In conclusion, miR-143 and miR-145 were indicated to show cooperative anti- cancer effects on glioblastoma cells via modulating AKT signaling as a new therapeutic approach.
Collapse
Affiliation(s)
- Sheyda Jodeiry Zaer
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Purazo ML, Ice RJ, Shimpi R, Hoenerhoff M, Pugacheva EN. NEDD9 Overexpression Causes Hyperproliferation of Luminal Cells and Cooperates with HER2 Oncogene in Tumor Initiation: A Novel Prognostic Marker in Breast Cancer. Cancers (Basel) 2023; 15:1119. [PMID: 36831460 PMCID: PMC9954084 DOI: 10.3390/cancers15041119] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/23/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
HER2 overexpression occurs in 10-20% of breast cancer patients. HER2+ tumors are characterized by an increase in Ki67, early relapse, and increased metastasis. Little is known about the factors influencing early stages of HER2- tumorigenesis and diagnostic markers. Previously, it was shown that the deletion of NEDD9 in mouse models of HER2 cancer interferes with tumor growth, but the role of NEDD9 upregulation is currently unexplored. We report that NEDD9 is overexpressed in a significant subset of HER2+ breast cancers and correlates with a limited response to anti-HER2 therapy. To investigate the mechanisms through which NEDD9 influences HER2-dependent tumorigenesis, we generated MMTV-Cre-NEDD9 transgenic mice. The analysis of mammary glands shows extensive ductal epithelium hyperplasia, increased branching, and terminal end bud expansion. The addition of oncogene Erbb2 (neu) leads to the earlier development of early hyperplastic benign lesions (~16 weeks), with a significantly shorter latency than the control mice. Similarly, NEDD9 upregulation in MCF10A-derived acini leads to hyperplasia-like DCIS. This phenotype is associated with activation of ERK1/2 and AURKA kinases, leading to an increased proliferation of luminal cells. These findings indicate that NEDD9 is setting permissive conditions for HER2-induced tumorigenesis, thus identifying this protein as a potential diagnostic marker for early detection.
Collapse
Affiliation(s)
- Marc L. Purazo
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
| | - Ryan J. Ice
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
| | - Rahul Shimpi
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
| | - Mark Hoenerhoff
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elena N. Pugacheva
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
- Department of Biochemistry & Molecular Medicine, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
| |
Collapse
|
5
|
Rajabi A, Kayedi M, Rahimi S, Dashti F, Mirazimi SMA, Homayoonfal M, Mahdian SMA, Hamblin MR, Tamtaji OR, Afrasiabi A, Jafari A, Mirzaei H. Non-coding RNAs and glioma: Focus on cancer stem cells. Mol Ther Oncolytics 2022; 27:100-123. [PMID: 36321132 PMCID: PMC9593299 DOI: 10.1016/j.omto.2022.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Glioblastoma and gliomas can have a wide range of histopathologic subtypes. These heterogeneous histologic phenotypes originate from tumor cells with the distinct functions of tumorigenesis and self-renewal, called glioma stem cells (GSCs). GSCs are characterized based on multi-layered epigenetic mechanisms, which control the expression of many genes. This epigenetic regulatory mechanism is often based on functional non-coding RNAs (ncRNAs). ncRNAs have become increasingly important in the pathogenesis of human cancer and work as oncogenes or tumor suppressors to regulate carcinogenesis and progression. These RNAs by being involved in chromatin remodeling and modification, transcriptional regulation, and alternative splicing of pre-mRNA, as well as mRNA stability and protein translation, play a key role in tumor development and progression. Numerous studies have been performed to try to understand the dysregulation pattern of these ncRNAs in tumors and cancer stem cells (CSCs), which show robust differentiation and self-regeneration capacity. This review provides recent findings on the role of ncRNAs in glioma development and progression, particularly their effects on CSCs, thus accelerating the clinical implementation of ncRNAs as promising tumor biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Ali Rajabi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mehrdad Kayedi
- Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shiva Rahimi
- School of Medicine,Fasa University of Medical Sciences, Fasa, Iran
| | - Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mina Homayoonfal
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mohammad Amin Mahdian
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Omid Reza Tamtaji
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Afrasiabi
- Department of Internal Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ameneh Jafari
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
6
|
Zhang Z, Li J, Yan B, Tu H, Huang C, Costa M. Loss of MEG3 and upregulation of miR-145 play an important role in the invasion and migration of Cr(VI)-transformed cells. Heliyon 2022; 8:e10086. [PMID: 36046536 PMCID: PMC9421329 DOI: 10.1016/j.heliyon.2022.e10086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/25/2022] [Accepted: 07/21/2022] [Indexed: 11/30/2022] Open
Abstract
Chronic exposure of human bronchial epithelial BEAS-2B cells to hexavalent chromium (Cr(VI)) causes malignant cell transformation. These transformed cells exhibit increases in migration and invasion. Neuronal precursor of developmentally downregulated protein 9 (NEDD9) is upregulated in Cr(VI)-transformed cells compared to that of passage-matched normal BEAS-2B cells. Knockdown of NEDD9 by its shRNA reduced invasion and migration of Cr(VI)-transformed cells. Maternally expressed gene 3 (MEG3), a long noncoding RNA, was lost and microRNA 145 (miR-145) was upregulated in Cr(VI)-transformed cells. MEG3 was bound to miR-145 and this binding reduced its activity. Overexpression of MEG3 or inhibition of miR-145 decreased invasion and migration of Cr(VI)-transformed cells. Overexpression of MEG3 was able to decrease miR-145 level and NEDD9 protein level in Cr(VI)-transformed cells. Ectopic expression of MEG3 was also shown to reduce β-catenin activation. Inhibition of miR-145 in Cr(VI)-transformed cells decreased Slug, an important transcription factor that regulates epithelial-to-mesenchymal transition (EMT). Inhibition of miR-145 was found to increase MEG3 in Cr(VI)-transformed cells. Further studies showed that mutation of MEG3 at the binding site for miR-145 did not change NEDD9 and failed to decrease invasion and migration. The present study demonstrated that loss of MEG3 and upregulation of miR-145 elevated NEDD9, resulting in activation of β-catenin and further upregulation of EMT, leading to increased invasion and migration of Cr(VI)-transformed cells.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Environmental Medicine, New York University Grossman School of Medicine, 341 East 25 Street, New York, New York, 10010, USA
| | - Jingxia Li
- Department of Environmental Medicine, New York University Grossman School of Medicine, 341 East 25 Street, New York, New York, 10010, USA
| | - Bo Yan
- Department of Environmental Medicine, New York University Grossman School of Medicine, 341 East 25 Street, New York, New York, 10010, USA
| | - Huailu Tu
- Department of Environmental Medicine, New York University Grossman School of Medicine, 341 East 25 Street, New York, New York, 10010, USA
| | - Chao Huang
- Department of Environmental Medicine, New York University Grossman School of Medicine, 341 East 25 Street, New York, New York, 10010, USA
| | - Max Costa
- Department of Environmental Medicine, New York University Grossman School of Medicine, 341 East 25 Street, New York, New York, 10010, USA
| |
Collapse
|
7
|
WAN J, HUANG M. Apigenin inhibits proliferation, migration, invasion and epithelial mesenchymal transition of glioma cells by regulating miR-103a-3p/NEED9/AKT axis. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.23022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Jing WAN
- Renmin Hospital of Wuhan University, China
| | - Min HUANG
- Renmin Hospital of Wuhan University, China
| |
Collapse
|
8
|
Hua S, Feng T, Yin L, Wang Q, Shao X. NEDD9 overexpression: Prognostic and guidance value in acute myeloid leukaemia. J Cell Mol Med 2021; 25:9331-9339. [PMID: 34432355 PMCID: PMC8500976 DOI: 10.1111/jcmm.16870] [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: 05/26/2021] [Revised: 07/22/2021] [Accepted: 08/02/2021] [Indexed: 12/11/2022] Open
Abstract
It has been demonstrated that neural precursor cell expressed developmentally downregulated protein (NEDD) plays crucial roles in tumorigenesis and may serve as potential biomarkers in cancer diagnosis and prognosis. However, few studies systematically investigated the expression of NEDD family members in acute myeloid leukaemia (AML). We systemically determined the expression of NEDD family members in AML and determined their clinical significance. We identified that NEDD9 expression was the only member among NEDD family which was significantly increased in AML. NEDD9 overexpression was more frequently classified as FAB‐M4/M5 (p = 0.008 and 0.013, respectively), hardly as FAB‐M2/M3. Moreover, NEDD9 overexpression was significantly associated with complex karyotype and TP53 mutation. The significant association between NEDD9 overexpression and survival was also observed in whole‐cohort AML and non‐M3 AML patients. Notably, AML patients with NEDD9 overexpression may benefit from hematopoietic stem cell transplantation (HSCT), whereas those cases without NEDD9 overexpression did not. Finally, a total of 822 mRNAs and 31 microRNAs were found to be differentially expressed between two groups. Among the microRNAs, miR‐381 was also identified as a microRNA that could direct target NEDD9. Taken together, our findings demonstrated that NEDD9 overexpression is associated with genetic abnormalities as well as prognosis and might act as a potential biomarker guiding the choice between HSCT and chemotherapy in patients with AML after achieving complete remission.
Collapse
Affiliation(s)
- Shenghao Hua
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, China
| | - Tao Feng
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, China
| | - Lei Yin
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, China
| | - Qi Wang
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, China
| | - Xuejun Shao
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
9
|
Jiawei Z, Min M, Yingru X, Xin Z, Danting L, Yafeng L, Jun X, Wangfa H, Lijun Z, Jing W, Dong H. Identification of Key Genes in Lung Adenocarcinoma and Establishment of Prognostic Mode. Front Mol Biosci 2020; 7:561456. [PMID: 33195408 PMCID: PMC7653064 DOI: 10.3389/fmolb.2020.561456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/07/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The development of human tumors is associated with the abnormal expression of various functional genes, and a massive tumor-based database needs to be deeply mined. Based on a multigene prediction model, access to urgent prognosis of patients has become possible. MATERIALS AND METHODS We selected three RNA expression profiles (GSE32863, GSE10072, and GSE43458) from the lung adenocarcinoma (LUAD) database of the Gene Expression Omnibus (GEO) and analyzed the differentially expressed genes (DEGs) between tumor and normal tissue using GEO2R program. After that, we analyzed the transcriptome data of 479 LUAD samples (54 normal tissue samples and 425 cancer tissue samples) and their clinical follow-up data from the (TCGA) database. Kaplan-Meier (KM) curve and receiver operating characteristic (ROC) were used to assess the prediction model. Multivariate Cox analysis was used to identify independent predictors. TCGA pancreatic adenocarcinoma datasets were used to establish a nomogram model. RESULTS We found 98 significantly prognosis-related genes using KM and COX analysis, among which six genes were found to be the DEGs in GEO. Using multivariate analysis, it was found that a single gene could not be used as an independent predictor of prognosis. However, the risk score calculated by weighting these six genes could serve as an independent prognosis predictor. COX analysis performed with multiple covariates such as age, gender, tumor stage, and TNM typing showed that risk score could still be utilized as an independent risk factor for patient survival rate (p = 0.013) and had an applicable reliability (area under the curve, AUC = 0.665). By combining risk score and various clinical features, the nomogram model was constructed, which had been proven to have high consistency for the prediction of 3- and 5-year survival rate (concordance = 0.751) and high accuracy as tested by ROC (AUC = 0.71;AUC = 0.708). CONCLUSION We proposed a method to predict the prognosis of LUAD by weighting multiple genes and constructed a nomogram model suitable for the prognostic evaluation of LUAD, which could provide a new tool for the identification of therapeutic targets and the efficacy evaluation of LUAD.
Collapse
Affiliation(s)
- Zhou Jiawei
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Mu Min
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, Huainan, China
| | - Xing Yingru
- Affiliated Cancer Hospital, Anhui University of Science and Technology, Huainan, China
| | - Zhang Xin
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Li Danting
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Liu Yafeng
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Xie Jun
- Affiliated Cancer Hospital, Anhui University of Science and Technology, Huainan, China
| | - Hu Wangfa
- Affiliated Cancer Hospital, Anhui University of Science and Technology, Huainan, China
| | - Zhang Lijun
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Wu Jing
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, Huainan, China
| | - Hu Dong
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, Huainan, China
| |
Collapse
|
10
|
Therapeutically Significant MicroRNAs in Primary and Metastatic Brain Malignancies. Cancers (Basel) 2020; 12:cancers12092534. [PMID: 32906592 PMCID: PMC7564168 DOI: 10.3390/cancers12092534] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary The overall survival of brain cancer patients remains grim, with conventional therapies such as chemotherapy and radiotherapy only providing marginal benefits to patient survival. Cancers are complex, with multiple pathways being dysregulated simultaneously. Non-coding RNAs such as microRNA (miRNAs) are gaining importance due to their potential in regulating a variety of targets implicated in the pathology of cancers. This could be leveraged for the development of targeted and personalized therapies for cancers. Since miRNAs can upregulate and/or downregulate proteins, this review aims to understand the role of these miRNAs in primary and metastatic brain cancers. Here, we discuss the regulatory mechanisms of ten miRNAs that are highly dysregulated in glioblastoma and metastatic brain tumors. This will enable researchers to develop miRNA-based targeted cancer therapies and identify potential prognostic biomarkers. Abstract Brain cancer is one among the rare cancers with high mortality rate that affects both children and adults. The most aggressive form of primary brain tumor is glioblastoma. Secondary brain tumors most commonly metastasize from primary cancers of lung, breast, or melanoma. The five-year survival of primary and secondary brain tumors is 34% and 2.4%, respectively. Owing to poor prognosis, tumor heterogeneity, increased tumor relapse, and resistance to therapies, brain cancers have high mortality and poor survival rates compared to other cancers. Early diagnosis, effective targeted treatments, and improved prognosis have the potential to increase the survival rate of patients with primary and secondary brain malignancies. MicroRNAs (miRNAs) are short noncoding RNAs of approximately 18–22 nucleotides that play a significant role in the regulation of multiple genes. With growing interest in the development of miRNA-based therapeutics, it is crucial to understand the differential role of these miRNAs in the given cancer scenario. This review focuses on the differential expression of ten miRNAs (miR-145, miR-31, miR-451, miR-19a, miR-143, miR-125b, miR-328, miR-210, miR-146a, and miR-126) in glioblastoma and brain metastasis. These miRNAs are highly dysregulated in both primary and metastatic brain tumors, which necessitates a better understanding of their role in these cancers. In the context of the tumor microenvironment and the expression of different genes, these miRNAs possess both oncogenic and/or tumor-suppressive roles within the same cancer.
Collapse
|
11
|
Tanooka H, Inoue A, Takahashi RU, Tatsumi K, Fujikawa K, Nagao T, Ishiai M, Chiwaki F, Aoyagi K, Sasaki H, Ochiya T. Bacterial SOS Genes mucAB/umuDC Promote Mouse Tumors by Activating Oncogenes Nedd9/Aurkb via a miR-145 Sponge. Mol Cancer Res 2020; 18:1271-1277. [PMID: 32513897 DOI: 10.1158/1541-7786.mcr-20-0137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/31/2020] [Accepted: 06/02/2020] [Indexed: 11/16/2022]
Abstract
The mechanism of cancer induction involves an aberrant expression of oncogenes whose functions can be controlled by RNAi with miRNA. Even foreign bacterial RNA may interfere with the expression of oncogenes. Here we show that bacterial plasmid mucAB and its Escherichia coli genomic homolog umuDC, carrying homologies that match the mouse anti-miR-145, sequestered the miR-145 function in mouse BALB 3T3 cells in a tetracycline (Tet)-inducible manner, activated oncogene Nedd9 and its downstream Aurkb, and further enhanced microcolony formation and cellular transformation as well as the short fragments of the bacterial gene containing the anti-miR-145 sequence. Furthermore, mucAB transgenic mice showed a 1.7-fold elevated tumor incidence compared with wild-type mice after treatments with 3-methylcolanthrene. However, the mutation frequency in intestinal stem cells of the mucAB transgenic mice was unchanged after treatment with X-rays or ethyl-nitrosourea, indicating that the target of mucAB/umuDC is the promotion stage in carcinogenesis. IMPLICATIONS: Foreign bacterial genes can exert oncogenic activity via RNAi, if endogenously expressed. VISUAL OVERVIEW: http://mcr.aacrjournals.org/content/molcanres/18/9/1271/F1.large.jpg.
Collapse
Affiliation(s)
- Hiroshi Tanooka
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan.
- Central Radioisotope Division, National Cancer Center Research Institute, Tokyo, Japan
- Biological Effects Research Group, Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
| | - Ayako Inoue
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Ryou-U Takahashi
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Kouichi Tatsumi
- Biological Effects Research Group, Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
| | - Kazuo Fujikawa
- Department of Life Science, Faculty of Science and Technology, Kindai University, Higashiosaka, Japan
| | - Tetsuji Nagao
- Department of Life Science, Faculty of Science and Technology, Kindai University, Higashiosaka, Japan
| | - Masamichi Ishiai
- Central Radioisotope Division, National Cancer Center Research Institute, Tokyo, Japan
| | - Fumiko Chiwaki
- Department of Translational Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Kazuhiko Aoyagi
- Department of Translational Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Hiroki Sasaki
- Department of Translational Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
- Department of Molecular and Cellular Medicine, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
12
|
Hassn Mesrati M, Behrooz AB, Y. Abuhamad A, Syahir A. Understanding Glioblastoma Biomarkers: Knocking a Mountain with a Hammer. Cells 2020; 9:E1236. [PMID: 32429463 PMCID: PMC7291262 DOI: 10.3390/cells9051236] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/18/2020] [Accepted: 03/24/2020] [Indexed: 12/14/2022] Open
Abstract
Gliomas are the most frequent and deadly form of human primary brain tumors. Among them, the most common and aggressive type is the high-grade glioblastoma multiforme (GBM), which rapidly grows and renders patients a very poor prognosis. Meanwhile, cancer stem cells (CSCs) have been determined in gliomas and play vital roles in driving tumor growth due to their competency in self-renewal and proliferation. Studies of gliomas have recognized CSCs via specific markers. This review comprehensively examines the current knowledge of the most significant CSCs markers in gliomas in general and in glioblastoma in particular and specifically focuses on their outlook and importance in gliomas CSCs research. We suggest that CSCs should be the superior therapeutic approach by directly targeting the markers. In addition, we highlight the association of these markers with each other in relation to their cascading pathways, and interactions with functional miRNAs, providing the role of the networks axes in glioblastoma signaling pathways.
Collapse
Affiliation(s)
| | | | | | - Amir Syahir
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia; (M.H.M.); (A.B.B.); (A.Y.A.)
| |
Collapse
|
13
|
Recent Trends of microRNA Significance in Pediatric Population Glioblastoma and Current Knowledge of Micro RNA Function in Glioblastoma Multiforme. Int J Mol Sci 2020; 21:ijms21093046. [PMID: 32349263 PMCID: PMC7246719 DOI: 10.3390/ijms21093046] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Central nervous system tumors are a significant problem for modern medicine because of their location. The explanation of the importance of microRNA (miRNA) in the development of cancerous changes plays an important role in this respect. The first papers describing the presence of miRNA were published in the 1990s. The role of miRNA has been pointed out in many medical conditions such as kidney disease, diabetes, neurodegenerative disorder, arthritis and cancer. There are several miRNAs responsible for invasiveness, apoptosis, resistance to treatment, angiogenesis, proliferation and immunology, and many others. The research conducted in recent years analyzing this group of tumors has shown the important role of miRNA in the course of gliomagenesis. These particles seem to participate in many stages of the development of cancer processes, such as proliferation, angiogenesis, regulation of apoptosis or cell resistance to cytostatics.
Collapse
|
14
|
A Peptide Nucleic Acid (PNA) Masking the miR-145-5p Binding Site of the 3'UTR of the Cystic Fibrosis Transmembrane Conductance Regulator ( CFTR) mRNA Enhances CFTR Expression in Calu-3 Cells. Molecules 2020; 25:molecules25071677. [PMID: 32260566 PMCID: PMC7181265 DOI: 10.3390/molecules25071677] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 01/19/2023] Open
Abstract
Peptide nucleic acids (PNAs) have been demonstrated to be very useful tools for gene regulation at different levels and with different mechanisms of action. In the last few years the use of PNAs for targeting microRNAs (anti-miRNA PNAs) has provided impressive advancements. In particular, targeting of microRNAs involved in the repression of the expression of the cystic fibrosis transmembrane conductance regulator (CFTR) gene, which is defective in cystic fibrosis (CF), is a key step in the development of new types of treatment protocols. In addition to the anti-miRNA therapeutic strategy, inhibition of miRNA functions can be reached by masking the miRNA binding sites present within the 3′UTR region of the target mRNAs. The objective of this study was to design a PNA masking the binding site of the microRNA miR-145-5p present within the 3′UTR of the CFTR mRNA and to determine its activity in inhibiting miR-145-5p function, with particular focus on the expression of both CFTR mRNA and CFTR protein in Calu-3 cells. The results obtained support the concept that the PNA masking the miR-145-5p binding site of the CFTR mRNA is able to interfere with miR-145-5p biological functions, leading to both an increase of CFTR mRNA and CFTR protein content.
Collapse
|
15
|
The Microrna-143/145 Cluster in Tumors: A Matter of Where and When. Cancers (Basel) 2020; 12:cancers12030708. [PMID: 32192092 PMCID: PMC7140083 DOI: 10.3390/cancers12030708] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/13/2020] [Accepted: 03/15/2020] [Indexed: 01/06/2023] Open
Abstract
The establishment and spreading of cancer involve the acquirement of many biological functions including resistance to apoptosis, enhanced proliferation and the ability to invade the surrounding tissue, extravasate from the primary site, survive in circulating blood, and finally extravasate and colonize distant organs giving origin to metastatic lesions, the major cause of cancer deaths. Dramatic changes in the expression of protein coding genes due to altered transcription factors activity or to epigenetic modifications orchestrate these events, intertwining with a microRNA regulatory network that is often disrupted in cancer cells. microRNAs-143 and -145 represent puzzling players of this game, with apparently contradictory functions. They were at first classified as tumor suppressive due to their frequently reduced levels in tumors, correlating with cell survival, proliferation, and migration. More recently, pro-oncogenic roles of these microRNAs have been described, challenging their simplistic definition as merely tumor-suppressive. Here we review their known activities in tumors, whether oncogenic or onco-suppressive, and highlight how their expression and functions are strongly dependent on their complex regulation downstream and upstream of cytokines and growth factors, on the cell type of expression and on the specific tumor stage.
Collapse
|
16
|
Wang Y, Dan L, Li Q, Li L, Zhong L, Shao B, Yu F, He S, Tian S, He J, Xiao Q, Putti TC, He X, Feng Y, Lin Y, Xiang T. ZMYND10, an epigenetically regulated tumor suppressor, exerts tumor-suppressive functions via miR145-5p/NEDD9 axis in breast cancer. Clin Epigenetics 2019; 11:184. [PMID: 31801619 PMCID: PMC6894283 DOI: 10.1186/s13148-019-0785-z] [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] [Received: 08/17/2019] [Accepted: 11/24/2019] [Indexed: 02/07/2023] Open
Abstract
Background Recent studies suggested that ZMYND10 is a potential tumor suppressor gene in multiple tumor types. However, the mechanism by which ZMYND10 inhibits breast cancer remains unclear. Here, we investigated the role and mechanism of ZMYND10 in breast cancer inhibition. Results ZMYND10 was dramatically reduced in multiple breast cancer cell lines and tissues, which was associated with promoter hypermethylation. Ectopic expression of ZMYND10 in silenced breast cancer cells induced cell apoptosis while suppressed cell growth, cell migration and invasion in vitro, and xenograft tumor growth in vivo. Furthermore, molecular mechanism studies indicated that ZMYND10 enhances expression of miR145-5p, which suppresses the expression of NEDD9 protein through directly targeting the 3'-untranslated region of NEDD9 mRNA. Conclusions Results from this study show that ZMYND10 suppresses breast cancer tumorigenicity by inhibiting the miR145-5p/NEDD9 signaling pathway. This novel discovered signaling pathway may be a valid target for small molecules that might help to develop new therapies to better inhibit the breast cancer metastasis.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liangying Dan
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,The People's Hospital of Tongliang District, Chongqing, China
| | - Qianqian Li
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lili Li
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Lan Zhong
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Bianfei Shao
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Yu
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Sanxiu He
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shaorong Tian
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jin He
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Xiao
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Thomas C Putti
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xiaoqian He
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yixiao Feng
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Lin
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | - Tingxiu Xiang
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
17
|
Gu Y, Lu J, Chen C, Zheng F. NEDD9 overexpression predicts poor prognosis in solid cancers: a meta-analysis. Onco Targets Ther 2019; 12:4213-4222. [PMID: 31213839 PMCID: PMC6549757 DOI: 10.2147/ott.s205760] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 04/26/2019] [Indexed: 12/24/2022] Open
Abstract
Background: The oncogenicity of neural precursor cell-expressed developmentally down-regulated 9 (NEDD9) has been demonstrated in multiple cancer types. However, the prognostic value of NEDD9 in some solid cancers remains controversial. Thus, this meta-analysis was conducted to evaluate the relationship between NEDD9 expression survival rates in solid tumors. Method: Our meta-analysis included studies searched from various search engines with specific inclusion criteria and exclusion criteria. Combined HRs for overall survival (OS) and disease-free survival (DFS) or progression-free survival (PFS) or recurrence-free survival (RFS) or cancer-specific survival (CSS) were assessed using fixed-effects and random-effects models. The source of heterogeneity was identified by subgroup analysis. Additionally, publication bias was assessed using funnel plot and Egger’s regression asymmetry test. Result: Eighteen studies with a total of 2,476 patients were retrieved for analysis. Pooled HRs and 95% CIs were calculated. Both OS (HR=1.82; 95% CI: 1.43–2.31) and DFS/PFS/RFS/CSS (HR=2.54; 95% CI: 1.93–3.33) indicated that NEDD9 overexpression is associated with poor OS in cancer patients with solid tumors. Conclusion: NEDD9 overexpression might be a potential marker to predict prognosis in solid cancer patients.
Collapse
Affiliation(s)
- Yang Gu
- Department of Orthopedics, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Jingjing Lu
- Department of Gynecology, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No.2 Hospital), Ningbo, People's Republic of China
| | - Chen Chen
- Department of Orthopedics, First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Fei Zheng
- Department of Gynecology, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No.2 Hospital), Ningbo, People's Republic of China
| |
Collapse
|
18
|
Sharifzad F, Ghavami S, Verdi J, Mardpour S, Mollapour Sisakht M, Azizi Z, Taghikhani A, Łos MJ, Fakharian E, Ebrahimi M, Hamidieh AA. Glioblastoma cancer stem cell biology: Potential theranostic targets. Drug Resist Updat 2019; 42:35-45. [PMID: 30877905 DOI: 10.1016/j.drup.2018.03.003] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/28/2018] [Accepted: 03/16/2018] [Indexed: 12/21/2022]
Abstract
Glioblastoma multiforme (GBM) is among the most incurable cancers. GBMs survival rate has not markedly improved, despite new radical surgery protocols, the introduction of new anticancer drugs, new treatment protocols, and advances in radiation techniques. The low efficacy of therapy, and short interval between remission and recurrence, could be attributed to the resistance of a small fraction of tumorigenic cells to treatment. The existence and importance of cancer stem cells (CSCs) is perceived by some as controversial. Experimental evidences suggest that the presence of therapy-resistant glioblastoma stem cells (GSCs) could explain tumor recurrence and metastasis. Some scientists, including most of the authors of this review, believe that GSCs are the driving force behind GBM relapses, whereas others however, question the existence of GSCs. Evidence has accumulated indicating that non-tumorigenic cancer cells with high heterogeneity, could undergo reprogramming and become GSCs. Hence, targeting GSCs as the "root cells" initiating malignancy has been proposed to eradicate this devastating disease. Most standard treatments fail to completely eradicate GSCs, which can then cause the recurrence of the disease. To effectively target GSCs, a comprehensive understanding of the biology of GSCs as well as the mechanisms by which these cells survive during treatment and develop into new tumor, is urgently needed. Herein, we provide an overview of the molecular features of GSCs, and elaborate how to facilitate their detection and efficient targeting for therapeutic interventions. We also discuss GBM classifications based on the molecular stem cell subtypes with a focus on potential therapeutic approaches.
Collapse
Affiliation(s)
- Farzaneh Sharifzad
- Department of Applied Cell Sciences, Kashan University of Medical Sciences, Kashan, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saeid Ghavami
- Department of Human Anatomy & Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada; Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, Canada
| | - Javad Verdi
- Department of Applied Cell Sciences, Kashan University of Medical Sciences, Kashan, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Soura Mardpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Zahra Azizi
- Heart Rhythm Program, Southlake Regional Health Centre, Toronto ON Canada
| | - Adeleh Taghikhani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Immunology, Faculty of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| | - Marek J Łos
- Biotechnology Center, Silesian University of Technology in Gliwice, Poland
| | - Esmail Fakharian
- Department of Neurosurgery, Kashan University of Medical Sciences, Kashan, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Amir Ali Hamidieh
- Pediatric Stem Cell Transplant Department, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Hua M, Qin Y, Sheng M, Cui X, Chen W, Zhong J, Yan J, Chen Y. miR‑145 suppresses ovarian cancer progression via modulation of cell growth and invasion by targeting CCND2 and E2F3. Mol Med Rep 2019; 19:3575-3583. [PMID: 30864742 PMCID: PMC6471561 DOI: 10.3892/mmr.2019.10004] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 01/28/2019] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNA/miRs) have been demonstrated to be critical post‑transcriptional modulators of gene expression during tumorigenesis. Numerous miRNAs have been revealed to be downregulated in human epithelial ovarian cancer (EOC). In the present study, it was observed that the expression of miR‑145 was decreased in EOC tissues and cell lines. Overexpression of miR‑145 inhibited the proliferation, migration and invasion of EOC cells. The D‑type cyclin 2, cyclin D2 (CCND2), and E2F transcription factor 3 (E2F3) were confirmed to be targets of miR‑145. In addition, restoration of these 2 genes significantly reversed the tumor suppressive effects of miR‑145. Collectively, the results indicated that miR‑145 serves a critical role in suppressing the biological behavior of EOC cells by targeting CCND2 and E2F3. Therefore, miR‑145 was suggested to be a potential miRNA‑based therapeutic target in ovarian cancer.
Collapse
Affiliation(s)
- Minhui Hua
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yongwei Qin
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Meihong Sheng
- Department of Radiology, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiaopeng Cui
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Weiguan Chen
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jianxin Zhong
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Junming Yan
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yan Chen
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
20
|
Xu L, Zhang Y, Tang J, Wang P, Li L, Yan X, Zheng X, Ren S, Zhang M, Xu M. The Prognostic Value and Regulatory Mechanisms of microRNA-145 in Various Tumors: A Systematic Review and Meta-analysis of 50 Studies. Cancer Epidemiol Biomarkers Prev 2019; 28:867-881. [PMID: 30602498 DOI: 10.1158/1055-9965.epi-18-0570] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/16/2018] [Accepted: 12/26/2018] [Indexed: 02/05/2023] Open
Abstract
Acting as an important tumor-related miRNA, the clinical significance and underlying mechanisms of miR-145 in various malignant tumors have been investigated by numerous studies. This study aimed to comprehensively estimate the prognostic value and systematically illustrate the regulatory mechanisms of miR-145 based on all eligible literature.Relevant studies were acquired from multiple online databases. Overall survival (OS) and progression-free survival (PFS) were used as primary endpoints. Detailed subgroup analyses were performed to decrease the heterogeneity among studies and recognize the prognostic value of miR-145. All statistical analyses were performed with RevMan software version 5.3 and STATA software version 14.1. A total of 48 articles containing 50 studies were included in the meta-analysis. For OS, the pooled results showed that low miR-145 expression in tumor tissues was significantly associated with worse OS in patients with various tumors [HR = 1.70; 95% confidence interval (CI), 1.46-1.99; P < 0.001). Subgroup analysis based on tumor type showed that the downregulation of miR-145 was associated with unfavorable OS in colorectal cancer (HR = 2.17; 95% CI, 1.52-3.08; P < 0.001), ovarian cancer (HR = 2.15; 95% CI, 1.29-3.59; P = 0.003), gastric cancer (HR = 1.78; 95% CI, 1.35-2.36; P < 0.001), glioma (HR = 1.65; 95% CI, 1.30-2.10; P < 0.001), and osteosarcoma (HR = 2.28; 95% CI, 1.50-3.47; P < 0.001). For PFS, the pooled results also showed that the downregulation of miR-145 was significantly associated with poor PFS in patients with multiple tumors (HR = 1.39; 95% CI, 1.16-1.67; P < 0.001), and the subgroup analyses further identified that the low miR-145 expression was associated with worse PFS in patients with lung cancer (HR = 1.97; 95% CI, 1.25-3.09; P = 0.003) and those of Asian descent (HR = 1.50; 95% CI, 1.23-1.82; P < 0.001). For the regulatory mechanisms, we observed that numerous tumor-related transcripts could be targeted by miR-145-5p or miR-145-3p, as well as the expression and function of miR-145-5p could be regulated by multiple molecules.This meta-analysis indicated that downregulated miR-145 in tumor tissues or peripheral blood predicted unfavorable prognostic outcomes for patients suffering from various malignant tumors. In addition, miR-145 was involved in multiple tumor-related pathways and the functioning of significant biological effects. miR-145 is a well-demonstrated tumor suppressor, and its expression level is significantly correlated with the prognosis of patients with multiple malignant tumors.
Collapse
Affiliation(s)
- Liangliang Xu
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yanfang Zhang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jianwei Tang
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Peng Wang
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Lian Li
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiaokai Yan
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiaobo Zheng
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Shengsheng Ren
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ming Zhang
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mingqing Xu
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
21
|
Clinical Significance of Serum NEDD9 Levels in Patients with Pancreatic Cancer. Biomolecules 2018; 8:biom8040169. [PMID: 30544746 PMCID: PMC6316687 DOI: 10.3390/biom8040169] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/26/2018] [Accepted: 12/03/2018] [Indexed: 12/11/2022] Open
Abstract
Introduction: Pancreatic cancer (PC) is a lethal malignancy. Various diagnostic, predictive, and prognostic biomarkers have been evaluated. This study was conducted to investigate the serum levels of neural precursor cell expressed developmentally downregulated protein 9 (NEDD9) in patients with PC and the relationship between tumor progression and known prognostic parameters. Materials and Methods: Serum samples were obtained on first admission before any treatment. Serum NEDD9 levels were determined using enzyme-linked immunosorbent assay (ELISA). Age- and sex-matched healthy controls were included in the analysis. Results: In a three year period, 32 patients with a pathologically-confirmed diagnosis of PC were enrolled in this study. The median age at diagnosis was 61 years, range 38 to 84 years; the majority of the patients in the group were men (n = 20, 62.5%). The tumor was located in the head of pancreas in 21 (65.6%) patients. Forty-one percent of 17 metastatic patients who received palliative CTx (chemotherapy) were CTx-responsive. The baseline serum NEDD9 levels were significantly higher in patients with PA than in the control group (p = 0.03). Median OS of the whole group were 27 ± 7.3 weeks. Alcohol intake, performance status, and LDH levels were found to be significant prognostic factors (p = 0.006, p < 0.001, and p < 0.001, respectively). However, serum NEDD9 levels had no significantly effect on progression free survival (PFS) and overall survival (OS) (p = 0.71 and p = 0.58, respectively). Conclusions: NEDD9 is identified as a secretory biomarker for PC but it has no prognostic role.
Collapse
|
22
|
Regulatory mechanisms of miR-145 expression and the importance of its function in cancer metastasis. Biomed Pharmacother 2018; 109:195-207. [PMID: 30396077 DOI: 10.1016/j.biopha.2018.10.037] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/05/2018] [Accepted: 10/09/2018] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs are post-transcriptional mediators of gene expression and regulation, which play influential roles in tumorigenesis and cancer metastasis. The expression of tumor suppressor miR-145 is reduced in various cancer cell lines, containing both solid tumors and blood malignancies. However, the responsible mechanisms of its down-regulation are a complicated network. miR-145 is potentially able to inhbit tumor cell metastasis by targeting of multiple oncogenes, including MUC1, FSCN1, Vimentin, Cadherin, Fibronectin, Metadherin, GOLM1, ARF6, SMAD3, MMP11, Snail1, ZEB1/2, HIF-1α and Rock-1. This distinctive role of miR-145 in the regulation of metastasis-related gene expression may introduce miR-145 as an ideal candidate for controlling of cancer metastasis by miRNA replacement therapy. The present review aims to discuss the current understanding of the different aspects of molecular mechanisms of miR-145 regulation as well as its role in r metastasis regulation.
Collapse
|
23
|
Salem SM, Hamed AR, Fayez AG, Nour Eldeen G. Non-target Genes Regulate miRNAs-Mediated Migration Steering of Colorectal Carcinoma. Pathol Oncol Res 2018; 25:559-566. [PMID: 30361904 DOI: 10.1007/s12253-018-0502-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 10/15/2018] [Indexed: 12/14/2022]
Abstract
MicroRNAs (miRNAs) trigger a two-layer regulatory network directly or through transcription factors and their co-regulators. Unlike miR-375, the role of miR-145 and miR-224 in inhibiting or driving cancer cell migration is controversial. This study is a step towards addressing the potential of miR-375, miR-145 and miR-224 expression modulation to inhibit colorectal carcinoma (CRC) cells migration in vitro through regulation of non-target genes VEGFA, TGFβ1, IGF1, CD105 and CD44. Transwell migration assay results revealed a significant subdue of migration ability of cells transfected with miR-375 and miR-145 mimics and miR-224 inhibitor. Real time PCR data showed that expression of VEGFA, TGFβ1, IGF1, CD105 and CD44 was downregulated as a consequence of exogenous re-expression of miR-375 and inhibition of miR-224. On the other hand, ectopic expression of miR-145 did not affect VEGFA, TGFβ1 and CD44 expression, while it elevated CD105 and suppressed IGF1 expression. MAP4K4, a predicted target of miR-145, was validated as a target that could play a role in miR-145-mediated regulation of migration. At mRNA level, no change was observed in expression of MAP4K4 in cells with restored expression of miR-145, while western blotting analysis revealed a 25% reduction of protein level. By applying luciferase reporter assay, a significant decrease in luciferase activity was observed, supporting that miR-145 directly target 3' UTR of MAP4K4. The study highlighted the involvement of non-target genes VEGFA, TGFβ1, IGF1, CD105 and CD44 in mediating anti- and pro-migratory effect of miR-375 and miR-224, respectively, and validated MAP4K4 as a direct target of anti-migratory miR-145.
Collapse
Affiliation(s)
- Sohair M Salem
- Molecular Genetics and Enzymology Department, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt.
| | - Ahmed R Hamed
- Phytochemistry Department, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt.,Biology Unit - Central Laboratory of Pharmaceutical and Drug Industries Research Division, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt
| | - Alaaeldin G Fayez
- Molecular Genetics and Enzymology Department, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt
| | - Ghada Nour Eldeen
- Molecular Genetics and Enzymology Department, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt.,Stem Cell Research Unit, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt
| |
Collapse
|
24
|
Glowacka WK, Jain H, Okura M, Maimaitiming A, Mamatjan Y, Nejad R, Farooq H, Taylor MD, Aldape K, Kongkham P. 5-Hydroxymethylcytosine preferentially targets genes upregulated in isocitrate dehydrogenase 1 mutant high-grade glioma. Acta Neuropathol 2018; 135:617-634. [PMID: 29428975 PMCID: PMC5978937 DOI: 10.1007/s00401-018-1821-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/18/2018] [Accepted: 02/07/2018] [Indexed: 01/12/2023]
Abstract
Gliomas demonstrate epigenetic dysregulation exemplified by the Glioma CpG Island Methylator Phenotype (G-CIMP) seen in IDH1 mutant tumors. 5-Hydroxymethylcytosine (5hmC) is implicated in glioma pathogenesis; however, its role in IDH1 mutant gliomas is incompletely understood. To characterize 5hmC in IDH1 mutant gliomas further, we examine 5hmC in a cohort of IDH1 mutant and wild-type high-grade gliomas (HGG) using a quantitative locus-specific approach. Regions demonstrating high 5hmC abundance and differentially hydroxymethylated regions (DHMR) enrich for enhancers implicated in glioma pathogenesis. Among these regions, IDH1 mutant tumors possess greater 5hmC compared to wild type. 5hmC contributes to overall methylation status of G-CIMP genes. 5hmC targeting gene body regions correlates significantly with increased gene expression. In particular, a strong correlation between increased 5hmC and increased gene expression is identified for genes highly expressed in the IDH1 mutant cohort. Overall, locus-specific gain of 5hmC targeting regulatory regions and associated with overexpressed genes suggests a significant role for 5hmC in IDH1 mutant HGG.
Collapse
|
25
|
Epigenetic modification of miR-141 regulates SKA2 by an endogenous 'sponge' HOTAIR in glioma. Oncotarget 2017; 7:30610-25. [PMID: 27121316 PMCID: PMC5058705 DOI: 10.18632/oncotarget.8895] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 03/31/2016] [Indexed: 12/21/2022] Open
Abstract
Aberrant expression of miR-141 has recently implicated in the occurrence and development of various types of malignant tumors. However whether the involvement of miR-141 in the pathogenesis of glioma remains unknown. Here, we showed that miR-141 was markedly downregulated in glioma tissues and cell lines compared with normal brain tissues, and its expression correlated with the pathological grading. Enforced expression of miR-141 in glioma cells significantly inhibited cell proliferation, migration and invasion, whereas knockdown of miR-141 exerted opposite effect. Mechanistic investigations revealed that HOTAIR might act as an endogenous 'sponge' of miR-141, thereby regulating the derepression of SKA2. Further, we explored the molecular mechanism by which miR-141 expression was regulated, and found that the miR-141 promoter was hypermethylated and that promoter methylation of miR-141 was mediated by DNMT1 in glioma cells. Finally, both overexpression of miR-141 and knockdown of HOTAIR in a mouse model of human glioma resulted in significant reduction of tumor growth in vivo. Collectively, these results suggest that epigenetic modification of miR-141 and the interaction of ceRNA regulatory network will provide a new approach for therapeutics against glioma.
Collapse
|
26
|
Abstract
Glioblastoma is the most aggressive brain tumor and, even with the current multimodal therapy, is an invariably lethal cancer with a life expectancy that depends on the tumor subtype but, even in the most favorable cases, rarely exceeds 2 years. Epigenetic factors play an important role in gliomagenesis, are strong predictors of outcome, and are important determinants for the resistance to radio- and chemotherapy. The latest addition to the epigenetic machinery is the noncoding RNA (ncRNA), that is, RNA molecules that are not translated into a protein and that exert their function by base pairing with other nucleic acids in a reversible and nonmutational mode. MicroRNAs (miRNA) are a class of ncRNA of about 22 bp that regulate gene expression by binding to complementary sequences in the mRNA and silence its translation into proteins. MicroRNAs reversibly regulate transcription through nonmutational mechanisms; accordingly, they can be considered as epigenetic effectors. In this review, we will discuss the role of miRNA in glioma focusing on their role in drug resistance and on their potential applications in the therapy of this tumor.
Collapse
|
27
|
Yang L, Mu Y, Cui H, Liang Y, Su X. MiR-9-3p augments apoptosis induced by H2O2 through down regulation of Herpud1 in glioma. PLoS One 2017; 12:e0174839. [PMID: 28430789 PMCID: PMC5400238 DOI: 10.1371/journal.pone.0174839] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 03/03/2017] [Indexed: 12/25/2022] Open
Abstract
MicroRNAs are short, single-stranded non-coding RNA molecules that function as regulators of tumor progression in various cancers, including glioma. The present study sought to investigate the biological functions of miR-9-3p in glioma progression. The results of a microRNA microarray indicated that microRNA-9-3p (miR-9-3p, miR-9*) is down-regulated in high-grade (grades III and IV) gliomas compared with non-tumor tissues. These results were confirmed with real-time PCR. The miR-9-3p expression level was associated with age and tumor grade. Herpud1 was regulated by miR-9-3p in glioma cells and tissues and was identified as a miR-9-3p target with luciferase reporter assays. Glioma cells transfected with miR-9-3p mimics or HERPUD1-RNAi had more apoptotic cells than them in control after induced by H2O2. Our results indicated that low expression of miR-9-3p results in a high level of Herpud1, which may protect against apoptosis in glioma.
Collapse
Affiliation(s)
- Ling Yang
- Clinical Medicine Research Center, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Yongping Mu
- Department of Clinical Laboratory, the Affiliated People’s Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Hongwei Cui
- Clinical Medicine Research Center, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Yabing Liang
- Clinical Medicine Research Center, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Xiulan Su
- Clinical Medicine Research Center, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
- * E-mail:
| |
Collapse
|
28
|
Zhao H, Shen J, Hodges TR, Song R, Fuller GN, Heimberger AB. Serum microRNA profiling in patients with glioblastoma: a survival analysis. Mol Cancer 2017; 16:59. [PMID: 28284220 PMCID: PMC5346242 DOI: 10.1186/s12943-017-0628-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/28/2017] [Indexed: 01/23/2023] Open
Abstract
Because circulating microRNAs (miRNAs) have drawn a great deal of attention as promising novel cancer diagnostics and prognostic biomarkers, we sought to identify serum miRNAs significantly associated with outcome in glioblastoma patients. To do this, we performed global miRNA profiling in serum samples from 106 primary glioblastoma patients. The study subjects were randomly divided into two sets: set one (n = 40) and set two (n = 66). Using a Cox regression model, 3 serum miRNAs (miR-106a-5p, miR-182, and miR-145-5p) and 5 serum miRNAs (miR-222-3p, miR-182, miR-20a-5p, miR-106a-5p, and miR-145-5p) were identified significantly associated with 2-year patient overall survival and disease-free survival (P < 0.05) in both sets and the combined set. We then created the miRNA risk scores to assess the total impact of the significant serum miRNAs on survival. The high risk scores were associated with poor patient survival (overall survival: HR = 1.92, 95% CI: 1.19, 10.23, and disease-free survival: HR = 2.03, 95%CI: 1.24, 4.28), and were independent of other clinicopathological factors. Our results suggest that serum miRNAs could serve as prognostic predictors of glioblastoma.
Collapse
Affiliation(s)
- Hua Zhao
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, Houston, TX, 77030, USA.
| | - Jie Shen
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, Houston, TX, 77030, USA
| | - Tiffany R Hodges
- Department of Neuro-Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Renduo Song
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, Houston, TX, 77030, USA
| | - Gregory N Fuller
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Amy B Heimberger
- Department of Neuro-Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
29
|
Synthetic miR-145 Mimic Enhances the Cytotoxic Effect of the Antiangiogenic Drug Sunitinib in Glioblastoma. Cell Biochem Biophys 2017; 72:551-7. [PMID: 25564360 DOI: 10.1007/s12013-014-0501-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Although aggressive therapeutic regimen has been applied in the treatment of Glioblastoma (GBM), the prognosis of patients with GBM remains poor. Preclinical studies have demonstrated the efficacy of Suntinib in GBM both in vitro and in vivo. In this study, we showed that the cytotoxicity was enhanced by transfection with miR-145 mimic. In addition, we suggested that the enhanced cytotoxicity of Sunitinib by miR-145 mimic was mediated by inhibition of both P-gp and Bcrp.
Collapse
|
30
|
The Prognostic Role of NEDD9 and P38 Protein Expression Levels in Urinary Bladder Transitional Cell Carcinoma. JOURNAL OF ONCOLOGY 2017; 2017:6095205. [PMID: 28194179 PMCID: PMC5282419 DOI: 10.1155/2017/6095205] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 10/15/2016] [Accepted: 10/18/2016] [Indexed: 11/18/2022]
Abstract
Background. The most common malignant tumor of the urinary bladder is transitional cell carcinoma (TCC). Neural precursor cell-expressed developmentally downregulated protein 9 (NEDD9) is found to be a cell adhesion mediator. P38 Mitogen-Activated Protein Kinase is a serine/threonine kinases member which can mediate carcinogenesis through intracellular signaling. Methods. To assess their prognostic role; NEDD9 and p38 protein were evaluated in sections from 50 paraffin blocks of TCC. Results. The high expressions of NEDD9 and p38 protein were significantly associated with grade, stage, distant metastasis (p < 0.001), number of tumors, lymph node metastasis, and tumor size (p < 0.001, 0.002; 0.018, <0.001; and 0.004, 0.007, respectively). High NEDD9 and p38 detection had a worse 3-year OS (p = 0.041 and <0.001, respectively). By multivariate analysis the NEDD9 and p38 protein expression levels and various clinicopathological criteria including gender, grade, stage of the tumor, and regional lymph node involvement were independent prognostic parameters of TCC of the urinary bladder patients' outcome. Conclusion. NEDD9 and p38 protein expressions were poor prognostic markers of TCC.
Collapse
|
31
|
Dai J, Van Wie PG, Fai LY, Kim D, Wang L, Poyil P, Luo J, Zhang Z. Downregulation of NEDD9 by apigenin suppresses migration, invasion, and metastasis of colorectal cancer cells. Toxicol Appl Pharmacol 2016; 311:106-112. [PMID: 27664007 DOI: 10.1016/j.taap.2016.09.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 09/16/2016] [Accepted: 09/20/2016] [Indexed: 02/07/2023]
Abstract
Apigenin is a natural flavonoid which possesses multiple anti-cancer properties such as anti-proliferation, anti-inflammation, and anti-metastasis in many types of cancers including colorectal cancer. Neural precursor cell expressed developmentally downregulated 9 (NEDD9) is a multi-domain scaffolding protein of the Cas family which has been shown to correlate with cancer metastasis and progression. The present study investigates the role of NEDD9 in apigenin-inhibited cell migration, invasion, and metastasis of colorectal adenocarcinoma DLD1 and SW480 cells. The results show that knockdown of NEDD9 inhibited cell migration, invasion, and metastasis and that overexpression of NEDD9 promoted cell migration and invasion of DLD1 cells and SW4890 cells. Apigenin treatment attenuated NEDD9 expression at protein level, resulting in reduced phosphorylations of FAK, Src, and Akt, leading to inhibition on cell migration, invasion, and metastasis of both DLD1 and SW480 cells. The present study has demonstrated that apigenin inhibits cell migration, invasion, and metastasis through NEDD9/Src/Akt cascade in colorectal cancer cells. NEDD9 may function as a biomarker for evaluation of cancer aggressiveness and for selection of therapeutic drugs against cancer progression.
Collapse
Affiliation(s)
- Jin Dai
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Peter G Van Wie
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Leonard Yenwong Fai
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Donghern Kim
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Lei Wang
- Center for Research on Environmental Disease, University of Kentucky, Lexington, KY 40536, USA
| | - Pratheeshkumar Poyil
- Center for Research on Environmental Disease, University of Kentucky, Lexington, KY 40536, USA
| | - Jia Luo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Zhuo Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
32
|
Suryo Rahmanto A, Savov V, Brunner A, Bolin S, Weishaupt H, Malyukova A, Rosén G, Čančer M, Hutter S, Sundström A, Kawauchi D, Jones DT, Spruck C, Taylor MD, Cho YJ, Pfister SM, Kool M, Korshunov A, Swartling FJ, Sangfelt O. FBW7 suppression leads to SOX9 stabilization and increased malignancy in medulloblastoma. EMBO J 2016; 35:2192-2212. [PMID: 27625374 PMCID: PMC5069553 DOI: 10.15252/embj.201693889] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 08/18/2016] [Indexed: 12/02/2022] Open
Abstract
SOX9 is a master transcription factor that regulates development and stem cell programs. However, its potential oncogenic activity and regulatory mechanisms that control SOX9 protein stability are poorly understood. Here, we show that SOX9 is a substrate of FBW7, a tumor suppressor, and a SCF (SKP1/CUL1/F‐box)‐type ubiquitin ligase. FBW7 recognizes a conserved degron surrounding threonine 236 (T236) in SOX9 that is phosphorylated by GSK3 kinase and consequently degraded by SCFFBW7α. Failure to degrade SOX9 promotes migration, metastasis, and treatment resistance in medulloblastoma, one of the most common childhood brain tumors. FBW7 is either mutated or downregulated in medulloblastoma, and in cases where FBW7 mRNA levels are low, SOX9 protein is significantly elevated and this phenotype is associated with metastasis at diagnosis and poor patient outcome. Transcriptional profiling of medulloblastoma cells expressing a degradation‐resistant SOX9 mutant reveals activation of pro‐metastatic genes and genes linked to cisplatin resistance. Finally, we show that pharmacological inhibition of PI3K/AKT/mTOR pathway activity destabilizes SOX9 in a GSK3/FBW7‐dependent manner, rendering medulloblastoma cells sensitive to cytostatic treatment.
Collapse
Affiliation(s)
| | - Vasil Savov
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Andrä Brunner
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sara Bolin
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Holger Weishaupt
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Alena Malyukova
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Gabriela Rosén
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Matko Čančer
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Sonja Hutter
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anders Sundström
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Daisuke Kawauchi
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David Tw Jones
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Charles Spruck
- Tumor Initiation and Maintenance Program, Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Michael D Taylor
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Yoon-Jae Cho
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Stefan M Pfister
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, University Hospital, Heidelberg, Germany
| | - Marcel Kool
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andrey Korshunov
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neuropathology, University Hospital, Heidelberg, Germany
| | - Fredrik J Swartling
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Olle Sangfelt
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
33
|
Namkung J, Kwon W, Choi Y, Yi SG, Han S, Kang MJ, Kim SW, Park T, Jang JY. Molecular subtypes of pancreatic cancer based on miRNA expression profiles have independent prognostic value. J Gastroenterol Hepatol 2016; 31:1160-7. [PMID: 26644397 DOI: 10.1111/jgh.13253] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 10/15/2015] [Accepted: 11/16/2015] [Indexed: 01/17/2023]
Abstract
BACKGROUND AND AIM Altered microRNAs (miRNA) expression, a typical feature of many cancers, is reportedly associated with prognosis according to several studies. Although numerous studies on miRNAs in pancreatic ductal adenocarcinoma have also attempted to identify prognostic biomarkers, more large-scale clinical studies are needed to establish the clinical significance of the results. Present study aimed to identify prognosis-related molecular subtypes of primary pancreas tumors using miRNA expression profiling. METHODS Expression profiles of 1733 miRNAs were obtained by using microarray analysis of 104 pancreatic tumors of Korean patients. To detect subgroups informative in predicting the patient's prognosis, we applied unsupervised clustering methods and then analyzed the association of the molecular subgroups with survival time. Then, we constructed a classifier to predict the subgroup using penalized regression models. RESULTS We have determined three pancreatic ductal adenocarcinoma tumor subtypes associated with prognosis based on miRNA expression profiles. These subtypes showed significantly different survival time for patients with the same clinical conditions. This demonstrates that our prognostic molecular subgroup has independent prognostic utility. The molecular subtypes can be predicted with a classifier of 19 miRNAs. Of the 19 signature miRNAs, miR-106b-star, miR-324-3p, and miR-615 were related to a p53 canonical pathway, and miR-324, miR-145-5p, miR-26b-5p, and miR-574-3p were related to a Cox-2 centered pathway. CONCLUSIONS Our prognostic molecular subtypes demonstrated that miRNA profiles could be used as prognostic markers. Additionally, we have constructed a classifier that may be used to determine the molecular subgroup of new patient sample data. Further studies are needed for validation.
Collapse
Affiliation(s)
- Junghyun Namkung
- Bioinformatics Tech. Lab, Healthcare group, Future Technology R&D Division, SK Telecom, Co., Seoul, Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yonwhan Choi
- Bioinformatics Tech. Lab, Healthcare group, Future Technology R&D Division, SK Telecom, Co., Seoul, Korea
| | - Sung Gon Yi
- Bioinformatics Tech. Lab, Healthcare group, Future Technology R&D Division, SK Telecom, Co., Seoul, Korea
| | - Sangjo Han
- Bioinformatics Tech. Lab, Healthcare group, Future Technology R&D Division, SK Telecom, Co., Seoul, Korea
| | - Mee Joo Kang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Sun-Whe Kim
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Taesung Park
- Department of Statistics, Seoul National University, Seoul, Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
34
|
Brito BDL, Lourenço SV, Damascena AS, Kowalski LP, Soares FA, Coutinho-Camillo CM. Expression of stem cell-regulating miRNAs in oral cavity and oropharynx squamous cell carcinoma. J Oral Pathol Med 2016; 45:647-654. [PMID: 26841253 DOI: 10.1111/jop.12424] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is the sixth most common tumor worldwide and is histologically heterogeneous. Studies have demonstrated the presence of stem cell markers in HNSCC, and microRNAs (miRNAs) have emerged as powerful regulators of differentiation, controlling the self-renewal of stem cells. miRNAs are non-coding RNA molecules that regulate gene expression post-transcriptionally. Many miRNAs have been described as regulators of stem cells in different types of cancer. METHODS We have analyzed the expression of let-7a, miR-34, miR-125b, miR-138, miR-145, miR-183, miR-200b, miR-203, and miR-205 by real-time RT-PCR (qPCR), in 35 oral cavity and oropharynx squamous cell carcinoma (SCC) samples and 10 non-neoplastic oral mucosa controls, to determine possible associations between the expression of these miRNAs and clinical and pathological features of these tumors. RESULTS We observed downregulation of miR-200b and miR-203 in 60.0% and 71.4% of the samples, respectively. Upregulation of miR-138 and miR-183 was observed in 50.0% of the samples. Downregulation of let-7a was associated with perineural invasion. Upregulation of miR-138, miRNA-145, and miR-205 was associated with advanced tumor stages, vascular invasion, and lymph node metastasis, respectively. CONCLUSIONS Our study provides evidence of the expression of miRNAs associated with stem cell regulation in oral cavity and oropharynx SCC and the association of these miRNAs with clinical and pathological features of these tumors.
Collapse
Affiliation(s)
- Bárbara de Lima Brito
- International Research Center, A.C. Camargo Cancer Center, São Paulo, SP, Brazil.,Department of Anatomic Pathology, A.C. Camargo Cancer Center, São Paulo, SP, Brazil
| | - Silvia Vanessa Lourenço
- Department of General Pathology, Dental School, University of São Paulo, São Paulo, SP, Brazil
| | | | - Luiz Paulo Kowalski
- Department of Head and Neck Surgery and Otorhinolaryngology, AC Camargo Cancer Center, São Paulo, SP, Brazil
| | - Fernando Augusto Soares
- Department of Anatomic Pathology, A.C. Camargo Cancer Center, São Paulo, SP, Brazil.,Department of General Pathology, Dental School, University of São Paulo, São Paulo, SP, Brazil
| | - Cláudia Malheiros Coutinho-Camillo
- International Research Center, A.C. Camargo Cancer Center, São Paulo, SP, Brazil. .,Department of Anatomic Pathology, A.C. Camargo Cancer Center, São Paulo, SP, Brazil.
| |
Collapse
|
35
|
Xue M, Zhao L, Yang F, Li Z, Li G. MicroRNA‑145 inhibits the malignant phenotypes of gastric carcinoma cells via downregulation of fascin 1 expression. Mol Med Rep 2015; 13:1033-9. [PMID: 26647829 DOI: 10.3892/mmr.2015.4609] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 11/05/2015] [Indexed: 11/06/2022] Open
Abstract
MicroRNA (miR)‑145 has been demonstrated to act as a tumor suppressor, and deregulation of fascin 1 (FSCN1) has been observed in several types of human malignancy, including gastric carcinoma. However, the molecular mechanism underlying the function of miR‑145, specifically its targets in gastric carcinoma have yet to be fully elucidated. In the present study, downregulation of miR‑145 and upregulation of FSCN1 was identified in gastric carcinoma cell lines, compared with normal gastric mucosal epithelial cells. A luciferase reporter assay demonstrated that miR‑145 was able to bind to the 3'‑untranslated region of FSCN1 mRNA. Overexpression of miR‑145 led to a significant decrease in FSCN1 expression levels, whereas knockdown of miR‑145 resulted in increased FSCN1 expression levels in gastric carcinoma cells. Furthermore, overexpression of miR‑145 inhibited proliferation, migration and invasion in gastric carcinoma cells. Similar effects were also observed in gastric carcinoma cells transfected with FSCN1 small interfering RNA. In addition, overexpression of FSCN1 reversed the suppressive effects of miR‑145 upregulation on proliferation, migration and invasion in gastric carcinoma cells, suggesting that FSCN1 is indeed involved in the miR‑145‑mediated malignant phenotype of gastric carcinoma cells. The present study revealed an anti‑oncogenic role of miR‑145 in gastric carcinoma via inhibition of FSCN1, and suggested that miR‑145 may be used for the treatment of gastric carcinoma.
Collapse
Affiliation(s)
- Minghui Xue
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Lunde Zhao
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Fang Yang
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Zhenjuan Li
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Guangyan Li
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| |
Collapse
|
36
|
Yan W, Li R, Liu Y, Yang P, Wang Z, Zhang C, Bao Z, Zhang W, You Y, Jiang T. MicroRNA expression patterns in the malignant progression of gliomas and a 5-microRNA signature for prognosis. Oncotarget 2015; 5:12908-15. [PMID: 25415048 PMCID: PMC4350338 DOI: 10.18632/oncotarget.2679] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 11/04/2014] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) are directly involved in the progression in various cancers. To date, no systematic researches have been performed on the expression pattern of miRNA during progression from low grade gliomas to anaplastic gliomas or secondary glioblastomas and those prognostic miRNAs in anaplastic gliomas and secondary glioblastomas. In the present study, high-throughput microarrays were used to measure miRNA expression levels in 116 samples in the different progression stages of glioma. We found that miRNA expression pattern totally altered when low grade gliomas progressed to anaplastic gliomas or secondary glioblastomas. However, anaplastic gliomas and secondary glioblastomas have similar expression pattern in miRNA level. Furthermore, we developed a five-miRNA signature (two protective miRNAs-miR-767-5p, miR-105; three risky miRNAs: miR-584, miR-296-5p and miR-196a) that could identify patients with a high risk of unfavorable outcome in anaplastic gliomas regardless of histology type. It should be highlighted that the five-miRNA signature can also identify patients who had a high risk of unfavorable outcome in secondary and TCGA Proneural glioblastomas, but not Neural, Classical and Mesenchymal glioblastomas. Taken together, our results demonstrate that miRNA expression patterns in the malignant progression of gliomas and a novel prognostic classifier, the five-miRNA signature, serve as a prognostic marker for patient risk stratification in anaplastic gliomas, Secondary and Proneural glioblastomas.
Collapse
Affiliation(s)
- Wei Yan
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China. Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Rui Li
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Yanwei Liu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China. Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Pei Yang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China. Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Zheng Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China. Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Chuanbao Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China. Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Zhaoshi Bao
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China. Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Wei Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China. Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Yongping You
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Tao Jiang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China. Beijing Institute for Brain Disorders Brain Tumor Center, Beijing, PR China. Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| |
Collapse
|
37
|
Das AV, Pillai RM. Implications of miR cluster 143/145 as universal anti-oncomiRs and their dysregulation during tumorigenesis. Cancer Cell Int 2015; 15:92. [PMID: 26425114 PMCID: PMC4588501 DOI: 10.1186/s12935-015-0247-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/22/2015] [Indexed: 02/07/2023] Open
Abstract
Tumorigenesis is a multistep process, de-regulated due to the imbalance of oncogenes as well as anti-oncogenes, resulting in disruption of tissue homeostasis. In many cases the effect of oncogenes and anti-oncogenes are mediated by various other molecules such as microRNAs. microRNAs are small non-coding RNAs established to post-transcriptionally regulate more than half of the protein coding genes. miR cluster 143/145 is one such cancer-related microRNA cluster which is down-regulated in most of the cancers and is able to hinder tumorigenesis by targeting tumor-associated genes. The fact that they could sensitize drug-resistant cancer cells by targeting multidrug resistant genes makes them potent tools to target cancer cells. Their low levels precede events which lead to cancer progression and therefore could be considered also as biomarkers to stage the disease. Interestingly, evidence suggests the existence of several in vivo mechanisms by which this cluster is differentially regulated at the molecular level to keep their levels low in cancer. In this review, we summarize the roles of miR cluster 143/145 in cancer, their potential prognostic applications and also their regulation during tumorigenesis.
Collapse
Affiliation(s)
- Ani V Das
- Cancer Research Program-9, Rajiv Gandhi Centre for Biotechnology, Thycaud.P.O., Thiruvananthapuram-14, Kerala India
| | - Radhakrishna M Pillai
- Cancer Research Program-9, Rajiv Gandhi Centre for Biotechnology, Thycaud.P.O., Thiruvananthapuram-14, Kerala India
| |
Collapse
|
38
|
Alvarado AG, Turaga SM, Sathyan P, Mulkearns-Hubert EE, Otvos B, Silver DJ, Hale JS, Flavahan WA, Zinn PO, Sinyuk M, Li M, Guda MR, Velpula KK, Tsung AJ, Nakano I, Vogelbaum MA, Majumder S, Rich JN, Lathia JD. Coordination of self-renewal in glioblastoma by integration of adhesion and microRNA signaling. Neuro Oncol 2015; 18:656-66. [PMID: 26374689 DOI: 10.1093/neuonc/nov196] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 08/10/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) provide an additional layer of complexity for tumor models and targets for therapeutic development. The balance between CSC self-renewal and differentiation is driven by niche components including adhesion, which is a hallmark of stemness. While studies have demonstrated that the reduction of adhesion molecules, such as integrins and junctional adhesion molecule-A (JAM-A), decreases CSC maintenance. The molecular circuitry underlying these interactions has yet to be resolved. METHODS MicroRNA screening predicted that microRNA-145 (miR-145) would bind to JAM-A. JAM-A overexpression in CSCs was evaluated both in vitro (proliferation and self-renewal) and in vivo (intracranial tumor initiation). miR-145 introduction into CSCs was similarly assessed in vitro. Additionally, The Cancer Genome Atlas dataset was evaluated for expression levels of miR-145 and overall survival of the different molecular groups. RESULTS Using patient-derived glioblastoma CSCs, we confirmed that JAM-A is suppressed by miR-145. CSCs expressed low levels of miR-145, and its introduction decreased self-renewal through reductions in AKT signaling and stem cell marker (SOX2, OCT4, and NANOG) expression; JAM-A overexpression rescued these effects. These findings were predictive of patient survival, with a JAM-A/miR-145 signature robustly predicting poor patient prognosis. CONCLUSIONS Our results link CSC-specific niche signaling to a microRNA regulatory network that is altered in glioblastoma and can be targeted to attenuate CSC self-renewal.
Collapse
Affiliation(s)
- Alvaro G Alvarado
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Soumya M Turaga
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Pratheesh Sathyan
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Erin E Mulkearns-Hubert
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Balint Otvos
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Daniel J Silver
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - James S Hale
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - William A Flavahan
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Pascal O Zinn
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Maksim Sinyuk
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Meizhang Li
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Maheedhara R Guda
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Kiran K Velpula
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Andrew J Tsung
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Ichiro Nakano
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Michael A Vogelbaum
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Sadhan Majumder
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Jeremy N Rich
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (A.G.A., S.M.T., E.E.M.-H., B.O., D.J.S., J.S.H., M.S., M.L., J.D.L.); Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (A.G.A., J.N.R., J.D.L.); Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, Texas (P.S., P.O.Z., S.M.); Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (W.A.F., J.N.R.); Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China (M.L.); Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois (M.R.G., K.K.V., A.J.T.); Department of Neurological Surgery, The Ohio State University, Columbus, Ohio (I.N.); Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., J.N.R., J.D.L.)
| |
Collapse
|
39
|
Xue L, Wang Y, Yue S, Zhang J. Low MiR-149 expression is associated with unfavorable prognosis and enhanced Akt/mTOR signaling in glioma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:11178-11184. [PMID: 26617839 PMCID: PMC4637654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/20/2015] [Indexed: 06/05/2023]
Abstract
MicroRNAs (miRs) play critical roles in the progression of glioma. Previous in vitro studies have described the anti-tumor role of miR-149 in cancer cells including glioma. In this study, we aimed to investigate whether miR-149 is associated with the prognosis of glioma patients. A total of 163 glioma patients who underwent tumor resection were included in our follow-up study. We found that the miR-149 expression was significantly lower in tumor tissues compared with that in normal tissues (P<0.05). Kaplan-Meier and analysis showed that the miR-149 expression status was significantly associated with the survival duration (logrank test, P<0.001), and multivariate Cox regression revealed that patients with low miR-149 expression were exposed to a 1.825 fold higher death risk (HR=1.825, 95% CI=1.031-3.229, P=0.039) compared with those with high miR-149 expression. Further study showed that Akt/mTOR signaling was hyperactive in low miR-149 expressing tissues. Our study thus demonstrates that miR-149 expression in glioma tissues is critically associated with the prognosis of patients, suggesting its potential clinical significance.
Collapse
Affiliation(s)
- Liang Xue
- Department of Neurosurgery, Tianjin Medical University General Hospital Tianjin 300052, P. R. China
| | - Yi Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital Tianjin 300052, P. R. China
| | - Shuyuan Yue
- Department of Neurosurgery, Tianjin Medical University General Hospital Tianjin 300052, P. R. China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital Tianjin 300052, P. R. China
| |
Collapse
|
40
|
ZHANG SISEN, WU LIHUA. Roles of neural precursor cell expressed, developmentally downregulated 9 in tumor-associated cellular processes (Review). Mol Med Rep 2015; 12:6415-21. [DOI: 10.3892/mmr.2015.4240] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 06/15/2015] [Indexed: 11/05/2022] Open
|
41
|
Shagisultanova E, Gaponova AV, Gabbasov R, Nicolas E, Golemis EA. Preclinical and clinical studies of the NEDD9 scaffold protein in cancer and other diseases. Gene 2015; 567:1-11. [PMID: 25967390 DOI: 10.1016/j.gene.2015.04.086] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 04/20/2015] [Accepted: 04/21/2015] [Indexed: 12/17/2022]
Abstract
Cancer progression requires a significant reprogramming of cellular signaling to support the essential tumor-specific processes that include hyperproliferation, invasion (for solid tumors) and survival of metastatic colonies. NEDD9 (also known as CasL and HEF1) encodes a multi-domain scaffolding protein that assembles signaling complexes regulating multiple cellular processes relevant to cancer. These include responsiveness to signals emanating from the T and B cell receptors, integrins, chemokine receptors, and receptor tyrosine kinases, as well as cytoplasmic oncogenes such as BCR-ABL and FAK- and SRC-family kinases. Downstream, NEDD9 regulation of partners including CRKL, WAVE, PI3K/AKT, ERK, E-cadherin, Aurora-A (AURKA), HDAC6, and others allow NEDD9 to influence functions as pleiotropic as migration, invasion, survival, ciliary resorption, and mitosis. In this review, we summarize a growing body of preclinical and clinical data that indicate that while NEDD9 is itself non-oncogenic, changes in expression of NEDD9 (most commonly elevation of expression) are common features of tumors, and directly impact tumor aggressiveness, metastasis, and response to at least some targeted agents inhibiting NEDD9-interacting proteins. These data strongly support the relevance of further development of NEDD9 as a biomarker for therapeutic resistance. Finally, we briefly discuss emerging evidence supporting involvement of NEDD9 in additional pathological conditions, including stroke and polycystic kidney disease.
Collapse
Affiliation(s)
- Elena Shagisultanova
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Anna V Gaponova
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Rashid Gabbasov
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; Department of Genetics, Kazan Federal University (Volga Region), Kazan, Tatarstan, Russia
| | - Emmanuelle Nicolas
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Erica A Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| |
Collapse
|
42
|
Doloff JC, Waxman DJ. Transcriptional profiling provides insights into metronomic cyclophosphamide-activated, innate immune-dependent regression of brain tumor xenografts. BMC Cancer 2015; 15:375. [PMID: 25952672 PMCID: PMC4523019 DOI: 10.1186/s12885-015-1358-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 04/23/2015] [Indexed: 02/07/2023] Open
Abstract
Background Cyclophosphamide treatment on a six-day repeating metronomic schedule induces a dramatic, innate immune cell-dependent regression of implanted gliomas. However, little is known about the underlying mechanisms whereby metronomic cyclophosphamide induces innate immune cell mobilization and recruitment, or about the role of DNA damage and cell stress response pathways in eliciting the immune responses linked to tumor regression. Methods Untreated and metronomic cyclophosphamide-treated human U251 glioblastoma xenografts were analyzed on human microarrays at two treatment time points to identify responsive tumor cell-specific factors and their upstream regulators. Mouse microarray analysis across two glioma models (human U251, rat 9L) was used to identify host factors and gene networks that contribute to the observed immune and tumor regression responses. Results Metronomic cyclophosphamide increased expression of tumor cell-derived DNA damage, cell stress, and cell death genes, which may facilitate innate immune activation. Increased expression of many host (mouse) immune networks was also seen in both tumor models, including complement components, toll-like receptors, interferons, and cytolysis pathways. Key upstream regulators activated by metronomic cyclophosphamide include members of the interferon, toll-like receptor, inflammatory response, and PPAR signaling pathways, whose activation may contribute to anti-tumor immunity. Many upstream regulators inhibited by metronomic cyclophosphamide, including hypoxia-inducible factors and MAP kinases, have glioma-promoting activity; their inhibition may contribute to the therapeutic effectiveness of the six-day repeating metronomic cyclophosphamide schedule. Conclusions Large numbers of responsive cytokines, chemokines and immune regulatory genes linked to innate immune cell recruitment and tumor regression were identified, as were several immunosuppressive factors that may contribute to the observed escape of some tumors from metronomic CPA-induced, immune-based regression. These factors may include useful biomarkers that facilitate discovery of clinically effective immunogenic metronomic drugs and treatment schedules, and the selection of patients most likely to be responsive to immunogenic drug scheduling. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1358-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joshua C Doloff
- Department of Biology, Division of Cell and Molecular Biology, Boston University, Boston, USA.
| | - David J Waxman
- Department of Biology, Division of Cell and Molecular Biology, Boston University, Boston, USA.
| |
Collapse
|
43
|
Han T, Yi XP, Liu B, Ke MJ, Li YX. MicroRNA-145 suppresses cell proliferation, invasion and migration in pancreatic cancer cells by targeting NEDD9. Mol Med Rep 2015; 11:4115-20. [PMID: 25646678 PMCID: PMC4394956 DOI: 10.3892/mmr.2015.3294] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 01/07/2015] [Indexed: 11/16/2022] Open
Abstract
MicroRNAs (miRNAs) represent a class of small non-coding RNAs regulating gene expression by inducing the degradation of RNA or interfering with translation. Aberrant miRNA expression has been described in several types of cancer in humans. In the present study, it was demonstrated that miR-145 is downregulated in pancreatic cancer tissues and the Panc-1 cell line. Restoration of miR-145 inhibited cell proliferation, invasion and migration in Panc-1 cells. Neural precursor cell expressed, developmentally down-regulated 9 (NEDD9) has been identified as a novel potential miR-145 target using bioinformatics. Using luciferase reporter constructs, it was observed that the NEDD9 3′-untranslated region is the location of the direct binding site for miR-145. Additionally, it was identified that miR-145 is inversely correlated with NEDD9 expression in pancreatic cancer tissues and that restoration of miR-145 in Panc-1 cells reduced NEDD9 mRNA and protein expression accompanied by inhibition of cell proliferation, invasion and migration. In conclusion, these findings indicate that miR-145 may be an effective target for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Tong Han
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiao-Ping Yi
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Bo Liu
- State Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan 410008, P.R. China
| | - Mu-Jing Ke
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yi-Xiong Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
44
|
Tay HL, Plank M, Collison A, Mattes J, Kumar RK, Foster PS. MicroRNA: potential biomarkers and therapeutic targets for allergic asthma? Ann Med 2014; 46:633-9. [PMID: 25307360 DOI: 10.3109/07853890.2014.958196] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs are small non-coding RNAs that bind to multiple target mRNAs to control gene expression post-transcriptionally by inhibiting translation. In mammalian cells, microRNAs play important roles in a diverse array of cellular processes (e.g. cell proliferation and differentiation). However, alterations in their levels may compromise cellular function, predisposing to disease. In this review, we discuss microRNAs that have been linked with pathogenesis of asthma and propose functional roles in the regulation of disease. MicroRNAs have the potential to be biomarkers for asthma and provide the platform for the development of new classes of therapeutic compounds.
Collapse
Affiliation(s)
- Hock L Tay
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle , Newcastle 2308 , Australia
| | | | | | | | | | | |
Collapse
|
45
|
Henriksen M, Johnsen KB, Andersen HH, Pilgaard L, Duroux M. MicroRNA expression signatures determine prognosis and survival in glioblastoma multiforme--a systematic overview. Mol Neurobiol 2014; 50:896-913. [PMID: 24619503 PMCID: PMC4225053 DOI: 10.1007/s12035-014-8668-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 02/25/2014] [Indexed: 12/21/2022]
Abstract
Despite advances in our knowledge about glioblastoma multiforme (GBM) pathology, clinical challenges still lie ahead with respect to treatment in GBM due to high prevalence, poor prognosis, and frequent tumor relapse. The implication of microRNAs (miRNAs) in GBM is a rapidly expanding field of research with the aim to develop more targeted molecular therapies. This review aims to present a comprehensive overview of all the available literature, evaluating miRNA signatures as a function of prognosis and survival in GBM. The results are presented with a focus on studies derived from clinical data in databases and independent tissue cohorts where smaller samples sizes were investigated. Here, miRNA associated to longer survival (protective) and miRNA with shorter survival (risk-associated) have been identified and their signatures based on different prognostic attributes are described. Finally, miRNAs associated with disease progression or survival in several studies are identified and functionally described. These miRNAs may be valuable for future determination of patient prognosis and could possibly serve as targets for miRNA-based therapies, which hold a great potential in the treatment of this severe malignant disease.
Collapse
Affiliation(s)
- Michael Henriksen
- Laboratory for Cancer Biology, Institute of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3B, 9220 Aalborg Ø, Denmark
| | - Kasper Bendix Johnsen
- Laboratory for Cancer Biology, Institute of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3B, 9220 Aalborg Ø, Denmark
| | - Hjalte Holm Andersen
- Laboratory for Cancer Biology, Institute of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3B, 9220 Aalborg Ø, Denmark
| | - Linda Pilgaard
- Laboratory for Cancer Biology, Institute of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3B, 9220 Aalborg Ø, Denmark
| | - Meg Duroux
- Laboratory for Cancer Biology, Institute of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3B, 9220 Aalborg Ø, Denmark
| |
Collapse
|
46
|
Cui SY, Wang R, Chen LB. MicroRNA-145: a potent tumour suppressor that regulates multiple cellular pathways. J Cell Mol Med 2014; 18:1913-26. [PMID: 25124875 PMCID: PMC4244007 DOI: 10.1111/jcmm.12358] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 05/30/2014] [Indexed: 12/26/2022] Open
Abstract
MicroRNAs are endogenous, small (18-25 nucleotides) non-coding RNAs, which regulate genes expression by directly binding to the 3'-untranslated regions of the target messenger RNAs. Emerging evidence shows that alteration of microRNAs is involved in cancer development. MicroRNA-145 is commonly down-regulated in many types of cancer, regulating various cellular processes, such as the cell cycle, proliferation, apoptosis and invasion, by targeting multiple oncogenes. This review aims to summarize the recent published literature on the role of microRNA-145 in regulating tumourigenesis and progression, and explore its potential for cancer diagnosis, prognosis and treatment.
Collapse
Affiliation(s)
- Shi-Yun Cui
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing UniversityNanjing, Jiangsu, China
| | - Rui Wang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing UniversityNanjing, Jiangsu, China
| | - Long-Bang Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing UniversityNanjing, Jiangsu, China
| |
Collapse
|
47
|
Loskutov YV, Kozyulina PY, Kozyreva VK, Ice RJ, Jones BC, Roston TJ, Smolkin MB, Ivanov AV, Wysolmerski RB, Pugacheva EN. NEDD9/Arf6-dependent endocytic trafficking of matrix metalloproteinase 14: a novel mechanism for blocking mesenchymal cell invasion and metastasis of breast cancer. Oncogene 2014; 34:3662-75. [PMID: 25241893 PMCID: PMC4369482 DOI: 10.1038/onc.2014.297] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 07/20/2014] [Accepted: 07/31/2014] [Indexed: 12/14/2022]
Abstract
NEDD9 is an established marker of invasive and metastatic cancers. NEDD9 downregulation has been shown to dramatically reduce cell invasion and metastasis in multiple tumors. The mechanisms by which NEDD9 regulates invasion are largely unknown. In the current study, we have found that NEDD9 is required for MMP14 enzymatic recovery/recycling through the late endosomes to enable disengagement of tissue inhibitor of matrix metalloproteinase 2 (TIMP2) and tumor invasion. Depletion of NEDD9 decreases targeting of the MMP14/TIMP2 complex to late endosomes and increases trafficking of MMP14 from early/sorting endosomes back to the surface in a small GTPase Arf6-dependent manner. NEDD9 directly binds to Arf6-GAP, ARAP3, and Arf6 effector GGA3 thereby facilitating the Arf6 inactivation required for MMP14/TIMP2 targeting to late endosomes. Re-expression of NEDD9 or a decrease in Arf6 activity is sufficient to restore MMP14 activity and the invasive properties of tumor cells. Importantly, NEDD9 inhibition by Vivo-Morpholinos, an antisense therapy, decreases primary tumor growth and metastasis in xenograft models of breast cancer. Collectively, our findings uncover a novel mechanism to control tumor cells dissemination through NEDD9/Arf6-dependent regulation of MMP14/TIMP2 trafficking, and validates NEDD9 as a clinically relevant therapeutic target to treat metastatic cancer.
Collapse
Affiliation(s)
- Y V Loskutov
- Mary Babb Randolph Cancer Center, West Virginia University School of Medicine, Morgantown, WV, USA
| | - P Y Kozyulina
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, WV, USA
| | - V K Kozyreva
- Mary Babb Randolph Cancer Center, West Virginia University School of Medicine, Morgantown, WV, USA
| | - R J Ice
- Mary Babb Randolph Cancer Center, West Virginia University School of Medicine, Morgantown, WV, USA
| | - B C Jones
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, WV, USA
| | - T J Roston
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, WV, USA
| | - M B Smolkin
- Department of Pathology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - A V Ivanov
- 1] Mary Babb Randolph Cancer Center, West Virginia University School of Medicine, Morgantown, WV, USA [2] Department of Biochemistry, West Virginia University School of Medicine, Morgantown, WV, USA
| | - R B Wysolmerski
- 1] Mary Babb Randolph Cancer Center, West Virginia University School of Medicine, Morgantown, WV, USA [2] Department of Neurobiology and Anatomy, West Virginia University School of Medicine, Morgantown, WV, USA
| | - E N Pugacheva
- 1] Mary Babb Randolph Cancer Center, West Virginia University School of Medicine, Morgantown, WV, USA [2] Department of Biochemistry, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
48
|
Yang J, Zhang JY, Chen J, Chen C, Song XM, Xu Y, Li J. Prognostic role of microRNA-145 in various human malignant neoplasms: a meta-analysis of 18 related studies. World J Surg Oncol 2014; 12:254. [PMID: 25106061 PMCID: PMC4249768 DOI: 10.1186/1477-7819-12-254] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 07/20/2014] [Indexed: 01/20/2023] Open
Abstract
Background Recent studies show that microRNA-145 (miR-145) might be an attractive tumor
biomarker of considerable prognostic value. To clarify the preliminary predictive
value of miR-145 for prognosis in various malignant neoplasms, we conducted a
meta-analysis of 18 relevant studies. Methods Eligible studies were identified by searching the online databases PubMed,
EMBASE, and Web of Science up to March 2014. Pooled hazard ratios (HRs) with 95%
confidence intervals (CIs) for patient survival and disease progress were
calculated to investigate the association with miR-145 expression. Results In total, 18 eligible studies were included in this meta-analysis. Our results
showed that upregulated miR-145 significantly predicted a favorable overall
survival (OS) (HR = 0.47, 95% CI 0.31 to 0.72), but failed to show a significant
relation with disease prognosis. In stratified analyses, high miR-145 expression
predicted favorable OS in both Whites and Asians but the intensity of the
association in Whites (HR = 0.67, 95% CI 0.47 to 0.95) was not as strong as in
Asians (HR = 0.35, 95% CI 0.19 to 0.64). High miR-145 expression also predicted
better progression-free survival (PFS) in Asians (HR = 0.43, 95% CI 0.21 to 0.89),
but not in Whites. In addition, a significantly favorable OS associated with
upregulated miR-145 expression was observed in both squamous cell (SCC)
(HR = 0.34, 95% CI 0.13 to 0.93) and glioblastoma (HR = 0.72, 95% CI 0.52 to
0.99). Conclusions Our findings indicate that high miR-145 expression is better at predicting
patient survival rather than disease progression for malignant tumors, especially
for SCC and glioblastoma in Asians. Considering the insufficient evidence, further
investigations and more studies are needed.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jie Li
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
49
|
Zhao B, Bian EB, Li J, Li J. New advances of microRNAs in glioma stem cells, with special emphasis on aberrant methylation of microRNAs. J Cell Physiol 2014; 229:1141-7. [PMID: 24374932 DOI: 10.1002/jcp.24540] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Accepted: 12/12/2013] [Indexed: 12/15/2022]
Abstract
Malignant brain tumors are thought to be originate from a small population of cells that display stem cell properties, including the capacity of self-renewal, multipotent differentiation, initiation of tumor tissues. Cancer stem cells (CSCs) have been identified in gliomas in which they are named as glioma stem cells (GSCs). GSCs, sharing some characteristics with normal neural stem cells (NSCs), contribute to the cellular origin for primary gliomas and the recurrence of malignant gliomas after current conventional therapy. Recently, increasing evidences have showed that miRNAs play a central role in GSCs. In this review we focus on the role of GSCs in gliomas and in the abnomal expression of miRNAs in GSCs. Furthermore, we also discuss epigenetic dysregulation of tumor-suppressor miRNAs by promoter DNA methylation is involved in the regulation of GSCs biology. Recent advances in understanding dysregulated expression of miRNAs and methylation of tumor-suppressor miRNAs in GSCs and their possible use as new therapeutic targets of gliomas.
Collapse
Affiliation(s)
- Bing Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | | | | | | |
Collapse
|
50
|
Zhang Q, Wang H, Ma Y, Zhang J, He X, Ma J, Zhao ZS. Overexpression of Nedd9 is a prognostic marker of human gastric cancer. Med Oncol 2014; 31:33. [PMID: 24906654 DOI: 10.1007/s12032-014-0033-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 05/10/2014] [Indexed: 01/06/2023]
Abstract
The present study was designed to evaluate the expression and prognostic significance of neural precursor cell-expressed, developmentally downregulated 9 (Nedd9) in patients with gastric cancer. Overexpression of Nedd9 was detected in a number of human cancers and was associated with progression and poor prognosis of the diseases. The expression of Nedd9 and focal adhesion kinase (FAK) were detected using the tissue microarray technique and immunohistochemical method and compared with clinicopathological parameters of patients with gastric cancer. The expressions of Nedd9 and FAK were upregulated in gastric cancer lesions compared with their expression in adjacent non-malignant tissues. High expression of Nedd9 correlated with age, location of tumor, tumor size, depth of invasion, vessel invasion, lymph node metastasis, and distant metastasis, and also with expression of FAK. Further, multivariate analysis suggested that expression of Nedd9 and FAK were independent prognostic indicators for gastric cancer. Cumulative 5-year survival rates of patients with high expression of both Nedd9 and FAK was significantly lower than those with low expression of both. Nedd9 was implicated in the progression of gastric cancer. Based on the TNM stage, Nedd9 and FAK proteins could be useful prognostic marker to predict tumor progression and prognosis in gastric cancer.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Surgery, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | | | | | | | | | | | | |
Collapse
|