51
|
Li C, Wang Z, Tang X, Zeng L, Fan X, Li Z. Molecular mechanisms and potential prognostic effects of REST and REST4 in glioma (Review). Mol Med Rep 2017; 16:3707-3712. [PMID: 29067465 DOI: 10.3892/mmr.2017.7071] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 05/24/2017] [Indexed: 11/06/2022] Open
Abstract
Glioma refers to a tumor of the brain and central nervous system, which is characterized by high incidence, high mortality and high recurrence rate. Although the association between glioma and the repressor element silencing transcription factor (REST) has been reported by numerous studies, the complicated regulatory mechanisms underlying REST remain unknown. REST is a transcriptional repressor that undergoes alternative splicing to produce splicing variants when transcribed. Previous studies have demonstrated that alternative splicing may serve a role in the outcome of glioma. The present review discussed the mutual relationship among REST, REST4 and glioma. It was concluded that increased REST expression in glioma may be associated with poor prognosis; and REST4, an AS variant of REST, also functions to regulate glioma by suppressing REST. In addition, the present review discussed the regulation of REST and its target genes in glioma, and identified factors that induce REST alternative splicing, particularly in glioma. These findings suggest that REST may be considered a prognostic factor, which can be predictive of patient outcome.
Collapse
Affiliation(s)
- Cuilin Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Zhifei Wang
- Department of Neurosurgery, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Xinyue Tang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Liu Zeng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xitang Fan
- Department of Neurosurgery, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
52
|
Inui K, Zhao Z, Yuan J, Jayaprakash S, Le LTM, Drakulic S, Sander B, Golas MM. Stepwise assembly of functional C-terminal REST/NRSF transcriptional repressor complexes as a drug target. Protein Sci 2017; 26:997-1011. [PMID: 28218430 DOI: 10.1002/pro.3142] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 02/05/2017] [Accepted: 02/10/2017] [Indexed: 01/15/2023]
Abstract
In human cells, thousands of predominantly neuronal genes are regulated by the repressor element 1 (RE1)-silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF). REST/NRSF represses transcription of these genes in stem cells and non-neuronal cells by tethering corepressor complexes. Aberrant REST/NRSF expression and intracellular localization are associated with cancer and neurodegeneration in humans. To date, detailed molecular analyses of REST/NRSF and its C-terminal repressor complex have been hampered largely by the lack of sufficient amounts of purified REST/NRSF and its complexes. Therefore, the aim of this study was to express and purify human REST/NRSF and its C-terminal interactors in a baculovirus multiprotein expression system as individual proteins and coexpressed complexes. All proteins were enriched in the nucleus, and REST/NRSF was isolated as a slower migrating form, characteristic of nuclear REST/NRSF in mammalian cells. Both REST/NRSF alone and its C-terminal repressor complex were functionally active in histone deacetylation and histone demethylation and bound to RE1/neuron-restrictive silencer element (NRSE) sites. Additionally, the mechanisms of inhibition of the small-molecule drugs 4SC-202 and SP2509 were analyzed. These drugs interfered with the viability of medulloblastoma cells, where REST/NRSF has been implicated in cancer pathogenesis. Thus, a resource for molecular REST/NRSF studies and drug development has been established.
Collapse
Affiliation(s)
- Ken Inui
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Zongpei Zhao
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Juan Yuan
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | | | - Le T M Le
- Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Srdja Drakulic
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Bjoern Sander
- Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Monika M Golas
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark.,Institute of Human Genetics, Hannover Medical School, D-30625 Hannover, Germany
| |
Collapse
|
53
|
Martin D, Grapin-Botton A. The Importance of REST for Development and Function of Beta Cells. Front Cell Dev Biol 2017; 5:12. [PMID: 28286748 PMCID: PMC5323410 DOI: 10.3389/fcell.2017.00012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 02/07/2017] [Indexed: 01/10/2023] Open
Abstract
Beta cells are defined by the genes they express, many of which are specific to this cell type, and ensure a specific set of functions. Beta cells are also defined by a set of genes they should not express (in order to function properly), and these genes have been called forbidden genes. Among these, the transcriptional repressor RE-1 Silencing Transcription factor (REST) is expressed in most cells of the body, excluding most populations of neurons, as well as pancreatic beta and alpha cells. In the cell types where it is expressed, REST represses the expression of hundreds of genes that are crucial for both neuronal and pancreatic endocrine function, through the recruitment of multiple transcriptional and epigenetic co-regulators. REST targets include genes encoding transcription factors, proteins involved in exocytosis, synaptic transmission or ion channeling, and non-coding RNAs. REST is expressed in the progenitors of both neurons and beta cells during development, but it is down-regulated as the cells differentiate. Although REST mutations and deregulation have yet to be connected to diabetes in humans, REST activation during both development and in adult beta cells leads to diabetes in mice.
Collapse
Affiliation(s)
- David Martin
- Service of Cardiology, Centre Hospitalier Universitaire Vaudois (CHUV) Lausanne, Switzerland
| | | |
Collapse
|
54
|
Cavadas MAS, Cheong A, Taylor CT. The regulation of transcriptional repression in hypoxia. Exp Cell Res 2017; 356:173-181. [PMID: 28219680 DOI: 10.1016/j.yexcr.2017.02.024] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/14/2017] [Accepted: 02/15/2017] [Indexed: 12/20/2022]
Abstract
A sufficient supply molecular oxygen is essential for the maintenance of physiologic metabolism and bioenergetic homeostasis for most metazoans. For this reason, mechanisms have evolved for eukaryotic cells to adapt to conditions where oxygen demand exceeds supply (hypoxia). These mechanisms rely on the modification of pre-existing proteins, translational arrest and transcriptional changes. The hypoxia inducible factor (HIF; a master regulator of gene induction in response to hypoxia) is responsible for the majority of induced gene expression in hypoxia. However, much less is known about the mechanism(s) responsible for gene repression, an essential part of the adaptive transcriptional response. Hypoxia-induced gene repression leads to a reduction in energy demanding processes and the redirection of limited energetic resources to essential housekeeping functions. Recent developments have underscored the importance of transcriptional repressors in cellular adaptation to hypoxia. To date, at least ten distinct transcriptional repressors have been reported to demonstrate sensitivity to hypoxia. Central among these is the Repressor Element-1 Silencing Transcription factor (REST), which regulates over 200 genes. In this review, written to honor the memory and outstanding scientific legacy of Lorenz Poellinger, we provide an overview of our existing knowledge with respect to transcriptional repressors and their target genes in hypoxia.
Collapse
Affiliation(s)
- Miguel A S Cavadas
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 2780-156 Oeiras, Portugal
| | - Alex Cheong
- Life and Health Sciences, Aston University, Birmingham B4 7ET, UK
| | - Cormac T Taylor
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland; Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences and Systems Biology Ireland, University College Dublin, Dublin 4, Ireland.
| |
Collapse
|
55
|
Jin WL, Mao XY, Qiu GZ. Targeting Deubiquitinating Enzymes in Glioblastoma Multiforme: Expectations and Challenges. Med Res Rev 2016; 37:627-661. [PMID: 27775833 DOI: 10.1002/med.21421] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 09/06/2016] [Accepted: 09/25/2016] [Indexed: 12/16/2022]
Abstract
Glioblastoma (GBM) is regarded as the most common primary intracranial neoplasm. Despite standard treatment with tumor resection and radiochemotherapy, the outcome remains gloomy. It is evident that a combination of oncogenic gain of function and tumor-suppressive loss of function has been attributed to glioma initiation and progression. The ubiquitin-proteasome system is a well-orchestrated system that controls the fate of most proteins by striking a dynamic balance between ubiquitination and deubiquitination of substrates, having a profound influence on the modulation of oncoproteins, tumor suppressors, and cellular signaling pathways. In recent years, deubiquitinating enzymes (DUBs) have emerged as potential anti-cancer targets due to their targeting several key proteins involved in the regulation of tumorigenesis, apoptosis, senescence, and autophagy. This review attempts to summarize recent studies of GBM-associated DUBs, their roles in various cellular processes, and discuss the relation between DUBs deregulation and gliomagenesis, especially how DUBs regulate glioma stem cells pluripotency, microenvironment, and resistance of radiation and chemotherapy through core stem-cell transcriptional factors. We also review recent achievements and progress in the development of potent and selective reversible inhibitors of DUBs, and attempted to find a potential GBM treatment by DUBs intervention.
Collapse
Affiliation(s)
- Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China.,National Centers for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Xiao-Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, P. R. China
| | - Guan-Zhong Qiu
- Department of Neurosurgery, General Hospital of Jinan Military Command, Jinan, 250031, P. R. China
| |
Collapse
|
56
|
An expression based REST signature predicts patient survival and therapeutic response for glioblastoma multiforme. Sci Rep 2016; 6:34556. [PMID: 27698411 PMCID: PMC5048293 DOI: 10.1038/srep34556] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 09/15/2016] [Indexed: 01/07/2023] Open
Abstract
Proper regulation of neuronal gene expression is crucial for the development and differentiation of the central nervous system. The transcriptional repressor REST (repressor element-1 silencing transcription factor) is a key regulator in differentiation of pluripotent stem cells to neuronal progenitors and mature neurons. Dysregulated REST activity has been implicated in various diseases, among which the most deadly is glioblastoma multiforme (GBM). Here we have developed an expression-based REST signature (EXPREST), a device providing quantitative measurements of REST activity for GBM tumors. EXPREST robustly quantifies REST activity (REST score) using gene expression profiles in absence of clinic-pathologic assessments of REST. Molecular characterization of REST activity identified global alterations at the DNA, RNA, protein and microRNA levels, suggesting a widespread role of REST in GBM tumorigenesis. Although originally aimed to capture REST activity, REST score was found to be a prognostic factor for overall survival. Further, cell lines with enhanced REST activity was found to be more sensitive to IGF1R, VEGFR and ABL inhibitors. In contrast, cell lines with low REST score were more sensitive to cytotoxic drugs including Mitomycin, Camptothecin and Cisplatin. Together, our work suggests that therapeutic targeting of REST provides a promising opportunity for GBM treatment.
Collapse
|
57
|
Liu Y, Lv H, Wu X, Zhou J, Shi Y, Wen J. Demethylation of Repressor Element-1 Silencing Transcription (REST) Suppresses the Malignant Phenotype of Breast Cancer via MMP9. Oncol Res 2016; 25:445-454. [PMID: 27697091 PMCID: PMC7841195 DOI: 10.3727/096504016x14747368729786] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Breast cancer is the leading cause of cancer deaths in females all over the world, mainly resulting from metastasis. Previous studies have revealed that repressor element-1 (RE-1) silencing transcription (REST) acted as a tumor suppressor in breast cancer. However, the mechanism by which REST is regulated remains unknown, and its role in the metastasis in breast cancer cells remains unclear. In the present study, we showed that the expression of REST was lower in breast cancer samples than that of adjacent samples by immunohistochemical analysis, which may be due to hypermethylation of the REST promoter. Low REST levels are significantly associated with malignant progression in breast cancer patients. Additionally, we elucidated the functions of REST on proliferation and invasion in breast cancer cells. Lentivirus transfection was used to overexpress REST in human breast MDA-MB-231 cells. Then the biologic consequences of overexpressing REST in regard to cell proliferation, apoptosis, and invasion were determined. Furthermore, we also determined matrix metalloproteinase-9 (MMP9) as a target of REST. These results demonstrate that downregulation of REST, a tumor suppressor in breast cancer, is associated with hypermethylation. Induced REST expression is capable of attenuating invasion ability of breast cancer cells, which may be a novel strategy for metastatic breast cancer treatment.
Collapse
|
58
|
Cavadas MAS, Mesnieres M, Crifo B, Manresa MC, Selfridge AC, Keogh CE, Fabian Z, Scholz CC, Nolan KA, Rocha LMA, Tambuwala MM, Brown S, Wdowicz A, Corbett D, Murphy KJ, Godson C, Cummins EP, Taylor CT, Cheong A. REST is a hypoxia-responsive transcriptional repressor. Sci Rep 2016; 6:31355. [PMID: 27531581 PMCID: PMC4987654 DOI: 10.1038/srep31355] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/18/2016] [Indexed: 12/15/2022] Open
Abstract
Cellular exposure to hypoxia results in altered gene expression in a range of physiologic and pathophysiologic states. Discrete cohorts of genes can be either up- or down-regulated in response to hypoxia. While the Hypoxia-Inducible Factor (HIF) is the primary driver of hypoxia-induced adaptive gene expression, less is known about the signalling mechanisms regulating hypoxia-dependent gene repression. Using RNA-seq, we demonstrate that equivalent numbers of genes are induced and repressed in human embryonic kidney (HEK293) cells. We demonstrate that nuclear localization of the Repressor Element 1-Silencing Transcription factor (REST) is induced in hypoxia and that REST is responsible for regulating approximately 20% of the hypoxia-repressed genes. Using chromatin immunoprecipitation assays we demonstrate that REST-dependent gene repression is at least in part mediated by direct binding to the promoters of target genes. Based on these data, we propose that REST is a key mediator of gene repression in hypoxia.
Collapse
Affiliation(s)
- Miguel A S Cavadas
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland.,Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland.,Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 2780-156 Oeiras, Portugal
| | - Marion Mesnieres
- Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| | - Bianca Crifo
- Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| | - Mario C Manresa
- Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| | - Andrew C Selfridge
- Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| | - Ciara E Keogh
- Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| | - Zsolt Fabian
- Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| | - Carsten C Scholz
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland.,Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland.,Institute of Physiology and Zurich Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Karen A Nolan
- Institute of Physiology and Zurich Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,Diabetes Complications Research Centre, School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| | - Liliane M A Rocha
- Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, University of Ulster, Coleraine, Co. Londonderry, BT52 1SA, Northern Ireland, UK
| | - Stuart Brown
- Center for Health Informatics and Bioinformatics, New York University School of Medicine, New York, NY 10016, USA
| | - Anita Wdowicz
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Danielle Corbett
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Keith J Murphy
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Catherine Godson
- Diabetes Complications Research Centre, School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| | - Eoin P Cummins
- Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| | - Cormac T Taylor
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland.,Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| | - Alex Cheong
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland.,Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland.,Life and Health Sciences, Aston University, Birmingham, B4 7ET, UK
| |
Collapse
|
59
|
Inhibition of REST Suppresses Proliferation and Migration in Glioblastoma Cells. Int J Mol Sci 2016; 17:ijms17050664. [PMID: 27153061 PMCID: PMC4881490 DOI: 10.3390/ijms17050664] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/26/2016] [Accepted: 04/27/2016] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary brain tumor, with poor prognosis and a lack of effective therapeutic options. The aberrant expression of transcription factor REST (repressor element 1-silencing transcription factor) had been reported in different kinds of tumors. However, the function of REST and its mechanisms in GBM remain elusive. Here, REST expression was inhibited by siRNA silencing in U-87 and U-251 GBM cells. Then CCK-8 assay showed significantly decreased cell proliferation, and the inhibition of migration was verified by scratch wound healing assay and transwell assay. Using cell cycle analysis and Annexin V/PI straining assay, G1 phase cell cycle arrest was found to be a reason for the suppression of cell proliferation and migration upon REST silencing, while apoptosis was not affected by REST silencing. Further, the detection of REST-downstream genes involved in cytostasis and migration inhibition demonstrated that CCND1 and CCNE1 were reduced; CDK5R1, BBC3, EGR1, SLC25A4, PDCD7, MAPK11, MAPK12, FADD and DAXX were enhanced, among which BBC3 and DAXX were direct targets of REST, as verified by ChIP (chromatin immunoprecipitation) and Western blotting. These data suggested that REST is a master regulator that maintains GBM cells proliferation and migration, partly through regulating cell cycle by repressing downstream genes, which might represent a potential target for GBM therapy.
Collapse
|
60
|
Singh DK, Bose S, Kumar S. Regulation of expression of microRNAs by DNA methylation in lung cancer. Biomarkers 2016; 21:589-99. [PMID: 27122255 DOI: 10.3109/1354750x.2016.1171906] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Differential expression of miRNAs has been linked with lung carcinogenesis. Recent studies have indicated that DNA hypermethylation can lead to silencing of tumor suppressor miRNA-encoding genes. Restoration of tumor suppressor miRNAs using inhibitors of DNA methyltransferases has been shown to suppress cell proliferation, angiogenesis, invasion and metastasis implying that modulation of methylation of specific miRNAs can be used as novel therapeutic targets in lung cancer. In this review, we highlight tremendous progress which has been made in the identification of methylation-mediated silencing of miRNAs and their contribution in lung carcinogenesis along with the clinical utility of methylated miRNAs.
Collapse
Affiliation(s)
- Dhirendra Kumar Singh
- a Amity Institute of Biotechnology , Amity University , Noida , Uttar Pradesh , India
| | - Sudeep Bose
- a Amity Institute of Biotechnology , Amity University , Noida , Uttar Pradesh , India
| | - Sachin Kumar
- b Amity Institute of Molecular Medicine and Stem Cell Research , Amity University , Noida , Uttar Pradesh , India
| |
Collapse
|
61
|
Nechiporuk T, McGann J, Mullendorff K, Hsieh J, Wurst W, Floss T, Mandel G. The REST remodeling complex protects genomic integrity during embryonic neurogenesis. eLife 2016; 5:e09584. [PMID: 26745185 PMCID: PMC4728133 DOI: 10.7554/elife.09584] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 10/20/2015] [Indexed: 01/01/2023] Open
Abstract
The timely transition from neural progenitor to post-mitotic neuron requires down-regulation and loss of the neuronal transcriptional repressor, REST. Here, we have used mice containing a gene trap in the Rest gene, eliminating transcription from all coding exons, to remove REST prematurely from neural progenitors. We find that catastrophic DNA damage occurs during S-phase of the cell cycle, with long-term consequences including abnormal chromosome separation, apoptosis, and smaller brains. Persistent effects are evident by latent appearance of proneural glioblastoma in adult mice deleted additionally for the tumor suppressor p53 protein (p53). A previous line of mice deleted for REST in progenitors by conventional gene targeting does not exhibit these phenotypes, likely due to a remaining C-terminal peptide that still binds chromatin and recruits co-repressors. Our results suggest that REST-mediated chromatin remodeling is required in neural progenitors for proper S-phase dynamics, as part of its well-established role in repressing neuronal genes until terminal differentiation. DOI:http://dx.doi.org/10.7554/eLife.09584.001 In the brain, cells called neurons connect to each other to form complex networks through which information is rapidly processed. These cells start to form in the developing brains of animal embryos when “neural” stem cells divide in a process called neurogenesis. For this process to proceed normally, particular genes in the stem cells have to be switched on or off at different times. This ensures that the protein products of the genes are only made when they are needed. Proteins called transcription factors can bind to DNA to activate or inactivate particular genes; for example, a transcription factor called REST inactivates thousands of genes that are needed by neurons. During neurogenesis, the production of REST normally declines, and some studies have shown that if the production of this protein is artificially increased, the formation of neurons is delayed. However, other studies suggest that REST may not play a major role in neurogenesis. Here, Nechiporuk et al. re-examine the role of REST in mice. The experiments used genetically modified mice in which the gene that encodes REST was prematurely switched off in neural stem cells. Compared with normal mice, these mutant mice had much smaller brains that contained fewer neurons because the stem cells stopped dividing earlier than normal. Unexpectedly, many genes that are normally switched off by REST, were not significantly changed, while genes that are not normally regulated by REST – such as the gene that encodes a protein called p53 – were active. It is known from previous work that p53 is expressed when cells are exposed to harmful conditions that can damage DNA. This helps to prevent cells from becoming cancerous. Nechiporuk et al. found that cells that lacked REST had higher levels of DNA damage than normal cells due to errors during the process of copying DNA before a cell divides. Furthermore, when both REST and p53 were absent, the neural stem cells became cancerous and formed tumors in the mice. Nechiporuk et al.’s findings suggest that REST protects the DNA of genes that are needed for neurons to form and work properly. The new challenge is to understand where in the genome the damage is occurring. DOI:http://dx.doi.org/10.7554/eLife.09584.002
Collapse
Affiliation(s)
- Tamilla Nechiporuk
- Vollum Institute, Howard Hughes Medical Institute, Oregon Health and Science University, Portland, United States
| | - James McGann
- Vollum Institute, Howard Hughes Medical Institute, Oregon Health and Science University, Portland, United States
| | - Karin Mullendorff
- Vollum Institute, Howard Hughes Medical Institute, Oregon Health and Science University, Portland, United States
| | - Jenny Hsieh
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, United States.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Technische Universität München, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians-Universität, Munich, Germany
| | - Thomas Floss
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Gail Mandel
- Vollum Institute, Howard Hughes Medical Institute, Oregon Health and Science University, Portland, United States
| |
Collapse
|
62
|
Brain REST/NRSF Is Not Only a Silent Repressor but Also an Active Protector. Mol Neurobiol 2016; 54:541-550. [DOI: 10.1007/s12035-015-9658-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/17/2015] [Indexed: 01/04/2023]
|
63
|
Schiffer D, Caldera V, Mellai M, Conforti P, Cattaneo E, Zuccato C. Repressor element-1 silencing transcription factor (REST) is present in human control and Huntington's disease neurones. Neuropathol Appl Neurobiol 2015; 40:899-910. [PMID: 24634989 DOI: 10.1111/nan.12137] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 03/12/2014] [Indexed: 01/25/2023]
Abstract
AIMS The repressor element-1 silencing transcription factor/neurone-restrictive silencer factor (REST/NRSF) is a master regulator of neuronal gene expression. REST/NRSF functions by recruiting other cofactors to genomic loci that contain the repressor element 1/neurone restrictive silencer element (RE1/NRSE) binding motif. In brain, demonstration of REST protein presence in neurones has remained controversial. However, RE1/NRSE containing neuronal genes are actively modulated and REST dysregulation is implicated in Huntington's disease (HD). We aimed to investigate REST distribution in autopsy brain from control and HD patients. METHODS Brain tissues from six controls and six HD cases (Vonsattel grade 3 and 4) were investigated using immunohistochemical analysis. RESULTS REST was present in neurones and glial cells of the cortex, caudate nucleus, hippocampus and cerebellum. REST labelling was mainly cytoplasmic in neurones while preferential nuclear staining of REST was found in glial cells. We also found that REST and huntingtin (HTT) colocalize in human neurones. Low levels of cytoplasmic REST were detected in neurones of the HD cortex and caudate but no direct relationship between decreased neuronal REST expression and disease grade was observed. CONCLUSIONS These data support the notion of REST presence in human brain neurones and glial cells and indicate the importance of developing compounds able to restore REST-regulated transcription of neuronal genes in HD.
Collapse
Affiliation(s)
- Davide Schiffer
- Neuro-Bio-Oncology Research Center, Policlinico di Monza Foundation, Vercelli; Consorzio per le Neuroscienze, University of Pavia, Pavia
| | | | | | | | | | | |
Collapse
|
64
|
Correction: REST Controls Self-Renewal and Tumorigenic Competence of Human Glioblastoma Cells. PLoS One 2015; 10:e0139645. [PMID: 26405802 PMCID: PMC4583477 DOI: 10.1371/journal.pone.0139645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
65
|
Alessandrini F, Cristofaro I, Di Bari M, Zasso J, Conti L, Tata AM. The activation of M2 muscarinic receptor inhibits cell growth and survival in human glioblastoma cancer stem cells. Int Immunopharmacol 2015; 29:105-9. [PMID: 26033491 DOI: 10.1016/j.intimp.2015.05.032] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/12/2015] [Accepted: 05/19/2015] [Indexed: 01/13/2023]
Abstract
The involvement of muscarinic receptors in cancer has been reported. Recently we have demonstrated that the activation of M2 muscarinic receptors, through arecaidine propargyl ester, arrests cell proliferation and induces apoptosis in primary and established glioblastoma cell lines. Considering the inability of conventional drugs to completely counteract the growth of glioblastoma cancer stem cells (GSCs), we have investigated the effect produced by arecaidine on GSC growth and survival. The expression of M2 receptors has been analyzed in GSC cell lines derived from human biopsies. Based on the M2 receptor expression levels, we have selected two gliolastoma cell lines (GB7 and GB8). In both cell lines the treatment with arecaidine decreased GCS cell growth. GB7 cells exhibited a time- and dose-dependent decrease of cell proliferation. Moreover arecaidine caused a reduced cell survival in particular in GB8 cell line. These effects appear to be mediated by M2 receptor activation as suggested by pharmacological experiments performed in the presence of M1 and M3 preferring antagonists (pirenzepine and 4-DAMP respectively) and M2/M4 antagonist methoctramine. M2 receptor silencing by siRNA has further confirmed that the inhibition of cell growth arecaidine-induced was mediated by the M2 receptor activation. These results suggest that the M2 receptors may represent a new interesting therapeutic tool to counteract glioblastoma cancer stem cell growth and survival.
Collapse
Affiliation(s)
- Francesco Alessandrini
- Dept. of Biology and Biotechnologies C. Darwin, Research Center of Neurobiology Daniel Bovet, "Sapienza" University of Rome, Roma, Italy
| | - Ilaria Cristofaro
- Dept. of Biology and Biotechnologies C. Darwin, Research Center of Neurobiology Daniel Bovet, "Sapienza" University of Rome, Roma, Italy
| | - Maria Di Bari
- Dept. of Biology and Biotechnologies C. Darwin, Research Center of Neurobiology Daniel Bovet, "Sapienza" University of Rome, Roma, Italy
| | - Jacopo Zasso
- Centre for Integrative Biology, Università degli Studi di Trento, Trento, Italy
| | - Luciano Conti
- Centre for Integrative Biology, Università degli Studi di Trento, Trento, Italy
| | - Ada Maria Tata
- Dept. of Biology and Biotechnologies C. Darwin, Research Center of Neurobiology Daniel Bovet, "Sapienza" University of Rome, Roma, Italy.
| |
Collapse
|
66
|
Ren H, Gao Z, Wu N, Zeng L, Tang X, Chen X, Liu Z, Zhang W, Wang L, Li Z. Expression of REST4 in human gliomas in vivo and influence of pioglitazone on REST in vitro. Biochem Biophys Res Commun 2015; 463:504-9. [PMID: 26003726 DOI: 10.1016/j.bbrc.2015.05.058] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 05/14/2015] [Indexed: 11/25/2022]
Abstract
The repressor element-1 (RE1) silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF) has an irreplaceable role during the differentiation of neurons. REST has multiple splice variants which link to various types of cancer. Previous work had highlighted the role of REST in glioma, where the expression of REST is enhanced. But whether alternative splicing of REST is expressed in glioma has not been described. Here, we show that a specific isoform REST4 is expressed in glioma specimens, and will influence the mRNA level of REST in vivo. Peroxisome proliferator-activated receptor-γ (PPARγ) agonists have a role of antineoplastic in various tumor cells, which including glioma cells. Moreover, study indicated that PPARγ agonist pioglitazone can promote alternative splicing of REST pre-mRNA. In this study, we selected pioglitazone as a tool drug to explore whether the role of pioglitazone in anti-glioma is mediated by regulating REST expression or promoting alternative splicing of REST in glioma cells. Results show that pioglitazone can inhibit proliferation and induce apoptosis of glioma cell in vitro, which may be mediated by down-regulating REST mRNA level but not by inducing alternative splicing of REST pre-mRNA. Our study firstly reports the expression of REST4 in glioma tissue samples. And we recommend that pioglitazone, which can reduce the expression level of REST, represents a promising drug for therapy of glioma.
Collapse
Affiliation(s)
- Huan Ren
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Zhangfeng Gao
- Department of Neurosurgery, Second Xiangya Hospital of Central South University, Changsha 410008, China
| | - Nayiyuan Wu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Liu Zeng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Xinyue Tang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Xiaoping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Liansheng Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Zhi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China.
| |
Collapse
|
67
|
Sun Y, Luo ZM, Guo XM, Su DF, Liu X. An updated role of microRNA-124 in central nervous system disorders: a review. Front Cell Neurosci 2015; 9:193. [PMID: 26041995 PMCID: PMC4438253 DOI: 10.3389/fncel.2015.00193] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 05/04/2015] [Indexed: 12/18/2022] Open
Abstract
MicroRNA-124 (miR-124) is the most abundant miRNA in the brain. Biogenesis of miR-124 displays specific temporal and spatial profiles in various cell and tissue types and affects a broad spectrum of biological functions in the central nervous system (CNS). Recently, the link between dysregulation of miR-124 and CNS disorders, such as neurodegeneration, CNS stress, neuroimmune disorders, stroke, and brain tumors, has become evident. Here, we provide an overview of the specific molecular function of miR-124 in the CNS and a revealing insight for the therapeutic potential of miR-124 in the treatment of human CNS diseases.
Collapse
Affiliation(s)
- Yang Sun
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai China
| | - Zhu-Min Luo
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai China
| | - Xiu-Ming Guo
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai China
| | - Ding-Feng Su
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai China
| | - Xia Liu
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai China
| |
Collapse
|
68
|
Fernández-Martínez P, Zahonero C, Sánchez-Gómez P. DYRK1A: the double-edged kinase as a protagonist in cell growth and tumorigenesis. Mol Cell Oncol 2015; 2:e970048. [PMID: 27308401 PMCID: PMC4905233 DOI: 10.4161/23723548.2014.970048] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 09/03/2014] [Accepted: 09/03/2014] [Indexed: 01/12/2023]
Abstract
DYRK1A (dual-specificity tyrosine-regulated kinase 1A) is a kinase with multiple implications for embryonic development, especially in the nervous system where it regulates the balance between proliferation and differentiation of neural progenitors. The DYRK1A gene is located in the Down syndrome critical region and may play a significant role in the developmental brain defects, early neurodegeneration, and cancer susceptibility of individuals with this syndrome. DYRK1A is also expressed in adults, where it might participate in the regulation of cell cycle, survival, and tumorigenesis, thus representing a potential therapeutic target for certain types of cancer. However, the final readout of DYRK1A overexpression or inhibition depends strongly on the cellular context, as it has both tumor suppressor and oncogenic activities. Here, we will discuss the functions and substrates of DYRK1A associated with the control of cell growth and tumorigenesis with a focus on the potential use of DYRK1A inhibitors in cancer therapy.
Collapse
Affiliation(s)
- P Fernández-Martínez
- Instituto de Medicina Molecular Aplicada; Universidad CEU-San Pablo ; Madrid, Spain
| | - C Zahonero
- Neuro-oncology Unit; Instituto de Salud Carlos III-UFIEC ; Madrid, Spain
| | - P Sánchez-Gómez
- Neuro-oncology Unit; Instituto de Salud Carlos III-UFIEC ; Madrid, Spain
| |
Collapse
|
69
|
NRSF: an Angel or a Devil in Neurogenesis and Neurological Diseases. J Mol Neurosci 2014; 56:131-44. [DOI: 10.1007/s12031-014-0474-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/18/2014] [Indexed: 12/12/2022]
|
70
|
Swartling FJ, Bolin S, Phillips JJ, Persson AI. Signals that regulate the oncogenic fate of neural stem cells and progenitors. Exp Neurol 2014; 260:56-68. [PMID: 23376224 PMCID: PMC3758390 DOI: 10.1016/j.expneurol.2013.01.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 01/19/2013] [Accepted: 01/24/2013] [Indexed: 12/16/2022]
Abstract
Brain tumors have frequently been associated with a neural stem cell (NSC) origin and contain stem-like tumor cells, so-called brain tumor stem cells (BTSCs) that share many features with normal NSCs. A stem cell state of BTSCs confers resistance to radiotherapy and treatment with alkylating agents. It is also a hallmark of aggressive brain tumors and is maintained by transcriptional networks that are also active in embryonic stem cells. Advances in reprogramming of somatic cells into induced pluripotent stem (iPS) cells have further identified genes that drive stemness. In this review, we will highlight the possible drivers of stemness in medulloblastoma and glioma, the most frequent types of primary malignant brain cancer in children and adults, respectively. Signals that drive expansion of developmentally defined neural precursor cells are also active in corresponding brain tumors. Transcriptomal subgroups of human medulloblastoma and glioma match features of NSCs but also more restricted progenitors. Lessons from genetically-engineered mouse (GEM) models show that temporally and regionally defined NSCs can give rise to distinct subgroups of medulloblastoma and glioma. We will further discuss how acquisition of stem cell features may drive brain tumorigenesis from a non-NSC origin. Genetic alterations, signaling pathways, and therapy-induced changes in the tumor microenvironment can drive reprogramming networks and induce stemness in brain tumors. Finally, we propose a model where dysregulation of microRNAs (miRNAs) that normally provide barriers against reprogramming plays an integral role in promoting stemness in brain tumors.
Collapse
Affiliation(s)
- Fredrik J Swartling
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Sara Bolin
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Joanna J Phillips
- Department of Neurological Surgery, Brain Tumor Research Center, University of California, San Francisco, USA; Department of Pathology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, USA
| | - Anders I Persson
- Department of Neurological Surgery, Brain Tumor Research Center, University of California, San Francisco, USA; Department of Neurology, Sandler Neurosciences Center, University of California, San Francisco, USA.
| |
Collapse
|
71
|
Das A, Chai JC, Jung KH, Das ND, Kang SC, Lee YS, Seo H, Chai YG. JMJD2A attenuation affects cell cycle and tumourigenic inflammatory gene regulation in lipopolysaccharide stimulated neuroectodermal stem cells. Exp Cell Res 2014; 328:361-78. [PMID: 25193078 DOI: 10.1016/j.yexcr.2014.08.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 07/21/2014] [Accepted: 08/19/2014] [Indexed: 01/05/2023]
Abstract
JMJD2A is a lysine trimethyl-specific histone demethylase that is highly expressed in a variety of tumours. The role of JMJD2A in tumour progression remains unclear. The objectives of this study were to identify JMJD2A-regulated genes and understand the function of JMJD2A in p53-null neuroectodermal stem cells (p53(-/-) NE-4Cs). We determined the effect of LPS as a model of inflammation in p53(-/-) NE-4Cs and investigated whether the epigenetic modifier JMJD2A alter the expression of tumourigenic inflammatory genes. Global gene expression was measured in JMJD2A knockdown (kd) p53(-/-) NE-4Cs and in LPS-stimulated JMJD2A-kd p53(-/-) NE-4C cells. JMJD2A attenuation significantly down-regulated genes were Cdca2, Ccnd2, Ccnd1, Crebbp, IL6rα, and Stat3 related with cell cycle, proliferation, and inflammatory-disease responses. Importantly, some tumour-suppressor genes including Dapk3, Timp2 and TFPI were significantly up-regulated but were not affected by silencing of the JMJD2B. Furthermore, we confirmed the attenuation of JMJD2A also down-regulated Cdca2, Ccnd2, Crebbp, and Rest in primary NSCs isolated from the forebrains of E15 embryos of C57/BL6J mice with effective p53 inhibitor pifithrin-α (PFT-α). Transcription factor (TF) motif analysis revealed known binding patterns for CDC5, MYC, and CREB, as well as three novel motifs in JMJD2A-regulated genes. IPA established molecular networks. The molecular network signatures and functional gene-expression profiling data from this study warrants further investigation as an effective therapeutic target, and studies to elucidate the molecular mechanism of JMJD2A-kd-dependent effects in neuroectodermal stem cells should be performed.
Collapse
Affiliation(s)
- Amitabh Das
- Department of Bionanotechnology, Hanyang University, Seoul 133-791, Republic of Korea.
| | - Jin Choul Chai
- Department of Molecular & Life Science, Hanyang University, 1271 Sa 3-dong, Ansan 426-791, Gyeonggi-do, Republic of Korea.
| | - Kyoung Hwa Jung
- Department of Molecular & Life Science, Hanyang University, 1271 Sa 3-dong, Ansan 426-791, Gyeonggi-do, Republic of Korea.
| | - Nando Dulal Das
- Clinical Research Centre, Inha University School of Medicine, Incheon 400-711, Republic of Korea.
| | - Sung Chul Kang
- Department of Molecular & Life Science, Hanyang University, 1271 Sa 3-dong, Ansan 426-791, Gyeonggi-do, Republic of Korea.
| | - Young Seek Lee
- Department of Molecular & Life Science, Hanyang University, 1271 Sa 3-dong, Ansan 426-791, Gyeonggi-do, Republic of Korea.
| | - Hyemyung Seo
- Department of Molecular & Life Science, Hanyang University, 1271 Sa 3-dong, Ansan 426-791, Gyeonggi-do, Republic of Korea.
| | - Young Gyu Chai
- Department of Bionanotechnology, Hanyang University, Seoul 133-791, Republic of Korea; Department of Molecular & Life Science, Hanyang University, 1271 Sa 3-dong, Ansan 426-791, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
72
|
Formisano L, Guida N, Laudati G, Boscia F, Esposito A, Secondo A, Di Renzo G, Canzoniero LMT. Extracellular signal-related kinase 2/specificity protein 1/specificity protein 3/repressor element-1 silencing transcription factor pathway is involved in Aroclor 1254-induced toxicity in SH-SY5Y neuronal cells. J Neurosci Res 2014; 93:167-77. [PMID: 25093670 DOI: 10.1002/jnr.23464] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 07/04/2014] [Accepted: 07/08/2014] [Indexed: 12/16/2022]
Abstract
Polychlorinated biphenyls (PCBs) cause a wide spectrum of toxic effects in the brain through undefined mechanisms. Exposure to the PCB mixture Aroclor-1254 (A1254) increases the repressor element-1 silencing transcription factor (REST) expression, leading to neuronal death. This study sought to understand the sequence of some molecular mechanisms to determine whether A1254 could increase REST expression and the cytoprotective effect of the phorbol ester tetradecanoylphorbol acetate (TPA) on A1254-induced toxicity in SH-SY5Y cells. As shown by Western blot analysis, A1254 (10 µg/ml) downregulates extracellular signal-related kinase 2 (ERK2) phosphorylation in a time-dependent manner, thereby triggering the binding of specificity protein 1 (Sp1) and Sp3 to the REST gene promoter as revealed by chromatin immunoprecipitation analysis. This chain of events results in an increase in REST mRNA and cell death, as assessed by quantitative real-time polymerase chain reaction and dimethylthiazolyl-2-5-diphenyltetrazolium-bromide assay, respectively. Accordingly, TPA prevented both the A1254-induced decrease in ERK2 phosphorylation and the A1254-induced increase in Sp1, Sp3, and REST protein expression. After 48 hr, TPA prevented A1254-induced cell death. ERK2 overexpression counteracted the A1254-induced increase in Sp1 and Sp3 protein expression and prevented A1254-induced Sp1 and Sp3 binding to the REST gene promoter, thus counteracting the increase in REST mRNA expression induced by the toxicant. In neuroblastoma SH-SY5Y cells, ERK2/Sp1/SP3/REST is a new pathway underlying the neurotoxic effect of PCB. The ERK2/Sp1/Sp3/REST pathway, which underlies A1254-induced neuronal death, might represent a new drug signaling cascade in PCB-induced neuronal toxicity.
Collapse
Affiliation(s)
- Luigi Formisano
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University of Naples Federico II, Naples, Italy; Division of Pharmacology, Department of Science and Technology, University of Sannio, Benevento, Italy
| | | | | | | | | | | | | | | |
Collapse
|
73
|
Lee M, Ji H, Furuta Y, Park JI, McCrea PD. p120-catenin regulates REST and CoREST, and modulates mouse embryonic stem cell differentiation. J Cell Sci 2014; 127:4037-51. [PMID: 25074806 DOI: 10.1242/jcs.151944] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Although the canonical Wnt pathway and β-catenin have been extensively studied, less is known about the role of p120-catenin (also known as δ1-catenin) in the nuclear compartment. Here, we report that p120-catenin binds and negatively regulates REST and CoREST (also known as Rcor1), a repressive transcriptional complex that has diverse developmental and pathological roles. Using mouse embryonic stem cells (mESCs), mammalian cell lines, Xenopus embryos and in vitro systems, we find that p120-catenin directly binds the REST-CoREST complex, displacing it from established gene targets to permit their transcriptional activation. Importantly, p120-catenin levels further modulate the mRNA and protein levels of Oct4 (also known as POU5F1), Nanog and Sox2, and have an impact upon the differentiation of mESCs towards neural fates. In assessing potential upstream inputs to this new p120-catenin-REST-CoREST pathway, REST gene targets were found to respond to the level of E-cadherin, with evidence suggesting that p120-catenin transduces signals between E-cadherin and the nucleus. In summary, we provide the first evidence for a direct upstream modulator and/or pathway regulating REST-CoREST, and reveal a substantial role for p120-catenin in the modulation of stem cell differentiation.
Collapse
Affiliation(s)
- Moonsup Lee
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA Program in Genes and Development, The University of Texas Graduate School of Biomedical Science-Houston, Houston, TX 77030, USA
| | - Hong Ji
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yasuhide Furuta
- Laboratory for Animal Resources and Genetic Engineering, Riken Center for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Jae-il Park
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA
| | - Pierre D McCrea
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA Program in Genes and Development, The University of Texas Graduate School of Biomedical Science-Houston, Houston, TX 77030, USA
| |
Collapse
|
74
|
Cell density modulates SHC3 expression and survival of human glioblastoma cells through Fak activation. J Neurooncol 2014; 120:245-56. [PMID: 25062668 DOI: 10.1007/s11060-014-1551-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 07/05/2014] [Indexed: 10/25/2022]
Abstract
Shc3 protein levels are high in human glioblastoma but they decrease in vitro. We found that SHC3 mRNA and protein increased when glioblastoma cells grew as multicellular tumor spheroid (MTS). Shc3 expression was also induced in adherent cultures by increasing cell density. Among the Shc family members, only Shc2 and Shc3 increased with cell density. Shc3 and focal adhesion kinase (Fak) interact as shown by co-immunoprecipitation. Inhibition of Fak activation reduced Shc3 increase and MTS formation and changed Shc3 phosphorylation pattern. Our results suggest that in gliomas cell density modulates Shc3 protein levels and its activity, at least in part, through Fak activation.
Collapse
|
75
|
Liang J, Tong P, Zhao W, Li Y, Zhang L, Xia Y, Yu Y. The REST gene signature predicts drug sensitivity in neuroblastoma cell lines and is significantly associated with neuroblastoma tumor stage. Int J Mol Sci 2014; 15:11220-33. [PMID: 24968265 PMCID: PMC4139778 DOI: 10.3390/ijms150711220] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 05/30/2014] [Accepted: 06/03/2014] [Indexed: 11/16/2022] Open
Abstract
Neuroblastoma is the most common and deadly solid tumor in children, and there is currently no effective treatment available for neuroblastoma patients. The repressor element-1 silencing transcription (REST) factor has been found to play important roles in the regulation of neural differentiation and tumorigenesis. Recently, a REST signature consisting of downstream targets of REST has been reported to have clinical relevance in both breast cancer and glioblastoma. However it remains unclear how the REST signature works in neuroblastoma. Publicly available datasets were mined and bioinformatic approaches were used to investigate the utility of the REST signature in neuroblastoma with both preclinical and real patient data. The REST signature was found to be associated with drug sensitivity in neuroblastoma cell lines. Further, neuroblastoma patients with enhanced REST activity are significantly associated with higher clinical stages. Loss of heterozygosity on chromosome 11q23, which occurs in a large subset of high-risk neuroblastomas, tends to be correlated with high REST activity, with marginal significance. In conclusion, the REST signature has important implications for targeted therapy, and it is a prognostic factor in neuroblastoma patients.
Collapse
Affiliation(s)
- Jianfeng Liang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Pan Tong
- Department of Bioinformatics and Computational Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Wanni Zhao
- Department of General Surgery, Jinan Central Hospital, Jinan 250013, China.
| | - Yaqiao Li
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| | - Li Zhang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Ying Xia
- The Vivan L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, TX 77030, USA.
| | - Yanbing Yu
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China.
| |
Collapse
|
76
|
Bai Y, Lathia JD, Zhang P, Flavahan W, Rich JN, Mattson MP. Molecular targeting of TRF2 suppresses the growth and tumorigenesis of glioblastoma stem cells. Glia 2014; 62:1687-98. [PMID: 24909307 DOI: 10.1002/glia.22708] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 05/23/2014] [Accepted: 05/23/2014] [Indexed: 12/23/2022]
Abstract
Glioblastoma is the most prevalent primary brain tumor and is essentially universally fatal within 2 years of diagnosis. Glioblastomas contain cellular hierarchies with self-renewing glioblastoma stem cells (GSCs) that are often resistant to chemotherapy and radiation therapy. GSCs express high amounts of repressor element 1 silencing transcription factor (REST), which may contribute to their resistance to standard therapies. Telomere repeat-binding factor 2 (TRF2) stablizes telomeres and REST to maintain self-renewal of neural stem cells and tumor cells. Here we show viral vector-mediated delivery of shRNAs targeting TRF2 mRNA depletes TRF2 and REST from GSCs isolated from patient specimens. As a result, GSC proliferation is reduced and the level of proteins normally expressed by postmitotic neurons (L1CAM and β3-tubulin) is increased, suggesting that loss of TRF2 engages a cell differentiation program in the GSCs. Depletion of TRF2 also sensitizes GSCs to temozolomide, a DNA-alkylating agent currently used to treat glioblastoma. Targeting TRF2 significantly increased the survival of mice bearing GSC xenografts. These findings reveal a role for TRF2 in the maintenance of REST-associated proliferation and chemotherapy resistance of GSCs, suggesting that TRF2 is a potential therapeutic target for glioblastoma.
Collapse
Affiliation(s)
- Yun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China; Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland
| | | | | | | | | | | |
Collapse
|
77
|
Stem cell niches in glioblastoma: a neuropathological view. BIOMED RESEARCH INTERNATIONAL 2014; 2014:725921. [PMID: 24834433 PMCID: PMC4009309 DOI: 10.1155/2014/725921] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/22/2014] [Accepted: 03/24/2014] [Indexed: 11/27/2022]
Abstract
Glioblastoma (GBM) stem cells (GSCs), responsible for tumor growth, recurrence, and resistance to therapies, are considered the real therapeutic target, if they had no molecular mechanisms of resistance, in comparison with the mass of more differentiated cells which are insensitive to therapies just because of being differentiated and nonproliferating. GSCs occur in tumor niches where both stemness status and angiogenesis are conditioned by the microenvironment. In both perivascular and perinecrotic niches, hypoxia plays a fundamental role. Fifteen glioblastomas have been studied by immunohistochemistry and immunofluorescence for stemness and differentiation antigens. It has been found that circumscribed necroses develop inside hyperproliferating areas that are characterized by high expression of stemness antigens. Necrosis developed inside them because of the imbalance between the proliferation of tumor cells and endothelial cells; it reduces the number of GSCs to a thin ring around the former hyperproliferating area. The perinecrotic GSCs are nothing else that the survivors remnants of those populating hyperproliferating areas. In the tumor, GSCs coincide with malignant areas so that the need to detect where they are located is not so urgent.
Collapse
|
78
|
Gebhardt ML, Reuter S, Mrowka R, Andrade-Navarro MA. Similarity in targets with REST points to neural and glioblastoma related miRNAs. Nucleic Acids Res 2014; 42:5436-46. [PMID: 24728992 PMCID: PMC4027192 DOI: 10.1093/nar/gku231] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
There are groups of genes that need coordinated repression in multiple contexts, for example if they code for proteins that work together in a pathway or in a protein complex. Redundancy of biological regulatory networks implies that such coordinated repression might occur at both the pre- and post-transcriptional level, though not necessarily simultaneously or under the same conditions. Here, we propose that such redundancy in the global regulatory network can be detected by the overlap between the putative targets of a transcriptional repressor, as identified by a ChIP-seq experiment, and predicted targets of a microRNA (miRNA). To test this hypothesis, we used publicly available ChIP-seq data of the neural transcriptional repressor RE1 silencing transcription factor (REST) from 15 different cell samples. We found 20 miRNAs, each of which shares a significant amount of predicted targets with REST. The set of predicted associations between these 20 miRNAs and the overlapping REST targets is enriched in known miRNA targets. Many of the detected miRNAs have functions related to neural identity and glioblastoma, which could be expected from their overlap in targets with REST. We propose that the integration of experimentally determined transcription factor binding sites with miRNA-target predictions provides functional information on miRNAs.
Collapse
Affiliation(s)
- Marie L Gebhardt
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany
| | - Stefanie Reuter
- Experimentelle Nephrologie, KIMIII, Universitätsklinikum Jena, Friedrich-Schiller-Universität, Jena 07743, Germany
| | - Ralf Mrowka
- Experimentelle Nephrologie, KIMIII, Universitätsklinikum Jena, Friedrich-Schiller-Universität, Jena 07743, Germany
| | | |
Collapse
|
79
|
Foudah D, Monfrini M, Donzelli E, Niada S, Brini AT, Orciani M, Tredici G, Miloso M. Expression of neural markers by undifferentiated mesenchymal-like stem cells from different sources. J Immunol Res 2014; 2014:987678. [PMID: 24741639 PMCID: PMC3987801 DOI: 10.1155/2014/987678] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 01/15/2014] [Indexed: 01/09/2023] Open
Abstract
The spontaneous expression of neural markers, already demonstrated in bone marrow (BM) mesenchymal stem cells (MSCs), has been considered as evidence of the MSCs' predisposition to differentiate toward neural lineages, supporting their use in stem cell-based therapy for neural repair. In this study we have evaluated, by immunocytochemistry, immunoblotting, and flow cytometry experiments, the expression of neural markers in undifferentiated MSCs from different sources: human adipose stem cells (hASCs), human skin-derived mesenchymal stem cells (hS-MSCs), human periodontal ligament stem cells (hPDLSCs,) and human dental pulp stem cells (hDPSCs). Our results demonstrate that the neuronal markers β III-tubulin and NeuN, unlike other evaluated markers, are spontaneously expressed by a very high percentage of undifferentiated hASCs, hS-MSCs, hPDLSCs, and hDPSCs. Conversely, the neural progenitor marker nestin is expressed only by a high percentage of undifferentiated hPDLSCs and hDPSCs. Our results suggest that the expression of β III-tubulin and NeuN could be a common feature of stem cells and not exclusive to neuronal cells. This could result in a reassessment of the use of β III-tubulin and NeuN as the only evidence proving neuronal differentiation. Further studies will be necessary to elucidate the relevance of the spontaneous expression of these markers in stem cells.
Collapse
Affiliation(s)
- Dana Foudah
- Department of Surgery and Translational Medicine, University of Milano-Bicocca, 20900 Monza, Italy
| | - Marianna Monfrini
- Department of Surgery and Translational Medicine, University of Milano-Bicocca, 20900 Monza, Italy
| | - Elisabetta Donzelli
- Department of Surgery and Translational Medicine, University of Milano-Bicocca, 20900 Monza, Italy
| | - Stefania Niada
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20129 Milano, Italy
- IRCCS Galeazzi Orthopaedic Institute, 20161 Milano, Italy
| | - Anna T. Brini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20129 Milano, Italy
- IRCCS Galeazzi Orthopaedic Institute, 20161 Milano, Italy
| | - Monia Orciani
- Department of Clinical and Molecular Sciences—Histology, University Politecnica delle Marche, 60126 Ancona, Italy
| | - Giovanni Tredici
- Department of Surgery and Translational Medicine, University of Milano-Bicocca, 20900 Monza, Italy
| | - Mariarosaria Miloso
- Department of Surgery and Translational Medicine, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
80
|
Tivnan A, Zhao J, Johns TG, Day BW, Stringer BW, Boyd AW, Tiwari S, Giles KM, Teo C, McDonald KL. The tumor suppressor microRNA, miR-124a, is regulated by epigenetic silencing and by the transcriptional factor, REST in glioblastoma. Tumour Biol 2013; 35:1459-65. [DOI: 10.1007/s13277-013-1200-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 09/11/2013] [Indexed: 01/23/2023] Open
|
81
|
Gorba T, Conti L. Neural stem cells as tools for drug discovery: novel platforms and approaches. Expert Opin Drug Discov 2013; 8:1083-94. [DOI: 10.1517/17460441.2013.805199] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
82
|
Negrini S, Prada I, D'Alessandro R, Meldolesi J. REST: an oncogene or a tumor suppressor? Trends Cell Biol 2013; 23:289-95. [PMID: 23414932 DOI: 10.1016/j.tcb.2013.01.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 01/10/2013] [Accepted: 01/15/2013] [Indexed: 12/21/2022]
Abstract
The Repressor Element-1 (RE-1) Silencing Transcription (REST) factor, which is highly expressed in stem cells and non-neural cells, with low expression in neurons and other neural cells, orchestrates neural differentiation and preserves the unique neural phenotype. REST also plays a role in proliferation, although its effect differs depending on the cell type. It acts as an oncogene in neural cells and tumors (medulloblastomas, neuroblastomas, glioblastomas) and as a tumor suppressor in carcinomas of the lung, breast, and colon. The mechanisms underlying this duality have started to emerge recently and new therapeutic approaches based on these findings are being developed. Here, we present the mechanisms proposed to account for the oncogenic and antioncogenic roles of REST and discuss the therapeutic perspective of recent advances, particularly for small-cell lung cancer.
Collapse
Affiliation(s)
- Sara Negrini
- San Raffaele Scientific Institute and Vita Salute San Raffaele University, Milan, Italy
| | | | | | | |
Collapse
|
83
|
A REST derived gene signature stratifies glioblastomas into chemotherapy resistant and responsive disease. BMC Genomics 2012; 13:686. [PMID: 23216891 PMCID: PMC3545737 DOI: 10.1186/1471-2164-13-686] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 11/28/2012] [Indexed: 11/10/2022] Open
Abstract
Background Glioblastomas are the most common central nervous system neoplasia in adults, with 9,000 cases in the US annually. Glioblastoma multiformae, the most aggressive glioma subtype, has an 18% one-year survival rate, and 3% two year survival rate. Recent work has highlighted the role of the transcription factor RE1 Silencing Transcription Factor, REST in glioblastoma but how REST function correlates with disease outcome has not been described. Method Using a bioinformatic approach and mining of publicly available microarray datasets, we describe an aggressive subtype of gliomas defined by a gene signature derived from REST. Using this REST gene signature we predict that REST function is enhanced in advanced glioblastoma. We compare disease outcomes between tumors based on REST status and treatment regimen, and describe downstream targets of REST that may contribute to the decreased benefits observed with high dose chemotherapy in REM tumors. Results We present human data showing that patients with “REST Enhanced Malignancies” (REM) tumors present with a shorter disease free survival compared to non-REM gliomas. Importantly, REM tumors are refractory to multiple rounds of chemotherapy and patients fail to respond to this line of treatment. Conclusions This report is the first to describe a REST gene signature that predicts response to multiple rounds of chemotherapy, the mainline therapy for this disease. The REST gene signature may have important clinical implications for the treatment of glioblastoma.
Collapse
|