Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38585789

ABSTRACT

The transcription repressor REST in the dorsal root ganglion (DRG) is upregulated by peripheral nerve injury and promotes the development of chronic pain. However, the genes targeted by REST in neuropathic pain development remain unclear. The expression levels of 4 opioid receptor (Oprm1, Oprd1, Oprl1, Oprk1) and the cannabinoid CB1 receptor (Cnr1) genes in the DRG regulate nociception. In this study, we determined the role of REST in the control of their expression in the DRG induced by spared nerve injury (SNI) in both male and female mice. Transcriptomic analyses of male mouse DRGs followed by quantitative reverse transcription polymerase chain reaction analyses of both male and female mouse DRGs showed that SNI upregulated expression of Rest and downregulated mRNA levels of all 4 opioid receptor and Cnr1 genes, but Oprm1 was upregulated in female mice. Analysis of publicly available bioinformatic data suggested that REST binds to the promoter regions of Oprm1 and Cnr1. Chromatin immunoprecipitation analyses indicated differing levels of REST at these promoters in male and female mice. Full-length Rest conditional knockout in primary sensory neurons reduced SNI-induced pain hypersensitivity and rescued the SNI-induced reduction in the expression of Oprd1 and Cnr1 in the DRG in both male and female mice. Our results suggest that nerve injury represses the transcription of Oprd1 and Cnr1 via REST in primary sensory neurons and that REST is a potential therapeutic target for neuropathic pain.

3.
Neuro Oncol ; 21(6): 775-785, 2019 06 10.
Article in English | MEDLINE | ID: mdl-30953587

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is a lethal, heterogeneous human brain tumor, with regulatory mechanisms that have yet to be fully characterized. Previous studies have indicated that the transcriptional repressor REST (repressor element-1 silencing transcription factor) regulates the oncogenic potential of GBM stem cells (GSCs) based on level of expression. However, how REST performs its regulatory role is not well understood. METHODS: We examined 2 independent high REST (HR) GSC lines using genome-wide assays, biochemical validations, gene knockdown analysis, and mouse tumor models. We analyzed in-house patient tumors and patient data present in The Cancer Genome Atlas (TCGA). RESULTS: Genome-wide transcriptome and DNA-binding analyses suggested the dopamine receptor D2 (DRD2) gene, a dominant regulator of neurotransmitter signaling, as a direct target of REST. Biochemical analyses and mouse intracranial tumor models using knockdown of REST and double knockdown of REST and DRD2 validated this target and suggested that DRD2 is a downstream target of REST regulating tumorigenesis, at least in part, through controlling invasion and apoptosis. Further, TCGA GBM data support the presence of the REST-DRD2 axis and reveal that high REST/low DRD2 (HRLD) and low REST/high DRD2 (LRHD) tumors are specific subtypes, are molecularly different from the known GBM subtypes, and represent functional groups with distinctive patterns of enrichment of gene sets and biological pathways. The inverse HRLD/LRHD expression pattern is also seen in in-house GBM tumors. CONCLUSIONS: These findings suggest that REST regulates neurotransmitter signaling pathways through DRD2 in HR-GSCs to impact tumorigenesis. They further suggest that the REST-DRD2 mechanism forms distinct subtypes of GBM.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Receptors, Dopamine D2/metabolism , Repressor Proteins/metabolism , Aged , Aged, 80 and over , Animals , Apoptosis , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Male , Mice , Middle Aged , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Prognosis , Receptors, Dopamine D2/genetics , Repressor Proteins/genetics , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Sci Rep ; 8(1): 12083, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30108242

ABSTRACT

Overexpression of REST has been implicated in brain tumors, ischemic insults, epilepsy, and movement disorders such as Huntington's disease. However, owing to the lack of a conditional REST overexpression animal model, the mechanism of action of REST overexpression in these disorders has not been established in vivo. We created a REST overexpression mouse model using the human REST (hREST) gene. Our results using these mice confirm that hREST expression parallels endogenous REST expression in embryonic mouse brains. Further analyses indicate that REST represses the dopamine receptor 2 (Drd2) gene, which encodes a critical nigrostriatal receptor involved in regulating movement, in vivo. Overexpression of REST using Drd2-Cre in adult mice results in increased REST and decreased DRD2 expression in the striatum, a major site of DRD2 expression, and phenocopies the spontaneous locomotion deficits seen upon global DRD2 deletion or specific DRD2 deletion from indirect-pathway medium spiny neurons. Thus, our studies using this mouse model not only reveal a new function of REST in regulating spontaneous locomotion but also suggest that REST overexpression in DRD2-expressing cells results in spontaneous locomotion deficits.


Subject(s)
Corpus Striatum/metabolism , Locomotion/physiology , Neurons/metabolism , Receptors, Dopamine D2/metabolism , Repressor Proteins/metabolism , Animals , Corpus Striatum/cytology , Embryo, Mammalian , Female , Gene Expression Profiling , Gene Knock-In Techniques , Male , Mice , Mice, Transgenic , Models, Animal , Receptors, Dopamine D2/genetics , Repressor Proteins/genetics , Sequence Analysis, RNA
5.
Cancer Cell ; 31(4): 591-606.e6, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28344040

ABSTRACT

The dynamic and reversible N6-methyladenosine (m6A) RNA modification installed and erased by N6-methyltransferases and demethylases regulates gene expression and cell fate. We show that the m6A demethylase ALKBH5 is highly expressed in glioblastoma stem-like cells (GSCs). Silencing ALKBH5 suppresses the proliferation of patient-derived GSCs. Integrated transcriptome and m6A-seq analyses revealed altered expression of certain ALKBH5 target genes, including the transcription factor FOXM1. ALKBH5 demethylates FOXM1 nascent transcripts, leading to enhanced FOXM1 expression. Furthermore, a long non-coding RNA antisense to FOXM1 (FOXM1-AS) promotes the interaction of ALKBH5 with FOXM1 nascent transcripts. Depleting ALKBH5 and FOXM1-AS disrupted GSC tumorigenesis through the FOXM1 axis. Our work uncovers a critical function for ALKBH5 and provides insight into critical roles of m6A methylation in glioblastoma.


Subject(s)
AlkB Homolog 5, RNA Demethylase/metabolism , Brain Neoplasms/pathology , Forkhead Box Protein M1/metabolism , Glioblastoma/pathology , 3' Untranslated Regions , AlkB Homolog 5, RNA Demethylase/genetics , Animals , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Cell Line, Tumor , Cell Proliferation , ELAV-Like Protein 1/genetics , ELAV-Like Protein 1/metabolism , Forkhead Box Protein M1/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioblastoma/genetics , Glioblastoma/mortality , Male , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Xenograft Model Antitumor Assays
6.
Neuro Oncol ; 19(4): 514-523, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28040710

ABSTRACT

Background: Glioblastoma (GBM) is one of the most common, aggressive, and invasive human brain tumors. There are few reliable mechanism-based therapeutic approaches for GBM patients. The transcriptional repressor RE1 silencing transcriptional factor (REST) regulates the oncogenic properties of a class of GBM stem-like cells (high-REST [HR]-GSCs) in humans. However, it has been unclear whether REST represses specific targets to regulate specific oncogenic functions or represses all targets with overlapping functions in GSCs. Methods: We used genome-wide, biochemical, and mouse intracranial tumorigenic assays to identify and determine functions of microRNA (miR) targets of REST in 2 independent HR-GSC lines. Results: Here we show that REST represses 2 major miR gene targets in HR-GSCs: miR-203, a new target, and miR-124, a known target. Gain of function of miR-124 or miR-203 in HR-GSCs increased survival in tumor-bearing mice. Importantly, the increased survival of tumor-bearing mice caused by knockdown of REST in HR-GSCs was reversed by double knockdown of REST and either miR-203 or miR-124, indicating that these 2 miRs are critical tumor suppressors that are repressed in REST-mediated tumorigenesis. We further show that while miR-124 and the REST-miR-124 pathways regulate self-renewal, apoptosis and invasion, miR-203 and the REST-miR-203 pathways regulate only invasion. We further identify and validate potential mRNA targets of miR-203 and miR-124 in REST-mediated HR-GSC tumor invasion. Conclusions: These findings indicate that REST regulates its miR gene targets with overlapping functions and suggest how REST maintains oncogenic competence in GSCs. These mechanisms could potentially be utilized to block REST-mediated GBM tumorigenesis.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Repressor Proteins/metabolism , Animals , Mice
7.
Neuro Oncol ; 18(5): 656-66, 2016 05.
Article in English | MEDLINE | ID: mdl-26374689

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.


Subject(s)
Brain Neoplasms/pathology , Cell Adhesion Molecules/metabolism , Cell Adhesion/physiology , Glioblastoma/pathology , MicroRNAs/metabolism , Neoplastic Stem Cells/pathology , Receptors, Cell Surface/metabolism , Animals , Brain Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Heterografts , Humans , Immunoblotting , Mice , Neoplastic Stem Cells/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Tumor Cells, Cultured
8.
J Neurosci ; 35(45): 15097-112, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26558781

ABSTRACT

Glioblastoma (GBM) is the most aggressive human brain tumor. Although several molecular subtypes of GBM are recognized, a robust molecular prognostic marker has yet to be identified. Here, we report that the stemness regulator Sox2 is a new, clinically important target of microRNA-21 (miR-21) in GBM, with implications for prognosis. Using the MiR-21-Sox2 regulatory axis, approximately half of all GBM tumors present in the Cancer Genome Atlas (TCGA) and in-house patient databases can be mathematically classified into high miR-21/low Sox2 (Class A) or low miR-21/high Sox2 (Class B) subtypes. This classification reflects phenotypically and molecularly distinct characteristics and is not captured by existing classifications. Supporting the distinct nature of the subtypes, gene set enrichment analysis of the TCGA dataset predicted that Class A and Class B tumors were significantly involved in immune/inflammatory response and in chromosome organization and nervous system development, respectively. Patients with Class B tumors had longer overall survival than those with Class A tumors. Analysis of both databases indicated that the Class A/Class B classification is a better predictor of patient survival than currently used parameters. Further, manipulation of MiR-21-Sox2 levels in orthotopic mouse models supported the longer survival of the Class B subtype. The MiR-21-Sox2 association was also found in mouse neural stem cells and in the mouse brain at different developmental stages, suggesting a role in normal development. Therefore, this mechanism-based classification suggests the presence of two distinct populations of GBM patients with distinguishable phenotypic characteristics and clinical outcomes. SIGNIFICANCE STATEMENT: Molecular profiling-based classification of glioblastoma (GBM) into four subtypes has substantially increased our understanding of the biology of the disease and has pointed to the heterogeneous nature of GBM. However, this classification is not mechanism based and its prognostic value is limited. Here, we identify a new mechanism in GBM (the miR-21-Sox2 axis) that can classify ∼50% of patients into two subtypes with distinct molecular, radiological, and pathological characteristics. Importantly, this classification can predict patient survival better than the currently used parameters. Further, analysis of the miR-21-Sox2 relationship in mouse neural stem cells and in the mouse brain at different developmental stages indicates that miR-21 and Sox2 are predominantly expressed in mutually exclusive patterns, suggesting a role in normal neural development.


Subject(s)
Brain Neoplasms/classification , Brain Neoplasms/metabolism , Glioblastoma/classification , Glioblastoma/metabolism , MicroRNAs/biosynthesis , SOXB1 Transcription Factors/biosynthesis , Animals , Biomarkers, Tumor/biosynthesis , Brain Neoplasms/diagnosis , Cells, Cultured , Glioblastoma/diagnosis , Humans , Male , Mice , Mice, Nude , Prognosis , Retrospective Studies , Survival Rate/trends
9.
Stem Cell Res ; 15(2): 305-11, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26209818

ABSTRACT

Our previous studies have shown that the regulatory network that maintains pluripotency in mouse embryonic stem cells (mESCs) is regulated in a context-dependent manner and can be modulated, at least in part, by re-calibration of an intracellular network of pluripotency factors as well as cues arising from the extracellular matrix. The transcriptional repressor REST represses miR-21 and, thus, regulates self-renewal in E14Tg2a.4 mESCs cultured in the absence of mouse embryonic fibroblast feeder cell effects. However, how miR-21 connects to the nuclear regulatory network has not been clear. Here, we show that miR-21, a direct target of REST-mediated repression, directly targets Sox2. Exogenously added miR-21 to mESCs decreases the expression of Sox2, decreasing mESC self-renewal, and this effect of miR-21 on mESC self-renewal can be blocked by expression of exogenous Sox2. Conversely, destabilization of Sox2 by miR-21 can be blocked by anti-miR-21. Thus, the REST-miR-21-Sox2 axis connects REST to the core nuclear pluripotency regulators in E14Tg2a.4 mESCs cultured in the absence of feeder cells.


Subject(s)
MicroRNAs/metabolism , Repressor Proteins/metabolism , SOXB1 Transcription Factors/metabolism , 3' Untranslated Regions , Animals , Base Sequence , Binding Sites , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Mice , MicroRNAs/chemistry , MicroRNAs/genetics , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , SOXB1 Transcription Factors/chemistry , SOXB1 Transcription Factors/genetics , Sequence Alignment
10.
Nat Commun ; 6: 6156, 2015 Feb 02.
Article in English | MEDLINE | ID: mdl-25639486

ABSTRACT

Aberrant activation of ß-catenin in the nucleus has been implicated in a variety of human cancers, but the fate of nuclear ß-catenin is unknown. Here we demonstrate that the tripartite motif-containing protein 33 (TRIM33), acting as an E3 ubiquitin ligase, reduces the abundance of nuclear ß-catenin protein. TRIM33-mediated ß-catenin is destabilized and is GSK-3ß or ß-TrCP independent. TRIM33 interacts with and ubiquitylates nuclear ß-catenin. Moreover, protein kinase Cδ, which directly phosphorylates ß-catenin at Ser715, is required for the TRIM33-ß-catenin interaction. The function of TRIM33 in suppressing tumour cell proliferation and brain tumour development depends on TRIM33-promoted ß-catenin degradation. In human glioblastoma specimens, endogenous TRIM33 levels are inversely correlated with ß-catenin. In summary, our findings identify TRIM33 as a tumour suppressor that can abolish tumour cell proliferation and tumorigenesis by degrading nuclear ß-catenin. This work suggests a new therapeutic strategy against human cancers caused by aberrant activation of ß-catenin.


Subject(s)
Transcription Factors/metabolism , beta Catenin/metabolism , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation , Humans , Mice , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Ubiquitination , beta Catenin/genetics
11.
Nat Commun ; 5: 5566, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25412762

ABSTRACT

Pyruvate kinase M2 (PKM2) is expressed at high levels during embryonic development and tumour progression and is important for cell growth. However, it is not known whether it directly controls cell division. Here, we found that Aurora B phosphorylates PKM2, but not PKM1, at T45; this phosphorylation is required for PKM2's localization and interaction with myosin light chain 2 (MLC2) in the contractile ring region of mitotic cells during cytokinesis. PKM2 phosphorylates MLC2 at Y118, which primes the binding of ROCK2 to MLC2 and subsequent ROCK2-dependent MLC2 S15 phosphorylation. PKM2-regulated MLC2 phosphorylation, which is greatly enhanced by EGF stimulation or EGFRvIII, K-Ras G12V and B-Raf V600E mutant expression, plays a pivotal role in cytokinesis, cell proliferation and brain tumour development. These findings underscore the instrumental function of PKM2 in oncogenic EGFR-, K-Ras- and B-Raf-regulated cytokinesis and tumorigenesis.


Subject(s)
Aurora Kinase B/metabolism , Cardiac Myosins/metabolism , Carrier Proteins/metabolism , Cytokinesis/physiology , Membrane Proteins/metabolism , Myosin Light Chains/metabolism , Pyruvate Kinase/metabolism , Thyroid Hormones/metabolism , Amides/pharmacology , Animals , Brain Neoplasms/genetics , Cardiac Myosins/genetics , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Leupeptins/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Nude , Mitosis/physiology , Myosin Light Chains/genetics , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras) , Pyridines/pharmacology , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/genetics , RNA Interference , RNA, Small Interfering , Thyroid Hormones/genetics , ras Proteins/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , Thyroid Hormone-Binding Proteins
12.
PLoS One ; 7(8): e43659, 2012.
Article in English | MEDLINE | ID: mdl-22952733

ABSTRACT

BACKGROUND: REST is abundantly expressed in mouse embryonic stem cells (ESCs). Many genome-wide analyses have found REST to be an integral part of the ESC pluripotency network. However, experimental systems have produced contradictory findings: (1) REST is required for the maintenance of ESC pluripotency and loss of REST causes increased expression of differentiation markers, (2) REST is not required for the maintenance of ESC pluripotency and loss of REST does not change expression of differentiation markers, and (3) REST is not required for the maintenance of ESC pluripotency but loss of REST causes decreased expression of differentiation markers. These reports highlight gaps in our knowledge of the ESC network. METHODS: Employing biochemical and genome-wide analyses of various culture conditions and ESC lines, we have attempted to resolve some of the discrepancies in the literature. RESULTS: We show that Rest+/- and Rest-/- AB-1 mutant ESCs, which did not exhibit a role of REST in ESC pluripotency when cultured in the presence of feeder cells, did show impaired self-renewal when compared with the parental cells under feeder-free culture conditions, but only in early passage cells. In late passage cells, both Rest+/- and Rest-/- AB-1 ESCs restored pluripotency, suggesting a passage and culture condition-dependent response. Genome-wide analysis followed by biochemical validation supported this response and further indicated that the restoration of pluripotency was associated by increased expression of the ESC pluripotency factors. E14Tg2a.4 ESCs with REST-knockdown, which earlier showed a REST-dependent pluripotency when cultured under feeder-free conditions, as well as Rest-/- AB-1 ESCs, showed no REST-dependent pluripotency when cultured in the presence of either feeder cells or laminin, indicating that extracellular matrix components can rescue REST's role in ESC pluripotency. CONCLUSIONS: REST regulates ESC pluripotency in culture condition- and ESC line-dependent fashion and ESC pluripotency needs to be evaluated in a context dependent manner.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Repressor Proteins/metabolism , Animals , Biomarkers/metabolism , Cell Culture Techniques , Cell Dedifferentiation , Cell Differentiation , Cell Line , Extracellular Space/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Genomics , Mice , Repressor Proteins/deficiency , Repressor Proteins/genetics , Time Factors
13.
PLoS One ; 7(8): e41522, 2012.
Article in English | MEDLINE | ID: mdl-22870228

ABSTRACT

BACKGROUND: Several studies have established Glioblastoma Multiforme (GBM) prognostic and predictive models based on age and Karnofsky Performance Status (KPS), while very few studies evaluated the prognostic and predictive significance of preoperative MR-imaging. However, to date, there is no simple preoperative GBM classification that also correlates with a highly prognostic genomic signature. Thus, we present for the first time a biologically relevant, and clinically applicable tumor Volume, patient Age, and KPS (VAK) GBM classification that can easily and non-invasively be determined upon patient admission. METHODS: We quantitatively analyzed the volumes of 78 GBM patient MRIs present in The Cancer Imaging Archive (TCIA) corresponding to patients in The Cancer Genome Atlas (TCGA) with VAK annotation. The variables were then combined using a simple 3-point scoring system to form the VAK classification. A validation set (N = 64) from both the TCGA and Rembrandt databases was used to confirm the classification. Transcription factor and genomic correlations were performed using the gene pattern suite and Ingenuity Pathway Analysis. RESULTS: VAK-A and VAK-B classes showed significant median survival differences in discovery (P = 0.007) and validation sets (P = 0.008). VAK-A is significantly associated with P53 activation, while VAK-B shows significant P53 inhibition. Furthermore, a molecular gene signature comprised of a total of 25 genes and microRNAs was significantly associated with the classes and predicted survival in an independent validation set (P = 0.001). A favorable MGMT promoter methylation status resulted in a 10.5 months additional survival benefit for VAK-A compared to VAK-B patients. CONCLUSIONS: The non-invasively determined VAK classification with its implication of VAK-specific molecular regulatory networks, can serve as a very robust initial prognostic tool, clinical trial selection criteria, and important step toward the refinement of genomics-based personalized therapy for GBM patients.


Subject(s)
Brain Neoplasms , Gene Expression Regulation , Glioblastoma , Magnetic Resonance Imaging , MicroRNAs , RNA, Neoplasm , Aged , Brain Neoplasms/classification , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/surgery , DNA Methylation , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Disease-Free Survival , Female , Glioblastoma/classification , Glioblastoma/diagnostic imaging , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/surgery , Humans , Male , MicroRNAs/biosynthesis , MicroRNAs/genetics , Middle Aged , Promoter Regions, Genetic , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Radiography , Survival Rate , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
14.
Stem Cells ; 30(3): 405-14, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22228704

ABSTRACT

Glioblastoma multiforme (GBM) tumors are the most common malignant primary brain tumors in adults. Although many GBM tumors are believed to be caused by self-renewing, glioblastoma-derived stem-like cells (GSCs), the mechanisms that regulate self-renewal and other oncogenic properties of GSCs are only now being unraveled. Here we showed that GSCs derived from GBM patient specimens express varying levels of the transcriptional repressor repressor element 1 silencing transcription factor (REST), suggesting heterogeneity across different GSC lines. Loss- and gain-of-function experiments indicated that REST maintains self-renewal of GSCs. High REST-expressing GSCs (HR-GSCs) produced tumors histopathologically distinct from those generated by low REST-expressing GSCs (LR-GSCs) in orthotopic mouse brain tumor models. Knockdown of REST in HR-GSCs resulted in increased survival in GSC-transplanted mice and produced tumors with higher apoptotic and lower invasive properties. Conversely, forced expression of exogenous REST in LR-GSCs produced decreased survival in mice and produced tumors with lower apoptotic and higher invasive properties, similar to HR-GSCs. Thus, based on our results, we propose that a novel function of REST is to maintain self-renewal and other oncogenic properties of GSCs and that REST can play a major role in mediating tumorigenicity in GBM.


Subject(s)
Glioblastoma/pathology , Neoplastic Stem Cells/metabolism , Repressor Proteins/physiology , Adipokines/metabolism , Animals , Apoptosis , Cell Movement , Cell Proliferation , Chitinase-3-Like Protein 1 , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioblastoma/metabolism , Lectins/metabolism , Male , Mice , Mice, Nude , Neoplasm Transplantation , RNA Interference , Repressor Proteins/genetics , Repressor Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Spheroids, Cellular/metabolism , Tumor Cells, Cultured
15.
PLoS One ; 6(10): e25451, 2011.
Article in English | MEDLINE | ID: mdl-21998659

ABSTRACT

BACKGROUND: Despite recent discoveries of new molecular targets and pathways, the search for an effective therapy for Glioblastoma Multiforme (GBM) continues. A newly emerged field, radiogenomics, links gene expression profiles with MRI phenotypes. MRI-FLAIR is a noninvasive diagnostic modality and was previously found to correlate with cellular invasion in GBM. Thus, our radiogenomic screen has the potential to reveal novel molecular determinants of invasion. Here, we present the first comprehensive radiogenomic analysis using quantitative MRI volumetrics and large-scale gene- and microRNA expression profiling in GBM. METHODS: Based on The Cancer Genome Atlas (TCGA), discovery and validation sets with gene, microRNA, and quantitative MR-imaging data were created. Top concordant genes and microRNAs correlated with high FLAIR volumes from both sets were further characterized by Kaplan Meier survival statistics, microRNA-gene correlation analyses, and GBM molecular subtype-specific distribution. RESULTS: The top upregulated gene in both the discovery (4 fold) and validation (11 fold) sets was PERIOSTIN (POSTN). The top downregulated microRNA in both sets was miR-219, which is predicted to bind to POSTN. Kaplan Meier analysis demonstrated that above median expression of POSTN resulted in significantly decreased survival and shorter time to disease progression (P<0.001). High POSTN and low miR-219 expression were significantly associated with the mesenchymal GBM subtype (P<0.0001). CONCLUSION: Here, we propose a novel diagnostic method to screen for molecular cancer subtypes and genomic correlates of cellular invasion. Our findings also have potential therapeutic significance since successful molecular inhibition of invasion will improve therapy and patient survival in GBM.


Subject(s)
Edema/genetics , Edema/pathology , Genomics/methods , Glioblastoma/genetics , Glioblastoma/pathology , Magnetic Resonance Imaging , Phenotype , Adult , Aged , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Edema/diagnosis , Female , Gene Expression Profiling , Glioblastoma/diagnosis , Humans , Image Processing, Computer-Assisted , Male , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness , Software , Survival Analysis
16.
Stem Cells ; 28(10): 1839-47, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20799335

ABSTRACT

Production of neurons from non-neural cells has far-reaching clinical significance. We previously found that myoblasts can be converted to a physiologically active neuronal phenotype by transferring a single recombinant transcription factor, REST-VP16, which directly activates target genes of the transcriptional repressor, REST. However, the neuronal subtype of M-RV cells and whether they can establish synaptic communication in the brain have remained unknown. M-RV cells engineered to express green fluorescent protein (M-RV-GFP) had functional ion channels but did not establish synaptic communication in vitro. However, when transplanted into newborn mice cerebella, a site of extensive postnatal neurogenesis, these cells expressed endogenous cerebellar granule precursors and neuron proteins, such as transient axonal glycoprotein-1, neurofilament, type-III ß-tubulin, superior cervical ganglia-clone 10, glutamate receptor-2, and glutamate decarboxylase. Importantly, they exhibited action potentials and were capable of receiving glutamatergic synaptic input, similar to the native cerebellar granule neurons. These results suggest that M-RV-GFP cells differentiate into glutamatergic neurons, an important neuronal subtype, in the postnatal cerebellar milieu. Our findings suggest that although activation of REST-target genes can reprogram myoblasts to assume a general neuronal phenotype, the subtype specificity may then be directed by the brain microenvironment.


Subject(s)
Cerebellum/cytology , Myoblasts/cytology , Neurons/cytology , Animals , Animals, Newborn , Calcium-Binding Proteins , Cell Differentiation , Contactin 2/genetics , Contactin 2/metabolism , Fluorescent Antibody Technique , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neurons/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stathmin , Tubulin/genetics , Tubulin/metabolism
17.
Nature ; 453(7192): 223-7, 2008 May 08.
Article in English | MEDLINE | ID: mdl-18362916

ABSTRACT

The neuronal repressor REST (RE1-silencing transcription factor; also called NRSF) is expressed at high levels in mouse embryonic stem (ES) cells, but its role in these cells is unclear. Here we show that REST maintains self-renewal and pluripotency in mouse ES cells through suppression of the microRNA miR-21. We found that, as with known self-renewal markers, the level of REST expression is much higher in self-renewing mouse ES cells than in differentiating mouse ES (embryoid body, EB) cells. Heterozygous deletion of Rest (Rest+/-) and its short-interfering-RNA-mediated knockdown in mouse ES cells cause a loss of self-renewal-even when these cells are grown under self-renewal conditions-and lead to the expression of markers specific for multiple lineages. Conversely, exogenously added REST maintains self-renewal in mouse EB cells. Furthermore, Rest+/- mouse ES cells cultured under self-renewal conditions express substantially reduced levels of several self-renewal regulators, including Oct4 (also called Pou5f1), Nanog, Sox2 and c-Myc, and exogenously added REST in mouse EB cells maintains the self-renewal phenotypes and expression of these self-renewal regulators. We also show that in mouse ES cells, REST is bound to the gene chromatin of a set of miRNAs that potentially target self-renewal genes. Whereas mouse ES cells and mouse EB cells containing exogenously added REST express lower levels of these miRNAs, EB cells, Rest+/- ES cells and ES cells treated with short interfering RNA targeting Rest express higher levels of these miRNAs. At least one of these REST-regulated miRNAs, miR-21, specifically suppresses the self-renewal of mouse ES cells, corresponding to the decreased expression of Oct4, Nanog, Sox2 and c-Myc. Thus, REST is a newly discovered element of the interconnected regulatory network that maintains the self-renewal and pluripotency of mouse ES cells.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Biomarkers , Cell Differentiation , Cell Line , Cell Lineage , Cell Proliferation , Chromatin/genetics , Chromatin/metabolism , Mice , Mice, Inbred C57BL , Repressor Proteins/genetics , Transcription Factors/deficiency , Transcription Factors/genetics
18.
Cell Cycle ; 5(17): 1929-35, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16929174

ABSTRACT

The repressor element 1 (RE-1)-silencing transcription factor (REST), also known as the neuron-restrictive silencer factor (NRSF), was originally discovered as a transcriptional repressor of a large number of primarily terminal neuronal differentiation genes in nonneuronal cells and neural stem cells (NSCs). Although REST is expressed in NSCs, its transcription is generally blocked as NSCs undergo differentiation, and it is rarely expressed in terminally differentiated neurons. In support of its function as a transcriptional repressor, REST was found to contain a DNA-binding domain and two repressor domains. The repressor domains were found to associate, directly or indirectly, with a large number of cellular repressor complexes. Thus, REST was considered a major epigenetic regulator controlling chromatin modification. However, REST is expressed in some differentiated neurons, and when bound to a double-stranded small RNA, REST was later found to also function as an activator of its same target neuronal differentiation genes in NSCs. In addition, REST has been found to regulate an evolving array of genes and cellular functions, making it a biological enigma. For example, REST was recently found to have a seemingly paradoxical role in both tumor suppressor activity and oncogenic activity. Current evidence suggests that the diverse cellular context generated by intrinsic factors in the cell, the amount of REST protein present in the cell, the affinity of the REST protein for its specific target gene, and the cellular niche dictate such behavior.


Subject(s)
Oncogene Proteins/physiology , Repressor Proteins/physiology , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Differentiation , Cerebellar Neoplasms/etiology , Humans , Medulloblastoma/etiology , Mice , Nerve Regeneration , Neuronal Plasticity , Neurons/cytology
19.
Mol Cell Biol ; 26(5): 1666-78, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16478988

ABSTRACT

Medulloblastoma, one of the most malignant brain tumors in children, is thought to arise from undifferentiated neural stem/progenitor cells (NSCs) present in the external granule layer of the cerebellum. However, the mechanism of tumorigenesis remains unknown for the majority of medulloblastomas. In this study, we found that many human medulloblastomas express significantly elevated levels of both myc oncogenes, regulators of neural progenitor proliferation, and REST/NRSF, a transcriptional repressor of neuronal differentiation genes. Previous studies have shown that neither c-Myc nor REST/NRSF alone could cause tumor formation. To determine whether c-Myc and REST/NRSF act together to cause medulloblastomas, we used a previously established cell line derived from external granule layer stem cells transduced with activated c-myc (NSC-M). These immortalized NSCs were able to differentiate into neurons in vitro. In contrast, when the cells were engineered to express a doxycycline-regulated REST/NRSF transgene (NSC-M-R), they no longer underwent terminal neuronal differentiation in vitro. When injected into intracranial locations in mice, the NSC-M cells did not form tumors either in the cerebellum or in the cerebral cortex. In contrast, the NSC-M-R cells did produce tumors in the cerebellum, the site of human medulloblastoma formation, but not when injected into the cerebral cortex. Furthermore, the NSC-M-R tumors were blocked from terminal neuronal differentiation. In addition, countering REST/NRSF function blocked the tumorigenic potential of NSC-M-R cells. To our knowledge, this is the first study in which abnormal expression of a sequence-specific DNA-binding transcriptional repressor has been shown to contribute directly to brain tumor formation. Our findings indicate that abnormal expression of REST/NRSF and Myc in NSCs causes cerebellum-specific tumors by blocking neuronal differentiation and thus maintaining the "stemness" of these cells. Furthermore, these results suggest that such a mechanism plays a role in the formation of human medulloblastoma.


Subject(s)
Cerebellar Neoplasms/genetics , Genes, myc , Medulloblastoma/genetics , Neurons/pathology , Repressor Proteins/genetics , Stem Cells/physiology , Transcription Factors/genetics , Adolescent , Adult , Animals , Carcinogenicity Tests , Cell Differentiation/genetics , Cerebellar Neoplasms/pathology , Child , Child, Preschool , Doxycycline/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Medulloblastoma/pathology , Mice , Mice, Nude , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/metabolism , Stem Cells/pathology , Transcription Factors/metabolism
20.
Mol Cancer Ther ; 4(3): 343-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15767543

ABSTRACT

Medulloblastoma, one of the most malignant pediatric brain tumors, is believed to arise from the undifferentiated external granule-layer cells in the cerebellum. It is a heterogeneous cancer, and the mechanism of tumorigenesis for the majority of types is unknown. Repressor element-1 silencing transcription/neuron-restrictive silencer factor (REST/NRSF) is a transcriptional repressor that can block transcription of a battery of neuronal differentiation genes by binding to a specific consensus DNA sequence present in their regulatory region. Previously, we found that some medulloblastoma cell lines express REST/NRSF at high levels compared with either neuronal progenitor cells or fully differentiated neurons. However, it is not known if REST/NRSF is indeed overexpressed in human medulloblastoma tumor specimens and in what frequency. Here, we did an immunohistochemical analysis of such tumor specimens using an anti-REST antibody. We show that among 21 human medulloblastoma tumors, 17 expressed REST/NRSF (6 strongly and 11 weakly). In contrast, adjacent normal cerebellum tissue sections and four of the tumor specimens did not express REST/NRSF, indicating that abnormal expression of REST/NRSF is observed in the majority of human medulloblastoma tumors. To determine whether countering REST/NRSF activity blocks tumorigenicity of medulloblastoma cells, especially in the intracranial (i.c.) environment, we found that adenovirus-mediated expression of REST-VP16, a recombinant transcription factor that can compete with REST/NRSF and activate REST/NRSF target genes instead of repressing them, blocked the i.c. tumorigenic potential of medulloblastoma cells and inhibited growth of established tumors in nude mice, suggesting that REST/NRSF may serve as a therapeutic target for medulloblastoma and that forced expression of neuronal differentiation genes in medulloblastoma cells through agents, such as REST-VP16, can interfere with their tumorigenicity.


Subject(s)
Brain Neoplasms/metabolism , Etoposide/pharmacology , Medulloblastoma/metabolism , Repressor Proteins/biosynthesis , Transcription Factors/biosynthesis , Adenoviridae/genetics , Animals , Apoptosis , Brain/metabolism , Brain/pathology , Cell Differentiation , Cell Line, Tumor , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Mice, Nude , Models, Biological , Neoplasm Transplantation , Neurons/metabolism , Stem Cells/metabolism , Time Factors , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...