Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Oncotarget ; 15: 175-198, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38484151

ABSTRACT

Our understanding of DNA G-quadruplexes (G4s) from in vitro studies has been complemented by genome-wide G4 landscapes from cultured cells. Conventionally, the formation of G4s is accepted to depend on G-repeats such that they form tetrads. However, genome-wide G4s characterized through high-throughput sequencing suggest that these structures form at a large number of regions with no such canonical G4-forming signatures. Many G4-binding proteins have been described with no evidence for any protein that binds to and stabilizes G4s. It remains unknown what fraction of G4s formed in human cells are protein-bound. The G4-chromatin immunoprecipitation (G4-ChIP) method hitherto employed to describe G4 landscapes preferentially reports G4s that get crosslinked to proteins in their proximity. Our current understanding of the G4 landscape is biased against representation of G4s which escape crosslinking as they are not stabilized by protein-binding and presumably transient. We report a protocol that captures G4s from the cells efficiently without any bias as well as eliminates the detection of G4s formed artifactually on crosslinked sheared chromatin post-fixation. We discover that G4s form sparingly at SINEs. An application of this method shows that depletion of a repeat-binding protein CGGBP1 enhances net G4 capture at CGGBP1-dependent CTCF-binding sites and regions of sharp interstrand G/C-skew transitions. Thus, we present an improved method for G4 landscape determination and by applying it we show that sequence property-specific constraints of the nuclear environment mitigate G4 formation.


Subject(s)
G-Quadruplexes , Humans , Chromatin , Genome , Antibodies , Protein Binding , DNA-Binding Proteins/genetics
2.
J Med Virol ; 95(3): e28675, 2023 03.
Article in English | MEDLINE | ID: mdl-36929720

ABSTRACT

Japanese encephalitis virus (JEV) is the foremost cause of viral encephalitis in Southeast Asia and Australia leading to approximately 68 000 clinical cases and about 13 600-20 400 deaths annually. Vaccination is not completely sure and safe. Despite this, no specific antiviral has been available or approved for JEV infection yet and treatment is generally symptomatic. Therefore, this study aims to examine the antiviral activity of natural compounds against JEV proteins. The antiviral activity of natural compounds was investigated via molecular docking, cytopathic effect (CPE) inhibition assay, western blotting, and indirect immunofluorescence assay. Physiochemical, pharmacokinetics, and toxicity analysis were evaluated for the safety and efficacy of natural compounds. Network pharmacology-based approaches have been used to study the molecular mechanisms of drug-target interactions. Molecular docking results suggested that the NS5 protein of JEV is the major target for natural compounds. Network pharmacology-based analysis revealed that these drugs majorly target IL6, AKT1, tumor necrosis factor (TNF), and PTGS2 to regulate key immune and inflammatory pathways such as nuclear factor kappa B, PI3K-Akt, and TNF signaling, during JEV infection. Our in vitro results show that among the natural compounds, curcumin provides the highest protection against JEV infection via reducing the JEV-induced CPE (IC50 = 5.90 ± 0.44 µM/mL), and reduces the expression of NS5 protein, IL6, AKT1, TNF-α, and PTGS2. However, other natural compounds also provide protection to some extent but their efficacy is lower compared to curcumin. Therefore, this study shows that natural compounds, mainly curcumin, may offer novel therapeutic avenues for the treatment of JEV via inhibiting key viral proteins and regulating crucial host pathways involved in JEV replication.


Subject(s)
Curcumin , Encephalitis Virus, Japanese , Encephalitis, Japanese , Humans , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/pharmacology , Cyclooxygenase 2/therapeutic use , Molecular Docking Simulation , Curcumin/pharmacology , Curcumin/therapeutic use , Interleukin-6 , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Virus Replication
3.
Oncotarget ; 13: 136-155, 2022.
Article in English | MEDLINE | ID: mdl-35070079

ABSTRACT

Genomes of somatic cells in culture are prone to spontaneous mutations due to errors in replication and DNA repair. Some of these errors, such as chromosomal fusions, are not rectifiable and subject to selection or elimination in growing cultures. Somatic cell cultures are thus expected to generate background levels of potentially stable chromosomal chimeras. A description of the landscape of such spontaneously generated chromosomal chimeras in cultured cells will help understand the factors affecting somatic mosaicism. Here we show that short homology-associated non-homologous chromosomal chimeras occur in normal human fibroblasts and HEK293T cells at genomic repeats. The occurrence of chromosomal chimeras is enhanced by heat stress and depletion of a repeat regulatory protein CGGBP1. We also present evidence of homologous chromosomal chimeras between allelic copies in repeat-rich DNA obtained by methylcytosine immunoprecipitation. The formation of homologous chromosomal chimeras at Alu and L1 repeats increases upon depletion of CGGBP1. Our data are derived from de novo sequencing from three different cell lines under different experimental conditions and our chromosomal chimera detection pipeline is applicable to long as well as short read sequencing platforms. These findings present significant information about the generation, sensitivity and regulation of somatic mosaicism in human cell cultures.


Subject(s)
Chimera , DNA , Cell Culture Techniques , Chromosomes , DNA-Binding Proteins , Genomics , HEK293 Cells , Humans
4.
Cell Cycle ; 20(22): 2387-2401, 2021 11.
Article in English | MEDLINE | ID: mdl-34585631

ABSTRACT

Binding sites of the chromatin regulator protein CTCF function as important landmarks in the human genome. The recently characterized CTCF-binding sites at LINE-1 repeats depend on another repeat-regulatory protein CGGBP1. These CGGBP1-dependent CTCF-binding sites serve as potential barrier elements for epigenetic marks such as H3K9me3. Such CTCF-binding sites are associated with asymmetric H3K9me3 levels as well as RNA levels in their flanks. The functions of these CGGBP1-dependent CTCF-binding sites remain unknown. By performing targeted studies on candidate CGGBP1-dependent CTCF-binding sites cloned in an SV40 promoter-enhancer episomal system we show that these regions act as inhibitors of ectopic transcription from the SV40 promoter. CGGBP1-dependent CTCF-binding sites that recapitulate their genomic function of loss of CTCF binding upon CGGBP1 depletion and H3K9me3 asymmetry in immediate flanks are also the ones that show the strongest inhibition of ectopic transcription. By performing a series of strand-specific reverse transcription PCRs we demonstrate that this ectopic transcription results in the synthesis of RNA from the SV40 promoter in a direction opposite to the downstream reporter gene in a strand-specific manner. The unleashing of the bidirectionality of the SV40 promoter activity and a breach of the transcription barrier seems to depend on depletion of CGGBP1 and loss of CTCF binding proximal to the SV40 promoter. RNA-sequencing reveals that CGGBP1-regulated CTCF-binding sites act as barriers to transcription at multiple locations genome-wide. These findings suggest a role of CGGBP1-dependent binding sites in restricting ectopic transcription.


Subject(s)
CCCTC-Binding Factor , Chromatin , DNA-Binding Proteins , Transcription Factors , Binding Sites , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genome, Human , Humans , Promoter Regions, Genetic , Transcription Factors/metabolism
5.
BMC Cancer ; 20(1): 1016, 2020 Oct 20.
Article in English | MEDLINE | ID: mdl-33081720

ABSTRACT

BACKGROUND: Inhibition of DNA-binding of proteins by small-molecule chemicals holds immense potential in manipulating the activities of DNA-binding proteins. Such a chemical inhibition of DNA-binding of proteins can be used to modulate processes such as replication, transcription, DNA repair and maintenance of epigenetic states. This prospect is currently challenged with the absence of robust and generic protocols to identify DNA-protein interactions. Additionally, much of the current approaches to designing inhibitors requires structural information of the target proteins. METHODS: We have developed a simple dot blot and immunodetection-based assay to screen chemical libraries for inhibitors of DNA-protein interactions. The assay has been applied to a library of 1685 FDA-approved chemicals to discover inhibitors of CGGBP1, a multifunctional DNA-binding protein with no known structure. Additional in vitro and in cellulo assays have been performed to verify and supplement the findings of the screen. RESULTS: Our primary screen has identified multiple inhibitors of direct or indirect interactions between CGGBP1 and genomic DNA. Of these, one inhibitor, Givinostat, was found to inhibit direct DNA-binding of CGGBP1 in the secondary screen using purified recombinant protein as the target. DNA and chromatin immunoprecipitation assays reinforced the findings of the screen that Givinostat inhibits CGGBP1-DNA binding. CONCLUSIONS: The assay we have described successfully identifies verifiable inhibitors of DNA-binding of protein; in this example, the human CGGBP1. This assay is customizable for a wide range of targets for which primary antibodies are available. It works with different sources of the target protein, cell lysates or purified recombinant preparations and does not require special equipment, DNA modifications or protein structural data. This assay is scalable and highly adaptable with the potential to discover inhibitors of transcription factors with implications in cancer biology.


Subject(s)
Carbamates/pharmacology , DNA-Binding Proteins/metabolism , DNA/metabolism , DNA-Binding Proteins/chemistry , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Immunoblotting , Protein Binding/drug effects , Small Molecule Libraries/pharmacology
6.
BMC Genet ; 21(1): 84, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32727353

ABSTRACT

BACKGROUND: The human CGGBP1 binds to GC-rich regions and interspersed repeats, maintains homeostasis of stochastic cytosine methylation and determines DNA-binding of CTCF. Interdependence between regulation of cytosine methylation and CTCF occupancy by CGGBP1 remains unknown. RESULTS: By analyzing methylated DNA-sequencing data obtained from CGGBP1-depleted cells, we report that some transcription factor-binding sites, including CTCF, resist stochastic changes in cytosine methylation. By analysing CTCF-binding sites we show that cytosine methylation changes at CTCF motifs caused by CGGBP1 depletion resist stochastic changes. These CTCF-binding sites are positioned at locations where the spread of cytosine methylation in cis depends on the levels of CGGBP1. CONCLUSION: Our findings suggest that CTCF occupancy and functions are determined by CGGBP1-regulated cytosine methylation patterns.


Subject(s)
Binding Sites , Cytosine/chemistry , DNA Methylation , DNA-Binding Proteins/genetics , Alleles , Chromosome Mapping , HEK293 Cells , Humans , Sequence Analysis, DNA , Transduction, Genetic
7.
Epigenetics Chromatin ; 12(1): 57, 2019 09 23.
Article in English | MEDLINE | ID: mdl-31547883

ABSTRACT

BACKGROUND: CGGBP1 is a repeat-binding protein with diverse functions in the regulation of gene expression, cytosine methylation, repeat silencing and genomic integrity. CGGBP1 has also been identified as a cooperator of histone-modifying enzymes and as a component of CTCF-containing complexes that regulate the enhancer-promoter looping. CGGBP1-CTCF cross talk in chromatin regulation has been hitherto unknown. RESULTS: Here, we report that the occupancy of CTCF at repeats depends on CGGBP1. Using ChIP-sequencing for CTCF, we describe its occupancy at repetitive DNA. Our results show that endogenous level of CGGBP1 ensures CTCF occupancy preferentially on repeats over canonical CTCF motifs. By combining CTCF ChIP-sequencing results with ChIP sequencing for three different kinds of histone modifications (H3K4me3, H3K9me3 and H3K27me3), we show that the CGGBP1-dependent repeat-rich CTCF-binding sites regulate histone marks in flanking regions. CONCLUSION: CGGBP1 affects the pattern of CTCF occupancy. Our results posit CGGBP1 as a regulator of CTCF and its binding sites in interspersed repeats.


Subject(s)
CCCTC-Binding Factor/metabolism , DNA-Binding Proteins/metabolism , Binding Sites , CCCTC-Binding Factor/chemistry , Cell Line , Cell Nucleus/metabolism , Chromatin/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Histones/metabolism , Humans , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering/metabolism
8.
Cancer Inform ; 18: 1176935119843835, 2019.
Article in English | MEDLINE | ID: mdl-31037026

ABSTRACT

Genome-wide occupancy of several histone modifications in various cell types has been studied using chromatin immunoprecipitation (ChIP) sequencing. Histone occupancy depends on DNA sequence features like inter-strand symmetry of base composition and periodic occurrence of TT/AT. However, whether DNA sequence motifs act as an additional effector of histone occupancy is not known. We have analyzed the presence of DNA sequence motifs in publicly available ChIP-sequence datasets for different histone modifications. Our results show that DNA sequence motifs are associated with histone occupancy, some of which are different between primary and transformed cells. The motifs for primary and transformed cells showed different levels of GC-richness and proximity to transcription start sites (TSSs). The TSSs associated with transformed or primary cell-specific motifs showed different levels of TSS flank transcription in primary and transformed cells. Interestingly, TSSs with a motif-linked occupancy of H2AFZ, a component of positioned nucleosomes, showed a distinct pattern of RNA Polymerase II (POLR2A) occupancy and TSS flank transcription in primary and transformed cells. These results indicate that DNA sequence features dictate differential histone occupancy in primary and transformed cells, and the DNA sequence motifs affect transcription through regulation of histone occupancy.

9.
BMC Res Notes ; 11(1): 419, 2018 Jul 02.
Article in English | MEDLINE | ID: mdl-29966527

ABSTRACT

OBJECTIVES: Although CpG methylation is well studied, mechanisms of non-CpG methylation in mammals remains elusive. Studying proteins with non-CpG cytosine methylation-sensitive DNA-binding, such as human CGGBP1, can unveil cytosine methylation regulatory mechanisms. Here we have resequenced a published genome-wide bisulfite sequencing library and analyzed it at base level resolution. CpG, CHG and CHH (where H is any nucleotide other than G) methylation states in non-targeting or CGGBP1-targeting shmiR lentivirus-transduced cells have been analyzed to identify how CGGBP1 regulates CpG and non-CpG methylation. RESULTS: We report that CGGBP1 acts as a dynamic bimodal balancer of methylation. Both gain and loss of methylation observed upon CGGBP1 depletion were spatially overlapping at annotated functional regions and not identifiable with any sequence motifs but clearly associated with GC-skew. CGGBP1 depletion caused clustered methylation changes in cis, upstream of R-loop forming promoters. This was complemented by clustered occurrences of methylation changes in proximity of transcription start sites of known cytosine methylation regulatory genes, altered expression of which can regulate cytosine methylation in trans. Despite low coverage, our data provide reliable estimates of the spectrum of methylation changes regulated by CGGBP1 in all cytosine contexts genome-wide through a combination of cis and trans-acting mechanisms.


Subject(s)
CpG Islands , DNA Methylation , DNA-Binding Proteins/genetics , Cytosine , Genome , Humans , Loss of Function Mutation , Promoter Regions, Genetic , Sequence Analysis, DNA
10.
Oncotarget ; 7(6): 6809-23, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26755663

ABSTRACT

Multiple myeloma (MM) is a malignancy of the antibody-producing plasma cells. MM is a highly heterogeneous disease, which has hampered the identification of a common underlying mechanism for disease establishment as well as the development of targeted therapy. Here we present the first genome-wide profiling of histone H3 lysine 27 and lysine 4 trimethylation in MM patient samples, defining a common set of active H3K4me3-enriched genes and silent genes marked by H3K27me3 (H3K27me3 alone or bivalent) unique to primary MM cells, when compared to normal bone marrow plasma cells. Using this epigenome profile, we found increased silencing of H3K27me3 targets in MM patients at advanced stages of the disease, and the expression pattern of H3K27me3-marked genes correlated with poor patient survival. We also demonstrated that pharmacological inhibition of EZH2 had anti-myeloma effects in both MM cell lines and CD138+ MM patient cells. In addition, EZH2 inhibition decreased the global H3K27 methylation and induced apoptosis. Taken together, these data suggest an important role for the Polycomb repressive complex 2 (PRC2) in MM, and highlights the PRC2 component EZH2 as a potential therapeutic target in MM.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Histones/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Polycomb-Group Proteins/genetics , Chromatin/metabolism , Down-Regulation , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Profiling , Histones/genetics , Humans , Lysine/metabolism , Methylation , Molecular Targeted Therapy , Multiple Myeloma/metabolism , Polycomb-Group Proteins/metabolism
11.
Cell Cycle ; 15(12): 1558-71, 2016 06 17.
Article in English | MEDLINE | ID: mdl-25483050

ABSTRACT

CGGBP1 (CGG triplet repeat-binding protein 1) regulates cell proliferation, stress response, cytokinesis, telomeric integrity and transcription. It could affect these processes by modulating target gene expression under different conditions. Identification of CGGBP1-target genes and their regulation could reveal how a transcription regulator affects such diverse cellular processes. Here we describe the mechanisms of differential gene expression regulation by CGGBP1 in quiescent or growing cells. By studying global gene expression patterns and genome-wide DNA-binding patterns of CGGBP1, we show that a possible mechanism through which it affects the expression of RNA Pol II-transcribed genes in trans depends on Alu RNA. We also show that it regulates Alu transcription in cis by binding to Alu promoter. Our results also indicate that potential phosphorylation of CGGBP1 upon growth stimulation facilitates its nuclear retention, Alu-binding and dislodging of RNA Pol III therefrom. These findings provide insights into how Alu transcription is regulated in response to growth signals.


Subject(s)
Alu Elements , DNA-Binding Proteins/genetics , Fibroblasts/metabolism , RNA Polymerase II/genetics , Transcription, Genetic , Base Sequence , Biological Factors/pharmacology , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Profiling , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Lentivirus/genetics , Lentivirus/metabolism , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , RNA Polymerase II/metabolism , Sequence Alignment , Serum/chemistry
12.
Ups J Med Sci ; 120(4): 219-32, 2015.
Article in English | MEDLINE | ID: mdl-26482656

ABSTRACT

The human genome contains multiple stretches of CGG trinucleotide repeats, which act as transcription- and translation-regulatory elements but at the same time form secondary structures that impede replication and give rise to sites of chromosome fragility. Proteins binding to such DNA elements may be involved in divergent cellular processes such as transcription, DNA damage, and epigenetic state of the chromatin. We review here the work done on CGG repeats and associated proteins with special focus on a factor called CGGBP1. CGGBP1 presents with an interesting example of factors that do not have any single dedicated function, but participate indispensably in multiple processes. Both experimental results and data from cancer genome sequencing have revealed that any alteration in CGGBP1 that compromises its function is not tolerated by normal or cancer cells alike. Based upon a large amount of published data, information from databases, and unpublished results, we decipher in this review how CGGBP1 is a classic example of the 'one factor, divergent functions' paradigm of cytoprotection. By taking cues from the studies on CGGBP1, more such factors can be discovered for a better understanding of the evolution of mechanisms of cellular survival.


Subject(s)
Brain Neoplasms/genetics , Cell Survival/genetics , Cytoprotection/genetics , DNA-Binding Proteins/genetics , Genetic Predisposition to Disease , Animals , Brain Neoplasms/physiopathology , Carcinogenesis/genetics , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Genome, Human , Humans , Sensitivity and Specificity
13.
BMC Genomics ; 16: 390, 2015 May 16.
Article in English | MEDLINE | ID: mdl-25981527

ABSTRACT

BACKGROUND: CGGBP1 is a repetitive DNA-binding transcription regulator with target sites at CpG-rich sequences such as CGG repeats and Alu-SINEs and L1-LINEs. The role of CGGBP1 as a possible mediator of CpG methylation however remains unknown. At CpG-rich sequences cytosine methylation is a major mechanism of transcriptional repression. Concordantly, gene-rich regions typically carry lower levels of CpG methylation than the repetitive elements. It is well known that at interspersed repeats Alu-SINEs and L1-LINEs high levels of CpG methylation constitute a transcriptional silencing and retrotransposon inactivating mechanism. RESULTS: Here, we have studied genome-wide CpG methylation with or without CGGBP1-depletion. By high throughput sequencing of bisulfite-treated genomic DNA we have identified CGGBP1 to be a negative regulator of CpG methylation at repetitive DNA sequences. In addition, we have studied CpG methylation alterations on Alu and L1 retrotransposons in CGGBP1-depleted cells using a novel bisulfite-treatment and high throughput sequencing approach. CONCLUSIONS: The results clearly show that CGGBP1 is a possible bidirectional regulator of CpG methylation at Alus, and acts as a repressor of methylation at L1 retrotransposons.


Subject(s)
Cytosine/chemistry , DNA Methylation/genetics , DNA-Binding Proteins/metabolism , DNA/metabolism , Repetitive Sequences, Nucleic Acid , Alu Elements/genetics , Cell Line , CpG Islands/genetics , Cytosine/metabolism , DNA/chemistry , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , High-Throughput Nucleotide Sequencing , Humans , Retroelements/genetics , Sequence Analysis, DNA
14.
Cell Cycle ; 13(1): 96-105, 2014.
Article in English | MEDLINE | ID: mdl-24196442

ABSTRACT

The shelterin proteins are required for telomere integrity. Shelterin dysfunction can lead to initiation of unwarranted DNA damage and repair pathways at chromosomal termini. Interestingly, many shelterin accessory proteins are involved in DNA damage signaling and repair. We demonstrate here that in normal human fibroblasts, telomeric ends are protected by phosphorylation of CGG triplet repeat-binding protein 1 (CGGBP1) at serine 164 (S164). We show that serine 164 is a major phosphorylation site on CGGBP1 with important functions. We provide evidence that one of the kinases that can phosphorylate S164 CGGBP1 is ATR. Overexpression of S164A phospho-deficient CGGBP1 exerted a dominant-negative effect, causing telomeric dysfunction, accelerated telomere shortening, enhanced fusion of telomeres, and crisis. However, overexpression of wild-type or phospho-mimicking S164E CGGBP1 did not cause these effects. This telomere damage was associated with reduced binding of the shelterin protein POT1 to telomeric DNA. Our results suggest that CGGBP1 phosphorylation at S164 is a novel telomere protection signal, which can affect telomere-protective function of the shelterin complex.


Subject(s)
DNA Damage/genetics , DNA-Binding Proteins/genetics , Serine/genetics , Telomere/genetics , Cell Cycle Proteins/genetics , Gene Expression Regulation , Humans , Phosphorylation , Shelterin Complex , Signal Transduction/genetics , Telomere-Binding Proteins/genetics
15.
DNA Repair (Amst) ; 12(11): 876-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24054699

ABSTRACT

Human POT1, a widely studied telomere protector protein is perceived to be expressed as a single 70kDa form. A survey of the literature as well as different commercially available antibodies against POT1 suggests occurrence of multiple forms of POT1. Knowledge about possible various forms of an important protein like POT1 is necessary for our understanding about its function. We have discovered that POT1 exists in at least three consistently occurring forms; 90, 70 and 45kDa. The unexpected molecular weights of POT1 seem to be associated with SUMO1 and ubiquitin conjugation; the latter occurring at a double lysine residue at 289-KK-290. We also present evidence that the relative abundance of the different POT1 forms can be altered by experimental modulation of POT1 nuclear localization. We thus present strong evidence that there are post-translational modifications of POT1 that can affect its molecular weight as well as intracellular localization and function.


Subject(s)
Protein Isoforms/metabolism , SUMO-1 Protein/metabolism , Telomere-Binding Proteins/chemistry , Telomere-Binding Proteins/metabolism , Cell Nucleus/metabolism , Gene Expression Regulation , Humans , Lysine/metabolism , Molecular Weight , Protein Isoforms/genetics , Shelterin Complex , Sumoylation , Telomere-Binding Proteins/genetics , Ubiquitin/metabolism
16.
BMC Cancer ; 12: 378, 2012 Aug 29.
Article in English | MEDLINE | ID: mdl-22931209

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) and their role during tumor development have been studied in great detail during the last decade, albeit their expression pattern and regulation during normal development are however not so well established. Previous studies have shown that miRNAs are differentially expressed in solid human tumors. Platelet-derived growth factor (PDGF) signaling is known to be involved in normal development of the brain as well as in malignant primary brain tumors, gliomas, but the complete mechanism is still lacking. We decided to investigate the expression of the oncogenic miR-21 during normal mouse development and glioma, focusing on PDGF signaling as a potential regulator of miR-21. METHODS: We generated mouse glioma using the RCAS/tv-a system for driving PDGF-BB expression in a cell-specific manner. Expression of miR-21 in mouse cell cultures and mouse brain were assessed using Northern blot analysis and in situ hybridization. Immunohistochemistry and Western blot analysis were used to investigate SOX2 expression. LNA-modified siRNA was used for irreversible depletion of miR-21. For inhibition of PDGF signaling Gleevec (imatinib mesylate), Rapamycin and U0126, as well as siRNA were used. Statistical significance was calculated using double-sided unpaired Student's t-test. RESULTS: We identified miR-21 to be highly expressed during embryonic and newborn brain development followed by a gradual decrease until undetectable at postnatal day 7 (P7), this pattern correlated with SOX2 expression. Furthermore, miR-21 and SOX2 showed up-regulation and overlapping expression pattern in RCAS/tv-a generated mouse brain tumor specimens. Upon irreversible depletion of miR-21 the expression of SOX2 was strongly diminished in both mouse primary glioma cultures and human glioma cell lines. Interestingly, in normal fibroblasts the expression of miR-21 was induced by PDGF-BB, and inhibition of PDGF signaling in mouse glioma primary cultures resulted in suppression of miR-21 suggesting that miR-21 is indeed regulated by PDGF signaling. CONCLUSIONS: Our data show that miR-21 and SOX2 are tightly regulated already during embryogenesis and define a distinct population with putative tumor cell of origin characteristics. Furthermore, we believe that miR-21 is a mediator of PDGF-driven brain tumors, which suggests miR-21 as a promising target for treatment of glioma.


Subject(s)
Brain/metabolism , Gene Expression Regulation, Developmental , Glioma/genetics , MicroRNAs/genetics , SOXB1 Transcription Factors/metabolism , Animals , Animals, Newborn , Apoptosis/genetics , Becaplermin , Blotting, Northern , Blotting, Western , Brain/embryology , Brain/growth & development , Cell Line, Transformed , Cell Line, Tumor , Chickens , Glioma/metabolism , Glioma/pathology , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/metabolism , RNA Interference , Signal Transduction/genetics , Transplantation, Heterologous
17.
BMC Mol Biol ; 12: 28, 2011 Jul 07.
Article in English | MEDLINE | ID: mdl-21733196

ABSTRACT

BACKGROUND: CGGBP1 is a CGG-triplet repeat binding protein, which affects transcription from CGG-triplet-rich promoters such as the FMR1 gene and the ribosomal RNA gene clusters. Earlier, we reported some previously unknown functions of CGGBP1 in gene expression during heat shock stress response. Recently we had found CGGBP1 to be a cell cycle regulatory midbody protein required for normal cytokinetic abscission in normal human fibroblasts, which have all the cell cycle regulatory mechanisms intact. RESULTS: In this study we explored the role of CGGBP1 in the cell cycle in various cancer cell lines. CGGBP1 depletion by RNA interference in tumor-derived cells caused an increase in the cell population at G0/G1 phase and reduced the number of cells in the S phase. CGGBP1 depletion also increased the expression of cell cycle regulatory genes CDKN1A and GAS1, associated with reductions in histone H3 lysine 9 trimethylation in their promoters. By combining RNA interference and genetic mutations, we found that the role of CGGBP1 in cell cycle involves multiple mechanisms, as single deficiencies of CDKN1A, GAS1 as well as TP53, INK4A or ARF failed to rescue the G0/G1 arrest caused by CGGBP1 depletion. CONCLUSIONS: Our results show that CGGBP1 expression is important for cell cycle progression through multiple parallel mechanisms including the regulation of CDKN1A and GAS1 levels.


Subject(s)
Cell Cycle , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Neoplasms/pathology , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA-Binding Proteins/genetics , G1 Phase , GPI-Linked Proteins/genetics , Humans , Mice , Neoplasms/genetics , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/genetics , Resting Phase, Cell Cycle , S Phase , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
18.
Exp Cell Res ; 317(2): 143-50, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20832400

ABSTRACT

Abscission marks the completion of cell division and its failure is associated with delayed cytokinesis and even tetraploidization. Aberrant abscission and consequential ploidy changes can underlie various diseases including cancer. Midbody, a transient structure formed in the intercellular bridge during telophase, contains several proteins including Aurora kinase B (AURKB), which participate in abscission. We report here an unexpected expression pattern and function of the transcription repressor protein CGG triplet repeat-binding protein 1 (CGGBP1), in normal human fibroblasts. We show that CGGBP1, a chromatin-associated protein, trans-localizes to spindle midzone and midbodies in a manner similar to that of AURKB. CGGBP1 depletion resulted in a cell cycle block at G2, characterized by failure of cells to undergo mitosis and also reduced entry into S phase. Consistent with its presence in the midbodies, live microscopy showed that CGGBP1 deficiency caused mitotic failure at abscission resulting in tetraploidy, which could be rescued by CGGBP1 overexpression. These results show that CGGBP1 is a bona fide midbody protein required for normal abscission and mitosis in general.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Cytokinesis/physiology , DNA-Binding Proteins/metabolism , Organelles/metabolism , Aurora Kinase B , Aurora Kinases , Cell Cycle/physiology , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Division/physiology , Cell Nucleus/chemistry , Cells, Cultured , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Male , Mitosis/physiology , Organelles/chemistry , Ploidies , Protein Serine-Threonine Kinases/physiology , S Phase/physiology , Skin/cytology , Spindle Apparatus/metabolism , Telophase/physiology , Tetraploidy
19.
PLoS One ; 4(4): e5050, 2009.
Article in English | MEDLINE | ID: mdl-19337383

ABSTRACT

BACKGROUND: Though the Nuclear factor 1 family member NFIX has been strongly implicated in PDGFB-induced glioblastoma, its molecular mechanisms of action remain unknown. HSF1, a heat shock-related transcription factor is also a powerful modifier of carcinogenesis by several factors, including PDGFB. How HSF1 transcription is controlled has remained largely elusive. METHODOLOGY/PRINCIPAL FINDINGS: By combining microarray expression profiling and a yeast-two-hybrid screen, we identified that NFIX and its interactions with CGGBP1 and HMGN1 regulate expression of HSF1. We found that CGGBP1 organizes a bifunctional transcriptional complex at small CGG repeats in the HSF1 promoter. Under chronic heat shock, NFIX uses CGGBP1 and HMGN1 to get recruited to this promoter and in turn affects their binding to DNA. Results show that the interactions of NFIX with CGGBP1 and HMGN1 in the soluble fraction are heat shock sensitive due to preferential localization of CGGBP1 to heterochromatin after heat shock. HSF1 in turn was found to bind to the NFIX promoter and repress its expression in a heat shock sensitive manner. CONCLUSIONS/SIGNIFICANCE: NFIX and HSF1 exert a mutual transcriptional repressive effect on each other which requires CGG repeat in HSF1 promoter and HSF1 binding site in NFIX promoter. We unravel a unique mechanism of heat shock sensitive DNA sequence-directed reciprocal transcriptional regulation between NFIX and HSF1. Our findings provide new insights into mechanisms of transcription regulation under stress.


Subject(s)
DNA-Binding Proteins/genetics , DNA/genetics , Gene Expression Regulation , Hot Temperature , NFI Transcription Factors/genetics , Transcription Factors/genetics , Transcription, Genetic , DNA-Binding Proteins/metabolism , Heat Shock Transcription Factors , NFI Transcription Factors/metabolism , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering , Transcription Factors/metabolism
20.
Arch Physiol Biochem ; 114(5): 301-30, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19085233

ABSTRACT

UNLABELLED: Interspecific hybridization in mammals causes hybrid dysgenesis effects, such as sterility and abnormal placentation. Here, we describe a novel obesity syndrome caused by interspecific hybridization in the genus Mus and show that this obesity, appearing sporadically in F1 littermates derived from inbred strains, has an epigenetic basis. Mus hybrids from various strains of M. musculus and M. spretus were generated and the sporadic obese phenotype was confirmed through assessment of physiological and biochemical parameters in littermates. To understand the underlying mechanisms, large-scale and candidate gene expression assays, global DNA methylation assays and allelic expression analysis were performed. Studies showed that obese hybrids are similar to other known models of obesity. While increased axial growth indicated a defect in POMC pathway, comparison of global gene expression patterns in brain of obese F1 and obese Pomc mutant mice showed little similarity. In F1 obese mice many genes involved in the maintenance of epigenetic states, as well as several imprinted genes, were differentially expressed. Global DNA methylation analysis in brain showed that increased methylation levels were associated with obesity. The imprinted gene Gnasxl, known to be important in lipid homeostasis, was found over expressed in the obese hybrids. Allelic expression and methylation analysis of Gnasxl showed that alterations of epigenetic marks underlying F1 obesity are probably many and multi-factorial. CONCLUSIONS: This model of obesity, which is both spontaneous and epigenetic, may be a useful tool to address the epigenetic aspects of clinical obesity.


Subject(s)
Hybridization, Genetic/genetics , Obesity/genetics , Animals , DNA Methylation/genetics , Epigenesis, Genetic , Female , Gene Expression Profiling , Homeostasis/genetics , Lipid Metabolism/genetics , Male , Mice , Mice, Obese , Obesity/metabolism , Obesity/pathology , Obesity/physiopathology , Oligonucleotide Array Sequence Analysis , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...