Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
2.
Nat Neurosci ; 26(11): 1868-1879, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37798411

ABSTRACT

The amygdala processes positive and negative valence and contributes to addiction, but the cell-type-specific gene regulatory programs involved are unknown. We generated an atlas of single-nucleus gene expression and chromatin accessibility in the amygdala of outbred rats with high and low cocaine addiction-like behaviors following prolonged abstinence. Differentially expressed genes between the high and low groups were enriched for energy metabolism across cell types. Rats with high addiction index (AI) showed increased relapse-like behaviors and GABAergic transmission in the amygdala. Both phenotypes were reversed by pharmacological inhibition of the glyoxalase 1 enzyme, which metabolizes methylglyoxal-a GABAA receptor agonist produced by glycolysis. Differences in chromatin accessibility between high and low AI rats implicated pioneer transcription factors in the basic helix-loop-helix, FOX, SOX and activator protein 1 families. We observed opposite regulation of chromatin accessibility across many cell types. Most notably, excitatory neurons had greater accessibility in high AI rats and inhibitory neurons had greater accessibility in low AI rats.


Subject(s)
Cocaine-Related Disorders , Cocaine , Humans , Rats , Animals , Amygdala/physiology , Neurons , Chromatin/metabolism , Cocaine/pharmacology
4.
Neuropsychopharmacology ; 47(12): 2071-2080, 2022 11.
Article in English | MEDLINE | ID: mdl-35995972

ABSTRACT

During adolescence, frequent and heavy cannabis use can lead to serious adverse health effects and cannabis use disorder (CUD). Rodent models of adolescent exposure to the main psychoactive component of cannabis, delta-9-tetrahydrocannabinol (THC), mimic the behavioral alterations observed in adolescent users. However, the underlying molecular mechanisms remain largely unknown. Here, we treated female and male C57BL6/N mice with high doses of THC during early adolescence and assessed their memory and social behaviors in late adolescence. We then profiled the transcriptome of five brain regions involved in cognitive and addiction-related processes. We applied gene coexpression network analysis and identified gene coexpression modules, termed cognitive modules, that simultaneously correlated with THC treatment and memory traits reduced by THC. The cognitive modules were related to endocannabinoid signaling in the female dorsal medial striatum, inflammation in the female ventral tegmental area, and synaptic transmission in the male nucleus accumbens. Moreover, cross-brain region module-module interaction networks uncovered intra- and inter-region molecular circuitries influenced by THC. Lastly, we identified key driver genes of gene networks associated with THC in mice and genetic susceptibility to CUD in humans. This analysis revealed a common regulatory mechanism linked to CUD vulnerability in the nucleus accumbens of females and males, which shared four key drivers (Hapln4, Kcnc1, Elavl2, Zcchc12). These genes regulate transcriptional subnetworks implicated in addiction processes, synaptic transmission, brain development, and lipid metabolism. Our study provides novel insights into disease mechanisms regulated by adolescent exposure to THC in a sex- and brain region-specific manner.


Subject(s)
Cannabis , Gene Expression , Hallucinogens , Sex Factors , Adolescent , Animals , Brain , Cannabinoid Receptor Agonists/pharmacology , Cannabis/adverse effects , Dronabinol/metabolism , Endocannabinoids/metabolism , Female , Gene Regulatory Networks , Hallucinogens/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Shaw Potassium Channels/metabolism
5.
Front Neurosci ; 16: 858427, 2022.
Article in English | MEDLINE | ID: mdl-35651629

ABSTRACT

Substance abuse and addiction represent a significant public health problem that impacts multiple dimensions of society, including healthcare, the economy, and the workforce. In 2021, over 100,000 drug overdose deaths were reported in the US, with an alarming increase in fatalities related to opioids and psychostimulants. Understanding the fundamental gene regulatory mechanisms underlying addiction and related behaviors could facilitate more effective treatments. To explore how repeated drug exposure alters gene regulatory networks in the brain, we combined capped small (cs)RNA-seq, which accurately captures nascent-like initiating transcripts from total RNA, with Hi-C and single nuclei (sn)ATAC-seq. We profiled initiating transcripts in two addiction-related brain regions, the prefrontal cortex (PFC) and the nucleus accumbens (NAc), from rats that were never exposed to drugs or were subjected to prolonged abstinence after oxycodone or cocaine intravenous self-administration (IVSA). Interrogating over 100,000 active transcription start regions (TSRs) revealed that most TSRs had hallmarks of bonafide enhancers and highlighted the KLF/SP1, RFX, and AP1 transcription factors families as central to establishing brain-specific gene regulatory programs. Analysis of rats with addiction-like behaviors versus controls identified addiction-associated repression of transcription at regulatory enhancers recognized by nuclear receptor subfamily 3 group C (NR3C) factors, including glucocorticoid receptors. Cell-type deconvolution analysis using snATAC-seq uncovered a potential role of glial cells in driving the gene regulatory programs associated with addiction-related phenotypes. These findings highlight the power of advanced transcriptomics methods to provide insight into how addiction perturbs gene regulatory programs in the brain.

6.
Cancers (Basel) ; 13(12)2021 Jun 20.
Article in English | MEDLINE | ID: mdl-34202984

ABSTRACT

Breast cancer is the most frequently occurred cancer type and the second cause of death in women worldwide. Alternative splicing (AS) is the process that generates more than one mRNA isoform from a single gene, and it plays a major role in expanding the human protein diversity. Aberrant AS contributes to breast cancer metastasis and resistance to chemotherapeutic interventions. Therefore, identifying cancer-specific isoforms is the prerequisite for therapeutic interventions intended to correct aberrantly expressed AS events. Here, we performed RNA-mediated oligonucleotide annealing, selection, and ligation coupled with next-generation sequencing (RASL-seq) in breast cancer cells, to identify global breast cancer-specific AS defects. By RT-PCR validation, we demonstrate the high accuracy of RASL-seq results. In addition, we analyzed identified AS events using the Cancer Genome Atlas (TCGA) database in a large number of non-pathological and breast tumor specimens and validated them in normal and breast cancer samples. Interestingly, aberrantly regulated AS cassette exons in cancer tissues do not encode for known functional domains but instead encode for amino acids constituting regions of intrinsically disordered protein portions characterized by high flexibility and prone to be subjected to post-translational modifications. Collectively, our results reveal novel AS errors occurring in human breast cancer, potentially affecting breast cancer-related biological processes.

7.
Cells ; 10(4)2021 04 09.
Article in English | MEDLINE | ID: mdl-33918758

ABSTRACT

Aberrant alternative splicing (AS) is a hallmark of cancer and a potential target for novel anti-cancer therapeutics. Breast cancer-associated AS events are known to be linked to disease progression, metastasis, and survival of breast cancer patients. To identify altered AS programs occurring in metastatic breast cancer, we perform a global analysis of AS events by using RNA-mediated oligonucleotide annealing, selection, and ligation coupled with next-generation sequencing (RASL-seq). We demonstrate that, relative to low-metastatic, high-metastatic breast cancer cells show different AS choices in genes related to cancer progression. Supporting a global reshape of cancer-related splicing profiles in metastatic breast cancer we found an enrichment of RNA-binding motifs recognized by several splicing regulators, which have aberrant expression levels or activity during breast cancer progression, including SRSF1. Among SRSF1-regulated targets we found DCUN1D5, a gene for which skipping of exon 4 in its pre-mRNA introduces a premature termination codon (PTC), thus generating an unstable transcript degraded by nonsense-mediated mRNA decay (NMD). Significantly, distinct breast cancer subtypes show different DCUN1D5 isoform ratios with metastatic breast cancer expressing the highest level of the NMD-insensitive DCUN1D5 mRNA, thus showing high DCUN1D5 expression levels, which are ultimately associated with poor overall and relapse-free survival in breast cancer patients. Collectively, our results reveal global AS features of metastatic breast tumors, which open new possibilities for the treatment of these aggressive tumor types.


Subject(s)
Alternative Splicing/genetics , Breast Neoplasms/genetics , Breast Neoplasms/secondary , Base Sequence , Cell Line, Tumor , Exons/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Nonsense Mediated mRNA Decay/genetics , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Peptide Synthases/genetics , Peptide Synthases/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , Survival Analysis
9.
Nature ; 583(7818): 711-719, 2020 07.
Article in English | MEDLINE | ID: mdl-32728246

ABSTRACT

Many proteins regulate the expression of genes by binding to specific regions encoded in the genome1. Here we introduce a new data set of RNA elements in the human genome that are recognized by RNA-binding proteins (RBPs), generated as part of the Encyclopedia of DNA Elements (ENCODE) project phase III. This class of regulatory elements functions only when transcribed into RNA, as they serve as the binding sites for RBPs that control post-transcriptional processes such as splicing, cleavage and polyadenylation, and the editing, localization, stability and translation of mRNAs. We describe the mapping and characterization of RNA elements recognized by a large collection of human RBPs in K562 and HepG2 cells. Integrative analyses using five assays identify RBP binding sites on RNA and chromatin in vivo, the in vitro binding preferences of RBPs, the function of RBP binding sites and the subcellular localization of RBPs, producing 1,223 replicated data sets for 356 RBPs. We describe the spectrum of RBP binding throughout the transcriptome and the connections between these interactions and various aspects of RNA biology, including RNA stability, splicing regulation and RNA localization. These data expand the catalogue of functional elements encoded in the human genome by the addition of a large set of elements that function at the RNA level by interacting with RBPs.


Subject(s)
RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Transcriptome/genetics , Alternative Splicing/genetics , Base Sequence , Binding Sites , Cell Line , Chromatin/genetics , Chromatin/metabolism , Databases, Genetic , Female , Gene Knockdown Techniques , Humans , Intracellular Space/genetics , Male , Protein Binding , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Substrate Specificity
10.
Cell ; 180(6): 1212-1227.e14, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32169215

ABSTRACT

The paternal genome undergoes a massive exchange of histone with protamine for compaction into sperm during spermiogenesis. Upon fertilization, this process is potently reversed, which is essential for parental genome reprogramming and subsequent activation; however, it remains poorly understood how this fundamental process is initiated and regulated. Here, we report that the previously characterized splicing kinase SRPK1 initiates this life-beginning event by catalyzing site-specific phosphorylation of protamine, thereby triggering protamine-to-histone exchange in the fertilized oocyte. Interestingly, protamine undergoes a DNA-dependent phase transition to gel-like condensates and SRPK1-mediated phosphorylation likely helps open up such structures to enhance protamine dismissal by nucleoplasmin (NPM2) and enable the recruitment of HIRA for H3.3 deposition. Remarkably, genome-wide assay for transposase-accessible chromatin sequencing (ATAC-seq) analysis reveals that selective chromatin accessibility in both sperm and MII oocytes is largely erased in early pronuclei in a protamine phosphorylation-dependent manner, suggesting that SRPK1-catalyzed phosphorylation initiates a highly synchronized reorganization program in both parental genomes.


Subject(s)
Chromatin/metabolism , Protamines/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Chromatin/physiology , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , Fertilization/genetics , Histones/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Oocytes/metabolism , Oocytes/physiology , Phosphorylation , Protamine Kinase/genetics , Protamine Kinase/metabolism , Protamines/genetics , Protein Serine-Threonine Kinases/physiology , RNA Splicing/genetics , RNA Splicing/physiology , Spermatozoa/metabolism , Transcription Factors/metabolism , Zygote/metabolism
11.
Proc Natl Acad Sci U S A ; 116(52): 26873-26880, 2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31843886

ABSTRACT

Primary liver cancer develops from multifactorial etiologies, resulting in extensive genomic heterogeneity. To probe the common mechanism of hepatocarcinogenesis, we interrogated temporal gene expression profiles in a group of mouse models with hepatic steatosis, fibrosis, inflammation, and, consequently, tumorigenesis. Instead of anticipated progressive changes, we observed a sudden molecular switch at a critical precancer stage, by developing analytical platform that focuses on transcription factor (TF) clusters. Coarse-grained network modeling demonstrated that an abrupt transcriptomic transition occurred once changes were accumulated to reach a threshold. Based on the experimental and bioinformatic data analyses as well as mathematical modeling, we derived a tumorigenic index (TI) to quantify tumorigenic signal strengths. The TI is powerful in predicting the disease status of patients with metabolic disorders and also the tumor stages and prognosis of liver cancer patients with diverse backgrounds. This work establishes a quantitative tool for triage of liver cancer patients and also for cancer risk assessment of chronic liver disease patients.

12.
Cell ; 178(1): 107-121.e18, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31251911

ABSTRACT

Increasing evidence suggests that transcriptional control and chromatin activities at large involve regulatory RNAs, which likely enlist specific RNA-binding proteins (RBPs). Although multiple RBPs have been implicated in transcription control, it has remained unclear how extensively RBPs directly act on chromatin. We embarked on a large-scale RBP ChIP-seq analysis, revealing widespread RBP presence in active chromatin regions in the human genome. Like transcription factors (TFs), RBPs also show strong preference for hotspots in the genome, particularly gene promoters, where their association is frequently linked to transcriptional output. Unsupervised clustering reveals extensive co-association between TFs and RBPs, as exemplified by YY1, a known RNA-dependent TF, and RBM25, an RBP involved in splicing regulation. Remarkably, RBM25 depletion attenuates all YY1-dependent activities, including chromatin binding, DNA looping, and transcription. We propose that various RBPs may enhance network interaction through harnessing regulatory RNAs to control transcription.


Subject(s)
Chromatin/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Transcription, Genetic/genetics , YY1 Transcription Factor/metabolism , Binding Sites , Gene Expression Regulation , Genome, Human/genetics , Hep G2 Cells , Humans , K562 Cells , Nuclear Proteins , Promoter Regions, Genetic/genetics , Protein Binding , RNA-Binding Proteins/genetics , RNA-Seq , Transcriptome , YY1 Transcription Factor/genetics
13.
Stem Cell Res Ther ; 10(1): 28, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30646941

ABSTRACT

BACKGROUND: Self-renewal is dependent on an intrinsic gene regulatory network centered on OCT4 and on an atypical cell cycle G1/S transition, which is also regulated by OCT4. p21, a gene negatively associated with self-renewal and a senescence marker, is a member of the universal cyclin-dependent kinase inhibitors (CDKIs) and plays critical roles in the regulation of the G1/S transition. The expression of p21 can be regulated by OCT4-targeted DNA methyltransferases (DNMTs), which play distinct roles in gene regulation and maintaining pluripotency properties. The aim of this study was to determine the role of OCT4 in the regulation of self-renewal and senescence in human hair follicle mesenchymal stem cells (hHFMSCs) and to characterize the molecular mechanisms involved. METHODS: A lentiviral vector was used to ectopically express OCT4. The influences of OCT4 on the self-renewal and senescence of hHFMSCs were investigated. Next-generation sequencing (NGS) was performed to identify the downstream genes of OCT4 in this process. Methylation-specific PCR (MSP) analysis was performed to measure the methylation level of the p21 promoter region. p21 was overexpressed in hHFMSCsOCT4 to test its downstream effect on OCT4. The regulatory effect of OCT4 on DNMTs was examined by ChIP assay. 5-aza-dC/zebularine was used to inhibit the expression of DNMTs, and then self-renewal properties and senescence in hHFMSCs were detected. RESULTS: The overexpression of OCT4 promoted proliferation, cell cycle progression, and osteogenic differentiation capacity of hHFMSCs. The cell senescence of hHFMSCs was markedly suppressed due to the ectopic expression of OCT4. Through NGS, we identified 2466 differentially expressed genes (DEGs) between hHFMSCsOCT4 and hHFMSCsEGFP, including p21, which was downregulated. The overexpression of p21 abrogated the proliferation and osteogenic differentiation capacity of hHFMSCsOCT4 and promoted cell senescence. OCT4 enhanced the transcription of DNMT genes, leading to an elevation in the methylation of the p21 promoter. The inhibition of DNMTs reversed the OCT4-induced p21 reduction, depleted the self-renewal of hHFMSCsOCT4, and triggered cell senescence. CONCLUSIONS: OCT4 maintains the self-renewal ability of hHFMSCs and reverses senescence by suppressing the expression of p21 through the upregulation of DNMTs.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Modification Methylases/metabolism , Hair Follicle/cytology , Mesenchymal Stem Cells/cytology , Octamer Transcription Factor-3/metabolism , Cell Line , Cellular Senescence/physiology , Down-Regulation , HEK293 Cells , Hair Follicle/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Octamer Transcription Factor-3/biosynthesis , Octamer Transcription Factor-3/genetics
15.
Mol Med Rep ; 17(5): 6211-6226, 2018 05.
Article in English | MEDLINE | ID: mdl-29512753

ABSTRACT

Multidrug resistance (MDR) is a major problem in the treatment of breast cancer. In the present study, next-generation sequencing technology was employed to identify differentially expressed genes in MCF­7/MDR cells and MCF­7 cells, and aimed to investigate the underlying molecular mechanisms of MDR in breast cancer. Differentially expressed genes between MCF­7/MDR and MCF­7 cells were selected using software; a total of 2085 genes were screened as differentially expressed in MCF­7/MDR cells. Furthermore, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed using the DAVID database. Finally, a protein­protein interaction network was constructed and the hub genes in the network were analyzed using the STRING database. GO annotation demonstrated that the differentially expressed genes were enriched in various biological processes, including 'regulation of cell differentiation', 'cell development', 'neuron development', 'movement of cell or subcellular component' and 'cell morphogenesis involved in neuron differentiation'. Cellular component analysis by GO revealed that differentially expressed genes were enriched in 'plasma membrane region' and 'extracellular matrix' terms. Furthermore, KEGG analysis demonstrated that the target genes were enriched in various pathways, including 'cell adhesion molecules (CAMs)', 'calcium signaling pathway', 'tight junction', 'Wnt signaling pathway' and 'pathways in cancer' terms. A protein­protein interaction network demonstrated that certain hub genes, including cyclin D1, nitric oxide synthase 3 (NOS3), NOTCH3, brain­derived neurotrophic factor (BDNF), paired box 6, neuropeptide Y, phospholipase C ß (PLCB) 4, PLCB2 and actin α cardiac muscle 1, may be associated with MDR in breast cancer. Subsequently, RT­qPCR confirmed that the expression of these 9 hub genes was higher in MCF­7/MDR cells compared with MCF­7 cells, consistent with the RNA­sequencing analysis. Additionally, a Cell Counting Kit­8 assay demonstrated that specific inhibitors of NOS3 and BDNF/neurotrophic receptor tyrosine kinase, type 2 signaling reduced the IC50 of MCF­7/MDR cells in response to various anticancer drugs, including adriamycin, cisplatin and 5­fluorouracil. The results of the present study provide novel insights into the mechanism underlying MDR in MCF­7 cells and may identify novel targets for the treatment of breast cancer.


Subject(s)
Breast Neoplasms , Genes, Neoplasm , Neoplasm Proteins , RNA, Neoplasm , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , High-Throughput Nucleotide Sequencing , Humans , MCF-7 Cells , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
16.
Mol Cell ; 69(3): 412-425.e6, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29395063

ABSTRACT

Mutations in several general pre-mRNA splicing factors have been linked to myelodysplastic syndromes (MDSs) and solid tumors. These mutations have generally been assumed to cause disease by the resultant splicing defects, but different mutations appear to induce distinct splicing defects, raising the possibility that an alternative common mechanism is involved. Here we report a chain of events triggered by multiple splicing factor mutations, especially high-risk alleles in SRSF2 and U2AF1, including elevated R-loops, replication stress, and activation of the ataxia telangiectasia and Rad3-related protein (ATR)-Chk1 pathway. We further demonstrate that enhanced R-loops, opposite to the expectation from gained RNA binding with mutant SRSF2, result from impaired transcription pause release because the mutant protein loses its ability to extract the RNA polymerase II (Pol II) C-terminal domain (CTD) kinase-the positive transcription elongation factor complex (P-TEFb)-from the 7SK complex. Enhanced R-loops are linked to compromised proliferation of bone-marrow-derived blood progenitors, which can be partially rescued by RNase H overexpression, suggesting a direct contribution of augmented R-loops to the MDS phenotype.


Subject(s)
Base Sequence/genetics , Myelodysplastic Syndromes/genetics , RNA Splicing Factors/genetics , Cell Cycle Checkpoints/genetics , HEK293 Cells , Humans , Mutation , Nuclear Proteins/genetics , Phosphoproteins/genetics , RNA Splicing/genetics , RNA Splicing Factors/metabolism , Ribonucleoproteins/genetics , Serine-Arginine Splicing Factors/genetics , Splicing Factor U2AF/genetics
17.
Hepatology ; 67(5): 1807-1822, 2018 05.
Article in English | MEDLINE | ID: mdl-29152756

ABSTRACT

Both activating and inactivating mutations in catenin ß1 (ctnnb1), which encodes ß-catenin, have been implicated in liver tumorigenesis in humans and mice, although the underlying mechanisms are not fully understood. Herein, we show that deletion of endogenous ß-catenin in hepatocytes aggravated hepatocellular carcinoma (HCC) development driven by an oncogenic version of ß-catenin (CAT) in combination with the hepatocyte growth factor receptor MET proto-oncogene receptor tyrosine kinase (MET). Although the mitogenic signaling and cell cycle progression was modestly impaired after CAT/MET transfection, the ß-catenin-deficient livers displayed changes in transcriptomes, increased DNA damage response, expanded Sox9+ cells, and up-regulation of protumorigenic cytokines, including interleukin-6 and transforming growth factor ß1. These events eventually exacerbated CAT/MET-driven hepatocarcinogenesis in ß-catenin-deficient livers, featured by up-regulation of extracellular signal-regulated kinase (Erk), protein kinase B (Akt), and Wnt/ß-catenin signaling and cyclin D1 expression. The resultant mouse tumors showed similar transcriptomes to human HCC samples with concomitant CTNNB1 mutations and MET overexpression. CONCLUSION: These data argue that while dominantly activating mutants of ß-catenin are oncogenic, inhibiting the oncogenic signaling pathway generates a pro-oncogenic microenvironment that may facilitate HCC recurrence following a targeted therapy of the primary tumor. An effective therapeutic strategy must require disruption of the oncogenic signaling in tumor cells and suppression of the secondary tumor-promoting stromal effects in the liver microenvironment. (Hepatology 2018;67:1807-1822).


Subject(s)
Carcinoma, Hepatocellular/genetics , Hepatocytes/metabolism , Liver Neoplasms/genetics , Proto-Oncogene Proteins c-met/genetics , beta Catenin/genetics , Animals , Carcinogenesis/genetics , Carcinoma, Hepatocellular/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver/pathology , Liver Neoplasms/metabolism , Mice , Oncogenes , Proto-Oncogene Mas , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , beta Catenin/metabolism
18.
Mol Cell ; 68(4): 745-757.e5, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29104020

ABSTRACT

R-loop, a three-stranded RNA/DNA structure, has been linked to induced genome instability and regulated gene expression. To enable precision analysis of R-loops in vivo, we develop an RNase-H-based approach; this reveals predominant R-loop formation near gene promoters with strong G/C skew and propensity to form G-quadruplex in non-template DNA, corroborating with all biochemically established properties of R-loops. Transcription perturbation experiments further indicate that R-loop induction correlates to transcriptional pausing. Interestingly, we note that most mapped R-loops are each linked to a nearby free RNA end; by using a ribozyme to co-transcriptionally cleave nascent RNA, we demonstrate that such a free RNA end coupled with a G/C-skewed sequence is necessary and sufficient to induce R-loop. These findings provide a topological solution for RNA invasion into duplex DNA and suggest an order for R-loop initiation and elongation in an opposite direction to that previously proposed.


Subject(s)
DNA/chemistry , Nucleic Acid Heteroduplexes/chemistry , Promoter Regions, Genetic/physiology , RNA/chemistry , Ribonuclease H/chemistry , Transcription, Genetic , DNA/biosynthesis , HEK293 Cells , Humans , K562 Cells , Nucleic Acid Heteroduplexes/metabolism , RNA/biosynthesis
19.
Int J Mol Sci ; 18(9)2017 Aug 30.
Article in English | MEDLINE | ID: mdl-28867761

ABSTRACT

The downregulation of tight junction protein CLDN6 promotes breast cancer cell migration and invasion; however, the exact mechanism underlying CLDN6 downregulation remains unclear. CLDN6 silence is associated with DNA methyltransferase 1 (DNMT1) mediated DNA methylation, and DNMT1 is regulated by the transforming growth factor beta (TGFß)/SMAD pathway. Therefore, we hypothesized that TGFß/SMAD pathway, specifically SMAD2, may play a critical role for CLDN6 downregulation through DNA methyltransferase 1 (DNMT1) mediated DNA methylation. To test this hypothesis, we blocked the SMAD2 pathway with SB431542 in two human breast cancer cell lines (MCF-7 and SKBR-3). Our results showed that treatment with SB431542 led to a decrease of DNMT1 expression and the binding activity for CLDN6 promoter. The methylation level of CLDN6 promoter was decreased, and simultaneously CLDN6 protein expression increased. Upregulation of CLDN6 inhibited epithelial to mesenchymal transition (EMT) and reduced the migration and invasion ability of both MCF-7 and SKBR-3 cells. Furthermore, knocked down of CLDN6 abolished SB431542 effects on suppression of EMT associated gene expression and inhibition of migration and invasion. Thus, we demonstrated that the downregulation of CLDN6 is regulated through promoter methylation by DNMT1, which depends on the SMAD2 pathway, and that CLDN6 is a key regulator in the SMAD2/DNMT1/CLDN6 pathway to inhibit EMT, migration and invasion of breast cancer cells.


Subject(s)
Breast Neoplasms/genetics , Claudins/genetics , Smad2 Protein/genetics , Breast Neoplasms/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Claudins/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA Methylation/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , MCF-7 Cells , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Signal Transduction/genetics
20.
Nat Biotechnol ; 35(10): 940-950, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28922346

ABSTRACT

Higher eukaryotic genomes are bound by a large number of coding and non-coding RNAs, but approaches to comprehensively map the identity and binding sites of these RNAs are lacking. Here we report a method to capture in situ global RNA interactions with DNA by deep sequencing (GRID-seq), which enables the comprehensive identification of the entire repertoire of chromatin-interacting RNAs and their respective binding sites. In human, mouse, and Drosophila cells, we detected a large set of tissue-specific coding and non-coding RNAs that are bound to active promoters and enhancers, especially super-enhancers. Assuming that most mRNA-chromatin interactions indicate the physical proximity of a promoter and an enhancer, we constructed a three-dimensional global connectivity map of promoters and enhancers, revealing transcription-activity-linked genomic interactions in the nucleus.


Subject(s)
Chromatin/metabolism , DNA/isolation & purification , High-Throughput Nucleotide Sequencing/methods , RNA/metabolism , Animals , Cell Line, Tumor , Drosophila , Enhancer Elements, Genetic , Humans , Promoter Regions, Genetic , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...