scispace - formally typeset
Search or ask a question

Showing papers on "Gene expression published in 2015"


Journal ArticleDOI
21 May 2015-Cell
TL;DR: This work has developed a high-throughput droplet-microfluidic approach for barcoding the RNA from thousands of individual cells for subsequent analysis by next-generation sequencing, which shows a surprisingly low noise profile and is readily adaptable to other sequencing-based assays.

2,894 citations


Journal ArticleDOI
29 Jan 2015-Nature
TL;DR: Structural-guided engineering of a CRISPR-Cas9 complex to mediate efficient transcriptional activation at endogenous genomic loci is described and the potential of Cas9-based activators as a powerful genetic perturbation technology is demonstrated.
Abstract: Systematic interrogation of gene function requires the ability to perturb gene expression in a robust and generalizable manner. Here we describe structure-guided engineering of a CRISPR-Cas9 complex to mediate efficient transcriptional activation at endogenous genomic loci. We used these engineered Cas9 activation complexes to investigate single-guide RNA (sgRNA) targeting rules for effective transcriptional activation, to demonstrate multiplexed activation of ten genes simultaneously, and to upregulate long intergenic non-coding RNA (lincRNA) transcripts. We also synthesized a library consisting of 70,290 guides targeting all human RefSeq coding isoforms to screen for genes that, upon activation, confer resistance to a BRAF inhibitor. The top hits included genes previously shown to be able to confer resistance, and novel candidates were validated using individual sgRNA and complementary DNA overexpression. A gene expression signature based on the top screening hits correlated with markers of BRAF inhibitor resistance in cell lines and patient-derived samples. These results collectively demonstrate the potential of Cas9-based activators as a powerful genetic perturbation technology.

2,186 citations


Journal ArticleDOI
04 Jun 2015-Cell
TL;DR: In a unified mechanism of m(6)A-based regulation in the cytoplasm, YTHDF2-mediated degradation controls the lifetime of target transcripts, whereasYTHDF1-mediated translation promotion increases translation efficiency, ensuring effective protein production from dynamic transcripts that are marked by m( 6)A.

2,179 citations


Journal ArticleDOI
24 Apr 2015-Science
TL;DR: This report reports multiplexed error-robust FISH (MERFISH), a single-molecule imaging method that allows thousands of RNA species to be imaged in single cells by using combinatorial FISH labeling with encoding schemes capable of detecting and/or correcting errors.
Abstract: INTRODUCTION: The copy number and in- tracellular localization of RNA are important regulators of gene expression. Measurement of these properties at the transcriptome scale in single cells will give answers to many ques- tions related to gene expression and regulation. Single-molecule RNA imaging approaches, such as single-molecule fluorescence in situ hybrid- ization(smFISH), are powerful toolsforcount- ing and mappingRNA; however, the number of RNA species that can be simultaneously im- aged in individual cells has been limited. This makes it challenging to perform transcriptomic analysis of single cells in a spatially resolved manner. Here, we report multiplexed error- robust FISH (MERFISH), a single-molecule im- aging method that allows thousands of RNA species to be imaged in single cells by using combinatorial FISH labeling with encoding schemes capable of detecting and/or correct- ing errors. RATIONALE: We labeled each cellular RNA with a set of encoding probes, which contain targeting sequences that bind the RNA and readout sequences that bind fluorescently la- beled readout probes. Each RNA species is encodedwithaparticular combinationofread- out sequences. We used successive rounds of hybridization and imaging, each with a differ- ent readout probe, to identify the readout se- quences bound to each RNA and to decode the RNA. In principle, combinatorial labeling al- lows the number of detectable RNA species to

1,576 citations


Journal ArticleDOI
26 Feb 2015-Nature
TL;DR: It is found that m6A alters the local structure in mRNA and long non-coding RNA (lncRNA) to facilitate binding of heterogeneous nuclear ribonucleoprotein C (HNRNPC), an abundant nuclear RNA-binding protein responsible for pre-mRNA processing.
Abstract: RNA-binding proteins control many aspects of cellular biology through binding single-stranded RNA binding motifs (RBMs). However, RBMs can be buried within their local RNA structures, thus inhibiting RNA-protein interactions. N(6)-methyladenosine (m(6)A), the most abundant and dynamic internal modification in eukaryotic messenger RNA, can be selectively recognized by the YTHDF2 protein to affect the stability of cytoplasmic mRNAs, but how m(6)A achieves its wide-ranging physiological role needs further exploration. Here we show in human cells that m(6)A controls the RNA-structure-dependent accessibility of RBMs to affect RNA-protein interactions for biological regulation; we term this mechanism 'the m(6)A-switch'. We found that m(6)A alters the local structure in mRNA and long non-coding RNA (lncRNA) to facilitate binding of heterogeneous nuclear ribonucleoprotein C (HNRNPC), an abundant nuclear RNA-binding protein responsible for pre-mRNA processing. Combining photoactivatable-ribonucleoside-enhanced crosslinking and immunoprecipitation (PAR-CLIP) and anti-m(6)A immunoprecipitation (MeRIP) approaches enabled us to identify 39,060 m(6)A-switches among HNRNPC-binding sites; and global m(6)A reduction decreased HNRNPC binding at 2,798 high-confidence m(6)A-switches. We determined that these m(6)A-switch-regulated HNRNPC-binding activities affect the abundance as well as alternative splicing of target mRNAs, demonstrating the regulatory role of m(6)A-switches on gene expression and RNA maturation. Our results illustrate how RNA-binding proteins gain regulated access to their RBMs through m(6)A-dependent RNA structural remodelling, and provide a new direction for investigating RNA-modification-coded cellular biology.

1,313 citations


Journal ArticleDOI
TL;DR: DIANA-TarBase v7.0 aims to provide for the first time hundreds of thousands of high-quality manually curated experimentally validated miRNA:gene interactions, enhanced with detailed meta-data, which enables users to easily identify positive or negative experimental results, the utilized experimental methodology, experimental conditions including cell/tissue type and treatment.
Abstract: microRNAs (miRNAs) are short non-coding RNA species, which act as potent gene expression regulators. Accurate identification of miRNA targets is crucial to understanding their function. Currently, hundreds of thousands of miRNA:gene interactions have been experimentally identified. However, this wealth of information is fragmented and hidden in thousands of manuscripts and raw next-generation sequencing data sets. DIANA-TarBase was initially released in 2006 and it was the first database aiming to catalog published experimentally validated miRNA:gene interactions. DIANA-TarBase v7.0 (http://www.microrna.gr/tarbase) aims to provide for the first time hundreds of thousands of high-quality manually curated experimentally validated miRNA:gene interactions, enhanced with detailed meta-data. DIANA-TarBase v7.0 enables users to easily identify positive or negative experimental results, the utilized experimental methodology, experimental conditions including cell/tissue type and treatment. The new interface provides also advanced information ranging from the binding site location, as identified experimentally as well as in silico, to the primer sequences used for cloning experiments. More than half a million miRNA:gene interactions have been curated from published experiments on 356 different cell types from 24 species, corresponding to 9- to 250-fold more entries than any other relevant database. DIANA-TarBase v7.0 is freely available.

659 citations


Journal ArticleDOI
TL;DR: The broad roles of m(6)A in regulating cell fates and embryonic development highlight the existence of another layer of epigenetic regulation at the RNA level, where mRNA is subjected to chemical modifications that affect protein expression.
Abstract: Both DNA and histone proteins undergo dynamic and reversible chemical modifications to control gene expression (Strahl and Allis 2000; Bird 2001; Suzuki and Bird 2008; Bhutani et al. 2011; Jones 2012; Kohli and Zhang 2013). Although post-transcriptional modifications are known to occur to RNAs, the impact of these modifications on gene expression regulation has only recently begun to be explored (He 2010). To date, more than a hundred structurally distinct chemical modifications have been found in eukaryotic RNAs (Cantara et al. 2011; Machnicka et al. 2013); however, the enzymes responsible for each modification and the biological consequences of these modified RNAs are largely unknown. RNA modifications were once considered to be static, but a flurry of recent discoveries has demonstrated that some chemical modifications can be dynamic and participate in the regulation of diverse physiological processes (Motorin and Helm 2011; Yi and Pan 2011; Chan et al. 2012; Fu et al. 2014; Meyer and Jaffrey 2014; Kirchner and Ignatova 2015). The presence of N6-methyladenosine (m6A) in polyadenylated mRNA was first discovered in the 1970s (Desrosiers et al. 1974; Perry and Kelley 1974; Lavi and Shatkin 1975; Wei et al. 1975; Schibler et al. 1977; Wei and Moss 1977) by researchers who were characterizing the 5′ cap structure of messenger RNA (mRNA) in mammalian cells. Since then, m6A has been identified as the most prevalent internal modification in mRNA and long noncoding RNA (lncRNA) in higher eukaryotes. It is widely conserved among eukaryotic species that range from yeast, plants, and flies to mammals as well as among viral mRNAs that replicate inside host nuclei (Krug et al. 1976; Beemon and Keith 1977; Horowitz et al. 1984; Bokar 2005). In addition to its occurrence in mRNA, m6A also exists in various classes of RNA in eukaryotes, bacteria, and archaea, including ribosomal RNAs, small nuclear RNAs, and transfer RNAs (Bjork et al. 1987; Maden 1990; Shimba et al. 1995; Gu et al. 1996; Agris et al. 2007; Piekna-Przybylska et al. 2008). Despite its widespread distribution in the mammalian transcriptome (on average, approximately three m6A sites per mRNA), functional insight has been lacking, possibly due to the low abundance of m6A mRNA and technical difficulties in global detection. Interest in the biological relevance of m6A in mRNA resurfaced after the discovery of two mammalian RNA demethylases, FTO (fat mass and obesity-associated protein) (Jia et al. 2011) and its homolog, ALKBH5 (Zheng et al. 2013), which selectively reverse m6A to adenosine in nuclear RNA. FTO is associated with human obesity (Dina et al. 2007; Frayling et al. 2007; Loos and Yeo 2014) and mental development (Hess et al. 2013), while ALKBH5 is shown to affect mouse spermatogenesis in a demethylation-dependent manner (Zheng et al. 2013), suggesting broad roles of m6A in various physiological processes. Shortly after these findings, YTHDF2 (YTH domain-containing family protein 2) was identified as the first m6A reader protein that preferentially recognizes m6A-containing mRNA (Dominissini et al. 2012; Wang et al. 2014a) and mediates mRNA decay (Wang et al. 2014a), thereby suggesting a role for m6A RNA as a negative regulator of gene expression. On the other hand, a transcriptome-wide m6A profiling method was developed to decipher the m6A RNA landscape (Dominissini et al. 2012; Meyer et al. 2012). Intriguingly, m6A sites in mammalian polyadenylated RNA are dominated by the conserved Pu[G > A]m6AC[A/C/U] motif that localizes near stop codons, in 3′ untranslated regions (UTRs), within long internal exons, and at 5′ UTRs (Dominissini et al. 2012; Meyer et al. 2012; Schwartz et al. 2013; Li et al. 2014; Luo et al. 2014), immediately raising the question of how this specificity is achieved. The m6A RNA landscape is initially sculptured by a methyltransferase complex, but for a long time, METTL3 (methyltransferase-like 3) was the only known SAM (S-adenosyl methionine)-binding subunit associated with mRNA methylation (Bokar et al. 1997). In 2014, a new mammalian methyltransferase, METTL14, was discovered to catalyze m6A methylation. Together with METTL3, these two proteins form a stable heterodimer complex that mediates cellular m6A deposition on mammalian mRNAs (Liu et al. 2014; Wang et al. 2014b). Recently, the mammalian splicing factor WTAP (Wilms’ tumor 1-associating protein) was identified as the third auxiliary factor of the core methyltransferase complex that affects cellular m6A methylation (Liu et al. 2014; Ping et al. 2014). The identification and characterization of the complete mammalian m6A methylation machinery are the first steps toward deciphering the selectivity and biological functions of m6A deposition in eukaryotic mRNAs. In this review, we mainly summarize recent progress in the study of m6A methylation in mRNA across different eukaryotes and discuss their newly discovered roles in post-transcriptional gene expression regulation. We first describe the features of m6A on a global scale and briefly introduce the mammalian m6A writers, erasers, and readers that specifically install, remove, or bind to m6A at defined sequence motifs (Fig. 1). We then discuss the evolutional conservation of the m6A methylation machinery across eukaryotic species that range from yeast, plants, and flies to mammals, highlighting the broad roles of methyltransferases and m6A in regulating cell status and embryonic development. Finally, we discuss the emerging functions of m6A in several mechanisms of post-transcriptional gene expression regulation with a special focus on the effects of m6A on differentiation and reprograming of stem cells. Figure 1. Illustration of the cellular pathways of m6A in nuclear RNAs. The m6A methyltransferases and demethylases dynamically control the m6A methylation landscape within the nucleus. The m6A reader proteins preferentially bind to the methylated RNA and mediate ...

659 citations


Journal ArticleDOI
26 Mar 2015-Nature
TL;DR: A novel biochemical approach, in vivo click selective 2′-hydroxyl acylation and profiling experiment (icSHAPE), is presented, which enables the first global view, to the authors' knowledge, of RNA secondary structures in living cells for all four bases.
Abstract: Visualizing the physical basis for molecular behaviour inside living cells is a great challenge for biology. RNAs are central to biological regulation, and the ability of RNA to adopt specific structures intimately controls every step of the gene expression program. However, our understanding of physiological RNA structures is limited; current in vivo RNA structure profiles include only two of the four nucleotides that make up RNA. Here we present a novel biochemical approach, in vivo click selective 2'-hydroxyl acylation and profiling experiment (icSHAPE), which enables the first global view, to our knowledge, of RNA secondary structures in living cells for all four bases. icSHAPE of the mouse embryonic stem cell transcriptome versus purified RNA folded in vitro shows that the structural dynamics of RNA in the cellular environment distinguish different classes of RNAs and regulatory elements. Structural signatures at translational start sites and ribosome pause sites are conserved from in vitro conditions, suggesting that these RNA elements are programmed by sequence. In contrast, focal structural rearrangements in vivo reveal precise interfaces of RNA with RNA-binding proteins or RNA-modification sites that are consistent with atomic-resolution structural data. Such dynamic structural footprints enable accurate prediction of RNA-protein interactions and N(6)-methyladenosine (m(6)A) modification genome wide. These results open the door for structural genomics of RNA in living cells and reveal key physiological structures controlling gene expression.

599 citations


Journal ArticleDOI
TL;DR: It is shown that MEG3 and EZH2 share common target genes, including the TGF-β pathway genes, and RNA–DNA triplex formation could be a general characteristic of target gene recognition by the chromatin-interacting lncRNAs.
Abstract: Long noncoding RNAs (lncRNAs) regulate gene expression by association with chromatin, but how they target chromatin remains poorly understood. We have used chromatin RNA immunoprecipitation-coupled high-throughput sequencing to identify 276 lncRNAs enriched in repressive chromatin from breast cancer cells. Using one of the chromatin-interacting lncRNAs, MEG3, we explore the mechanisms by which lncRNAs target chromatin. Here we show that MEG3 and EZH2 share common target genes, including the TGF-β pathway genes. Genome-wide mapping of MEG3 binding sites reveals that MEG3 modulates the activity of TGF-β genes by binding to distal regulatory elements. MEG3 binding sites have GA-rich sequences, which guide MEG3 to the chromatin through RNA–DNA triplex formation. We have found that RNA–DNA triplex structures are widespread and are present over the MEG3 binding sites associated with the TGF-β pathway genes. Our findings suggest that RNA–DNA triplex formation could be a general characteristic of target gene recognition by the chromatin-interacting lncRNAs.

526 citations


Journal ArticleDOI
04 Jun 2015-Nature
TL;DR: A genome-wide length-dependent increase in gene expression is identified in MeCP2 mutant mouse models and human RTT brains, and it is found that long genes as a population are enriched for neuronal functions and selectively expressed in the brain.
Abstract: Rett syndrome is caused by mutation of the MECP2 gene that codes for a protein that binds methylated DNA; this study reveals that MeCP2 affects the expression of long genes, which often serve neuronal functions Autism-related Rett syndrome is caused by disruption of the MECP2 gene, which codes for a methyl-DNA binding protein, but how MECP2 may control transcription of other genes has remained unclear Now Michael Greenberg and colleagues show that disruption of the Mecp2 gene in a mouse model and in human Rett syndrome leads to preferential upregulation of longer genes, and that these often serve neuronal functions Further data indicate that decreasing the expression of long genes, via hypomethylation of the dinucleotide CA, attenuates Rett-related dysfunctions in cultured neurons lacking MECP2 Disruption of the MECP2 gene leads to Rett syndrome (RTT), a severe neurological disorder with features of autism1 MECP2 encodes a methyl-DNA-binding protein2 that has been proposed to function as a transcriptional repressor, but despite numerous mouse studies examining neuronal gene expression in Mecp2 mutants, no clear model has emerged for how MeCP2 protein regulates transcription3,4,5,6,7,8,9 Here we identify a genome-wide length-dependent increase in gene expression in MeCP2 mutant mouse models and human RTT brains We present evidence that MeCP2 represses gene expression by binding to methylated CA sites within long genes, and that in neurons lacking MeCP2, decreasing the expression of long genes attenuates RTT-associated cellular deficits In addition, we find that long genes as a population are enriched for neuronal functions and selectively expressed in the brain These findings suggest that mutations in MeCP2 may cause neurological dysfunction by specifically disrupting long gene expression in the brain

511 citations


Journal ArticleDOI
TL;DR: Embryonic stem cell culture conditions are important for maintaining long-term self-renewal, and they influence cellular pluripotency state, with 2i being the most similar to blastocyst cells and including a subpopulation resembling the two-cell embryo state.

Journal ArticleDOI
18 Jun 2015-Cell
TL;DR: It is reported that neuronal activity stimulation triggers the formation of DNA double strand breaks (DSBs) in the promoters of a subset of early-response genes, including Fos, Npas4, and Egr1.

Journal ArticleDOI
TL;DR: It is shown that transcript abundance correlates with cellular volume at the single-cell level due to increased global transcription in larger cells, and a separate mechanism for gene dosage compensation after DNA replication that enables proper transcriptional output during early and late S phase is revealed.

Journal ArticleDOI
TL;DR: Microglia priming induces a highly conserved transcriptional signature with aging- and disease-specific aspects that prominently differed from the acute inflammatory gene network induced by lipopolysaccharide (LPS).
Abstract: Microglia are tissue macrophages of the central nervous system that monitor brain homeostasis and react upon neuronal damage and stress. Aging and neurodegeneration induce a hypersensitive, pro-inflammatory phenotype, referred to as primed microglia. To determine the gene expression signature of priming, the transcriptomes of microglia in aging, Alzheimer’s disease (AD), and amyotrophic lateral sclerosis (ALS) mouse models were compared using Weighted Gene Co-expression Network Analysis (WGCNA). A highly consistent consensus transcriptional profile of up-regulated genes was identified, which prominently differed from the acute inflammatory gene network induced by lipopolysaccharide (LPS). Where the acute inflammatory network was significantly enriched for NF-κB signaling, the primed microglia profile contained key features related to phagosome, lysosome, antigen presentation, and AD signaling. In addition, specific signatures for aging, AD, and ALS were identified. Microglia priming induces a highly conserved transcriptional signature with aging- and disease-specific aspects.

Journal ArticleDOI
TL;DR: A protocol for genome-wide profiling of gene expression in situ in fixed cells and tissues, in which RNA is converted into cross-linked cDNA amplicons and sequenced manually on a confocal microscope, which enriches for context-specific transcripts over housekeeping and/or structural RNA.
Abstract: RNA-sequencing (RNA-seq) measures the quantitative change in gene expression over the whole transcriptome, but it lacks spatial context. In contrast, in situ hybridization provides the location of gene expression, but only for a small number of genes. Here we detail a protocol for genome-wide profiling of gene expression in situ in fixed cells and tissues, in which RNA is converted into cross-linked cDNA amplicons and sequenced manually on a confocal microscope. Unlike traditional RNA-seq, our method enriches for context-specific transcripts over housekeeping and/or structural RNA, and it preserves the tissue architecture for RNA localization studies. Our protocol is written for researchers experienced in cell microscopy with minimal computing skills. Library construction and sequencing can be completed within 14 d, with image analysis requiring an additional 2 d.

Journal ArticleDOI
TL;DR: ABF1 is a functional homolog of AREB1, AREB2, and ABF3 in ABA-dependent gene expression from a comparative analysis between the areb 1 areb2 abf3 abf1 and areb1 areb 2 abF3 mutants.
Abstract: Under osmotic stress conditions such as drought and high salinity, the plant hormone abscisic acid (ABA) plays important roles in stress-responsive gene expression mainly through three bZIP transcription factors, AREB1/ABF2, AREB2/ABF4 and ABF3, which are activated by SNF1-related kinase 2s (SnRK2s) such as SRK2D/SnRK2.2, SRK2E/SnRK2.6 and SRK2I/SnRK2.3 (SRK2D/E/I). However, since the three AREB/ABFs are crucial, but not exclusive, for the SnRK2-mediated gene expression, transcriptional pathways governed by SRK2D/E/I are not fully understood. Here, we show that a bZIP transcription factor, ABF1, is a functional homolog of AREB1, AREB2 and ABF3 in ABA-dependent gene expression in Arabidopsis. Despite lower expression levels of ABF1 than those of the three AREB/ABFs, the areb1 areb2 abf3 abf1 mutant plants displayed increased sensitivity to drought and decreased sensitivity to ABA in primary root growth compared with the areb1 areb2 abf3 mutant. Genome-wide transcriptome analyses revealed that expression of downstream genes of SRK2D/E/I, which include many genes functioning in osmotic stress responses and tolerance such as transcription factors and LEA proteins, was mostly impaired in the quadruple mutant. Thus, these results indicate that the four AREB/ABFs are the predominant transcription factors downstream of SRK2D/E/I in ABA signalling in response to osmotic stress during vegetative growth.

Journal ArticleDOI
06 Mar 2015-Science
TL;DR: In this article, a quantitative genomic model of gene expression in lipopolysaccharide-stimulated mouse dendritic cells is presented, which combines measurements of protein production and degradation and mRNA dynamics.
Abstract: Protein expression is regulated by the production and degradation of messenger RNAs (mRNAs) and proteins, but their specific relationships remain unknown. We combine measurements of protein production and degradation and mRNA dynamics so as to build a quantitative genomic model of the differential regulation of gene expression in lipopolysaccharide-stimulated mouse dendritic cells. Changes in mRNA abundance play a dominant role in determining most dynamic fold changes in protein levels. Conversely, the preexisting proteome of proteins performing basic cellular functions is remodeled primarily through changes in protein production or degradation, accounting for more than half of the absolute change in protein molecules in the cell. Thus, the proteome is regulated by transcriptional induction for newly activated cellular functions and by protein life-cycle changes for remodeling of preexisting functions.

Journal ArticleDOI
TL;DR: It is found that differentially expressed mRNAs correlate significantly better with their protein product than non-differentially expressed MNCs, providing optimism for the usefulness of inferences from mRNA expression in general.
Abstract: Differential mRNA expression studies implicitly assume that changes in mRNA expression have biological meaning, most likely mediated by corresponding changes in protein levels. Yet studies into mRNA-protein correspondence have shown notoriously poor correlation between mRNA and protein expression levels, creating concern for inferences from only mRNA expression data. However, none of these studies have examined in particular differentially expressed mRNA. Here, we examined this question in an ovarian cancer xenograft model. We measured protein and mRNA expression for twenty-nine genes in four drug-treatment conditions and in untreated controls. We identified mRNAs differentially expressed between drug-treated xenografts and controls, then analysed mRNA-protein expression correlation across a five-point time-course within each of the four experimental conditions. We evaluated correlations between mRNAs and their protein products for mRNAs differentially expressed within an experimental condition compared to those that are not. We found that differentially expressed mRNAs correlate significantly better with their protein product than non-differentially expressed mRNAs. This result increases confidence for the use of differential mRNA expression for biological discovery in this system, as well as providing optimism for the usefulness of inferences from mRNA expression in general.

Journal ArticleDOI
TL;DR: For example, this article found that gene expression patterns in mouse models closely recapitulate those in human inflammatory conditions and strongly argue for the utility of mice as animal models of human disorders.
Abstract: The use of mice as animal models has long been considered essential in modern biomedical research, but the role of mouse models in research was challenged by a recent report that genomic responses in mouse models poorly mimic human inflammatory diseases. Here we reevaluated the same gene expression datasets used in the previous study by focusing on genes whose expression levels were significantly changed in both humans and mice. Contrary to the previous findings, the gene expression levels in the mouse models showed extraordinarily significant correlations with those of the human conditions (Spearman’s rank correlation coefficient: 0.43–0.68; genes changed in the same direction: 77–93%; P = 6.5 × 10−11 to 1.2 × 10−35). Moreover, meta-analysis of those datasets revealed a number of pathways/biogroups commonly regulated by multiple conditions in humans and mice. These findings demonstrate that gene expression patterns in mouse models closely recapitulate those in human inflammatory conditions and strongly argue for the utility of mice as animal models of human disorders.

Journal ArticleDOI
17 Sep 2015-Nature
TL;DR: It is found that oncogenic MYC confers a collateral stress on splicing, and that components of the spliceosome may be therapeutic entry points for aggressive MYC-driven cancers.
Abstract: MYC (also known as c-MYC) overexpression or hyperactivation is one of the most common drivers of human cancer Despite intensive study, the MYC oncogene remains recalcitrant to therapeutic inhibition MYC is a transcription factor, and many of its pro-tumorigenic functions have been attributed to its ability to regulate gene expression programs Notably, oncogenic MYC activation has also been shown to increase total RNA and protein production in many tissue and disease contexts While such increases in RNA and protein production may endow cancer cells with pro-tumour hallmarks, this increase in synthesis may also generate new or heightened burden on MYC-driven cancer cells to process these macromolecules properly Here we discover that the spliceosome is a new target of oncogenic stress in MYC-driven cancers We identify BUD31 as a MYC-synthetic lethal gene in human mammary epithelial cells, and demonstrate that BUD31 is a component of the core spliceosome required for its assembly and catalytic activity Core spliceosomal factors (such as SF3B1 and U2AF1) associated with BUD31 are also required to tolerate oncogenic MYC Notably, MYC hyperactivation induces an increase in total precursor messenger RNA synthesis, suggesting an increased burden on the core spliceosome to process pre-mRNA In contrast to normal cells, partial inhibition of the spliceosome in MYC-hyperactivated cells leads to global intron retention, widespread defects in pre-mRNA maturation, and deregulation of many essential cell processes Notably, genetic or pharmacological inhibition of the spliceosome in vivo impairs survival, tumorigenicity and metastatic proclivity of MYC-dependent breast cancers Collectively, these data suggest that oncogenic MYC confers a collateral stress on splicing, and that components of the spliceosome may be therapeutic entry points for aggressive MYC-driven cancers

Journal ArticleDOI
TL;DR: Findings in the field are summarized, highlighting those noncoding RNAs that regulate inflammation, with emphasis on recognized mediators such as TNF‐α, IL‐1,IL‐6, IL-18, intercellular adhesion molecule 1, VCAM‐1 and plasminogen activator inhibitor 1.
Abstract: Chronic inflammation is involved in the onset and development of many diseases, including obesity, atherosclerosis, type 2 diabetes, osteoarthritis, autoimmune and degenerative diseases, asthma, periodontitis, and cirrhosis. The inflammation process is mediated by chemokines, cytokines, and different inflammatory cells. Although the molecules and mechanisms that regulate this primary defense mechanism are not fully understood, recent findings offer a putative role of noncoding RNAs, especially microRNAs (miRNAs), in the progression and management of the inflammatory response. These noncoding RNAs are crucial for the stability and maintenance of gene expression patterns that characterize some cell types, tissues, and biologic responses. Several miRNAs, such as miR-126, miR-132, miR-146, miR-155, and miR-221, have emerged as important transcriptional regulators of some inflammation-related mediators. Additionally, little is known about the involvement of long noncoding RNAs, long intergenic noncoding RNAs, and circular RNAs in inflammation-mediated processes and the homeostatic imbalance associated with metabolic disorders. These noncoding RNAs are emerging as biomarkers with diagnosis value, in prognosis protocols, or in the personalized treatment of inflammation-related alterations. In this context, this review summarizes findings in the field, highlighting those noncoding RNAs that regulate inflammation, with emphasis on recognized mediators such as TNF-α, IL-1, IL-6, IL-18, intercellular adhesion molecule 1, VCAM-1, and plasminogen activator inhibitor 1. The down-regulation or antagonism of the noncoding RNAs and the administration of exogenous miRNAs could be, in the near future, a promising therapeutic strategy in the treatment of inflammation-related diseases.

Journal ArticleDOI
TL;DR: It is suggested that TEs provide an important source of allelic regulatory variation in gene response to abiotic stress in maize and may provide local enhancer activities that stimulate stress-responsive gene expression.
Abstract: Transposable elements (TEs) account for a large portion of the genome in many eukaryotic species. Despite their reputation as “junk” DNA or genomic parasites deleterious for the host, TEs have complex interactions with host genes and the potential to contribute to regulatory variation in gene expression. It has been hypothesized that TEs and genes they insert near may be transcriptionally activated in response to stress conditions. The maize genome, with many different types of TEs interspersed with genes, provides an ideal system to study the genome-wide influence of TEs on gene regulation. To analyze the magnitude of the TE effect on gene expression response to environmental changes, we profiled gene and TE transcript levels in maize seedlings exposed to a number of abiotic stresses. Many genes exhibit up- or down-regulation in response to these stress conditions. The analysis of TE families inserted within upstream regions of up-regulated genes revealed that between four and nine different TE families are associated with up-regulated gene expression in each of these stress conditions, affecting up to 20% of the genes up-regulated in response to abiotic stress, and as many as 33% of genes that are only expressed in response to stress. Expression of many of these same TE families also responds to the same stress conditions. The analysis of the stress-induced transcripts and proximity of the transposon to the gene suggests that these TEs may provide local enhancer activities that stimulate stress-responsive gene expression. Our data on allelic variation for insertions of several of these TEs show strong correlation between the presence of TE insertions and stress-responsive up-regulation of gene expression. Our findings suggest that TEs provide an important source of allelic regulatory variation in gene response to abiotic stress in maize.

Journal ArticleDOI
TL;DR: It is proposed that increased ER transcriptional activity may be a reactive mechanism that limits the activity of PI3K inhibitors and that combinedPI3K and ER inhibition is a rational approach to target these tumors.
Abstract: Activating mutations of PIK3CA are the most frequent genomic alterations in estrogen receptor (ER)–positive breast tumors, and selective phosphatidylinositol 3-kinase a (PI3Ka) inhibitors are in clinical development. The activity of these agents, however, is not homogeneous, and only a fraction of patients bearing PIK3CA-mutant ER-positive tumors benefit from single-agent administration. Searching for mechanisms of resistance, we observed that suppression of PI3K signaling results in induction of ER-dependent transcriptional activity, as demonstrated by changes in expression of genes containing ER-binding sites and increased occupancy by the ER of promoter regions of upregulated genes. Furthermore, expression of ESR1 mRNA and ER protein were also increased upon PI3K inhibition. These changes in gene expression were confirmed in vivo in xenografts and patient-derived models and in tumors from patients undergoing treatment with the PI3Ka inhibitor BYL719. The observed effects on transcription were enhanced by the addition of estradiol and suppressed by the anti-ER therapies fulvestrant and tamoxifen. Fulvestrant markedly sensitized ER-positive tumors to PI3Ka inhibition, resulting in major tumor regressions in vivo. We propose that increased ER transcriptional activity may be a reactive mechanism that limits the activity of PI3K inhibitors and that combined PI3K and ER inhibition is a rational approach to target these tumors.

Journal ArticleDOI
23 Apr 2015-Cell
TL;DR: A simple and powerful approach for performing native elongating transcript sequencing (NET-seq) in human cells that globally maps strand-specific Pol II density at nucleotide resolution and reveals stereotypic Pol II pausing coincident with transcription factor occupancy.

Journal ArticleDOI
TL;DR: The comprehensive identification of 2,006 genes producing mobile RNAs in Arabidopsis thaliana allowed the identification of mRNAs moving between various organs under normal or nutrient-limiting conditions and suggests that a postulated tissue-specific gene expression profile might not be predictive for the actual plant body part in which a transcript exerts its function.
Abstract: The concept that proteins and small RNAs can move to and function in distant body parts is well established. However, non-cell-autonomy of small RNA molecules raises the question: To what extent are protein-coding messenger RNAs (mRNAs) exchanged between tissues in plants? Here we report the comprehensive identification of 2,006 genes producing mobile RNAs in Arabidopsis thaliana. The analysis of variant ecotype transcripts that were present in heterografted plants allowed the identification of mRNAs moving between various organs under normal or nutrient-limiting conditions. Most of these mobile transcripts seem to follow the phloem-dependent allocation pathway transporting sugars from photosynthetic tissues to roots via the vasculature. Notably, a high number of transcripts also move in the opposite, root-to-shoot direction and are transported to specific tissues including flowers. Proteomic data on grafted plants indicate the presence of proteins from mobile RNAs, allowing the possibility that they may be translated at their destination site. The mobility of a high number of mRNAs suggests that a postulated tissue-specific gene expression profile might not be predictive for the actual plant body part in which a transcript exerts its function.

Journal ArticleDOI
TL;DR: This work reanalyzed expression profiles and observed a limited set of 'transcriptional components' that describe well-known biology, explain the vast majority of variation in gene expression and enable us to predict the biological function of genes.
Abstract: Many cancer-associated somatic copy number alterations (SCNAs) are known. Currently, one of the challenges is to identify the molecular downstream effects of these variants. Although several SCNAs are known to change gene expression levels, it is not clear whether each individual SCNA affects gene expression. We reanalyzed 77,840 expression profiles and observed a limited set of 'transcriptional components' that describe well-known biology, explain the vast majority of variation in gene expression and enable us to predict the biological function of genes. On correcting expression profiles for these components, we observed that the residual expression levels (in 'functional genomic mRNA' profiling) correlated strongly with copy number. DNA copy number correlated positively with expression levels for 99% of all abundantly expressed human genes, indicating global gene dosage sensitivity. By applying this method to 16,172 patient-derived tumor samples, we replicated many loci with aberrant copy numbers and identified recurrently disrupted genes in genomically unstable cancers.

Journal ArticleDOI
TL;DR: Repression of ncRNAs derived from the DLK1-DIO3 imprinted locus is associated with reduced neural lineage differentiation potential in hESCs.
Abstract: Pluripotent stem cells are increasingly used to build therapeutic models, including the transplantation of neural progenitors derived from human embryonic stem cells (hESCs). Recently, long non-coding RNAs (lncRNAs), including delta-like homolog 1 gene and the type III iodothyronine deiodinase gene (DLK1-DIO3) imprinted locus-derived maternally expressed gene 3 (MEG3), were found to be expressed during neural development. The deregulation of these lncRNAs is associated with various neurological diseases. The imprinted locus DLK1-DIO3 encodes abundant non-coding RNAs (ncRNAs) that are regulated by differential methylation of the locus. We aim to study the correlation between the DLK1-DIO3-derived ncRNAs and the capacity of hESCs to differentiate into neural lineages. We classified hESC sublines into MEG3-ON and MEG3-OFF based on the expression levels of MEG3 and its downstream microRNAs as detected by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). A cDNA microarray was used to analyze the gene expression profiles of hESCs. To investigate the capacity of neural differentiation in MEG3-ON and MEG3-OFF hESCs, we performed neural lineage differentiation followed by neural lineage marker expression and neurite formation analyses via qRT-PCR and immunocytochemistry, respectively. MEG3-knockdown via small interfering RNA (siRNA) and small hairpin RNA (shRNA) was used to investigate the potential causative effect of MEG3 in regulating neural lineage-related gene expression. DLK1-DIO3-derived ncRNAs were repressed in MEG3-OFF hESCs compared with those in the MEG3-ON hESCs. The transcriptome profile indicated that many genes related to nervous system development and neural-type tumors were differentially expressed in MEG3-OFF hESCs. Three independent MEG3-knockdown assays using different siRNA and shRNA constructs consistently resulted in downregulation of some neural lineage genes. Lower expression levels of stage-specific neural lineage markers and reduced neurite formation were observed in neural lineage-like cells derived from MEG3-OFF-associated hESCs compared with those in the MEG3-ON groups at the same time points after differentiation. Repression of ncRNAs derived from the DLK1-DIO3 imprinted locus is associated with reduced neural lineage differentiation potential in hESCs.

Journal ArticleDOI
TL;DR: Results of this study indicate that LncRNA MALAT1 regulates glucose‐induced up‐regulation of inflammatory mediators IL‐6 and TNF‐α through activation of SAA3.
Abstract: To examine whether the long non-coding RNA (lncRNA) metastasis associated lung adenocarcinoma transcript 1 (MALAT1) is altered in the endothelial cells in response to glucose and the significance of such alteration. We incubated human umbilical vein endothelial cells with media containing various glucose levels. We found an increase in MALAT1 expression peaking after 12 hrs of incubation in high glucose. This increase was associated with parallel increase in serum amyloid antigen 3 (SAA3), an inflammatory ligand and target of MALAT1 and was further accompanied by increase in mRNAs and proteins of inflammatory mediators, tumour necrosis factor alpha (TNF-α) and interleukin 6 (IL-6). Renal tissue from the diabetic animals showed similar changes. Such cellular alterations were prevented following MALAT1 specific siRNA transfection. Results of this study indicate that LncRNA MALAT1 regulates glucose-induced up-regulation of inflammatory mediators IL-6 and TNF-α through activation of SAA3. Identification of such novel mechanism may lead to the development of RNA-based therapeutics targeting MALAT1 for diabetes-induced micro and macro vascular complications.

Journal ArticleDOI
04 Jun 2015-Nature
TL;DR: The genome-wide discovery of human transcripts that interact with eIF3 using photoactivatable ribonucleoside-enhanced crosslinking and immunoprecipitation (PAR-CLIP) is reported, illuminating a new role for eif3 in governing a specialized repertoire of gene expression and suggesting that binding of e IF3 to specific mRNAs could be targeted to control carcinogenesis.
Abstract: Regulation of protein synthesis is fundamental for all aspects of eukaryotic biology by controlling development, homeostasis and stress responses. The 13-subunit, 800-kilodalton eukaryotic initiation factor 3 (eIF3) organizes initiation factor and ribosome interactions required for productive translation. However, current understanding of eIF3 function does not explain genetic evidence correlating eIF3 deregulation with tissue-specific cancers and developmental defects. Here we report the genome-wide discovery of human transcripts that interact with eIF3 using photoactivatable ribonucleoside-enhanced crosslinking and immunoprecipitation (PAR-CLIP). eIF3 binds to a highly specific program of messenger RNAs involved in cell growth control processes, including cell cycling, differentiation and apoptosis, via the mRNA 5' untranslated region. Surprisingly, functional analysis of the interaction between eIF3 and two mRNAs encoding the cell proliferation regulators c-JUN and BTG1 reveals that eIF3 uses different modes of RNA stem-loop binding to exert either translational activation or repression. Our findings illuminate a new role for eIF3 in governing a specialized repertoire of gene expression and suggest that binding of eIF3 to specific mRNAs could be targeted to control carcinogenesis.

Journal ArticleDOI
21 Jul 2015-eLife
TL;DR: In this paper, the authors used whole genome bisulfite sequencing to assess the spatio-temporal dynamics of mC in rice grown under phosphate starvation and recovery conditions, and identified widespread phosphate starvation-induced changes in mC, preferentially localized in transposable elements (TEs) close to highly induced genes.
Abstract: Cytosine DNA methylation (mC) is a genome modification that can regulate the expression of coding and non-coding genetic elements. However, little is known about the involvement of mC in response to environmental cues. Using whole genome bisulfite sequencing to assess the spatio-temporal dynamics of mC in rice grown under phosphate starvation and recovery conditions, we identified widespread phosphate starvation-induced changes in mC, preferentially localized in transposable elements (TEs) close to highly induced genes. These changes in mC occurred after changes in nearby gene transcription, were mostly DCL3a-independent, and could partially be propagated through mitosis, however no evidence of meiotic transmission was observed. Similar analyses performed in Arabidopsis revealed a very limited effect of phosphate starvation on mC, suggesting a species-specific mechanism. Overall, this suggests that TEs in proximity to environmentally induced genes are silenced via hypermethylation, and establishes the temporal hierarchy of transcriptional and epigenomic changes in response to stress.