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










Publication year range
1.
Nucleic Acids Res ; 52(8): 4483-4501, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38587191

ABSTRACT

Messenger RNA precursors (pre-mRNA) generally undergo 3' end processing by cleavage and polyadenylation (CPA), which is specified by a polyadenylation site (PAS) and adjacent RNA sequences and regulated by a large variety of core and auxiliary CPA factors. To date, most of the human CPA factors have been discovered through biochemical and proteomic studies. However, genetic identification of the human CPA factors has been hampered by the lack of a reliable genome-wide screening method. We describe here a dual fluorescence readthrough reporter system with a PAS inserted between two fluorescent reporters. This system enables measurement of the efficiency of 3' end processing in living cells. Using this system in combination with a human genome-wide CRISPR/Cas9 library, we conducted a screen for CPA factors. The screens identified most components of the known core CPA complexes and other known CPA factors. The screens also identified CCNK/CDK12 as a potential core CPA factor, and RPRD1B as a CPA factor that binds RNA and regulates the release of RNA polymerase II at the 3' ends of genes. Thus, this dual fluorescence reporter coupled with CRISPR/Cas9 screens reliably identifies bona fide CPA factors and provides a platform for investigating the requirements for CPA in various contexts.


Subject(s)
CRISPR-Cas Systems , Genes, Reporter , RNA Precursors , mRNA Cleavage and Polyadenylation Factors , Humans , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/genetics , Genome, Human , HEK293 Cells , mRNA Cleavage and Polyadenylation Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics , Polyadenylation , RNA Cleavage , RNA Polymerase II/metabolism , RNA Precursors/metabolism , RNA Precursors/genetics
2.
Mol Cell ; 82(17): 3135-3150.e9, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35914531

ABSTRACT

Alternative polyadenylation (APA) enhances gene regulatory potential by increasing the diversity of mRNA transcripts. 3' UTR shortening through APA correlates with enhanced cellular proliferation and is a widespread phenomenon in tumor cells. Here, we show that the ubiquitously expressed transcription factor Sp1 binds RNA in vivo and is a common repressor of distal poly(A) site usage. RNA sequencing identified 2,344 genes (36% of the total mapped mRNA transcripts) with lengthened 3' UTRs upon Sp1 depletion. Sp1 preferentially binds the 3' UTRs of such lengthened transcripts and inhibits cleavage at distal sites by interacting with the subunits of the core cleavage and polyadenylation (CPA) machinery. The 3' UTR lengths of Sp1 target genes in breast cancer patient RNA-seq data correlate with Sp1 expression levels, implicating Sp1-mediated APA regulation in modulating tumorigenic properties. Taken together, our findings provide insights into the mechanism for dynamic APA regulation by unraveling a previously unknown function of the DNA-binding transcription factor Sp1.


Subject(s)
Poly A , Polyadenylation , 3' Untranslated Regions , Humans , Poly A/metabolism , RNA, Messenger/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Zinc/metabolism
3.
Nat Commun ; 12(1): 335, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436550

ABSTRACT

Previous transcriptomic profiling studies have typically focused on separately analyzing mRNA expression, alternative splicing and alternative polyadenylation differences between cell and tissue types. However, the relative contribution of these three transcriptomic regulatory layers to cell type specification is poorly understood. This question is particularly relevant to neurons, given their extensive heterogeneity associated with brain location, morphology and function. In the present study, we generated profiles for the three regulatory layers from developmentally and regionally distinct subpopulations of neurons from the mouse hippocampus and broader nervous system. Multi-omics factor analyses revealed differing contributions of each transcriptomic layer in the discrimination of neurons based on their stage of development, region, and function. Importantly, profiles of differential alternative splicing and polyadenylation better discriminated specific neuronal subtype populations than gene expression patterns. These results provide evidence for differential relative contributions of coordinated gene regulatory layers in the specification of neuronal subtypes.


Subject(s)
Gene Expression Regulation , Neurons/metabolism , Transcriptome/genetics , 3' Untranslated Regions/genetics , Alternative Splicing/genetics , Animals , Down-Regulation/genetics , Hippocampus/anatomy & histology , Hippocampus/cytology , Mice , Polyadenylation/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Transcription, Genetic , Up-Regulation/genetics
4.
Nat Biotechnol ; 38(5): 638-648, 2020 05.
Article in English | MEDLINE | ID: mdl-32249828

ABSTRACT

Systematic mapping of genetic interactions (GIs) and interrogation of the functions of sizable genomic segments in mammalian cells represent important goals of biomedical research. To advance these goals, we present a CRISPR (clustered regularly interspaced short palindromic repeats)-based screening system for combinatorial genetic manipulation that employs coexpression of CRISPR-associated nucleases 9 and 12a (Cas9 and Cas12a) and machine-learning-optimized libraries of hybrid Cas9-Cas12a guide RNAs. This system, named Cas Hybrid for Multiplexed Editing and screening Applications (CHyMErA), outperforms genetic screens using Cas9 or Cas12a editing alone. Application of CHyMErA to the ablation of mammalian paralog gene pairs reveals extensive GIs and uncovers phenotypes normally masked by functional redundancy. Application of CHyMErA in a chemogenetic interaction screen identifies genes that impact cell growth in response to mTOR pathway inhibition. Moreover, by systematically targeting thousands of alternative splicing events, CHyMErA identifies exons underlying human cell line fitness. CHyMErA thus represents an effective screening approach for GI mapping and the functional analysis of sizable genomic regions, such as alternative exons.


Subject(s)
Bacterial Proteins/metabolism , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Proteins/metabolism , Endodeoxyribonucleases/metabolism , Gene Editing/methods , Gene Regulatory Networks , Alternative Splicing , Animals , CRISPR-Cas Systems , Cell Line , Genetic Fitness , Humans , Machine Learning , Male , Mice , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
5.
Cell Rep ; 30(12): 4179-4196.e11, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32209477

ABSTRACT

Regulation of translation during human development is poorly understood, and its dysregulation is associated with Rett syndrome (RTT). To discover shifts in mRNA ribosomal engagement (RE) during human neurodevelopment, we use parallel translating ribosome affinity purification sequencing (TRAP-seq) and RNA sequencing (RNA-seq) on control and RTT human induced pluripotent stem cells, neural progenitor cells, and cortical neurons. We find that 30% of transcribed genes are translationally regulated, including key gene sets (neurodevelopment, transcription and translation factors, and glycolysis). Approximately 35% of abundant intergenic long noncoding RNAs (lncRNAs) are ribosome engaged. Neurons translate mRNAs more efficiently and have longer 3' UTRs, and RE correlates with elements for RNA-binding proteins. RTT neurons have reduced global translation and compromised mTOR signaling, and >2,100 genes are translationally dysregulated. NEDD4L E3-ubiquitin ligase is translationally impaired, ubiquitinated protein levels are reduced, and protein targets accumulate in RTT neurons. Overall, the dynamic translatome in neurodevelopment is disturbed in RTT and provides insight into altered ubiquitination that may have therapeutic implications.


Subject(s)
Nervous System/growth & development , Nervous System/pathology , Rett Syndrome/genetics , Ribosomes/metabolism , Ubiquitination , 3' Untranslated Regions/genetics , Animals , Base Sequence , Female , Gene Expression Regulation, Developmental , Glycolysis/genetics , Induced Pluripotent Stem Cells/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Mice , Nedd4 Ubiquitin Protein Ligases/metabolism , Neurons/metabolism , Protein Binding , Protein Biosynthesis , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , RNA-Binding Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Transcription Factors/metabolism , Ubiquitination/genetics
6.
Mol Cell ; 72(1): 187-200.e6, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30220560

ABSTRACT

Alternative splicing (AS) is a widespread process underlying the generation of transcriptomic and proteomic diversity and is frequently misregulated in human disease. Accordingly, an important goal of biomedical research is the development of tools capable of comprehensively, accurately, and efficiently profiling AS. Here, we describe Whippet, an easy-to-use RNA-seq analysis method that rapidly-with hardware requirements compatible with a laptop-models and quantifies AS events of any complexity without loss of accuracy. Using an entropic measure of splicing complexity, Whippet reveals that one-third of human protein coding genes produce transcripts with complex AS events involving co-expression of two or more principal splice isoforms. We observe that high-entropy AS events are more prevalent in tumor relative to matched normal tissues and correlate with increased expression of proto-oncogenic splicing factors. Whippet thus affords the rapid and accurate analysis of AS events of any complexity, and as such will facilitate future biomedical research.


Subject(s)
Alternative Splicing/genetics , Proteomics , RNA Splicing/genetics , Sequence Analysis, RNA/methods , Gene Expression Profiling/methods , Humans , Molecular Sequence Annotation , RNA, Messenger/genetics , Transcriptome
7.
Genome Biol ; 19(1): 45, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29592814

ABSTRACT

Alternative polyadenylation (APA) affects most mammalian genes. The genome-wide investigation of APA has been hampered by an inability to reliably profile it using conventional RNA-seq. We describe 'Quantification of APA' (QAPA), a method that infers APA from conventional RNA-seq data. QAPA is faster and more sensitive than other methods. Application of QAPA reveals discrete, temporally coordinated APA programs during neurogenesis and that there is little overlap between genes regulated by alternative splicing and those by APA. Modeling of these data uncovers an APA sequence code. QAPA thus enables the discovery and characterization of programs of regulated APA using conventional RNA-seq.


Subject(s)
Polyadenylation , Sequence Analysis, RNA/methods , Alternative Splicing , Animals , Cell Differentiation/genetics , Gene Expression Profiling , High-Throughput Nucleotide Sequencing/methods , Humans , Mice , Neurons/metabolism , Transcription Initiation Site
8.
Genome Res ; 27(10): 1759-1768, 2017 10.
Article in English | MEDLINE | ID: mdl-28855263

ABSTRACT

Alternative splicing (AS) generates remarkable regulatory and proteomic complexity in metazoans. However, the functions of most AS events are not known, and programs of regulated splicing remain to be identified. To address these challenges, we describe the Vertebrate Alternative Splicing and Transcription Database (VastDB), the largest resource of genome-wide, quantitative profiles of AS events assembled to date. VastDB provides readily accessible quantitative information on the inclusion levels and functional associations of AS events detected in RNA-seq data from diverse vertebrate cell and tissue types, as well as developmental stages. The VastDB profiles reveal extensive new intergenic and intragenic regulatory relationships among different classes of AS and previously unknown and conserved landscapes of tissue-regulated exons. Contrary to recent reports concluding that nearly all human genes express a single major isoform, VastDB provides evidence that at least 48% of multiexonic protein-coding genes express multiple splice variants that are highly regulated in a cell/tissue-specific manner, and that >18% of genes simultaneously express multiple major isoforms across diverse cell and tissue types. Isoforms encoded by the latter set of genes are generally coexpressed in the same cells and are often engaged by translating ribosomes. Moreover, they are encoded by genes that are significantly enriched in functions associated with transcriptional control, implying they may have an important and wide-ranging role in controlling cellular activities. VastDB thus provides an unprecedented resource for investigations of AS function and regulation.


Subject(s)
Alternative Splicing , Databases, Nucleic Acid , Exons , Gene Regulatory Networks , Protein Isoforms , Animals , Chickens , Humans , Mice , Protein Isoforms/biosynthesis , Protein Isoforms/genetics
9.
Cancer Cell ; 32(1): 101-114.e8, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28697339

ABSTRACT

Global transcriptomic imbalance is a ubiquitous feature associated with cancer, including hepatocellular carcinoma (HCC). Analyses of 1,225 clinical HCC samples revealed that a large numbers of RNA binding proteins (RBPs) are dysregulated and that RBP dysregulation is associated with poor prognosis. We further identified that oncogenic activation of a top candidate RBP, negative elongation factor E (NELFE), via somatic copy-number alterations enhanced MYC signaling and promoted HCC progression. Interestingly, NELFE induces a unique tumor transcriptome by selectively regulating MYC-associated genes. Thus, our results revealed NELFE as an oncogenic protein that may contribute to transcriptome imbalance in HCC through the regulation of MYC signaling.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/physiology , Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Methods ; 126: 18-28, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28651966

ABSTRACT

RNA-binding proteins recognize RNA sequences and structures, but there is currently no systematic and accurate method to derive large (>12base) motifs de novo that reflect a combination of intrinsic preference to both sequence and structure. To address this absence, we introduce RNAcompete-S, which couples a single-step competitive binding reaction with an excess of random RNA 40-mers to a custom computational pipeline for interrogation of the bound RNA sequences and derivation of SSMs (Sequence and Structure Models). RNAcompete-S confirms that HuR, QKI, and SRSF1 prefer binding sites that are single stranded, and recapitulates known 8-10bp sequence and structure preferences for Vts1p and RBMY. We also derive an 18-base long SSM for Drosophila SLBP, which to our knowledge has not been previously determined by selections from pure random sequence, and accurately discriminates human replication-dependent histone mRNAs. Thus, RNAcompete-S enables accurate identification of large, intrinsic sequence-structure specificities with a uniform assay.


Subject(s)
Base Sequence/genetics , High-Throughput Nucleotide Sequencing/methods , RNA-Binding Proteins/genetics , Humans , RNA-Binding Proteins/chemistry , Sequence Analysis, RNA/methods
11.
Mol Cell ; 65(3): 539-553.e7, 2017 Feb 02.
Article in English | MEDLINE | ID: mdl-28157508

ABSTRACT

Networks of coordinated alternative splicing (AS) events play critical roles in development and disease. However, a comprehensive knowledge of the factors that regulate these networks is lacking. We describe a high-throughput system for systematically linking trans-acting factors to endogenous RNA regulatory events. Using this system, we identify hundreds of factors associated with diverse regulatory layers that positively or negatively control AS events linked to cell fate. Remarkably, more than one-third of the regulators are transcription factors. Further analyses of the zinc finger protein Zfp871 and BTB/POZ domain transcription factor Nacc1, which regulate neural and stem cell AS programs, respectively, reveal roles in controlling the expression of specific splicing regulators. Surprisingly, these proteins also appear to regulate target AS programs via binding RNA. Our results thus uncover a large "missing cache" of splicing regulators among annotated transcription factors, some of which dually regulate AS through direct and indirect mechanisms.


Subject(s)
Alternative Splicing , Gene Regulatory Networks , Sequence Analysis, RNA/methods , Transcription Factors/metabolism , Animals , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , HEK293 Cells , Humans , Mice , Neurons/cytology , Neurons/metabolism , RNA, Messenger/genetics
12.
Methods ; 118-119: 3-15, 2017 04 15.
Article in English | MEDLINE | ID: mdl-27956239

ABSTRACT

RNA-binding proteins (RBPs) participate in diverse cellular processes and have important roles in human development and disease. The human genome, and that of many other eukaryotes, encodes hundreds of RBPs that contain canonical sequence-specific RNA-binding domains (RBDs) as well as numerous other unconventional RNA binding proteins (ucRBPs). ucRBPs physically associate with RNA but lack common RBDs. The degree to which these proteins bind RNA, in a sequence specific manner, is unknown. Here, we provide a detailed description of both the laboratory and data processing methods for RNAcompete, a method we have previously used to analyze the RNA binding preferences of hundreds of RBD-containing RBPs, from diverse eukaryotes. We also determine the RNA-binding preferences for two human ucRBPs, NUDT21 and CNBP, and use this analysis to exemplify the RNAcompete pipeline. The results of our RNAcompete experiments are consistent with independent RNA-binding data for these proteins and demonstrate the utility of RNAcompete for analyzing the growing repertoire of ucRBPs.


Subject(s)
Cleavage And Polyadenylation Specificity Factor/genetics , Microarray Analysis/methods , RNA-Binding Proteins/genetics , RNA/chemistry , Animals , Base Sequence , Binding Sites , Cleavage And Polyadenylation Specificity Factor/metabolism , Cloning, Molecular , DNA Primers/chemistry , DNA Primers/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Protein Binding , Protein Domains , RNA/genetics , RNA/metabolism , RNA-Binding Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment
13.
Cell Rep ; 17(3): 720-734, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27732849

ABSTRACT

A progressive increase in MECP2 protein levels is a crucial and precisely regulated event during neurodevelopment, but the underlying mechanism is unclear. We report that MECP2 is regulated post-transcriptionally during in vitro differentiation of human embryonic stem cells (hESCs) into cortical neurons. Using reporters to identify functional RNA sequences in the MECP2 3' UTR and genetic manipulations to explore the role of interacting factors on endogenous MECP2, we discover combinatorial mechanisms that regulate RNA stability and translation. The RNA-binding protein PUM1 and pluripotent-specific microRNAs destabilize the long MECP2 3' UTR in hESCs. Hence, the 3' UTR appears to lengthen during differentiation as the long isoform becomes stable in neurons. Meanwhile, translation of MECP2 is repressed by TIA1 in hESCs until HuC predominates in neurons, resulting in a switch to translational enhancement. Ultimately, 3' UTR-directed translational fine-tuning differentially modulates MECP2 protein in the two cell types to levels appropriate for normal neurodevelopment.


Subject(s)
Gene Expression Regulation , Methyl-CpG-Binding Protein 2/genetics , MicroRNAs/metabolism , Neurons/metabolism , RNA-Binding Proteins/metabolism , Transcription, Genetic , 3' Untranslated Regions/genetics , AU Rich Elements/genetics , Base Sequence , Cell Lineage , Cell Proliferation , Conserved Sequence/genetics , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Methyl-CpG-Binding Protein 2/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Prosencephalon/embryology , Protein Binding/genetics , Protein Biosynthesis , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
Hum Mutat ; 34(10): 1366-70, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23878101

ABSTRACT

Isolated cytochrome c oxidase (COX) deficiency is a common cause of mitochondrial disease, yet its genetic basis remains unresolved in many patients. Here, we identified novel compound heterozygous mutations in SCO1 (p.M294V, p.Val93*) in one such patient with fatal encephalopathy. The patient lacked the severe hepatopathy (p.P174L) or hypertrophic cardiomyopathy (p.G132S) observed in previously reported SCO1 cases, so we investigated whether allele-specific defects in SCO1 function might underlie the genotype-phenotype relationships. Fibroblasts expressing p.M294V had a relatively modest decrease in COX activity compared with those expressing p.P174L, whereas both SCO1 lines had marked copper deficiencies. Overexpression of known pathogenic variants in SCO1 fibroblasts showed that p.G132S exacerbated the COX deficiency, whereas COX activity was partially or fully restored by p.P174L and p.M294V, respectively. These data suggest that the clinical phenotypes in SCO1 patients might reflect the residual capacity of the pathogenic alleles to perform one or both functions of SCO1.


Subject(s)
Acidosis, Lactic/genetics , Membrane Proteins/genetics , Mutation , Olivopontocerebellar Atrophies/genetics , Acidosis, Lactic/metabolism , Alleles , Amino Acid Sequence , DNA Mutational Analysis , Fatal Outcome , Gene Order , Humans , Infant , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Molecular Chaperones , Molecular Sequence Data , Olivopontocerebellar Atrophies/metabolism , Sequence Alignment
15.
J Med Genet ; 50(5): 330-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23434736

ABSTRACT

BACKGROUND: Congenital nephrotic syndrome arises from a defect in the glomerular filtration barrier that permits the unrestricted passage of protein across the barrier, resulting in proteinuria, hypoalbuminaemia, and severe oedema. While most cases are due to mutations in one of five genes, in up to 15% of cases, a genetic cause is not identified. We investigated two sisters with a presumed recessive form of congenital nephrotic syndrome. METHODS AND RESULTS: Whole exome sequencing identified five genes with diallelic mutations that were shared by the sisters, and Sanger sequencing revealed that ARHGDIA that encodes Rho GDP (guanosine diphosphate) dissociation inhibitor α (RhoGDIα, OMIM 601925) was the most likely candidate. Mice with targeted inactivation of ARHGDIA are known to develop severe proteinuria and nephrotic syndrome, therefore this gene was pursued in functional studies. The sisters harbour a homozygous in-frame deletion that is predicted to remove a highly conserved aspartic acid residue within the interface where the protein, RhoGDIα, interacts with the Rho family of small GTPases (c.553_555del(p.Asp185del)). Rho-GTPases are critical regulators of the actin cytoskeleton and when bound to RhoGDIα, they are sequestered in an inactive, cytosolic pool. In the mouse kidney, RhoGDIα was highly expressed in podocytes, a critical cell within the glomerular filtration barrier. When transfected in HEK293T cells, the mutant RhoGDIα was unable to bind to the Rho-GTPases, RhoA, Rac1, and Cdc42, unlike the wild-type construct. When RhoGDIα was knocked down in podocytes, RhoA, Rac1, and Cdc42 were hyperactivated and podocyte motility was impaired. The proband's fibroblasts demonstrated mislocalisation of RhoGDIα to the nucleus, hyperactivation of the three Rho-GTPases, and impaired cell motility, suggesting that the in-frame deletion leads to a loss of function. CONCLUSIONS: Mutations in ARHGDIA need to be considered in the aetiology of heritable forms of nephrotic syndrome.


Subject(s)
Exome/genetics , Kidney/pathology , Nephrotic Syndrome/genetics , Nephrotic Syndrome/pathology , rho Guanine Nucleotide Dissociation Inhibitor alpha/genetics , Amino Acid Sequence , Analysis of Variance , Animals , Base Sequence , Computational Biology , DNA Primers/genetics , Fatal Outcome , Female , Fluorescent Antibody Technique , HEK293 Cells , Humans , Immunohistochemistry , Infant, Newborn , Mice , Molecular Sequence Data , Pakistan , Pedigree , Sequence Analysis, DNA
16.
BMC Med Genomics ; 4: 75, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-22032724

ABSTRACT

BACKGROUND: Gene fusions arising from chromosomal translocations have been implicated in cancer. However, the role of gene fusions in BRCA1-related breast cancers is not well understood. Mutations in BRCA1 are associated with an increased risk for breast cancer (up to 80% lifetime risk) and ovarian cancer (up to 50%). We sought to identify putative gene fusions in the transcriptomes of these cancers using high-throughput RNA sequencing (RNA-Seq). METHODS: We used Illumina sequencing technology to sequence the transcriptomes of five BRCA1-mutated breast cancer cell lines, three BRCA1-mutated primary tumors, two secretory breast cancer primary tumors and one non-tumorigenic breast epithelial cell line. Using a bioinformatics approach, our initial attempt at discovering putative gene fusions relied on analyzing single-end reads and identifying reads that aligned across exons of two different genes. Subsequently, latter samples were sequenced with paired-end reads and at longer cycles (producing longer reads). We then refined our approach by identifying misaligned paired reads, which may flank a putative gene fusion junction. RESULTS: As a proof of concept, we were able to identify two previously characterized gene fusions in our samples using both single-end and paired-end approaches. In addition, we identified three novel in-frame fusions, but none were recurrent. Two of the candidates, WWC1-ADRBK2 in HCC3153 cell line and ADNP-C20orf132 in a primary tumor, were confirmed by Sanger sequencing and RT-PCR. RNA-Seq expression profiling of these two fusions showed a distinct overexpression of the 3' partner genes, suggesting that its expression may be under the control of the 5' partner gene's regulatory elements. CONCLUSIONS: In this study, we used both single-end and paired-end sequencing strategies to discover gene fusions in breast cancer transcriptomes with BRCA1 mutations. We found that the use of paired-end reads is an effective tool for transcriptome profiling of gene fusions. Our findings suggest that while gene fusions are present in some BRCA1-mutated breast cancers, they are infrequent and not recurrent. However, private fusions may still be valuable as potential patient-specific biomarkers for diagnosis and treatment.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/pathology , Gene Expression Profiling/methods , Gene Fusion/genetics , High-Throughput Nucleotide Sequencing/methods , Mutation , Sequence Analysis, RNA/methods , Base Sequence , Breast Neoplasms/genetics , Cell Line, Tumor , Chromosome Breakpoints , DNA Copy Number Variations/genetics , Feasibility Studies , Female , Humans , Molecular Sequence Data , RNA, Messenger/genetics
17.
J Med Genet ; 48(9): 602-5, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21785126

ABSTRACT

BACKGROUND: Combined Malonic and Methylmalonic Aciduria (CMAMMA) is a rare recessive inborn error of metabolism characterised by elevations of urine malonic acid (MA) and methylmalonic acid (MMA). Nearly all reported cases are caused by malonyl-CoA decarboxylase (MCD) deficiency. Most patients have metabolic acidosis, developmental delay, seizures and cardiomyopathy. CMAMMA was also described in symptomatic patients with normal MCD activity, suggesting heterogeneity in this disorder. METHODS AND RESULTS: We identified two probands with a non-classical CMAMMA variant through the Quebec newborn urine screening program. While they share the biochemical phenotype of elevated MA and MMA, the MMA excretion was higher than MA, the clinical courses were benign, MYLCD gene sequencing was normal and MCD activity, measured in one proband, was normal. Using exome sequencing in the single consanguineous proband, we identified a homozygous missense allele in the ACSF3 gene, encoding an Acyl-CoA Synthetase (ACS) with unknown substrate and function. The second proband was homozygous for a different ACSF3 missense allele. Both substitutions were in conserved residues and were identified in less than 0.5% of their respective ethnic control populations. CONCLUSION: These results suggest that ACSF3 is a candidate gene for non-classical CMAMMA observed in our patients and document the value of exome sequencing of a limited number of patients for the identification of novel disease genes.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Coenzyme A Ligases/genetics , Exome , Metabolism, Inborn Errors/genetics , Mutation , Alleles , Base Sequence , Carboxy-Lyases/deficiency , Carboxy-Lyases/genetics , Genetic Association Studies , Humans , Infant , Malonates/urine , Malonyl Coenzyme A , Methylmalonic Acid/urine , Molecular Sequence Data , Pedigree , Phenotype , Sequence Analysis, DNA
18.
Genome Res ; 21(4): 545-54, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21173033

ABSTRACT

Expression levels of many human genes are under the genetic control of expression quantitative trait loci (eQTLs). Despite technological advances, the precise molecular mechanisms underlying most eQTLs remain elusive. Here, we use deep mRNA sequencing of two CEU individuals to investigate those mechanisms, with particular focus on the role of splicing control loci (sQTLs). We identify a large number of genes that are differentially spliced between the two samples and associate many of those differences with nearby single nucleotide polymorphisms (SNPs). Subsequently, we investigate the potential effect of splicing SNPs on eQTL control in general. We find a significant enrichment of alternative splicing (AS) events within a set of highly confident eQTL targets discovered in previous studies, suggesting a role of AS in regulating overall gene expression levels. Next, we demonstrate high correlation between the levels of mature (exonic) and unprocessed (intronic) RNA, implying that ∼75% of eQTL target variance can be explained by control at the level of transcription, but that the remaining 25% may be regulated co- or post-transcriptionally. We focus on eQTL targets with discordant mRNA and pre-mRNA expression patterns and use four examples: USMG5, MMAB, MRPL43, and OAS1, to dissect the exact downstream effects of the associated genetic variants.


Subject(s)
Gene Expression Regulation , Polymorphism, Genetic , RNA Splicing/genetics , Sequence Analysis, RNA , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Cell Line , Exons , Gene Order , Humans , Introns , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Quantitative Trait Loci/genetics , Transcription, Genetic
19.
Am J Hum Genet ; 87(4): 553-9, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20887961

ABSTRACT

Van Den Ende-Gupta syndrome (VDEGS) is an extremely rare autosomal-recessive disorder characterized by distinctive craniofacial features, which include blepharophimosis, malar and/or maxillary hypoplasia, a narrow and beaked nose, and an everted lower lip. Other features are arachnodactyly, camptodactyly, peculiar skeletal abnormalities, and normal development and intelligence. We present molecular data on four VDEGS patients from three consanguineous Qatari families belonging to the same highly inbred Bedouin tribe. The patients were genotyped with SNP microarrays, and a 2.4 Mb homozygous region was found on chromosome 22q11 in an area overlapping the DiGeorge critical region. This region contained 44 genes, including SCARF2, a gene that is expressed during development in a number of mouse tissues relevant to the symptoms described above. Sanger sequencing identified a missense change, c.773G>A (p.C258Y), in exon 4 in the two closely related patients and a 2 bp deletion in exon 8, c.1328_1329delTG (p.V443DfsX83), in two unrelated individuals. In parallel with the candidate gene approach, complete exome sequencing was used to confirm that SCARF2 was the gene responsible for VDEGS. SCARF2 contains putative epidermal growth factor-like domains in its extracellular domain, along with a number of positively charged residues in its intracellular domain, indicating that it may be involved in intracellular signaling. However, the function of SCARF2 has not been characterized, and this study reports that phenotypic effects can be associated with defects in the scavenger receptor F family of genes.


Subject(s)
Abnormalities, Multiple/genetics , Blepharophimosis/genetics , Chromosomes, Human, Pair 22/genetics , Ethnicity/genetics , Musculoskeletal Abnormalities/genetics , Scavenger Receptors, Class F/genetics , Amino Acid Sequence , Base Sequence , Female , Genes, Recessive , Genotype , Humans , Male , Microarray Analysis , Molecular Sequence Data , Mutation, Missense/genetics , Pedigree , Polymorphism, Single Nucleotide/genetics , Qatar , Scavenger Receptors, Class F/metabolism , Sequence Analysis, DNA , Syndrome
20.
Hum Mutat ; 31(8): 918-23, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20518025

ABSTRACT

Protein coding genes constitute approximately 1% of the human genome but harbor 85% of the mutations with large effects on disease-related traits. Therefore, efficient strategies for selectively sequencing complete coding regions (i.e., "whole exome") have the potential to contribute our understanding of human diseases. We used a method for whole-exome sequencing coupling Agilent whole-exome capture to the Illumina DNA-sequencing platform, and investigated two unrelated fetuses from nonconsanguineous families with Fowler Syndrome (FS), a stereotyped phenotype lethal disease. We report novel germline mutations in feline leukemia virus subgroup C cellular-receptor-family member 2, FLVCR2, which has recently been shown to cause FS. Using this technology, we identified three types of genetic abnormalities: point-mutations, insertions-deletions, and intronic splice-site changes (first pathogenic report using this technology), in the fetuses who both were compound heterozygotes for the disease. Although revealing a high level of allelic heterogeneity and mutational spectrum in FS, this study further illustrates the successful application of whole-exome sequencing to uncover genetic defects in rare Mendelian disorders. Of importance, we show that we can identify genes underlying rare, monogenic and recessive diseases using a limited number of patients (n=2), in the absence of shared genetic heritage and in the presence of allelic heterogeneity.


Subject(s)
Abnormalities, Multiple/genetics , Alleles , DNA Mutational Analysis/methods , Exons/genetics , Genetic Heterogeneity , Mutation/genetics , Abnormalities, Multiple/etiology , Abnormalities, Multiple/pathology , Female , Humans , Reproducibility of Results , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...