Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nat Commun ; 15(1): 749, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38272908

ABSTRACT

Transposable elements (TEs) are prevalent repeats in the human genome, play a significant role in the regulome, and their disruption can contribute to tumorigenesis. However, TE influence on gene expression in cancer remains unclear. Here, we analyze 275 normal colon and 276 colorectal cancer samples from the SYSCOL cohort, discovering 10,231 and 5,199 TE-expression quantitative trait loci (eQTLs) in normal and tumor tissues, respectively, of which 376 are colorectal cancer specific eQTLs, likely due to methylation changes. Tumor-specific TE-eQTLs show greater enrichment of transcription factors, compared to shared TE-eQTLs suggesting specific regulation of their expression in tumor. Bayesian networks reveal 1,766 TEs as mediators of genetic effects, altering the expression of 1,558 genes, including 55 known cancer driver genes and show that tumor-specific TE-eQTLs trigger the driver capability of TEs. These insights expand our knowledge of cancer drivers, deepening our understanding of tumorigenesis and presenting potential avenues for therapeutic interventions.


Subject(s)
Colorectal Neoplasms , DNA Transposable Elements , Humans , DNA Transposable Elements/genetics , Bayes Theorem , Transcription Factors/metabolism , Carcinogenesis/genetics , Colorectal Neoplasms/genetics
2.
BMC Genomics ; 24(1): 442, 2023 Aug 05.
Article in English | MEDLINE | ID: mdl-37543566

ABSTRACT

BACKGROUND: Expression quantitative trait loci (eQTL) studies provide insights into regulatory mechanisms underlying disease risk. Expanding studies of gene regulation to underexplored populations and to medically relevant tissues offers potential to reveal yet unknown regulatory variants and to better understand disease mechanisms. Here, we performed eQTL mapping in subcutaneous (S) and visceral (V) adipose tissue from 106 Greek individuals (Greek Metabolic study, GM) and compared our findings to those from the Genotype-Tissue Expression (GTEx) resource. RESULTS: We identified 1,930 and 1,515 eGenes in S and V respectively, over 13% of which are not observed in GTEx adipose tissue, and that do not arise due to different ancestry. We report additional context-specific regulatory effects in genes of clinical interest (e.g. oncogene ST7) and in genes regulating responses to environmental stimuli (e.g. MIR21, SNX33). We suggest that a fraction of the reported differences across populations is due to environmental effects on gene expression, driving context-specific eQTLs, and suggest that environmental effects can determine the penetrance of disease variants thus shaping disease risk. We report that over half of GM eQTLs colocalize with GWAS SNPs and of these colocalizations 41% are not detected in GTEx. We also highlight the clinical relevance of S adipose tissue by revealing that inflammatory processes are upregulated in individuals with obesity, not only in V, but also in S tissue. CONCLUSIONS: By focusing on an understudied population, our results provide further candidate genes for investigation regarding their role in adipose tissue biology and their contribution to disease risk and pathogenesis.


Subject(s)
Genetic Predisposition to Disease , Quantitative Trait Loci , Humans , Greece , Gene Expression Regulation , Genotype , Polymorphism, Single Nucleotide , Genome-Wide Association Study/methods
3.
Mol Psychiatry ; 27(12): 5177-5185, 2022 12.
Article in English | MEDLINE | ID: mdl-36114277

ABSTRACT

Schizophrenia is a polygenic psychiatric disorder with limited understanding about the mechanistic changes in gene expression regulation. To elucidate on this, we integrate interindividual variability of regulatory activity (ChIP-sequencing for H3K27ac histone mark) with gene expression and genotype data captured from the prefrontal cortex of 272 cases and controls. By measuring interindividual correlation among proximal chromatin peaks, we show that regulatory element activity is structured into 10,936 and 10,376 cis-regulatory domains in cases and controls, respectively. The schizophrenia-specific cis-regulatory domains are enriched for fetal-specific (p = 0.0014, OR = 1.52) and depleted of adult-specific regulatory activity (p = 3.04 × 10-50, OR = 0.57) and are enriched for SCZ heritability (p = 0.001). By studying the interplay among genetic variants, gene expression, and cis-regulatory domains, we ascertain that changes in coordinated regulatory activity tag alterations in gene expression levels (p = 3.43 × 10-5, OR = 1.65), unveil case-specific QTL effects, and identify regulatory machinery changes for genes affecting synaptic function and dendritic spine morphology in schizophrenia. Altogether, we show that accounting for coordinated regulatory activity provides a novel mechanistic approach to reduce the search space for unveiling genetically perturbed regulation of gene expression in schizophrenia.


Subject(s)
Schizophrenia , Adult , Humans , Schizophrenia/genetics , Gene Expression Regulation , Prefrontal Cortex/metabolism , Chromatin/metabolism , Multifactorial Inheritance , Genetic Predisposition to Disease
4.
PLoS Genet ; 18(6): e1010212, 2022 06.
Article in English | MEDLINE | ID: mdl-35666741

ABSTRACT

The Human Leukocyte Antigen (HLA) is a critical genetic system for different outcomes after solid organ and hematopoietic cell transplantation. Its polymorphism is usually determined by molecular technologies at the DNA level. A potential role of HLA allelic expression remains under investigation in the context of the allogenic immune response between donors and recipients. In this study, we quantified the allelic expression of all three HLA class I loci (HLA-A, B and C) by RNA sequencing and conducted an analysis of expression quantitative traits loci (eQTL) to investigate whether HLA expression regulation could be associated with non-coding gene variations. HLA-B alleles exhibited the highest expression levels followed by HLA-C and HLA-A alleles. The max fold expression variation was observed for HLA-C alleles. The expression of HLA class I loci of distinct individuals demonstrated a coordinated and paired expression of both alleles of the same locus. Expression of conserved HLA-A~B~C haplotypes differed in distinct PBMC's suggesting an individual regulated expression of both HLA class I alleles and haplotypes. Cytokines TNFα /IFNß, which induced a very similar upregulation of HLA class I RNA and cell surface expression across alleles did not modify the individually coordinated expression at the three HLA class I loci. By identifying cis eQTLs for the HLA class I genes, we show that the non-coding eQTLs explain 29%, 13%, and 31% of the respective HLA-A, B, C expression variance in unstimulated cells, and 9%, 23%, and 50% of the variance in cytokine-stimulated cells. The eQTLs have significantly higher effect sizes in stimulated cells compared to unstimulated cells for HLA-B and HLA-C genes expression. Our data also suggest that the identified eQTLs are independent from the coding variation which defines HLA alleles and thus may be influential on intra-allele expression variability although they might not represent the causal eQTLs.


Subject(s)
HLA-C Antigens , Leukocytes, Mononuclear , Alleles , Gene Frequency , HLA Antigens , HLA-A Antigens/genetics , HLA-B Antigens/genetics , HLA-C Antigens/genetics , Haplotypes , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class II/genetics , Humans
6.
Nat Commun ; 11(1): 2025, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32332866

ABSTRACT

Transcriptional characterization and classification has potential to resolve the inter-tumor heterogeneity of colorectal cancer and improve patient management. Yet, robust transcriptional profiling is difficult using formalin-fixed, paraffin-embedded (FFPE) samples, which complicates testing in clinical and archival material. We present MethCORR, an approach that allows uniform molecular characterization and classification of fresh-frozen and FFPE samples. MethCORR identifies genome-wide correlations between RNA expression and DNA methylation in fresh-frozen samples. This information is used to infer gene expression information in FFPE samples from their methylation profiles. MethCORR is here applied to methylation profiles from 877 fresh-frozen/FFPE samples and comparative analysis identifies the same two subtypes in four independent cohorts. Furthermore, subtype-specific prognostic biomarkers that better predicts relapse-free survival (HR = 2.66, 95%CI [1.67-4.22], P value < 0.001 (log-rank test)) than UICC tumor, node, metastasis (TNM) staging and microsatellite instability status are identified and validated using DNA methylation-specific PCR. The MethCORR approach is general, and may be similarly successful for other cancer types.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/mortality , Epigenome/genetics , Models, Genetic , Neoplasm Recurrence, Local/diagnosis , Aged , Colon/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Methylation , Datasets as Topic , Disease-Free Survival , Female , Formaldehyde , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Intestinal Mucosa/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Paraffin Embedding , Prognosis , Rectum/pathology , Risk Assessment/methods , Tissue Fixation
7.
Ann Rheum Dis ; 78(8): 1079-1089, 2019 08.
Article in English | MEDLINE | ID: mdl-31167757

ABSTRACT

OBJECTIVES: Systemic lupus erythematosus (SLE) diagnosis and treatment remain empirical and the molecular basis for its heterogeneity elusive. We explored the genomic basis for disease susceptibility and severity. METHODS: mRNA sequencing and genotyping in blood from 142 patients with SLE and 58 healthy volunteers. Abundances of cell types were assessed by CIBERSORT and cell-specific effects by interaction terms in linear models. Differentially expressed genes (DEGs) were used to train classifiers (linear discriminant analysis) of SLE versus healthy individuals in 80% of the dataset and were validated in the remaining 20% running 1000 iterations. Transcriptome/genotypes were integrated by expression-quantitative trail loci (eQTL) analysis; tissue-specific genetic causality was assessed by regulatory trait concordance (RTC). RESULTS: SLE has a 'susceptibility signature' present in patients in clinical remission, an 'activity signature' linked to genes that regulate immune cell metabolism, protein synthesis and proliferation, and a 'severity signature' best illustrated in active nephritis, enriched in druggable granulocyte and plasmablast/plasma-cell pathways. Patients with SLE have also perturbed mRNA splicing enriched in immune system and interferon signalling genes. A novel transcriptome index distinguished active versus inactive disease-but not low disease activity-and correlated with disease severity. DEGs discriminate SLE versus healthy individuals with median sensitivity 86% and specificity 92% suggesting a potential use in diagnostics. Combined eQTL analysis from the Genotype Tissue Expression (GTEx) project and SLE-associated genetic polymorphisms demonstrates that susceptibility variants may regulate gene expression in the blood but also in other tissues. CONCLUSION: Specific gene networks confer susceptibility to SLE, activity and severity, and may facilitate personalised care.


Subject(s)
Gene Expression Profiling/methods , Genetic Predisposition to Disease/epidemiology , Interferon Type I/genetics , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Adult , Case-Control Studies , Disease Progression , Female , Genetic Variation , Genome-Wide Association Study , Genotype , Humans , Male , Middle Aged , Phenotype , RNA, Messenger/genetics , Reference Values , Transcriptome/genetics , Young Adult
8.
Nat Commun ; 9(1): 3664, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30202008

ABSTRACT

Point mutations in cancer have been extensively studied but chromosomal gains and losses have been more challenging to interpret due to their unspecific nature. Here we examine high-resolution allelic imbalance (AI) landscape in 1699 colorectal cancers, 256 of which have been whole-genome sequenced (WGSed). The imbalances pinpoint 38 genes as plausible AI targets based on previous knowledge. Unbiased CRISPR-Cas9 knockout and activation screens identified in total 79 genes within AI peaks regulating cell growth. Genetic and functional data implicate loss of TP53 as a sufficient driver of AI. The WGS highlights an influence of copy number aberrations on the rate of detected somatic point mutations. Importantly, the data reveal several associations between AI target genes, suggesting a role for a network of lineage-determining transcription factors in colorectal tumorigenesis. Overall, the results unravel the contribution of AI in colorectal cancer and provide a plausible explanation why so few genes are commonly affected by point mutations in cancers.


Subject(s)
Allelic Imbalance , Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , CRISPR-Cas Systems , Chromosome Aberrations , Chromosomes, Human, Pair 8 , Colorectal Neoplasms/pathology , DNA Copy Number Variations , Denmark , Gene Expression Profiling , Genomics , Genotype , Humans , Loss of Heterozygosity , Microsatellite Repeats , Phenotype , Point Mutation , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Small Interfering/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Whole Genome Sequencing
9.
EMBO Mol Med ; 10(9)2018 09.
Article in English | MEDLINE | ID: mdl-30108113

ABSTRACT

Microsatellite instability (MSI) leads to accumulation of an excessive number of mutations in the genome, mostly small insertions and deletions. MSI colorectal cancers (CRCs), however, also contain more point mutations than microsatellite-stable (MSS) tumors, yet they have not been as comprehensively studied. To identify candidate driver genes affected by point mutations in MSI CRC, we ranked genes based on mutation significance while correcting for replication timing and gene expression utilizing an algorithm, MutSigCV Somatic point mutation data from the exome kit-targeted area from 24 exome-sequenced sporadic MSI CRCs and respective normals, and 12 whole-genome-sequenced sporadic MSI CRCs and respective normals were utilized. The top 73 genes were validated in 93 additional MSI CRCs. The MutSigCV ranking identified several well-established MSI CRC driver genes and provided additional evidence for previously proposed CRC candidate genes as well as shortlisted genes that have to our knowledge not been linked to CRC before. Two genes, SMARCB1 and STK38L, were also functionally scrutinized, providing evidence of a tumorigenic role, for SMARCB1 mutations in particular.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Microsatellite Instability , Point Mutation , Gene Regulatory Networks , Humans , Molecular Sequence Annotation , Sequence Analysis, DNA
10.
Nat Genet ; 50(7): 956-967, 2018 07.
Article in English | MEDLINE | ID: mdl-29955180

ABSTRACT

We apply integrative approaches to expression quantitative loci (eQTLs) from 44 tissues from the Genotype-Tissue Expression project and genome-wide association study data. About 60% of known trait-associated loci are in linkage disequilibrium with a cis-eQTL, over half of which were not found in previous large-scale whole blood studies. Applying polygenic analyses to metabolic, cardiovascular, anthropometric, autoimmune, and neurodegenerative traits, we find that eQTLs are significantly enriched for trait associations in relevant pathogenic tissues and explain a substantial proportion of the heritability (40-80%). For most traits, tissue-shared eQTLs underlie a greater proportion of trait associations, although tissue-specific eQTLs have a greater contribution to some traits, such as blood pressure. By integrating information from biological pathways with eQTL target genes and applying a gene-based approach, we validate previously implicated causal genes and pathways, and propose new variant and gene associations for several complex traits, which we replicate in the UK BioBank and BioVU.


Subject(s)
Disease/genetics , Gene Expression Regulation , Gene Expression , Gene Expression Profiling/methods , Genome-Wide Association Study/methods , Genotype , Humans , Phenotype , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Quantitative Trait, Heritable
11.
Gut ; 67(3): 521-533, 2018 03.
Article in English | MEDLINE | ID: mdl-28634199

ABSTRACT

OBJECTIVE: To elucidate the genetic architecture of gene expression in pancreatic tissues. DESIGN: We performed expression quantitative trait locus (eQTL) analysis in histologically normal pancreatic tissue samples (n=95) using RNA sequencing and the corresponding 1000 genomes imputed germline genotypes. Data from pancreatic tumour-derived tissue samples (n=115) from The Cancer Genome Atlas were included for comparison. RESULTS: We identified 38 615 cis-eQTLs (in 484 genes) in histologically normal tissues and 39 713 cis-eQTL (in 237 genes) in tumour-derived tissues (false discovery rate <0.1), with the strongest effects seen near transcriptional start sites. Approximately 23% and 42% of genes with significant cis-eQTLs appeared to be specific for tumour-derived and normal-derived tissues, respectively. Significant enrichment of cis-eQTL variants was noted in non-coding regulatory regions, in particular for pancreatic tissues (1.53-fold to 3.12-fold, p≤0.0001), indicating tissue-specific functional relevance. A common pancreatic cancer risk locus on 9q34.2 (rs687289) was associated with ABO expression in histologically normal (p=5.8×10-8) and tumour-derived (p=8.3×10-5) tissues. The high linkage disequilibrium between this variant and the O blood group generating deletion variant in ABO (exon 6) suggested that nonsense-mediated decay (NMD) of the 'O' mRNA might explain this finding. However, knockdown of crucial NMD regulators did not influence decay of the ABO 'O' mRNA, indicating that a gene regulatory element influenced by pancreatic cancer risk alleles may underlie the eQTL. CONCLUSIONS: We have identified cis-eQTLs representing potential functional regulatory variants in the pancreas and generated a rich data set for further studies on gene expression and its regulation in pancreatic tissues.


Subject(s)
ABO Blood-Group System/genetics , Gene Expression , Pancreas , Pancreatic Neoplasms/genetics , Quantitative Trait Loci , RNA, Neoplasm/analysis , Transcriptome , Alleles , Chromosomes, Human, Pair 9 , Genome-Wide Association Study , Genotype , Humans , Nonsense Mediated mRNA Decay , Polymorphism, Single Nucleotide , Regulatory Sequences, Nucleic Acid , Sequence Analysis, RNA
12.
Nat Genet ; 49(12): 1676-1683, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29058715

ABSTRACT

How to interpret the biological causes underlying the predisposing markers identified through genome-wide association studies (GWAS) remains an open question. One direct and powerful way to assess the genetic causality behind GWAS is through analysis of expression quantitative trait loci (eQTLs). Here we describe a new approach to estimate the tissues behind the genetic causality of a variety of GWAS traits, using the cis-eQTLs in 44 tissues from the Genotype-Tissue Expression (GTEx) Consortium. We have adapted the regulatory trait concordance (RTC) score to measure the probability of eQTLs being active in multiple tissues and to calculate the probability that a GWAS-associated variant and an eQTL tag the same functional effect. By normalizing the GWAS-eQTL probabilities by the tissue-sharing estimates for eQTLs, we generate relative tissue-causality profiles for GWAS traits. Our approach not only implicates the gene likely mediating individual GWAS signals, but also highlights tissues where the genetic causality for an individual trait is likely manifested.


Subject(s)
Gene Expression Profiling , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Quantitative Trait Loci/genetics , Genetic Association Studies , Genotype , Humans , Organ Specificity/genetics , Phenotype , Polymorphism, Single Nucleotide
13.
Cell Rep ; 19(6): 1268-1280, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28494874

ABSTRACT

Colorectal cancer (CRC) is characterized by major inter-tumor diversity that complicates the prediction of disease and treatment outcomes. Recent efforts help resolve this by sub-classification of CRC into natural molecular subtypes; however, this strategy is not yet able to provide clinicians with improved tools for decision making. We here present an extended framework for CRC stratification that specifically aims to improve patient prognostication. Using transcriptional profiles from 1,100 CRCs, including >300 previously unpublished samples, we identify cancer cell and tumor archetypes and suggest the tumor microenvironment as a major prognostic determinant that can be influenced by the microbiome. Notably, our subtyping strategy allowed identification of archetype-specific prognostic biomarkers that provided information beyond and independent of UICC-TNM staging, MSI status, and consensus molecular subtyping. The results illustrate that our extended subtyping framework, combining subtyping and subtype-specific biomarkers, could contribute to improved patient prognostication and may form a strong basis for future studies.


Subject(s)
Biomarkers, Tumor/classification , Colorectal Neoplasms/genetics , Transcriptome , Biomarkers, Tumor/genetics , Case-Control Studies , Colorectal Neoplasms/classification , Colorectal Neoplasms/pathology , Humans , Microbiota , Tumor Microenvironment
14.
Nat Commun ; 8: 15452, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28516912

ABSTRACT

Population scale studies combining genetic information with molecular phenotypes (for example, gene expression) have become a standard to dissect the effects of genetic variants onto organismal phenotypes. These kinds of data sets require powerful, fast and versatile methods able to discover molecular Quantitative Trait Loci (molQTL). Here we propose such a solution, QTLtools, a modular framework that contains multiple new and well-established methods to prepare the data, to discover proximal and distal molQTLs and, finally, to integrate them with GWAS variants and functional annotations of the genome. We demonstrate its utility by performing a complete expression QTL study in a few easy-to-perform steps. QTLtools is open source and available at https://qtltools.github.io/qtltools/.


Subject(s)
Algorithms , Chromosome Mapping/methods , Genome, Human , High-Throughput Nucleotide Sequencing/statistics & numerical data , Quantitative Trait Loci , Genome-Wide Association Study , Genotype , Humans , Phenotype , Polymorphism, Single Nucleotide , Quality Control
16.
Nat Commun ; 8: 14418, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28195176

ABSTRACT

Genome-wide association studies have identified a great number of non-coding risk variants for colorectal cancer (CRC). To date, the majority of these variants have not been functionally studied. Identification of allele-specific transcription factor (TF) binding is of great importance to understand regulatory consequences of such variants. A recently developed proteome-wide analysis of disease-associated SNPs (PWAS) enables identification of TF-DNA interactions in an unbiased manner. Here we perform a large-scale PWAS study to comprehensively characterize TF-binding landscape that is associated with CRC, which identifies 731 allele-specific TF binding at 116 CRC risk loci. This screen identifies the A-allele of rs1800734 within the promoter region of MLH1 as perturbing the binding of TFAP4 and consequently increasing DCLK3 expression through a long-range interaction, which promotes cancer malignancy through enhancing expression of the genes related to epithelial-to-mesenchymal transition.


Subject(s)
Colonic Neoplasms/genetics , Colorectal Neoplasms/genetics , Disease Progression , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Alleles , CRISPR-Cas Systems , Cell Line, Tumor , Colorectal Neoplasms/metabolism , DNA Methylation , DNA-Binding Proteins , Doublecortin-Like Kinases , Epigenesis, Genetic , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , MutL Protein Homolog 1/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Proteome , Proteomics , Transcription Factors
17.
Nat Genet ; 49(1): 17-26, 2017 01.
Article in English | MEDLINE | ID: mdl-27841877

ABSTRACT

Insulin resistance is a key mediator of obesity-related cardiometabolic disease, yet the mechanisms underlying this link remain obscure. Using an integrative genomic approach, we identify 53 genomic regions associated with insulin resistance phenotypes (higher fasting insulin levels adjusted for BMI, lower HDL cholesterol levels and higher triglyceride levels) and provide evidence that their link with higher cardiometabolic risk is underpinned by an association with lower adipose mass in peripheral compartments. Using these 53 loci, we show a polygenic contribution to familial partial lipodystrophy type 1, a severe form of insulin resistance, and highlight shared molecular mechanisms in common/mild and rare/severe insulin resistance. Population-level genetic analyses combined with experiments in cellular models implicate CCDC92, DNAH10 and L3MBTL3 as previously unrecognized molecules influencing adipocyte differentiation. Our findings support the notion that limited storage capacity of peripheral adipose tissue is an important etiological component in insulin-resistant cardiometabolic disease and highlight genes and mechanisms underpinning this link.


Subject(s)
Adipose Tissue/pathology , Cardiovascular Diseases/physiopathology , Genomics/methods , Insulin Resistance/genetics , Metabolic Diseases/physiopathology , Obesity/complications , Adipose Tissue/metabolism , Animals , Blood Glucose/analysis , Body Mass Index , Case-Control Studies , Disease Models, Animal , Female , Genome-Wide Association Study , Humans , Male , Mice , Obesity/genetics , Phenotype
18.
Mol Oncol ; 10(8): 1266-82, 2016 10.
Article in English | MEDLINE | ID: mdl-27396952

ABSTRACT

It is well established that lncRNAs are aberrantly expressed in cancer where they have been shown to act as oncogenes or tumor suppressors. RNA profiling of 314 colorectal adenomas/adenocarcinomas and 292 adjacent normal colon mucosa samples using RNA-sequencing demonstrated that the snoRNA host gene 16 (SNHG16) is significantly up-regulated in adenomas and all stages of CRC. SNHG16 expression was positively correlated to the expression of Wnt-regulated transcription factors, including ASCL2, ETS2, and c-Myc. In vitro abrogation of Wnt signaling in CRC cells reduced the expression of SNHG16 indicating that SNHG16 is regulated by the Wnt pathway. Silencing of SNHG16 resulted in reduced viability, increased apoptotic cell death and impaired cell migration. The SNHG16 silencing particularly affected expression of genes involved in lipid metabolism. A connection between SNHG16 and genes involved in lipid metabolism was also observed in clinical tumors. Argonaute CrossLinking and ImmunoPrecipitation (AGO-CLIP) demonstrated that SNHG16 heavily binds AGO and has 27 AGO/miRNA target sites along its length, indicating that SNHG16 may act as a competing endogenous RNA (ceRNA) "sponging" miRNAs off their cognate targets. Most interestingly, half of the miRNA families with high confidence targets on SNHG16 also target the 3'UTR of Stearoyl-CoA Desaturase (SCD). SCD is involved in lipid metabolism and is down-regulated upon SNHG16 silencing. In conclusion, up-regulation of SNHG16 is a frequent event in CRC, likely caused by deregulated Wnt signaling. In vitro analyses demonstrate that SNHG16 may play an oncogenic role in CRC and that it affects genes involved in lipid metabolism, possible through ceRNA related mechanisms.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Lipid Metabolism/genetics , RNA, Long Noncoding/metabolism , Wnt Signaling Pathway/genetics , Apoptosis/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Colorectal Neoplasms/pathology , Cytoplasm/metabolism , Gene Knockdown Techniques , HCT116 Cells , Humans , Nucleotide Motifs/genetics , Polyribosomes/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Long Noncoding/genetics , RNA, Small Nucleolar/metabolism , Transcription Factors/metabolism , Up-Regulation/genetics
19.
Bioinformatics ; 32(10): 1479-85, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26708335

ABSTRACT

MOTIVATION: In order to discover quantitative trait loci, multi-dimensional genomic datasets combining DNA-seq and ChiP-/RNA-seq require methods that rapidly correlate tens of thousands of molecular phenotypes with millions of genetic variants while appropriately controlling for multiple testing. RESULTS: We have developed FastQTL, a method that implements a popular cis-QTL mapping strategy in a user- and cluster-friendly tool. FastQTL also proposes an efficient permutation procedure to control for multiple testing. The outcome of permutations is modeled using beta distributions trained from a few permutations and from which adjusted P-values can be estimated at any level of significance with little computational cost. The Geuvadis & GTEx pilot datasets can be now easily analyzed an order of magnitude faster than previous approaches. AVAILABILITY AND IMPLEMENTATION: Source code, binaries and comprehensive documentation of FastQTL are freely available to download at http://fastqtl.sourceforge.net/ CONTACT: emmanouil.dermitzakis@unige.ch or olivier.delaneau@unige.ch SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Quantitative Trait Loci , Genomics , Phenotype , Software , Statistical Distributions
20.
Am J Hum Genet ; 97(4): 567-75, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26430802

ABSTRACT

With the advent of RNA-sequencing technology, we can detect different types of alternative splicing and determine how DNA variation regulates splicing. However, given the short read lengths used in most population-based RNA-sequencing experiments, quantifying transcripts accurately remains a challenge. Here we present a method, Altrans, for discovery of alternative splicing quantitative trait loci (asQTLs). To assess the performance of Altrans, we compared it to Cufflinks and MISO in simulations and Cufflinks for asQTL discovery. Simulations show that in the presence of unannotated transcripts, Altrans performs better in quantifications than Cufflinks and MISO. We have applied Altrans and Cufflinks to the Geuvadis dataset, which comprises samples from European and African populations, and discovered (FDR = 1%) 1,427 and 166 asQTLs with Altrans and 1,737 and 304 asQTLs with Cufflinks for Europeans and Africans, respectively. We show that, by discovering a set of asQTLs in a smaller subset of European samples and replicating these in the remaining larger subset of Europeans, both methods achieve similar replication levels (95% for both methods). We find many Altrans-specific asQTLs, which replicate to a high degree (93%). This is mainly due to junctions absent from the annotations and hence not tested with Cufflinks. The asQTLs are significantly enriched for biochemically active regions of the genome, functional marks, and variants in splicing regions, highlighting their biological relevance. We present an approach for discovering asQTLs that is a more direct assessment of splicing compared to other methods and is complementary to other transcript quantification methods.


Subject(s)
Alternative Splicing , Black People/genetics , Gene Expression Profiling , High-Throughput Nucleotide Sequencing/methods , Quantitative Trait Loci , Sequence Analysis, RNA/methods , White People/genetics , Algorithms , Cohort Studies , Genome, Human , Humans , Protein Isoforms , Software
SELECTION OF CITATIONS
SEARCH DETAIL
...