Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Publication year range
1.
Cell ; 186(18): 3945-3967.e26, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37582358

ABSTRACT

Post-translational modifications (PTMs) play key roles in regulating cell signaling and physiology in both normal and cancer cells. Advances in mass spectrometry enable high-throughput, accurate, and sensitive measurement of PTM levels to better understand their role, prevalence, and crosstalk. Here, we analyze the largest collection of proteogenomics data from 1,110 patients with PTM profiles across 11 cancer types (10 from the National Cancer Institute's Clinical Proteomic Tumor Analysis Consortium [CPTAC]). Our study reveals pan-cancer patterns of changes in protein acetylation and phosphorylation involved in hallmark cancer processes. These patterns revealed subsets of tumors, from different cancer types, including those with dysregulated DNA repair driven by phosphorylation, altered metabolic regulation associated with immune response driven by acetylation, affected kinase specificity by crosstalk between acetylation and phosphorylation, and modified histone regulation. Overall, this resource highlights the rich biology governed by PTMs and exposes potential new therapeutic avenues.


Subject(s)
Neoplasms , Protein Processing, Post-Translational , Proteomics , Humans , Acetylation , Histones/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Phosphorylation , Proteomics/methods
2.
Cell ; 186(18): 3921-3944.e25, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37582357

ABSTRACT

Cancer driver events refer to key genetic aberrations that drive oncogenesis; however, their exact molecular mechanisms remain insufficiently understood. Here, our multi-omics pan-cancer analysis uncovers insights into the impacts of cancer drivers by identifying their significant cis-effects and distal trans-effects quantified at the RNA, protein, and phosphoprotein levels. Salient observations include the association of point mutations and copy-number alterations with the rewiring of protein interaction networks, and notably, most cancer genes converge toward similar molecular states denoted by sequence-based kinase activity profiles. A correlation between predicted neoantigen burden and measured T cell infiltration suggests potential vulnerabilities for immunotherapies. Patterns of cancer hallmarks vary by polygenic protein abundance ranging from uniform to heterogeneous. Overall, our work demonstrates the value of comprehensive proteogenomics in understanding the functional states of oncogenic drivers and their links to cancer development, surpassing the limitations of studying individual cancer types.


Subject(s)
Neoplasms , Proteogenomics , Humans , Neoplasms/genetics , Oncogenes , Cell Transformation, Neoplastic/genetics , DNA Copy Number Variations
3.
Cell ; 186(7): 1493-1511.e40, 2023 03 30.
Article in English | MEDLINE | ID: mdl-37001506

ABSTRACT

Understanding how genetic variants impact molecular phenotypes is a key goal of functional genomics, currently hindered by reliance on a single haploid reference genome. Here, we present the EN-TEx resource of 1,635 open-access datasets from four donors (∼30 tissues × âˆ¼15 assays). The datasets are mapped to matched, diploid genomes with long-read phasing and structural variants, instantiating a catalog of >1 million allele-specific loci. These loci exhibit coordinated activity along haplotypes and are less conserved than corresponding, non-allele-specific ones. Surprisingly, a deep-learning transformer model can predict the allele-specific activity based only on local nucleotide-sequence context, highlighting the importance of transcription-factor-binding motifs particularly sensitive to variants. Furthermore, combining EN-TEx with existing genome annotations reveals strong associations between allele-specific and GWAS loci. It also enables models for transferring known eQTLs to difficult-to-profile tissues (e.g., from skin to heart). Overall, EN-TEx provides rich data and generalizable models for more accurate personal functional genomics.


Subject(s)
Epigenome , Quantitative Trait Loci , Genome-Wide Association Study , Genomics , Phenotype , Polymorphism, Single Nucleotide
4.
Cell ; 184(10): 2633-2648.e19, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33864768

ABSTRACT

Long non-coding RNA (lncRNA) genes have well-established and important impacts on molecular and cellular functions. However, among the thousands of lncRNA genes, it is still a major challenge to identify the subset with disease or trait relevance. To systematically characterize these lncRNA genes, we used Genotype Tissue Expression (GTEx) project v8 genetic and multi-tissue transcriptomic data to profile the expression, genetic regulation, cellular contexts, and trait associations of 14,100 lncRNA genes across 49 tissues for 101 distinct complex genetic traits. Using these approaches, we identified 1,432 lncRNA gene-trait associations, 800 of which were not explained by stronger effects of neighboring protein-coding genes. This included associations between lncRNA quantitative trait loci and inflammatory bowel disease, type 1 and type 2 diabetes, and coronary artery disease, as well as rare variant associations to body mass index.


Subject(s)
Disease/genetics , Multifactorial Inheritance/genetics , Population/genetics , RNA, Long Noncoding/genetics , Transcriptome , Coronary Artery Disease/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/genetics , Gene Expression Profiling , Genetic Variation , Humans , Inflammatory Bowel Diseases/genetics , Organ Specificity/genetics , Quantitative Trait Loci
5.
Nature ; 625(7996): 735-742, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38030727

ABSTRACT

Noncoding DNA is central to our understanding of human gene regulation and complex diseases1,2, and measuring the evolutionary sequence constraint can establish the functional relevance of putative regulatory elements in the human genome3-9. Identifying the genomic elements that have become constrained specifically in primates has been hampered by the faster evolution of noncoding DNA compared to protein-coding DNA10, the relatively short timescales separating primate species11, and the previously limited availability of whole-genome sequences12. Here we construct a whole-genome alignment of 239 species, representing nearly half of all extant species in the primate order. Using this resource, we identified human regulatory elements that are under selective constraint across primates and other mammals at a 5% false discovery rate. We detected 111,318 DNase I hypersensitivity sites and 267,410 transcription factor binding sites that are constrained specifically in primates but not across other placental mammals and validate their cis-regulatory effects on gene expression. These regulatory elements are enriched for human genetic variants that affect gene expression and complex traits and diseases. Our results highlight the important role of recent evolution in regulatory sequence elements differentiating primates, including humans, from other placental mammals.


Subject(s)
Conserved Sequence , Evolution, Molecular , Genome , Primates , Animals , Female , Humans , Pregnancy , Conserved Sequence/genetics , Deoxyribonuclease I/metabolism , DNA/genetics , DNA/metabolism , Genome/genetics , Mammals/classification , Mammals/genetics , Placenta , Primates/classification , Primates/genetics , Regulatory Sequences, Nucleic Acid/genetics , Reproducibility of Results , Transcription Factors/metabolism , Proteins/genetics , Gene Expression Regulation/genetics
6.
Nature ; 608(7923): 569-577, 2022 08.
Article in English | MEDLINE | ID: mdl-35922514

ABSTRACT

A major challenge in human genetics is to identify the molecular mechanisms of trait-associated and disease-associated variants. To achieve this, quantitative trait locus (QTL) mapping of genetic variants with intermediate molecular phenotypes such as gene expression and splicing have been widely adopted1,2. However, despite successes, the molecular basis for a considerable fraction of trait-associated and disease-associated variants remains unclear3,4. Here we show that ADAR-mediated adenosine-to-inosine RNA editing, a post-transcriptional event vital for suppressing cellular double-stranded RNA (dsRNA)-mediated innate immune interferon responses5-11, is an important potential mechanism underlying genetic variants associated with common inflammatory diseases. We identified and characterized 30,319 cis-RNA editing QTLs (edQTLs) across 49 human tissues. These edQTLs were significantly enriched in genome-wide association study signals for autoimmune and immune-mediated diseases. Colocalization analysis of edQTLs with disease risk loci further pinpointed key, putatively immunogenic dsRNAs formed by expected inverted repeat Alu elements as well as unexpected, highly over-represented cis-natural antisense transcripts. Furthermore, inflammatory disease risk variants, in aggregate, were associated with reduced editing of nearby dsRNAs and induced interferon responses in inflammatory diseases. This unique directional effect agrees with the established mechanism that lack of RNA editing by ADAR1 leads to the specific activation of the dsRNA sensor MDA5 and subsequent interferon responses and inflammation7-9. Our findings implicate cellular dsRNA editing and sensing as a previously underappreciated mechanism of common inflammatory diseases.


Subject(s)
Adenosine Deaminase , Genetic Predisposition to Disease , Immune System Diseases , Inflammation , RNA Editing , RNA, Double-Stranded , Adenosine/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Alu Elements/genetics , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Genome-Wide Association Study , Humans , Immune System Diseases/genetics , Immune System Diseases/immunology , Immune System Diseases/pathology , Immunity, Innate , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inosine/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , Interferons/genetics , Interferons/immunology , Quantitative Trait Loci/genetics , RNA Editing/genetics , RNA, Double-Stranded/genetics , RNA-Binding Proteins/metabolism
7.
Nature ; 608(7922): 353-359, 2022 08.
Article in English | MEDLINE | ID: mdl-35922509

ABSTRACT

Regulation of transcript structure generates transcript diversity and plays an important role in human disease1-7. The advent of long-read sequencing technologies offers the opportunity to study the role of genetic variation in transcript structure8-16. In this Article, we present a large human long-read RNA-seq dataset using the Oxford Nanopore Technologies platform from 88 samples from Genotype-Tissue Expression (GTEx) tissues and cell lines, complementing the GTEx resource. We identified just over 70,000 novel transcripts for annotated genes, and validated the protein expression of 10% of novel transcripts. We developed a new computational package, LORALS, to analyse the genetic effects of rare and common variants on the transcriptome by allele-specific analysis of long reads. We characterized allele-specific expression and transcript structure events, providing new insights into the specific transcript alterations caused by common and rare genetic variants and highlighting the resolution gained from long-read data. We were able to perturb the transcript structure upon knockdown of PTBP1, an RNA binding protein that mediates splicing, thereby finding genetic regulatory effects that are modified by the cellular environment. Finally, we used this dataset to enhance variant interpretation and study rare variants leading to aberrant splicing patterns.


Subject(s)
Alleles , Gene Expression Profiling , Organ Specificity , RNA-Seq , Transcriptome , Alternative Splicing/genetics , Cell Line , Datasets as Topic , Genotype , Heterogeneous-Nuclear Ribonucleoproteins/deficiency , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Organ Specificity/genetics , Polypyrimidine Tract-Binding Protein/deficiency , Polypyrimidine Tract-Binding Protein/genetics , Reproducibility of Results , Transcriptome/genetics
8.
Am J Hum Genet ; 111(1): 133-149, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38181730

ABSTRACT

Bulk-tissue molecular quantitative trait loci (QTLs) have been the starting point for interpreting disease-associated variants, and context-specific QTLs show particular relevance for disease. Here, we present the results of mapping interaction QTLs (iQTLs) for cell type, age, and other phenotypic variables in multi-omic, longitudinal data from the blood of individuals of diverse ancestries. By modeling the interaction between genotype and estimated cell-type proportions, we demonstrate that cell-type iQTLs could be considered as proxies for cell-type-specific QTL effects, particularly for the most abundant cell type in the tissue. The interpretation of age iQTLs, however, warrants caution because the moderation effect of age on the genotype and molecular phenotype association could be mediated by changes in cell-type composition. Finally, we show that cell-type iQTLs contribute to cell-type-specific enrichment of diseases that, in combination with additional functional data, could guide future functional studies. Overall, this study highlights the use of iQTLs to gain insights into the context specificity of regulatory effects.


Subject(s)
Gene Expression Regulation , Quantitative Trait Loci , Humans , Quantitative Trait Loci/genetics , Genotype , Phenotype
9.
Cell ; 150(3): 495-507, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22863004

ABSTRACT

Coated pits assemble by growth of a clathrin lattice, which is linked by adaptors to the underlying membrane. How does this process start? We used live-cell TIRF imaging with single-molecule EGFP sensitivity and high temporal resolution to detect arrival of the clathrin triskelions and AP2 adaptors that initiate coat assembly. Unbiased object identification and trajectory tracking, together with a statistical model, yield the arrival times and numbers of individual proteins, as well as experimentally confirmed estimates of the extent of substitution of endogenous by expressed, fluorescently tagged proteins. Pits initiate by coordinated arrival of clathrin and AP2, which is usually detected as two sequential steps, each of one triskelion with two adaptors. PI-4,5-P2 is essential for initiation. The accessory proteins FCHo1/2 are not; instead, they are required for sustained growth. This objective picture of coated pit initiation also shows that methods outlined here will be broadly useful for studies of dynamic assemblies in living cells.


Subject(s)
Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , Membrane Proteins/metabolism , Adaptor Protein Complex 2/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Proteins/metabolism
10.
Nature ; 590(7845): 290-299, 2021 02.
Article in English | MEDLINE | ID: mdl-33568819

ABSTRACT

The Trans-Omics for Precision Medicine (TOPMed) programme seeks to elucidate the genetic architecture and biology of heart, lung, blood and sleep disorders, with the ultimate goal of improving diagnosis, treatment and prevention of these diseases. The initial phases of the programme focused on whole-genome sequencing of individuals with rich phenotypic data and diverse backgrounds. Here we describe the TOPMed goals and design as well as the available resources and early insights obtained from the sequence data. The resources include a variant browser, a genotype imputation server, and genomic and phenotypic data that are available through dbGaP (Database of Genotypes and Phenotypes)1. In the first 53,831 TOPMed samples, we detected more than 400 million single-nucleotide and insertion or deletion variants after alignment with the reference genome. Additional previously undescribed variants were detected through assembly of unmapped reads and customized analysis in highly variable loci. Among the more than 400 million detected variants, 97% have frequencies of less than 1% and 46% are singletons that are present in only one individual (53% among unrelated individuals). These rare variants provide insights into mutational processes and recent human evolutionary history. The extensive catalogue of genetic variation in TOPMed studies provides unique opportunities for exploring the contributions of rare and noncoding sequence variants to phenotypic variation. Furthermore, combining TOPMed haplotypes with modern imputation methods improves the power and reach of genome-wide association studies to include variants down to a frequency of approximately 0.01%.


Subject(s)
Genetic Variation/genetics , Genome, Human/genetics , Genomics , National Heart, Lung, and Blood Institute (U.S.) , Precision Medicine , Cytochrome P-450 CYP2D6/genetics , Haplotypes/genetics , Heterozygote , Humans , INDEL Mutation , Loss of Function Mutation , Mutagenesis , Phenotype , Polymorphism, Single Nucleotide , Population Density , Precision Medicine/standards , Quality Control , Sample Size , United States , Whole Genome Sequencing/standards
11.
Nature ; 586(7831): 763-768, 2020 10.
Article in English | MEDLINE | ID: mdl-33057201

ABSTRACT

Age is the dominant risk factor for most chronic human diseases, but the mechanisms through which ageing confers this risk are largely unknown1. The age-related acquisition of somatic mutations that lead to clonal expansion in regenerating haematopoietic stem cell populations has recently been associated with both haematological cancer2-4 and coronary heart disease5-this phenomenon is termed clonal haematopoiesis of indeterminate potential (CHIP)6. Simultaneous analyses of germline and somatic whole-genome sequences provide the opportunity to identify root causes of CHIP. Here we analyse high-coverage whole-genome sequences from 97,691 participants of diverse ancestries in the National Heart, Lung, and Blood Institute Trans-omics for Precision Medicine (TOPMed) programme, and identify 4,229 individuals with CHIP. We identify associations with blood cell, lipid and inflammatory traits that are specific to different CHIP driver genes. Association of a genome-wide set of germline genetic variants enabled the identification of three genetic loci associated with CHIP status, including one locus at TET2 that was specific to individuals of African ancestry. In silico-informed in vitro evaluation of the TET2 germline locus enabled the identification of a causal variant that disrupts a TET2 distal enhancer, resulting in increased self-renewal of haematopoietic stem cells. Overall, we observe that germline genetic variation shapes haematopoietic stem cell function, leading to CHIP through mechanisms that are specific to clonal haematopoiesis as well as shared mechanisms that lead to somatic mutations across tissues.


Subject(s)
Clonal Hematopoiesis/genetics , Genetic Predisposition to Disease , Genome, Human/genetics , Whole Genome Sequencing , Adult , Africa/ethnology , Aged , Aged, 80 and over , Black People/genetics , Cell Self Renewal/genetics , DNA-Binding Proteins/genetics , Dioxygenases , Female , Germ-Line Mutation/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Middle Aged , National Heart, Lung, and Blood Institute (U.S.) , Phenotype , Precision Medicine , Proto-Oncogene Proteins/genetics , Tripartite Motif Proteins/genetics , United States , alpha Karyopherins/genetics
12.
PLoS Genet ; 19(5): e1010517, 2023 05.
Article in English | MEDLINE | ID: mdl-37216410

ABSTRACT

Integrative approaches that simultaneously model multi-omics data have gained increasing popularity because they provide holistic system biology views of multiple or all components in a biological system of interest. Canonical correlation analysis (CCA) is a correlation-based integrative method designed to extract latent features shared between multiple assays by finding the linear combinations of features-referred to as canonical variables (CVs)-within each assay that achieve maximal across-assay correlation. Although widely acknowledged as a powerful approach for multi-omics data, CCA has not been systematically applied to multi-omics data in large cohort studies, which has only recently become available. Here, we adapted sparse multiple CCA (SMCCA), a widely-used derivative of CCA, to proteomics and methylomics data from the Multi-Ethnic Study of Atherosclerosis (MESA) and Jackson Heart Study (JHS). To tackle challenges encountered when applying SMCCA to MESA and JHS, our adaptations include the incorporation of the Gram-Schmidt (GS) algorithm with SMCCA to improve orthogonality among CVs, and the development of Sparse Supervised Multiple CCA (SSMCCA) to allow supervised integration analysis for more than two assays. Effective application of SMCCA to the two real datasets reveals important findings. Applying our SMCCA-GS to MESA and JHS, we identified strong associations between blood cell counts and protein abundance, suggesting that adjustment of blood cell composition should be considered in protein-based association studies. Importantly, CVs obtained from two independent cohorts also demonstrate transferability across the cohorts. For example, proteomic CVs learned from JHS, when transferred to MESA, explain similar amounts of blood cell count phenotypic variance in MESA, explaining 39.0% ~ 50.0% variation in JHS and 38.9% ~ 49.1% in MESA. Similar transferability was observed for other omics-CV-trait pairs. This suggests that biologically meaningful and cohort-agnostic variation is captured by CVs. We anticipate that applying our SMCCA-GS and SSMCCA on various cohorts would help identify cohort-agnostic biologically meaningful relationships between multi-omics data and phenotypic traits.


Subject(s)
Canonical Correlation Analysis , Proteomics , Humans , Proteomics/methods , Multiomics , Cohort Studies
13.
Am J Hum Genet ; 109(7): 1286-1297, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35716666

ABSTRACT

Despite the growing number of genome-wide association studies (GWASs), it remains unclear to what extent gene-by-gene and gene-by-environment interactions influence complex traits in humans. The magnitude of genetic interactions in complex traits has been difficult to quantify because GWASs are generally underpowered to detect individual interactions of small effect. Here, we develop a method to test for genetic interactions that aggregates information across all trait-associated loci. Specifically, we test whether SNPs in regions of European ancestry shared between European American and admixed African American individuals have the same causal effect sizes. We hypothesize that in African Americans, the presence of genetic interactions will drive the causal effect sizes of SNPs in regions of European ancestry to be more similar to those of SNPs in regions of African ancestry. We apply our method to two traits: gene expression in 296 African Americans and 482 European Americans in the Multi-Ethnic Study of Atherosclerosis (MESA) and low-density lipoprotein cholesterol (LDL-C) in 74K African Americans and 296K European Americans in the Million Veteran Program (MVP). We find significant evidence for genetic interactions in our analysis of gene expression; for LDL-C, we observe a similar point estimate, although this is not significant, most likely due to lower statistical power. These results suggest that gene-by-gene or gene-by-environment interactions modify the effect sizes of causal variants in human complex traits.


Subject(s)
Genome-Wide Association Study , Multifactorial Inheritance , Cholesterol, LDL , Gene Expression , Humans , Multifactorial Inheritance/genetics , Polymorphism, Single Nucleotide/genetics , White People/genetics
14.
Nature ; 569(7757): 503-508, 2019 05.
Article in English | MEDLINE | ID: mdl-31068700

ABSTRACT

Large panels of comprehensively characterized human cancer models, including the Cancer Cell Line Encyclopedia (CCLE), have provided a rigorous framework with which to study genetic variants, candidate targets, and small-molecule and biological therapeutics and to identify new marker-driven cancer dependencies. To improve our understanding of the molecular features that contribute to cancer phenotypes, including drug responses, here we have expanded the characterizations of cancer cell lines to include genetic, RNA splicing, DNA methylation, histone H3 modification, microRNA expression and reverse-phase protein array data for 1,072 cell lines from individuals of various lineages and ethnicities. Integration of these data with functional characterizations such as drug-sensitivity, short hairpin RNA knockdown and CRISPR-Cas9 knockout data reveals potential targets for cancer drugs and associated biomarkers. Together, this dataset and an accompanying public data portal provide a resource for the acceleration of cancer research using model cancer cell lines.


Subject(s)
Cell Line, Tumor , Neoplasms/genetics , Neoplasms/pathology , Antineoplastic Agents/pharmacology , Biomarkers, Tumor , DNA Methylation , Drug Resistance, Neoplasm , Ethnicity/genetics , Gene Editing , Histones/metabolism , Humans , MicroRNAs/genetics , Molecular Targeted Therapy , Neoplasms/metabolism , Protein Array Analysis , RNA Splicing
15.
PLoS Genet ; 18(1): e1009719, 2022 01.
Article in English | MEDLINE | ID: mdl-35100260

ABSTRACT

Tens of thousands of genetic variants associated with gene expression (cis-eQTLs) have been discovered in the human population. These eQTLs are active in various tissues and contexts, but the molecular mechanisms of eQTL variability are poorly understood, hindering our understanding of genetic regulation across biological contexts. Since many eQTLs are believed to act by altering transcription factor (TF) binding affinity, we hypothesized that analyzing eQTL effect size as a function of TF level may allow discovery of mechanisms of eQTL variability. Using GTEx Consortium eQTL data from 49 tissues, we analyzed the interaction between eQTL effect size and TF level across tissues and across individuals within specific tissues and generated a list of 10,098 TF-eQTL interactions across 2,136 genes that are supported by at least two lines of evidence. These TF-eQTLs were enriched for various TF binding measures, supporting with orthogonal evidence that these eQTLs are regulated by the implicated TFs. We also found that our TF-eQTLs tend to overlap genes with gene-by-environment regulatory effects and to colocalize with GWAS loci, implying that our approach can help to elucidate mechanisms of context-specificity and trait associations. Finally, we highlight an interesting example of IKZF1 TF regulation of an APBB1IP gene eQTL that colocalizes with a GWAS signal for blood cell traits. Together, our findings provide candidate TF mechanisms for a large number of eQTLs and offer a generalizable approach for researchers to discover TF regulators of genetic variant effects in additional QTL datasets.


Subject(s)
Quantitative Trait Loci , Transcription Factors/physiology , Alleles , Binding Sites , Gene Knockdown Techniques , Gene-Environment Interaction , Genome-Wide Association Study , Humans , Interferon Regulatory Factor-1/genetics , Models, Genetic , Phenotype , Transcription Factors/metabolism
17.
Int J Obes (Lond) ; 47(2): 109-116, 2023 02.
Article in English | MEDLINE | ID: mdl-36463326

ABSTRACT

BACKGROUND/OBJECTIVES: Obesity, defined as excessive fat accumulation that represents a health risk, is increasing in adults and children, reaching global epidemic proportions. Body mass index (BMI) correlates with body fat and future health risk, yet differs in prediction by fat distribution, across populations and by age. Nonetheless, few genetic studies of BMI have been conducted in ancestrally diverse populations. Gene expression association with BMI was assessed in the Multi-Ethnic Study of Atherosclerosis (MESA) in four self-identified race and ethnicity (SIRE) groups to identify genes associated with obesity. SUBJECTS/METHODS: RNA-sequencing was performed on 1096 MESA participants (37.8% white, 24.3% Hispanic, 28.4% African American, and 9.5% Chinese American) and linear models were used to assess the association of expression from each gene for its effect on BMI, adjusting for age, sex, sequencing center, study site, five expression and four genetic principal components in each self-identified race group. Sample-size-weighted meta-analysis was performed to identify genes with BMI-associated expression across ancestry groups. RESULTS: Within individual SIRE groups, there were zero to three genes whose expression is significantly (p < 1.97 × 10-6) associated with BMI. Across all groups, 45 genes were identified by meta-analysis whose expression was significantly associated with BMI, explaining 29.7% of BMI variation. The 45 genes are expressed in a variety of tissues and cell types and are enriched for obesity-related processes including erythrocyte function, oxygen binding and transport, and JAK-STAT signaling. CONCLUSIONS: We have identified genes whose expression is significantly associated with obesity in a multi-ethnic cohort. We have identified novel genes associated with BMI as well as confirmed previously identified genes from earlier genetic analyses. These novel genes and their biological pathways represent new targets for understanding the biology of obesity as well as new therapeutic intervention to reduce obesity and improve global public health.


Subject(s)
Body Mass Index , Gene Expression , Obesity , Adult , Child , Humans , Atherosclerosis , Obesity/epidemiology , Obesity/genetics
18.
Brief Bioinform ; 22(6)2021 11 05.
Article in English | MEDLINE | ID: mdl-34015820

ABSTRACT

Large datasets of hundreds to thousands of individuals measuring RNA-seq in observational studies are becoming available. Many popular software packages for analysis of RNA-seq data were constructed to study differences in expression signatures in an experimental design with well-defined conditions (exposures). In contrast, observational studies may have varying levels of confounding transcript-exposure associations; further, exposure measures may vary from discrete (exposed, yes/no) to continuous (levels of exposure), with non-normal distributions of exposure. We compare popular software for gene expression-DESeq2, edgeR and limma-as well as linear regression-based analyses for studying the association of continuous exposures with RNA-seq. We developed a computation pipeline that includes transformation, filtering and generation of empirical null distribution of association P-values, and we apply the pipeline to compute empirical P-values with multiple testing correction. We employ a resampling approach that allows for assessment of false positive detection across methods, power comparison and the computation of quantile empirical P-values. The results suggest that linear regression methods are substantially faster with better control of false detections than other methods, even with the resampling method to compute empirical P-values. We provide the proposed pipeline with fast algorithms in an R package Olivia, and implemented it to study the associations of measures of sleep disordered breathing with RNA-seq in peripheral blood mononuclear cells in participants from the Multi-Ethnic Study of Atherosclerosis.


Subject(s)
Benchmarking/methods , RNA-Seq , Sequence Analysis, RNA , Software , Algorithms , Atherosclerosis/epidemiology , Atherosclerosis/etiology , Atherosclerosis/metabolism , Computer Simulation , Disease Susceptibility , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Mutation , Phenotype , Risk Assessment , Risk Factors , Web Browser
19.
Respir Res ; 24(1): 30, 2023 Jan 25.
Article in English | MEDLINE | ID: mdl-36698131

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) varies significantly in symptomatic and physiologic presentation. Identifying disease subtypes from molecular data, collected from easily accessible blood samples, can help stratify patients and guide disease management and treatment. METHODS: Blood gene expression measured by RNA-sequencing in the COPDGene Study was analyzed using a network perturbation analysis method. Each COPD sample was compared against a learned reference gene network to determine the part that is deregulated. Gene deregulation values were used to cluster the disease samples. RESULTS: The discovery set included 617 former smokers from COPDGene. Four distinct gene network subtypes are identified with significant differences in symptoms, exercise capacity and mortality. These clusters do not necessarily correspond with the levels of lung function impairment and are independently validated in two external cohorts: 769 former smokers from COPDGene and 431 former smokers in the Multi-Ethnic Study of Atherosclerosis (MESA). Additionally, we identify several genes that are significantly deregulated across these subtypes, including DSP and GSTM1, which have been previously associated with COPD through genome-wide association study (GWAS). CONCLUSIONS: The identified subtypes differ in mortality and in their clinical and functional characteristics, underlining the need for multi-dimensional assessment potentially supplemented by selected markers of gene expression. The subtypes were consistent across cohorts and could be used for new patient stratification and disease prognosis.


Subject(s)
Gene Regulatory Networks , Pulmonary Disease, Chronic Obstructive , Humans , Gene Regulatory Networks/genetics , Smokers , Genome-Wide Association Study/methods , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/genetics , Prognosis
20.
Nature ; 548(7667): 343-346, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28792927

ABSTRACT

Mammalian genomes contain thousands of loci that transcribe long noncoding RNAs (lncRNAs), some of which are known to carry out critical roles in diverse cellular processes through a variety of mechanisms. Although some lncRNA loci encode RNAs that act non-locally (in trans), there is emerging evidence that many lncRNA loci act locally (in cis) to regulate the expression of nearby genes-for example, through functions of the lncRNA promoter, transcription, or transcript itself. Despite their potentially important roles, it remains challenging to identify functional lncRNA loci and distinguish among these and other mechanisms. Here, to address these challenges, we developed a genome-scale CRISPR-Cas9 activation screen that targets more than 10,000 lncRNA transcriptional start sites to identify noncoding loci that influence a phenotype of interest. We found 11 lncRNA loci that, upon recruitment of an activator, mediate resistance to BRAF inhibitors in human melanoma cells. Most candidate loci appear to regulate nearby genes. Detailed analysis of one candidate, termed EMICERI, revealed that its transcriptional activation resulted in dosage-dependent activation of four neighbouring protein-coding genes, one of which confers the resistance phenotype. Our screening and characterization approach provides a CRISPR toolkit with which to systematically discover the functions of noncoding loci and elucidate their diverse roles in gene regulation and cellular function.


Subject(s)
Drug Resistance, Neoplasm/genetics , Genetic Loci/genetics , Genome, Human/genetics , Indoles/pharmacology , Melanoma/genetics , RNA, Long Noncoding/genetics , Sulfonamides/pharmacology , Transcriptional Activation/genetics , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Genetic Loci/drug effects , Hippo Signaling Pathway , Humans , Indoles/therapeutic use , Melanoma/drug therapy , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Phenotype , Promoter Regions, Genetic/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Signal Transduction/drug effects , Sulfonamides/therapeutic use , Transcription Initiation Site , Vemurafenib
SELECTION OF CITATIONS
SEARCH DETAIL