Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nat Commun ; 11(1): 5206, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33060586

ABSTRACT

Variation in the human gut microbiome can reflect host lifestyle and behaviors and influence disease biomarker levels in the blood. Understanding the relationships between gut microbes and host phenotypes are critical for understanding wellness and disease. Here, we examine associations between the gut microbiota and ~150 host phenotypic features across ~3,400 individuals. We identify major axes of taxonomic variance in the gut and a putative diversity maximum along the Firmicutes-to-Bacteroidetes axis. Our analyses reveal both known and unknown associations between microbiome composition and host clinical markers and lifestyle factors, including host-microbe associations that are composition-specific. These results suggest potential opportunities for targeted interventions that alter the composition of the microbiome to improve host health. By uncovering the interrelationships between host diet and lifestyle factors, clinical blood markers, and the human gut microbiome at the population-scale, our results serve as a roadmap for future studies on host-microbe interactions and interventions.


Subject(s)
Biomarkers , Disease , Gastrointestinal Microbiome/physiology , Health , Host Microbial Interactions/physiology , Adult , Biodiversity , Diet , Female , Firmicutes , Gastrointestinal Microbiome/genetics , Humans , Life Style , Male , Middle Aged , RNA, Ribosomal, 16S/genetics , Systems Biology
2.
Proc Natl Acad Sci U S A ; 117(24): 13839-13845, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32471946

ABSTRACT

The Pioneer 100 Wellness Project involved quantitatively profiling 108 participants' molecular physiology over time, including genomes, gut microbiomes, blood metabolomes, blood proteomes, clinical chemistries, and data from wearable devices. Here, we present a longitudinal analysis focused specifically around the Pioneer 100 gut microbiomes. We distinguished a subpopulation of individuals with reduced gut diversity, elevated relative abundance of the genus Prevotella, and reduced levels of the genus Bacteroides We found that the relative abundances of Bacteroides and Prevotella were significantly correlated with certain serum metabolites, including omega-6 fatty acids. Primary dimensions in distance-based redundancy analysis of clinical chemistries explained 18.5% of the variance in bacterial community composition, and revealed a Bacteroides/Prevotella dichotomy aligned with inflammation and dietary markers. Finally, longitudinal analysis of gut microbiome dynamics within individuals showed that direct transitions between Bacteroides-dominated and Prevotella-dominated communities were rare, suggesting the presence of a barrier between these states. One implication is that interventions seeking to transition between Bacteroides- and Prevotella-dominated communities will need to identify permissible paths through ecological state-space that circumvent this apparent barrier.


Subject(s)
Bacteria/isolation & purification , Gastrointestinal Microbiome , Adult , Aged , Bacteria/classification , Bacteria/genetics , Bacteroides/classification , Bacteroides/genetics , Bacteroides/isolation & purification , Cohort Studies , Feces/microbiology , Female , Humans , Longitudinal Studies , Male , Middle Aged , Phylogeny , Prevotella/classification , Prevotella/genetics , Prevotella/isolation & purification
3.
Nat Biotechnol ; 37(10): 1217-1228, 2019 10.
Article in English | MEDLINE | ID: mdl-31477923

ABSTRACT

Depleted gut microbiome α-diversity is associated with several human diseases, but the extent to which this is reflected in the host molecular phenotype is poorly understood. We attempted to predict gut microbiome α-diversity from ~1,000 blood analytes (laboratory tests, proteomics and metabolomics) in a cohort enrolled in a consumer wellness program (N = 399). Although 77 standard clinical laboratory tests and 263 plasma proteins could not accurately predict gut α-diversity, we found that 45% of the variance in α-diversity was explained by a subset of 40 plasma metabolites (13 of the 40 of microbial origin). The prediction capacity of these 40 metabolites was confirmed in a separate validation cohort (N = 540) and across disease states, showing that our findings are robust. Several of the metabolite biomarkers that are reported here are linked with cardiovascular disease, diabetes and kidney function. Associations between host metabolites and gut microbiome α-diversity were modified in those with extreme obesity (body mass index ≥ 35), suggesting metabolic perturbation. The ability of the blood metabolome to predict gut microbiome α-diversity could pave the way to the development of clinical tests for monitoring gut microbial health.


Subject(s)
Bacteria/classification , Gastrointestinal Microbiome , Metabolome , Bacteria/genetics , Cohort Studies , Genetic Variation , Humans , Metabolomics , RNA, Ribosomal, 16S/blood , RNA, Ribosomal, 16S/genetics
4.
Cell Rep ; 24(4): 935-946, 2018 07 24.
Article in English | MEDLINE | ID: mdl-30044989

ABSTRACT

Trimethylamine N-oxide (TMAO) is a circulating metabolite that has been implicated in the development of atherosclerosis and cardiovascular disease (CVD). In this paper, we identify blood markers, metabolites, proteins, gut microbiota patterns, and diets that are significantly associated with levels of plasma TMAO. We find that kidney markers are strongly associated with TMAO and identify CVD-related proteins that are positively correlated with TMAO. We show that metabolites derived by the gut microbiota are strongly correlated with TMAO and that the magnitude of this correlation varies with kidney function. Moreover, we identify diet-associated patterns in the microbiome that are correlated with TMAO. These findings suggest that both the process of TMAO accumulation and the mechanism by which TMAO promotes atherosclerosis are a complex interplay between diet and the microbiome on one hand and other system-level factors such as circulating proteins, metabolites, and kidney function.


Subject(s)
Atherosclerosis/metabolism , Cardiovascular Diseases/metabolism , Gastrointestinal Microbiome/genetics , Methylamines/adverse effects , Female , Humans , Male , Microbiota , Middle Aged
5.
Microbiome ; 5(1): 19, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28179006

ABSTRACT

BACKGROUND: Recent metagenomic analyses of the human gut microbiome identified striking variability in its taxonomic composition across individuals. Notably, however, these studies often reported marked functional uniformity, with relatively little variation in the microbiome's gene composition or in its overall metabolic capacity. RESULTS: Here, we address this surprising discrepancy between taxonomic and functional variations and set out to track its origins. Specifically, we demonstrate that the functional uniformity observed in microbiome studies can be attributed, at least partly, to common computational metagenomic processing procedures that mask true functional variation across microbiome samples. We identify several such procedures, including commonly used practices for gene abundance normalization, mapping of gene families to functional pathways, and gene family aggregation. We show that accounting for these factors and using revised metagenomic processing procedures uncovers such hidden functional variation, significantly increasing observed variation in the abundance of functional elements across samples. Importantly, we find that this uncovered variation is biologically meaningful and that it is associated with both host identity and health. CONCLUSIONS: Accurate characterization of functional variation in the microbiome is essential for comparative metagenomic analyses in health and disease. Our finding that metagenomic processing procedures mask underlying and biologically meaningful functional variation therefore highlights an important challenge such studies may face. Alternative schemes for metagenomic processing that uncover this hidden functional variation can facilitate improved metagenomic analysis and help pinpoint disease- and host-associated shifts in the microbiome's functional capacity.


Subject(s)
Bacteria/genetics , Computational Biology/methods , Gastrointestinal Microbiome/genetics , Gastrointestinal Tract/microbiology , Metagenomics/methods , DNA, Bacterial/genetics , Genetic Variation/genetics , Humans
6.
PLoS One ; 12(2): e0171017, 2017.
Article in English | MEDLINE | ID: mdl-28152044

ABSTRACT

The gut microbiome community structure and development are associated with several health outcomes in young children. To determine the household influences of gut microbiome structure, we assessed microbial sharing within households in western Kenya by sequencing 16S rRNA libraries of fecal samples from children and cattle, cloacal swabs from chickens, and swabs of household surfaces. Among the 156 households studied, children within the same household significantly shared their gut microbiome with each other, although we did not find significant sharing of gut microbiome across host species or household surfaces. Higher gut microbiome diversity among children was associated with lower wealth status and involvement in livestock feeding chores. Although more research is necessary to identify further drivers of microbiota development, these results suggest that the household should be considered as a unit. Livestock activities, health and microbiome perturbations among an individual child may have implications for other children in the household.


Subject(s)
Gastrointestinal Microbiome , Livestock/microbiology , Animals , Biodiversity , Cattle/microbiology , Chickens/microbiology , Child, Preschool , Family Characteristics , Feces/microbiology , Female , Gastrointestinal Microbiome/genetics , Humans , Kenya , Male , Poultry/microbiology , RNA, Ribosomal, 16S , Rural Population
7.
Cell Host Microbe ; 21(2): 254-267, 2017 Feb 08.
Article in English | MEDLINE | ID: mdl-28111203

ABSTRACT

Comparative analyses of the human microbiome have identified both taxonomic and functional shifts that are associated with numerous diseases. To date, however, microbiome taxonomy and function have mostly been studied independently and the taxonomic drivers of functional imbalances have not been systematically identified. Here, we present FishTaco, an analytical and computational framework that integrates taxonomic and functional comparative analyses to accurately quantify taxon-level contributions to disease-associated functional shifts. Applying FishTaco to several large-scale metagenomic cohorts, we show that shifts in the microbiome's functional capacity can be traced back to specific taxa. Furthermore, the set of taxa driving functional shifts and their contribution levels vary markedly between functions. We additionally find that similar functional imbalances in different diseases are driven by both disease-specific and shared taxa. Such integrated analysis of microbiome ecological and functional dynamics can inform future microbiome-based therapy, pinpointing putative intervention targets for manipulating the microbiome's functional capacity.


Subject(s)
Computational Biology , Microbiota , Software , Actinobacteria/classification , Bacteroides/classification , Communicable Diseases/microbiology , Databases, Genetic , Diabetes Mellitus, Type 2/microbiology , Firmicutes/classification , Genome, Microbial , Humans , Metagenomics , Phylogeny , Proteobacteria/classification
8.
Genetics ; 204(4): 1391-1396, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27729424

ABSTRACT

Gene expression levels are dynamic molecular phenotypes that respond to biological, environmental, and technical perturbations. Here we use a novel replicate-classifier approach for discovering transcriptional signatures and apply it to the Genotype-Tissue Expression data set. We identified many factors contributing to expression heterogeneity, such as collection center and ischemia time, and our approach of scoring replicate classifiers allows us to statistically stratify these factors by effect strength. Strikingly, from transcriptional expression in blood alone we detect markers that help predict heart disease and stroke in some patients. Our results illustrate the challenges and opportunities of interpreting patterns of transcriptional variation in large-scale data sets.


Subject(s)
Algorithms , Datasets as Topic/standards , Gene Expression Profiling/standards , Genetic Heterogeneity , Phenotype , Humans , Organ Specificity , Transcriptome
9.
Sci Rep ; 6: 22493, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26940651

ABSTRACT

Cystic fibrosis (CF) results in inflammation, malabsorption of fats and other nutrients, and obstruction in the gastrointestinal (GI) tract, yet the mechanisms linking these disease manifestations to microbiome composition remain largely unexplored. Here we used metagenomic analysis to systematically characterize fecal microbiomes of children with and without CF, demonstrating marked CF-associated taxonomic dysbiosis and functional imbalance. We further showed that these taxonomic and functional shifts were especially pronounced in young children with CF and diminished with age. Importantly, the resulting dysbiotic microbiomes had significantly altered capacities for lipid metabolism, including decreased capacity for overall fatty acid biosynthesis and increased capacity for degrading anti-inflammatory short-chain fatty acids. Notably, these functional differences correlated with fecal measures of fat malabsorption and inflammation. Combined, these results suggest that enteric fat abundance selects for pro-inflammatory GI microbiota in young children with CF, offering novel strategies for improving the health of children with CF-associated fat malabsorption.


Subject(s)
Actinobacteria/genetics , Cystic Fibrosis/microbiology , Dysbiosis/microbiology , Gastrointestinal Microbiome/genetics , Gastrointestinal Tract/microbiology , Metagenome , Proteobacteria/genetics , Biodiversity , Child, Preschool , Cystic Fibrosis/genetics , DNA Barcoding, Taxonomic , Dysbiosis/genetics , Feces/microbiology , Humans , Infant , Infant, Newborn , Leukocyte L1 Antigen Complex/metabolism
10.
Genome Biol ; 16: 53, 2015 Mar 25.
Article in English | MEDLINE | ID: mdl-25885687

ABSTRACT

Functional metagenomic analyses commonly involve a normalization step, where measured levels of genes or pathways are converted into relative abundances. Here, we demonstrate that this normalization scheme introduces marked biases both across and within human microbiome samples, and identify sample- and gene-specific properties that contribute to these biases. We introduce an alternative normalization paradigm, MUSiCC, which combines universal single-copy genes with machine learning methods to correct these biases and to obtain an accurate and biologically meaningful measure of gene abundances. Finally, we demonstrate that MUSiCC significantly improves downstream discovery of functional shifts in the microbiome.MUSiCC is available at http://elbo.gs.washington.edu/software.html .


Subject(s)
Metagenomics , Microbiota/genetics , Sequence Analysis, DNA/methods , Algorithms , Cell Proliferation/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Machine Learning
11.
Bioinformatics ; 31(11): 1848-50, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25637557

ABSTRACT

UNLABELLED: Understanding the effect of single nucleotide polymorphisms (SNPs) on the expression level of genes is an important goal. We recently published a study in which we devised a multi-SNP predictive model for gene expression in Lymphoblastoid cell lines (LCL), and showed that it can robustly predict the expression of a small number of genes in test individuals. Here, we validate the generality of our models by predicting expression profiles for genes in LCL in an independent study, and extend the pool of predictable genes for which we are able to explain more than 25% of their expression variability to 232 genes across 14 different cell types. As the number of people who obtained their SNP profiles through companies such as 23andMe is rising rapidly, we developed GenoExp, a web-based tool in which users can upload their individual SNP data and obtain predicted expression levels for the set of predictable genes across the 14 different cell types. Our tool thus allows users with biological knowledge to study the possible effects that their set of SNPs might have on these genes and predict their cell-specific expression levels relative to the population average. AVAILABILITY AND IMPLEMENTATION: GenoExp is freely available at http://genie.weizmann.ac.il/pubs/GenoExp/.


Subject(s)
Gene Expression Profiling , Polymorphism, Single Nucleotide , Software , Cell Line , Humans , Internet , Models, Genetic
12.
Cell Metab ; 20(5): 742-752, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25176148

ABSTRACT

The human gut microbiome is a major contributor to human metabolism and health, yet the metabolic processes that are carried out by various community members, the way these members interact with each other and with the host, and the impact of such interactions on the overall metabolic machinery of the microbiome have not yet been mapped. Here, we discuss recent efforts to study the metabolic inner workings of this complex ecosystem. We will specifically highlight two interrelated lines of work, the first aiming to deconvolve the microbiome and to characterize the metabolic capacity of various microbiome species and the second aiming to utilize computational modeling to infer and study metabolic interactions between these species.


Subject(s)
Metabolomics , Metagenomics , Microbiota , Bacteria/genetics , Bacteria/metabolism , Computer Simulation , Fungi/genetics , Fungi/metabolism , Humans , Metabolic Networks and Pathways , Metabolome , Metabolomics/methods , Metagenome , Metagenomics/methods , Microbial Interactions , Models, Biological
13.
Genome Res ; 24(10): 1698-706, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25030889

ABSTRACT

Genetically identical cells exhibit large variability (noise) in gene expression, with important consequences for cellular function. Although the amount of noise decreases with and is thus partly determined by the mean expression level, the extent to which different promoter sequences can deviate away from this trend is not fully known. Here, we present a high-throughput method for measuring promoter-driven noise for thousands of designed synthetic promoters in parallel. We use it to investigate how promoters encode different noise levels and find that the noise levels of promoters with similar mean expression levels can vary more than one order of magnitude, with nucleosome-disfavoring sequences resulting in lower noise and more transcription factor binding sites resulting in higher noise. We propose a kinetic model of gene expression that takes into account the nonspecific DNA binding and one-dimensional sliding along the DNA, which occurs when transcription factors search for their target sites. We show that this assumption can improve the prediction of the mean-independent component of expression noise for our designed promoter sequences, suggesting that a transcription factor target search may affect gene expression noise. Consistent with our findings in designed promoters, we find that binding-site multiplicity in native promoters is associated with higher expression noise. Overall, our results demonstrate that small changes in promoter DNA sequence can tune noise levels in a manner that is predictable and partly decoupled from effects on the mean expression levels. These insights may assist in designing promoters with desired noise levels.


Subject(s)
Computational Biology/methods , DNA/metabolism , Gene Expression , Promoter Regions, Genetic , Saccharomyces cerevisiae/genetics , Binding Sites , Genes, Fungal , Linear Models , Molecular Sequence Data , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism
14.
Curr Biol ; 24(11): R516-7, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24892909

ABSTRACT

The number of applicants vastly outnumbers the available academic faculty positions. What makes a successful academic job market candidate is the subject of much current discussion [1-4]. Yet, so far there has been no quantitative analysis of who becomes a principal investigator (PI). We here use a machine-learning approach to predict who becomes a PI, based on data from over 25,000 scientists in PubMed. We show that success in academia is predictable. It depends on the number of publications, the impact factor (IF) of the journals in which those papers are published, and the number of papers that receive more citations than average for the journal in which they were published (citations/IF). However, both the scientist's gender and the rank of their university are also of importance, suggesting that non-publication features play a statistically significant role in the academic hiring process. Our model (www.pipredictor.com) allows anyone to calculate their likelihood of becoming a PI.


Subject(s)
Artificial Intelligence , Journal Impact Factor , Research Personnel/statistics & numerical data , Universities/statistics & numerical data , Faculty , Humans , Models, Theoretical , Probability , Sex Factors
15.
Sci Signal ; 7(327): ra50, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24866020

ABSTRACT

Type I interferons (IFNs), including various IFN-α isoforms and IFN-ß, are a family of homologous, multifunctional cytokines. IFNs activate different cellular responses by binding to a common receptor that consists of two subunits, IFNAR1 and IFNAR2. In addition to stimulating antiviral responses, they also inhibit cell proliferation and modulate other immune responses. We characterized various IFNs, including a mutant IFN-α2 (IFN-1ant) that bound tightly to IFNAR2 but had markedly reduced binding to IFNAR1. Whereas IFN-1ant stimulated antiviral activity in a range of cell lines, it failed to elicit immunomodulatory and antiproliferative activities. The antiviral activities of the various IFNs tested depended on a set of IFN-sensitive genes (the "robust" genes) that were controlled by canonical IFN response elements and responded at low concentrations of IFNs. Conversely, these elements were not found in the promoters of genes required for the antiproliferative responses of IFNs (the "tunable" genes). The extent of expression of tunable genes was cell type-specific and correlated with the magnitude of the antiproliferative effects of the various IFNs. Although IFN-1ant induced the expression of robust genes similarly in five different cell lines, its antiviral activity was virus- and cell type-specific. Our findings suggest that IFN-1ant may be a therapeutic candidate for the treatment of specific viral infections without inducing the immunomodulatory and antiproliferative functions of wild-type IFN.


Subject(s)
Gene Expression Regulation/immunology , Interferon Type I/immunology , Receptor, Interferon alpha-beta/metabolism , Virus Diseases/immunology , Cell Line, Tumor , Cell Proliferation/physiology , Cluster Analysis , Flow Cytometry , Humans , Interferon Type I/metabolism , Principal Component Analysis , RNA, Small Interfering/genetics
16.
Nature ; 505(7485): 706-9, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24476892

ABSTRACT

In parallel to the genetic code for protein synthesis, a second layer of information is embedded in all RNA transcripts in the form of RNA structure. RNA structure influences practically every step in the gene expression program. However, the nature of most RNA structures or effects of sequence variation on structure are not known. Here we report the initial landscape and variation of RNA secondary structures (RSSs) in a human family trio (mother, father and their child). This provides a comprehensive RSS map of human coding and non-coding RNAs. We identify unique RSS signatures that demarcate open reading frames and splicing junctions, and define authentic microRNA-binding sites. Comparison of native deproteinized RNA isolated from cells versus refolded purified RNA suggests that the majority of the RSS information is encoded within RNA sequence. Over 1,900 transcribed single nucleotide variants (approximately 15% of all transcribed single nucleotide variants) alter local RNA structure. We discover simple sequence and spacing rules that determine the ability of point mutations to impact RSSs. Selective depletion of 'riboSNitches' versus structurally synonymous variants at precise locations suggests selection for specific RNA shapes at thousands of sites, including 3' untranslated regions, binding sites of microRNAs and RNA-binding proteins genome-wide. These results highlight the potentially broad contribution of RNA structure and its variation to gene regulation.


Subject(s)
Nucleic Acid Conformation , RNA/chemistry , RNA/genetics , Transcriptome/genetics , 3' Untranslated Regions/genetics , Base Sequence , Binding Sites , Child , Female , Gene Expression Regulation/genetics , Genome, Human/genetics , Humans , Male , MicroRNAs/chemistry , MicroRNAs/genetics , MicroRNAs/metabolism , Open Reading Frames/genetics , Point Mutation/genetics , RNA/metabolism , RNA Splice Sites/genetics , RNA-Binding Proteins/metabolism
17.
Dev Neurobiol ; 74(3): 365-81, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24127433

ABSTRACT

RNA localization is a regulatory mechanism that is conserved from bacteria to mammals. Yet, little is known about the mechanism and the logic that govern the distribution of RNA transcripts within the cell. Here, we present a novel organ culture system, which enables the isolation of RNA specifically from NGF dependent re-growing peripheral axons of mouse embryo, sensory neurons. In combination with massive parallel sequencing technology, we determine the subcellular localization of most transcripts in the transcriptome. We found that the axon is enriched in mRNAs that encode secreted proteins, transcription factors, and the translation machinery. In contrast, the axon was largely depleted from mRNAs encoding transmembrane proteins, a particularly interesting finding, since many of these gene products are specifically expressed in the tip of the axon at the protein level. Comparison of the mitochondrial mRNAs encoded in the nucleus with those encoded in the mitochondria, uncovered completely different localization pattern, with the latter much enriched in the axon fraction. This discovery is intriguing since the protein products encoded by the nuclear and mitochondrial genome form large co-complexes. Finally, focusing on alternative splice variants that are specific to axonal fractions, we find short sequence motifs that are enriched in the axonal transcriptome. Together our findings shed light on the extensive role of RNA localization and its characteristics.


Subject(s)
Axons/metabolism , Ganglia, Spinal/metabolism , RNA, Messenger/metabolism , Sensory Receptor Cells/metabolism , Transcriptome , Alternative Splicing , Animals , Cell Nucleus/metabolism , High-Throughput Nucleotide Sequencing , Mice , Mice, Inbred ICR , Mitochondria/metabolism , Presynaptic Terminals/metabolism , RNA, Mitochondrial , Tissue Culture Techniques
18.
PLoS Comput Biol ; 9(8): e1003200, 2013.
Article in English | MEDLINE | ID: mdl-23990773

ABSTRACT

Genome-wide association studies (GWAS) are widely used to search for genetic loci that underlie human disease. Another goal is to predict disease risk for different individuals given their genetic sequence. Such predictions could either be used as a "black box" in order to promote changes in life-style and screening for early diagnosis, or as a model that can be studied to better understand the mechanism of the disease. Current methods for risk prediction typically rank single nucleotide polymorphisms (SNPs) by the p-value of their association with the disease, and use the top-associated SNPs as input to a classification algorithm. However, the predictive power of such methods is relatively poor. To improve the predictive power, we devised BootRank, which uses bootstrapping in order to obtain a robust prioritization of SNPs for use in predictive models. We show that BootRank improves the ability to predict disease risk of unseen individuals in the Wellcome Trust Case Control Consortium (WTCCC) data and results in a more robust set of SNPs and a larger number of enriched pathways being associated with the different diseases. Finally, we show that combining BootRank with seven different classification algorithms improves performance compared to previous studies that used the WTCCC data. Notably, diseases for which BootRank results in the largest improvements were recently shown to have more heritability than previously thought, likely due to contributions from variants with low minimum allele frequency (MAF), suggesting that BootRank can be beneficial in cases where SNPs affecting the disease are poorly tagged or have low MAF. Overall, our results show that improving disease risk prediction from genotypic information may be a tangible goal, with potential implications for personalized disease screening and treatment.


Subject(s)
Algorithms , Cluster Analysis , Computational Biology/methods , Disease/genetics , Genetic Predisposition to Disease , Area Under Curve , Genotype , Humans , Models, Genetic , Polymorphism, Single Nucleotide , Reproducibility of Results , Sequence Analysis, DNA/methods
19.
PLoS Genet ; 9(3): e1003396, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23555302

ABSTRACT

Many genetic variants that are significantly correlated to gene expression changes across human individuals have been identified, but the ability of these variants to predict expression of unseen individuals has rarely been evaluated. Here, we devise an algorithm that, given training expression and genotype data for a set of individuals, predicts the expression of genes of unseen test individuals given only their genotype in the local genomic vicinity of the predicted gene. Notably, the resulting predictions are remarkably robust in that they agree well between the training and test sets, even when the training and test sets consist of individuals from distinct populations. Thus, although the overall number of genes that can be predicted is relatively small, as expected from our choice to ignore effects such as environmental factors and trans sequence variation, the robust nature of the predictions means that the identity and quantitative degree to which genes can be predicted is known in advance. We also present an extension that incorporates heterogeneous types of genomic annotations to differentially weigh the importance of the various genetic variants, and we show that assigning higher weights to variants with particular annotations such as proximity to genes and high regional G/C content can further improve the predictions. Finally, genes that are successfully predicted have, on average, higher expression and more variability across individuals, providing insight into the characteristics of the types of genes that can be predicted from their cis genetic variation.


Subject(s)
Gene Expression , Genotype , Quantitative Trait Loci/genetics , HapMap Project , Humans , Polymorphism, Single Nucleotide
20.
Blood ; 121(6): 1016-27, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23212522

ABSTRACT

The mononuclear phagocyte system comprises cells as diverse as monocytes, macrophages, and dendritic cells (DCs), which collectively play key roles in innate immune responses and the triggering of adaptive immunity. Recent studies have highlighted the role of growth and transcription factors in defining developmental pathways and lineage relations within this cellular compartment. However, contributions of miRNAs to the development of mononuclear phagocytes remain largely unknown. In the present study, we report a comprehensive map of miRNA expression profiles for distinct myeloid populations, including BM-resident progenitors, monocytes, and mature splenic DCs. Each of the analyzed cell populations displayed a distinctive miRNA profile, suggesting a role for miRNAs in defining myeloid cell identities. Focusing on DC development, we found miR-142 to be highly expressed in classic FLT3-L­dependent CD4+ DCs, whereas reduced expression was observed in closely related CD8α+ or CD4- CD8α- DCs. Moreover, mice deficient for miR-142 displayed an impairment of CD4+ DC homeostasis both in vitro and in vivo. Furthermore, loss of miR-142­dependent CD4+ DCs was accompanied by a severe and specific defect in the priming of CD4+ T cells. The results of our study establish a novel role for miRNAs in myeloid cell specification and define miR-142 as a pivotal genetic component in the maintenance of CD4+ DCs.


Subject(s)
Dendritic Cells/metabolism , Homeostasis/genetics , MicroRNAs/genetics , Phagocytes/metabolism , Transcriptome/genetics , Animals , CD4 Antigens/immunology , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Dendritic Cells/immunology , Female , Flow Cytometry , Gene Expression Profiling , Homeostasis/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MicroRNAs/immunology , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , Phagocytes/immunology , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...