Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters

Publication year range
1.
Cell ; 172(5): 952-965.e18, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29474921

ABSTRACT

Viruses that are typically benign sometimes invade the brainstem in otherwise healthy children. We report bi-allelic DBR1 mutations in unrelated patients from different ethnicities, each of whom had brainstem infection due to herpes simplex virus 1 (HSV1), influenza virus, or norovirus. DBR1 encodes the only known RNA lariat debranching enzyme. We show that DBR1 expression is ubiquitous, but strongest in the spinal cord and brainstem. We also show that all DBR1 mutant alleles are severely hypomorphic, in terms of expression and function. The fibroblasts of DBR1-mutated patients contain higher RNA lariat levels than control cells, this difference becoming even more marked during HSV1 infection. Finally, we show that the patients' fibroblasts are highly susceptible to HSV1. RNA lariat accumulation and viral susceptibility are rescued by wild-type DBR1. Autosomal recessive, partial DBR1 deficiency underlies viral infection of the brainstem in humans through the disruption of tissue-specific and cell-intrinsic immunity to viruses.


Subject(s)
Brain Diseases, Metabolic, Inborn/genetics , Brain Stem/metabolism , Brain Stem/virology , RNA/chemistry , RNA/metabolism , Alleles , Amino Acid Sequence , Animals , Brain Diseases, Metabolic, Inborn/pathology , Brain Stem/pathology , Encephalitis, Viral/genetics , Escherichia coli/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/virology , Herpesvirus 1, Human , Humans , Interferons/metabolism , Introns/genetics , Male , Mice , Mutant Proteins/metabolism , Mutation/genetics , Open Reading Frames/genetics , Pedigree , RNA Nucleotidyltransferases/chemistry , RNA Nucleotidyltransferases/deficiency , RNA Nucleotidyltransferases/genetics , Toll-Like Receptor 3/metabolism , Virus Replication
2.
Cell ; 168(5): 789-800.e10, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28235196

ABSTRACT

The molecular basis of the incomplete penetrance of monogenic disorders is unclear. We describe here eight related individuals with autosomal recessive TIRAP deficiency. Life-threatening staphylococcal disease occurred during childhood in the proband, but not in the other seven homozygotes. Responses to all Toll-like receptor 1/2 (TLR1/2), TLR2/6, and TLR4 agonists were impaired in the fibroblasts and leukocytes of all TIRAP-deficient individuals. However, the whole-blood response to the TLR2/6 agonist staphylococcal lipoteichoic acid (LTA) was abolished only in the index case individual, the only family member lacking LTA-specific antibodies (Abs). This defective response was reversed in the patient, but not in interleukin-1 receptor-associated kinase 4 (IRAK-4)-deficient individuals, by anti-LTA monoclonal antibody (mAb). Anti-LTA mAb also rescued the macrophage response in mice lacking TIRAP, but not TLR2 or MyD88. Thus, acquired anti-LTA Abs rescue TLR2-dependent immunity to staphylococcal LTA in individuals with inherited TIRAP deficiency, accounting for incomplete penetrance. Combined TIRAP and anti-LTA Ab deficiencies underlie staphylococcal disease in this patient.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Lipopolysaccharides/metabolism , Membrane Glycoproteins/deficiency , Receptors, Interleukin-1/deficiency , Staphylococcal Infections/genetics , Staphylococcal Infections/immunology , Teichoic Acids/metabolism , Adaptive Immunity , Child , Female , Fibroblasts/metabolism , Humans , Immunity, Innate , Lipopolysaccharides/immunology , Macrophages/immunology , Male , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Monocytes/metabolism , Myeloid Differentiation Factor 88/metabolism , Pedigree , Phagocytes/metabolism , Point Mutation , Protein Isoforms/analysis , Protein Isoforms/genetics , Receptors, Interleukin-1/analysis , Receptors, Interleukin-1/genetics , Staphylococcal Infections/drug therapy , Teichoic Acids/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism
3.
Nat Immunol ; 19(9): 973-985, 2018 09.
Article in English | MEDLINE | ID: mdl-30127434

ABSTRACT

Human inborn errors of IFN-γ immunity underlie mycobacterial diseases. We describe patients with Mycobacterium bovis (BCG) disease who are homozygous for loss-of-function mutations of SPPL2A. This gene encodes a transmembrane protease that degrades the N-terminal fragment (NTF) of CD74 (HLA invariant chain) in antigen-presenting cells. The CD74 NTF therefore accumulates in the HLA class II+ myeloid and lymphoid cells of SPPL2a-deficient patients. This toxic fragment selectively depletes IL-12- and IL-23-producing CD1c+ conventional dendritic cells (cDC2s) and their circulating progenitors. Moreover, SPPL2a-deficient memory TH1* cells selectively fail to produce IFN-γ when stimulated with mycobacterial antigens in vitro. Finally, Sppl2a-/- mice lack cDC2s, have CD4+ T cells that produce small amounts of IFN-γ after BCG infection, and are highly susceptible to infection with BCG or Mycobacterium tuberculosis. These findings suggest that inherited SPPL2a deficiency in humans underlies mycobacterial disease by decreasing the numbers of cDC2s and impairing IFN-γ production by mycobacterium-specific memory TH1* cells.


Subject(s)
Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Dendritic Cells/immunology , Membrane Proteins/metabolism , Mycobacterium Infections/immunology , Mycobacterium bovis/physiology , Mycobacterium tuberculosis/physiology , Th1 Cells/immunology , Tuberculosis/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/metabolism , Cells, Cultured , HLA Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Immunity , Immunologic Memory , Infant , Interferon-gamma/metabolism , Lymphadenopathy , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Mycobacterium Infections/genetics , Vaccination
4.
Cell ; 152(4): 691-702, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23415220

ABSTRACT

An adaptive variant of the human Ectodysplasin receptor, EDARV370A, is one of the strongest candidates of recent positive selection from genome-wide scans. We have modeled EDAR370A in mice and characterized its phenotype and evolutionary origins in humans. Our computational analysis suggests the allele arose in central China approximately 30,000 years ago. Although EDAR370A has been associated with increased scalp hair thickness and changed tooth morphology in humans, its direct biological significance and potential adaptive role remain unclear. We generated a knockin mouse model and find that, as in humans, hair thickness is increased in EDAR370A mice. We identify new biological targets affected by the mutation, including mammary and eccrine glands. Building on these results, we find that EDAR370A is associated with an increased number of active eccrine glands in the Han Chinese. This interdisciplinary approach yields unique insight into the generation of adaptive variation among modern humans.


Subject(s)
Biological Evolution , Edar Receptor/genetics , Exocrine Glands/physiology , Hair/physiology , Mice , Models, Animal , Adolescent , Adult , Amino Acid Sequence , Animals , Evolution, Molecular , Gene Knock-In Techniques , Genetic Pleiotropy , Haplotypes , Humans , Mice, Inbred C57BL , Middle Aged , Molecular Sequence Data , Polymorphism, Single Nucleotide , Scalp/physiology , Sequence Alignment , Young Adult
5.
Am J Hum Genet ; 108(12): 2301-2318, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34762822

ABSTRACT

Identifying whether a given genetic mutation results in a gene product with increased (gain-of-function; GOF) or diminished (loss-of-function; LOF) activity is an important step toward understanding disease mechanisms because they may result in markedly different clinical phenotypes. Here, we generated an extensive database of documented germline GOF and LOF pathogenic variants by employing natural language processing (NLP) on the available abstracts in the Human Gene Mutation Database. We then investigated various gene- and protein-level features of GOF and LOF variants and applied machine learning and statistical analyses to identify discriminative features. We found that GOF variants were enriched in essential genes, for autosomal-dominant inheritance, and in protein binding and interaction domains, whereas LOF variants were enriched in singleton genes, for protein-truncating variants, and in protein core regions. We developed a user-friendly web-based interface that enables the extraction of selected subsets from the GOF/LOF database by a broad set of annotated features and downloading of up-to-date versions. These results improve our understanding of how variants affect gene/protein function and may ultimately guide future treatment options.


Subject(s)
Databases, Genetic , Gain of Function Mutation , Loss of Function Mutation , Proteins/genetics , Cloud Computing , Genetic Predisposition to Disease , Genome, Human , Germ-Line Mutation , Humans , Internet-Based Intervention , Machine Learning
6.
Am J Hum Genet ; 108(6): 1012-1025, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34015270

ABSTRACT

The human genetic dissection of clinical phenotypes is complicated by genetic heterogeneity. Gene burden approaches that detect genetic signals in case-control studies are underpowered in genetically heterogeneous cohorts. We therefore developed a genome-wide computational method, network-based heterogeneity clustering (NHC), to detect physiological homogeneity in the midst of genetic heterogeneity. Simulation studies showed our method to be capable of systematically converging genes in biological proximity on the background biological interaction network, and capturing gene clusters harboring presumably deleterious variants, in an efficient and unbiased manner. We applied NHC to whole-exome sequencing data from a cohort of 122 individuals with herpes simplex encephalitis (HSE), including 13 individuals with previously published monogenic inborn errors of TLR3-dependent IFN-α/ß immunity. The top gene cluster identified by our approach successfully detected and prioritized all causal variants of five TLR3 pathway genes in the 13 previously reported individuals. This approach also suggested candidate variants of three reported genes and four candidate genes from the same pathway in another ten previously unstudied individuals. TLR3 responsiveness was impaired in dermal fibroblasts from four of the five individuals tested, suggesting that the variants detected were causal for HSE. NHC is, therefore, an effective and unbiased approach for unraveling genetic heterogeneity by detecting physiological homogeneity.


Subject(s)
Computational Biology/methods , Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/pathology , Fibroblasts/immunology , Gene Regulatory Networks , Genetic Heterogeneity , Genetic Predisposition to Disease , Case-Control Studies , Encephalitis, Herpes Simplex/immunology , Fibroblasts/metabolism , Humans , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/immunology , Toll-Like Receptor 3/metabolism , Exome Sequencing
7.
Brief Bioinform ; 23(5)2022 09 20.
Article in English | MEDLINE | ID: mdl-35598327

ABSTRACT

Distinguishing pathogenic variants from non-pathogenic ones remains a major challenge in clinical genetic testing of primary immunodeficiency (PID) patients. Most of the existing mutation pathogenicity prediction tools treat all mutations as homogeneous entities, ignoring the differences in characteristics of different genes, and use the same model for genes in different diseases. In this study, we developed a single nucleotide variant (SNV) pathogenicity prediction tool, Variant Impact Predictor for PIDs (VIPPID; https://mylab.shinyapps.io/VIPPID/), which was tailored for PIDs genes and used a specific model for each of the most prevalent PID known genes. It employed a Conditional Inference Forest model and utilized information of 85 features of SNVs and scores from 20 existing prediction tools. Evaluation of VIPPID showed that it had superior performance (area under the curve = 0.91) over non-specific conventional tools. In addition, we also showed that the gene-specific model outperformed the non-gene-specific models. Our study demonstrated that disease-specific and gene-specific models can improve SNV pathogenicity prediction performance. This observation supports the notion that each feature of mutations in the model can be potentially used, in a new algorithm, to investigate the characteristics and function of the encoded proteins.


Subject(s)
Polymorphism, Single Nucleotide , Primary Immunodeficiency Diseases , Algorithms , Humans , Nucleotides , Virulence
8.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Article in English | MEDLINE | ID: mdl-34426522

ABSTRACT

The construction of population-based variomes has contributed substantially to our understanding of the genetic basis of human inherited disease. Here, we investigated the genetic structure of Turkey from 3,362 unrelated subjects whose whole exomes (n = 2,589) or whole genomes (n = 773) were sequenced to generate a Turkish (TR) Variome that should serve to facilitate disease gene discovery in Turkey. Consistent with the history of present-day Turkey as a crossroads between Europe and Asia, we found extensive admixture between Balkan, Caucasus, Middle Eastern, and European populations with a closer genetic relationship of the TR population to Europeans than hitherto appreciated. We determined that 50% of TR individuals had high inbreeding coefficients (≥0.0156) with runs of homozygosity longer than 4 Mb being found exclusively in the TR population when compared to 1000 Genomes Project populations. We also found that 28% of exome and 49% of genome variants in the very rare range (allele frequency < 0.005) are unique to the modern TR population. We annotated these variants based on their functional consequences to establish a TR Variome containing alleles of potential medical relevance, a repository of homozygous loss-of-function variants and a TR reference panel for genotype imputation using high-quality haplotypes, to facilitate genome-wide association studies. In addition to providing information on the genetic structure of the modern TR population, these data provide an invaluable resource for future studies to identify variants that are associated with specific phenotypes as well as establishing the phenotypic consequences of mutations in specific genes.


Subject(s)
Genetic Variation/genetics , Genome, Human/genetics , Alleles , Consanguinity , Exome , Gene Frequency/genetics , Genetic Drift , Genetics, Population/methods , Genome-Wide Association Study/methods , Genotype , Haplotypes/genetics , Human Migration/trends , Humans , Turkey/ethnology , Exome Sequencing/methods
9.
Hum Genet ; 142(2): 275-288, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36352240

ABSTRACT

Epilepsy (EP) and congenital heart disease (CHD) are two apparently unrelated diseases that nevertheless display substantial mutual comorbidity. Thus, while congenital heart defects are associated with an elevated risk of developing epilepsy, the incidence of epilepsy in CHD patients correlates with CHD severity. Although genetic determinants have been postulated to underlie the comorbidity of EP and CHD, the precise genetic etiology is unknown. We performed variant and gene association analyses on EP and CHD patients separately, using whole exomes of genetically identified Europeans from the UK Biobank and Mount Sinai BioMe Biobank. We prioritized biologically plausible candidate genes and investigated the enriched pathways and other identified comorbidities by biological proximity calculation, pathway analyses, and gene-level phenome-wide association studies. Our variant- and gene-level results point to the Voltage-Gated Calcium Channels (VGCC) pathway as being a unifying framework for EP and CHD comorbidity. Additionally, pathway-level analyses indicated that the functions of disease-associated genes partially overlap between the two disease entities. Finally, phenome-wide association analyses of prioritized candidate genes revealed that cerebral blood flow and ulcerative colitis constitute the two main traits associated with both EP and CHD.


Subject(s)
Epilepsy , Heart Defects, Congenital , Humans , European People , Heart Defects, Congenital/genetics , Epilepsy/epidemiology , Epilepsy/genetics , Genetic Association Studies , Phenotype
10.
Gastroenterology ; 160(5): 1709-1724, 2021 04.
Article in English | MEDLINE | ID: mdl-33421512

ABSTRACT

BACKGROUND & AIMS: Recent literature has implicated a key role for mast cells in murine models of colonic inflammation, but their role in human ulcerative colitis (UC) is not well established. A major advance has been the identification of mrgprb2 (human orthologue, MRGPX2) as mediating IgE-independent mast cell activation. We sought to define mechanisms of mast cell activation and MRGPRX2 in human UC. METHODS: Colon tissues were collected from patients with UC for bulk RNA sequencing and lamina propria cells were isolated for MRGPRX2 activation studies and single-cell RNA sequencing. Genetic association of all protein-altering G-protein coupled receptor single-nucleotide polymorphism was performed in an Ashkenazi Jewish UC case-control cohort. Variants of MRGPRX2 were transfected into Chinese hamster ovary (CHO) and human mast cell (HMC) 1.1 cells to detect genotype-dependent effects on ß-arrestin recruitment, IP-1 accumulation, and phosphorylated extracellular signal-regulated kinase. RESULTS: Mast cell-specific mediators and adrenomedullin (proteolytic precursor of PAMP-12, an MRGPRX2 agonist) are up-regulated in inflamed compared to uninflamed UC. MRGPRX2 stimulation induces carboxypeptidase secretion from inflamed UC. Of all protein-altering GPCR alleles, a unique variant of MRGPRX2, Asn62Ser, was most associated with and was bioinformatically predicted to alter arrestin recruitment. We validated that the UC protective serine allele enhances ß-arrestin recruitment, decreases IP-1, and increases phosphorylated extracellular signal-regulated kinase with MRGPRX2 agonists. Single-cell RNA sequencing defines that adrenomedullin is expressed by activated fibroblasts and epithelial cells and that interferon gamma is a key upstream regulator of mast cell gene expression. CONCLUSION: Inflamed UC regions are distinguished by MRGPRX2-mediated activation of mast cells, with decreased activation observed with a UC-protective genetic variant. These results define cell modules of UC activation and a new therapeutic target.


Subject(s)
Cell Degranulation , Colitis, Ulcerative/metabolism , Colon/metabolism , Mast Cells/metabolism , Nerve Tissue Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Adrenomedullin/genetics , Adrenomedullin/metabolism , Animals , CHO Cells , Case-Control Studies , Colitis, Ulcerative/genetics , Colitis, Ulcerative/immunology , Colon/immunology , Cricetulus , Extracellular Signal-Regulated MAP Kinases/metabolism , Genetic Variation , Humans , Inositol Phosphates/metabolism , Ligands , Mast Cells/immunology , Nerve Tissue Proteins/genetics , Phosphorylation , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism
11.
Gastroenterology ; 160(5): 1546-1557, 2021 04.
Article in English | MEDLINE | ID: mdl-33359885

ABSTRACT

BACKGROUND AND AIMS: Polygenic risk scores (PRS) may soon be used to predict inflammatory bowel disease (IBD) risk in prevention efforts. We leveraged exome-sequence and single nucleotide polymorphism (SNP) array data from 29,358 individuals in the multiethnic, randomly ascertained health system-based BioMe biobank to define effects of common and rare IBD variants on disease prediction and pathophysiology. METHODS: PRS were calculated from European, African American, and Ashkenazi Jewish (AJ) reference case-control studies, and a meta-GWAS run using all three association datasets. PRS were then combined using regression to assess which combination of scores best predicted IBD status in European, AJ, Hispanic, and African American cohorts in BioMe. Additionally, rare variants were assessed in genes associated with very early-onset IBD (VEO-IBD), by estimating genetic penetrance in each BioMe population. RESULTS: Combining risk scores based on association data from distinct ancestral populations improved IBD prediction for every population in BioMe and significantly improved prediction among European ancestry UK Biobank individuals. Lower predictive power for non-Europeans was observed, reflecting in part substantially lower African IBD case-control reference sizes. We replicated associations for two VEO-IBD genes, ADAM17 and LRBA, with high dominant model penetrance in BioMe. Autosomal recessive LRBA risk alleles are associated with severe, early-onset autoimmunity; we show that heterozygous carriage of an African-predominant LRBA protein-altering allele is associated with significantly decreased LRBA and CTLA-4 expression with T-cell activation. CONCLUSIONS: Greater genetic diversity in African populations improves prediction across populations, and generalizes some VEO-IBD genes. Increasing African American IBD case-collections should be prioritized to reduce health disparities and enhance pathophysiological insight.


Subject(s)
Black or African American/genetics , Colitis, Ulcerative/genetics , Crohn Disease/genetics , Hispanic or Latino/genetics , Jews/genetics , Multifactorial Inheritance , Penetrance , Polymorphism, Single Nucleotide , White People/genetics , Age of Onset , Case-Control Studies , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/ethnology , Crohn Disease/diagnosis , Crohn Disease/ethnology , Europe/epidemiology , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Phenotype , Prevalence , Race Factors , Risk Assessment , Risk Factors , United States/epidemiology
12.
Proc Natl Acad Sci U S A ; 116(3): 950-959, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30591557

ABSTRACT

Computational analyses of human patient exomes aim to filter out as many nonpathogenic genetic variants (NPVs) as possible, without removing the true disease-causing mutations. This involves comparing the patient's exome with public databases to remove reported variants inconsistent with disease prevalence, mode of inheritance, or clinical penetrance. However, variants frequent in a given exome cohort, but absent or rare in public databases, have also been reported and treated as NPVs, without rigorous exploration. We report the generation of a blacklist of variants frequent within an in-house cohort of 3,104 exomes. This blacklist did not remove known pathogenic mutations from the exomes of 129 patients and decreased the number of NPVs remaining in the 3,104 individual exomes by a median of 62%. We validated this approach by testing three other independent cohorts of 400, 902, and 3,869 exomes. The blacklist generated from any given cohort removed a substantial proportion of NPVs (11-65%). We analyzed the blacklisted variants computationally and experimentally. Most of the blacklisted variants corresponded to false signals generated by incomplete reference genome assembly, location in low-complexity regions, bioinformatic misprocessing, or limitations inherent to cohort-specific private alleles (e.g., due to sequencing kits, and genetic ancestries). Finally, we provide our precalculated blacklists, together with ReFiNE, a program for generating customized blacklists from any medium-sized or large in-house cohort of exome (or other next-generation sequencing) data via a user-friendly public web server. This work demonstrates the power of extracting variant blacklists from private databases as a specific in-house but broadly applicable tool for optimizing exome analysis.


Subject(s)
Databases, Nucleic Acid , Exome , Genetic Variation , Genome, Human , Sequence Analysis, DNA , Software , Cohort Studies , Female , Humans , Male
13.
Nature ; 517(7532): 89-93, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25307056

ABSTRACT

Intracellular ISG15 is an interferon (IFN)-α/ß-inducible ubiquitin-like modifier which can covalently bind other proteins in a process called ISGylation; it is an effector of IFN-α/ß-dependent antiviral immunity in mice. We previously published a study describing humans with inherited ISG15 deficiency but without unusually severe viral diseases. We showed that these patients were prone to mycobacterial disease and that human ISG15 was non-redundant as an extracellular IFN-γ-inducing molecule. We show here that ISG15-deficient patients also display unanticipated cellular, immunological and clinical signs of enhanced IFN-α/ß immunity, reminiscent of the Mendelian autoinflammatory interferonopathies Aicardi-Goutières syndrome and spondyloenchondrodysplasia. We further show that an absence of intracellular ISG15 in the patients' cells prevents the accumulation of USP18, a potent negative regulator of IFN-α/ß signalling, resulting in the enhancement and amplification of IFN-α/ß responses. Human ISG15, therefore, is not only redundant for antiviral immunity, but is a key negative regulator of IFN-α/ß immunity. In humans, intracellular ISG15 is IFN-α/ß-inducible not to serve as a substrate for ISGylation-dependent antiviral immunity, but to ensure USP18-dependent regulation of IFN-α/ß and prevention of IFN-α/ß-dependent autoinflammation.


Subject(s)
Cytokines/metabolism , Inflammation/prevention & control , Interferon Type I/immunology , Intracellular Space/metabolism , Ubiquitins/metabolism , Adolescent , Alleles , Child , Cytokines/deficiency , Cytokines/genetics , Endopeptidases/chemistry , Endopeptidases/metabolism , Female , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/immunology , Interferon Type I/metabolism , Male , Pedigree , S-Phase Kinase-Associated Proteins/metabolism , Signal Transduction , Ubiquitin Thiolesterase , Ubiquitination , Ubiquitins/deficiency , Ubiquitins/genetics , Viruses/immunology
14.
Nucleic Acids Res ; 47(W1): W623-W631, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31045209

ABSTRACT

Human whole-genome-sequencing reveals about 4 000 000 genomic variants per individual. These data are mostly stored as VCF-format files. Although many variant analysis methods accept VCF as input, many other tools require DNA or protein sequences, particularly for splicing prediction, sequence alignment, phylogenetic analysis, and structure prediction. However, there is no existing webserver capable of extracting DNA/protein sequences for genomic variants from VCF files in a user-friendly and efficient manner. We developed the SeqTailor webserver to bridge this gap, by enabling rapid extraction of (i) DNA sequences around genomic variants, with customizable window sizes and options to annotate the splice sites closest to the variants and to consider the neighboring variants within the window; and (ii) protein sequences encoded by the DNA sequences around genomic variants, with built-in SnpEff annotator and customizable window sizes. SeqTailor supports 11 species, including: human (GRCh37/GRCh38), chimpanzee, mouse, rat, cow, chicken, lizard, zebrafish, fruitfly, Arabidopsis and rice. Standalone programs are provided for command-line-based needs. SeqTailor streamlines the sequence extraction process, and accelerates the analysis of genomic variants with software requiring DNA/protein sequences. It will facilitate the study of genomic variation, by increasing the feasibility of sequence-based analysis and prediction. The SeqTailor webserver is freely available at http://shiva.rockefeller.edu/SeqTailor/.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, DNA/methods , Sequence Analysis, Protein/methods , Software , Animals , Cattle , Genetic Variation , Humans , INDEL Mutation , Internet , Mice , Rats
15.
Hum Genet ; 139(6-7): 769-776, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32405658

ABSTRACT

Over the last decade next generation sequencing (NGS) has been extensively used to identify new pathogenic mutations and genes causing rare genetic diseases. The efficient analyses of NGS data is not trivial and requires a technically and biologically rigorous pipeline that addresses data quality control, accurate variant filtration to minimize false positives and false negatives, and prioritization of the remaining genes based on disease genomics and physiological knowledge. This review provides a pipeline including all these steps, describes popular software for each step of the analysis, and proposes a general framework for the identification of causal mutations and genes in individual patients of rare genetic diseases.


Subject(s)
Computational Biology/methods , Genes/genetics , Genetic Diseases, Inborn/etiology , Genome, Human , Mutation , Precision Medicine , Rare Diseases/etiology , Genetic Diseases, Inborn/pathology , High-Throughput Nucleotide Sequencing , Humans , Rare Diseases/pathology , Software
16.
Proc Natl Acad Sci U S A ; 114(4): E514-E523, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28069966

ABSTRACT

Most members of the Toll-like receptor (TLR) and interleukin-1 receptor (IL-1R) families transduce signals via a canonical pathway involving the MyD88 adapter and the interleukin-1 receptor-associated kinase (IRAK) complex. This complex contains four molecules, including at least two (IRAK-1 and IRAK-4) active kinases. In mice and humans, deficiencies of IRAK-4 or MyD88 abolish most TLR (except for TLR3 and some TLR4) and IL-1R signaling in both leukocytes and fibroblasts. TLR and IL-1R responses are weak but not abolished in mice lacking IRAK-1, whereas the role of IRAK-1 in humans remains unclear. We describe here a boy with X-linked MECP2 deficiency-related syndrome due to a large de novo Xq28 chromosomal deletion encompassing both MECP2 and IRAK1 Like many boys with MECP2 null mutations, this child died very early, at the age of 7 mo. Unlike most IRAK-4- or MyD88-deficient patients, he did not suffer from invasive bacterial diseases during his short life. The IRAK-1 protein was completely absent from the patient's fibroblasts, which responded very poorly to all TLR2/6 (PAM2CSK4, LTA, FSL-1), TLR1/2 (PAM3CSK4), and TLR4 (LPS, MPLA) agonists tested but had almost unimpaired responses to IL-1ß. By contrast, the patient's peripheral blood mononuclear cells responded normally to all TLR1/2, TLR2/6, TLR4, TLR7, and TLR8 (R848) agonists tested, and to IL-1ß. The death of this child precluded long-term evaluations of the clinical consequences of inherited IRAK-1 deficiency. However, these findings suggest that human IRAK-1 is essential downstream from TLRs but not IL-1Rs in fibroblasts, whereas it plays a redundant role downstream from both TLRs and IL-1Rs in leukocytes.


Subject(s)
Fibroblasts/metabolism , Interleukin-1 Receptor-Associated Kinases/deficiency , Toll-Like Receptors/metabolism , Chromosome Deletion , Chromosomes, Human, X/genetics , Humans , Infant , Interleukin-1 Receptor-Associated Kinases/genetics , Leukocytes/metabolism , Male , Methyl-CpG-Binding Protein 2/genetics , Receptors, Interleukin-1/metabolism , Signal Transduction , Toll-Like Receptors/genetics
17.
Am J Hum Genet ; 98(1): 5-21, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26748513

ABSTRACT

Human genes governing innate immunity provide a valuable tool for the study of the selective pressure imposed by microorganisms on host genomes. A comprehensive, genome-wide study of how selective constraints and adaptations have driven the evolution of innate immunity genes is missing. Using full-genome sequence variation from the 1000 Genomes Project, we first show that innate immunity genes have globally evolved under stronger purifying selection than the remainder of protein-coding genes. We identify a gene set under the strongest selective constraints, mutations in which are likely to predispose individuals to life-threatening disease, as illustrated by STAT1 and TRAF3. We then evaluate the occurrence of local adaptation and detect 57 high-scoring signals of positive selection at innate immunity genes, variation in which has been associated with susceptibility to common infectious or autoimmune diseases. Furthermore, we show that most adaptations targeting coding variation have occurred in the last 6,000-13,000 years, the period at which populations shifted from hunting and gathering to farming. Finally, we show that innate immunity genes present higher Neandertal introgression than the remainder of the coding genome. Notably, among the genes presenting the highest Neandertal ancestry, we find the TLR6-TLR1-TLR10 cluster, which also contains functional adaptive variation in Europeans. This study identifies highly constrained genes that fulfill essential, non-redundant functions in host survival and reveals others that are more permissive to change-containing variation acquired from archaic hominins or adaptive variants in specific populations-improving our understanding of the relative biological importance of innate immunity pathways in natural conditions.


Subject(s)
Immunity, Innate/genetics , Neanderthals/immunology , Adaptation, Physiological/genetics , Animals , Genome-Wide Association Study , Humans , Neanderthals/genetics , Polymorphism, Single Nucleotide , Selection, Genetic
18.
Bioinformatics ; 34(24): 4307-4309, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30535305

ABSTRACT

Summary: Next-generation sequencing (NGS) generates large amounts of genomic data and reveals about 20 000 genetic coding variants per individual studied. Several mutation damage prediction scores are available to prioritize variants, but there is currently no application to help investigators to determine the relevance of the candidate genes and variants quickly and visually from population genetics data and deleteriousness scores. Here, we present PopViz, a user-friendly, rapid, interactive, mobile-compatible webserver providing a gene-centric visualization of the variants of any human gene, with (i) population-specific minor allele frequencies from the gnomAD population genetic database; (ii) mutation damage prediction scores from CADD, EIGEN and LINSIGHT and (iii) amino-acid positions and protein domains. This application will be particularly useful in investigations of NGS data for new disease-causing genes and variants, by reinforcing or rejecting the plausibility of the candidate genes, and by selecting and prioritizing, the candidate variants for experimental testing. Availability and implementation: PopViz webserver is freely accessible from http://shiva.rockefeller.edu/PopViz/. Supplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Gene Frequency , Genetic Variation , Software , Computational Biology , Genomics , High-Throughput Nucleotide Sequencing , Humans
19.
Proc Natl Acad Sci U S A ; 113(24): 6713-8, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27247391

ABSTRACT

Principal component analysis (PCA), homozygosity rate estimations, and linkage studies in humans are classically conducted through genome-wide single-nucleotide variant arrays (GWSA). We compared whole-exome sequencing (WES) and GWSA for this purpose. We analyzed 110 subjects originating from different regions of the world, including North Africa and the Middle East, which are poorly covered by public databases and have high consanguinity rates. We tested and applied a number of quality control (QC) filters. Compared with GWSA, we found that WES provided an accurate prediction of population substructure using variants with a minor allele frequency > 2% (correlation = 0.89 with the PCA coordinates obtained by GWSA). WES also yielded highly reliable estimates of homozygosity rates using runs of homozygosity with a 1,000-kb window (correlation = 0.94 with the estimates provided by GWSA). Finally, homozygosity mapping analyses in 15 families including a single offspring with high homozygosity rates showed that WES provided 51% less genome-wide linkage information than GWSA overall but 97% more information for the coding regions. At the genome-wide scale, 76.3% of linked regions were found by both GWSA and WES, 17.7% were found by GWSA only, and 6.0% were found by WES only. For coding regions, the corresponding percentages were 83.5%, 7.4%, and 9.1%, respectively. With appropriate QC filters, WES can be used for PCA and adjustment for population substructure, estimating homozygosity rates in individuals, and powerful linkage analyses, particularly in coding regions.


Subject(s)
Consanguinity , Genetic Linkage , Genome-Wide Association Study , Homozygote , Female , Humans , Male , Middle East , North America
20.
Proc Natl Acad Sci U S A ; 113(51): E8277-E8285, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27930337

ABSTRACT

Chronic mucocutaneous candidiasis (CMC) is defined as recurrent or persistent infection of the skin, nails, and/or mucosae with commensal Candida species. The first genetic etiology of isolated CMC-autosomal recessive (AR) IL-17 receptor A (IL-17RA) deficiency-was reported in 2011, in a single patient. We report here 21 patients with complete AR IL-17RA deficiency, including this first patient. Each patient is homozygous for 1 of 12 different IL-17RA alleles, 8 of which create a premature stop codon upstream from the transmembrane domain and have been predicted and/or shown to prevent expression of the receptor on the surface of circulating leukocytes and dermal fibroblasts. Three other mutant alleles create a premature stop codon downstream from the transmembrane domain, one of which encodes a surface-expressed receptor. Finally, the only known missense allele (p.D387N) also encodes a surface-expressed receptor. All of the alleles tested abolish cellular responses to IL-17A and -17F homodimers and heterodimers in fibroblasts and to IL-17E/IL-25 in leukocytes. The patients are currently aged from 2 to 35 y and originate from 12 unrelated kindreds. All had their first CMC episode by 6 mo of age. Fourteen patients presented various forms of staphylococcal skin disease. Eight were also prone to various bacterial infections of the respiratory tract. Human IL-17RA is, thus, essential for mucocutaneous immunity to Candida and Staphylococcus, but otherwise largely redundant. A diagnosis of AR IL-17RA deficiency should be considered in children or adults with CMC, cutaneous staphylococcal disease, or both, even if IL-17RA is detected on the cell surface.


Subject(s)
Bacterial Infections/immunology , Candidiasis/immunology , Mycoses/immunology , Receptors, Interleukin-17/deficiency , Receptors, Interleukin-17/genetics , Alleles , Candida , Cell Membrane , Child , Child, Preschool , Family Health , Female , Fibroblasts/metabolism , Genes, Recessive , Genome-Wide Association Study , HEK293 Cells , Homozygote , Humans , Immunophenotyping , Infant , Infant, Newborn , Interleukin-17/metabolism , Male , Mutation , Open Reading Frames , Pedigree , Receptors, Interleukin-17/metabolism , Skin/microbiology , T-Lymphocytes/cytology
SELECTION OF CITATIONS
SEARCH DETAIL