62 results on '"Yuval Itan"'
Search Results
2. Identifying shared genetic factors underlying epilepsy and congenital heart disease in Europeans
- Author
-
Yiming Wu, Cigdem Sevim Bayrak, Bosi Dong, Shixu He, Peter D. Stenson, David N. Cooper, Lei Chen, and Yuval Itan
- Subjects
Genetics ,Genetics (clinical) - Abstract
Epilepsy (EP) and congenital heart disease (CHD) are two apparently unrelated diseases that nevertheless display substantial mutual comorbidity. Thus, whilst 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 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. Lastly, 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.
- Published
- 2022
- Full Text
- View/download PDF
3. Identifying high-impact variants and genes in exomes of Ashkenazi Jewish inflammatory bowel disease patients
- Author
-
Yiming Wu, Kyle Gettler, Meltem Ece Kars, Mamta Giri, Dalin Li, Cigdem Sevim Bayrak, Peng Zhang, Aayushee Jain, Patrick Maffucci, Ksenija Sabic, Tielman Van Vleck, Girish Nadkarni, Lee A. Denson, Harry Ostrer, Adam P. Levine, Elena R. Schiff, Anthony W. Segal, Subra Kugathasan, Peter D. Stenson, David N. Cooper, L. Philip Schumm, Scott Snapper, Mark J. Daly, Talin Haritunians, Richard H. Duerr, Mark S. Silverberg, John D. Rioux, Steven R. Brant, Dermot P. B. McGovern, Judy H. Cho, and Yuval Itan
- Subjects
Multidisciplinary ,General Physics and Astronomy ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology - Abstract
Inflammatory bowel disease (IBD) is a group of chronic digestive tract inflammatory conditions whose genetic etiology is still poorly understood. The incidence of IBD is particularly high among Ashkenazi Jews. Here, we identify 8 novel and plausible IBD-causing genes from the exomes of 4453 genetically identified Ashkenazi Jewish IBD cases (1734) and controls (2719). Various biological pathway analyses are performed, along with bulk and single-cell RNA sequencing, to demonstrate the likely physiological relatedness of the novel genes to IBD. Importantly, we demonstrate that the rare and high impact genetic architecture of Ashkenazi Jewish adult IBD displays significant overlap with very early onset-IBD genetics. Moreover, by performing biobank phenome-wide analyses, we find that IBD genes have pleiotropic effects that involve other immune responses. Finally, we show that polygenic risk score analyses based on genome-wide high impact variants have high power to predict IBD susceptibility.
- Published
- 2023
- Full Text
- View/download PDF
4. Identification of discriminative gene-level and protein-level features associated with pathogenic gain-of-function and loss-of-function variants
- Author
-
Cigdem Sevim Bayrak, Avner Schlessinger, Satoshi Okada, Peter D. Stenson, Girish N. Nadkarni, Tielman Van Vleck, David Neil Cooper, Kumardeep Chaudhary, David Stein, Stéphanie Boisson-Dupuis, Aayushee Jain, Anne Puel, and Yuval Itan
- Subjects
Genome, Human ,Inheritance (genetic algorithm) ,Proteins ,Computational biology ,Cloud Computing ,Biology ,Gene mutation ,Phenotype ,Article ,Germline ,Machine Learning ,Discriminative model ,Loss of Function Mutation ,Gain of Function Mutation ,Databases, Genetic ,Genetics ,Humans ,Genetic Predisposition to Disease ,Identification (biology) ,Gene ,Germ-Line Mutation ,Internet-Based Intervention ,Genetics (clinical) ,Loss function - Abstract
Summary 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.
- Published
- 2021
- Full Text
- View/download PDF
5. Impact of SARS-CoV-2 infection and COVID-19 on patients with inborn errors of immunity
- Author
-
Stuart G. Tangye, Laurent Abel, Salah Al-Muhsen, Alessandro Aiuti, Saleh Al-Muhsen, Fahd Al-Mulla, Mark S. Anderson, Evangelos Andreakos, Antonio Novelli, Andrés A. Arias, Hagit Baris Feldman, Alexandre Belot, Catherine M. Biggs, Ahmed A. Bousfiha, Petter Brodin, John Christodoulou, Antonio Condino-Neto, Clifton L. Dalgard, Sara Espinosa-Padilla, Jacques Fellay, Carlos Flores, José Luis Franco, Antoine Froidure, Filomeen Haerynck, Rabih Halwani, Lennart Hammarström, Sarah E. Henrickson, Elena W.Y. Hsieh, Yuval Itan, Timokratis Karamitros, Yu-Lung Lau, Davood Mansouri, Isabelle Meyts, Trine H. Mogensen, Tomohiro Morio, Lisa F.P. Ng, Luigi D. Notarangelo, Giuseppe Novelli, Satoshi Okada, Tayfun Ozcelik, Qiang Pan-Hammarström, Rebeca Perez de Diego, Carolina Prando, Aurora Pujol, Laurent Renia, Igor Resnick, Carlos Rodríguez-Gallego, Vanessa Sancho-Shimizu, Mikko R.J. Seppänen, Anna Shcherbina, Andrew L. Snow, Pere Soler-Palacín, András N. Spaan, Ivan Tancevski, Ahmad Abou Tayoun, Sehime G. Temel, Stuart E. Turvey, Mohammed J. Uddin, Donald C. Vinh, Mayana Zatz, Keisuke Okamoto, David S. Pelin, Graziano Pesole, Diederik van de Beek, Roger Colobran, Joost Wauters, Helen C. Su, and Jean-Laurent Casanova
- Subjects
Settore MED/03 ,inborn errors of immunity ,SARS-CoV-2 ,immune dysregulation ,Immunology ,cytokine storm ,primary immune deficiencies ,Immunology and Allergy ,COVID-19 ,type I IFN signaling - Abstract
Since the arrival of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in December 2019, its characterization as a novel human pathogen, and the resulting coronavirus disease 2019 (COVID-19) pandemic, over 6.5 million people have died worldwide—a stark and sobering reminder of the fundamental and nonredundant roles of the innate and adaptive immune systems in host defense against emerging pathogens. Inborn errors of immunity (IEI) are caused by germline variants, typically in single genes. IEI are characterized by defects in development and/or function of cells involved in immunity and host defense, rendering individuals highly susceptible to severe, recurrent, and sometimes fatal infections, as well as immune dysregulatory conditions such as autoinflammation, autoimmunity, and allergy. The study of IEI has revealed key insights into the molecular and cellular requirements for immune-mediated protection against infectious diseases. Indeed, this has been exemplified by assessing the impact of SARS-CoV-2 infection in individuals with previously diagnosed IEI, as well as analyzing rare cases of severe COVID-19 in otherwise healthy individuals. This approach has defined fundamental aspects of mechanisms of disease pathogenesis, immunopathology in the context of infection with a novel pathogen, and therapeutic options to mitigate severe disease. This review summarizes these findings and illustrates how the study of these rare experiments of nature can inform key features of human immunology, which can then be leveraged to improve therapies for treating emerging and established infectious diseases.
- Published
- 2022
6. A computational approach for detecting physiological homogeneity in the midst of genetic heterogeneity
- Author
-
Peng Zhang, Shen-Ying Zhang, Laurent Abel, Clémentine Boccon-Gibod, Yuval Itan, Jean-Laurent Casanova, Burkhard Stüve, Cigdem Sevim Bayrak, Lazaro Lorenzo, Louis Vallée, Daniela Matuozzo, Yoon-Seung Lee, Soraya Boucherit, Yiming Wu, Aurélie Cobat, Aayushee Jain, and Stéphane Chabrier
- Subjects
Candidate gene ,Computational biology ,Biology ,Article ,DNA sequencing ,Genetic Heterogeneity ,03 medical and health sciences ,0302 clinical medicine ,Exome Sequencing ,Gene cluster ,Genetics ,Humans ,Gene Regulatory Networks ,Genetic Predisposition to Disease ,Gene ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,Genetic heterogeneity ,Computational Biology ,Fibroblasts ,Phenotype ,Penetrance ,Toll-Like Receptor 3 ,Case-Control Studies ,Encephalitis, Herpes Simplex ,030217 neurology & neurosurgery ,Biological network - 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/β 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.
- Published
- 2021
- Full Text
- View/download PDF
7. A comprehensive knowledgebase of known and predicted human genetic variants associated with COVID-19 susceptibility and severity
- Author
-
Meltem Ece Kars, David Stein, Çiğdem Sevim Bayrak, Peter D Stenson, David N Cooper, and Yuval Itan
- Subjects
Immunology ,Immunology and Allergy - Abstract
Host genetic susceptibility is a key risk factor for severe illness associated with COVID-19. Despite numerous studies of COVID-19 host genetics, our knowledge of COVID-19-associated variants is still limited, and there is no resource comprising all the published variants and categorizing them based on their confidence level. Also, there are currently no computational tools available to predict novel COVID-19 severity variants. Therefore, we collated 820 host genetic variants reported to affect COVID-19 susceptibility by means of a systematic literature search and confidence evaluation, and obtained 196 high-confidence variants. We then developed the first machine learning classifier of severe COVID-19 variants to perform a genome-wide prediction of COVID-19 severity for 82,468,698 missense variants in the human genome. We further evaluated the classifier’s predictions using feature importance analyses to investigate the biological properties of COVID-19 susceptibility variants, which identified conservation scores as the most impactful predictive features. The results of enrichment analyses revealed that genes carrying high-confidence COVID-19 susceptibility variants shared pathways, networks, diseases and biological functions, with the immune system and infectious disease being the most significant categories. Additionally, we investigated the pleiotropic effects of COVID-19-associated variants using phenome-wide association studies (PheWAS) in ∼40,000 BioMe BioBank genotyped individuals, revealing pre-existing conditions that could serve to increase the risk of severe COVID-19 such as chronic liver disease and thromboembolism. Lastly, we generated a web-based interface for exploring, downloading and submitting genetic variants associated with COVID-19 susceptibility for use in both research and clinical settings (https://itanlab.shinyapps.io/COVID19webpage/). Taken together, our work provides the most comprehensive COVID-19 host genetics knowledgebase to date for the known and predicted genetic determinants of severe COVID-19, a resource that should further contribute to our understanding of the biology underlying COVID-19 susceptibility and facilitate the identification of individuals at high risk for severe COVID-19.
- Published
- 2023
- Full Text
- View/download PDF
8. Inflamed Ulcerative Colitis Regions Associated With MRGPRX2-Mediated Mast Cell Degranulation and Cell Activation Modules, Defining a New Therapeutic Target
- Author
-
Yuval Itan, Mark S. Silverberg, Steven R. Brant, Ujunwa M. Korie, Aayushee Jain, Inga Peter, Shikha Nayar, Ling-Shiang Chuang, Talin Haritunians, Ksenija Sabic, John D. Rioux, Gerardus Bongers, Nicole Villaverde, Colleen C. Chasteau, Arden Moscati, Richard H. Duerr, Ernie Chen, Nai Yun Hsu, Tin Htwe Thin, Judy H. Cho, Sari Joshowitz, Isaac L. Alter, Siarhei Dzedzik, Zhi Chai, Mamta Giri, Kyle Gettler, L. Philip Schumm, and Dermot P.B. McGovern
- Subjects
Receptors, Neuropeptide ,0301 basic medicine ,Colon ,Inositol Phosphates ,Cell ,Nerve Tissue Proteins ,Inflammation ,CHO Cells ,Biology ,Ligands ,Article ,Cell Degranulation ,Receptors, G-Protein-Coupled ,Adrenomedullin ,03 medical and health sciences ,Cricetulus ,0302 clinical medicine ,medicine ,Animals ,Humans ,Mast Cells ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Hepatology ,Chinese hamster ovary cell ,Gastroenterology ,Degranulation ,Genetic Variation ,Transfection ,Mast cell ,beta-Arrestin 2 ,030104 developmental biology ,medicine.anatomical_structure ,Case-Control Studies ,Cancer research ,Colitis, Ulcerative ,030211 gastroenterology & hepatology ,Tumor necrosis factor alpha ,medicine.symptom ,Cell activation - 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 β-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 β-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.
- Published
- 2021
- Full Text
- View/download PDF
9. Sudden Cardiac Arrest in a Patient With Mitral Valve Prolapse and LMNA and SCN5A Mutations
- Author
-
Lior Jankelson, Larry A. Chinitz, Marina Cerrone, Asha M. Mahajan, Yuval Itan, Linda Borneman, and James M. Horowitz
- Subjects
mitral valve ,0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,CADD, Combined Annotation Dependent Depletion ,030105 genetics & heredity ,LMNA ,03 medical and health sciences ,0302 clinical medicine ,Mitral valve ,Internal medicine ,medicine ,Diseases of the circulatory (Cardiovascular) system ,Mitral valve prolapse ,Mini-Focus Issue: Imaging ,genetic disorders ,VF - Ventricular fibrillation ,business.industry ,MVP - Mitral valve prolapse ,Sudden cardiac arrest ,MSC, mutation significance cutoff ,ventricular fibrillation ,medicine.disease ,MAD, mitral annular disjunction ,LV, left ventricle ,medicine.anatomical_structure ,Increased risk ,MVP, mitral valve prolapse ,RC666-701 ,Ventricular fibrillation ,cardiovascular system ,Cardiology ,ECG, electrocardiogram ,Case Report: Clinical Case ,VF, ventricular fibrillation ,QTc, corrected QT ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,MRI, magnetic resonance imaging ,030217 neurology & neurosurgery - Abstract
Bileaflet mitral valve prolapse (Bi-MVP) is associated with increased risk for cardiac arrest. We describe a patient who presented after a cardiac arrest with Bi-MVP and variants in Lamin A/C (LMNA) and the sodium channel alpha-subunit 5a (SCN5A). Genetic variants may be the culprit for arrhythmogenesis in Bi-MVP patients. (Level of Difficulty: Intermediate.), Central Illustration
- Published
- 2021
- Full Text
- View/download PDF
10. Genome-wide prediction of pathogenic gain- and loss-of-function variants from ensemble learning of a diverse feature set
- Author
-
David Stein, Çiğdem Sevim Bayrak, Yiming Wu, Meltem Ece Kars, Peter D. Stenson, David N. Cooper, Avner Schlessinger, and Yuval Itan
- Abstract
Gain-of-function (GOF) variants give rise to increased or novel protein functions whereas loss-of-function (LOF) variants lead to diminished protein function. GOF and LOF variants can result in markedly varying phenotypes, even when occurring in the same gene. However, experimental approaches for identifying GOF and LOF are generally slow and costly, whilst currently available computational methods have not been optimized to discriminate between GOF and LOF variants. We have developed LoGoFunc, an ensemble machine learning method for predicting pathogenic GOF, pathogenic LOF, and neutral genetic variants. LoGoFunc was trained on a broad range of gene-, protein-, and variant-level features describing diverse biological characteristics, as well as network features summarizing the protein-protein interactome and structural features calculated from AlphaFold2 protein models. We analyzed GOF, LOF, and neutral variants in terms of local protein structure and function, splicing disruption, and phenotypic associations, thereby revealing previously unreported relationships between various biological phenomena and variant functional outcomes. For example, GOF and LOF variants exhibit contrasting enrichments in protein structural and functional regions, whilst LOF variants are more likely to disrupt canonical splicing as indicated by splicing-related features employed by the model. Further, by performing phenome-wide association studies (PheWAS), we identified strong associations between relevant phenotypes and high-confidence predicted GOF and LOF variants. LoGoFunc outperforms other tools trained solely to predict pathogenicity or general variant impact for the identification of pathogenic GOF and LOF variants.
- Published
- 2022
- Full Text
- View/download PDF
11. Human CRY1 variants associate with attention deficit/hyperactivity disorder
- Author
-
I Halil Kavaklı, Arianna Goracci, Yuval Itan, Ayse Ozhan, Chiara Fallerini, Jean-Laurent Casanova, Cihan Aydin, M Ece Kars, O Emre Onat, Yiming Wu, Cem Atbaşoğlu, A. Nazli Basak, Kaya Bilguvar, Alessandra Renieri, Tayfun Ozcelik, M Allegra Trusso, Meram Can Saka, Seref Gul, Gül, Şeref, Aydın, Cihan (ORCID 0000-0003-0560-1895 & YÖK ID 214696), Başak, Ayşe Nazlı (ORCID 0000-0001-9257-3540 & YÖK ID 1512), Kavaklı, İbrahim Halil (ORCID 0000-0001-6624-3505 & YÖK ID 40319), Onat, O. Emre, Kars, M. Ece, Bilguvar, Kaya, Wu, Yiming, Özhan, Ayşe, Trusso, M. Allegra, Goracci, Arianna, Fallerini, Chiara, Renieri, Alessandra, Casanova, Jean Laurent, Itan, Yuval, Atbaşoğlu, Cem E., Saka, Meram C., Özçelik, Tayfun, Koç University Research Center for Translational Medicine (KUTTAM) / Koç Üniversitesi Translasyonel Tıp Araştırma Merkezi (KUTTAM), Graduate School of Sciences and Engineering, College of Engineering, College of Sciences, Department of Chemical and Biological Engineering, Department of Molecular Biology and Genetics, and Başak, A. N.
- Subjects
Adult ,Male ,0301 basic medicine ,Genetic diseases ,Genetics ,Monogenic diseases ,Psychiatric diseases ,CLOCK Proteins ,Delayed sleep phase ,Bioinformatics ,ARNTL Transcription Factors ,Attention Deficit Disorder with Hyperactivity ,Cryptochromes ,Female ,Genetic Association Studies ,HEK293 Cells ,Humans ,Sleep Disorders, Circadian Rhythm ,Mutation ,03 medical and health sciences ,0302 clinical medicine ,mental disorders ,Insomnia ,Medicine ,Attention deficit hyperactivity disorder ,Circadian rhythm ,Exome sequencing ,Depression (differential diagnoses) ,business.industry ,General Medicine ,medicine.disease ,Circadian Rhythm ,Biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Anxiety ,Major depressive disorder ,medicine.symptom ,Sleep Disorders ,business ,Research Article - Abstract
Attention deficit/hyperactivity disorder (ADHD) is a common and heritable phenotype frequently accompanied by insomnia, anxiety, and depression. Here, using a reverse phenotyping approach, we report heterozygous coding variations in the core circadian clock gene cryptochrome 1 in 15 unrelated multigenerational families with combined ADHD and insomnia. The variants led to functional alterations in the circadian molecular rhythms, providing a mechanistic link to the behavioral symptoms. One variant, CRY1Δ11 c.1657+3A>C, is present in approximately 1% of Europeans, therefore standing out as a diagnostic and therapeutic marker. We showed by exome sequencing in an independent cohort of patients with combined ADHD and insomnia that 8 of 62 patients and 0 of 369 controls carried CRY1Δ11. Also, we identified a variant, CRY1Δ6 c.825+1G>A, that shows reduced affinity for BMAL1/CLOCK and causes an arrhythmic phenotype. Genotype-phenotype correlation analysis revealed that this variant segregated with ADHD and delayed sleep phase disorder (DSPD) in the affected family. Finally, we found in a phenome-wide association study involving 9438 unrelated adult Europeans that CRY1Δ11 was associated with major depressive disorder, insomnia, and anxiety. These results defined a distinctive group of circadian psychiatric phenotypes that we propose to designate as "circiatric" disorders., NIH Clinical and Translational Science Award (CTSA) Program; NIH; French National Research Agency (ANR) under the “Investments for the future” Program; Integrative Biology of Emerging Infectious Diseases Laboratoire d’Excellence; IEIHSEER Grant; SEAe-Host Factors Grant; PNEUMOID Project Grant; INCA/Cancéropole Ile-de-France; Turkish Academy of Sciences (TÜBA); National Center for Advancing Translational Sciences (NCAST); Rockefeller University, INSERM; HHMI, University of Paris; St. Giles Foundation; Charles Bronfman Institute for Personalized Medicine at the Icahn School of Medicine at Mount Sinai
- Published
- 2020
- Full Text
- View/download PDF
12. Identifying disease-causing mutations in genomes of single patients by computational approaches
- Author
-
Yuval Itan and Cigdem Sevim Bayrak
- Subjects
False positives and false negatives ,Genomics ,Computational biology ,Biology ,Genome ,DNA sequencing ,03 medical and health sciences ,Rare Diseases ,Genetics ,Humans ,Precision Medicine ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,Genome, Human ,030305 genetics & heredity ,Genetic Diseases, Inborn ,Computational Biology ,High-Throughput Nucleotide Sequencing ,Precision medicine ,Human genetics ,Genes ,Mutation ,Mutation (genetic algorithm) ,Identification (biology) ,Software - 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.
- Published
- 2020
- Full Text
- View/download PDF
13. VIPPID: a gene-specific single nucleotide variant pathogenicity prediction tool for primary immunodeficiency diseases
- Author
-
Mingyan Fang, Zheng Su, Hassan Abolhassani, Yuval Itan, Xin Jin, and Lennart Hammarström
- Subjects
Virulence ,Nucleotides ,Primary Immunodeficiency Diseases ,Humans ,Molecular Biology ,Polymorphism, Single Nucleotide ,Algorithms ,Information Systems - 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.
- Published
- 2022
14. The genetic structure of the Turkish population reveals high levels of variation and admixture
- Author
-
Jean-Laurent Casanova, A. Nazli Basak, David Neil Cooper, Jungmin Choi, M Ece Kars, Peter D. Stenson, Hakan Buluş, Robin Palvadeau, Alper Yavuz, Caner Çağlar, O Emre Onat, Kaya Bilguvar, Tayfun Ozcelik, Yuval Itan, Murat Gunel, Jeffrey M. Friedman, Kars, Meltem Ece, Onat, Onur Emre, Özçelik, Tayfun, Acibadem University Dspace, and ÇAĞLAR, CANER
- Subjects
Genotype ,Turkey ,Human Migration ,Population ,Variation ,Admixture ,Biology ,Consanguinity ,Gene Frequency ,Exome Sequencing ,Turkish Variome ,Sequencing ,Humans ,Exome ,Allele ,1000 Genomes Project ,education ,Allele frequency ,Alleles ,Genetic association ,Genetics ,education.field_of_study ,Multidisciplinary ,Genome, Human ,Genetic Drift ,population genetics ,Genetic Variation ,Correction ,sequencing ,Variome ,Genetics, Population ,Haplotypes ,Genetic structure ,admixture ,Sopulation genetics ,variation ,Genome-Wide Association Study - 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.
- Published
- 2021
15. SARS-CoV-2 susceptibility and COVID-19 disease severity are associated with genetic variants affecting gene expression in a variety of tissues
- Author
-
Matteo D’Antonio, Jennifer P. Nguyen, Timothy D. Arthur, Hiroko Matsui, Agnieszka D’Antonio-Chronowska, Kelly A. Frazer, Benjamin M. Neale, Mark Daly, Andrea Ganna, Christine Stevens, Gita A. Pathak, Shea J. Andrews, Masahiro Kanai, Mattia Cordioli, Juha Karjalainen, Renato Polimanti, Matti Pirinen, Nadia Harerimana, Kumar Veerapen, Brooke Wolford, Huy Nguyen, Matthew Solomonson, Rachel G. Liao, Karolina Chwialkowska, Amy Trankiem, Mary K. Balaconis, Caroline Hayward, Anne Richmond, Archie Campbell, Marcela Morris, Chloe Fawns-Ritchie, Joseph T. Glessner, Douglas M. Shaw, Xiao Chang, Hannah Polikowski, Petty E. Lauren, Hung-Hsin Chen, Zhu Wanying, Hakon Hakonarson, David J. Porteous, Jennifer Below, Kari North, Joseph B. McCormick, Paul R.H.J. Timmers, James F. Wilson, Albert Tenesa, Kenton D’Mellow, Shona M. Kerr, Mari E.K. Niemi, Lindokuhle Nkambul, Kathrin Aprile von Hohenstaufen, Ali Sobh, Madonna M. Eltoukhy, Amr M. Yassen, Mohamed A.F. Hegazy, Kamal Okasha, Mohammed A. Eid, Hanteera S. Moahmed, Doaa Shahin, Yasser M. El-Sherbiny, Tamer A. Elhadidy, Mohamed S. Abd Elghafar, Jehan J. El-Jawhari, Attia A.S. Mohamed, Marwa H. Elnagdy, Amr Samir, Mahmoud Abdel-Aziz, Walid T. Khafaga, Walaa M. El-Lawaty, Mohamed S. Torky, Mohamed R. El-shanshory, Chiara Batini, Paul H. Lee, Nick Shrine, Alexander T. Williams, Martin D. Tobin, Anna L. Guyatt, Catherine John, Richard J. Packer, Altaf Ali, Robert C. Free, Xueyang Wang, Louise V. Wain, Edward J. Hollox, Laura D. Venn, Catherine E. Bee, Emma L. Adams, Ahmadreza Niavarani, Bahareh Sharififard, Rasoul Aliannejad, Ali Amirsavadkouhi, Zeinab Naderpour, Hengameh Ansari Tadi, Afshar Etemadi Aleagha, Saeideh Ahmadi, Seyed Behrooz Mohseni Moghaddam, Alireza Adamsara, Morteza Saeedi, Hamed Abdollahi, Abdolmajid Hosseini, Pajaree Chariyavilaskul, Monpat Chamnanphon, Thitima B. Suttichet, Vorasuk Shotelersuk, Monnat Pongpanich, Chureerat Phokaew, Wanna Chetruengchai, Watsamon Jantarabenjakul, Opass Putchareon, Pattama Torvorapanit, Thanyawee Puthanakit, Pintip Suchartlikitwong, Nattiya Hirankarn, Voraphoj Nilaratanakul, Pimpayao Sodsai, Ben M. Brumpton, Kristian Hveem, Cristen Willer, Wei Zhou, Tormod Rogne, Erik Solligard, Bjørn Olav Åsvold, Malak Abedalthagafi, Manal Alaamery, Saleh Alqahtani, Dona Baraka, Fawz Al Harthi, Ebtehal Alsolm, Leen Abu Safieh, Albandary M. Alowayn, Fatimah Alqubaishi, Amal Al Mutairi, Serghei Mangul, Abdulraheem Alshareef, Mona Sawaji, Mansour Almutairi, Nora Aljawini, Nour Albesher, Yaseen M. Arabi, Ebrahim S. Mahmoud, Amin K. Khattab, Roaa T. Halawani, Ziab Z. Alahmadey, Jehad K. Albakri, Walaa A. Felemban, Bandar A. Suliman, Rana Hasanato, Laila Al-Awdah, Jahad Alghamdi, Deema AlZahrani, Sameera AlJohani, Hani Al-Afghani, May Alrashed, Nouf AlDhawi, Hadeel AlBardis, Sarah Alkwai, Moneera Alswailm, Faisal Almalki, Maha Albeladi, Iman Almohammed, Eman Barhoush, Anoud Albader, Salam Massadeh, Abdulaziz AlMalik, Sara Alotaibi, Bader Alghamdi, Junghyun Jung, Mohammad S. Fawzy, Yunsung Lee, Per Magnus, Lill-Iren S. Trogstad, Øyvind Helgeland, Jennifer R. Harris, Massimo Mangino, Tim D. Spector, Duncan Emma, Sandra P. Smieszek, Bartlomiej P. Przychodzen, Christos Polymeropoulos, Vasilios Polymeropoulos, Mihael H. Polymeropoulos, Israel Fernandez-Cadenas, Jordi Perez-Tur, Laia Llucià-Carol, Natalia Cullell, Elena Muiño, Jara Cárcel-Márquez, Marta L. DeDiego, Lara Lloret Iglesias, Anna M. Planas, Alex Soriano, Veronica Rico, Daiana Agüero, Josep L. Bedini, Francisco Lozano, Carlos Domingo, Veronica Robles, Francisca Ruiz-Jaén, Leonardo Márquez, Juan Gomez, Eliecer Coto, Guillermo M. Albaiceta, Marta García-Clemente, David Dalmau, Maria J. Arranz, Beatriz Dietl, Alex Serra-Llovich, Pere Soler, Roger Colobrán, Andrea Martín-Nalda, Alba Parra Martínez, David Bernardo, Silvia Rojo, Aida Fiz-López, Elisa Arribas, Paloma de la Cal-Sabater, Tomás Segura, Esther González-Villa, Gemma Serrano-Heras, Joan Martí-Fàbregas, Elena Jiménez-Xarrié, Alicia de Felipe Mimbrera, Jaime Masjuan, Sebastian García-Madrona, Anna Domínguez-Mayoral, Joan Montaner Villalonga, Paloma Menéndez-Valladares, Daniel I. Chasman, Julie E. Buring, Paul M. Ridker, Giulianini Franco, Howard D. Sesso, JoAnn E. Manson, Joseph R. Glessner, Carolina Medina-Gomez, Andre G. Uitterlinden, M. Arfan Ikram, Kati Kristiansson, Sami Koskelainen, Markus Perola, Kati Donner, Katja Kivinen, Aarno Palotie, Samuli Ripatti, Sanni Ruotsalainen, Mari Kaunisto, null FinnGen, Tomoko Nakanishi, Guillaume Butler-Laporte, Vincenzo Forgetta, David R. Morrison, Biswarup Ghosh, Laetitia Laurent, Alexandre Belisle, Danielle Henry, Tala Abdullah, Olumide Adeleye, Noor Mamlouk, Nofar Kimchi, Zaman Afrasiabi, Nardin Rezk Branka Vulesevic, Meriem Bouab, Charlotte Guzman, Louis Petitjean, Chris Tselios, Xiaoqing Xue, Erwin Schurr, Jonathan Afilalo, Marc Afilalo, Maureen Oliveira, Bluma Brenner, Pierre Lepage, Jiannis Ragoussis, Daniel Auld, Nathalie Brassard, Madeleine Durand, Michaël Chassé, Daniel E. Kaufmann, G. Mark Lathrop, Vincent Mooser, J. Brent Richards, Rui Li, Darin Adra, Souad Rahmouni, Michel Georges, Michel Moutschen, Benoit Misset, Gilles Darcis, Julien Guiot, Julien Guntz, Samira Azarzar, Stéphanie Gofflot, Yves Beguin, Sabine Claassen, Olivier Malaise, Pascale Huynen, Christelle Meuris, Marie Thys, Jessica Jacques, Philippe Léonard, Frederic Frippiat, Jean-Baptiste Giot, Anne-Sophie Sauvage, Christian Von Frenckell, Yasmine Belhaj, Bernard Lambermont, Sara Pigazzini, Lindokuhle Nkambule, Michelle Daya, Jonathan Shortt, Nicholas Rafaels, Stephen J. Wicks, Kristy Crooks, Kathleen C. Barnes, Christopher R. Gignoux, Sameer Chavan, Triin Laisk, Kristi Läll, Maarja Lepamets, Reedik Mägi, Tõnu Esko, Ene Reimann, Lili Milani, Helene Alavere, Kristjan Metsalu, Mairo Puusepp, Andres Metspalu, Paul Naaber, Edward Laane, Jaana Pesukova, Pärt Peterson, Kai Kisand, Jekaterina Tabri, Raili Allos, Kati Hensen, Joel Starkopf, Inge Ringmets, Anu Tamm, Anne Kallaste, Pierre-Yves Bochud, Carlo Rivolta, Stéphanie Bibert, Mathieu Quinodoz, Dhryata Kamdar, Noémie Boillat, Semira Gonseth Nussle, Werner Albrich, Noémie Suh, Dionysios Neofytos, Véronique Erard, Cathy Voide, null FHoGID, null RegCOVID, null P-PredictUs, null SeroCOVID, null CRiPSI, Rafael de Cid, Iván Galván-Femenía, Natalia Blay, Anna Carreras, Beatriz Cortés, Xavier Farré, Lauro Sumoy, Victor Moreno, Josep Maria Mercader, Marta Guindo-Martinez, David Torrents, Manolis Kogevinas, Judith Garcia-Aymerich, Gemma Castaño-Vinyals, Carlota Dobaño, Alessandra Renieri, Francesca Mari, Chiara Fallerini, Sergio Daga, Elisa Benetti, Margherita Baldassarri, Francesca Fava, Elisa Frullanti, Floriana Valentino, Gabriella Doddato, Annarita Giliberti, Rossella Tita, Sara Amitrano, Mirella Bruttini, Susanna Croci, Ilaria Meloni, Maria Antonietta Mencarelli, Caterina Lo Rizzo, Anna Maria Pinto, Giada Beligni, Andrea Tommasi, Laura Di Sarno, Maria Palmieri, Miriam Lucia Carriero, Diana Alaverdian, Stefano Busani, Raffaele Bruno, Marco Vecchia, Mary Ann Belli, Nicola Picchiotti, Maurizio Sanarico, Marco Gori, Simone Furini, Stefania Mantovani, Serena Ludovisi, Mario Umberto Mondelli, Francesco Castelli, Eugenia Quiros-Roldan, Melania Degli Antoni, Isabella Zanella, Massimo Vaghi, Stefano Rusconi, Matteo Siano, Francesca Montagnani, Arianna Emiliozzi, Massimiliano Fabbiani, Barbara Rossetti, Elena Bargagli, Laura Bergantini, Miriana D’Alessandro, Paolo Cameli, David Bennett, Federico Anedda, Simona Marcantonio, Sabino Scolletta, Federico Franchi, Maria Antonietta Mazzei, Susanna Guerrini, Edoardo Conticini, Luca Cantarini, Bruno Frediani, Danilo Tacconi, Chiara Spertilli, Marco Feri, Alice Donati, Raffaele Scala, Luca Guidelli, Genni Spargi, Marta Corridi, Cesira Nencioni, Leonardo Croci, Maria Bandini, Gian Piero Caldarelli, Paolo Piacentini, Elena Desanctis, Silvia Cappelli, Anna Canaccini, Agnese Verzuri, Valentina Anemoli, Agostino Ognibene, Alessandro Pancrazzi, Maria Lorubbio, Antonella D’Arminio Monforte, Federica Gaia Miraglia, Massimo Girardis, Sophie Venturelli, Andrea Cossarizza, Andrea Antinori, Alessandra Vergori, Arianna Gabrieli, Agostino Riva, Daniela Francisci, Elisabetta Schiaroli, Francesco Paciosi, Pier Giorgio Scotton, Francesca Andretta, Sandro Panese, Renzo Scaggiante, Francesca Gatti, Saverio Giuseppe Parisi, Stefano Baratti, Matteo Della Monica, Carmelo Piscopo, Mario Capasso, Roberta Russo, Immacolata Andolfo, Achille Iolascon, Giuseppe Fiorentino, Massimo Carella, Marco Castori, Giuseppe Merla, Gabriella Maria Squeo, Filippo Aucella, Pamela Raggi, Carmen Marciano, Rita Perna, Matteo Bassetti, Antonio Di Biagio, Maurizio Sanguinetti, Luca Masucci, Serafina Valente, Marco Mandalà, Alessia Giorli, Lorenzo Salerni, Patrizia Zucchi, Pierpaolo Parravicini, Elisabetta Menatti, Tullio Trotta, Ferdinando Giannattasio, Gabriella Coiro, Fabio Lena, Domenico A. Coviello, Cristina Mussini, Enrico Martinelli, Sandro Mancarella, Luisa Tavecchia, Lia Crotti, Chiara Gabbi, Marco Rizzi, Franco Maggiolo, Diego Ripamonti, Tiziana Bachetti, Maria Teresa La Rovere, Simona Sarzi-Braga, Maurizio Bussotti, Stefano Ceri, Pietro Pinoli, Francesco Raimondi, Filippo Biscarini, Alessandra Stella, Kristina Zguro, Katia Capitani, Claudia Suardi, Simona Dei, Gianfranco Parati, Sabrina Ravaglia, Rosangela Artuso, Giordano Bottà, Paolo Di Domenico, Ilaria Rancan, Antonio Perrella Francesco Bianchi, Davide Romani, Paola Bergomi, Emanuele Catena, Riccardo Colombo, Marco Tanfoni, Antonella Vincenti, Claudio Ferri, Davide Grassi, Gloria Pessina, Mario Tumbarello, Massimo Di Pietro, Ravaglia Sabrina, Sauro Luchi, Chiara Barbieri, Donatella Acquilini, Elena Andreucci, Francesco Vladimiro Segala, Giusy Tiseo, Marco Falcone, Mirjam Lista, Monica Poscente, Oreste De Vivo, Paola Petrocelli, Alessandra Guarnaccia, Silvia Baroni, Albert V. Smith, Andrew P. Boughton, Kevin W. Li, Jonathon LeFaive, Aubrey Annis, Anne E. Justice, Tooraj Mirshahi, Geetha Chittoor, Navya Shilpa Josyula, Jack A. Kosmicki, Manuel A.R. Ferreira, Joseph B. Leader, Dave J. Carey, Matthew C. Gass, Julie E. Horowitz, Michael N. Cantor, Ashish Yadav, Aris Baras, Goncalo R. Abecasis, David A. van Heel, Karen A. Hunt, Dan Mason, Qin Qin Huang, Sarah Finer, null Genes & Health Research Team, Bhavi Trivedi, Christopher J. Griffiths, Hilary C. Martin, John Wright, Richard C. Trembath, Nicole Soranzo, Jing Hua Zhao, Adam S. Butterworth, John Danesh, Emanuele Di Angelantonio, Lude Franke Marike Boezen, Patrick Deelen, Annique Claringbould, Esteban Lopera, Robert Warmerdam, Judith.M. Vonk, Irene van Blokland, Pauline Lanting, Anil P.S. Ori, Brooke Wolford Sebastian Zöllner, Jiongming Wang, Andrew Beck, Gina Peloso, Yuk-Lam Ho, Yan V. Sun, Jennifer E. Huffman, Christopher J. O’Donnell, Kelly Cho, Phil Tsao, J. Michael Gaziano, Michel (M.G.) Nivard, Eco (E.J.C.) de geus, Meike Bartels, Jouke Jan Hottenga, Scott T. Weiss, Elizabeth W. Karlson, Jordan W. Smoller, Robert C. Green, Yen-Chen Anne Feng, Josep Mercader, Shawn N. Murphy, James B. Meigs, Ann E. Woolley, Emma F. Perez, Daniel Rader, Anurag Verma, Marylyn D. Ritchie, Binglan Li, Shefali S. Verma, Anastasia Lucas, Yuki Bradford, Hugo Zeberg, Robert Frithiof, Michael Hultström, Miklos Lipcsey, Lindo Nkambul, Nicolas Tardif, Olav Rooyackers, Jonathan Grip, Tomislav Maricic, Konrad J. Karczewski, Elizabeth G. Atkinson, Kristin Tsuo, Nikolas Baya, Patrick Turley, Rahul Gupta, Shawneequa Callier, Raymond K. Walters, Duncan S. Palmer, Gopal Sarma, Nathan Cheng, Wenhan Lu, Sam Bryant, Claire Churchhouse, Caroline Cusick, Jacqueline I. Goldstein, Daniel King, Cotton Seed, Hilary Finucane, Alicia R. Martin, F. Kyle Satterstrom, Daniel J. Wilson, Jacob Armstrong, Justine K. Rudkin, Gavin Band, Sarah G. Earle, Shang-Kuan Lin, Nicolas Arning, Derrick W. Crook, David H. Wyllie, Anne Marie O’Connell, Chris C.A. Spencer, Nils Koelling, Mark J. Caulfield, Richard H. Scott, Tom Fowler, Loukas Moutsianas, Athanasios Kousathanas, Dorota Pasko, Susan Walker, Augusto Rendon, Alex Stuckey, Christopher A. Odhams, Daniel Rhodes, Georgia Chan, Prabhu Arumugam, Catherine A. Ball, Eurie L. Hong, Kristin Rand, Ahna Girshick, Harendra Guturu, Asher Haug Baltzell, Genevieve Roberts, Danny Park, Marie Coignet, Shannon McCurdy, Spencer Knight, Raghavendran Partha, Brooke Rhead, Miao Zhang, Nathan Berkowitz, Michael Gaddis, Keith Noto, Luong Ruiz, Milos Pavlovic, Laura G. Sloofman, Alexander W. Charney, Noam D. Beckmann, Eric E. Schadt, Daniel M. Jordan, Ryan C. Thompson, Kyle Gettler, Noura S. Abul-Husn, Steven Ascolillo, Joseph D. Buxbaum, Kumardeep Chaudhary, Judy H. Cho, Yuval Itan, Eimear E. Kenny, Gillian M. Belbin, Stuart C. Sealfon, Robert P. Sebra, Irene Salib, Brett L. Collins, Tess Levy, Bari Britvan, Katherine Keller, Lara Tang, Michael Peruggia, Liam L. Hiester, Kristi Niblo, Alexandra Aksentijevich, Alexander Labkowsky, Avromie Karp, Menachem Zlatopolsky, Michael Preuss, Ruth J.F. Loos, Girish N. Nadkarni, Ron Do, Clive Hoggart, Sam Choi, Slayton J. Underwood, Paul O’Reilly, Laura M. Huckins, Marissa Zyndorf, AII - Infectious diseases, Amsterdam Neuroscience - Neuroinfection & -inflammation, and Neurology
- Subjects
Medical Physiology ,Gene Expression ,Genome-wide association study ,Genome ,Severity of Illness Index ,colocalization ,Gene expression ,Databases, Genetic ,Ethnicity ,2.1 Biological and endogenous factors ,GWAS ,Aetiology ,Biology (General) ,Lung ,Genetics ,Chromosome Mapping ,Single Nucleotide ,Organ Specificity ,Biotechnology ,Cell type ,QH301-705.5 ,Quantitative Trait Loci ,Single-nucleotide polymorphism ,Biology ,eQTL ,Polymorphism, Single Nucleotide ,Article ,General Biochemistry, Genetics and Molecular Biology ,Databases ,Genetic ,SNP ,Humans ,Genetic Predisposition to Disease ,COVID-19 Host Genetics Initiative ,Polymorphism ,Gene ,COVID-19 ,SARS-CoV-2 ,Gene Expression Profiling ,Prevention ,Human Genome ,Computational Biology ,Genetic Variation ,Good Health and Well Being ,Expression quantitative trait loci ,Biochemistry and Cell Biology ,Transcriptome ,Genome-Wide Association Study - Abstract
Variability in SARS-CoV-2 susceptibility and COVID-19 disease severity between individuals is partly due to genetic factors. Here, we identify 4 genomic loci with suggestive associations for SARS-CoV-2 susceptibility and 19 for COVID-19 disease severity. Four of these 23 loci likely have an ethnicity-specific component. Genome-wide association study (GWAS) signals in 11 loci colocalize with expression quantitative trait loci (eQTLs) associated with the expression of 20 genes in 62 tissues/cell types (range: 1:43 tissues/gene), including lung, brain, heart, muscle, and skin as well as the digestive system and immune system. We perform genetic fine mapping to compute 99% credible SNP sets, which identify 10 GWAS loci that have eight or fewer SNPs in the credible set, including three loci with one single likely causal SNP. Our study suggests that the diverse symptoms and disease severity of COVID-19 observed between individuals is associated with variants across the genome, affecting gene expression levels in a wide variety of tissue types., Graphical abstract, D’Antonio et al. characterize associations between GWAS signals for COVID-19 disease and eQTLs in 69 human tissues to identify causal variants and their underlying molecular mechanisms. They show that diverse symptoms and disease severity of COVID-19 are associated with variants affecting gene expression in a wide variety of tissues.
- Published
- 2022
- Full Text
- View/download PDF
16. Identification of Discriminative Gene-level and Protein-level Features Associated with Gain-of-Function and Loss-of-Function Mutations
- Author
-
Cigdem Sevim Bayrak, Aayushee Jain, David Neil Cooper, Kumardeep Chaudhary, Avner Schlessinger, Yuval Itan, Tielman Van Vleck, Girish N. Nadkarni, Stéphanie Boisson-Dupuis, David Stein, and Peter D. Stenson
- Subjects
Discriminative model ,Identification (biology) ,Computational biology ,Plasma protein binding ,Gene mutation ,Biology ,Phenotype ,Gene ,Loss function ,Germline - Abstract
Gain-of-function (GOF) and loss-of-function (LOF) mutations in the same gene may result in markedly different clinical phenotypes and hence require different therapeutic treatments. Identifying the functional consequences of mutations is an important step toward understanding disease mechanisms. While there are numerous computational tools (e.g., CADD, SIFT, PolyPhen-2) to predict the pathogenicity of a variant, there are currently no methods to predict whether a given genetic mutation results in a gene product with increased (gain-of-function; GOF) or diminished (loss-of-function; LOF) activity. Here, we investigated various gene- and protein-level features of GOF and LOF mutations. We generated the first extensive database of all currently known germline GOF and LOF pathogenic mutations by employing natural language processing (NLP) on the available abstracts in the Human Gene Mutation Database. Machine learning and statistical analyses of gene- and protein-level features associated with GOF and LOF mutations indicated significant differences. For example, GOF mutations were enriched in essential genes, autosomal dominant inheritance, protein binding and interaction domains, whereas LOF mutations were enriched in singleton genes, protein-truncating variants, and protein core regions. These results are consistent with the notion that mutations underlying recessive and dominant disorders have significantly different structural and functional properties. These findings could ultimately improve our understanding of how mutations affect gene/protein function thereby guiding future treatment options.
- Published
- 2021
- Full Text
- View/download PDF
17. TLR3 controls constitutive IFN-β antiviral immunity in human fibroblasts and cortical neurons
- Author
-
Isabelle Meyts, Xin Mu, Emmanuelle Jouanguy, Daxing Gao, Aurélie Cobat, Lazaro Lorenzo, Mary Hasek, Damien Chaussabel, Luigi D. Notarangelo, Darragh Duffy, Vanessa Sancho-Shimizu, Jean-Laurent Casanova, Lorenz Studer, Gregory A. Smith, Sun Hur, Jie Chen, Peng Zhang, Michael J. Ciancanelli, Jessica L. McAlpine, Gabriele Ciceri, Shen-Ying Zhang, Oliver Harschnitz, Yuval Itan, Michael S. Diamond, Benedetta Bigio, Vincent Bondet, Esperanza Anguiano, Laurent Abel, Rockefeller University [New York], University of Science and Technology of China [Hefei] (USTC), Turnstone Biologics [New York], Memorial Sloane Kettering Cancer Center [New York], Immunologie Translationnelle - Translational Immunology lab, Institut Pasteur [Paris], Harvard Medical School [Boston] (HMS), Icahn School of Medicine at Mount Sinai [New York] (MSSM), Human genetics of infectious diseases: Complex predisposition (Equipe Inserm U1163), Imagine - Institut des maladies génétiques (IHU) (Imagine - U1163), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), Imperial College London, Baylor Institute for Immunology Research (BIIR), Benaroya Research Institute [Seattle] (BRI), Sidra Medicine [Doha, Qatar], Catholic University of Leuven - Katholieke Universiteit Leuven (KU Leuven), University Hospitals Leuven [Leuven], Washington University School of Medicine in St. Louis, Washington University in Saint Louis (WUSTL), Feinberg School of Medicine, Northwestern University [Evanston], National Institute of Allergy and Infectious Diseases [Bethesda] (NIAID-NIH), National Institutes of Health [Bethesda] (NIH), Service d'immuno-hématologie pédiatrique [CHU Necker], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Howard Hughes Medical Institute (HHMI), This work was funded in part by the National Center for Advancing Translational Sciences, NIH Clinical and Translational Science Award program (UL1TR001866), NIH (R01NS072381, R01AI088364, and R21AI151663), the French National Research Agency (ANR) under the 'Investments for the future' program (ANR-10-IAHU-01), Integrative Biology of Emerging Infectious Diseases Laboratoire d’Excellence (ANR-10-LABX-62-IBEID), and grants ANR-14-CE14-0008-01 and ANR-18-CE15-0020-02, The Rockefeller University, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Descartes University, and the St. Giles Foundation. DG is supported by the Charles H. Revson Senior Fellowship in Biomedical Sciences and the National Natural Science Foundation of China (grant 31970855). IM is supported by KU Leuven C1 grant C16/18/007 and Fonds Wetenschappelijk Onderzoek Vlaanderen grant G0C8517N., ANR-10-IAHU-0001,Imagine,Institut Hospitalo-Universitaire Imagine(2010), ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), ANR-14-CE14-0008,IEIHSEER,L'encéphalite Herpétique de l'enfant résulte de déficits héréditaires d'immunité contre l'HSV-1: une exception ou une règle?(2014), ANR-18-CE15-0020,SEAe-HostFactors,Facteurs de susceptibilité de l'hôte à l'encéphalite pédiatrique en Asie du Sud Est(2018), Institut Pasteur [Paris] (IP), and Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité)
- Subjects
0301 basic medicine ,viruses ,NF-KAPPA-B ,Herpesvirus 1, Human ,Research & Experimental Medicine ,RIG-I ,Mice ,0302 clinical medicine ,ADAPTER PROTEIN ,Induced pluripotent stem cell ,GENE-EXPRESSION ,Cerebral Cortex ,Mice, Knockout ,Neurons ,Innate immunity ,Infectious disease ,biology ,virus diseases ,VESICULAR STOMATITIS-VIRUS ,General Medicine ,3. Good health ,Cell biology ,INTERFERON-ALPHA/BETA ,Medicine, Research & Experimental ,Vesicular stomatitis virus ,030220 oncology & carcinogenesis ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,MESSENGER-RNA ,Life Sciences & Biomedicine ,Research Article ,HERPES-SIMPLEX ENCEPHALITIS ,Immunology ,chemical and pharmacologic phenomena ,DOUBLE-STRANDED-RNA ,Cell Line ,03 medical and health sciences ,Immunity ,Animals ,Humans ,Secretion ,Messenger RNA ,Science & Technology ,Innate immune system ,TOLL-LIKE RECEPTOR-3 ,Interferon-beta ,Vesiculovirus ,Fibroblasts ,biology.organism_classification ,Embryonic stem cell ,Toll-Like Receptor 3 ,030104 developmental biology ,TLR3 - Abstract
Human herpes simplex virus 1 (HSV-1) encephalitis can be caused by inborn errors of the TLR3 pathway, resulting in impairment of CNS cell-intrinsic antiviral immunity. Deficiencies of the TLR3 pathway impair cell-intrinsic immunity to vesicular stomatitis virus (VSV) and HSV-1 in fibroblasts, and to HSV-1 in cortical but not trigeminal neurons. The underlying molecular mechanism is thought to involve impaired IFN-α/β induction by the TLR3 recognition of dsRNA viral intermediates or by-products. However, we show here that human TLR3 controls constitutive levels of IFNB mRNA and secreted bioactive IFN-β protein, and thereby also controls constitutive mRNA levels for IFN-stimulated genes (ISGs) in fibroblasts. Tlr3-/- mouse embryonic fibroblasts also have lower basal ISG levels. Moreover, human TLR3 controls basal levels of IFN-β secretion and ISG mRNA in induced pluripotent stem cell-derived cortical neurons. Consistently, TLR3-deficient human fibroblasts and cortical neurons are vulnerable not only to both VSV and HSV-1, but also to several other families of viruses. The mechanism by which TLR3 restricts viral growth in human fibroblasts and cortical neurons in vitro and, by inference, by which the human CNS prevents infection by HSV-1 in vivo, is therefore based on the control of early viral infection by basal IFN-β immunity. ispartof: JOURNAL OF CLINICAL INVESTIGATION vol:131 issue:1 ispartof: location:United States status: published
- Published
- 2021
- Full Text
- View/download PDF
18. Identifying novel high-impact rare disease-causing mutations, genes and pathways in exomes of Ashkenazi Jewish inflammatory bowel disease patients
- Author
-
Dalin Li, Lee A. Denson, Talin Haritunians, Scott B. Snapper, Harry Ostrer, Aayushee Jain, Kyle Gettler, John D. Rioux, David Neil Cooper, Cigdem Sevim Bayrak, Yiming Wu, Ksenija Sabic, Peter D. Stenson, Subra Kugathasan, Mamta Giri, Dermot P.B. McGovern, Mark S. Silverberg, Patrick Maffucci, L. Philip Schumm, Steven R. Brant, Yuval Itan, Richard H. Duerr, Girish N. Nadkarni, Judy H. Cho, Tielman Van Vleck, and Mark J. Daly
- Subjects
Genetics ,0303 health sciences ,education.field_of_study ,Population ,Genome-wide association study ,Disease ,Biology ,digestive system diseases ,Ashkenazi jews ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Missing heritability problem ,030211 gastroenterology & hepatology ,education ,Exome sequencing ,030304 developmental biology ,Genetic association ,Rare disease - Abstract
Inflammatory bowel disease (IBD) is a group of chronic diseases, affecting different parts of the gastrointestinal tract, that mainly comprises Crohn’s Disease (CD) and Ulcerative Colitis (UC). Most IBD genomic research to date has involved genome-wide association studies (GWAS) of common genetic variants, mostly in Europeans, resulting in the identification of over 200 risk loci. The incidence of IBD in Ashkenazi Jews (AJ) is particularly high compared to other population groups and rare protein-coding variants are significantly enriched in AJ. These variants are expected to have a larger phenotypic effect and are hypothesized to complement the missing heritability that cannot be fully addressed by GWAS in IBD. Therefore, we genetically identified 4,974 AJs IBD cases and controls from whole exome sequencing (WES) data from the NIDDK IBD Genetics Consortium (IBDGC). We selected credible rare variants with high predicted impact, aggregated them into genes, and performed gene burden and pathway enrichment analyses to identify 7 novel plausible IBD-causing genes:NCF1, CES1, ICAM1, INPP5D, ABCB1, IL33andTLR4. We further perform bulk and single-cell RNA sequencing, demonstrating the likely relatedness of the novel genes to IBD. Importantly, we demonstrate that the rare and high impact genetic architecture of AJ adult IBD displays a significant overlap with very early onset IBD (VEOIBD) genetics. At the variant level, we performed Phenome-wide association studies (PheWAS) in the UK Biobank to replicate risk sites in IBD and reveal shared risk sites with other diseases. Finally, we showed that a polygenic risk score (PRS) has high power to differentiate AJ IBD cases from controls when using rare and high impact variants.
- Published
- 2020
- Full Text
- View/download PDF
19. Non-syndromic Oculocutaneous Albinism: Novel Genetic Variants and Clinical Follow Up of a Brazilian Pediatric Cohort
- Author
-
Laire Schidlowski, Fernando Liebert, Pérola Grupenmacher Iankilevich, Priscila Regina Orso Rebellato, Rafaela Andrade Rocha, Nadia Aparecida Pereira Almeida, Aayushee Jain, Yiming Wu, Yuval Itan, Roberto Rosati, and Carolina Prando
- Subjects
melanogenesis ,0301 basic medicine ,SLC45A2 ,medicine.medical_specialty ,lcsh:QH426-470 ,Genetic counseling ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,Medicine ,Genetics (clinical) ,Hypopigmentation ,TYR ,biology ,business.industry ,Genetic disorder ,sequencing ,Brief Research Report ,medicine.disease ,Oculocutaneous albinism ,Dermatology ,eye diseases ,lcsh:Genetics ,030104 developmental biology ,030220 oncology & carcinogenesis ,Chromosomal region ,Albinism ,biology.protein ,Molecular Medicine ,medicine.symptom ,Skin cancer ,business ,oculocutaneous albinism ,exome - Abstract
Oculocutaneous albinism (OCA) is a genetic disorder characterized by skin, hair, and eye hypopigmentation due to a reduction or absence of melanin. Clinical manifestations include vision problems and a high susceptibility to skin cancer. In its non-syndromic form, OCA is associated with six genes and one chromosomal region. Because OCA subtypes are not always clinically distinguishable, molecular analysis has become an important tool for classifying types of OCA, which facilitates genetic counseling and can guide the development of new therapies. We studied eight Brazilian individuals aged 1.5–18 years old with clinical diagnosis of OCA. Assessment of ophthalmologic characteristics showed results consistent with albinism, including reduced visual acuity, nystagmus, and loss of stereoscopic vision. We also observed the appearance of the strabismus and changes in static refraction over a 2-year period. Dermatologic evaluation showed that no participants had preneoplastic skin lesions, despite half of the participants reporting insufficient knowledge about skin care in albinism. Whole-exome and Sanger sequencing revealed eight different mutations: six in the TYR gene and two in the SLC45A2 gene, of which one was novel and two were described in a population study but were not previously associated with the OCA phenotype. We performed two ophthalmological evaluations, 2 years apart; and one dermatological evaluation. To the best of our knowledge, this is the first study to perform clinical follow-up and genetic analysis of a Brazilian cohort with albinism. Here, we report three new OCA causing mutations.
- Published
- 2020
- Full Text
- View/download PDF
20. Rescue of recurrent deep intronic mutation underlying cell type–dependent quantitative NEMO deficiency
- Author
-
Maya Chrabieh, Anne Puel, Yuri Kawasaki, Hong Hur, Kazushi Izawa, Bertrand Boisson, Osamu Ohara, Takahiro Yasumi, Mitsujiro Osawa, Matthieu Bendavid, Ryuta Nishikomori, Benedetta Bigio, Masatoshi Hagiwara, Yuval Itan, Masahiko Ajiro, Hiroshi Nihira, Toshio Heike, Andrew R. Gennery, Megumu K. Saito, Jacinta Bustamante, Jean-Laurent Casanova, Takayuki Tanaka, Jose Ichishima, Satoshi Okada, Yupu Liang, Laurent Abel, Yoshitaka Honda, and Takeshi Shiba
- Subjects
Male ,0301 basic medicine ,Cell type ,Biology ,Frameshift mutation ,03 medical and health sciences ,0302 clinical medicine ,Ectodermal Dysplasia ,IKBKG ,medicine ,Humans ,Incontinentia Pigmenti ,Frameshift Mutation ,Induced pluripotent stem cell ,Gene knockdown ,Macrophages ,Immunologic Deficiency Syndromes ,Intron ,General Medicine ,Incontinentia pigmenti ,Fibroblasts ,medicine.disease ,Molecular biology ,Introns ,I-kappa B Kinase ,030104 developmental biology ,030220 oncology & carcinogenesis ,RNA splicing ,Female ,Research Article - Abstract
X-linked dominant incontinentia pigmenti (IP) and X-linked recessive anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID) are caused by loss-of-function and hypomorphic IKBKG (also known as NEMO) mutations, respectively. We describe a European mother with mild IP and a Japanese mother without IP, whose 3 boys with EDA-ID died from ID. We identify the same private variant in an intron of IKBKG, IVS4+866 C>T, which was inherited from and occurred de novo in the European mother and Japanese mother, respectively. This mutation creates a new splicing donor site, giving rise to a 44-nucleotide pseudoexon (PE) generating a frameshift. Its leakiness accounts for NF-κB activation being impaired but not abolished in the boys’ cells. However, aberrant splicing rates differ between cell types, with WT NEMO mRNA and protein levels ranging from barely detectable in leukocytes to residual amounts in induced pluripotent stem cell–derived (iPSC-derived) macrophages, and higher levels in fibroblasts and iPSC-derived neuronal precursor cells. Finally, SRSF6 binds to the PE, facilitating its inclusion. Moreover, SRSF6 knockdown or CLK inhibition restores WT NEMO expression and function in mutant cells. A recurrent deep intronic splicing mutation in IKBKG underlies a purely quantitative NEMO defect in males that is most severe in leukocytes and can be rescued by the inhibition of SRSF6 or CLK.
- Published
- 2018
- Full Text
- View/download PDF
21. Disruption of an antimycobacterial circuit between dendritic and helper T cells in human SPPL2a deficiency
- Author
-
Katia Abarca, Fabienne Jabot-Hanin, Rubén Martínez-Barricarte, David Langlais, Kimberly Lucero, Stéphanie Boisson-Dupuis, Laurent Abel, Federico Mele, Xiao-Fei Kong, Capucine Picard, Cindy S. Ma, Cecilia S. Lindestam Arlehamn, Aziz Bousfiha, Dewton Moraes-Vasconcelos, Fatima Ailal, Figen Dogu, Caner Aytekin, Caroline Deswarte, Yuval Itan, Gaëlle Breton, Michel C. Nussenzweig, Geetha Rao, Elissa K. Deenick, Théo Lasseau, J. Kennedy, Jacinta Bustamante, Bernd Schröder, Stuart G. Tangye, Daniela Latorre, Sophie Hambleton, Ibtihal Benhsaien, Kang Liu, Frederic Geissmann, Alessandro Sette, Federica Sallusto, Aydan Ikinciogullari, Janet Markle, Tomi Lazarov, Céline Trouillet, Philippe Gros, and Jean-Laurent Casanova
- Subjects
Male ,0301 basic medicine ,Myeloid ,CD74 ,Immunology ,Lymphadenopathy ,Human leukocyte antigen ,Biology ,medicine.disease_cause ,Interferon-gamma ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,HLA Antigens ,Immunity ,medicine ,Animals ,Aspartic Acid Endopeptidases ,Humans ,Tuberculosis ,Immunology and Allergy ,Interferon gamma ,Cells, Cultured ,Mice, Knockout ,Mycobacterium Infections ,Mutation ,Vaccination ,Histocompatibility Antigens Class II ,Infant ,Membrane Proteins ,Dendritic Cells ,Mycobacterium tuberculosis ,Dendritic cell ,Th1 Cells ,Mycobacterium bovis ,Research Highlight ,3. Good health ,Antigens, Differentiation, B-Lymphocyte ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunologic Memory ,medicine.drug - 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. Primary immunodeficiency can predispose patients to mycobacterial disease. Casanova and colleagues identify novel human mutations in the enzyme SPPL2A that result in selective accumulation of CD74 in a dendritic cell subset and lead to their death and the failure to mount effective TH1 responses.
- Published
- 2018
- Full Text
- View/download PDF
22. Alanine-scanning mutagenesis of human signal transducer and activator of transcription 1 to estimate loss- or gain-of-function variants
- Author
-
Masao Kobayashi, Reiko Kagawa, Hidetoshi Takada, Osamu Ohara, Jamila El Baghdadi, Maiko Ikeda, Yuval Itan, Silvia Danielian, Jean-Laurent Casanova, Kaori Fujiwara, Ryoji Fujiki, Osamu Hirata, Zenichiro Kato, Satoshi Saito, Shiho Nishimura, Aziz Bousfiha, Jacinta Bustamante, Fatima Ailal, Stéphanie Boisson-Dupuis, Satoshi Okada, Anne Puel, Matías Oleastro, Laura Perez, Xiao-Fei Kong, Toshiro Hara, Hidenori Ohnishi, Judith Yancoski, Sonoko Sakata, and Miyuki Tsumura
- Subjects
0301 basic medicine ,Genetics ,Immunology ,Mutant ,Wild type ,DNA-binding domain ,Alanine scanning ,Biology ,medicine.disease ,Germline ,03 medical and health sciences ,030104 developmental biology ,medicine ,Immunology and Allergy ,Missense mutation ,Chronic mucocutaneous candidiasis ,Loss function - Abstract
Background Germline heterozygous mutations in human signal transducer and activator of transcription 1 (STAT1) can cause loss of function (LOF), as in patients with Mendelian susceptibility to mycobacterial diseases, or gain of function (GOF), as in patients with chronic mucocutaneous candidiasis. LOF and GOF mutations are equally rare and can affect the same domains of STAT1, especially the coiled-coil domain (CCD) and DNA-binding domain (DBD). Moreover, 6% of patients with chronic mucocutaneous candidiasis with a GOF STAT1 mutation have mycobacterial disease, obscuring the functional significance of the identified STAT1 mutations. Current computational approaches, such as combined annotation-dependent depletion, do not distinguish LOF and GOF variants. Objective We estimated variations in the CCD/DBD of STAT1. Methods We mutagenized 342 individual wild-type amino acids in the CCD/DBD (45.6% of full-length STAT1) to alanine and tested the mutants for STAT1 transcriptional activity. Results Of these 342 mutants, 201 were neutral, 30 were LOF, and 111 were GOF mutations in a luciferase assay. This assay system correctly estimated all previously reported LOF mutations (100%) and slightly fewer GOF mutations (78.1%) in the CCD/DBD of STAT1. We found that GOF alanine mutants occurred at the interface of the antiparallel STAT1 dimer, suggesting that they destabilize this dimer. This assay also precisely predicted the effect of 2 hypomorphic and dominant negative mutations, E157K and G250E, in the CCD of STAT1 that we found in 2 unrelated patients with Mendelian susceptibility to mycobacterial diseases. Conclusion The systematic alanine-scanning assay is a useful tool to estimate the GOF or LOF status and the effect of heterozygous missense mutations in STAT1 identified in patients with severe infectious diseases, including mycobacterial and fungal diseases.
- Published
- 2017
- Full Text
- View/download PDF
23. PopViz: a webserver for visualizing minor allele frequencies and damage prediction scores of human genetic variations
- Author
-
Jean-Laurent Casanova, Laurent Abel, Peng Zhang, Yuval Itan, Shen-Ying Zhang, Franck Rapaport, Bertrand Boisson, and Benedetta Bigio
- Subjects
0301 basic medicine ,Statistics and Probability ,Candidate gene ,Population ,Population genetics ,Computational biology ,Biology ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Gene Frequency ,Genetic variation ,Humans ,education ,Molecular Biology ,Gene ,education.field_of_study ,Computational Biology ,Genetic Variation ,High-Throughput Nucleotide Sequencing ,Genomics ,Genetic code ,Applications Notes ,Computer Science Applications ,Minor allele frequency ,Computational Mathematics ,030104 developmental biology ,Computational Theory and Mathematics ,Mutation (genetic algorithm) ,Software ,030217 neurology & neurosurgery - 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.
- Published
- 2018
- Full Text
- View/download PDF
24. Chronic mucocutaneous candidiasis and connective tissue disorder in humans with impaired JNK1-dependent responses to IL-17A/F and TGF-β
- Author
-
Marco Ritelli, Harry C. Dietz, Danielle T. Avery, Bertrand Boisson, Soraya Boucherit, Lucie Grodecká, Stuart G. Tangye, Romain Lévy, Kathryn Payne, Tomáš Freiberger, Sophie Cypowyj, Juan Li, Valérie Cormier-Daire, Nicoletta Zoppi, Laurent Abel, Geetha Rao, Vivien Béziat, Andrea Guennoun, Benedetta Bigio, Maya Chrabieh, Salim Bougarn, Marina Colombi, Lei Shang, Emilie Corvilain, Yuval Itan, Anne Puel, Franck Rapaport, Nico Marr, Fransiska Malfait, Delfien Syx, Mélanie Migaud, Tanwir Habib, Sabri Boughorbel, Jean-Laurent Casanova, and Cindy S. Ma
- Subjects
Male ,0301 basic medicine ,Connective Tissue Disorder ,MAPK8 ,Immunology ,Mucocutaneous zone ,Connective tissue ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Transforming Growth Factor beta ,medicine ,Humans ,Mitogen-Activated Protein Kinase 8 ,Chronic mucocutaneous candidiasis ,Connective Tissue Diseases ,Alleles ,Cells, Cultured ,business.industry ,Candidiasis, Chronic Mucocutaneous ,Interleukin-17 ,General Medicine ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Female ,Signal transduction ,Haploinsufficiency ,business - Abstract
Genetic etiologies of chronic mucocutaneous candidiasis (CMC) disrupt human IL-17A/F-dependent immunity at mucosal surfaces, whereas those of connective tissue disorders (CTDs) often impair the TGF-β-dependent homeostasis of connective tissues. The signaling pathways involved are incompletely understood. We report a three-generation family with an autosomal dominant (AD) combination of CMC and a previously undescribed form of CTD that clinically overlaps with Ehlers-Danlos syndrome (EDS). The patients are heterozygous for a private splice-site variant of MAPK8, the gene encoding c-Jun N-terminal kinase 1 (JNK1), a component of the MAPK signaling pathway. This variant is loss-of-expression and loss-of-function in the patients' fibroblasts, which display AD JNK1 deficiency by haploinsufficiency. These cells have impaired, but not abolished, responses to IL-17A and IL-17F. Moreover, the development of the patients' TH17 cells was impaired ex vivo and in vitro, probably due to the involvement of JNK1 in the TGF-β-responsive pathway and further accounting for the patients' CMC. Consistently, the patients' fibroblasts displayed impaired JNK1- and c-Jun/ATF-2-dependent induction of key extracellular matrix (ECM) components and regulators, but not of EDS-causing gene products, in response to TGF-β. Furthermore, they displayed a transcriptional pattern in response to TGF-β different from that of fibroblasts from patients with Loeys-Dietz syndrome caused by mutations of TGFBR2 or SMAD3, further accounting for the patients' complex and unusual CTD phenotype. This experiment of nature indicates that the integrity of the human JNK1-dependent MAPK signaling pathway is essential for IL-17A- and IL-17F-dependent mucocutaneous immunity to Candida and for the TGF-β-dependent homeostasis of connective tissues.
- Published
- 2019
- Full Text
- View/download PDF
25. 374 INFLAMED ULCERATIVE COLITIS REGIONS ASSOCIATED TO MRGPRX2-MEDIATED MAST CELL DEGRANULATION AND CELL ACTIVATION MODULES, DEFINING A NEW THERAPEUTIC TARGET
- Author
-
Ling-shiang F. Chuang, Ernie Chen, Mamta Giri, Nicole Villaverde, Nai-yun Hsu, Ksenija Sabic, Sari Joshowitz, Kyle Gettler, Shikha Nayar, Zhi Chai, Isaac Alter, Colleen Chasteau, Ujunwa Korie, Siarhei Dzedzik, Tin Htwe Thin, Aayushee Jain, Arden Moscati, Gerold Bongers, Richard H. Duerr, Mark S. Silverberg, Steven R. Brant, John D. Rioux, Inga Peter, L. Philip Schumm, Talin Haritunians, Dermot P.B. Mcgovern, Yuval Itan, and Judy H. Cho
- Subjects
Hepatology ,Gastroenterology - Published
- 2021
- Full Text
- View/download PDF
26. 776 IDENTIFYING NOVEL HIGH-IMPACT RARE DISEASE-CAUSING MUTATIONS, GENES AND PATHWAYS IN EXOMES OF ASHKENAZI JEWISH INFLAMMATORY BOWEL DISEASE PATIENT
- Author
-
Yiming Wu, Kyle Gettler, Mamta Giri, Ksenija Sabic, L. Philip Schumm, Mark J. Daly, Richard H. Duerr, Mark S. Silverberg, John D. Rioux, Steven R. Brant, Dermot P.B. Mcgovern, Judy H. Cho, and Yuval Itan
- Subjects
Hepatology ,Gastroenterology - Published
- 2021
- Full Text
- View/download PDF
27. Gain-of-function mutation in PIK3R1 in a patient with a narrow clinical phenotype of respiratory infections
- Author
-
José R. Regueiro, Sonia García-Gómez, Jean-Laurent Casanova, Ana Domínguez Acosta, Rebeca Pérez de Diego, Bertrand Boisson, Jesús Poch Páez, Yuval Itan, Juan Jesús Mendoza Quintana, Rubén Martínez-Barricarte, Carlos Rodríguez-Gallego, María Teresa Martínez-Saavedra, Eduardo J. García-Reino, Gracián Camps, and Silvia Sánchez-Ramón
- Subjects
0301 basic medicine ,Immunology ,Biology ,medicine.disease ,Hypogammaglobulinemia ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immunophenotyping ,PIK3R1 ,Mutation (genetic algorithm) ,medicine ,biology.protein ,Immunology and Allergy ,Respiratory system ,Antibody ,Clinical phenotype ,Exome sequencing ,030215 immunology - Abstract
Antibody deficiencies can be caused by a variety of defects that interfere with B-cell development, maturation, and/or function. Using whole-exome sequencing we found a PIK3R1 mutation in a patient with hypogammaglobulinemia and a narrow clinical phenotype of respiratory infections. Early diagnosis is crucial; careful analysis of B and T-cells followed by genetic analyses may help to distinguish activated PI3K-delta syndrome (APDS) from other, less severe, predominantly antibody deficiencies.
- Published
- 2016
- Full Text
- View/download PDF
28. Mo1112 IDENTIFYING NOVEL HIGH-IMPACT RARE DISEASE-CAUSING MUTATIONS, GENES AND PATHWAYS IN EXOMES OF ASHKENAZI JEWISH INFLAMMATORY BOWEL DISEASE PATIENTS
- Author
-
Richard H. Duerr, Judy H. Cho, Dermot P.B. McGovern, Mark J. Daly, Steven R. Brant, Mark S. Silverberg, Yuval Itan, Ksenija Sabic, L. Philip Schumm, Yiming Wu, Kyle Gettler, Mamta Giri, and John D. Rioux
- Subjects
Hepatology ,business.industry ,Immunology ,Gastroenterology ,medicine ,Ashkenazi Jewish ,medicine.disease ,business ,Inflammatory bowel disease ,Gene ,Exome sequencing ,Rare disease - Published
- 2020
- Full Text
- View/download PDF
29. Human IFN-γ immunity to mycobacteria is governed by both IL-12 and IL-23
- Author
-
Cindy S. Ma, Jean-Laurent Casanova, Monika Schmidt, Federico Mele, Gaspard Kerner, James P. Di Santo, Cecilia S. Lindestam Arlehamn, Avneet Heer, Lluis Quintana-Murci, Alessandro Sette, Bernhard Fleckenstein, Tomi Lazarov, Sandra Jovic, Natalie Wong, Stéphanie Boisson-Dupuis, Daniela Latorre, Julia K. Joseph, Bertrand Boisson, Rubén Martínez-Barricarte, Caner Aytekin, Danielle T. Avery, Aydan Ikinciogullari, Figen Dogu, Jean-François Emile, Etienne Patin, Federica Sallusto, Yoann Seeleuthner, Yuval Itan, Laurent Abel, Franck Rapaport, Alejandro Nieto-Patlán, Frederic Geissmann, Satoshi Okada, Fabienne Jabot-Hanin, Stuart G. Tangye, Esther van de Vosse, Geetha Rao, Elissa K. Deenick, Jacinta Bustamante, Mélanie Migaud, Caroline Deswarte, Mohammed Reza Bloursaz, Laura Surace, Benedetta Bigio, Davood Mansouri, Anne Puel, Payam Tabarsi, Xiao-Fei Kong, Gönül Tanır, Vanessa L. Bryant, Noé Ramírez-Alejo, Seyed Alireza Mahdaviani, Janet Markle, St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller University [New York], University of New South Wales [Sydney] (UNSW), Università della Svizzera italiana = University of Italian Switzerland (USI), Shahid Beheshti University of Medical Sciences [Tehran] (SBUMS), Sami Ulus Maternity and Children Training and Research Hospital, The Walter and Eliza Hall Institute of Medical Research (WEHI), University of Melbourne, The Royal Melbourne Hospital, CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Imagine - Institut des maladies génétiques (IHU) (Imagine - U1163), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), Immunité Innée - Innate Immunity, Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM), Icahn School of Medicine at Mount Sinai [New York] (MSSM), Génomique évolutive, modélisation et santé (GEMS), Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Génétique Evolutive Humaine - Human Evolutionary Genetics, Centre de Bioinformatique, Biostatistique et Biologie Intégrative (C3BI), Hiroshima University, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Ankara University School of Medicine [Turkey], Memorial Sloane Kettering Cancer Center [New York], Weill Medical College of Cornell University [New York], King‘s College London, La Jolla Institute for Immunology [La Jolla, CA, États-Unis], University of California [San Diego] (UC San Diego), University of California (UC), Hôpital Ambroise Paré [AP-HP], Leiden University Medical Center (LUMC), Universiteit Leiden, Eidgenössische Technische Hochschule - Swiss Federal Institute of Technology [Zürich] (ETH Zürich), Howard Hughes Medical Institute [New York] (HHMI), Howard Hughes Medical Institute (HHMI)-New York University School of Medicine, NYU System (NYU)-NYU System (NYU)-Rockefeller University [New York]-Columbia University Irving Medical Center (CUIMC), The Laboratory of Human Genetics of Infectious Diseases was supported by grants from the National Institute of Allergy and Infectious Diseases (NIAID) grant numbers 5R37AI095983, R01AI089970, and K99AI127932, the National Center for Research Resources and the National Center for Advancing Sciences (NCATS) of the NIH grant number UL1TR001866, the Rockefeller University, the St. Giles Foundation, the European Research Council (ERC-2010-AdG-268777 and grant no. 323183), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Descartes University, the Integrative Biology of Emerging Infectious Diseases Laboratory of Excellence (ANR-10-LABX-62-IBEID), and the French National Research Agency (ANR) under the 'Investissement d’avenir' program (grant ANR-10-IAHU-01), ANR-TBPATHGEN (grant ANR-14-CE14-0007-01), ANR-IFNPHOX (grant ANR13-ISV3-0001-01), and ANR-GENMSMD (grant ANR16-CE17-0005-01). This work was supported by NIAID award no. U19AI118626 (to A.S. and F.S.). J.G.M. was funded by the Canadian Institutes of Health Research, the NIH Translational Science Award (CTSA) program (no. UL1 TR000043), the Swiss National Science Foundation (grant no. IZKOZ3_173586), the Charles H. Revson Foundation, and the NIAID (1K99AI127932-01A1). R.M.-B. was supported by the European Molecular Biology Organization (EMBO). N.R.-A. was supported by the National Council of Science and Technology of Mexico (CONACYT, 264011) and the Stony Wold-Herbert Fund Fellowship Grant. Y.I. was supported by the AXA Research Fund. S.G.T., E.K.D., and C.S.M. are supported by research grants and fellowships from the National Health and Medical Research Council of Australia (S.G.T., C.S.M., and E.K.D.) and the Office for Health and Medical Research of the State Government of NSW Australia (C.S.M.). A.S. is supported by NIH research grant HHSN272200900044C. J.B. is supported by SRC2017. The Institute for Research in Biomedicine and F.S. are supported by the Helmut Horten Foundation. S.O. was supported by the Aid for Scientific Research Grant from the Japanese Society for the Promotion of Science (16H05355) and the Practical Research Project for Rare/Intractable Diseases from the Japan Agency for Medical Research and Development., ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), ANR-10-IAHU-0001,Imagine,Institut Hospitalo-Universitaire Imagine(2010), ANR-14-CE14-0007,TBPATHGEN,Dissection de la pathogenèse de la tuberculose par l'identification de défauts monogéniques de l'immunité dans les formes pédiatriques sévères de la maladie(2014), ANR-13-ISV3-0001,IFNGPHOX,L'immunité anti-tuberculeuse dépendante de l'IFN-gamma chez l'homme opère via la NADPH oxydase phagocytaire(2013), ANR-16-CE17-0005,GENMSMD,Dissection génétique de la Susceptibilité Mendélienne aux infections mycobactériennes chez l'homme(2016), European Project: 268777,EC:FP7:ERC,ERC-2010-AdG_20100317,GENTB(2011), European Project: 323183,EC:FP7:ERC,ERC-2012-ADG_20120314,PREDICT(2013), Shahid Beheshti University, Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), Institut Pasteur [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Génomique évolutive, modélisation et santé (CNRS-UMR2000), Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), University of California, The Rockefeller University, St Giles laboratory of Human Genetics and Infectious Diseases, rockefeller university, Garvan Institute of Medical Research [Darlinghurst, Australia], Dis Training & Res Ctr, Shahid Beheshti University of Medical Sciences, Ecole d'Ingénieurs de Purpan (INPT - EI Purpan), Institut National Polytechnique (Toulouse) (Toulouse INP), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées, Immunité Innée, Imagine - Institut des maladies génétiques (IMAGINE - U1163), Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Centre de Bioinformatique (CBIO), MINES ParisTech - École nationale supérieure des mines de Paris-PSL Research University (PSL), Institut de Génomique d'Evry (IG), Institut de Biologie François JACOB (JACOB), Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay, Sami Ulus Children Hlth & Dis Training & Res Ctr, Ankara, Génétique Humaine des Maladies Infectieuses (Inserm U980), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Descartes - Paris 5 (UPD5), Service d'anatomie pathologique [CHU Necker], CHU Necker - Enfants Malades [AP-HP]-Assistance publique - Hôpitaux de Paris (AP-HP) (APHP), Univ Erlangen Nurnberg, La Jolla Institute for Allergy and Immunology, Laboratoire épidémiologie et oncogénèse des tumeurs digestives, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Garvan Institute for Medical Research, and Institute for Research in Biomedicine
- Subjects
MESH: Interleukin-12 ,0301 basic medicine ,MESH: Interferon-gamma ,MESH: Pedigree ,medicine.medical_treatment ,Immunology ,Population ,Mycobacterium Infections, Nontuberculous ,Biology ,Interleukin-23 ,Article ,Mycobacterium ,Interferon-gamma ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunity ,MESH: Mycobacterium ,medicine ,Humans ,Interferon gamma ,education ,Interleukin 12 receptor, beta 1 subunit ,MESH: Interleukin-23 ,education.field_of_study ,MESH: Humans ,General Medicine ,MESH: Mycobacterium Infections, Nontuberculous ,Natural killer T cell ,Interleukin-12 ,Immunity, Innate ,Pedigree ,3. Good health ,030104 developmental biology ,Cytokine ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Interleukin 12 ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,MESH: Immunity, Innate ,030215 immunology ,medicine.drug - Abstract
International audience; Hundreds of patients with autosomal recessive, complete IL-12p40 or IL-12Rβ1 deficiency have been diagnosed over the last 20 years. They typically suffer from invasive mycobacteriosis and, occasionally, from mucocutaneous candidiasis. Susceptibility to these infections is thought to be due to impairments of IL-12-dependent IFN-γ immunity and IL-23-dependent IL-17A/IL-17F immunity, respectively. We report here patients with autosomal recessive, complete IL-12Rβ2 or IL-23R deficiency, lacking responses to IL-12 or IL-23 only, all of whom, unexpectedly, display mycobacteriosis without candidiasis. We show that αβ T, γδ T, B, NK, ILC1, and ILC2 cells from healthy donors preferentially produce IFN-γ in response to IL-12, whereas NKT cells and MAIT cells preferentially produce IFN-γ in response to IL-23. We also show that the development of IFN-γ-producing CD4 + T cells, including, in particular , mycobacterium-specific TH1* cells (CD45RA− CCR6+), is dependent on both IL-12 and IL-23. Last, we show that IL12RB1, IL12RB2, and IL23R have similar frequencies of deleterious variants in the general population. The comparative rarity of symptomatic patients with IL-12Rβ2 or IL-23R deficiency, relative to IL-12Rβ1 deficiency , is, therefore, due to lower clinical penetrance. There are fewer symptomatic IL-23R-and IL-12Rβ2-deficient than IL-12Rβ1-deficient patients, not because these genetic disorders are rarer, but because the isolated absence of IL-12 or IL-23 is, in part, compensated by the other cytokine for the production of IFN-γ, thereby providing some protection against mycobacteria. These experiments of nature show that human IL-12 and IL-23 are both required for optimal IFN-γ dependent immunity to mycobacteria, both individually and much more so cooperatively.
- Published
- 2018
- Full Text
- View/download PDF
30. SeqTailor: a user-friendly webserver for the extraction of DNA or protein sequences from next-generation sequencing data
- Author
-
Peng Zhang, Jean-Laurent Casanova, Yuval Itan, Bertrand Boisson, and Laurent Abel
- Subjects
chemistry.chemical_compound ,chemistry ,Phylogenetic tree ,Computer science ,RNA splicing ,Sequence alignment ,Computational biology ,Genome ,DNA ,DNA sequencing - Abstract
Human whole-genome sequencing generally reveals about 4,000,000 genetic variants, including 20,000 coding variants, in each individual studied. These data are mostly stored as VCF-format files. Although many variant analysis methods accept VCF files as input, many other tools require DNA or protein sequences, particularly for splicing prediction, sequence alignment, phylogenetic analysis, and structure prediction. However, there is currently no existing online tool for extracting DNA or protein sequences for genomic variants from VCF files with user-defined parameters in a user-friendly, efficient, and standardized manner. We developed the SeqTailor webserver to bridge this gap. It can be used for the rapid extraction of (1) DNA sequences around genetic variants, with customizable window sizes, from the hg19 or hg38 human reference genomes; and (2) protein sequences encoded by the DNA sequences around genetic variants, with built-in SnpEff annotation and customizable window sizes, from human canonical transcripts. The SeqTailor webserver streamlines the sequence extraction process, and accelerates the analysis of genetic variant data with software requiring DNA or protein sequences. SeqTailor will facilitate the study of human genomic variation, by increasing the feasibility of sequence-based analysis and prediction. The SeqTailor webserver is freely available from http://shiva.rockefeller.edu/SeqTailor/.
- Published
- 2018
- Full Text
- View/download PDF
31. Severe influenza pneumonitis in children with inherited TLR3 deficiency
- Author
-
Mary Hasek, Isabelle Meyts, Michael D. Keller, Flore Rozenberg, Fabien G. Lafaille, Priya Luthra, Lazaro Lorenzo, Marie Celard, Jie Chen, Eduardo J. Garcia Reino, Tatiana Kochetkov, Laurent Abel, Catherine Vedrinne, Yuval Itan, Olivier Gilliaux, Hye Kyung Lim, Soraya Boucherit, Benedetta Bigio, Jordan S. Orange, Nicholas Hernandez, Nicolas Goudin, Paul Bastard, Sarah X.L. Huang, Hans-Willem Snoeck, Jean-Laurent Casanova, Michael J. Ciancanelli, Gaspard Kerner, Kerry Dobbs, Shen-Ying Zhang, Luigi D. Notarangelo, Qian Zhang, and Adolfo García-Sastre
- Subjects
0301 basic medicine ,Male ,viruses ,NF-KAPPA-B ,Inheritance Patterns ,STAT2 DEFICIENCY ,Research & Experimental Medicine ,medicine.disease_cause ,RIG-I ,0302 clinical medicine ,Fatal Outcome ,Loss of Function Mutation ,Influenza A virus ,Immunology and Allergy ,Child ,Lung ,Research Articles ,virus diseases ,hemic and immune systems ,Penetrance ,3. Good health ,VIRUS ENCEPHALITIS ,Protein Transport ,Medicine, Research & Experimental ,Child, Preschool ,Female ,Life Sciences & Biomedicine ,Encephalitis ,STRUCTURAL BASIS ,Heterozygote ,HERPES-SIMPLEX ENCEPHALITIS ,Immunology ,Induced Pluripotent Stem Cells ,Mutation, Missense ,chemical and pharmacologic phenomena ,DOUBLE-STRANDED-RNA ,PANDEMIC INFLUENZA ,Article ,03 medical and health sciences ,Immunity ,Influenza, Human ,medicine ,Humans ,Alleles ,Pneumonitis ,Science & Technology ,business.industry ,TOLL-LIKE RECEPTOR-3 ,Infant, Newborn ,Infant ,Heterozygote advantage ,Epithelial Cells ,Pneumonia ,Fibroblasts ,medicine.disease ,Toll-Like Receptor 3 ,030104 developmental biology ,Poly I-C ,IRF7 ,Interferons ,business ,030217 neurology & neurosurgery ,GROWTH-RETARDATION - Abstract
The authors report three unrelated children with inherited TLR3 deficiency, impaired TLR3-dependent, IFN-α/β– and/or -λ–mediated, pulmonary epithelial cell–intrinsic immunity to influenza A virus, and life-threatening influenza pneumonitis., Autosomal recessive IRF7 and IRF9 deficiencies impair type I and III IFN immunity and underlie severe influenza pneumonitis. We report three unrelated children with influenza A virus (IAV) infection manifesting as acute respiratory distress syndrome (IAV-ARDS), heterozygous for rare TLR3 variants (P554S in two patients and P680L in the third) causing autosomal dominant (AD) TLR3 deficiency. AD TLR3 deficiency can underlie herpes simplex virus-1 (HSV-1) encephalitis (HSE) by impairing cortical neuron-intrinsic type I IFN immunity to HSV-1. TLR3-mutated leukocytes produce normal levels of IFNs in response to IAV. In contrast, TLR3-mutated fibroblasts produce lower levels of IFN-β and -λ, and display enhanced viral susceptibility, upon IAV infection. Moreover, the patients’ iPSC-derived pulmonary epithelial cells (PECs) are susceptible to IAV. Treatment with IFN-α2b or IFN-λ1 rescues this phenotype. AD TLR3 deficiency may thus underlie IAV-ARDS by impairing TLR3-dependent, type I and/or III IFN–mediated, PEC-intrinsic immunity. Its clinical penetrance is incomplete for both IAV-ARDS and HSE, consistent with their typically sporadic nature.
- Published
- 2018
32. Genomic Signatures of Selective Pressures and Introgression from Archaic Hominins at Human Innate Immunity Genes
- Author
-
Etienne Patin, Laurent Abel, Lluis Quintana-Murci, Yuval Itan, Jean-Laurent Casanova, Matthieu Deschamps, Guillaume Laval, Maud Fagny, Génétique Evolutive Humaine - Human Evolutionary Genetics, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Cellule Pasteur UPMC, Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut Pasteur [Paris] (IP), Rockefeller University [New York], Human genetics of infectious diseases: Complex predisposition (Equipe Inserm U1163), Imagine - Institut des maladies génétiques (IMAGINE - U1163), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Howard Hughes Medical Institute (HHMI), Service d'immuno-hématologie pédiatrique [CHU Necker], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), This work was supported by the Institut Pasteur, the Centre National de la Recherche Scientifique (CNRS), and the Agence Nationale de la Recherche (ANR) grants: 'DEMOCHIPS' ANR-12-BSV7-0012, 'IEIHSEER' ANR-14-CE14-0008-02, and 'TBPATHGEN' ANR-14-CE14-0007-02. The laboratory of L.Q.-M. has received funding from the French Government’s Investissement d’Avenir program, Laboratoire d’Excellence 'Integrative Biology of Emerging Infectious Diseases' (grant no. ANR-10-LABX-62-IBEID), and from the European Research Council under the European Union’s Seventh Framework Programme (FP/2007–2013)/ERC Grant Agreement No. 281297., ANR-12-BSV7-0012,demochips,Inférence de l'histoire démographique à partie des grands jeux de données de polymorphisme A.D.N.(2012), ANR-14-CE14-0008,IEIHSEER,L'encéphalite Herpétique de l'enfant résulte de déficits héréditaires d'immunité contre l'HSV-1: une exception ou une règle?(2014), ANR-14-CE14-0007,TBPATHGEN,Dissection de la pathogenèse de la tuberculose par l'identification de défauts monogéniques de l'immunité dans les formes pédiatriques sévères de la maladie(2014), ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), European Project: 281297,EC:FP7:ERC,ERC-2011-StG_20101109,EVOIMMUNOPOP(2012), Fagny, Maud, BLANC - Inférence de l'histoire démographique à partie des grands jeux de données de polymorphisme A.D.N. - - demochips2012 - ANR-12-BSV7-0012 - BLANC - VALID, Appel à projets générique - L'encéphalite Herpétique de l'enfant résulte de déficits héréditaires d'immunité contre l'HSV-1: une exception ou une règle? - - IEIHSEER2014 - ANR-14-CE14-0008 - Appel à projets générique - VALID, Appel à projets générique - Dissection de la pathogenèse de la tuberculose par l'identification de défauts monogéniques de l'immunité dans les formes pédiatriques sévères de la maladie - - TBPATHGEN2014 - ANR-14-CE14-0007 - Appel à projets générique - VALID, Integrative Biology of Emerging Infectious Diseases - - IBEID2010 - ANR-10-LABX-0062 - LABX - VALID, and Human Evolutionary Immunogenomics: population genetic variation in immune responses - EVOIMMUNOPOP - - EC:FP7:ERC2012-01-01 - 2017-08-31 - 281297 - VALID
- Subjects
0301 basic medicine ,MESH: Selection, Genetic ,Biology ,Polymorphism, Single Nucleotide ,Genome ,Article ,03 medical and health sciences ,Negative selection ,Immunity ,Genetics ,Animals ,Humans ,Genetics(clinical) ,MESH: Animals ,Selection, Genetic ,1000 Genomes Project ,Gene ,Genetics (clinical) ,Neanderthals ,MESH: Neanderthals ,MESH: Humans ,[SDV.GEN.GPO]Life Sciences [q-bio]/Genetics/Populations and Evolution [q-bio.PE] ,Innate immune system ,MESH: Polymorphism, Single Nucleotide ,Adaptation, Physiological ,MESH: Adaptation, Physiological ,Immunity, Innate ,030104 developmental biology ,Evolutionary biology ,MESH: Genome-Wide Association Study ,[SDV.GEN.GPO] Life Sciences [q-bio]/Genetics/Populations and Evolution [q-bio.PE] ,MESH: Immunity, Innate ,Human genome ,Adaptation ,Genome-Wide Association Study - Abstract
International audience; 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.
- Published
- 2016
- Full Text
- View/download PDF
33. Impairment of immunity to Candida and Mycobacterium in humans with bi-allelic RORC mutations
- Author
-
Jean-Pierre de Villartay, Luigi D. Notarangelo, Caroline Deswarte, Danielle T. Avery, Rubén Martínez-Barricarte, Masao Kobayashi, Claire Soudais, Dina Averbuch, Fabienne Jabot-Hanin, Egídio Torrado, Chizuru Okada, Stuart G. Tangye, Yuval Itan, Despina Moshous, Bertrand Boisson, Cindy S. Ma, Anne Puel, M. Lagos, Capucine Picard, Mohammed Alzahrani, Jamie Rossjohn, Rabih Halwani, Elissa K. Deenick, Jeffrey J. Fountain, Jamal Shamma, Jacinta Bustamante, Mélanie Migaud, Natalie Wong, Dan Engelhard, Daniela Latorre, Aziz Belkadi, Sylvain Breton, Sylvain Latour, Federico Mele, Janet Markle, Marcela Gonzalez, Hiyam Marzouqa, Sophie Hambleton, Stéphanie Boisson-Dupuis, Laurent Abel, James McCluskey, Federica Sallusto, Alessandro Sette, Peter D. Arkwright, Saleh Al-Muhsen, Olivier Lantz, Jean-Laurent Casanova, Lauren A. Henderson, Andrea M. Cooper, Cecilia S. Lindestam Arlehamn, and Satoshi Okada
- Subjects
0303 health sciences ,Multidisciplinary ,biology ,T cell ,Gene rearrangement ,Mucocutaneous Candidiasis ,medicine.disease ,biology.organism_classification ,Chronic Candidiasis ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,RAR-related orphan receptor gamma ,Immunity ,Immunology ,medicine ,Chronic mucocutaneous candidiasis ,Candida albicans ,030304 developmental biology ,030215 immunology - Abstract
A surprising immune twist for RORC The immune system needs its full array of soldiers—including cells and the molecules they secrete—to optimally protect the host. When this isn't the case, minor infections can become chronic or even deadly. Markle et al. report the discovery of seven individuals carrying loss-of-function mutations in RORC, which encodes the transcription factors RORγ and RORγT. These individuals lacked immune cells that produce the cytokine interleukin-17, causing them to suffer from chronic candidiasis. RORC-deficient individuals also exhibited impaired immunity to mycobacterium, probably due to reduced production of the cytokine interferon-γ, a molecule not known to require RORC for its induction. Science , this issue p. 606
- Published
- 2015
- Full Text
- View/download PDF
34. Inherited IL-17RC deficiency in patients with chronic mucocutaneous candidiasis
- Author
-
Miguel Galicchio, Yun Ling, Figen Dogu, Aydan Ikinciogullari, Capucine Picard, Anne Puel, Romain Lévy, Yuval Itan, Laurent Abel, Julien Cottineau, Sophie Cypowyj, Yildiz Camcioglu, Jacinta Bustamante, Mélanie Migaud, Nicolas Goudin, Alexandre Bolze, Jean-Laurent Casanova, Serdar Nepesov, Bertrand Boisson, Aziz Belkadi, and Caner Aytekin
- Subjects
Adult ,Male ,Immunology ,Mucocutaneous zone ,macromolecular substances ,Article ,Immunity ,Candida albicans ,Humans ,Immunology and Allergy ,Medicine ,Sex organ ,Allele ,Chronic mucocutaneous candidiasis ,Child ,Adaptor Proteins, Signal Transducing ,biology ,business.industry ,Candidiasis, Chronic Mucocutaneous ,Homozygote ,Interleukin-17 ,Skin Diseases, Genetic ,Receptors, Interleukin ,medicine.disease ,biology.organism_classification ,Tumor Necrosis Factor Receptor-Associated Peptides and Proteins ,Corpus albicans ,Female ,Interleukin 17 ,business - Abstract
Autosomal-recessive IL-17RA, IL-17RC, and ACT1 deficiencies and autosomal-dominant IL-17F deficiency in humans underlie susceptibility to chronic mucocutaneous candidiasis., Chronic mucocutaneous candidiasis (CMC) is characterized by recurrent or persistent infections of the skin, nail, oral, and genital mucosae with Candida species, mainly C. albicans. Autosomal-recessive (AR) IL-17RA and ACT1 deficiencies and autosomal-dominant IL-17F deficiency, each reported in a single kindred, underlie CMC in otherwise healthy patients. We report three patients from unrelated kindreds, aged 8, 12, and 37 yr with isolated CMC, who display AR IL-17RC deficiency. The patients are homozygous for different nonsense alleles that prevent the expression of IL-17RC on the cell surface. The defect is complete, abolishing cellular responses to IL-17A and IL-17F homo- and heterodimers. However, in contrast to what is observed for the IL-17RA– and ACT1-deficient patients tested, the response to IL-17E (IL-25) is maintained in these IL-17RC–deficient patients. These experiments of nature indicate that human IL-17RC is essential for mucocutaneous immunity to C. albicans but is otherwise largely redundant.
- Published
- 2015
- Full Text
- View/download PDF
35. Whole exome sequencing in inborn errors of immunity: use the power but mind the limits
- Author
-
Leen Moens, Yuval Itan, Giorgia Bucciol, Erika Van Nieuwenhove, and Isabelle Meyts
- Subjects
0301 basic medicine ,Genetics ,business.industry ,Genome, Human ,Immunology ,DNA Mutational Analysis ,Immunity ,Infant, Newborn ,High-Throughput Nucleotide Sequencing ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,030104 developmental biology ,Neonatal Screening ,Exome Sequencing ,Immunology and Allergy ,Medicine ,Humans ,Exome ,business ,Exome sequencing - Abstract
Next-generation sequencing, especially whole exome sequencing (WES), has revolutionized the molecular diagnosis of inborn errors of immunity. This review summarizes the generation and analysis of next-generation sequencing data.The focus is on prioritizing strategies for unveiling the potential disease-causing variant. We also highlighted oversights and imperfections of WES and targeted panel sequencing, as well as the need for functional validation.The information is crucial for a judicious use of WES by researchers, but even more so by the clinical immunologist.
- Published
- 2017
36. Inherited BCL10 deficiency impairs hematopoietic and nonhematopoietic immunity
- Author
-
Rita Álvarez, Eduardo López-Collazo, Lucia del Pino, Rubén Martínez-Barricarte, Antonio Ferreira, Mariana Díaz-Almirón, Victor Toledano, Carolina Cubillos-Zapata, Enrique Hernández-Jiménez, Rebeca Rodríguez-Pena, Marina S. Mazariegos, Bertrand Boisson, Sonia García-Gómez, Yuval Itan, Juan Torres, Eduardo López-Granados, José R. Regueiro, Anaïs Jiménez-Reinoso, José L. Unzueta-Roch, Rebeca Pérez de Diego, Jean-Laurent Casanova, and Silvia Sánchez-Ramón
- Subjects
Cell type ,T-Lymphocytes ,T cell ,CARD11 ,Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immunity ,medicine ,Animals ,Humans ,Myeloid Cells ,Immunodeficiency ,B cell ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,B-Lymphocytes ,0303 health sciences ,Genetic Diseases, Inborn ,Immunologic Deficiency Syndromes ,NF-kappa B ,General Medicine ,Fibroblasts ,B-Cell CLL-Lymphoma 10 Protein ,Acquired immune system ,medicine.disease ,BCL10 ,Hematopoiesis ,3. Good health ,Disease Models, Animal ,medicine.anatomical_structure ,Gene Expression Regulation ,Child, Preschool ,030220 oncology & carcinogenesis ,Immunology ,Research Article - Abstract
Heterotrimers composed of B cell CLL/lymphoma 10 (BCL10), mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1), and caspase recruitment domain-containing (CARD) family adaptors play a role in NF-κB activation and have been shown to be involved in both the innate and the adaptive arms of immunity in murine models. Moreover, individuals with inherited defects of MALT1, CARD9, and CARD11 present with immunological and clinical phenotypes. Here, we characterized a case of autosomal-recessive, complete BCL10 deficiency in a child with a broad immunodeficiency, including defects of both hematopoietic and nonhematopoietic immunity. The patient died at 3 years of age and was homozygous for a loss-of-expression, loss-of-function BCL10 mutation. The effect of BCL10 deficiency was dependent on the signaling pathway, and, for some pathways, the cell type affected. Despite the noted similarities to BCL10 deficiency in mice, including a deficient adaptive immune response, human BCL10 deficiency in this patient resulted in a number of specific features within cell populations. Treatment of the patient's myeloid cells with a variety of pathogen-associated molecular pattern molecules (PAMPs) elicited a normal response; however, NF-κB-mediated fibroblast functions were dramatically impaired. The results of this study indicate that inherited BCL10 deficiency should be considered in patients with combined immunodeficiency with B cell, T cell, and fibroblast defects.
- Published
- 2014
- Full Text
- View/download PDF
37. Defining Risk Groups to Yellow Fever Vaccine-Associated Viscerotropic Disease in the Absence of Denominator Data
- Author
-
John C. Pezzullo, Joel E. Cohen, Yuval Itan, Stephen J. Seligman, and Jean-Laurent Casanova
- Subjects
Adult ,Male ,Pediatrics ,medicine.medical_specialty ,Adolescent ,Population ,Yellow fever vaccine ,Disease ,Vaccines, Attenuated ,Young Adult ,Risk Factors ,Virology ,Yellow Fever ,Confidence Intervals ,Odds Ratio ,Cluster Analysis ,Humans ,Medicine ,Fraction (mathematics) ,Risk factor ,Child ,education ,Aged ,education.field_of_study ,Denominator data ,business.industry ,Vaccination ,Yellow Fever Vaccine ,Infant ,Articles ,Odds ratio ,Middle Aged ,Confidence interval ,Infectious Diseases ,Child, Preschool ,Female ,Parasitology ,Yellow fever virus ,business ,Demography ,medicine.drug - Abstract
Several risk groups are known for the rare but serious, frequently fatal, viscerotropic reactions following live yellow fever virus vaccine (YEL-AVD). Establishing additional risk groups is hampered by ignorance of the numbers of vaccinees in factor-specific risk groups thus preventing their use as denominators in odds ratios (ORs). Here, we use an equation to calculate ORs using the prevalence of the factor-specific risk group in the population who remain well. The 95% confidence limits and P values can also be calculated. Moreover, if the estimate of the prevalence is imprecise, discrimination analysis can indicate the prevalence at which the confidence interval results in an OR of ~1 revealing if the prevalence might be higher without yielding a non-significant result. These methods confirm some potential risk groups for YEL-AVD and cast doubt on another. They should prove useful in situations in which factor-specific risk group denominator data are not available. ated to assist in the documentation of other risk groups. Odds ratios (ORs) are frequently used to identify risk groups, and to quantify the magnitude of the risk. Their calcu- lation typically requires knowledge of the numbers of subjects who had an outcome and had or did not have the predisposing factor (numerator data) and the numbers of subjects in both categories who remained unaffected (denominator data). Con- sequently, if denominator data are not available, ORs cannot be calculated by the traditional method. The current report illustrates how estimates of the fraction of the exposed popula- tion with the suspected risk factor who remain well can be used instead to calculate the OR and confidence interval (CI), how the effect of uncertainty in the estimate of that fraction can be evaluated, and how P values can be calculated using the CI.
- Published
- 2014
- Full Text
- View/download PDF
38. Correction to: Use of Genetic Testing for Primary Immunodeficiency Patients
- Author
-
Sergio D. Rosenzweig, Janet Chou, Joud Hajjar, David Hagin, Elena W Y Hsieh, Roshini S. Abraham, Jennifer Heimall, Hillary S. Hernandez-Trujillo, Rebecca A. Marsh, James W. Verbsky, Lisa Kobrynski, Yuval Itan, Monica G. Lawrence, Jack J. Bleesing, Richard L. Wasserman, Kenneth Paris, Sarah E. Henrickson, Jordan S. Orange, and Troy R. Torgerson
- Subjects
medicine.medical_specialty ,medicine.diagnostic_test ,GeneralLiterature_INTRODUCTORYANDSURVEY ,business.industry ,InformationSystems_INFORMATIONSYSTEMSAPPLICATIONS ,Published Erratum ,Immunology ,MEDLINE ,medicine.disease ,GeneralLiterature_MISCELLANEOUS ,Family medicine ,medicine ,Primary immunodeficiency ,Immunology and Allergy ,business ,Genetic testing - Abstract
The original version of this article unfortunately contained mistakes in some of the author names and affiliations. The correct list of author names and affiliations is below, with the corrections in bold.
- Published
- 2018
- Full Text
- View/download PDF
39. Su1028 – Whole-Genome Sequencing of African Americans Identifies Novel Rare Variants Associated with Inflammatory Bowel Disease
- Author
-
Hari K. Somineni, Talin Haritunians, Claire L. Simpson, David J. Cutler, David Okou, Yuval Itan, Suresh Venkateswaran, Christine Stevens, Lisa W. Datta, Tanvi A. Dhere, Mark Lazarev, Emory African American IBD Consortium, Michael E. Zwick, Judy H. Cho, Mark J. Daly, Dermot McGovern, Steven R. Brant, and Subra Kugathasan
- Subjects
Genetics ,Whole genome sequencing ,Hepatology ,Gastroenterology ,medicine ,Biology ,medicine.disease ,Inflammatory bowel disease - Published
- 2019
- Full Text
- View/download PDF
40. Genetic, immunological, and clinical features of patients with bacterial and fungal infections due to inherited IL-17RA deficiency
- Author
-
Nobuyuki Hyakuna, Saleh Al Muhsen, Takaki Asano, Caner Aytekin, Didier Bronnimann, Yuval Itan, Erika Della Mina, Figen Dogu, Chittibabu Vatte, Laurent Abel, Amein Al Ali, Caini Liu, Vivien Béziat, Xiaoxia Li, Julie Toubiana, Marie Ferneiny, Romain Lévy, Masao Kobayashi, Anne Puel, Ekrem Unal, Suzan A AlKhater, Cyril Cyrus, Louis Yi Ann Chai, Fatih Celmeli, Fabian Hauck, Satoshi Okada, Stéphanie Boisson-Dupuis, Osamu Ohara, Turkan Patiroglu, Jacinta Bustamante, Mélanie Migaud, Kunihiko Moriya, Matías Oleastro, Jean-Laurent Casanova, Aydan Ikinciogullari, Florian Gothe, Natalia Tahuil, Yildiz Camcioglu, Christoph Klein, Aicha Salhi, Ling Yun, and Serdar Nepesov
- Subjects
Male ,0301 basic medicine ,T-Lymphocytes ,0302 clinical medicine ,Receptors ,2.1 Biological and endogenous factors ,Missense mutation ,Aetiology ,Chronic mucocutaneous candidiasis ,Child ,Receptor ,Immunodeficiency ,Skin ,Candida ,Pediatric ,Receptors, Interleukin-17 ,Multidisciplinary ,Interleukin-17 ,Homozygote ,Candidiasis ,Bacterial Infections ,Pedigree ,Transmembrane domain ,Infectious Diseases ,PNAS Plus ,Child, Preschool ,Female ,Infection ,Mucocutaneous zone ,Genes, Recessive ,Biology ,Immunophenotyping ,Open Reading Frames ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,Genetics ,medicine ,Humans ,Recessive ,Allele ,Preschool ,Gene ,Alleles ,Family Health ,Cell Membrane ,Infant, Newborn ,Infant ,Fibroblasts ,Newborn ,medicine.disease ,HEK293 Cells ,Emerging Infectious Diseases ,Good Health and Well Being ,030104 developmental biology ,Mycoses ,Genes ,Mutation ,Immunology ,immunodeficiency ,Genome-Wide Association Study ,030215 immunology - 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.
- Published
- 2016
- Full Text
- View/download PDF
41. Autosomal Recessive Cardiomyopathy Presenting as Acute Myocarditis
- Author
-
Soraya Boucherit, Damien Bonnet, Lazaro Lorenzo, Raphaelle Quint, Serkan Belkaya, Laurent Abel, Aurélie Cobat, Yuval Itan, Fanny Bajolle, Amy R Kontorovich, Cecile Stoven, Minji Byun, Shen-Ying Zhang, Bruce D. Gelb, Sylvie Di Filippo, Rebecca Josowitz, Sonia Mulero-Navarro, and Jean-Laurent Casanova
- Subjects
0301 basic medicine ,Male ,Pathology ,medicine.medical_specialty ,Myocarditis ,Viral Myocarditis ,viruses ,Induced Pluripotent Stem Cells ,Cardiomyopathy ,030204 cardiovascular system & hematology ,Compound heterozygosity ,Virus ,TNNI3 ,03 medical and health sciences ,0302 clinical medicine ,Immunity ,Interferon ,Medicine ,Humans ,Myocytes, Cardiac ,business.industry ,medicine.disease ,Enterovirus B, Human ,Toll-Like Receptor 3 ,030104 developmental biology ,STAT1 Transcription Factor ,Case-Control Studies ,Immunology ,Host-Pathogen Interactions ,Female ,Cardiology and Cardiovascular Medicine ,business ,Cardiomyopathies ,medicine.drug - Abstract
Background Myocarditis is inflammation of the heart muscle that can follow various viral infections. Why children only rarely develop life-threatening acute viral myocarditis (AVM), given that the causal viral infections are common, is unknown. Genetic lesions might underlie such susceptibilities. Mouse genetic studies demonstrated that interferon (IFN)-α/β immunity defects increased susceptibility to virus-induced myocarditis. Moreover, variations in human TLR3, a potent inducer of IFNs, were proposed to underlie AVM. Objectives This study sought to evaluate the hypothesis that human genetic factors may underlie AVM in previously healthy children. Methods We tested the role of TLR3-IFN immunity using human induced pluripotent stem cell-derived cardiomyocytes. We then performed whole-exome sequencing of 42 unrelated children with acute myocarditis (AM), some with proven viral causes. Results We found that TLR3- and STAT1-deficient cardiomyocytes were not more susceptible to Coxsackie virus B3 (CVB3) infection than control cells. Moreover, CVB3 did not induce IFN-α/β and IFN-α/β-stimulated genes in control cardiomyocytes. Finally, exogenous IFN-α did not substantially protect cardiomyocytes against CVB3. We did not observe a significant enrichment of rare variations in TLR3- or IFN-α/β-related genes. Surprisingly, we found that homozygous but not heterozygous rare variants in genes associated with inherited cardiomyopathies were significantly enriched in AM-AVM patients compared with healthy individuals (p = 2.22E-03) or patients with other diseases (p = 1.08E-04). Seven of 42 patients (16.7%) carried rare biallelic (homozygous or compound heterozygous) nonsynonymous or splice-site variations in 6 cardiomyopathy-associated genes (BAG3, DSP, PKP2, RYR2, SCN5A, or TNNI3). Conclusions Previously silent recessive defects of the myocardium may predispose to acute heart failure presenting as AM, notably after common viral infections in children.
- Published
- 2016
42. Genetic Diagnosis Using Whole Exome Sequencing in Common Variable Immunodeficiency
- Author
-
Charles A. Filion, Patrick Maffucci, Yuval Itan, Jean-Laurent Casanova, Bertrand Boisson, Lei Shang, and Charlotte Cunningham-Rundles
- Subjects
0301 basic medicine ,Immunology ,Population ,Biology ,Bioinformatics ,DNA sequencing ,whole exome sequencing ,LRBA ,Hypogammaglobulinemia ,genetic diagnosis ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Immunology and Allergy ,education ,Gene ,Exome sequencing ,Original Research ,Genetics ,education.field_of_study ,Common variable immunodeficiency ,common variable immunodeficiency ,medicine.disease ,Phenotype ,3. Good health ,030104 developmental biology ,next-generation sequencing ,primary immunodeficiencies ,030215 immunology - Abstract
Whole exome sequencing (WES) has proven an effective tool for the discovery of genetic defects in patients with primary immunodeficiencies (PIDs). However, success in dissecting the genetic etiology of common variable immunodeficiency (CVID) has been limited. We outline a practical framework for using WES to identify causative genetic defects in these subjects. Whole Exome Sequencing (WES) was performed on 50 subjects diagnosed with CVID who had at least one of the following criteria: early onset, autoimmune/inflammatory manifestations, low B lymphocytes, and/or familial history of hypogammaglobulinemia. Following alignment and variant calling, exomes were screened for mutations in 269 PID-causing genes. Variants were filtered based on the mode of inheritance and reported frequency in the general population. Each variant was assessed by study of familial segregation and computational predictions of deleteriousness. Out of 433 variations in PID-associated genes, we identified 17 probable disease-causing mutations in 15 patients (30%). These variations were rare or private and included mono-allelic mutations in NFKB1, STAT3, CTLA4, PIK3CD, and IKZF1, and bi-allelic mutations in LRBA and STXBP2. Forty-two other damaging variants were found but were not considered likely disease-causing based on mode of inheritance and/or patient phenotype. WES combined with analysis of PID-associated genes is a cost-effective approach to identify disease-causing mutations in CVID patients with severe phenotypes and was successful in 30% of our cohort. As targeted therapeutics are becoming the mainstay of treatment for non-infectious manifestations in CVID, this approach will improve management of patients with more severe phenotypes.
- Published
- 2016
- Full Text
- View/download PDF
43. Contrasting exome constancy and regulatory region variation in the gene encoding CYP3A4: an examination of the extent and potential implications
- Author
-
Naser Ansari-Pour, Yuval Itan, Ripudaman K. Bains, Olivia J. Creemer, Ayele Tarekegn, Christopher A Plaster, Endashaw Bekele, Neil Bradman, and Rosemary Ekong
- Subjects
0301 basic medicine ,Adult ,Linkage disequilibrium ,Population ,Biology ,Regulatory Sequences, Nucleic Acid ,030226 pharmacology & pharmacy ,Linkage Disequilibrium ,03 medical and health sciences ,0302 clinical medicine ,Genetic variation ,Genetics ,Coding region ,Cytochrome P-450 CYP3A ,Humans ,Exome ,General Pharmacology, Toxicology and Pharmaceutics ,1000 Genomes Project ,Allele ,education ,Molecular Biology ,Genetics (clinical) ,education.field_of_study ,Haplotype ,Genetic Variation ,030104 developmental biology ,Haplotypes ,Molecular Medicine - Abstract
Objective CYP3A4 expression varies up to 100-fold among individuals, and, to date, genetic causes remain elusive. As a major drug-metabolizing enzyme, elucidation of such genetic causes would increase the potential for introducing personalized dose adjustment of therapies involving CYP3A4 drug substrates. The foetal CYP3A isoform, CYP3A7, is reported to be expressed in ∼10% of European adults and may thus contribute towards the metabolism of endogenous substances and CYP3A drug substrates. However, little is known about the distribution of the variant expressed in the adult. Methods We resequenced the exons, flanking introns, regulatory elements and 3'UTR of CYP3A4 in five Ethiopian populations and incorporated data from the 1000 Genomes Project. Using bioinformatic analysis, we assessed likely consequences of observed CYP3A4 genomic variation. We also conducted the first extensive geographic survey of alleles associated with adult expression of CYP3A7 - that is, CYP3A7*1B and CYP3A7*1C. Results and conclusion Ethiopia contained 60 CYP3A4 variants (26 novel) and more variants (>1%) than all non-African populations combined. No nonsynonymous mutation was found in the homozygous form or at more than 2.8% in any population. Seventy-nine per cent of haplotypes contained 3'UTR and/or regulatory region variation with striking pairwise population differentiation, highlighting the potential for interethnic variation in CYP3A4 expression. Conversely, coding region variation showed that significant interethnic variation is unlikely at the protein level. CYP3A7*1C was found at up to 17.5% in North African populations and in significant linkage disequilibrium with CYP3A5*3, indicating that adult expression of the foetal isoform is likely to be accompanied by reduced or null expression of CYP3A5.
- Published
- 2016
44. Evolutionary genetic dissection of human interferons
- Author
-
Lluis Quintana-Murci, Christiane Bouchier, Luis B. Barreiro, Jean-Laurent Casanova, Etienne Patin, Simona Fornarino, Manuela Sironi, Yuval Itan, Matteo Fumagalli, Jeremy Manry, Magali Tichit, and Guillaume Laval
- Subjects
Male ,Immunology ,Population ,Mutation, Missense ,Biology ,medicine.disease_cause ,Article ,Evolution, Molecular ,Negative selection ,Immune system ,Genetic variation ,medicine ,Humans ,Immunology and Allergy ,Selection, Genetic ,education ,Gene ,Genetics ,Mutation ,education.field_of_study ,Natural selection ,Codon, Nonsense ,Virus Diseases ,Female ,Interferons ,Function (biology) - Abstract
As revealed by population genetic analyses, different human interferon genes evolved under distinct selective constraints and signatures of positive selection vary according to geographic region, suggesting that some sequence changes may have conferred an advantage by increasing resistance to viral infection., Interferons (IFNs) are cytokines that play a key role in innate and adaptive immune responses. Despite the large number of immunological studies of these molecules, the relative contributions of the numerous IFNs to human survival remain largely unknown. Here, we evaluated the extent to which natural selection has targeted the human IFNs and their receptors, to provide insight into the mechanisms that govern host defense in the natural setting. We found that some IFN-α subtypes, such as IFN-α6, IFN-α8, IFN-α13, and IFN-α14, as well as the type II IFN-γ, have evolved under strong purifying selection, attesting to their essential and nonredundant function in immunity to infection. Conversely, selective constraints have been relaxed for other type I IFNs, particularly for IFN-α10 and IFN-ε, which have accumulated missense or nonsense mutations at high frequencies within the population, suggesting redundancy in host defense. Finally, type III IFNs display geographically restricted signatures of positive selection in European and Asian populations, indicating that genetic variation at these genes has conferred a selective advantage to the host, most likely by increasing resistance to viral infection. Our population genetic analyses show that IFNs differ widely in their biological relevance, and highlight evolutionarily important determinants of host immune responsiveness.
- Published
- 2011
- Full Text
- View/download PDF
45. Evolution of lactase persistence: an example of human niche construction
- Author
-
Joachim Burger, Dallas M. Swallow, Yuval Itan, Mark G. Thomas, Adam Powell, Anke Liebert, Pascale Gerbault, and Mathias Currat
- Subjects
Adult ,0106 biological sciences ,Asia ,Natural selection ,medicine.medical_treatment ,Lactose ,Biology ,010603 evolutionary biology ,01 natural sciences ,Gene Expression Regulation, Enzymologic ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Gene Frequency ,Lactase persistence ,medicine ,Animals ,Humans ,Computer Simulation ,Neolithic ,Allele ,Domestication ,Lactase ,ddc:599.9 ,030304 developmental biology ,2. Zero hunger ,Genetics ,0303 health sciences ,Genetic Variation ,Articles ,Biological Evolution ,Europe ,Domestic animals ,Dairying ,Niche construction ,Genetics, Population ,Milk ,Africa ,Trait ,Local environment ,Cattle ,General Agricultural and Biological Sciences ,Single mutation - Abstract
Niche construction is the process by which organisms construct important components of their local environment in ways that introduce novel selection pressures. Lactase persistence is one of the clearest examples of niche construction in humans. Lactase is the enzyme responsible for the digestion of the milk sugar lactose and its production decreases after the weaning phase in most mammals, including most humans. Some humans, however, continue to produce lactase throughout adulthood, a trait known as lactase persistence. In European populations, a single mutation (−13910*T) explains the distribution of the phenotype, whereas several mutations are associated with it in Africa and the Middle East. Current estimates for the age of lactase persistence-associated alleles bracket those for the origins of animal domestication and the culturally transmitted practice of dairying. We report new data on the distribution of−13910*Tand summarize genetic studies on the diversity of lactase persistence worldwide. We review relevant archaeological data and describe three simulation studies that have shed light on the evolution of this trait in Europe. These studies illustrate how genetic and archaeological information can be integrated to bring new insights to the origins and spread of lactase persistence. Finally, we discuss possible improvements to these models.
- Published
- 2011
- Full Text
- View/download PDF
46. Detecting Gene Duplications in the Human Lineage
- Author
-
Mark G. Thomas, Kevin Bryson, and Yuval Itan
- Subjects
Chimpanzee genome project ,Genetics ,Lineage (genetic) ,Gene duplication ,Human genome ,Gene Annotation ,Computational biology ,Biology ,Genome ,Gene ,Genetics (clinical) ,Segmental duplication - Abstract
Gene duplications represent an important class of evolutionary events that is likely to have contributed to the unique human phenotype in the short evolutionary time since the human-chimpanzee divergence. With the availability of both human and chimpanzee genome drafts in high coverage re-sequencing assemblies and the high annotation quality of most human genes, it should now be possible to identify all human lineage-specific gene duplication events (human inparalogues) and a few pioneering studies have attempted to do that. However, the different levels of coverage in the human and chimpanzee's genomes assemblies, and the differing levels of gene annotation, have led to problematic assumptions and oversimplifications in the algorithms and the datasets used to detect human lineage-specific gene duplications. In this study, we have developed a set of bioinformatic tools to overcome a number of the conceptual problems that are prevalent in previous studies and have collected a reliable and representative set of human inparalogues.
- Published
- 2010
- Full Text
- View/download PDF
47. Lactose digestion and the evolutionary genetics of lactase persistence
- Author
-
Catherine J. E. Ingram, Charlotte A. Mulcare, Yuval Itan, Dallas M. Swallow, and Mark G. Thomas
- Subjects
medicine.medical_treatment ,Molecular Sequence Data ,Population ,Population genetics ,Cell Cycle Proteins ,Lactose ,Single-nucleotide polymorphism ,Biology ,Polymorphism, Single Nucleotide ,Evolution, Molecular ,Lactose Intolerance ,Lactose Tolerance Test ,Genetics ,medicine ,Humans ,Allele ,education ,Alleles ,Genetics (clinical) ,Lactase ,education.field_of_study ,Base Sequence ,Human evolutionary genetics ,Haplotype ,Minichromosome Maintenance Complex Component 6 ,Lactase persistence ,Genetics, Population ,Haplotypes ,Africa ,Digestion - Abstract
It has been known for some 40 years that lactase production persists into adult life in some people but not in others. However, the mechanism and evolutionary significance of this variation have proved more elusive, and continue to excite the interest of investigators from different disciplines. This genetically determined trait differs in frequency worldwide and is due to cis-acting polymorphism of regulation of lactase gene expression. A single nucleotide polymorphism located 13.9 kb upstream from the lactase gene (C-13910 > T) was proposed to be the cause, and the −13910*T allele, which is widespread in Europe was found to be located on a very extended haplotype of 500 kb or more. The long region of haplotype conservation reflects a recent origin, and this, together with high frequencies, is evidence of positive selection, but also means that −13910*T might be an associated marker, rather than being causal of lactase persistence itself. Doubt about function was increased when it was shown that the original SNP did not account for lactase persistence in most African populations. However, the recent discovery that there are several other SNPs associated with lactase persistence in close proximity (within 100 bp), and that they all reside in a piece of sequence that has enhancer function in vitro, does suggest that they may each be functional, and their occurrence on different haplotype backgrounds shows that several independent mutations led to lactase persistence. Here we provide access to a database of worldwide distributions of lactase persistence and of the C-13910*T allele, as well as reviewing lactase molecular and population genetics and the role of selection in determining present day distributions of the lactase persistence phenotype.
- Published
- 2008
- Full Text
- View/download PDF
48. Loss of B Cells in Patients with Heterozygous Mutations in IKAROS
- Author
-
Asbjørg Stray-Pedersen, Jeana S. Bush, William K. Dolen, Julie E. Niemela, Harry R. Hill, Monica G. Lawrence, Nancy H. Augustine, Charlotte Cunningham-Rundles, Sarah T. South, Katherine R. Calvo, Keith R. Pierce, Jordan K. Abbott, Aurélie Cobat, Erwin W. Gelfand, Betty B. Wray, Sundar Ganesan, Attila Kumánovics, Sergio D. Rosenzweig, Jordan S. Orange, Yuval Itan, Patrick Maffucci, Hanne Sørmo Sorte, Mary Ellen Conley, Karl V. Voelkerding, Janine Reichenbach, Hye Sun Kuehn, Thomas B. Martins, Thomas A. Fleisher, Jean-Laurent Casanova, Bertrand Boisson, Seraina Prader, University of Zurich, and Conley, Mary Ellen
- Subjects
0301 basic medicine ,Proband ,Adult ,Male ,Heterozygote ,Adolescent ,610 Medicine & health ,2700 General Medicine ,medicine.disease_cause ,Article ,Hypogammaglobulinemia ,03 medical and health sciences ,Ikaros Transcription Factor ,Antigens, CD ,Bone Marrow ,medicine ,Humans ,Exome ,Lymphocyte Count ,Child ,Gene ,Genetics ,Mutation ,B-Lymphocytes ,business.industry ,Common variable immunodeficiency ,Bone Marrow Examination ,General Medicine ,Sequence Analysis, DNA ,medicine.disease ,Pedigree ,030104 developmental biology ,Common Variable Immunodeficiency ,10036 Medical Clinic ,Child, Preschool ,Immunoglobulin G ,Female ,business ,Chromosomes, Human, Pair 7 ,Comparative genomic hybridization - Abstract
Common variable immunodeficiency (CVID) is characterized by late-onset hypogammaglobulinemia in the absence of predisposing factors. The genetic cause is unknown in the majority of cases, and less than 10% of patients have a family history of the disease. Most patients have normal numbers of B cells but lack plasma cells.We used whole-exome sequencing and array-based comparative genomic hybridization to evaluate a subset of patients with CVID and low B-cell numbers. Mutant proteins were analyzed for DNA binding with the use of an electrophoretic mobility-shift assay (EMSA) and confocal microscopy. Flow cytometry was used to analyze peripheral-blood lymphocytes and bone marrow aspirates.Six different heterozygous mutations in IKZF1, the gene encoding the transcription factor IKAROS, were identified in 29 persons from six families. In two families, the mutation was a de novo event in the proband. All the mutations, four amino acid substitutions, an intragenic deletion, and a 4.7-Mb multigene deletion involved the DNA-binding domain of IKAROS. The proteins bearing missense mutations failed to bind target DNA sequences on EMSA and confocal microscopy; however, they did not inhibit the binding of wild-type IKAROS. Studies in family members showed progressive loss of B cells and serum immunoglobulins. Bone marrow aspirates in two patients had markedly decreased early B-cell precursors, but plasma cells were present. Acute lymphoblastic leukemia developed in 2 of the 29 patients.Heterozygous mutations in the transcription factor IKAROS caused an autosomal dominant form of CVID that is associated with a striking decrease in B-cell numbers. (Funded by the National Institutes of Health and others.).
- Published
- 2016
- Full Text
- View/download PDF
49. Whole-exome sequencing for detecting inborn errors of immunity: overview and perspectives
- Author
-
Yuval Itan, Barbara Bosch, and Isabelle Meyts
- Subjects
0301 basic medicine ,Open science ,inborn errors of immunity ,medicine.medical_treatment ,Genetic counseling ,Review ,Bioinformatics ,General Biochemistry, Genetics and Molecular Biology ,Targeted therapy ,03 medical and health sciences ,Immunity ,Genetic etiology ,Genetics of the Immune System ,Medicine ,whole-exome sequencing ,General Pharmacology, Toxicology and Pharmaceutics ,Exome sequencing ,General Immunology and Microbiology ,business.industry ,Articles ,General Medicine ,3. Good health ,Open data ,030104 developmental biology ,primary immune deficiencies ,business - Abstract
The study of inborn errors of immunity is based on a comprehensive clinical description of the patient's phenotype and the elucidation of the underlying molecular mechanisms and their genetic etiology. Deciphering the pathogenesis is key to genetic counseling and the development of targeted therapy. This review shows the power of whole-exome sequencing in detecting inborn errors of immunity along five central steps taken in whole-exome sequencing analysis. In parallel, we highlight the challenges for the clinical and scientific use of the method and how these hurdles are currently being addressed. We end by ruminating on major areas in the field open to future research. ispartof: F1000Research vol:6 pages:2056- ispartof: location:England status: published
- Published
- 2017
- Full Text
- View/download PDF
50. IMMUNODEFICIENCIES. Impairment of immunity to Candida and Mycobacterium in humans with bi-allelic RORC mutations
- Author
-
Satoshi, Okada, Janet G, Markle, Elissa K, Deenick, Federico, Mele, Dina, Averbuch, Macarena, Lagos, Mohammed, Alzahrani, Saleh, Al-Muhsen, Rabih, Halwani, Cindy S, Ma, Natalie, Wong, Claire, Soudais, Lauren A, Henderson, Hiyam, Marzouqa, Jamal, Shamma, Marcela, Gonzalez, Rubén, Martinez-Barricarte, Chizuru, Okada, Danielle T, Avery, Daniela, Latorre, Caroline, Deswarte, Fabienne, Jabot-Hanin, Egidio, Torrado, Jeffrey, Fountain, Aziz, Belkadi, Yuval, Itan, Bertrand, Boisson, Mélanie, Migaud, Cecilia S Lindestam, Arlehamn, Alessandro, Sette, Sylvain, Breton, James, McCluskey, Jamie, Rossjohn, Jean-Pierre, de Villartay, Despina, Moshous, Sophie, Hambleton, Sylvain, Latour, Peter D, Arkwright, Capucine, Picard, Olivier, Lantz, Dan, Engelhard, Masao, Kobayashi, Laurent, Abel, Andrea M, Cooper, Luigi D, Notarangelo, Stéphanie, Boisson-Dupuis, Anne, Puel, Federica, Sallusto, Jacinta, Bustamante, Stuart G, Tangye, and Jean-Laurent, Casanova
- Subjects
Receptors, Antigen, T-Cell, alpha-beta ,T-Lymphocytes ,DNA Mutational Analysis ,Thymus Gland ,Article ,Interferon-gamma ,Mice ,Candida albicans ,Animals ,Humans ,Exome ,Child ,Tuberculosis, Pulmonary ,Alleles ,Candidiasis, Chronic Mucocutaneous ,Interleukin-17 ,Immunity ,Receptors, Antigen, T-Cell, gamma-delta ,Mycobacterium tuberculosis ,Nuclear Receptor Subfamily 1, Group F, Member 3 ,Mycobacterium bovis ,Pedigree ,Child, Preschool ,Mutation ,Cattle ,Female ,Severe Combined Immunodeficiency ,Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor ,Tuberculosis, Bovine - Abstract
Human inborn errors of immunity mediated by the cytokines interleukin-17A and interleukin-17F (IL-17A/F) underlie mucocutaneous candidiasis, whereas inborn errors of interferon-γ (IFN-γ) immunity underlie mycobacterial disease. We report the discovery of bi-allelic RORC loss-of-function mutations in seven individuals from three kindreds of different ethnic origins with both candidiasis and mycobacteriosis. The lack of functional RORγ and RORγT isoforms resulted in the absence of IL-17A/F-producing T cells in these individuals, probably accounting for their chronic candidiasis. Unexpectedly, leukocytes from RORγ- and RORγT-deficient individuals also displayed an impaired IFN-γ response to Mycobacterium. This principally reflected profoundly defective IFN-γ production by circulating γδ T cells and CD4(+)CCR6(+)CXCR3(+) αβ T cells. In humans, both mucocutaneous immunity to Candida and systemic immunity to Mycobacterium require RORγ, RORγT, or both.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.