17 research outputs found

    Human IFN-Îł immunity to mycobacteria is governed by both IL-12 and IL-23

    Get PDF
    Hundreds of patients with autosomal recessive, complete IL-12p40 or IL-12Rß1 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-? immunity and IL-23–dependent IL-17A/IL-17F immunity, respectively. We report here patients with autosomal recessive, complete IL- 12Rß2 or IL-23R deficiency, lacking responses to IL-12 or IL- 23 only, all of whom, unexpectedly, display mycobacteriosis without candidiasis. We show that aß T, ?d T, B, NK, ILC1, and ILC2 cells from healthy donors preferentially produce IFN-? in response to IL-12, whereas NKT cells and MAIT cells preferentially produce IFN-? in response to IL-23. We also show that the development of IFN-?–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-12Rß2 or IL-23R deficiency, relative to IL-12Rß1 deficiency, is, therefore, due to lower clinical penetrance. There are fewer symptomatic IL-23R– and IL-12Rß2–deficient than IL-12Rß1–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-?, thereby providing some protection against mycobacteria. These experiments of nature show that human IL-12 and IL-23 are both required for optimal IFN-?–dependent immunity to mycobacteria, both individually and much more so cooperatively

    Recherche des facteurs gĂ©nĂ©tiques contrĂŽlant la rĂ©ponse Ă  l’infection par Mycobacterium tuberculosis et le dĂ©veloppement d’une tuberculose maladie

    No full text
    Tuberculosis remains a major public health concern, with approximately 10.4 million new cases and 1.8 million deaths due to the disease in 2015 according to WHO. While an estimated one third of the world population is estimated to be infected with Mycobacterium tuberculosis, only about 10% of infected individuals go on to develop a clinical disease. Among them, half will declare the disease in the 2 years following infection, which is generally considered as primary tuberculosis. The other patients will develop the disease more distant in time of primary infection, sometimes several tens of years latter; these are classical pulmonary forms in adults. In humans, the role of genetic factors have been demonstrated in the development of active tuberculosis, in pulmonary forms as in disseminated forms in childhood, et also in the control of M.tuberculosis infection. Nevertheless, most of these genetic factors remain to identify. The first aim of my PhD was to identify genetic factors controlling in vitro interferon-gamma production phenotypes (IGRA) after exposure to M.tuberculosis in a sample of 590 subjects who were in contact with a proven tuberculous patient in Val-de-Marne, Paris suburbs, and in a second time, to try to replicate the findings in a south African familial sample where the tuberculosis is highly endemic. For this purpose, I first performed genome-wide genetic linkage analysis for several quantitative IGRA phenotypes. They led to identify 2 major loci (p<10-4) replicated in South-Africa and linked to the interferon-gamma production induced by live BCG for the first one, and for the second one, by the specific part of the ESAT6 antigen of M.tuberculosis (absent from most of environmental mycobacteria and from BCG), independently of intrinsic ability to respond to mycobacteria. The second step was an association study in the identified linkage regions. A variant associated to the specific ESAT6 phenotype was found (p<10-5), which was significantly contributing to the linkage peak (p<0.001) and previously reported as eQTL of ZXDC gene. The second objective of my PhD was the identification of rare genetic variants underlying the development of pulmonary tuberculosis in infected individuals. To this end, I compared exome data from 120 tuberculous patients and 136 infected individuals without any clinical symptoms. All of them were from Morocco. This study resulted in the lighting of BTNL2 gene, very closed to the HLA region, in which around 10% of patients had a rare loss of function variant whereas the controls didn’t have any.La tuberculose, causĂ©e par Mycobacterium tuberculosis, connaĂźt actuellement une rĂ©surgence inquiĂ©tante, et l’OMS estime Ă  plus de 10 millions le nombre de nouveaux cas cliniques en 2015 avec environ 1,8 millions de dĂ©cĂšs dus Ă  la maladie. Environ un tiers de la population mondiale est exposĂ©e Ă  M.tuberculosis, et aprĂšs exposition, la plupart des individus sont infectĂ©s par la mycobactĂ©rie. La grande majoritĂ© (~90%) des individus infectĂ©s ne prĂ©sentera jamais de symptomatologie clinique. Parmi les 10% qui dĂ©veloppent la maladie, environ la moitiĂ© le fera dans les deux annĂ©es suivant l’infection, ce qui est en gĂ©nĂ©ral considĂ©rĂ© comme une forme primaire de tuberculose. Les autres patients prĂ©senteront leur maladie Ă  distance de l’infection primaire (parfois plusieurs dizaines d’annĂ©es plus tard) ; il s’agit des formes pulmonaires classiques de l’adulte. Chez l’homme, le rĂŽle de certains facteurs gĂ©nĂ©tiques a Ă©tĂ© maintenant dĂ©montrĂ© dans le dĂ©veloppement d’une tuberculose active, Ă  la fois la tuberculose pulmonaire de l’adulte et les formes plus dissĂ©minĂ©es de l’enfant, et aussi dans le contrĂŽle de l’infection tuberculeuse. Cependant, la plus grande part de ces facteurs gĂ©nĂ©tiques reste Ă  identifier. Le premier objectif de ma thĂšse Ă©tait d'identifier les facteurs gĂ©nĂ©tiques de l'hĂŽte modulant les phĂ©notypes immunologiques de production d'InterfĂ©ron gamma in vitro (IGRA) aprĂšs exposition à M. tuberculosis dans un Ă©chantillon de 590 individus ayant Ă©tĂ© en contact avec un cas avĂ©rĂ© de tuberculose dans le Val de Marne, en rĂ©gion parisienne. Puis, dans un second temps, de voir si les facteurs trouvĂ©s pouvaient ĂȘtre rĂ©pliquĂ©es dans un Ă©chantillon familial d'Afrique du Sud, zone de trĂšs forte endĂ©mie tuberculeuse. Pour cela, j'ai tout d'abord rĂ©alisĂ© des analyses de liaison gĂ©nĂ©tique Ă  l'Ă©chelle du gĂ©nome entier sur plusieurs phĂ©notypes quantitatifs d'IGRA. Celles-ci ont permis de mettre en Ă©vidence 2 loci majeurs (p < 10-4) rĂ©pliquĂ©s en Afrique du Sud et liĂ©s Ă  la production d'interfĂ©ron gamma induite pour l’un par le bacille du BCG, et pour l’autre, par la part spĂ©cifique de l'antigĂšne ESAT6 de M. tuberculosis (absent de la plupart des mycobactĂ©ries environnementales et du BCG), indĂ©pendamment de la capacitĂ© intrinsĂšque de rĂ©ponse aux mycobactĂ©ries. La seconde Ă©tape a consistĂ© en la rĂ©alisation d'une Ă©tude d'association sur les rĂ©gions de liaison ainsi identifiĂ©es. Un variant associĂ© au phĂ©notype spĂ©cifique de l’ESAT6 (p < 10-5) a ainsi Ă©tĂ© trouvĂ©, variant contribuant de maniĂšre significative au pic de liaison prĂ©cĂ©demment dĂ©couvert (p<0.001) et ayant Ă©tĂ© rapportĂ© comme modulant l’expression du gĂšne ZXDC. Le second objectif de la thĂšse concernait l’identification de variants gĂ©nĂ©tiques rares sous-jacents Ă  la dĂ©claration d’une tuberculose pulmonaire chez les individus infectĂ©s par le bacille. A cette fin, j’ai comparĂ© les exomes de 120 patients tuberculeux Ă  ceux de 136 individus infectĂ©s par le bacille mais non malades, tous originaires du Maroc. Cette Ă©tude m’a permis d’identifier le gĂšne BTNL2, en bordure de la rĂ©gion HLA, dans lequel prĂšs de 10% des patients comportaient un variant rare perte de fonction contrairement aux contrĂŽles qui n’en prĂ©sentaient aucun

    Search for genetic factors controlling the response to infection by Mycobacterium tuberculosis and the development of clinical tuberculosis

    No full text
    La tuberculose, causĂ©e par Mycobacterium tuberculosis, connaĂźt actuellement une rĂ©surgence inquiĂ©tante, et l’OMS estime Ă  plus de 10 millions le nombre de nouveaux cas cliniques en 2015 avec environ 1,8 millions de dĂ©cĂšs dus Ă  la maladie. Environ un tiers de la population mondiale est exposĂ©e Ă  M.tuberculosis, et aprĂšs exposition, la plupart des individus sont infectĂ©s par la mycobactĂ©rie. La grande majoritĂ© (~90%) des individus infectĂ©s ne prĂ©sentera jamais de symptomatologie clinique. Parmi les 10% qui dĂ©veloppent la maladie, environ la moitiĂ© le fera dans les deux annĂ©es suivant l’infection, ce qui est en gĂ©nĂ©ral considĂ©rĂ© comme une forme primaire de tuberculose. Les autres patients prĂ©senteront leur maladie Ă  distance de l’infection primaire (parfois plusieurs dizaines d’annĂ©es plus tard) ; il s’agit des formes pulmonaires classiques de l’adulte. Chez l’homme, le rĂŽle de certains facteurs gĂ©nĂ©tiques a Ă©tĂ© maintenant dĂ©montrĂ© dans le dĂ©veloppement d’une tuberculose active, Ă  la fois la tuberculose pulmonaire de l’adulte et les formes plus dissĂ©minĂ©es de l’enfant, et aussi dans le contrĂŽle de l’infection tuberculeuse. Cependant, la plus grande part de ces facteurs gĂ©nĂ©tiques reste Ă  identifier. Le premier objectif de ma thĂšse Ă©tait d'identifier les facteurs gĂ©nĂ©tiques de l'hĂŽte modulant les phĂ©notypes immunologiques de production d'InterfĂ©ron gamma in vitro (IGRA) aprĂšs exposition à M. tuberculosis dans un Ă©chantillon de 590 individus ayant Ă©tĂ© en contact avec un cas avĂ©rĂ© de tuberculose dans le Val de Marne, en rĂ©gion parisienne. Puis, dans un second temps, de voir si les facteurs trouvĂ©s pouvaient ĂȘtre rĂ©pliquĂ©es dans un Ă©chantillon familial d'Afrique du Sud, zone de trĂšs forte endĂ©mie tuberculeuse. Pour cela, j'ai tout d'abord rĂ©alisĂ© des analyses de liaison gĂ©nĂ©tique Ă  l'Ă©chelle du gĂ©nome entier sur plusieurs phĂ©notypes quantitatifs d'IGRA. Celles-ci ont permis de mettre en Ă©vidence 2 loci majeurs (p < 10-4) rĂ©pliquĂ©s en Afrique du Sud et liĂ©s Ă  la production d'interfĂ©ron gamma induite pour l’un par le bacille du BCG, et pour l’autre, par la part spĂ©cifique de l'antigĂšne ESAT6 de M. tuberculosis (absent de la plupart des mycobactĂ©ries environnementales et du BCG), indĂ©pendamment de la capacitĂ© intrinsĂšque de rĂ©ponse aux mycobactĂ©ries. La seconde Ă©tape a consistĂ© en la rĂ©alisation d'une Ă©tude d'association sur les rĂ©gions de liaison ainsi identifiĂ©es. Un variant associĂ© au phĂ©notype spĂ©cifique de l’ESAT6 (p < 10-5) a ainsi Ă©tĂ© trouvĂ©, variant contribuant de maniĂšre significative au pic de liaison prĂ©cĂ©demment dĂ©couvert (p<0.001) et ayant Ă©tĂ© rapportĂ© comme modulant l’expression du gĂšne ZXDC. Le second objectif de la thĂšse concernait l’identification de variants gĂ©nĂ©tiques rares sous-jacents Ă  la dĂ©claration d’une tuberculose pulmonaire chez les individus infectĂ©s par le bacille. A cette fin, j’ai comparĂ© les exomes de 120 patients tuberculeux Ă  ceux de 136 individus infectĂ©s par le bacille mais non malades, tous originaires du Maroc. Cette Ă©tude m’a permis d’identifier le gĂšne BTNL2, en bordure de la rĂ©gion HLA, dans lequel prĂšs de 10% des patients comportaient un variant rare perte de fonction contrairement aux contrĂŽles qui n’en prĂ©sentaient aucun.Tuberculosis remains a major public health concern, with approximately 10.4 million new cases and 1.8 million deaths due to the disease in 2015 according to WHO. While an estimated one third of the world population is estimated to be infected with Mycobacterium tuberculosis, only about 10% of infected individuals go on to develop a clinical disease. Among them, half will declare the disease in the 2 years following infection, which is generally considered as primary tuberculosis. The other patients will develop the disease more distant in time of primary infection, sometimes several tens of years latter; these are classical pulmonary forms in adults. In humans, the role of genetic factors have been demonstrated in the development of active tuberculosis, in pulmonary forms as in disseminated forms in childhood, et also in the control of M.tuberculosis infection. Nevertheless, most of these genetic factors remain to identify. The first aim of my PhD was to identify genetic factors controlling in vitro interferon-gamma production phenotypes (IGRA) after exposure to M.tuberculosis in a sample of 590 subjects who were in contact with a proven tuberculous patient in Val-de-Marne, Paris suburbs, and in a second time, to try to replicate the findings in a south African familial sample where the tuberculosis is highly endemic. For this purpose, I first performed genome-wide genetic linkage analysis for several quantitative IGRA phenotypes. They led to identify 2 major loci (p<10-4) replicated in South-Africa and linked to the interferon-gamma production induced by live BCG for the first one, and for the second one, by the specific part of the ESAT6 antigen of M.tuberculosis (absent from most of environmental mycobacteria and from BCG), independently of intrinsic ability to respond to mycobacteria. The second step was an association study in the identified linkage regions. A variant associated to the specific ESAT6 phenotype was found (p<10-5), which was significantly contributing to the linkage peak (p<0.001) and previously reported as eQTL of ZXDC gene. The second objective of my PhD was the identification of rare genetic variants underlying the development of pulmonary tuberculosis in infected individuals. To this end, I compared exome data from 120 tuberculous patients and 136 infected individuals without any clinical symptoms. All of them were from Morocco. This study resulted in the lighting of BTNL2 gene, very closed to the HLA region, in which around 10% of patients had a rare loss of function variant whereas the controls didn’t have any

    Calculating rfPred scores with package rfPred

    No full text
    Exome sequencing is becoming a standard tool for gene mapping of monogenic diseases. Given the vast amount of data generated by Next Generation Sequencing techniques, identification of disease causal variants is like finding a needle in a haystack. The impact assessment and the prioritization of potential pathogenic variants are expected to reduce work in biological validation, which is long and costly. One of the possible approaches to determine the most probable deleterious variants in individual exomes is to use protein function alteration prediction algorithms. This package proposes a new method [1] based on five previously described algorithms (SIFT [2], Polyphen2 [3], LRT [4], PhyloP [5] and MutationTaster [6]) compiled in the dbNSFP database [7]. A functional meta-score is derived from a random forest method trained on a dataset of 61,500 nonsynonymous SNPs. On Two independent validation datasets, the random forest method appears to be globally better than each of the algorithms separately or in combination in a logistic regression model. rfPred scores have been precalculated and made available for all the possible non-synonymous SNPs of human exome. 2 Computing rfPred scores After having launched the R software, the user must load the rfPred package with:&gt; library(rfPred) Several packages whose rfPred is depending on will also be loaded (especially Rsamtools). The package contains one main function- rfPred_scores()- which returns the rfPred scores. It works as follows: the user gives as input a list of variants for which he/she wants the corresponding scores and the function looks for these variants in a data base which stores the pre-calculated scores. It avoids re-calculating the scores for each request. The list of variants can ben either a data.frame, or the path to a VCF (Variant Call Format) file as a character string or a GRanges object (see package GenomicRanges on Bioconductor [8]). For example, the list of variants can be the data frame variant_list_Y:&gt; data(variant_list_Y)&gt; print(variant_list_Y) chr pos ref alt unipro

    Association Between Plasma Rituximab Concentration and the Risk of Major Relapse in Antineutrophil Cytoplasmic Antibody–Associated Vasculitides During Rituximab Maintenance Therapy

    No full text
    International audienceObjective Interindividual variability in response to rituximab remains unexplored in antineutrophil cytoplasmic antibody (ANCA)–associated vasculitides. Rituximab pharmacokinetics (PK) and pharmacodynamics (PD) as well as genetic polymorphisms could contribute to variability. This ancillary study of the MAINRITSAN 2 trial aimed to explore the relationship between rituximab plasma concentration, genetic polymorphisms in PK/PD candidate genes, and clinical outcomes. Methods Patients included in the MAINRITSAN2 trial ( ClinicalTrials.gov identifier: NCT01731561) were randomized to receive a 500‐mg fixed‐schedule rituximab infusion or an individually tailored regimen. Rituximab plasma concentrations at month 3 (CM3) were assessed. DNA samples (n = 53) were genotyped for single‐nucleotide polymorphisms within 88 putative PK/PD candidate genes. The relationship between PK/PD outcomes and genetic variants was investigated using logistic linear regression in additive and recessive genetic models. Results One hundred and thirty‐five patients were included. The frequency of underexposed patients (<4 ÎŒg/ml) in the fixed‐schedule group was statistically lower compared to that in the tailored‐infusion group (2.0% versus 18.0%; P = 0.02, respectively). Low rituximab plasma concentration at 3 months (CM3 <4 ÎŒg/ml) was an independent risk factor for major relapse (odds ratio 6.56 [95% confidence interval (95% CI) 1.26–34.09]; P = 0.025) at month 28 (M28). A sensitivity survival analysis also identified CM3 <4 ÎŒg/ml as an independent risk factor for major relapse (hazard ratio [HR] 4.81 [95% CI 1.56–14.82]; P = 0.006) and relapse (HR 2.70 [95% CI 1.02–7.15]; P = 0.046). STAT4 rs2278940 and PRKCA rs8076312 were significantly associated with CM3 but not with major relapse onset at M28. Conclusion These results suggest that drug monitoring could be useful to individualize the schedule of rituximab administration within the maintenance phase

    Major Loci on Chromosomes 8q and 3q Control Interferon Îł Production Triggered by Bacillus Calmette-Guerin and 6-kDa Early Secretory Antigen Target, Respectively, in Various Populations

    No full text
    Background. Interferon Îł (IFN-Îł) release assays (IGRAs) provide an in vitro measurement of antimycobacterial immunity that is widely used as a test for Mycobacterium tuberculosis infection. IGRA outcomes are highly heritable in various populations, but the nature of the involved genetic factors remains unknown. Methods. We conducted a genome-wide linkage analysis of IGRA phenotypes in families from a tuberculosis household contact study in France and a replication study in families from South Africa to confirm the loci identified. Results. We identified a major locus on chromosome 8q controlling IFN-Îł production in response to stimulation with live bacillus Calmette-Guerin (BCG; LOD score, 3.81; P = 1.40 × 10(−5)). We also detected a second locus, on chromosome 3q, that controlled IFN-Îł levels in response to stimulation with 6-kDa early secretory antigen target, when accounting for the IFN-Îł production shared with that induced by BCG (LOD score, 3.72; P = 1.8 × 10(−5)). Both loci were replicated in South African families, where tuberculosis is hyperendemic. These loci differ from those previously identified as controlling the response to the tuberculin skin test (TST1 and TST2) and the production of TNF-α (TNF1). Conclusions. The identification of 2 new linkage signals in populations of various ethnic origins living in different M. tuberculosis exposure settings provides new clues about the genetic control of human antimycobacterial immunity

    TUBB1 mutations cause thyroid dysgenesis associated with abnormal platelet physiology

    Get PDF
    International audienceThe genetic causes of congenital hypothyroidism due to thyroid dysgenesis (TD) remain largely unknown. We identified three novel TUBB1 gene mutations that co-segregated with TD in three distinct families leading to 1.1% of TUBB1 mutations in TD study cohort. TUBB1 (Tubulin, Beta 1 Class VI) encodes for a member of the b-tubulin protein family. TUBB1 gene is expressed in the developing and adult thyroid in humans and mice. All three TUBB1 mutations lead to non-functional a/b-tubulin dimers that cannot be incorporated into microtubules. In mice, Tubb1 knockout disrupted micro-tubule integrity by preventing b1-tubulin incorporation and impaired thyroid migration and thyroid hormone secretion. In addition, TUBB1 mutations caused the formation of macroplatelets and hyperaggregation of human platelets after stimulation by low doses of agonists. Our data highlight unexpected roles for b1-tubulin in thyroid development and in platelet physiology. Finally, these findings expand the spectrum of the rare paediatric diseases related to mutations in tubulin-coding genes and provide new insights into the genetic background and mechanisms involved in congenital hypothyroidism and thyroid dysgenesis

    Identification of Germline Non-coding Deletions in XIAP Gene Causing XIAP Deficiency Reveals a Key Promoter Sequence

    No full text
    International audiencePurposeX-linked inhibitor of apoptosis protein (XIAP) deficiency, also known as the X-linked lymphoproliferative syndrome of type 2 (XLP-2), is a rare immunodeficiency characterized by recurrent hemophagocytic lymphohistiocytosis, splenomegaly, and inflammatory bowel disease. Variants in XIAP including missense, non-sense, frameshift, and deletions of coding exons have been reported to cause XIAP deficiency. We studied three young boys with immunodeficiency displaying XLP-2-like clinical features. No genetic variation in the coding exons of XIAP was identified by whole-exome sequencing (WES), although the patients exhibited a complete loss of XIAP expression.MethodsTargeted next-generation sequencing (NGS) of the entire locus of XIAP was performed on DNA samples from the three patients. Molecular investigations were assessed by gene reporter expression assays in HEK cells and CRISPR-Cas9 genome editing in primary T cells.ResultsNGS of XIAP identified three distinct non-coding deletions in the patients that were predicted to be driven by repetitive DNA sequences. These deletions share a common region of 839 bp that encompassed the first non-coding exon of XIAP and contained regulatory elements and marks specific of an active promoter. Moreover, we showed that among the 839 bp, the exon was transcriptionally active. Finally, deletion of the exon by CRISPR-Cas9 in primary cells reduced XIAP protein expression.ConclusionsThese results identify a key promoter sequence contained in the first non-coding exon of XIAP. Importantly, this study highlights that sequencing of the non-coding exons that are not currently captured by WES should be considered in the genetic diagnosis when no variation is found in coding exons

    Genome-wide association study of resistance to Mycobacterium tuberculosis infection identifies a locus at 10q26.2 in three distinct populations.

    No full text
    The natural history of tuberculosis (TB) is characterized by a large inter-individual outcome variability after exposure to Mycobacterium tuberculosis. Specifically, some highly exposed individuals remain resistant to M. tuberculosis infection, as inferred by tuberculin skin test (TST) or interferon-gamma release assays (IGRAs). We performed a genome-wide association study of resistance to M. tuberculosis infection in an endemic region of Southern Vietnam. We enrolled household contacts (HHC) of pulmonary TB cases and compared subjects who were negative for both TST and IGRA (n = 185) with infected individuals (n = 353) who were either positive for both TST and IGRA or had a diagnosis of TB. We found a genome-wide significant locus on chromosome 10q26.2 with a cluster of variants associated with strong protection against M. tuberculosis infection (OR = 0.42, 95%CI 0.35-0.49, P = 3.71×10-8, for the genotyped variant rs17155120). The locus was replicated in a French multi-ethnic HHC cohort and a familial admixed cohort from a hyper-endemic area of South Africa, with an overall OR for rs17155120 estimated at 0.50 (95%CI 0.45-0.55, P = 1.26×10-9). The variants are located in intronic regions and upstream of C10orf90, a tumor suppressor gene which encodes an ubiquitin ligase activating the transcription factor p53. In silico analysis showed that the protective alleles were associated with a decreased expression in monocytes of the nearby gene ADAM12 which could lead to an enhanced response of Th17 lymphocytes. Our results reveal a novel locus controlling resistance to M. tuberculosis infection across different populations
    corecore