9 research outputs found

    A comprehensive assessment of demographic, environmental, and host genetic associations with gut microbiome diversity in healthy individuals.

    Get PDF
    BACKGROUND: The gut microbiome is an important determinant of human health. Its composition has been shown to be influenced by multiple environmental factors and likely by host genetic variation. In the framework of the Milieu Intérieur Consortium, a total of 1000 healthy individuals of western European ancestry, with a 1:1 sex ratio and evenly stratified across five decades of life (age 20-69), were recruited. We generated 16S ribosomal RNA profiles from stool samples for 858 participants. We investigated genetic and non-genetic factors that contribute to individual differences in fecal microbiome composition. RESULTS: Among 110 demographic, clinical, and environmental factors, 11 were identified as significantly correlated with α-diversity, ß-diversity, or abundance of specific microbial communities in multivariable models. Age and blood alanine aminotransferase levels showed the strongest associations with microbiome diversity. In total, all non-genetic factors explained 16.4% of the variance. We then searched for associations between > 5 million single nucleotide polymorphisms and the same indicators of fecal microbiome diversity, including the significant non-genetic factors as covariates. No genome-wide significant associations were identified after correction for multiple testing. A small fraction of previously reported associations between human genetic variants and specific taxa could be replicated in our cohort, while no replication was observed for any of the diversity metrics. CONCLUSION: In a well-characterized cohort of healthy individuals, we identified several non-genetic variables associated with fecal microbiome diversity. In contrast, host genetics only had a negligible influence. Demographic and environmental factors are thus the main contributors to fecal microbiome composition in healthy individuals. TRIAL REGISTRATION: ClinicalTrials.gov identifier NCT01699893

    Human genetic variants and age are the strongest predictors of humoral immune responses to common pathogens and vaccines.

    Get PDF
    Humoral immune responses to infectious agents or vaccination vary substantially among individuals, and many of the factors responsible for this variability remain to be defined. Current evidence suggests that human genetic variation influences (i) serum immunoglobulin levels, (ii) seroconversion rates, and (iii) intensity of antigen-specific immune responses. Here, we evaluated the impact of intrinsic (age and sex), environmental, and genetic factors on the variability of humoral response to common pathogens and vaccines. We characterized the serological response to 15 antigens from common human pathogens or vaccines, in an age- and sex-stratified cohort of 1000 healthy individuals (Milieu Intérieur cohort). Using clinical-grade serological assays, we measured total IgA, IgE, IgG, and IgM levels, as well as qualitative (serostatus) and quantitative IgG responses to cytomegalovirus, Epstein-Barr virus, herpes simplex virus 1 and 2, varicella zoster virus, Helicobacter pylori, Toxoplasma gondii, influenza A virus, measles, mumps, rubella, and hepatitis B virus. Following genome-wide genotyping of single nucleotide polymorphisms and imputation, we examined associations between ~ 5 million genetic variants and antibody responses using single marker and gene burden tests. We identified age and sex as important determinants of humoral immunity, with older individuals and women having higher rates of seropositivity for most antigens. Genome-wide association studies revealed significant associations between variants in the human leukocyte antigen (HLA) class II region on chromosome 6 and anti-EBV and anti-rubella IgG levels. We used HLA imputation to fine map these associations to amino acid variants in the peptide-binding groove of HLA-DRβ1 and HLA-DPβ1, respectively. We also observed significant associations for total IgA levels with two loci on chromosome 2 and with specific KIR-HLA combinations. Using extensive serological testing and genome-wide association analyses in a well-characterized cohort of healthy individuals, we demonstrated that age, sex, and specific human genetic variants contribute to inter-individual variability in humoral immunity. By highlighting genes and pathways implicated in the normal antibody response to frequently encountered antigens, these findings provide a basis to better understand disease pathogenesis. ClinicalTrials.gov , NCT01699893

    Genetic determinants of healthy human immune variance: from humoral response to microbiome diversity

    No full text
    Identifying the drivers of the observed interindividual variability of the human immune system is crucial to our understanding of infectious and immune-mediated diseases. The contribution of genetic and non-genetic factors to immunological differences between humans remains largely undefined. The Milieu Intérieur Consortium has established a 1000-person healthy population-based cohort (evenly stratified by sex and age), which represents an unparalleled opportunity for assessing the determinants of human immunologic variance. In this thesis, three population-based studies are presented, all benefiting from the samples and data collected by investigators of the Milieu Intérieur Consortium. Human genome-wide genotyping data, more than 100 environmental, lifestyle and physiological variables, and their combination have been tested for their impact on multiple immune phenotypes. Firstly, we identified the respective contributions of age, sex, and genetics to humoral responses to vaccination and persistent viral infection. We observed that specific variants in the human leukocyte antigen (HLA) region are the strongest genetic determinant of antibody response to common antigens. In the second study, investigation of 166 immuno-phenotypes revealed 15 genetic loci associated with variation of immune cell parameters, mainly of innate immune cells. We attributed an important role to genetic variation in the major histocompatibility complex (MHC) region for these phenotypes and narrowed the signals to probable causal associations in HLA genes. In the third work, forces shaping the gut microbiome composition were investigated. We found a strong influence of several non-genetic factors on overall microbiome diversity and on the abundance of specific bacterial species. We showed as well that genetic factors only play a minor role in gut microbiome composition. Together, these studies quantified the effects of demographic, environment and genetics on the interindividual variability of phenotypes central to the human immune system. Furthermore, they constitute a valuable resource for further explorations of the impact of immune diversity on the individual risk of infections or of immune diseases

    Human genomics of acute liver failure due to hepatitis B virus infection: An exome sequencing study in liver transplant recipients

    Full text link
    Acute liver failure (ALF) or fulminant hepatitis is a rare, yet severe outcome of infection with hepatitis B virus (HBV) that carries a high mortality rate. The occurrence of a life-threatening condition upon infection with a prevalent virus in individuals without known risk factors is suggestive of pathogen-specific immune dysregulation. In the absence of established differences in HBV virulence, we hypothesized that ALF upon primary infection with HBV could be due to rare deleterious variants in the human genome. To search for such variants, we performed exome sequencing in 21 previously healthy adults who required liver transplantation upon fulminant HBV infection and 172 controls that were positive for anti-HBc and anti-HBs but had no clinical history of jaundice or liver disease. After a series of hypothesis-driven filtering steps, we searched for putatively pathogenic variants that were significantly associated with case-control status. We did not find any causal variant or gene, a result that does not support the hypothesis of a shared monogenic basis for human susceptibility to HBV-related ALF in adults. This study represents a first attempt at deciphering the human genetic contribution to the most severe clinical presentation of acute HBV infection in previously healthy individuals

    Additional file 2: of Human genetic variants and age are the strongest predictors of humoral immune responses to common pathogens and vaccines

    No full text
    Figure S1. Principal Component Analysis. Figure S2. Distribution of serological variables, and clinical thresholds. Figure S3. Seroprevalence data. Figure S4. Impact of non-genetic factors on serostatus. Figure S5. Evolution of serostatus with age and sex. Figure S6. Correlations between age and IgG specific to Rubella and T. gondii. Figure S7. QQ plots for logistic regressions preformed in the study. Figure S8. QQ plots for linear regressions preformed on total Ig levels. Figure S9. QQ plots for linear regressions preformed for pathogen-specific IgG levels. Figure S10. QQ plots for burden testing analyses preformed for all binary phenotypes. Figure S11. QQ plots for burden testing analyses preformed for total Ig levels. Figure S12. QQ plots for burden testing analyses preformed for pathogen-specific IgG levels. (DOCX 89996 kb

    Natural variation in the parameters of innate immune cells is preferentially driven by genetic factors

    Get PDF
    A Publisher Correction to this article was published on 03 May 2018International audienceThe quantification and characterization of circulating immune cells provide key indicators of human health and disease. To identify the relative effects of environmental and genetic factors on variation in the parameters of innate and adaptive immune cells in homeostatic conditions, we combined standardized flow cytometry of blood leukocytes and genome-wide DNA genotyping of 1,000 healthy, unrelated people of Western European ancestry. We found that smoking, together with age, sex and latent infection with cytomegalovirus, were the main non-genetic factors that affected variation in parameters of human immune cells. Genome-wide association studies of 166 immunophenotypes identified 15 loci that showed enrichment for disease-associated variants. Finally, we demonstrated that the parameters of innate cells were more strongly controlled by genetic variation than were those of adaptive cells, which were driven by mainly environmental exposure. Our data establish a resource that will generate new hypotheses in immunology and highlight the role of innate immunity in susceptibility to common autoimmune diseases

    Human genetic variants and age are the strongest predictors of humoral immune responses to common pathogens and vaccines

    No full text
    Background: Humoral immune responses to infectious agents or vaccination vary substantially among individuals, and many of the factors responsible for this variability remain to be defined. Current evidence suggests that human genetic variation influences (i) serum immunoglobulin levels, (ii) seroconversion rates, and (iii) intensity of antigen-specific immune responses. Here, we evaluated the impact of intrinsic (age and sex), environmental, and genetic factors on the variability of humoral response to common pathogens and vaccines. Methods: We characterized the serological response to 15 antigens from common human pathogens or vaccines, in an age- and sex-stratified cohort of 1000 healthy individuals (Milieu Intérieur cohort). Using clinical-grade serological assays, we measured total IgA, IgE, IgG, and IgM levels, as well as qualitative (serostatus) and quantitative IgG responses to cytomegalovirus, Epstein-Barr virus, herpes simplex virus 1 and 2, varicella zoster virus, Helicobacter pylori, Toxoplasma gondii, influenza A virus, measles, mumps, rubella, and hepatitis B virus. Following genome-wide genotyping of single nucleotide polymorphisms and imputation, we examined associations between ~5 million genetic variants and antibody responses using single marker and gene burden tests. Results: We identified age and sex as important determinants of humoral immunity, with older individuals and women having higher rates of seropositivity for most antigens. Genome-wide association studies revealed significant associations between variants in the human leukocyte antigen (HLA) class II region on chromosome 6 and anti-EBV and anti-rubella IgG levels. We used HLA imputation to fine map these associations to amino acid variants in the peptide-binding groove of HLA-DRβ1 and HLA-DPβ1, respectively. We also observed significant associations for total IgA levels with two loci on chromosome 2 and with specific KIR-HLA combinations. Conclusions: Using extensive serological testing and genome-wide association analyses in a well-characterized cohort of healthy individuals, we demonstrated that age, sex, and specific human genetic variants contribute to inter-individual variability in humoral immunity. By highlighting genes and pathways implicated in the normal antibody response to frequently encountered antigens, these findings provide a basis to better understand disease pathogenesis
    corecore