Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Cell ; 167(3): 643-656.e17, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27768888

ABSTRACT

Humans differ in the outcome that follows exposure to life-threatening pathogens, yet the extent of population differences in immune responses and their genetic and evolutionary determinants remain undefined. Here, we characterized, using RNA sequencing, the transcriptional response of primary monocytes from Africans and Europeans to bacterial and viral stimuli-ligands activating Toll-like receptor pathways (TLR1/2, TLR4, and TLR7/8) and influenza virus-and mapped expression quantitative trait loci (eQTLs). We identify numerous cis-eQTLs that contribute to the marked differences in immune responses detected within and between populations and a strong trans-eQTL hotspot at TLR1 that decreases expression of pro-inflammatory genes in Europeans only. We find that immune-responsive regulatory variants are enriched in population-specific signals of natural selection and show that admixture with Neandertals introduced regulatory variants into European genomes, affecting preferentially responses to viral challenges. Together, our study uncovers evolutionarily important determinants of differences in host immune responsiveness between human populations.


Subject(s)
Adaptation, Physiological/genetics , Adaptation, Physiological/immunology , Adaptive Immunity , Neanderthals/genetics , Neanderthals/immunology , Adaptive Immunity/genetics , Alleles , Animals , Bacterial Infections/genetics , Bacterial Infections/immunology , Base Sequence , Biological Evolution , Black People/genetics , Gene Expression Regulation , Genetic Variation , Humans , Immune System , Quantitative Trait Loci , RNA/genetics , Selection, Genetic , Sequence Analysis, RNA , Toll-Like Receptors/genetics , Transcription, Genetic , Virus Diseases/genetics , Virus Diseases/immunology , White People/genetics
3.
Nat Immunol ; 19(3): 302-314, 2018 03.
Article in English | MEDLINE | ID: mdl-29476184

ABSTRACT

The 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.


Subject(s)
Genetic Variation/immunology , Immunity, Innate/genetics , Adaptive Immunity/genetics , Adult , Aged , Female , Genome-Wide Association Study , Humans , Immunophenotyping , Male , Middle Aged , Young Adult
4.
Proc Natl Acad Sci U S A ; 115(3): E488-E497, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29282317

ABSTRACT

The contribution of host genetic and nongenetic factors to immunological differences in humans remains largely undefined. Here, we generated bacterial-, fungal-, and viral-induced immune transcriptional profiles in an age- and sex-balanced cohort of 1,000 healthy individuals and searched for the determinants of immune response variation. We found that age and sex affected the transcriptional response of most immune-related genes, with age effects being more stimulus-specific relative to sex effects, which were largely shared across conditions. Although specific cell populations mediated the effects of age and sex on gene expression, including CD8+ T cells for age and CD4+ T cells and monocytes for sex, we detected a direct effect of these intrinsic factors for the majority of immune genes. The mapping of expression quantitative trait loci (eQTLs) revealed that genetic factors had a stronger effect on immune gene regulation than age and sex, yet they affected a smaller number of genes. Importantly, we identified numerous genetic variants that manifested their regulatory effects exclusively on immune stimulation, including a Candida albicans-specific master regulator at the CR1 locus. These response eQTLs were enriched in disease-associated variants, particularly for autoimmune and inflammatory disorders, indicating that differences in disease risk may result from regulatory variants exerting their effects only in the presence of immune stress. Together, this study quantifies the respective effects of age, sex, genetics, and cellular heterogeneity on the interindividual variability of immune responses and constitutes a valuable resource for further exploration in the context of different infection risks or disease outcomes.


Subject(s)
Aging , Gene Expression Regulation/immunology , Genetic Variation , Adult , Aged , Bacteria/immunology , Cohort Studies , Enterotoxins/immunology , Female , Fungi/immunology , Genotype , Humans , Influenza A virus/immunology , Male , Middle Aged , Quantitative Trait Loci , Young Adult
5.
Am J Hum Genet ; 101(6): 977-984, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-29129317

ABSTRACT

From the eighth century onward, the Indian Ocean was the scene of extensive trade of sub-Saharan African slaves via sea routes controlled by Muslim Arab and Swahili traders. Several populations in present-day Pakistan and India are thought to be the descendants of such slaves, yet their history of admixture and natural selection remains largely undefined. Here, we studied the genome-wide diversity of the African-descent Makranis, who reside on the Arabian Sea coast of Pakistan, as well that of four neighboring Pakistani populations, to investigate the genetic legacy, population dynamics, and tempo of the Indian Ocean slave trade. We show that the Makranis are the result of an admixture event between local Baluch tribes and Bantu-speaking populations from eastern or southeastern Africa; we dated this event to ∼300 years ago during the Omani Empire domination. Levels of parental relatedness, measured through runs of homozygosity, were found to be similar across Pakistani populations, suggesting that the Makranis rapidly adopted the traditional practice of endogamous marriages. Finally, we searched for signatures of post-admixture selection at traits evolving under positive selection, including skin color, lactase persistence, and resistance to malaria. We demonstrate that the African-specific Duffy-null blood group-believed to confer resistance against Plasmodium vivax infection-was recently introduced to Pakistan through the slave trade and evolved adaptively in this P. vivax malaria-endemic region. Our study reconstructs the genetic and adaptive history of a neglected episode of the African Diaspora and illustrates the impact of recent admixture on the diffusion of adaptive traits across human populations.


Subject(s)
Asian People/genetics , Black People/genetics , Duffy Blood-Group System/genetics , Enslaved Persons , Malaria, Vivax/immunology , Plasmodium vivax/immunology , Population Dynamics , Quantitative Trait, Heritable , Gene Frequency , Genetic Variation/genetics , Genetics, Population , Humans , Indian Ocean , Pakistan/epidemiology
6.
Hum Mol Genet ; 22(23): 4829-40, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23851028

ABSTRACT

The study of the genetic and selective landscape of immunity genes across primates can provide insight into the existing differences in susceptibility to infection observed between human and non-human primates. Here, we explored how selection has driven the evolution of a key family of innate immunity receptors, the Toll-like receptors (TLRs), in African great ape species. We sequenced the 10 TLRs in various populations of chimpanzees and gorillas, and analysed these data jointly with a human data set. We found that purifying selection has been more pervasive in great apes than in humans. Furthermore, in chimpanzees and gorillas, purifying selection has targeted TLRs irrespectively of whether they are endosomal or cell surface, in contrast to humans where strong selective constraints are restricted to endosomal TLRs. These observations suggest important differences in the relative importance of TLR-mediated pathogen sensing, such as that of recognition of flagellated bacteria by TLR5, between humans and great apes. Lastly, we used a population genetics-phylogenetics method that jointly analyses polymorphism and divergence data to detect fine-scale variation in selection pressures at specific codons within TLR genes. We identified different codons at different TLRs as being under positive selection in each species, highlighting that functional variation at these genes has conferred a selective advantage in immunity to infection to specific primate species. Overall, this study showed that the degree of selection driving the evolution of TLRs has largely differed between human and non-human primates, increasing our knowledge on their respective biological contribution to host defence in the natural setting.


Subject(s)
Evolution, Molecular , Gorilla gorilla/genetics , Pan troglodytes/genetics , Toll-Like Receptors/genetics , Animals , Base Sequence , Genetic Variation , Genome , Gorilla gorilla/classification , Gorilla gorilla/immunology , Humans , Immunity, Innate/genetics , Molecular Sequence Data , Pan troglodytes/classification , Pan troglodytes/immunology , Phylogeny , Polymorphism, Single Nucleotide , Selection, Genetic , Sequence Analysis, DNA , Species Specificity
7.
Am J Hum Genet ; 91(1): 27-37, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22748209

ABSTRACT

Host-pathogen interactions are generally initiated by host recognition of microbial components or danger signals triggered by microbial invasion. This recognition involves germline-encoded microbial sensors or pattern-recognition receptors (PRRs). By studying the way in which natural selection has driven the evolution of these microbial sensors in humans, we can identify genes playing an essential role and distinguish them from other, more redundant genes. We characterized the sequence diversity of the NOD-like receptor family, including the NALP and NOD/IPAF subfamilies, in various populations worldwide and compared this diversity with that of other PRR families, such as Toll-like receptors (TLRs) and RIG-I-like receptors (RLRs). We found that most NALPs had evolved under strong selective constraints, suggesting that their functions are essential and possibly much broader than previously thought. Conversely, most NOD/IPAF subfamily members were subject to more relaxed selective constraints, suggesting greater redundancy. Furthermore, some NALP genes, including NLRP1, NLRP14, and CIITA, were found to have evolved adaptively. We identified those variants conferring a selective advantage on some human populations as the most likely targets of positive selection. More generally, the strength of selection differed considerably between the major families of microbial sensors. Endosomal TLRs and most NALPs were found to evolve under stronger purifying selection than most NOD/IPAF subfamily members and cell-surface TLRs and RLRs, suggesting some degree of redundancy in the signaling pathways triggered by these molecules. This study provides novel perspectives and experimentally testable hypotheses concerning the relative biological relevance of the various families of microbial sensors in humans.


Subject(s)
Evolution, Molecular , Immunity, Innate/genetics , Receptors, Pattern Recognition/genetics , Adaptor Proteins, Signal Transducing/genetics , CARD Signaling Adaptor Proteins/genetics , Calcium-Binding Proteins/genetics , Cytosol/metabolism , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Genetic Variation , Humans , Polymorphism, Single Nucleotide , Receptors, Immunologic , Selection, Genetic , Toll-Like Receptors/genetics
8.
Am J Hum Genet ; 86(4): 611-20, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20346436

ABSTRACT

The study of recently admixed populations provides unique tools for understanding recent population dynamics, socio-cultural factors associated with the founding of emerging populations, and the genetic basis of disease by means of admixture mapping. Historical records and recent autosomal data indicate that the South African Coloured population forms a unique highly admixed population, resulting from the encounter of different peoples from Africa, Europe, and Asia. However, little is known about the mode by which this admixed population was recently founded. Here we show, through detailed phylogeographic analyses of mitochondrial DNA and Y-chromosome variation in a large sample of South African Coloured individuals, that this population derives from at least five different parental populations (Khoisan, Bantus, Europeans, Indians, and Southeast Asians), who have differently contributed to the foundation of the South African Coloured. In addition, our analyses reveal extraordinarily unbalanced gender-specific contributions of the various population genetic components, the most striking being the massive maternal contribution of Khoisan peoples (more than 60%) and the almost negligible maternal contribution of Europeans with respect to their paternal counterparts. The overall picture of gender-biased admixture depicted in this study indicates that the modern South African Coloured population results mainly from the early encounter of European and African males with autochthonous Khoisan females of the Cape of Good Hope around 350 years ago.


Subject(s)
Black People/genetics , Chromosomes, Human, Y/genetics , DNA, Mitochondrial/genetics , Genetics, Population , Polymorphism, Single Nucleotide/genetics , Female , Genetic Linkage , Humans , Male , Mothers , Sex Factors
9.
Nat Genet ; 34(1): 27-9, 2003 May.
Article in English | MEDLINE | ID: mdl-12669065

ABSTRACT

Many studies have supported a genetic etiology for autism. Here we report mutations in two X-linked genes encoding neuroligins NLGN3 and NLGN4 in siblings with autism-spectrum disorders. These mutations affect cell-adhesion molecules localized at the synapse and suggest that a defect of synaptogenesis may predispose to autism.


Subject(s)
Autistic Disorder/genetics , Carrier Proteins/genetics , Chromosomes, Human, X/genetics , Membrane Proteins/genetics , Mutation , Nerve Tissue Proteins/genetics , Amino Acid Sequence , Autistic Disorder/metabolism , Base Sequence , Brain/metabolism , Cell Adhesion Molecules, Neuronal , DNA, Complementary/genetics , Female , Gene Expression Profiling , Genetic Linkage , Humans , Male , Molecular Sequence Data , Pedigree , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid
10.
Am J Hum Genet ; 84(3): 316-27, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19232555

ABSTRACT

MicroRNAs (miRNAs) are noncoding RNAs involved in posttranscriptional gene repression, and their role in diverse physiological processes is increasingly recognized. Yet, few efforts have been devoted to evolutionary studies of human miRNAs. Knowledge about the way in which natural selection has targeted miRNAs should provide insight into their functional relevance as well as their mechanisms of action. Here we used miRNAs as a model system for investigating the influence of natural selection on gene regulation by characterizing the full spectrum of naturally occurring sequence variation of 117 human miRNAs from different populations worldwide. We found that purifying selection has globally constrained the diversity of miRNA-containing regions and has strongly targeted the mature miRNA. This observation emphasizes that mutations in these molecules are likely to be deleterious, and therefore they can have severe phenotypic consequences on human health. More importantly, we obtained evidence of population-specific events of positive selection acting on a number of miRNA-containing regions. Notably, our analysis revealed that positive selection has targeted a "small-RNA-rich island" on chromosome 14, harboring both miRNAs and small nucleolar RNAs, in Europeans and East Asians. These observations support the notion that the tuning of gene expression contributes to the processes by which populations adapt to specific environments. These findings will fuel future investigations exploring how genetic and functional variation of miRNAs under selection affects the repression of their mRNA targets, increasing our understanding of the role of gene regulation in population adaptation and human disease.


Subject(s)
Evolution, Molecular , Genetic Variation , MicroRNAs/genetics , Selection, Genetic , Base Sequence , Chromosomes, Human, Pair 14/genetics , Genetics, Population , Humans , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , RNA, Untranslated/genetics
11.
PLoS Genet ; 5(7): e1000562, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19609346

ABSTRACT

Infectious diseases have been paramount among the threats to health and survival throughout human evolutionary history. Natural selection is therefore expected to act strongly on host defense genes, particularly on innate immunity genes whose products mediate the direct interaction between the host and the microbial environment. In insects and mammals, the Toll-like receptors (TLRs) appear to play a major role in initiating innate immune responses against microbes. In humans, however, it has been speculated that the set of TLRs could be redundant for protective immunity. We investigated how natural selection has acted upon human TLRs, as an approach to assess their level of biological redundancy. We sequenced the ten human TLRs in a panel of 158 individuals from various populations worldwide and found that the intracellular TLRs -- activated by nucleic acids and particularly specialized in viral recognition -- have evolved under strong purifying selection, indicating their essential non-redundant role in host survival. Conversely, the selective constraints on the TLRs expressed on the cell surface -- activated by compounds other than nucleic acids -- have been much more relaxed, with higher rates of damaging nonsynonymous and stop mutations tolerated, suggesting their higher redundancy. Finally, we tested whether TLRs have experienced spatially-varying selection in human populations and found that the region encompassing TLR10-TLR1-TLR6 has been the target of recent positive selection among non-Africans. Our findings indicate that the different TLRs differ in their immunological redundancy, reflecting their distinct contributions to host defense. The insights gained in this study foster new hypotheses to be tested in clinical and epidemiological genetics of infectious disease.


Subject(s)
Evolution, Molecular , Genetics, Population , Immunity/genetics , Toll-Like Receptors/genetics , Ethnicity/genetics , Humans , Infections/immunology , Kinetics , Mutation , Selection, Genetic , Sequence Analysis, DNA , Toll-Like Receptors/immunology
12.
Hum Mutat ; 32(6): 643-52, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21618349

ABSTRACT

Toll-like receptors (TLRs) are considered an essential component of the innate immune system, initiating inflammatory responses following infection of the host. Humans have 10 functional TLRs, differing in their subcellular distributions and the microbial agonists they sense. The phylogenetically conserved TLR1-2-6 family is unique in that TLR1 and TLR6 form heterodimers with TLR2 to mediate signalling in response to agonists. Epidemiological genetic studies have identified several TLR variants that appear to influence susceptibility to infectious diseases, but the functional consequences of which remain largely unknown. Here, we assessed the functional impact of the TLR1-2-6 variants with altered amino acid sequences segregating naturally in the human population. We used an NF-κB reporter assay in TLR-transfected human embryonic kidney 293T cells stimulated with the corresponding TLR agonists. We found that among the 41 naturally occurring variants with amino acid alterations identified in the TLR1-2-6 family, 14 of them (five TLR1, four TLR2, and five TLR6 variants) displayed marked impairment of NF-κB activation. Most of these variants are present at very low population frequencies and are population-specific. These observations suggest that rare, nonsynonymous TLR mutations are likely to have deleterious effects on immune responses and may therefore contribute to complex susceptibility to infection at the population level.


Subject(s)
Genetic Variation , Toll-Like Receptor 1/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 6/genetics , Evolution, Molecular , HEK293 Cells , Humans , Mutation/genetics , Population/genetics , Toll-Like Receptor 1/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 6/immunology
13.
Proc Natl Acad Sci U S A ; 105(5): 1596-601, 2008 Feb 05.
Article in English | MEDLINE | ID: mdl-18216239

ABSTRACT

Two groups of populations with completely different lifestyles-the Pygmy hunter-gatherers and the Bantu-speaking farmers-coexist in Central Africa. We investigated the origins of these two groups and the interactions between them, by analyzing mtDNA variation in 1,404 individuals from 20 farming populations and 9 Pygmy populations from Central Africa, with the aim of shedding light on one of the most fascinating cultural transitions in human evolution (the transition from hunting and gathering to agriculture). Our data indicate that this region was colonized gradually, with an initial L1c-rich ancestral population ultimately giving rise to current-day farmers, who display various L1c clades, and to Pygmies, in whom L1c1a is the only surviving clade. Detailed phylogenetic analysis of complete mtDNA sequences for L1c1a showed this clade to be autochthonous to Central Africa, with its most recent branches shared between farmers and Pygmies. Coalescence analyses revealed that these two groups arose through a complex evolutionary process characterized by (i) initial divergence of the ancestors of contemporary Pygmies from an ancestral Central African population no more than approximately 70,000 years ago, (ii) a period of isolation between the two groups, accounting for their phenotypic differences, (iii) long-standing asymmetric maternal gene flow from Pygmies to the ancestors of the farming populations, beginning no more than approximately 40,000 years ago and persisting until a few thousand years ago, and (iv) enrichment of the maternal gene pool of the ancestors of the farming populations by the arrival and/or subsequent demographic expansion of L0a, L2, and L3 carriers.


Subject(s)
Black People/genetics , Gene Flow , Genes, Mitochondrial/genetics , Genetic Variation , Population/genetics , Africa, Central , Base Sequence , DNA, Mitochondrial/classification , DNA, Mitochondrial/genetics , Female , Haploidy , Humans , Male , Molecular Sequence Data , Phylogeny , Sequence Analysis, DNA
14.
Front Immunol ; 12: 768189, 2021.
Article in English | MEDLINE | ID: mdl-34912340

ABSTRACT

There is considerable inter-individual and inter-population variability in response to viruses. The potential of monocytes to elicit type-I interferon responses has attracted attention to their role in viral infections. Here, we use single-cell RNA-sequencing to characterize the role of cellular heterogeneity in human variation of monocyte responses to influenza A virus (IAV) exposure. We show widespread inter-individual variability in the percentage of IAV-infected monocytes. Notably, individuals with high cellular susceptibility to IAV are characterized by a lower activation at basal state of an IRF/STAT-induced transcriptional network, which includes antiviral genes such as IFITM3, MX1 and OAS3. Upon IAV challenge, we find that cells escaping viral infection display increased mRNA expression of type-I interferon stimulated genes and decreased expression of ribosomal genes, relative to both infected cells and those never exposed to IAV. We also uncover a stronger resistance of CD16+ monocytes to IAV infection, together with CD16+ -specific mRNA expression of IL6 and TNF in response to IAV. Finally, using flow cytometry and bulk RNA-sequencing across 200 individuals of African and European ancestry, we observe a higher number of CD16+ monocytes and lower susceptibility to IAV infection among monocytes from individuals of African-descent. Based on these data, we hypothesize that higher basal monocyte activation, driven by environmental factors and/or weak-effect genetic variants, underlies the lower cellular susceptibility to IAV infection of individuals of African ancestry relative to those of European ancestry. Further studies are now required to investigate how such cellular differences in IAV susceptibility translate into population differences in clinical outcomes and susceptibility to severe influenza.


Subject(s)
Influenza A virus , Influenza, Human/ethnology , Monocytes/immunology , Sequence Analysis, RNA , Single-Cell Analysis , Adult , Black People , Cytokines/physiology , GPI-Linked Proteins/analysis , Humans , Middle Aged , Monocytes/virology , Receptors, IgG/analysis , Receptors, IgG/genetics , Ribosomes/physiology , White People , Young Adult
15.
Genome Biol ; 21(1): 187, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32731901

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are key regulators of the immune system, yet their variation and contribution to intra- and inter-population differences in immune responses is poorly characterized. RESULTS: We generate 977 miRNA-sequencing profiles from primary monocytes from individuals of African and European ancestry following activation of three TLR pathways (TLR4, TLR1/2, and TLR7/8) or infection with influenza A virus. We find that immune activation leads to important modifications in the miRNA and isomiR repertoire, particularly in response to viral challenges. These changes are much weaker than those observed for protein-coding genes, suggesting stronger selective constraints on the miRNA response to stimulation. This is supported by the limited genetic control of miRNA expression variability (miR-QTLs) and the lower occurrence of gene-environment interactions, in stark contrast with eQTLs that are largely context-dependent. We also detect marked differences in miRNA expression between populations, which are mostly driven by non-genetic factors. On average, miR-QTLs explain approximately 60% of population differences in expression of their cognate miRNAs and, in some cases, evolve adaptively, as shown in Europeans for a miRNA-rich cluster on chromosome 14. Finally, integrating miRNA and mRNA data from the same individuals, we provide evidence that the canonical model of miRNA-driven transcript degradation has a minor impact on miRNA-mRNA correlations, which are, in our setting, mainly driven by co-transcription. CONCLUSION: Together, our results shed new light onto the factors driving miRNA and isomiR diversity at the population level and constitute a useful resource for evaluating their role in host differences of immunity to infection.


Subject(s)
Immunity , Infections/immunology , MicroRNAs/metabolism , Monocytes/metabolism , RNA Isoforms/metabolism , Black People , Genome, Human , Humans , Infections/metabolism , MicroRNAs/immunology , Quantitative Trait Loci , RNA Isoforms/immunology , White People
16.
Nat Commun ; 10(1): 1671, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30975994

ABSTRACT

Host and environmental factors contribute to variation in human immune responses, yet the genetic and evolutionary drivers of alternative splicing in response to infection remain largely uncharacterised. Leveraging 970 RNA-sequencing profiles of resting and stimulated monocytes from 200 individuals of African- and European-descent, we show that immune activation elicits a marked remodelling of the isoform repertoire, while increasing the levels of erroneous splicing. We identify 1,464 loci associated with variation in isoform usage (sQTLs), 9% of them being stimulation-specific, which are enriched in disease-related loci. Furthermore, we detect a longstanding increased plasticity of immune gene splicing, and show that positive selection and Neanderthal introgression have both contributed to diversify the splicing landscape of human populations. Together, these findings suggest that differential isoform usage has been an important substrate of innovation in the long-term evolution of immune responses and a more recent vehicle of population local adaptation.


Subject(s)
Alternative Splicing/immunology , Immunity/genetics , Infections/immunology , Selection, Genetic/immunology , Transcriptome/immunology , Animals , Biological Evolution , Black People/genetics , Genetic Variation/immunology , Healthy Volunteers , Humans , Male , Neanderthals/genetics , Polymorphism, Single Nucleotide , Protein Isoforms/genetics , Protein Isoforms/immunology , Quantitative Trait Loci/immunology , Sequence Analysis, RNA , White People/genetics , Exome Sequencing
17.
Genome Biol ; 19(1): 222, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30563547

ABSTRACT

BACKGROUND: DNA methylation is influenced by both environmental and genetic factors and is increasingly thought to affect variation in complex traits and diseases. Yet, the extent of ancestry-related differences in DNA methylation, their genetic determinants, and their respective causal impact on immune gene regulation remain elusive. RESULTS: We report extensive population differences in DNA methylation between 156 individuals of African and European descent, detected in primary monocytes that are used as a model of a major innate immunity cell type. Most of these differences (~ 70%) are driven by DNA sequence variants nearby CpG sites, which account for ~ 60% of the variance in DNA methylation. We also identify several master regulators of DNA methylation variation in trans, including a regulatory hub nearby the transcription factor-encoding CTCF gene, which contributes markedly to ancestry-related differences in DNA methylation. Furthermore, we establish that variation in DNA methylation is associated with varying gene expression levels following mostly, but not exclusively, a canonical model of negative associations, particularly in enhancer regions. Specifically, we find that DNA methylation highly correlates with transcriptional activity of 811 and 230 genes, at the basal state and upon immune stimulation, respectively. Finally, using a Bayesian approach, we estimate causal mediation effects of DNA methylation on gene expression in ~ 20% of the studied cases, indicating that DNA methylation can play an active role in immune gene regulation. CONCLUSION: Using a system-level approach, our study reveals substantial ancestry-related differences in DNA methylation and provides evidence for their causal impact on immune gene regulation.


Subject(s)
Black People/genetics , DNA Methylation , Gene Expression Regulation , Immunity, Innate , White People/genetics , Adult , Epigenesis, Genetic , Humans , Male , Monocytes , Quantitative Trait Loci
18.
Nat Ecol Evol ; 2(4): 721-730, 2018 04.
Article in English | MEDLINE | ID: mdl-29531345

ABSTRACT

Understanding how deleterious genetic variation is distributed across human populations is of key importance in evolutionary biology and medical genetics. However, the impact of population size changes and gene flow on the corresponding mutational load remains a controversial topic. Here, we report high-coverage exomes from 300 rainforest hunter-gatherers and farmers of central Africa, whose distinct subsistence strategies are expected to have impacted their demographic pasts. Detailed demographic inference indicates that hunter-gatherers and farmers recently experienced population collapses and expansions, respectively, accompanied by increased gene flow. We show that the distribution of deleterious alleles across these populations is compatible with a similar efficacy of selection to remove deleterious variants with additive effects, and predict with simulations that their present-day additive mutation load is almost identical. For recessive mutations, although an increased load is predicted for hunter-gatherers, this increase has probably been partially counteracted by strong gene flow from expanding farmers. Collectively, our predicted and empirical observations suggest that the impact of the recent population decline of African hunter-gatherers on their mutation load has been modest and more restrained than would be expected under a fully recessive model of dominance.


Subject(s)
Exome/genetics , Gene Flow , Mutation , Africa , Farmers , Humans , Life Style , Population Dynamics , Rainforest
19.
J Exp Med ; 214(4): 877-894, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28351985

ABSTRACT

More than a decade after the sequencing of the human genome, a deluge of genome-wide population data are generating a portrait of human genetic diversity at an unprecedented level of resolution. Genomic studies have provided new insight into the demographic and adaptive history of our species, Homo sapiens, including its interbreeding with other hominins, such as Neanderthals, and the ways in which natural selection, in its various guises, has shaped genome diversity. These studies, combined with functional genomic approaches, such as the mapping of expression quantitative trait loci, have helped to identify genes, functions, and mechanisms of prime importance for host survival and involved in phenotypic variation and differences in disease risk. This review summarizes new findings in this rapidly developing field, focusing on the human immune response. We discuss the importance of defining the genetic and evolutionary determinants driving immune response variation, and highlight the added value of population genomic approaches in settings relevant to immunity and infection.


Subject(s)
Adaptation, Biological , Genome, Human , Immunity , Genetic Variation , Humans , Mutation , Quantitative Trait Loci , Selection, Genetic
20.
Science ; 356(6337): 543-546, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28473590

ABSTRACT

Bantu languages are spoken by about 310 million Africans, yet the genetic history of Bantu-speaking populations remains largely unexplored. We generated genomic data for 1318 individuals from 35 populations in western central Africa, where Bantu languages originated. We found that early Bantu speakers first moved southward, through the equatorial rainforest, before spreading toward eastern and southern Africa. We also found that genetic adaptation of Bantu speakers was facilitated by admixture with local populations, particularly for the HLA and LCT loci. Finally, we identified a major contribution of western central African Bantu speakers to the ancestry of African Americans, whose genomes present no strong signals of natural selection. Together, these results highlight the contribution of Bantu-speaking peoples to the complex genetic history of Africans and African Americans.


Subject(s)
Adaptation, Physiological/genetics , Black or African American/genetics , Genetic Loci , HLA Antigens/genetics , Lactase/genetics , Language , Africa, Central , Human Migration , Humans , North America , Polymorphism, Single Nucleotide , Rainforest , Speech
SELECTION OF CITATIONS
SEARCH DETAIL