Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Annu Rev Genet ; 51: 413-433, 2017 11 27.
Article in English | MEDLINE | ID: mdl-28934590

ABSTRACT

The body's microbiome, composed of microbial cells that number in the trillions, is involved in human health and disease in ways that are just starting to emerge. The microbiome is assembled at birth, develops with its host, and is greatly influenced by environmental factors such as diet and other exposures. Recently, a role for human genetic variation has emerged as also influential in accounting for interpersonal differences in microbiomes. Thus, human genes may influence health directly or by promoting a beneficial microbiome. Studies of the heritability of gut microbiotas reveal a subset of microbes whose abundances are partly genetically determined by the host. However, the use of genome-wide association studies (GWASs) to identify human genetic variants associated with microbiome phenotypes has proven challenging. Studies to date are small by GWAS standards, and cross-study comparisons are hampered by differences in analytical approaches. Nevertheless, associations between microbes or microbial genes and human genes have emerged that are consistent between human populations. Most notably, higher levels of beneficial gut bacteria called Bifidobacteria are associated with the human lactase nonpersister genotype, which typically confers lactose intolerance, in several different human populations. It is time for the microbiome to be incorporated into studies that quantify interactions among genotype, environment, and the microbiome in order to predict human disease susceptibility.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Gastrointestinal Microbiome/physiology , Genome, Human , Lactose Intolerance/genetics , Obesity/genetics , Schizophrenia/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/microbiology , Amyotrophic Lateral Sclerosis/pathology , Bifidobacterium/growth & development , Bifidobacterium/metabolism , Diet/methods , Gastrointestinal Tract/microbiology , Genetic Variation , Genome-Wide Association Study , Genotype , Human Genetics , Humans , Lactose Intolerance/metabolism , Lactose Intolerance/microbiology , Lactose Intolerance/pathology , Obesity/metabolism , Obesity/microbiology , Obesity/pathology , Phenotype , Quantitative Trait, Heritable , Schizophrenia/metabolism , Schizophrenia/microbiology , Schizophrenia/pathology
2.
Am J Hum Genet ; 108(1): 49-67, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33326753

ABSTRACT

Although thousands of loci have been associated with human phenotypes, the role of gene-environment (GxE) interactions in determining individual risk of human diseases remains unclear. This is partly because of the severe erosion of statistical power resulting from the massive number of statistical tests required to detect such interactions. Here, we focus on improving the power of GxE tests by developing a statistical framework for assessing quantitative trait loci (QTLs) associated with the trait means and/or trait variances. When applying this framework to body mass index (BMI), we find that GxE discovery and replication rates are significantly higher when prioritizing genetic variants associated with the variance of the phenotype (vQTLs) compared to when assessing all genetic variants. Moreover, we find that vQTLs are enriched for associations with other non-BMI phenotypes having strong environmental influences, such as diabetes or ulcerative colitis. We show that GxE effects first identified in quantitative traits such as BMI can be used for GxE discovery in disease phenotypes such as diabetes. A clear conclusion is that strong GxE interactions mediate the genetic contribution to body weight and diabetes risk.


Subject(s)
Biological Variation, Population/genetics , Genome-Wide Association Study/methods , Gene-Environment Interaction , Genotype , Humans , Phenotype , Quantitative Trait Loci/genetics , Quantitative Trait, Heritable
3.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Article in English | MEDLINE | ID: mdl-34161260

ABSTRACT

Individuals who are minoritized as a result of race, sexual identity, gender, or socioeconomic status experience a higher prevalence of many diseases. Understanding the biological processes that cause and maintain these socially driven health inequities is essential for addressing them. The gut microbiome is strongly shaped by host environments and affects host metabolic, immune, and neuroendocrine functions, making it an important pathway by which differences in experiences caused by social, political, and economic forces could contribute to health inequities. Nevertheless, few studies have directly integrated the gut microbiome into investigations of health inequities. Here, we argue that accounting for host-gut microbe interactions will improve understanding and management of health inequities, and that health policy must begin to consider the microbiome as an important pathway linking environments to population health.


Subject(s)
Gastrointestinal Microbiome , Health Status Disparities , Disease , Health , Humans , Mental Health , Publications
4.
Trends Immunol ; 41(1): 1-3, 2020 01.
Article in English | MEDLINE | ID: mdl-31806285

ABSTRACT

Host genetics influence gut microbiome composition. However, determining the specific physiological mechanisms and microbes affected has been difficult due to 'cage' and 'legacy' effects in model systems. Recently, Khan et al. cleverly colonized ex-germ-free mice to demonstrate that immune genes regulate select bacterial lineages in the mouse gut.


Subject(s)
Gastrointestinal Microbiome , Animals , Bacteria/genetics , Genetic Variation , Humans , Immunity/genetics , Mice , Symbiosis
5.
Hum Genet ; 140(5): 747-760, 2021 May.
Article in English | MEDLINE | ID: mdl-33221945

ABSTRACT

Despite the growing knowledge surrounding host-microbiome interactions, we are just beginning to understand how the gut microbiome influences-and is influenced by-host gene expression. Here, we review recent literature that intersects these two fields, summarizing themes across studies. Work in model organisms, human biopsies, and cell culture demonstrate that the gut microbiome is an important regulator of several host pathways relevant for disease, including immune development and energy metabolism, and vice versa. The gut microbiome remodels host chromatin, causes differential splicing, alters the epigenetic landscape, and directly interrupts host signaling cascades. Emerging techniques like single-cell RNA sequencing and organoid generation have the potential to refine our understanding of the relationship between the gut microbiome and host gene expression in the future. By intersecting microbiome and host gene expression, we gain a window into the physiological processes important for fostering the extensive cross-kingdom interactions and ultimately our health.


Subject(s)
Gastrointestinal Microbiome/physiology , Gene Expression Regulation/genetics , Gene Expression/genetics , Energy Metabolism/genetics , Humans , RNA, Ribosomal, 16S/genetics
6.
PLoS Biol ; 16(11): e2005396, 2018 11.
Article in English | MEDLINE | ID: mdl-30439937

ABSTRACT

The composition of the gut microbiome in industrialized populations differs from those living traditional lifestyles. However, it has been difficult to separate the contributions of human genetic and geographic factors from lifestyle. Whether shifts away from the foraging lifestyle that characterize much of humanity's past influence the gut microbiome, and to what degree, remains unclear. Here, we characterize the stool bacterial composition of four Himalayan populations to investigate how the gut community changes in response to shifts in traditional human lifestyles. These groups led seminomadic hunting-gathering lifestyles until transitioning to varying levels of agricultural dependence upon farming. The Tharu began farming 250-300 years ago, the Raute and Raji transitioned 30-40 years ago, and the Chepang retain many aspects of a foraging lifestyle. We assess the contributions of dietary and environmental factors on their gut-associated microbes and find that differences in the lifestyles of Himalayan foragers and farmers are strongly correlated with microbial community variation. Furthermore, the gut microbiomes of all four traditional Himalayan populations are distinct from that of the Americans, indicating that industrialization may further exacerbate differences in the gut community. The Chepang foragers harbor an elevated abundance of taxa associated with foragers around the world. Conversely, the gut microbiomes of the populations that have transitioned to farming are more similar to those of Americans, with agricultural dependence and several associated lifestyle and environmental factors correlating with the extent of microbiome divergence from the foraging population. The gut microbiomes of Raute and Raji reveal an intermediate state between the Chepang and Tharu, indicating that divergence from a stereotypical foraging microbiome can occur within a single generation. Our results also show that environmental factors such as drinking water source and solid cooking fuel are significantly associated with the gut microbiome. Despite the pronounced differences in gut bacterial composition across populations, we found little differences in alpha diversity across lifestyles. These findings in genetically similar populations living in the same geographical region establish the key role of lifestyle in determining human gut microbiome composition and point to the next challenging steps of determining how large-scale gut microbiome reconfiguration impacts human biology.


Subject(s)
Gastrointestinal Microbiome/genetics , Life Style/ethnology , Microbiota/genetics , Adult , Bacteria/genetics , Diet , Diet, Paleolithic , Feces/microbiology , Female , Gastrointestinal Microbiome/physiology , Genetics, Population/methods , Geography , Humans , Male , Middle Aged , Nepal/ethnology , RNA, Ribosomal, 16S/genetics , Rural Population
7.
BMC Biol ; 15(1): 127, 2017 12 27.
Article in English | MEDLINE | ID: mdl-29282061

ABSTRACT

The trillions of microbes living in the gut-the gut microbiota-play an important role in human biology and disease. While much has been done to explore its diversity, a full understanding of our microbiomes demands an evolutionary perspective. In this review, we compare microbiomes from human populations, placing them in the context of microbes from humanity's near and distant animal relatives. We discuss potential mechanisms to generate host-specific microbiome configurations and the consequences of disrupting those configurations. Finally, we propose that this broader phylogenetic perspective is useful for understanding the mechanisms underlying human-microbiome interactions.


Subject(s)
Biological Evolution , Microbiota/physiology , Animals , Gastrointestinal Microbiome/physiology , Host Specificity , Humans , Phylogeny
8.
BMC Genomics ; 17(1): 941, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27871240

ABSTRACT

BACKGROUND: Host genetics is one of several factors known to shape human gut microbiome composition, however, the physiological processes underlying the heritability are largely unknown. Inter-individual differences in host factors secreted into the gut lumen may lead to variation in microbiome composition. One such factor is the ABO antigen. This molecule is not only expressed on the surface of red blood cells, but is also secreted from mucosal surfaces in individuals containing an intact FUT2 gene (secretors). Previous studies report differences in microbiome composition across ABO and secretor genotypes. However, due to methodological limitations, the specific bacterial taxa involved remain unknown. RESULTS: Here, we sought to determine the relationship of the microbiota to ABO blood group and secretor status in a large panel of 1503 individuals from a cohort of twins from the United Kingdom. Contrary to previous reports, robust associations between either ABO or secretor phenotypes and gut microbiome composition were not detected. Overall community structure, diversity, and the relative abundances of individual taxa were not significantly associated with ABO or secretor status. Additionally, joint-modeling approaches were unsuccessful in identifying combinations of taxa that were predictive of ABO or secretor status. CONCLUSIONS: Despite previous reports, the taxonomic composition of the microbiota does not appear to be strongly associated with ABO or secretor status in 1503 individuals from the United Kingdom. These results highlight the importance of replicating microbiome-associated traits in large, well-powered cohorts to ensure results are robust.


Subject(s)
ABO Blood-Group System/immunology , Biodiversity , Gastrointestinal Microbiome , Twins , ABO Blood-Group System/genetics , Adult , Aged , Female , Genotype , Humans , Male , Middle Aged , Phenotype , United Kingdom
9.
Nat Microbiol ; 8(12): 2315-2325, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38030898

ABSTRACT

The prevalence of chronic, non-communicable diseases has risen sharply in recent decades, especially in industrialized countries. While several studies implicate the microbiome in this trend, few have examined the evolutionary history of industrialized microbiomes. Here we sampled 235 ancient dental calculus samples from individuals living in Great Britain (∼2200 BCE to 1853 CE), including 127 well-contextualized London adults. We reconstructed their microbial history spanning the transition to industrialization. After controlling for oral geography and technical biases, we identified multiple oral microbial communities that coexisted in Britain for millennia, including a community associated with Methanobrevibacter, an anaerobic Archaea not commonly prevalent in the oral microbiome of modern industrialized societies. Calculus analysis suggests that oral hygiene contributed to oral microbiome composition, while microbial functions reflected past differences in diet, specifically in dairy and carbohydrate consumption. In London samples, Methanobrevibacter-associated microbial communities are linked with skeletal markers of systemic diseases (for example, periostitis and joint pathologies), and their disappearance is consistent with temporal shifts, including the arrival of the Second Plague Pandemic. This suggests pre-industrialized microbiomes were more diverse than previously recognized, enhancing our understanding of chronic, non-communicable disease origins in industrialized populations.


Subject(s)
Dental Calculus , Microbiota , Adult , Humans , United Kingdom/epidemiology , Dental Calculus/epidemiology , Diet , Life Style
11.
Annu Rev Anim Biosci ; 10: 203-226, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35167316

ABSTRACT

Animals harbor diverse communities of microbes within their gastrointestinal tracts. Phylogenetic relationship, diet, gut morphology, host physiology, and ecology all influence microbiome composition within and between animal clades. Emerging evidence points to host genetics as also playing a role in determining gut microbial composition within species. Here, we discuss recent advances in the study of microbiome heritability across a variety of animal species. Candidate gene and discovery-based studies in humans, mice, Drosophila, Caenorhabditis elegans, cattle, swine, poultry, and baboons reveal trends in the types of microbes that are heritable and the host genes and pathways involved in shaping the microbiome. Heritable gut microbes within a host species tend to be phylogenetically restricted. Host genetic variation in immune- and growth-related genes drives the abundances of these heritable bacteria within the gut. With only a small slice of the metazoan branch of the tree of life explored to date, this is an area rife with opportunities to shed light into the mechanisms governing host-microbe relationships.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Animals , Bacteria , Caenorhabditis elegans/genetics , Caenorhabditis elegans/microbiology , Cattle , Gastrointestinal Microbiome/genetics , Mice , Phylogeny , Swine
12.
Elife ; 102021 03 23.
Article in English | MEDLINE | ID: mdl-33755018

ABSTRACT

Changes in diet associated with domestication may have shaped the composition of microbes found in the guts of animals.


Subject(s)
Gastrointestinal Microbiome , Animals , Diet , Domestication , Humans , Industrial Development
13.
Nat Genet ; 51(3): 548-559, 2019 03.
Article in English | MEDLINE | ID: mdl-30778225

ABSTRACT

Streptococcus pyogenes causes 700 million human infections annually worldwide, yet, despite a century of intensive effort, there is no licensed vaccine against this bacterium. Although a number of large-scale genomic studies of bacterial pathogens have been published, the relationships among the genome, transcriptome, and virulence in large bacterial populations remain poorly understood. We sequenced the genomes of 2,101 emm28 S. pyogenes invasive strains, from which we selected 492 phylogenetically diverse strains for transcriptome analysis and 50 strains for virulence assessment. Data integration provided a novel understanding of the virulence mechanisms of this model organism. Genome-wide association study, expression quantitative trait loci analysis, machine learning, and isogenic mutant strains identified and confirmed a one-nucleotide indel in an intergenic region that significantly alters global transcript profiles and ultimately virulence. The integrative strategy that we used is generally applicable to any microbe and may lead to new therapeutics for many human pathogens.


Subject(s)
Genome, Bacterial/genetics , Streptococcus pyogenes/genetics , Transcriptome/genetics , Virulence/genetics , Gene Expression Regulation, Bacterial/genetics , Genome-Wide Association Study/methods , Genomics/methods , Phylogeny , Quantitative Trait Loci/genetics
15.
Cell Host Microbe ; 22(3): 251-253, 2017 09 13.
Article in English | MEDLINE | ID: mdl-28910628

ABSTRACT

The mix of bacteria that coat our teeth impact oral health, but it remains unclear what factors govern their composition. In this issue of Cell Host & Microbe, Gomez et al. (2017) examine the relationship between host genetics and the oral microbiome in the context of health and disease.


Subject(s)
Bacteria , Microbiota , Humans
16.
Microbiome ; 5(1): 16, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28143570

ABSTRACT

BACKGROUND: The degree to which host genetic variation can modulate microbial communities in humans remains an open question. Here, we performed a genetic mapping study of the microbiome in two accessible upper airway sites, the nasopharynx and the nasal vestibule, during two seasons in 144 adult members of a founder population of European decent. RESULTS: We estimated the relative abundances (RAs) of genus level bacteria from 16S rRNA gene sequences and examined associations with 148,653 genetic variants (linkage disequilibrium [LD] r 2 < 0.5) selected from among all common variants discovered in genome sequences in this population. We identified 37 microbiome quantitative trait loci (mbQTLs) that showed evidence of association with the RAs of 22 genera (q < 0.05) and were enriched for genes in mucosal immunity pathways. The most significant association was between the RA of Dermacoccus (phylum Actinobacteria) and a variant 8 kb upstream of TINCR (rs117042385; p = 1.61 × 10-8; q = 0.002), a long non-coding RNA that binds to peptidoglycan recognition protein 3 (PGLYRP3) mRNA, a gene encoding a known antimicrobial protein. A second association was between a missense variant in PGLYRP4 (rs3006458) and the RA of an unclassified genus of family Micrococcaceae (phylum Actinobacteria) (p = 5.10 × 10-7; q = 0.032). CONCLUSIONS: Our findings provide evidence of host genetic influences on upper airway microbial composition in humans and implicate mucosal immunity genes in this relationship.


Subject(s)
Actinobacteria/classification , Actinobacteria/genetics , Immunity, Mucosal/genetics , Microbiota/genetics , Nasopharynx/microbiology , Nose/microbiology , Actinobacteria/isolation & purification , Adolescent , Adult , Aged , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chromosome Mapping , Female , Genetic Variation/genetics , Humans , Immunity, Mucosal/immunology , Male , Microbiota/immunology , Middle Aged , Quantitative Trait Loci/genetics , RNA, Ribosomal, 16S/genetics , Young Adult
17.
Gut Microbes ; 7(2): 178-84, 2016.
Article in English | MEDLINE | ID: mdl-26939746

ABSTRACT

A major goal of microbiome research is to identify the factors that determine bacterial composition within and upon a host. Environmental factors are thought to play a large role, such as diet in determining gut microbiome composition and moisture in determining skin microbiome composition. The role of host genetics, however, has been a source of debate in the literature. Recently, we examined the association of host genetics with human gut microbiome composition in the Hutterites, a population that lives and eats communally. We identified heritable bacterial taxa and host genetic loci associated with their abundances. In this addendum, I put these results into a broader context along with other recent studies of microbiome heritability, and synthesize common themes that appear across organisms and tissues, such as the relatively small extent genetics plays compared to environment and the role of host genetic variation in immune response and barrier integrity.


Subject(s)
Bacteria/genetics , Bacterial Infections/genetics , Gastrointestinal Microbiome , Animals , Bacteria/classification , Bacteria/isolation & purification , Bacteria/metabolism , Bacterial Infections/microbiology , Gastrointestinal Tract/microbiology , Host-Pathogen Interactions , Humans
18.
Science ; 352(6285): 532-5, 2016 Apr 29.
Article in English | MEDLINE | ID: mdl-27126034

ABSTRACT

Recent studies in human populations and mouse models reveal notable congruences in gut microbial taxa whose abundances are partly regulated by host genotype. Host genes associating with these taxa are related to diet sensing, metabolism, and immunity. These broad patterns are further validated in similar studies of nonmammalian microbiomes. The next generation of genome-wide association studies will expand the size of the data sets and refine the microbial phenotypes to fully capture these intriguing signatures of host-microbiome coevolution.


Subject(s)
Bacteria/classification , Genome-Wide Association Study , Microbiota/physiology , Quantitative Trait Loci , Animals , Bacteria/genetics , Diet , Genotype , Humans , Mice , Microbiota/genetics , Phenotype , Species Specificity
19.
Cell Host Microbe ; 19(5): 731-43, 2016 May 11.
Article in English | MEDLINE | ID: mdl-27173935

ABSTRACT

Studies in mice and humans have revealed intriguing associations between host genetics and the microbiome. Here we report a 16S rRNA-based analysis of the gut microbiome in 1,126 twin pairs, a subset of which was previously reported. Tripling the sample narrowed the confidence intervals around heritability estimates and uncovered additional heritable taxa, some of which are validated in other studies. Repeat sampling of subjects showed heritable taxa to be temporally stable. A candidate gene approach uncovered associations between heritable taxa and genes related to diet, metabolism, and olfaction. We replicate an association between Bifidobacterium and the lactase (LCT) gene locus and identify an association between the host gene ALDH1L1 and the bacteria SHA-98, suggesting a link between formate production and blood pressure. Additional genes detected are involved in barrier defense and self/non-self recognition. Our results indicate that diet-sensing, metabolism, and immune defense are important drivers of human-microbiome co-evolution.


Subject(s)
Gastrointestinal Microbiome/genetics , Microbial Consortia/genetics , Twins , Base Sequence , Female , Genetic Variation , Humans , Male , United Kingdom
20.
Genome Biol ; 17(1): 189, 2016 Sep 26.
Article in English | MEDLINE | ID: mdl-27666579

ABSTRACT

BACKGROUND: Variation in the human fecal microbiota has previously been associated with body mass index (BMI). Although obesity is a global health burden, the accumulation of abdominal visceral fat is the specific cardio-metabolic disease risk factor. Here, we explore links between the fecal microbiota and abdominal adiposity using body composition as measured by dual-energy X-ray absorptiometry in a large sample of twins from the TwinsUK cohort, comparing fecal 16S rRNA diversity profiles with six adiposity measures. RESULTS: We profile six adiposity measures in 3666 twins and estimate their heritability, finding novel evidence for strong genetic effects underlying visceral fat and android/gynoid ratio. We confirm the association of lower diversity of the fecal microbiome with obesity and adiposity measures, and then compare the association between fecal microbial composition and the adiposity phenotypes in a discovery subsample of twins. We identify associations between the relative abundances of fecal microbial operational taxonomic units (OTUs) and abdominal adiposity measures. Most of these results involve visceral fat associations, with the strongest associations between visceral fat and Oscillospira members. Using BMI as a surrogate phenotype, we pursue replication in independent samples from three population-based cohorts including American Gut, Flemish Gut Flora Project and the extended TwinsUK cohort. Meta-analyses across the replication samples indicate that 8 OTUs replicate at a stringent threshold across all cohorts, while 49 OTUs achieve nominal significance in at least one replication sample. Heritability analysis of the adiposity-associated microbial OTUs prompted us to assess host genetic-microbe interactions at obesity-associated human candidate loci. We observe significant associations of adiposity-OTU abundances with host genetic variants in the FHIT, TDRG1 and ELAVL4 genes, suggesting a potential role for host genes to mediate the link between the fecal microbiome and obesity. CONCLUSIONS: Our results provide novel insights into the role of the fecal microbiota in cardio-metabolic disease with clear potential for prevention and novel therapies.

SELECTION OF CITATIONS
SEARCH DETAIL