Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 31(23): 3945-3966, 2022 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-35848942

RESUMEN

Given the highly variable clinical phenotype of Coronavirus disease 2019 (COVID-19), a deeper analysis of the host genetic contribution to severe COVID-19 is important to improve our understanding of underlying disease mechanisms. Here, we describe an extended genome-wide association meta-analysis of a well-characterized cohort of 3255 COVID-19 patients with respiratory failure and 12 488 population controls from Italy, Spain, Norway and Germany/Austria, including stratified analyses based on age, sex and disease severity, as well as targeted analyses of chromosome Y haplotypes, the human leukocyte antigen region and the SARS-CoV-2 peptidome. By inversion imputation, we traced a reported association at 17q21.31 to a ~0.9-Mb inversion polymorphism that creates two highly differentiated haplotypes and characterized the potential effects of the inversion in detail. Our data, together with the 5th release of summary statistics from the COVID-19 Host Genetics Initiative including non-Caucasian individuals, also identified a new locus at 19q13.33, including NAPSA, a gene which is expressed primarily in alveolar cells responsible for gas exchange in the lung.


Asunto(s)
COVID-19 , Humanos , COVID-19/genética , SARS-CoV-2/genética , Estudio de Asociación del Genoma Completo , Haplotipos , Polimorfismo Genético
2.
Gastroenterology ; 160(5): 1784-1798.e0, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33387530

RESUMEN

BACKGROUND & AIMS: To influence host and disease phenotype, compositional microbiome changes, which have been demonstrated in patients with primary sclerosing cholangitis (PSC), must be accompanied by functional changes. We therefore aimed to characterize the genetic potential of the gut microbiome in patients with PSC compared with healthy controls (HCs) and patients with inflammatory bowel disease (IBD). METHODS: Fecal DNA from 2 cohorts (1 Norwegian and 1 German), in total comprising 136 patients with PSC (58% with IBD), 158 HCs, and 93 patients with IBD without PSC, were subjected to metagenomic shotgun sequencing, generating 17 billion paired-end sequences, which were processed using HUMAnN2 and MetaPhlAn2, and analyzed using generalized linear models and random effects meta-analyses. RESULTS: Patients with PSC had fewer microbial genes compared with HCs (P < .0001). Compared with HCs, patients with PSC showed enrichment and increased prevalence of Clostridium species and a depletion of, for example, Eubacterium spp and Ruminococcus obeum. Patients with PSC showed marked differences in the abundance of genes related to vitamin B6 synthesis and branched-chain amino acid synthesis (Qfdr < .05). Targeted metabolomics of plasma from an independent set of patients with PSC and controls found reduced concentrations of vitamin B6 and branched-chain amino acids in PSC (P < .0001), which strongly associated with reduced liver transplantation-free survival (log-rank P < .001). No taxonomic or functional differences were detected between patients with PSC with and without IBD. CONCLUSIONS: The gut microbiome in patients with PSC exhibits large functional differences compared with that in HCs, including microbial metabolism of essential nutrients. Alterations in related circulating metabolites associated with disease course, suggesting that microbial functions may be relevant for the disease process in PSC.


Asunto(s)
Bacterias/metabolismo , Colangitis Esclerosante/microbiología , Microbioma Gastrointestinal , Metaboloma , Metagenoma , Adolescente , Adulto , Anciano , Bacterias/genética , Estudios de Casos y Controles , Colangitis Esclerosante/sangre , Colangitis Esclerosante/diagnóstico , Colangitis Esclerosante/cirugía , Estudios Transversales , Disbiosis , Heces/microbiología , Femenino , Alemania , Humanos , Trasplante de Hígado , Masculino , Metabolómica , Metagenómica , Persona de Mediana Edad , Noruega , Filogenia , Supervivencia sin Progresión , Adulto Joven
3.
BMC Microbiol ; 21(1): 276, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34635060

RESUMEN

Compared to the huge microbial diversity in most mammals, human gut microbiomes have lost diversity while becoming specialized for animal-based diets - especially compared to chimps, their genetically closest ancestors. The lowered microbial diversity within the gut of westernized populations has also been associated with different kinds of chronic inflammatory diseases in humans. To further deepen our knowledge on phylogenetic and ecologic impacts on human health and fitness, we established the herein presented biobank as well as its comprehensive microbiota analysis. In total, 368 stool samples from 38 different animal species, including Homo sapiens, belonging to four diverse mammalian orders were collected at seven different locations and analyzed by 16S rRNA gene amplicon sequencing. Comprehensive data analysis was performed to (i) determine the overall impact of host phylogeny vs. diet, location, and ecology and to (ii) examine the general pattern of fecal bacterial diversity across captive mammals and humans.By using a controlled study design with captive mammals we could verify that host phylogeny is the most dominant driver of mammalian gut microbiota composition. However, the effect of ecology appears to be able to overcome host phylogeny and should therefore be studied in more detail in future studies. Most importantly, our study could observe a remarkable decrease of Spirochaetes and Prevotella in westernized humans and platyrrhines, which is probably not only due to diet, but also to the social behavior and structure in these communities.Our study highlights the importance of phylogenetic relationship and ecology within the evolution of mammalian fecal microbiota composition. Particularly, the observed decrease of Spirochaetes and Prevotella in westernized communities might be associated to lifestyle dependent rapid evolutionary changes, potentially involved in the establishment of dysbiotic microbiomes, which promote the etiology of chronic diseases.


Asunto(s)
Ecosistema , Heces/microbiología , Microbioma Gastrointestinal/fisiología , Prevotella/fisiología , Spirochaetales/fisiología , Población Urbana , Bacterias/clasificación , Bacterias/genética , Biodiversidad , Dieta , Humanos , Filogenia , ARN Ribosómico 16S/genética
4.
BMC Microbiol ; 21(1): 162, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34078289

RESUMEN

BACKGROUND: Human well-being has been linked to the composition and functional capacity of the intestinal microbiota. As regular exercise is known to improve human health, it is not surprising that exercise was previously described to positively modulate the gut microbiota, too. However, most previous studies mainly focused on either elite athletes or animal models. Thus, we conducted a randomised intervention study that focused on the effects of different types of training (endurance and strength) in previously physically inactive, healthy adults in comparison to controls that did not perform regular exercise. Overall study duration was ten weeks including six weeks of intervention period. In addition to 16S rRNA gene amplicon sequencing of longitudinally sampled faecal material of participants (six time points), detailed body composition measurements and analysis of blood samples (at baseline and after the intervention) were performed to obtain overall physiological changes within the intervention period. Activity tracker devices (wrist-band wearables) provided activity status and sleeping patterns of participants as well as exercise intensity and heart measurements. RESULTS: Different biometric responses between endurance and strength activities were identified, such as a significant increase of lymphocytes and decrease of mean corpuscular haemoglobin concentration (MCHC) only within the strength intervention group. In the endurance group, we observed a significant reduction in hip circumference and an increase in physical working capacity (PWC). Though a large variation of microbiota changes were observed between individuals of the same group, we did not find specific collective alterations in the endurance nor the strength groups, arguing for microbiome variations specific to individuals, and therefore, were not captured in our analysis. CONCLUSIONS: We could show that different types of exercise have distinct but moderate effects on the overall physiology of humans and very distinct microbial changes in the gut. The observed overall changes during the intervention highlight the importance of physical activity on well-being. Future studies should investigate the effect of exercise on a longer timescale, investigate different training intensities and consider high-resolution shotgun metagenomics technology. TRIAL REGISTRATION: DRKS, DRKS00015873 . Registered 12 December 2018; Retrospectively registered.


Asunto(s)
Ejercicio Físico , Microbioma Gastrointestinal , Adulto , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , ADN Bacteriano/genética , Dieta , Heces/microbiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , ARN Ribosómico 16S/genética , Adulto Joven
5.
Gut ; 69(4): 665-672, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31243055

RESUMEN

BACKGROUND: Patients with primary sclerosing cholangitis (PSC) display an altered colonic microbiome compared with healthy controls. However, little is known on the bile duct microbiome and its interplay with bile acid metabolism in PSC. METHODS: Patients with PSC (n=43) and controls without sclerosing cholangitis (n=22) requiring endoscopic retrograde cholangiography were included prospectively. Leading indications in controls were sporadic choledocholithiasis and papillary adenoma. A total of 260 biospecimens were collected from the oral cavity, duodenal fluid and mucosa and ductal bile. Microbiomes of the upper alimentary tract and ductal bile were profiled by sequencing the 16S-rRNA-encoding gene (V1-V2). Bile fluid bile acid composition was measured by high-performance liquid chromatography mass spectrometry and validated in an external cohort (n=20). RESULTS: The bile fluid harboured a diverse microbiome that was distinct from the oral cavity, the duodenal fluid and duodenal mucosa communities. The upper alimentary tract microbiome differed between PSC patients and controls. However, the strongest differences between PSC patients and controls were observed in the ductal bile fluid, including reduced biodiversity (Shannon entropy, p=0.0127) and increase of pathogen Enterococcus faecalis (FDR=4.18×10-5) in PSC. Enterococcus abundance in ductal bile was strongly correlated with concentration of the noxious secondary bile acid taurolithocholic acid (r=0.60, p=0.0021). CONCLUSION: PSC is characterised by an altered microbiome of the upper alimentary tract and bile ducts. Biliary dysbiosis is linked with increased concentrations of the proinflammatory and potentially cancerogenic agent taurolithocholic acid.


Asunto(s)
Bilis/microbiología , Colangitis Esclerosante/microbiología , Disbiosis/complicaciones , Microbiota , Adulto , Anciano , Anciano de 80 o más Años , Conductos Biliares/microbiología , Estudios de Casos y Controles , Estudios de Cohortes , Duodeno/microbiología , Disbiosis/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mucosa Bucal/microbiología , Adulto Joven
6.
Nucleic Acids Res ; 46(4): e23, 2018 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-29194524

RESUMEN

The vast majority of microorganisms on Earth reside in often-inseparable environment-specific communities-microbiomes. Meta-genomic/-transcriptomic sequencing could reveal the otherwise inaccessible functionality of microbiomes. However, existing analytical approaches focus on attributing sequencing reads to known genes/genomes, often failing to make maximal use of available data. We created faser (functional annotation of sequencing reads), an algorithm that is optimized to map reads to molecular functions encoded by the read-correspondent genes. The mi-faser microbiome analysis pipeline, combining faser with our manually curated reference database of protein functions, accurately annotates microbiome molecular functionality. mi-faser's minutes-per-microbiome processing speed is significantly faster than that of other methods, allowing for large scale comparisons. Microbiome function vectors can be compared between different conditions to highlight environment-specific and/or time-dependent changes in functionality. Here, we identified previously unseen oil degradation-specific functions in BP oil-spill data, as well as functional signatures of individual-specific gut microbiome responses to a dietary intervention in children with Prader-Willi syndrome. Our method also revealed variability in Crohn's Disease patient microbiomes and clearly distinguished them from those of related healthy individuals. Our analysis highlighted the microbiome role in CD pathogenicity, demonstrating enrichment of patient microbiomes in functions that promote inflammation and that help bacteria survive it.


Asunto(s)
Metagenómica/métodos , Microbiota , Anotación de Secuencia Molecular/métodos , Algoritmos , Proteínas Bacterianas/fisiología , Niño , Enfermedad de Crohn/microbiología , Humanos , Síndrome de Prader-Willi/microbiología , Alineación de Secuencia
7.
J Allergy Clin Immunol ; 141(5): 1668-1676.e16, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29421277

RESUMEN

BACKGROUND: Genomic approaches have revealed characteristic site specificities of skin bacterial community structures. In addition, in children with atopic dermatitis (AD), characteristic shifts were described at creases and, in particular, during flares, which have been postulated to mirror disturbed skin barrier function, cutaneous inflammation, or both. OBJECTIVE: We sought to comprehensively analyze microbial configurations in patients with AD across body sites and to explore the effect of distinct abnormalities of epidermal barrier function. METHODS: The skin microbiome was determined by using bacterial 16S rRNA sequencing at 4 nonlesional body sites, as well as acute and chronic lesions of 10 patients with AD and 10 healthy control subjects matched for age, sex, and filaggrin (FLG) mutation status. Nonlesional sampling sites were characterized for skin physiology parameters, including chromatography-based lipid profiling. RESULTS: Epidermal lipid composition, in particular levels of long-chain unsaturated free fatty acids, strongly correlated with bacterial composition, in particular Propionibacteria and Corynebacteria abundance. AD displayed a distinct community structure, with increased abundance and altered composition of staphylococcal species across body sites, the strongest loss of diversity and increase in Staphylococcus aureus seen on chronic lesions, and a progressive shift from nonlesional skin to acute and chronic lesions. FLG-deficient skin showed a distinct microbiome composition resembling in part the AD-related pattern. CONCLUSION: Epidermal barrier integrity and function affect the skin microbiome composition. AD shows an altered microbial configuration across diverse body sites, which is most pronounced at sites of predilection and AD. Eczematous affection appears to be a more important determinant than body site.


Asunto(s)
Eccema/microbiología , Eccema/patología , Epidermis/microbiología , Epidermis/patología , Inflamación/microbiología , Inflamación/patología , Lípidos/fisiología , Adulto , Estudios de Casos y Controles , Dermatitis Atópica/genética , Dermatitis Atópica/microbiología , Dermatitis Atópica/patología , Eccema/genética , Femenino , Proteínas Filagrina , Humanos , Inflamación/genética , Proteínas de Filamentos Intermediarios/genética , Masculino , Microbiota/fisiología , Persona de Mediana Edad , ARN Ribosómico 16S/genética , Fenómenos Fisiológicos de la Piel/genética , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/genética , Adulto Joven
8.
Front Med (Lausanne) ; 10: 1200317, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37588005

RESUMEN

Inflammatory Bowel Disease (IBD) affects approximately 0.3% of the global population, with incidence rates rising dramatically worldwide. Emerging evidence points to an interplay between exposome factors such as diet and gut microbiota, host genetics, and the immune system as crucial elements in IBD development. ATP-binding cassette (ABC) transporters, including human p-glycoprotein encoded by the Abcb1 gene, influence intestinal inflammation, and their expression may interact with environmental factors such as diet and gut microbes. Our study aimed to examine the impact of protein sources on a genetic colitis mouse model. Methods: Abcb1a-deficient colitis mice were fed either casein or red meat-supplemented diets to investigate potential colitis-aggravating components in red meat and their effects on host-microbiota interactions. We conducted deep label free quantitative proteomic inflammation profiling of gastrointestinal tissue (colon, ileum) and urine, and determined the overall microbiome in feces using 16S rRNA gene sequencing. Microbiota shifts by diet and protein transporter impairment were addressed by multivariate statistical analysis. Colon and systemic gut inflammation were validated through histology and immune assays, respectively. Results: A quantitative discovery based proteomic analysis of intestinal tissue and urine revealed associations between ileum and urine proteomes in relation to Abcb1a deficiency. The absence of Abcb1a efflux pump function and diet-induced intestinal inflammation impacted multiple systemic immune processes, including extensive neutrophil extracellular trap (NET) components observed in relation to neutrophil degranulation throughout the gastrointestinal tract. The colitis model's microbiome differed significantly from that of wild-type mice, indicating the substantial influence of efflux transporter deficiency on microbiota. Conclusion: The proteomic and microbiota analyzes of a well-established murine model enabled the correlation of gastrointestinal interactions not readily identifiable in human cohorts. Insights into dysregulated biological pathways in this disease model might offer translational biomarkers based on NETs and improved understanding of IBD pathogenesis in human patients. Our findings demonstrate that drug transporter deficiency induces substantial changes in the microbiota, leading to increased levels of IBD-associated strains and resulting in intestinal inflammation. GRAPHICAL ABSTRACT.

9.
Sci Rep ; 13(1): 5968, 2023 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-37045850

RESUMEN

The role of the human gut microbiome in colorectal cancer (CRC) is unclear as most studies on the topic are unable to discern correlation from causation. We apply two-sample Mendelian randomization (MR) to estimate the causal relationship between the gut microbiome and CRC. We used summary-level data from independent genome-wide association studies to estimate the causal effect of 14 microbial traits (n = 3890 individuals) on overall CRC (55,168 cases, 65,160 controls) and site-specific CRC risk, conducting several sensitivity analyses to understand the nature of results. Initial MR analysis suggested that a higher abundance of Bifidobacterium and presence of an unclassified group of bacteria within the Bacteroidales order in the gut increased overall and site-specific CRC risk. However, sensitivity analyses suggested that instruments used to estimate relationships were likely complex and involved in many potential horizontal pleiotropic pathways, demonstrating that caution is needed when interpreting MR analyses with gut microbiome exposures. In assessing reverse causality, we did not find strong evidence that CRC causally affected these microbial traits. Whilst our study initially identified potential causal roles for two microbial traits in CRC, importantly, further exploration of these relationships highlighted that these were unlikely to reflect causality.


Asunto(s)
Neoplasias Colorrectales , Microbioma Gastrointestinal , Humanos , Microbioma Gastrointestinal/genética , Análisis de la Aleatorización Mendeliana/métodos , Estudio de Asociación del Genoma Completo , Causalidad , Neoplasias Colorrectales/genética , Polimorfismo de Nucleótido Simple
10.
PLoS Negl Trop Dis ; 15(3): e0009232, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33657123

RESUMEN

Intestinal parasitic infections, caused by helminths and protozoa, are globally distributed and major causes of worldwide morbidity. The gut microbiota may modulate parasite virulence and host response upon infection. The complex interplay between parasites and the gut microbiota is poorly understood, partly due to sampling difficulties in remote areas with high parasite burden. In a large study of children in Guinea-Bissau, we found high prevalence of intestinal parasites. By sequencing of the 16S rRNA genes of fecal samples stored on filter paper from a total of 1,204 children, we demonstrate that the bacterial microbiota is not significantly altered by helminth infections, whereas it is shaped by the presence of both pathogenic and nonpathogenic protozoa, including Entamoeba (E.) spp. and Giardia (G.) lamblia. Within-sample diversity remains largely unaffected, whereas overall community composition is significantly affected by infection with both nonpathogenic E. coli (R2 = 0.0131, P = 0.0001) and Endolimax nana (R2 = 0.00902, P = 0.0001), and by pathogenic E. histolytica (R2 = 0.0164, P = 0.0001) and G. lamblia (R2 = 0.00676, P = 0.0001). Infections with multiple parasite species induces more pronounced shifts in microbiota community than mild ones. A total of 31 bacterial genera across all four major bacterial phyla were differentially abundant in protozoan infection as compared to noninfected individuals, including increased abundance of Prevotella, Campylobacter and two Clostridium clades, and decreased abundance of Collinsella, Lactobacillus, Ruminococcus, Veillonella and one Clostridium clade. In the present study, we demonstrate that the fecal bacterial microbiota is shaped by intestinal parasitic infection, with most pronounced associations for protozoan species. Our results provide insights into the interplay between the microbiota and intestinal parasites, which are valuable to understand infection biology and design further studies aimed at optimizing treatment strategies.


Asunto(s)
Microbioma Gastrointestinal , Infecciones por Protozoos/microbiología , Infecciones por Protozoos/parasitología , Adolescente , Animales , Bacterias/clasificación , Bacterias/genética , Niño , Preescolar , Coinfección/microbiología , Coinfección/parasitología , Entamoeba/aislamiento & purificación , Heces/microbiología , Heces/parasitología , Femenino , Giardia/aislamiento & purificación , Guinea Bissau , Helmintos/aislamiento & purificación , Humanos , Parasitosis Intestinales/epidemiología , Parasitosis Intestinales/parasitología , Masculino , ARN Ribosómico 16S
11.
Nat Genet ; 53(2): 156-165, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33462485

RESUMEN

To study the effect of host genetics on gut microbiome composition, the MiBioGen consortium curated and analyzed genome-wide genotypes and 16S fecal microbiome data from 18,340 individuals (24 cohorts). Microbial composition showed high variability across cohorts: only 9 of 410 genera were detected in more than 95% of samples. A genome-wide association study of host genetic variation regarding microbial taxa identified 31 loci affecting the microbiome at a genome-wide significant (P < 5 × 10-8) threshold. One locus, the lactase (LCT) gene locus, reached study-wide significance (genome-wide association study signal: P = 1.28 × 10-20), and it showed an age-dependent association with Bifidobacterium abundance. Other associations were suggestive (1.95 × 10-10 < P < 5 × 10-8) but enriched for taxa showing high heritability and for genes expressed in the intestine and brain. A phenome-wide association study and Mendelian randomization identified enrichment of microbiome trait loci in the metabolic, nutrition and environment domains and suggested the microbiome might have causal effects in ulcerative colitis and rheumatoid arthritis.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Variación Genética , Sitios de Carácter Cuantitativo , Adolescente , Adulto , Bifidobacterium/genética , Niño , Preescolar , Estudios de Cohortes , Femenino , Microbioma Gastrointestinal/genética , Estudio de Asociación del Genoma Completo , Humanos , Lactasa/genética , Desequilibrio de Ligamiento , Masculino , Análisis de la Aleatorización Mendeliana , Metabolismo/genética , ARN Ribosómico 16S
12.
Sci Total Environ ; 734: 139471, 2020 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-32464382

RESUMEN

The translocation of non-indigenous species (NIS) around the world, especially in marine systems, is increasingly being recognized as a matter of concern. Species translocations have been shown to lead to wide ranging changes in food web structure and functioning. In addition to the direct effects of NIS, they could facilitate the accumulation or translocation of bacteria as part of their microbiomes. The Baltic Sea harbours many non-indigenous species, with most recent detection of the jellyfish Blackfordia virginica and the comb jelly Mnemiopsis leidyi in the low saline southwestern Baltic Sea. In this study, we used a multidisciplinary approach and investigated three gelatinous zooplankton species that co-occur in the same environment and feed on similar zooplankton food sources but show different histories of origin. The aim was to conduct a comparative microbiome analysis of indigenous and non-indigenous gelatinous zooplankton species in the low-saline southwestern Baltic Sea. Next-generation 16S rRNA marker gene sequencing of the V1/V2 region was employed to study the bacterial microbiome compositions. All tested species showed significant differences in their microbiome compositions (one way ANOSIM, R = 1, P < 0.008) with dissimilarities ranging from 85 to 92%. The indigenous jellyfish Aurelia aurita showed the highest bacterial operational taxonomic unit (OTU) richness. The overall differentiation between microbiomes was driven by eight indicator OTUs, which included Mycoplasma and Vibrio species. These bacteria can be problematic, as they include known pathogenic strains that are relevant to human health and aquaculture activities. Our results suggest that the impact assessment of NIS should consider potential pathogenic bacteria, enriched in the environment due to invasion, as potential risks to aquaculture activities.


Asunto(s)
Microbiota , Animales , ARN Ribosómico 16S , Escifozoos , Vibrio , Zooplancton
13.
Front Cell Infect Microbiol ; 10: 536282, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33194786

RESUMEN

Background: Offspring of mothers with gestational diabetes mellitus (GDM) have increased risk of developing metabolic disorders as they grow up. Microbial colonization of the newborn gut and environmental exposures affecting the configuration of the gut microbiota during infancy have been linked to increased risk of developing disease during childhood and adulthood. In a convenience sample, we examined whether the intestinal tract of children born to mothers with GDM is differentially colonized in early life compared to offspring of mothers with normal gestational glucose regulation. Secondly, we examined whether any such difference persists during infancy, thus potentially conferring increased risk of developing metabolic disease later in life. Methods: Fecal samples were collected from children of mothers with (n = 43) and without GDM (n = 82) during the first week of life and again at an average age of 9 months. The gut microbiota was characterized by 16S rRNA gene amplicon sequencing (V1-V2). Differences in diversity and composition according to maternal GDM status were assessed, addressing potential confounding by mode of delivery, perinatal antibiotics treatment, feeding and infant sex. Results: Children of mothers with GDM were featured by a differential composition of the gut microbiota, both during the first week of life and at 9 months, at higher taxonomic and OTU levels. Sixteen and 15 OTUs were differentially abundant after correction for multiple testing during the first week of life and at 9 months, respectively. Two OTUs remained differentially abundant after adjustment for potential confounders both during the first week of life and at 9 months. Richness (OTU) was decreased in neonates born to mothers with GDM; however, at 9 months no difference in richness was observed. There was no difference in Shannon's diversity or Pielou's evenness at any timepoint. Longitudinally, we detected differential changes in the gut microbiota composition from birth to infancy according to GDM status. Conclusion: Differences in glycaemic regulation in late pregnancy is linked with relatively modest variation in the gut microbiota composition of the offspring during the first week of life and 9 months after birth.


Asunto(s)
Diabetes Gestacional , Microbioma Gastrointestinal , Adulto , Glucemia , Niño , Femenino , Humanos , Lactante , Recién Nacido , Madres , Embarazo , ARN Ribosómico 16S/genética
14.
Nat Microbiol ; 5(9): 1079-1087, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32572223

RESUMEN

Recent population-based1-4 and clinical studies5 have identified a range of factors associated with human gut microbiome variation. Murine quantitative trait loci6, human twin studies7 and microbiome genome-wide association studies1,3,8-12 have provided evidence for genetic contributions to microbiome composition. Despite this, there is still poor overlap in genetic association across human studies. Using appropriate taxon-specific models, along with support from independent cohorts, we show an association between human host genotype and gut microbiome variation. We also suggest that interpretation of applied analyses using genetic associations is complicated by the probable overlap between genetic contributions and heritable components of host environment. Using faecal 16S ribosomal RNA gene sequences and host genotype data from the Flemish Gut Flora Project (n = 2,223) and two German cohorts (FoCus, n = 950; PopGen, n = 717), we identify genetic associations involving multiple microbial traits. Two of these associations achieved a study-level threshold of P = 1.57 × 10-10; an association between Ruminococcus and rs150018970 near RAPGEF1 on chromosome 9, and between Coprococcus and rs561177583 within LINC01787 on chromosome 1. Exploratory analyses were undertaken using 11 other genome-wide associations with strong evidence for association (P < 2.5 × 10-8) and a previously reported signal of association between rs4988235 (MCM6/LCT) and Bifidobacterium. Across these 14 single-nucleotide polymorphisms there was evidence of signal overlap with other genome-wide association studies, including those for age at menarche and cardiometabolic traits. Mendelian randomization analysis was able to estimate associations between microbial traits and disease (including Bifidobacterium and body composition); however, in the absence of clear microbiome-driven effects, caution is needed in interpretation. Overall, this work marks a growing catalogue of genetic associations that will provide insight into the contribution of host genotype to gut microbiome. Despite this, the uncertain origin of association signals will likely complicate future work looking to dissect function or use associations for causal inference analysis.


Asunto(s)
Microbioma Gastrointestinal/genética , Estudio de Asociación del Genoma Completo , Microbiota/genética , Animales , Bifidobacterium/genética , Heces/microbiología , Genotipo , Humanos , Ratones , Fenotipo , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , ARN Ribosómico 16S/genética
15.
Artículo en Inglés | MEDLINE | ID: mdl-31624178

RESUMEN

OBJECTIVE: To determine whether the gut microbiota shows overabundance of commensal bacteria species in patients with anti-NMDA receptor (NMDAR) encephalitis, similar to patients with MS or neuromyelitis optica where they potentially balance pro- and anti-inflammatory immune responses or participate in disease pathogenesis by molecular mimicry. METHODS: Intestinal microbiota was characterized in patients with NMDAR encephalitis (n = 23, mean age: 34 ± 12.7 years; 21 females) and age/sex/environment-matched healthy controls (n = 24, 40 ± 14.2 years; 22 females) using stool bacteria 16S rDNA sequencing and classification in operational taxonomic units (OTUs). Statistical analyses focused on intraindividual and interindividual bacterial diversity and identification of differentially abundant taxa. RESULTS: Patients with NMDAR encephalitis and controls had similar microbiome profiles of the gut microbiota regarding intraindividual bacterial diversity, OTU distribution, ratio between regional and local species diversity when testing all OTUs, and genera with a relative abundance greater than 0.5%. Similarly, the subgroup of NMDAR encephalitis patients with an ovarian teratoma (n = 3) showed no differences in microbiome variation compared with controls. Patients in the acute encephalitis stage (n = 8) showed significant differences in the numbers of Clostridium XVIII, Clostridium IV, Oscillibacter, Prevotella, and Blautia; however, significance was lost after correction for multiple testing. CONCLUSION: Patients with NMDAR encephalitis and controls both had a normal gut microbiome. The lack of overabundance of certain bacterial species in patients suggests that microbiome changes are no major contributors to the pathogenesis, disease course, or prognosis in NMDAR encephalitis. Despite the small sample size and heterogeneous groups, findings indicate differences to other neuroimmunologic diseases.


Asunto(s)
Encefalitis Antirreceptor N-Metil-D-Aspartato/microbiología , Microbioma Gastrointestinal , Enfermedad Aguda , Adulto , Heces/microbiología , Femenino , Humanos , Masculino , Persona de Mediana Edad
16.
Sci Rep ; 9(1): 19008, 2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31831829

RESUMEN

The gut microbiota is recognized as having major impact in health and disease. Sample storage is an important aspect to obtain reliable results. Mostly recommended is immediate freezing, however, this is not always feasible. Faecal occult blood test (FOBT) papers are an appealing solution in such situations, and most studies find these to be applicable, showing no major changes within 7 days storage at room temperature (RT). As fieldwork often requires RT storage for longer periods, evaluation of this is warranted. We performed 16S rRNA gene sequencing of 19 paired faecal samples immediately frozen or kept five weeks and five months at RT on FOBT papers. Alpha-diversity evaluation revealed no effect of FOBT storage, and evaluation of beta-diversity showed that host explained 65% of community variation, while storage method explained 5%. Evaluation of community dispersion and the Firmicutes/Bacteroidetes ratio revealed a larger effect of storage time for fresh-frozen samples. Single taxa evaluation (order-to-genus level) showed significant alterations of four (of 37) genera after five weeks and five genera after five months. When comparing the two timepoints, alterations were only detectable for fresh-frozen samples. Our findings reveal that long term storage on FOBT papers is an applicable approach for microbiota research.


Asunto(s)
Heces/microbiología , Filtración , Microbiota , Preservación Biológica , Temperatura , Adulto , Bacterias/clasificación , Biodiversidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Papel , Filogenia , Adulto Joven
17.
J Innate Immun ; 11(5): 393-404, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30566939

RESUMEN

Animals are usually regarded as independent entities within their respective environments. However, within an organism, eukaryotes and prokaryotes interact dynamically to form the so-called metaorganism or holobiont, where each partner fulfils its versatile and crucial role. This review focuses on the interplay between microorganisms and multicellular eukaryotes in the context of host physiology, in particular aging and mucus-associated crosstalk. In addition to the interactions between bacteria and the host, we highlight the importance of viruses and nonmodel organisms. Moreover, we discuss current culturing and computational methodologies that allow a deeper understanding of underlying mechanisms controlling the physiology of metaorganisms.


Asunto(s)
Interacciones Microbiota-Huesped/fisiología , Microbiota/fisiología , Envejecimiento , Animales , Biología Computacional , Estado de Salud , Humanos , Modelos Biológicos , Moco/microbiología , Moco/virología , Simbiosis/fisiología
18.
Cell Host Microbe ; 26(2): 252-264.e10, 2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31399369

RESUMEN

Obesity and type 2 diabetes (T2D) are metabolic disorders that are linked to microbiome alterations. However, their co-occurrence poses challenges in disentangling microbial features unique to each condition. We analyzed gut microbiomes of lean non-diabetic (n = 633), obese non-diabetic (n = 494), and obese individuals with T2D (n = 153) from German population and metabolic disease cohorts. Microbial taxonomic and functional profiles were analyzed along with medical histories, serum metabolomics, biometrics, and dietary data. Obesity was associated with alterations in microbiome composition, individual taxa, and functions with notable changes in Akkermansia, Faecalibacterium, Oscillibacter, and Alistipes, as well as in serum metabolites that correlated with gut microbial patterns. However, microbiome associations were modest for T2D, with nominal increases in Escherichia/Shigella. Medications, including antihypertensives and antidiabetics, along with dietary supplements including iron, were significantly associated with microbiome variation. These results differentiate microbial components of these interrelated metabolic diseases and identify dietary and medication exposures to consider in future studies.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Microbioma Gastrointestinal/fisiología , Obesidad/complicaciones , Obesidad/microbiología , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Biodiversidad , Dieta , Suplementos Dietéticos , Heces/microbiología , Femenino , Alemania , Humanos , Hierro/metabolismo , Magnesio/metabolismo , Masculino , Enfermedades Metabólicas/complicaciones , Metagenómica , Ratones , Ratones Endogámicos C57BL , Análisis Multivariante , Evaluación Nutricional , Suero/metabolismo
19.
Gut Microbes ; 9(1): 68-75, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-28816579

RESUMEN

Factors shaping the human intestinal microbiota range from environmental influences, like smoking and exercise, over dietary patterns and disease to the host's genetic variation. Recently, we could show in a microbiome genome-wide association study (mGWAS) targeting genetic variation influencing the ß diversity of gut microbial communities, that approximately 10% of the overall gut microbiome variation can be explained by host genetics. Here, we report on the application of a new method for genotype-ß-diversity association testing, the distance-based F (DBF) test. With this we identified 4 loci with genome-wide significant associations, harboring the genes CBEP4, SLC9A8, TNFSF4, and SP140, respectively. Our findings highlight the utility of the high-performance DBF test in ß diversity GWAS and emphasize the important role of host genetics and immunity in shaping the human intestinal microbiota.


Asunto(s)
Bacterias/genética , Biodiversidad , Microbioma Gastrointestinal , Sitios Genéticos/genética , Estudio de Asociación del Genoma Completo , Modelos Estadísticos , Antígenos Nucleares/genética , Bacterias/clasificación , Variación Genética , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad/genética , Ligando OX40/genética , Proteínas de Unión al ARN/genética , Reproducibilidad de los Resultados , Intercambiadores de Sodio-Hidrógeno/genética , Factores de Transcripción/genética
20.
Microbiome ; 6(1): 89, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29764499

RESUMEN

BACKGROUND: Imbalances of gut microbiota composition are linked to a range of metabolic perturbations. In the present study, we examined the gut microbiota of women with gestational diabetes mellitus (GDM) and normoglycaemic pregnant women in late pregnancy and about 8 months postpartum. METHODS: Gut microbiota profiles of women with GDM (n = 50) and healthy (n = 157) pregnant women in the third trimester and 8 months postpartum were assessed by 16S rRNA gene amplicon sequencing of the V1-V2 region. Insulin and glucose homeostasis were evaluated by a 75 g 2-h oral glucose tolerance test during and after pregnancy. RESULTS: Gut microbiota of women with GDM was aberrant at multiple levels, including phylum and genus levels, compared with normoglycaemic pregnant women. Actinobacteria at phylum level and Collinsella, Rothia and Desulfovibrio at genus level had a higher abundance in the GDM cohort. Difference in abundance of 17 species-level operational taxonomic units (OTUs) during pregnancy was associated with GDM. After adjustment for pre-pregnancy body mass index (BMI), 5 of the 17 OTUs showed differential abundance in the GDM cohort compared with the normoglycaemic pregnant women with enrichment of species annotated to Faecalibacterium and Anaerotruncus and depletion of species annotated to Clostridium (sensu stricto) and to Veillonella. OTUs assigned to Akkermansia were associated with lower insulin sensitivity while Christensenella OTUs were associated with higher fasting plasma glucose concentration. OTU richness and Shannon index decreased from late pregnancy to postpartum regardless of metabolic status. About 8 months after delivery, the microbiota of women with previous GDM was still characterised by an aberrant composition. Thirteen OTUs were differentially abundant in women with previous GDM compared with women with previous normoglycaemic pregnancy. CONCLUSION: GDM diagnosed in the third trimester of pregnancy is associated with a disrupted gut microbiota composition compared with normoglycaemic pregnant women, and 8 months after pregnancy, differences in the gut microbiota signatures are still detectable. The gut microbiota composition of women with GDM, both during and after pregnancy, resembles the aberrant microbiota composition reported in non-pregnant individuals with type 2 diabetes and associated intermediary metabolic traits.


Asunto(s)
Diabetes Gestacional/microbiología , Disbiosis/microbiología , Microbioma Gastrointestinal/genética , Tracto Gastrointestinal/microbiología , Periodo Posparto/sangre , Tercer Trimestre del Embarazo/sangre , Actinobacteria/genética , Actinobacteria/aislamiento & purificación , Adulto , Glucemia , Índice de Masa Corporal , Clostridium/genética , Clostridium/aislamiento & purificación , Desulfovibrio/genética , Desulfovibrio/aislamiento & purificación , Faecalibacterium/genética , Faecalibacterium/aislamiento & purificación , Femenino , Glucosa/metabolismo , Humanos , Embarazo , ARN Ribosómico 16S/genética , Encuestas y Cuestionarios
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA