Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 544
Filtrer
1.
Ann Clin Microbiol Antimicrob ; 23(1): 72, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138497

RÉSUMÉ

BACKGROUND: Alongside microbiota development, the evolution of the resistome is crucial in understanding the early-life acquisition and persistence of Antibiotic Resistance Genes (ARGs). Therefore, the aim of this study is to provide a comprehensive view of the evolution and dynamics of the neonatal resistome from 7 days to 4 months of age using a high-throughput qPCR platform. METHODS: In the initial phase, a massive screening of 384 ARGs using a high-throughput qPCR in pooled healthy mother-infant pairs feces from the MAMI cohort was carried out to identify the most abundant and prevalent ARGs in infants and in mothers. This pre-analysis allowed for later targeted profiling in a large number of infants in a longitudinal manner during the first 4 months of life. 16S rRNA V3-V4 amplicon sequencing was performed to asses microbial composition longitudinally. Potential factors influencing the microbiota and ARGs in this period were also considered, such as mode of birth and breastfeeding type. RESULTS: Following the massive screening, the top 45 abundant ARGs and mobile genetic elements were identified and studied in 72 infants during their first months of life (7 days, 1, 2, and 4 months). These genes were associated with resistance to aminoglycosides, beta-lactams and tetracyclines, among others, as well as integrons, and other mobile genetic elements. Changes in both ARG composition and quantity were observed during the first 4 months of life: most ARGs abundance increased over time, but mobile genetic elements decreased significantly. Further exploration of modulating factors highlighted the effect on ARG composition of specific microbial genus, and the impact of mode of birth at 7 days and 4 months. The influence of infant formula feeding was observed at 4-month-old infants, who exhibited a distinctive resistome composition. CONCLUSIONS: This study illustrates the ARG evolution and dynamics in the infant gut by use of a targeted, high-throughput, quantitative PCR-based method. An increase in antibiotic resistance over the first months of life were observed with a fundamental role of delivery mode in shaping resistance profiles. Further, we highlighted the influence of feeding methods on the resistome development. These findings offer pivotal insights into dynamics of and factors influencing early-life resistome, with potential avenues for intervention strategies.


Sujet(s)
Antibactériens , Fèces , Microbiome gastro-intestinal , ARN ribosomique 16S , Humains , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Microbiome gastro-intestinal/génétique , Nourrisson , Nouveau-né , Fèces/microbiologie , Femelle , Antibactériens/pharmacologie , ARN ribosomique 16S/génétique , Mâle , Allaitement naturel , Bactéries/génétique , Bactéries/effets des médicaments et des substances chimiques , Bactéries/classification , Gènes bactériens/génétique , Résistance bactérienne aux médicaments/génétique , Évolution moléculaire , Résistance microbienne aux médicaments/génétique , Études longitudinales
2.
Food Res Int ; 192: 114840, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39147525

RÉSUMÉ

Bifidobacterium pseudocatenulatum is a prevalent gut microbe in humans of all ages and plays a crucial role in host health. However, its adaptive evolutionary characteristics remain poorly understood. This study analyzed the genome of 247 B. pseudocatenulatum isolates from Chinese, Vietnamese, Japanese and other region populations using population genomics and functional genomics. Our findings revealed high genetic heterogeneity and regional clustering within B. pseudocatenulatum isolates. Significant differences were observed in genome characteristics, phylogeny, and functional genes. Specifically, Chinese and Vietnamese isolates exhibited a higher abundance of genes involved in the metabolism of plant-derived carbohydrates (GH13, GH43, and GH5 enzyme families), aligning with the predominantly vegetable-, wheat- and fruit-based diets of these populations. Additionally, we found widespread transmission of antibiotic resistance genes (tetO and tetW) through mobile genetic elements, such as genomic islands (GIs), resulting in substantial intra-regional differences. Our findings highlight distinct adaptive evolution in B. pseudocatenulatum driven by gene specialization, possibly in response to regional variations in diet and lifestyle. This study sheds light on bifidobacteria colonization mechanisms in the host gut. IMPORTANCE: Gut microbiota, as a key link in the gut-brain axis, helps to maintain the health of the organism, among which, Bifidobacterium pseudocatenulatum (B. pseudocatenulatum) is an important constituent member of the gut microbiota, which plays an important role in maintaining the balance of gut microbiota. The probiotic properties of B. pseudocatenulatum have been widely elaborated, and in order to excavate its evolutionary features at the genomic level, here we focused on the genetic background and evolutionary mechanism of the B. pseudocatenulatum genomes isolated from the intestinal tracts of different populations. Ultimately, based on the phylogenetic tree, we found that B. pseudocatenulatum has high genetic diversity and regional clustering phenomenon, in which plant-derived carbohydrate metabolism genes (GH13, GH43, GH5) showed significant regional differences, and this genetic differentiation drove the adaptive evolution, which likely shaped by diet and lifestyle.


Sujet(s)
Bifidobacterium pseudocatenulatum , Microbiome gastro-intestinal , Génome bactérien , Phylogenèse , Microbiome gastro-intestinal/génétique , Humains , Bifidobacterium pseudocatenulatum/génétique , Bifidobacterium pseudocatenulatum/métabolisme , Variation génétique , Génomique , Régime alimentaire
3.
Microbiome ; 12(1): 155, 2024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39175056

RÉSUMÉ

BACKGROUND: Despite being among the most abundant biological entities on earth, bacteriophage (phage) remain an understudied component of host-associated systems. One limitation to studying host-associated phage is the lack of consensus on methods for sampling phage communities. Here, we compare paired total metagenomes and viral size fraction metagenomes (viromes) as methods for investigating the dsDNA viral communities associated with the GI tract of two bee species: the European honey bee Apis mellifera and the eastern bumble bee Bombus impatiens. RESULTS: We find that viromes successfully enriched for phage, thereby increasing phage recovery, but only in honey bees. In contrast, for bumble bees, total metagenomes recovered greater phage diversity. Across both bee species, viromes better sampled low occupancy phage, while total metagenomes were biased towards sampling temperate phage. Additionally, many of the phage captured by total metagenomes were absent altogether from viromes. Comparing between bees, we show that phage communities in commercially reared bumble bees are significantly reduced in diversity compared to honey bees, likely reflecting differences in bacterial titer and diversity. In a broader context, these results highlight the complementary nature of total metagenomes and targeted viromes, especially when applied to host-associated environments. CONCLUSIONS: Overall, we suggest that studies interested in assessing total communities of host-associated phage should consider using both approaches. However, given the constraints of virome sampling, total metagenomes may serve to sample phage communities with the understanding that they will preferentially sample dominant and temperate phage. Video Abstract.


Sujet(s)
Bactériophages , Métagénome , Virome , Abeilles/virologie , Abeilles/microbiologie , Animaux , Bactériophages/génétique , Bactériophages/isolement et purification , Bactériophages/classification , Microbiome gastro-intestinal/génétique , Métagénomique/méthodes , Tube digestif/microbiologie , Tube digestif/virologie
4.
Front Cell Infect Microbiol ; 14: 1398756, 2024.
Article de Anglais | MEDLINE | ID: mdl-39176264

RÉSUMÉ

Background: Gut microbiota is closely related to the occurrence and development of sepsis. However, the causal effects between the gut microbiota and sepsis, and whether circulating inflammatory proteins act as mediators, remain unclear. Methods: Gut microbiota, circulating inflammatory proteins, and four sepsis-related outcomes were identified from large-scale genome wide association studies (GWAS) summary data. Inverse Variance Weighted (IVW) was the primary statistical method. Additionally, we investigated whether circulating inflammatory proteins play a mediating role in the pathway from gut microbiota to the four sepsis-related outcomes. Results: There were 14 positive and 15 negative causal effects between genetic liability in the gut microbiota and four sepsis-related outcomes. Additionally, eight positive and four negative causal effects were observed between circulating inflammatory proteins and the four sepsis-related outcomes. Circulating inflammatory proteins do not act as mediators. Conclusions: Gut microbiota and circulating inflammatory proteins were causally associated with the four sepsis-related outcomes. However, circulating inflammatory proteins did not appear to mediate the pathway from gut microbiota to the four sepsis-related outcomes.


Sujet(s)
Microbiome gastro-intestinal , Étude d'association pangénomique , Analyse de randomisation mendélienne , Sepsie , Sepsie/microbiologie , Sepsie/sang , Humains , Microbiome gastro-intestinal/génétique , Inflammation/sang , Polymorphisme de nucléotide simple
5.
Ren Fail ; 46(2): 2393754, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39177227

RÉSUMÉ

OBJECTIVE: The aim of this study was to investigate the characteristics and related functional pathways of the gut microbiota in patients with IgA nephropathy (IgAN) through metagenomic sequencing technology. METHODS: We enrolled individuals with primary IgAN, including patients with normal and abnormal renal function. Additionally, we recruited healthy volunteers as the healthy control group. Stool samples were collected, and species and functional annotation were performed through fecal metagenome sequencing. We employed linear discriminant analysis effect size (LEfSe) analysis to identify significantly different bacterial microbiota and functional pathways. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis was used to annotate microbiota functions, and redundancy analysis (RDA) was performed to analyze the factors affecting the composition and distribution of the gut microbiota. RESULTS: LEfSe analysis revealed differences in the gut microbiota between IgAN patients and healthy controls. The characteristic microorganisms in the IgAN group were classified as Escherichia coli, with a significantly greater abundance than that in the healthy control group (p < 0.05). The characteristic microorganisms in the IgAN group with abnormal renal function were identified as Enterococcaceae, Moraxella, Moraxella, and Acinetobacter. KEGG functional analysis demonstrated that the functional pathways of the microbiota that differed between IgAN patients and healthy controls were related primarily to bile acid metabolism. CONCLUSIONS: The status of the gut microbiota is closely associated not only with the onset of IgAN but also with the renal function of IgAN patients. The characteristic gut microbiota may serve as a promising diagnostic biomarker and therapeutic target for IgAN.


Sujet(s)
Fèces , Microbiome gastro-intestinal , Glomérulonéphrite à dépôts d'IgA , Métagénomique , Humains , Glomérulonéphrite à dépôts d'IgA/microbiologie , Microbiome gastro-intestinal/génétique , Mâle , Femelle , Adulte , Fèces/microbiologie , Métagénomique/méthodes , Études cas-témoins , Adulte d'âge moyen , Moraxella/isolement et purification , Moraxella/génétique , Escherichia coli/isolement et purification , Escherichia coli/génétique , Acinetobacter/isolement et purification , Acinetobacter/génétique , Métagénome , Jeune adulte
6.
Front Endocrinol (Lausanne) ; 15: 1364280, 2024.
Article de Anglais | MEDLINE | ID: mdl-39157683

RÉSUMÉ

Background: Gut microbiota (GM) homeostasis in the human body is closely associated with health, which can be used as a regulator for preventing the onset and progression of disease. Diabetic microvascular complications bring about not only a huge economic burden to society, but also miserable mental and physical pain. Thus, alteration of the GM may be a method to delay diabetic microvascular complications. Objective: A two-sample Mendelian randomization (MR) analysis was conducted to reveal the causal inference between GM and three core diabetic microvascular complications, namely, diabetic kidney disease (DKD), diabetic retinopathy (DR), and diabetic neuropathy (DNP). Methods: First, genome-wide association study (GWAS) summary statistics for GM from the MiBioGen consortium and three main diabetic microvascular complications acquired from the FinnGen research project were assessed. Second, a forward MR analysis was conducted to assess the causality of GM on the risk of DKD, DR, and DNP. Third, a series of sensitivity studies, such as heterogeneity tests, pleiotropy evaluations, and leave-one-out analyses, were further conducted to assess the accuracy of MR analysis. Finally, Steiger tests and reverse MR analyses were performed to appraise the possibility of reverse causation. Results: A total of 2,092 single-nucleotide polymorphisms related to 196 bacterial traits were selected as instrumental variables. This two-sample MR analysis provided strongly reasonable evidence that 28 genetically predicted abundance of specific GM that played non-negligible roles in the occurrence of DKD, DR, and DNP complications were causally associated with 23 GM, the odds ratio of which generally ranged from 0.9 to 1.1. Further sensitivity analysis indicated low heterogeneity, low pleiotropy, and high reliability of the causal estimates. Conclusion: The study raised the possibility that GM may be a potential target to prevent and delay the progression of diabetic microvascular complications. Further experiments of GM therapy on diabetic microvascular complications are warranted to clarify their effects and specific mechanisms.


Sujet(s)
Angiopathies diabétiques , Microbiome gastro-intestinal , Étude d'association pangénomique , Analyse de randomisation mendélienne , Humains , Microbiome gastro-intestinal/génétique , Angiopathies diabétiques/génétique , Angiopathies diabétiques/microbiologie , Néphropathies diabétiques/génétique , Néphropathies diabétiques/microbiologie , Polymorphisme de nucléotide simple , Neuropathies diabétiques/génétique , Neuropathies diabétiques/microbiologie , Neuropathies diabétiques/étiologie , Rétinopathie diabétique/génétique , Rétinopathie diabétique/microbiologie , Rétinopathie diabétique/étiologie
7.
Microb Genom ; 10(8)2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39166974

RÉSUMÉ

Although the production of carbon monoxide (CO) within the human body has been detected, only two CO-utilizing prokaryotes (CO utilizers) have been reported in the human gut. Therefore, the phylogenetic diversity of the human gut CO-utilizing prokaryotes remains unclear. Here, we unveiled more than a thousand representative genomes containing genes for putative nickel-containing CO dehydrogenase (pCODH), an essential enzyme for CO utilization. The taxonomy of genomes encoding pCODH was expanded to include 8 phyla, comprising 82 genera and 248 species. In contrast, putative molybdenum-containing CODH genes were not detected in the human gut microbial genomes. pCODH transcripts were detected in 97.3 % (n=110) of public metatranscriptome datasets derived from healthy human faeces, suggesting the ubiquitous presence of prokaryotes bearing transcriptionally active pCODH genes in the human gut. More than half of the pCODH-encoding genomes contain a set of genes for the autotrophic Wood-Ljungdahl pathway (WLP). However, 79 % of these genomes commonly lack a key gene for the WLP, which encodes the enzyme that synthesizes formate from CO2, suggesting that potential human gut CO-utilizing prokaryotes share a degenerated gene set for WLP. In the other half of the pCODH-encoding genomes, seven genes, including putative genes for flavin adenine dinucleotide-dependent NAD(P) oxidoreductase (FNOR), ABC transporter and Fe-hydrogenase, were found adjacent to the pCODH gene. None of the putative genes associated with CO-oxidizing respiratory machinery, such as energy-converting hydrogenase genes, were found in pCODH-encoding genomes. This suggests that the human gut CO utilization is not for CO removal, but potentially for fixation and/or biosynthesis, consistent with the harmless yet continuous production of CO in the human gut. Our findings reveal the diversity and distribution of prokaryotes with pCODH in the human gut microbiome, suggesting their potential contribution to microbial ecosystems in human gut environments.


Sujet(s)
Aldehyde oxidoreductases , Bactéries , Monoxyde de carbone , Microbiome gastro-intestinal , Complexes multienzymatiques , Nickel , Phylogenèse , Humains , Aldehyde oxidoreductases/génétique , Aldehyde oxidoreductases/métabolisme , Microbiome gastro-intestinal/génétique , Nickel/métabolisme , Monoxyde de carbone/métabolisme , Complexes multienzymatiques/génétique , Bactéries/génétique , Bactéries/classification , Bactéries/enzymologie , Bactéries/isolement et purification , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme
8.
Front Cell Infect Microbiol ; 14: 1404745, 2024.
Article de Anglais | MEDLINE | ID: mdl-39165915

RÉSUMÉ

Background: Recent studies have shown that gut microbiota may be related to the occurrence of brain tumors, but direct evidence is lacking. This study used the Mendelian randomization study (MR) method to explore the potential causal link between gut microbiota and brain tumors. Method: We analyzed the genome-wide association data between 211 gut microbiota taxa and brain tumors, using the largest existing gut microbiota Genome-Wide Association Studies meta-analysis data (n=13266) and combining it with brain tumor data in the IEU OpenGWAS database. We use inverse-variance weighted analysis, supplemented by methods such as Mendelian randomization-Egger regression, weighted median estimator, simple mode, and weighted mode, to assess causality. In addition, we also conducted the Mendelian randomization-Egger intercept test, Cochran's Q test, and Mendelian randomization Steiger directionality test to ensure the accuracy of the analysis. Quality control includes sensitivity analysis, horizontal gene pleiotropy test, heterogeneity test, and MR Steiger directionality test. Result: Our study found that specific gut microbial taxa, such as order Lactobacillales and family Clostridiaceae1, were positively correlated with the occurrence of brain tumors, while genus Defluviitaleaceae UCG011 and genus Flavonifractor were negatively correlated with the occurrence of brain tumors. The Mendelian randomization-Egger intercept test showed that our analysis was not affected by pleiotropy (P>0.05). Conclusion: This study reveals for the first time the potential causal relationship between gut microbiota and brain tumors, providing a new perspective for the prevention and treatment of early brain tumors. These findings may help develop new clinical intervention strategies and point the way for future research.


Sujet(s)
Tumeurs du cerveau , Microbiome gastro-intestinal , Étude d'association pangénomique , Analyse de randomisation mendélienne , Humains , Microbiome gastro-intestinal/génétique , Tumeurs du cerveau/génétique , Tumeurs du cerveau/microbiologie , Bactéries/génétique , Bactéries/classification , Bactéries/isolement et purification
9.
Sci Rep ; 14(1): 17933, 2024 08 02.
Article de Anglais | MEDLINE | ID: mdl-39095393

RÉSUMÉ

The microbiome is a key factor in the health, well-being, and success of vertebrates, contributing to the adaptive capacity of the host. However, the impact of geographic and biotic factors that may affect the microbiome of wild birds in polar environments is not well defined. To address this, we determined the bacterial 16S rRNA gene sequence profiles in faecal samples from pygoscelid penguin populations in the Scotia Arc, focusing on gentoo penguins. This mesopredatory group breeds in defined colonies across a wide geographic range. Since diet could influence microbiome structure, we extracted dietary profiles from a eukaryotic 18S rRNA gene sequence profile. The bacterial microbiome profiles were considered in the context of a diverse set of environmental and ecological measures. Integrating wide geographic sampling with bacterial 16S and eukaryotic 18S rRNA gene sequencing of over 350 faecal samples identified associations between the microbiome profile and a suite of geographic and ecological factors. Microbiome profiles differed according to host species, colony identity, distance between colonies, and diet. Interestingly there was also a relationship between the proportion of host DNA (in relation to total 18S rRNA gene signal) and the microbiome, which may reflect gut passage time. Colony identity provided the strongest association with differences in microbiome profiles indicating that local factors play a key role in the microbiome structure of these polar seabirds. This may reflect the influence of local transfer of microbes either via faecal-oral routes, during chick feeding or other close contact events. Other factors including diet and host species also associate with variation in microbiome profile, and in at least some locations, the microbiome composition varies considerably between individuals. Given the variation in penguin microbiomes associated with diverse factors there is potential for disruption of microbiome associations at a local scale that could influence host health, productivity, and immunological competence. The microbiome represents a sensitive indicator of changing conditions, and the implications of any changes need to be considered in the wider context of environmental change and other stressors.


Sujet(s)
Fèces , Microbiote , ARN ribosomique 16S , Spheniscidae , Animaux , Spheniscidae/microbiologie , ARN ribosomique 16S/génétique , Microbiote/génétique , Fèces/microbiologie , Bactéries/classification , Bactéries/génétique , Bactéries/isolement et purification , ARN ribosomique 18S/génétique , Régime alimentaire , Microbiome gastro-intestinal/génétique
10.
BMC Cancer ; 24(1): 970, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39118079

RÉSUMÉ

BACKGROUND: Several studies have explored the potential link between gut microbiota and breast cancer; nevertheless, the causal relationship between gut microbiota and breast cancer remains unclear. METHODS: We utilized summary statistics from genome-wide association studies (GWAS) of the gut microbiome from the MiBioGen project with summary data from GWAS on breast cancer from the FinnGen consortium and the IEU database, with the IEU data sourced from the Biobank Japan. Preliminary statistical analyses were conducted using inverse variance weighting (IVW), supplemented by various sensitivity analysis methods, including MR-Egger regression, weighted median, weighted mode, simple median, and simple mode, to ensure the robustness of our findings. Heterogeneity and pleiotropy were assessed to avoid misleading conclusions caused by unconsidered confounders or non-specific effects of genetic variants, ensuring that the results reflect a genuine causal relationship. RESULTS: In European populations, four types of gut microbiota were associated with breast cancer. The genus Erysipelatoclostridium was positively associated with the risk of breast cancer, with an odds ratio (OR) of 1.21 (95% confidence interval [CI] 1.083-1.358), false discovery rate (FDR) = 0.0039. The class Coriobacteriia, order Coriobacteriales, and family Coriobacteriaceae, which belong to the same phylogenetic system, showed a consistent inversely association with breast cancer risk, with an OR of 0.757 (95% CI 0.616-0.930), FDR = 0.0281. In East Asian populations, three types of gut microbiota were related to breast cancer. The Eubacterium ruminantium group was positively associated with breast cancer risk, with an OR of 1.259 (95% CI 1.056-1.499), FDR = 0.0497. The families Porphyromonadaceae and Ruminococcaceae were inversely associated with breast cancer risk, with ORs of 0.304 (95% CI 0.155-0.596), FDR = 0.0005, and 0.674 (95% CI 0.508-0.895), FDR = 0.03173, respectively. However, these two taxa had limited instrumental variables, restricting the statistical power and potentially affecting the interpretation of the results. CONCLUSION: This MR analysis demonstrated a probable causal link between specific gut microbiota and breast cancer. This study, through Mendelian randomization analysis comparing European and East Asian populations, reveals that gut microbiota may influence breast cancer risk differently across populations, providing potential directions for developing targeted prevention and treatment methods.


Sujet(s)
Tumeurs du sein , Microbiome gastro-intestinal , Étude d'association pangénomique , Analyse de randomisation mendélienne , Femelle , Humains , Tumeurs du sein/génétique , Tumeurs du sein/microbiologie , Tumeurs du sein/épidémiologie , Peuples d'Asie de l'Est/génétique , Microbiome gastro-intestinal/génétique , Polymorphisme de nucléotide simple , Facteurs de risque , /génétique
11.
Sci Rep ; 14(1): 18306, 2024 08 07.
Article de Anglais | MEDLINE | ID: mdl-39112529

RÉSUMÉ

Gut bacteria might play an important role in male reproductive disorders, such as male infertility and sperm abnormalities; however, their causal role is unclear. Herein, Mendelian randomization (MR)-Egger, weighted median, inverse variance weighting, Simple mode, and Weighted mode were used to test the causal relationship between gut microbes and male reproductive diseases. The MR results were validated using various metrics. The MR results were also consolidated using reverse causality speculation, conducted using two-way MR analysis and Steiger filtering. Biological function was analysed using enrichment analyses. The results suggested that eight intestinal microflorae were causally associated with male infertility. The Eubacterium oxidoreducens group was associated with an increased risk of male infertility, while the family Bacteroidaceae was negatively associated with male reproductive diseases. Eight intestinal microflorae were causally associated with abnormal spermatozoa. The family Streptococcaceae was associated with a high risk of abnormal spermatozoa, whereas the family Porphyromonadaceae was associated with a low risk of abnormal spermatozoa. No pleiotropy was observed, this study identified a high correlation between the gut flora and the likelihood of male reproductive diseases. Future research will attempt to advance microbial-focused treatments for such diseases.


Sujet(s)
Microbiome gastro-intestinal , Infertilité masculine , Analyse de randomisation mendélienne , Mâle , Humains , Microbiome gastro-intestinal/génétique , Infertilité masculine/microbiologie , Infertilité masculine/génétique , Spermatozoïdes/microbiologie
12.
Sci Rep ; 14(1): 18405, 2024 08 08.
Article de Anglais | MEDLINE | ID: mdl-39117770

RÉSUMÉ

Evidence from previous studies have demonstrated that gut microbiota are closely associated with occurrence of interstitial cystitis/bladder pain syndrome (IC/BPS), yet the causal link between the two is not well known. In this study, we performed a two-sample Mendelian randomization (MR) analysis to determine the possible causal association between gut microbiota with IC/BPS. Gut microbiota summary level data were derived from the genome-wide association study (GWAS) conducted by MiBioGen and the IC/BPS GWAS summary level data were obtained from the GWAS Catalog. Next, we performed an MR study to investigate the causal link between gut microbiota and IC/BPS. The primary method for causal analysis was the inverse variance weighted (IVW), and the MR results were validated through multiple sensitivity analyses. A positive association was found between IC/BPS and eight gut microbial taxa, including genus Bacteroides, genus Haemophilus, genus Veillonella, genus Coprococcus1, genus Butyricimonas, family Bacteroidaceae, family Christensenellaceae, and order Lactobacillales. Sensitivity analysis revealed lack of significant pleiotropy or heterogeneity in the obtained results. This MR analysis reveals that a causal association exists between some gut microbiota with IC/BPS. This finding may is expected to guide future research and development of IC/BPS preventions and treatments based on the bladder-gut axis. However, given the clinical complexity and diagnostic challenges of IC/BPS, along with the limitations of using large-scale GWAS summary data for analysis, our MR results require further validation through additional research.


Sujet(s)
Cystite interstitielle , Microbiome gastro-intestinal , Étude d'association pangénomique , Analyse de randomisation mendélienne , Cystite interstitielle/microbiologie , Cystite interstitielle/génétique , Humains , Microbiome gastro-intestinal/génétique , Polymorphisme de nucléotide simple
13.
Nat Commun ; 15(1): 6924, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138199

RÉSUMÉ

Plasmids are small DNA molecules that enable bacteria to share beneficial traits, influencing microbial communities. However, their role within the human gut microbiome remains largely unknown. In this study, we investigate the gut microbiomes of 34 mother-child cohorts, employing a plasmid analysis workflow to understand the impact of plasmids on the gut microbiome. We create a plasmid phylogenetic tree, devise a method for assigning plasmid hosts, and examine potential plasmid transfer networks. Our research discovers a wide variety of previously unidentified plasmid sequences, indicating that current databases do not fully represent the gut plasmidome. Interestingly, infants display greater plasmid diversity compared to mothers and other healthy adults. We find that Bacteroidota, a major bacterial phylum, serves as the primary host for gut plasmids and plays a dominant role in gut plasmid transfer events. Additionally, plasmids broaden the genetic capabilities of bacteria, with their influence on bacterial function becoming more apparent as children's gut microbiomes develop. This study sheds light on the role of plasmids in the infant gut microbiome, making a significant contribution to our understanding of plasmid biology.


Sujet(s)
Microbiome gastro-intestinal , Phylogenèse , Plasmides , Plasmides/génétique , Humains , Microbiome gastro-intestinal/génétique , Nourrisson , Femelle , Adulte , Bactéries/génétique , Bactéries/classification , Mâle , Fèces/microbiologie , Bacteroidetes/génétique , Nouveau-né
14.
Sci Adv ; 10(33): eadn3316, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39141729

RÉSUMÉ

Genetic variations are instrumental for unraveling phage evolution and deciphering their functional implications. Here, we explore the underlying fine-scale genetic variations in the gut phageome, especially structural variations (SVs). By using virome-enriched long-read metagenomic sequencing across 91 individuals, we identified a total of 14,438 nonredundant phage SVs and revealed their prevalence within the human gut phageome. These SVs are mainly enriched in genes involved in recombination, DNA methylation, and antibiotic resistance. Notably, a substantial fraction of phage SV sequences share close homology with bacterial fragments, with most SVs enriched for horizontal gene transfer (HGT) mechanism. Further investigations showed that these SV sequences were genetic exchanged between specific phage-bacteria pairs, particularly between phages and their respective bacterial hosts. Temperate phages exhibit a higher frequency of genetic exchange with bacterial chromosomes and then virulent phages. Collectively, our findings provide insights into the genetic landscape of the human gut phageome.


Sujet(s)
Bactéries , Bactériophages , Microbiome gastro-intestinal , Transfert horizontal de gène , Bactériophages/génétique , Humains , Microbiome gastro-intestinal/génétique , Bactéries/virologie , Bactéries/génétique , Métagénomique/méthodes , Variation génétique , Virome/génétique , Génome viral , Séquençage nucléotidique à haut débit
15.
Nat Commun ; 15(1): 6980, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39143045

RÉSUMÉ

Antibiotics may alter the gut microbiome, and this is one of the mechanisms by which antimicrobial resistance may be promoted. Suboptimal antimicrobial stewardship in Asia has been linked to antimicrobial resistance. We aim to examine the relationship between oral antibiotic use and composition and antimicrobial resistance in the gut microbiome in 1093 Bangladeshi infants. We leverage a trial of 8-month-old infants in rural Bangladesh: 61% of children were cumulatively exposed to antibiotics (most commonly cephalosporins and macrolides) over the 12-month study period, including 47% in the first 3 months of the study, usually for fever or respiratory infection. 16S rRNA amplicon sequencing in 11-month-old infants reveals that alpha diversity of the intestinal microbiome is reduced in children who received antibiotics within the previous 7 days; these samples also exhibit enrichment for Enterococcus and Escherichia/Shigella genera. No effect is seen in children who received antibiotics earlier. Using shotgun metagenomics, overall abundance of antimicrobial resistance genes declines over time. Enrichment for an Enterococcus-related antimicrobial resistance gene is observed in children receiving antibiotics within the previous 7 days, but not earlier. Presence of antimicrobial resistance genes is correlated to microbiome composition. In Bangladeshi children, community use of antibiotics transiently reprofiles the gut microbiome.


Sujet(s)
Antibactériens , Microbiome gastro-intestinal , ARN ribosomique 16S , Humains , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Microbiome gastro-intestinal/génétique , Bangladesh/épidémiologie , Nourrisson , Antibactériens/administration et posologie , Antibactériens/pharmacologie , ARN ribosomique 16S/génétique , Mâle , Femelle , Administration par voie orale , Résistance bactérienne aux médicaments/génétique , Fèces/microbiologie , Métagénomique/méthodes , Bactéries/génétique , Bactéries/effets des médicaments et des substances chimiques , Bactéries/classification , Bactéries/isolement et purification , Céphalosporines/administration et posologie , Céphalosporines/pharmacologie , Céphalosporines/usage thérapeutique , Enterococcus/effets des médicaments et des substances chimiques , Enterococcus/génétique , Enterococcus/isolement et purification , Gestion responsable des antimicrobiens
16.
Sci Rep ; 14(1): 18906, 2024 08 14.
Article de Anglais | MEDLINE | ID: mdl-39143178

RÉSUMÉ

The human gut microbiome composition has been linked to Parkinson's disease (PD). However, knowledge of the gut microbiota on the genome level is still limited. Here we performed deep metagenomic sequencing and binning to build metagenome-assembled genomes (MAGs) from 136 human fecal microbiomes (68 PD samples and 68 control samples). We constructed 952 non-redundant high-quality MAGs and compared them between PD and control groups. Among these MAGs, there were 22 different genomes of Collinsella and Prevotella, indicating high variability of those genera in the human gut environment. Microdiversity analysis indicated that Ruminococcus bromii was statistically significantly (p < 0.002) more diverse on the strain level in the control samples compared to the PD samples. In addition, by clustering all genes and performing presence-absence analysis between groups, we identified several control-specific (p < 0.05) related genes, such as speF and Fe-S oxidoreductase. We also report detailed annotation of MAGs, including Clusters of Orthologous Genes (COG), Cas operon type, antiviral gene, prophage, and secondary metabolites biosynthetic gene clusters, which can be useful for providing a reference for future studies.


Sujet(s)
Fèces , Microbiome gastro-intestinal , Métagénome , Maladie de Parkinson , Maladie de Parkinson/génétique , Maladie de Parkinson/microbiologie , Humains , Fèces/microbiologie , Microbiome gastro-intestinal/génétique , Métagénomique/méthodes , Génome bactérien , Mâle , Sujet âgé , Femelle , Génome microbien , Adulte d'âge moyen , Séquençage nucléotidique à haut débit
17.
Sci Rep ; 14(1): 18912, 2024 08 14.
Article de Anglais | MEDLINE | ID: mdl-39143364

RÉSUMÉ

There is a significant focus on the role of the host microbiome in different outcomes of human parasitic diseases, including cystic echinococcosis (CE). This study was conducted to identify the intestinal microbiome of patients with CE at different stages of hydatid cyst compared to healthy individuals. Stool samples from CE patients as well as healthy individuals were collected. The samples were divided into three groups representing various stages of hepatic hydatid cyst: active (CE1 and CE2), transitional (CE3), and inactive (CE4 and CE5). One family member from each group was selected to serve as a control. The gut microbiome of patients with different stages of hydatid cysts was investigated using metagenomic next-generation amplicon sequencing of the V3-V4 region of the 16S rRNA gene. In this study, we identified 4862 Operational Taxonomic Units from three stages of hydatid cysts in CE patients and healthy individuals with a combined frequency of 2,955,291. The most abundant genera observed in all the subjects were Blautia, Agathobacter, Faecalibacterium, Bacteroides, Bifidobacterium, and Prevotella. The highest microbial frequency was related to inactive forms of CE, and the lowest frequency was observed in the group with active forms. However, the lowest OTU diversity was found in patients with inactive cysts compared with those with active and transitional cyst stages. The genus Agatobacter had the highest OTU frequency. Pseudomonas, Gemella, and Ligilactobacillus showed significant differences among the patients with different stages of hydatid cysts. Additionally, Anaerostipes and Candidatus showed significantly different reads in CE patients compared to healthy individuals. Our findings indicate that several bacterial genera can play a role in the fate of hydatid cysts in patients at different stages of the disease.


Sujet(s)
Échinococcose hépatique , Microbiome gastro-intestinal , ARN ribosomique 16S , Humains , Microbiome gastro-intestinal/génétique , Échinococcose hépatique/microbiologie , Échinococcose hépatique/parasitologie , Mâle , Femelle , ARN ribosomique 16S/génétique , Adulte , Adulte d'âge moyen , Fèces/microbiologie , Fèces/parasitologie , Bactéries/classification , Bactéries/génétique , Bactéries/isolement et purification , Biodiversité , Séquençage nucléotidique à haut débit
18.
J Hazard Mater ; 477: 135387, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39094311

RÉSUMÉ

Urban parks play a significant role in urban ecosystems and are strongly associated with human health. Nevertheless, the biological contamination of urban parks - opportunistic pathogens and antibiotic resistance genes (ARGs) - has been poorly reported. Here, metagenomic and 16 S rRNA sequencing methods were used to study the distribution and assembly of opportunistic pathogens and ARGs in soil and water from nine parks in Lanzhou city, and further compared them with local human gut microbiomes to investigate the potential transmission risk. Our results revealed that the most important type of drug resistance in urban parks was multidrug resistance, with various resistance mechanisms. Approximately half of ARGs were shared between human gut and park environment, and it was noteworthy that cross-species transmission might exist among some high-risk ARGs, such as mepA and mdtE, with a significant enrichment in human gut. Metagenomic binning uncovered several bacterial genomes carrying adjacent ARGs, MGEs, and virulence genes, indicating a possibility that these genes may jointly transfer among different environments, particularly from park environment to human. Our results provided a reference point for the management of environmental pollutants in urban parks.


Sujet(s)
Métagénomique , Humains , Parcs de loisirs , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Microbiome gastro-intestinal/génétique , ARN ribosomique 16S/génétique , Chine , Bactéries/génétique , Bactéries/effets des médicaments et des substances chimiques , Bactéries/classification , Microbiologie du sol , Villes , Résistance bactérienne aux médicaments/génétique , Microbiologie de l'eau , Gènes bactériens
19.
BMC Gastroenterol ; 24(1): 271, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39160466

RÉSUMÉ

BACKGROUND: Constipation is one of the most common gastrointestinal disorders afflicting the population, with recent observational studies implicating dysfunction of the gut microbiota in constipation. Despite observational studies indicating a relationship, a clear causality remains unclear. This study aims to use two-sample Mendelian randomization (MR) to establish a clearer causal relationship between the two. METHODS: A two-sample Mendelian randomization (MR) study was performed using the gut microbiota summary Genome-Wide Association Studies (GWAS) statistics from MiBioGen consortium (n = 13,266) and constipation GWAS summary statistics from the IEU OpenGWAS database. The causality between gut microbiota and constipation is primarily analyzed using the inverse-variance weighted (IVW) method and reinforced by an additional four methods, including MR-Egger, Weighted Median, Simple Mode, and Weighted Mode. Finally, funnel plot, heterogeneity test, horizontal pleiotropy test, and leave-one-out test were used to evaluate the reliability of MR results. RESULTS: IVW estimates suggested that the bacterial species Anaerotruncus, Butyricimonas, and Hungatella were causally associated with constipation. The odds ratio (OR) values of Anaerotruncus, Butyricimonas, and Hungatella were 1.08 (95% CI = 1.02-1.13; P = 0.007), 1.07 (95% CI = 1.01-1.13; P = 0.015), 1.03 (95% CI = 1.00-1.06; P = 0.037) respectively. Meanwhile, Ruminiclostridium 9 and Intestinibacter have been shown to be associated with a reduced risk of constipation. The OR of Ruminiclostridium 9 = 0.75(95% CI = 0.73-0.78, P < 0.001 and Intestinibacter of OR = 0.89 (95% CI = 0.86-0.93, P < 0.001). Furthermore, validation by funnel plot, heterogeneity test, and horizontal pleiotropy test showed that MR results were reliable. CONCLUSION: This is the first Mendelian randomization study to explore the causalities between specific gut microbiota taxa and constipation, and as such may be useful in providing insights into the unclear pathology of constipation which can in turn aid in the search for prevention and treatment.


Sujet(s)
Constipation , Microbiome gastro-intestinal , Étude d'association pangénomique , Analyse de randomisation mendélienne , Constipation/microbiologie , Humains , Microbiome gastro-intestinal/génétique , Causalité
20.
Ren Fail ; 46(2): 2385065, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39090986

RÉSUMÉ

Over recent years, the prevalence of diabetes has been on the rise, paralleling improvements in living standards. Diabetic nephropathy (DN), a prevalent complication of diabetes, has also exhibited a growing incidence. While some clinical studies and reviews have hinted at a link between diabetic nephropathy and gut microbiota (GM), the nature of this connection, specifically its causative nature, remains uncertain. Investigating the causal relationship between diabetic nephropathy and gut microbiota holds the promise of aiding in disease screening and identifying novel biomarkers. In this study, we employed a two-sample Mendelian randomization analysis. Our dataset encompassed 4,111 DN patients from the GWAS database, juxtaposed with 308,539 members forming a control group. The aim was to pinpoint specific categories within the vast spectrum of the 211 known gut microbiota types that may have a direct causal relationship with diabetic nephropathy. Rigorous measures, including extensive heterogeneity and sensitivity analyses, were implemented to mitigate the influence of confounding variables on our experimental outcomes. Ultimately, our comprehensive analysis revealed 15 distinct categories of gut microbiota that exhibit a causal association with diabetic nephropathy. In summary, the phyla Bacteroidota and Verrucomicrobiae, the families Peptostreptococcaceae and Veillonellaceae, the genus Akkermansia, and the species Catenibacterium, Lachnoclostridium, Parasutterella, along with the orders Bacteroidales and Verrucomicrobiales, and the class Bacteroidetes were identified as correlates of increased risk for DN. Conversely, the family Victivallaceae, the species Eubacterium coprostanoligenes, and the Clostridium sensu stricto 1 group were found to be associated with a protective effect against the development of DN.These findings not only provide valuable insights but also open up novel avenues for clinical research, offering fresh directions for potential treatments.


Sujet(s)
Néphropathies diabétiques , Microbiome gastro-intestinal , Séquençage du génome entier , Humains , Néphropathies diabétiques/microbiologie , Microbiome gastro-intestinal/génétique , Analyse de randomisation mendélienne , Étude d'association pangénomique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE