Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 323
Filtrar
1.
PLoS One ; 17(2): e0263109, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35202432

RESUMO

BACKGROUND: Autism spectrum disorder (ASD) is a neurological and developmental condition that begins in infancy or earlier and lasts through the individual's lifetime. The aetiology and mechanisms of ASD are not yet fully understood, and current treatment comprises mainly education and rehabilitation, without significant improvement in the core symptoms. Recent studies suggest that microbiota change in children with ASD after the ingestion of probiotics may improve the balance of microbiota and thus ASD symptoms. OBJECTIVE: The objectives of this study are to evaluate the efficacy of probiotics on the symptoms of children with ASD and the possible mechanisms involved. METHODS: This is a prospective controlled trial. A total of 160 children with ASD will be stratified and allocated to placebo and probiotics groups randomised according to the severity of their ASD symptoms. The probiotics group will be given probiotics supplements orally twice a day for 3 months and the control group will be given a placebo at the same amount, in addition to the baseline therapy of education and rehabilitation. All the children will be evaluated systematically by using different scales, questionnaires before, during, and after 3 months' treatment, as well as 3 months after discontinuation. The potential impact of probiotics on immunity and inflammation, metabolism, and metagenome will also be investigated. DISCUSSION: Our previous study showed that the abundance of intestinal flora was greatly different in children with ASD, and that Bifidobacterium was associated with the severity of ASD. In the present study, we will investigate the impact of probiotics supplementation on the symptoms of Children with ASD, with the purpose of evaluating the possible therapeutic effects of additives on ASD and of providing a reference for clinical treatment. The results will help to disclose as yet unknown relationship between probiotics and ASD. TRIAL REGISTRATION: This study has been registered with Chinese Clinical Trial Registry (ChiCTR-2000037941).


Assuntos
Transtorno do Espectro Autista/tratamento farmacológico , Microbioma Gastrointestinal/efeitos dos fármacos , Metagenoma/genética , Probióticos/administração & dosagem , Transtorno do Espectro Autista/microbiologia , Transtorno do Espectro Autista/patologia , Bifidobacterium/genética , Bifidobacterium/patogenicidade , Criança , Pré-Escolar , Feminino , Microbioma Gastrointestinal/genética , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Humanos , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/imunologia , Inflamação/microbiologia , Masculino , Metagenoma/efeitos dos fármacos , Placebos , Probióticos/efeitos adversos
2.
Nature ; 600(7887): 110-115, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34819672

RESUMO

The human microbiome encodes a large repertoire of biochemical enzymes and pathways, most of which remain uncharacterized. Here, using a metagenomics-based search strategy, we discovered that bacterial members of the human gut and oral microbiome encode enzymes that selectively phosphorylate a clinically used antidiabetic drug, acarbose1,2, resulting in its inactivation. Acarbose is an inhibitor of both human and bacterial α-glucosidases3, limiting the ability of the target organism to metabolize complex carbohydrates. Using biochemical assays, X-ray crystallography and metagenomic analyses, we show that microbiome-derived acarbose kinases are specific for acarbose, provide their harbouring organism with a protective advantage against the activity of acarbose, and are widespread in the microbiomes of western and non-western human populations. These results provide an example of widespread microbiome resistance to a non-antibiotic drug, and suggest that acarbose resistance has disseminated in the human microbiome as a defensive strategy against a potential endogenous producer of a closely related molecule.


Assuntos
Acarbose/farmacologia , Farmacorresistência Bacteriana/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Inativação Metabólica , Metagenoma/genética , Boca/microbiologia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Acarbose/metabolismo , Amilases/metabolismo , Animais , Humanos , Hipoglicemiantes/metabolismo , Metagenoma/efeitos dos fármacos , Modelos Moleculares , Boca/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/química , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo
3.
Nutrients ; 13(11)2021 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-34836145

RESUMO

The gut microbiota consists of a community of microorganisms that inhabit the large intestine. These microbes play important roles in maintaining gut barrier integrity, inflammation, lipid and carbohydrate metabolism, immunity, and protection against pathogens. However, recent studies have shown that dysfunction in the gut microbiota composition can lead to the development of several diseases. Urolithin A has recently been approved as a functional food ingredient. In this study, we examined the potentials of urolithin A (Uro-A) and B (Uro-B) in improving metabolic functions and their impact on gut microbiota composition under a metabolically unchallenged state in normal rats. Male Wistar rats (n = 18) were randomly segregated into three groups, with Group 1 serving as the control group. Groups 2 and 3 were administered with 2.5 mg/kg Uro-A and Uro-B, respectively, for four weeks. Our results showed that both Uro-A and B improved liver and kidney functions without affecting body weight. Metagenomic analysis revealed that both Uro-A and B induced the growth of Akkermansia. However, Uro-A decreased species diversity and microbial richness and negatively impacted the composition of pathogenic microbes in normal rats. Taken together, this study showed the differential impacts of Uro-A and B on the gut microbiota composition in normal rats and would thus serve as a guide in the choice of these metabolites as a functional food ingredient or prebiotic.


Assuntos
Cumarínicos/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Metagenoma/efeitos dos fármacos , Animais , Ingredientes de Alimentos , Alimento Funcional , Masculino , Ratos , Ratos Wistar
4.
EBioMedicine ; 71: 103566, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34492446

RESUMO

BACKGROUND: Shortening antibiotic-treatment durations is a key recommendation of antibiotic-stewardship programmes, yet it is based on weak evidence. We investigated whether halving antibiotic courses would reduce antibiotic-resistance genes (ARG) in the intestinal microbiomes of patients treated for gram-negative bacteraemia. METHODS: This nested prospective cohort study included adult patients hospitalized at Geneva University Hospitals (Switzerland) participating in the PIRATE randomized trial assessing non-inferiority of shorter antibiotic courses (7 versus 14 days) for gram-negative bacteraemia ('cases') and, simultaneously, hospitalized patients with similar demography and comorbidity yet no antibiotic therapy ('controls'). Stool was collected from case and control patients on days 7, 14, 30 and 90 after antibiotic initiation (day 1) and days 7 and 14 after admission, respectively, and analysed by whole-metagenome shotgun sequencing. The primary outcome was ARG abundance at day 30; secondary outcomes included microbiota-species composition and clustering over time. FINDINGS: Forty-five patients and 11 controls were included and evaluable; ARG analyses were conducted on the 29 per-protocol patients receiving 7 (±2) days or 14 (±3) days of antibiotic therapy. At day 30, ARGs were not detected at similar abundance in patients receiving 7 and 14 days (median counts/million [mCPM]: 96 versus [vs] 71; p=.38). By day 30, total ARG content between both groups was not significantly different from that of controls at D7 (362 and 370 mCPM vs 314 mCPM, p=.24 and 0.19). There were no significant differences amongst antibiotic-treated patients at any timepoint in bacterial diversity or clustering, but Shannon species diversity was significantly reduced compared to controls through day 14 (median 3.12 and 3.24 in the 7-day and 14-day groups vs 3.61 [controls]; p=.04 and 0.012). Patients treated for 14 days had reduced faecal phage content during and after therapy compared to other patient groups. INTERPRETATION: Reducing antibiotic durations by half did not result in decreased abundance of ARGs in patients treated for gram-negative bacteraemia, nor did it improve microbiota species diversity. FUNDING: The study was funded by the University of Geneva's Louis-Jeantet Foundation (grant no. S04_12) and the Swiss National Science Foundation (NRP Smarter Healthcare, grant no. 407,440_167359).


Assuntos
Antibacterianos/administração & dosagem , Farmacorresistência Bacteriana/genética , Microbioma Gastrointestinal/efeitos dos fármacos , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Bacteriemia/tratamento farmacológico , Esquema de Medicação , Microbioma Gastrointestinal/genética , Genes Bacterianos , Infecções por Bactérias Gram-Negativas/tratamento farmacológico , Humanos , Metagenoma/efeitos dos fármacos , Metagenoma/genética
5.
Nat Microbiol ; 6(8): 1043-1054, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34226711

RESUMO

Antimicrobial resistance poses a substantial threat to human health. The gut microbiome is considered a reservoir for potential spread of resistance genes from commensals to pathogens, termed the gut resistome. The impact of probiotics, commonly consumed by many in health or in conjunction with the administration of antibiotics, on the gut resistome is elusive. Reanalysis of gut metagenomes from healthy antibiotics-naïve humans supplemented with an 11-probiotic-strain preparation, allowing direct assessment of the gut resistome in situ along the gastrointestinal (GI) tract, demonstrated that probiotics reduce the number of antibiotic resistance genes exclusively in the gut of colonization-permissive individuals. In mice and in a separate cohort of humans, a course of antibiotics resulted in expansion of the lower GI tract resistome, which was mitigated by autologous faecal microbiome transplantation or during spontaneous recovery. In contrast, probiotics further exacerbated resistome expansion in the GI mucosa by supporting the bloom of strains carrying vancomycin resistance genes but not resistance genes encoded by the probiotic strains. Importantly, the aforementioned effects were not reflected in stool samples, highlighting the importance of direct sampling to analyse the effect of probiotics and antibiotics on the gut resistome. Analysing antibiotic resistance gene content in additional published clinical trials with probiotics further highlighted the importance of person-specific metagenomics-based profiling of the gut resistome using direct sampling. Collectively, these findings suggest opposing person-specific and antibiotic-dependent effects of probiotics on the resistome, whose contribution to the spread of antimicrobial resistance genes along the human GI tract merit further studies.


Assuntos
Antibacterianos/administração & dosagem , Bactérias/efeitos dos fármacos , Bactérias/genética , Proteínas de Bactérias/genética , Farmacorresistência Bacteriana , Microbioma Gastrointestinal/efeitos dos fármacos , Probióticos/administração & dosagem , Adulto , Bactérias/classificação , Bactérias/isolamento & purificação , Proteínas de Bactérias/metabolismo , Estudos de Coortes , Transplante de Microbiota Fecal , Fezes/microbiologia , Feminino , Trato Gastrointestinal/microbiologia , Humanos , Masculino , Metagenoma/efeitos dos fármacos , Pessoa de Meia-Idade , Adulto Jovem
6.
Front Endocrinol (Lausanne) ; 12: 626359, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33815284

RESUMO

Effects of metformin, the first-line drug for type 2 diabetes therapy, on gut microbiome composition in type 2 diabetes have been described in various studies both in human subjects and animals. However, the details of the molecular mechanisms of metformin action have not been fully understood. Moreover, there is a significant lack of information on how metformin affects gut microbiome composition in female mouse models, depending on sex and metabolic status in well controlled experimental setting. Our study aimed to examine metformin-induced alterations in gut microbiome diversity, composition, and functional implications of high-fat diet-induced type 2 diabetes mouse model, using, for the first time in mice study, the shotgun metagenomic sequencing that allows estimation of microorganisms at species level. We also employed a randomized block, factorial study design, and including 24 experimental units allocated to 8 treatment groups to systematically evaluate the effect of sex and metabolic status on metformin interaction with microbiome. We used DNA obtained from fecal samples representing gut microbiome before and after ten weeks-long metformin treatment. We identified 100 metformin-related differentially abundant species in high-fat diet-fed mice before and after the treatment, with most of the species relative abundances increased. In contrast, no significant changes were observed in control diet-fed mice. Functional analysis targeted to carbohydrate, lipid, and amino acid metabolism pathways revealed 14 significantly altered hierarchies. We also observed sex-specific differences in response to metformin treatment. Males experienced more pronounced changes in metabolic markers, while in females the extent of changes in gut microbiome representatives was more marked, indicated by 53 differentially abundant species with more remarkable Log fold changes compared to the combined-sex analysis. The same pattern manifested regarding the functional analysis, where we discovered 5 significantly affected hierarchies in female groups but not in males. Our results suggest that both sexes of animals should be included in future studies focusing on metformin effects on the gut microbiome.


Assuntos
Diabetes Mellitus Tipo 2/microbiologia , Dieta Hiperlipídica , Microbioma Gastrointestinal/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Metformina/farmacologia , Animais , Modelos Animais de Doenças , Feminino , Masculino , Metagenoma/efeitos dos fármacos , Camundongos
7.
Gut Microbes ; 13(1): 1-19, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33779498

RESUMO

Oral antibiotics are commonly prescribed to non-hospitalized adults. However, antibiotic-induced changes in the human gut microbiome are often investigated in cohorts with preexisting health conditions and/or concomitant medication, leaving the effects of antibiotics not completely understood. We used a combination of omic approaches to comprehensively assess the effects of antibiotics on the gut microbiota and particularly the gut resistome of a small cohort of healthy adults. We observed that 3 to 19 species per individual proliferated during antibiotic treatment and Gram-negative species expanded significantly in relative abundance. While the overall relative abundance of antibiotic resistance gene homologs did not significantly change, antibiotic-specific gene homologs with presumed resistance toward the administered antibiotics were common in proliferating species and significantly increased in relative abundance. Virome sequencing and plasmid analysis showed an expansion of antibiotic-specific resistance gene homologs even 3 months after antibiotic administration, while paired-end read analysis suggested their dissemination among different species. These results suggest that antibiotic treatment can lead to a persistent expansion of antibiotic resistance genes in the human gut microbiota and provide further data in support of good antibiotic stewardship.Abbreviation: ARG - Antibiotic resistance gene homolog; AsRG - Antibiotic-specific resistance gene homolog; AZY - Azithromycin; CFX - Cefuroxime; CIP - Ciprofloxacin; DOX - Doxycycline; FDR - False discovery rate; GRiD - Growth rate index value; HGT - Horizontal gene transfer; NMDS - Non-metric multidimensional scaling; qPCR - Quantitative polymerase chain reaction; RPM - Reads per million mapped reads; TA - Transcriptional activity; TE - Transposable element; TPM - Transcripts per million mapped reads.


Assuntos
Antibacterianos/uso terapêutico , Resistência Microbiana a Medicamentos , Fezes/microbiologia , Fezes/virologia , Microbioma Gastrointestinal/efeitos dos fármacos , Microbiota/efeitos dos fármacos , Adolescente , Adulto , Idoso , Bactérias/virologia , Bacteriófagos/efeitos dos fármacos , Guerra Biológica , Estudos de Coortes , Transferência Genética Horizontal/efeitos dos fármacos , Humanos , Metagenoma/efeitos dos fármacos , Pessoa de Meia-Idade , Plasmídeos/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Viroma/efeitos dos fármacos , Adulto Jovem
8.
Microbiome ; 9(1): 32, 2021 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-33517907

RESUMO

BACKGROUND: While indoor microbiomes impact our health and well-being, much remains unknown about taxonomic and functional transitions that occur in human-derived microbial communities once they are transferred away from human hosts. Toothbrushes are a model to investigate the potential response of oral-derived microbiota to conditions of the built environment. Here, we characterize metagenomes of toothbrushes from 34 subjects to define the toothbrush microbiome and resistome and possible influential factors. RESULTS: Toothbrush microbiomes often comprised a dominant subset of human oral taxa and less abundant or site-specific environmental strains. Although toothbrushes contained lower taxonomic diversity than oral-associated counterparts (determined by comparison with the Human Microbiome Project), they had relatively broader antimicrobial resistance gene (ARG) profiles. Toothbrush resistomes were enriched with a variety of ARGs, notably those conferring multidrug efflux and putative resistance to triclosan, which were primarily attributable to versatile environmental taxa. Toothbrush microbial communities and resistomes correlated with a variety of factors linked to personal health, dental hygiene, and bathroom features. CONCLUSIONS: Selective pressures in the built environment may shape the dynamic mixture of human (primarily oral-associated) and environmental microbiota that encounter each other on toothbrushes. Harboring a microbial diversity and resistome distinct from human-associated counterparts suggests toothbrushes could potentially serve as a reservoir that may enable the transfer of ARGs. Video abstract.


Assuntos
Ambiente Construído , Microbiota , Boca/microbiologia , Escovação Dentária , Adolescente , Adulto , Idoso , Resistência Microbiana a Medicamentos/efeitos dos fármacos , Resistência Microbiana a Medicamentos/genética , Humanos , Metagenoma/efeitos dos fármacos , Metagenoma/genética , Microbiota/efeitos dos fármacos , Microbiota/genética , Pessoa de Meia-Idade , Boca/efeitos dos fármacos , Triclosan/farmacologia , Adulto Jovem
9.
Sci Rep ; 11(1): 3753, 2021 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-33580146

RESUMO

In the fight to limit the global spread of antibiotic resistance, the assembly of environmental metagenomes has the potential to provide rich contextual information (e.g., taxonomic hosts, carriage on mobile genetic elements) about antibiotic resistance genes (ARG) in the environment. However, computational challenges associated with assembly can impact the accuracy of downstream analyses. This work critically evaluates the impact of assembly leveraging short reads, nanopore MinION long-reads, and a combination of the two (hybrid) on ARG contextualization for ten environmental metagenomes using seven prominent assemblers (IDBA-UD, MEGAHIT, Canu, Flye, Opera-MS, metaSpades and HybridSpades). While short-read and hybrid assemblies produced similar patterns of ARG contextualization, raw or assembled long nanopore reads produced distinct patterns. Based on an in-silico spike-in experiment using real and simulated reads, we show that low to intermediate coverage species are more likely to be incorporated into chimeric contigs across all assemblers and sequencing technologies, while more abundant species produce assemblies with a greater frequency of inversions and insertion/deletions (indels). In sum, our analyses support hybrid assembly as a valuable technique for boosting the reliability and accuracy of assembly-based analyses of ARGs and neighboring genes at environmentally-relevant coverages, provided that sufficient short-read sequencing depth is achieved.


Assuntos
Resistência Microbiana a Medicamentos/genética , Metagenoma/genética , Águas Residuárias/análise , Antibacterianos/farmacologia , Biologia Computacional/métodos , Microbiologia Ambiental , Monitoramento Ambiental/métodos , Genoma Bacteriano/efeitos dos fármacos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Metagenoma/efeitos dos fármacos , Metagenômica/métodos , Reprodutibilidade dos Testes , Análise de Sequência de DNA/métodos
10.
PLoS One ; 16(1): e0244876, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33411719

RESUMO

Characterizing the gut microbiota in terms of their capacity to interfere with drug metabolism is necessary to achieve drug efficacy and safety. Although examples of drug-microbiome interactions are well-documented, little has been reported about a computational pipeline for systematically identifying and characterizing bacterial enzymes that process particular classes of drugs. The goal of our study is to develop a computational approach that compiles drugs whose metabolism may be influenced by a particular class of microbial enzymes and that quantifies the variability in the collective level of those enzymes among individuals. The present paper describes this approach, with microbial ß-glucuronidases as an example, which break down drug-glucuronide conjugates and reactivate the drugs or their metabolites. We identified 100 medications that may be metabolized by ß-glucuronidases from the gut microbiome. These medications included morphine, estrogen, ibuprofen, midazolam, and their structural analogues. The analysis of metagenomic data available through the Sequence Read Archive (SRA) showed that the level of ß-glucuronidase in the gut metagenomes was higher in males than in females, which provides a potential explanation for the sex-based differences in efficacy and toxicity for several drugs, reported in previous studies. Our analysis also showed that infant gut metagenomes at birth and 12 months of age have higher levels of ß-glucuronidase than the metagenomes of their mothers and the implication of this observed variability was discussed in the context of breastfeeding as well as infant hyperbilirubinemia. Overall, despite important limitations discussed in this paper, our analysis provided useful insights on the role of the human gut metagenome in the variability in drug response among individuals. Importantly, this approach exploits drug and metagenome data available in public databases as well as open-source cheminformatics and bioinformatics tools to predict drug-metagenome interactions.


Assuntos
Previsões/métodos , Microbioma Gastrointestinal/efeitos dos fármacos , Metagenômica/métodos , Adulto , Bactérias/genética , Biologia Computacional/métodos , Gerenciamento de Dados , Feminino , Microbioma Gastrointestinal/genética , Glucuronidase/genética , Glucuronidase/metabolismo , Humanos , Recém-Nascido , Masculino , Metagenoma/efeitos dos fármacos , Metagenoma/genética , Microbiota/efeitos dos fármacos , Microbiota/genética , Mães
11.
Gut ; 70(6): 1162-1173, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32998876

RESUMO

OBJECTIVE: Altered bacterial composition is associated with disease progression in cirrhosis but the role of virome, especially phages, is unclear. DESIGN: Cross-sectional and pre/post rifaximin cohorts were enrolled. Cross-sectional: controls and cirrhotic outpatients (compensated, on lactulose (Cirr-L), on rifaximin (Cirr-LR)) were included and followed for 90-day hospitalisations. Pre/post: compensated cirrhotics underwent stool collection pre/post 8 weeks of rifaximin. Stool metagenomics for bacteria and phages and their correlation networks were analysed in controls versus cirrhosis, within cirrhotics, hospitalised/not and pre/post rifaximin. RESULTS: Cross-sectional: 40 controls and 163 cirrhotics (63 compensated, 43 Cirr-L, 57 Cirr-LR) were enrolled. Cirr-L/LR groups were similar on model for end-stage liver disease (MELD) score but Cirr-L developed greater hospitalisations versus Cirr-LR (56% vs 30%, p=0.008). Bacterial alpha/beta diversity worsened from controls through Cirr-LR. While phage alpha diversity was similar, beta diversity was different between groups. Autochthonous bacteria linked negatively, pathobionts linked positively with MELD but only modest phage-MELD correlations were seen. Phage-bacterial correlation network complexity was highest in controls, lowest in Cirr-L and increased in Cirr-LR. Microviridae and Faecalibacterium phages were linked with autochthonous bacteria in Cirr-LR, but not Cirr-L hospitalised patients had greater pathobionts, lower commensal bacteria and phages focused on Streptococcus, Lactococcus and Myoviridae. Pre/post: No changes in alpha/beta diversity of phages or bacteria were seen postrifaximin. Phage-bacterial linkages centred around urease-producing Streptococcus species collapsed postrifaximin. CONCLUSION: Unlike bacteria, faecal phages are sparsely linked with cirrhosis characteristics and 90-day outcomes. Phage and bacterial linkages centred on urease-producing, ammonia-generating Streptococcus species were affected by disease progression and rifaximin therapy and were altered in patients who experienced 90-day hospitalisations.


Assuntos
Antibacterianos/uso terapêutico , Doença Hepática Terminal/microbiologia , Firmicutes/virologia , Encefalopatia Hepática/microbiologia , Cirrose Hepática/microbiologia , Rifaximina/uso terapêutico , Idoso , Antibacterianos/farmacologia , Estudos Transversais , Progressão da Doença , Doença Hepática Terminal/etiologia , Faecalibacterium/genética , Faecalibacterium/virologia , Fezes/microbiologia , Feminino , Firmicutes/genética , Fármacos Gastrointestinais/uso terapêutico , Hospitalização , Humanos , Lactococcus/genética , Lactococcus/virologia , Lactulose/uso terapêutico , Cirrose Hepática/complicações , Cirrose Hepática/tratamento farmacológico , Masculino , Metagenoma/efeitos dos fármacos , Metagenômica , Interações Microbianas , Microviridae/genética , Pessoa de Meia-Idade , Myoviridae/genética , Gravidade do Paciente , Rifaximina/farmacologia , Streptococcus/genética , Streptococcus/virologia , Viroma/efeitos dos fármacos
12.
Microbiome ; 8(1): 164, 2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-33213522

RESUMO

BACKGROUND: The global threat of antimicrobial resistance (AMR) is a One Health problem impacted by antimicrobial use (AMU) for human and livestock applications. Extensive Iberian swine production is based on a more sustainable and eco-friendly management system, providing an excellent opportunity to evaluate how sustained differences in AMU impact the resistome, not only in the animals but also on the farm environment. Here, we evaluate the resistome footprint of an extensive pig farming system, maintained for decades, as compared to that of industrialized intensive pig farming by analyzing 105 fecal, environmental and slurry metagenomes from 38 farms. RESULTS: Our results evidence a significantly higher abundance of antimicrobial resistance genes (ARGs) on intensive farms and a link between AMU and AMR to certain antimicrobial classes. We observed differences in the resistome across sample types, with a higher richness and dispersion of ARGs within environmental samples than on those from feces or slurry. Indeed, a deeper analysis revealed that differences among the three sample types were defined by taxa-ARGs associations. Interestingly, mobilome analyses revealed that the observed AMR differences between intensive and extensive farms could be linked to differences in the abundance of mobile genetic elements (MGEs). Thus, while there were no differences in the abundance of chromosomal-associated ARGs between intensive and extensive herds, a significantly higher abundance of integrons in the environment and plasmids, regardless of the sample type, was detected on intensive farms. CONCLUSIONS: Overall, this study shows how AMU, production system, and sample type influence, mainly through MGEs, the profile and dispersion of ARGs in pig production. Video Abstract.


Assuntos
Animais Domésticos/microbiologia , Anti-Infecciosos/farmacologia , Resistência Microbiana a Medicamentos/efeitos dos fármacos , Resistência Microbiana a Medicamentos/genética , Microbiologia Ambiental , Fazendas , Fezes/microbiologia , Suínos/microbiologia , Animais , Genes Bacterianos/genética , Metagenoma/efeitos dos fármacos , Metagenoma/genética
13.
Pharmacol Res ; 161: 105225, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33007417

RESUMO

Cardiovascular disease (CVD) is the most wide-spread disorder all over the world. The personalized and precision diagnosis, treatment and prevention of CVD is still a challenge. With the developing of metagenome sequencing technologies and the paradigm shifting to data-driven discovery in life science, the computer aided microbiota biomarker discovery for CVD is becoming reality. We here summarize the data resources, knowledgebases and computational models available for CVD microbiota biomarker discovery, and review the present status of the findings about the microbiota patterns associated with the therapeutic effects on CVD. The future challenges and opportunities of the translational informatics on the personalized drug usages in CVD diagnosis, prognosis and treatment are also discussed.


Assuntos
Bactérias/efeitos dos fármacos , Biomarcadores/metabolismo , Fármacos Cardiovasculares/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Metagenoma/efeitos dos fármacos , Metagenômica , Medicina de Precisão , Animais , Bactérias/genética , Bactérias/metabolismo , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/microbiologia , Disbiose , Interações Hospedeiro-Patógeno , Humanos , Metagenoma/genética , Valor Preditivo dos Testes , Medição de Risco , Fatores de Risco
14.
Nat Commun ; 11(1): 5015, 2020 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-33024120

RESUMO

Human gut microbiome is a promising target for managing type 2 diabetes (T2D). Measures altering gut microbiota like oral intake of probiotics or berberine (BBR), a bacteriostatic agent, merit metabolic homoeostasis. We hence conducted a randomized, double-blind, placebo-controlled trial with newly diagnosed T2D patients from 20 centres in China. Four-hundred-nine eligible participants were enroled, randomly assigned (1:1:1:1) and completed a 12-week treatment of either BBR-alone, probiotics+BBR, probiotics-alone, or placebo, after a one-week run-in of gentamycin pretreatment. The changes in glycated haemoglobin, as the primary outcome, in the probiotics+BBR (least-squares mean [95% CI], -1.04[-1.19, -0.89]%) and BBR-alone group (-0.99[-1.16, -0.83]%) were significantly greater than that in the placebo and probiotics-alone groups (-0.59[-0.75, -0.44]%, -0.53[-0.68, -0.37]%, P < 0.001). BBR treatment induced more gastrointestinal side effects. Further metagenomics and metabolomic studies found that the hypoglycaemic effect of BBR is mediated by the inhibition of DCA biotransformation by Ruminococcus bromii. Therefore, our study reports a human microbial related mechanism underlying the antidiabetic effect of BBR on T2D. (Clinicaltrial.gov Identifier: NCT02861261).


Assuntos
Berberina/farmacologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Probióticos/uso terapêutico , Berberina/uso terapêutico , Feminino , Microbioma Gastrointestinal/fisiologia , Hemoglobinas Glicadas/metabolismo , Humanos , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Masculino , Metagenoma/efeitos dos fármacos , Metagenoma/genética , Pessoa de Meia-Idade , Placebos , Resultado do Tratamento
15.
Brain Res ; 1747: 147067, 2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-32827548

RESUMO

Research focusing on the gut-brain axis is growing, but the interplay of ethanol (alcohol molecule), the gut microbiome, the brain and behavior is poorly understood. In the current study, we remodeled the gut microbiota by providing adult male C57BL/6J mice with a non-absorbable antibiotic cocktail (ABX) in the drinking water and tested ethanol consumption behavior in a binge-like "Drinking in the Dark" model. Notably, 2 weeks of ABX pre-treatment significantly increased ethanol consumption during the 6 weeks of ethanol exposure in the DID paradigm. ABX treatment also appeared to prevent anxiety-like behavior during ethanol withdrawal period. ABX-treated mice expressed reduced bacterial diversity and modified microbiota compositions within cecal samples. There were drastically reduced levels of commensal Firmicutes and increases in the Bacteroidetes and Verrucomicrobia populations. Importantly, the relative abundance of Firmicutes inversely correlated to ethanol intake levels regardless of antibiotic treatment, whereas Bacteroidetes and Verrucomicrobia populations negatively correlated to ethanol intake levels. This is the first report demonstrating that ABX-induced disruption of the gut commensal microbiota leads to increased ethanol consumption in mice. This work reveals an important relationship between the gut microbiota and ethanol consumption behavior and supports the use of microbial-targeted approaches to study gut-brain interactions during alcohol use disorder.


Assuntos
Consumo de Bebidas Alcoólicas , Antibacterianos/farmacologia , Metagenoma/efeitos dos fármacos , Microbiota/efeitos dos fármacos , Animais , Etanol/sangue , Masculino , Camundongos
16.
Thorax ; 75(9): 780-790, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32631930

RESUMO

RATIONALE: The most common antibiotic used to treat people with cystic fibrosis (PWCF) is inhaled tobramycin, administered as maintenance therapy for chronic Pseudomonas aeruginosa lung infections. While the effects of inhaled tobramycin on P. aeruginosa abundance and lung function diminish with continued therapy, this maintenance treatment is known to improve long-term outcomes, underscoring how little is known about why antibiotics work in CF infections, what their effects are on complex CF sputum microbiomes and how to improve these treatments. OBJECTIVES: To rigorously define the effect of maintenance tobramycin on CF sputum microbiome characteristics. METHODS AND MEASUREMENTS: We collected sputum from 30 PWCF at standardised times before, during and after a single month-long course of maintenance inhaled tobramycin. We used traditional culture, quantitative PCR and metagenomic sequencing to define the dynamic effects of this treatment on sputum microbiomes, including abundance changes in both clinically targeted and untargeted bacteria, as well as functional gene categories. MAIN RESULTS: CF sputum microbiota changed most markedly by 1 week of antibiotic therapy and plateaued thereafter, and this shift was largely driven by changes in non-dominant taxa. The genetically conferred functional capacities (ie, metagenomes) of subjects' sputum communities changed little with antibiotic perturbation, despite taxonomic shifts, suggesting functional redundancy within the CF sputum microbiome. CONCLUSIONS: Maintenance treatment with inhaled tobramycin, an antibiotic with demonstrated long-term mortality benefit, primarily impacted clinically untargeted bacteria in CF sputum, highlighting the importance of monitoring the non-canonical effects of antibiotics and other treatments to accurately define and improve their clinical impact.


Assuntos
Antibacterianos/farmacologia , Bactérias , Fibrose Cística/microbiologia , Microbiota/efeitos dos fármacos , Escarro/microbiologia , Tobramicina/farmacologia , Administração por Inalação , Adolescente , Adulto , Idoso , Antibacterianos/uso terapêutico , Bactérias/genética , Bactérias/isolamento & purificação , Infecções Bacterianas/prevenção & controle , Criança , Fibrose Cística/fisiopatologia , Volume Expiratório Forçado , Humanos , Quimioterapia de Manutenção , Metagenoma/efeitos dos fármacos , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Fatores de Tempo , Tobramicina/uso terapêutico , Adulto Jovem
17.
Microbiome ; 8(1): 75, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32482165

RESUMO

BACKGROUND: Antibiotic resistance developed by bacteria is a significant threat to global health. Antibiotic resistance genes (ARGs) spread across different bacterial populations through multiple dissemination routes, including horizontal gene transfer mediated by bacteriophages. ARGs carried by bacteriophages are considered especially threatening due to their prolonged persistence in the environment, fast replication rates, and ability to infect diverse bacterial hosts. Several studies employing qPCR and viral metagenomics have shown that viral fraction and viral sequence reads in clinical and environmental samples carry many ARGs. However, only a few ARGs have been found in viral contigs assembled from metagenome reads, with most of these genes lacking effective antibiotic resistance phenotypes. Owing to the wide application of viral metagenomics, nevertheless, different classes of ARGs are being continuously found in viral metagenomes acquired from diverse environments. As such, the presence and functionality of ARGs encoded by bacteriophages remain up for debate. RESULTS: We evaluated ARGs excavated from viral contigs recovered from urban surface water viral metagenome data. In virome reads and contigs, diverse ARGs, including polymyxin resistance genes, multidrug efflux proteins, and ß-lactamases, were identified. In particular, when a lenient threshold of e value of ≤ 1 × e-5 and query coverage of ≥ 60% were employed in the Resfams database, the novel ß-lactamases blaHRV-1 and blaHRVM-1 were found. These genes had unique sequences, forming distinct clades of class A and subclass B3 ß-lactamases, respectively. Minimum inhibitory concentration analyses for E. coli strains harboring blaHRV-1 and blaHRVM-1 and catalytic kinetics of purified HRV-1 and HRVM-1 showed reduced susceptibility to penicillin, narrow- and extended-spectrum cephalosporins, and carbapenems. These genes were also found in bacterial metagenomes, indicating that they were harbored by actively infecting phages. CONCLUSION: Our results showed that viruses in the environment carry as-yet-unreported functional ARGs, albeit in small quantities. We thereby suggest that environmental bacteriophages could be reservoirs of widely variable, unknown ARGs that could be disseminated via virus-host interactions. Video abstract.


Assuntos
Bacteriófagos , Metagenoma , Antibacterianos/farmacologia , Bacteriófagos/genética , Farmacorresistência Bacteriana/genética , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Água Doce/virologia , Metagenoma/efeitos dos fármacos , Metagenômica , Vírus/genética
18.
Chemosphere ; 255: 127032, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32417519

RESUMO

Antibiotics can affect microbial community structure and promote antibiotic resistance. However, the course of microbial community recovery in wastewater treatment systems after antibiotic disturbance remains unclear. Herein, multiple molecular biology tools, including 16S amplicon sequencing, GeoChip 5.0, quantitative polymerase chain reaction (qPCR), and metagenomic sequencing, were used to investigate the year-long (352 d) recovery of the microbial community functional structure in an aerobic biofilm reactor. Nitrification was completely inhibited under 50 mg/L of streptomycin spiking (STM_50) due to the significant reduction of ammonia-oxidizing bacteria, but recovered to original pre-disturbance levels after streptomycin removal, indicating the high resilience of ammonia-oxidizing bacteria. Bacterial community richness and diversity decreased significantly under STM_50 (p < 0.05), but recovered to levels similar to those observed before disturbance after 352 d. In contrast, bacterial composition did not recover to the original structure. The carbon degradation and nitrogen cycling functional community significantly changed after recovery compared to that observed pre-disturbance (p < 0.05), thus indicating functional redundancy. Additionally, levels of aminoglycoside and total antibiotic resistance genes under STM_50 (relative abundance, 0.33 and 0.80, respectively) and after one year of recovery (0.12 and 0.29, respectively) were higher than the levels detected pre-disturbance (0.04 and 0.24, respectively). This study provides an overall depiction of the recovery of the microbial community functional structure after antibiotic exposure. Our findings give notice that recovery caused by antibiotic disturbance in the water environment should be taken more seriously, and that engineering control strategies should be implemented to prevent the antibiotic pollution of wastewater.


Assuntos
Antibacterianos/toxicidade , Biofilmes/efeitos dos fármacos , Reatores Biológicos/microbiologia , Microbiota/efeitos dos fármacos , Estreptomicina/toxicidade , Poluentes Químicos da Água/toxicidade , Purificação da Água/métodos , Aerobiose , Antibacterianos/análise , Biofilmes/crescimento & desenvolvimento , Carbono/metabolismo , Farmacorresistência Bacteriana/efeitos dos fármacos , Farmacorresistência Bacteriana/genética , Metagenoma/efeitos dos fármacos , Microbiota/genética , Nitrificação , Nitrogênio/metabolismo , Estreptomicina/análise , Águas Residuárias/química , Águas Residuárias/microbiologia , Poluentes Químicos da Água/análise
19.
Sci Total Environ ; 727: 138708, 2020 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-32334231

RESUMO

Antibiotic resistance genes (ARGs) harbored by plant microbiomes have been implicated as a potential risk to public health via food chain, especially directly edible fruits and vegetables. Here, we investigated the microbiome and antibiotic resistome in soil-strawberry ecosystem using shotgun metagenomic sequencing. The results showed that the enterobacterial population dominated the endophytes of strawberry fruits. Moreover, 85 subtypes of ARGs, including several clinically important ARGs, were detected in the strawberry fruit metagenomes. Additionally, host tracking analysis in combination with antibiotic-resistant bacterial isolate screening suggested that fruit-borne ARGs were mainly carried by members of the Enterobacteriaceae family. Unexpectedly, most of fruit-borne isolates were found to be resistant to several clinically important antimicrobials, e.g., erythromycin and cephalexin. Our findings provide broad insights into endophytic antibiotic resistomes of direct edible strawberry fruits and their potential hosts, and highlight the potential exposure risks of plant microbiomes to the human food chain.


Assuntos
Fragaria , Microbiota/efeitos dos fármacos , Antibacterianos/farmacologia , Resistência Microbiana a Medicamentos/efeitos dos fármacos , Enterobacteriaceae , Genes Bacterianos/efeitos dos fármacos , Humanos , Metagenoma/efeitos dos fármacos
20.
Environ Pollut ; 263(Pt B): 114470, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32247114

RESUMO

Erhai Lake in the Yunnan-Guizhou Plateau, a typical subtropical deep lake, experienced long-time input of nutrients and pollutants, which provides beneficial conditions for evolution and spread of resistance genes. In this study, the profile of bacterial community, antibiotic and metal resistance genes in sediments of Erhai Lake were analyzed via high-thought sequencing of 16S rRNA and metagenomic DNA. Proteobacteria, Firmicutes, Nitrospirae, and Bacteroidetes were found to be the main composition of the bacterial community. Multidrug-, bacitracin-, macrolide-lincosamide-streptogramin (MLS)- and tetracycline resistance genes were the primary antibiotic resistance gene (ARG) types with high relative abundance, whereas Multi-metal-, and arsenic- resistance genes were the primary metal resistance gene (MRG) types. The effects of nitrogen and phosphorus on the abundance of ARGs and MRGs depended on the different types. Some specific ARG (fosmidomycin resistance genes) and MRG types (multi-metal resistance genes) also showed significant geographic distribution. Bacterial community was the main driver shaping the resistome. Nutrients also played an essential role in structuring the bacterial community and resistome in the sediments of Erhai Lake. This study sheds light on the distribution and fate of resistome under a high load of nitrogen and phosphorus in a deep lake.


Assuntos
Lagos , Metagenoma/efeitos dos fármacos , Antibacterianos/farmacologia , China , Resistência Microbiana a Medicamentos/efeitos dos fármacos , Genes Bacterianos/efeitos dos fármacos , RNA Ribossômico 16S
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA