Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.544
Filtrer
1.
J Microbiol Biotechnol ; 34(7): 1484-1490, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-38960872

RÉSUMÉ

The gut microbiota is a key factor significantly impacting host health by influencing metabolism and immune function. Its composition can be altered by genetic factors, as well as environmental factors such as the host's surroundings, diet, and antibiotic usage. This study aims to examine how the characteristics of the gut microbiota in pigs, used as source animals for xenotransplantation, vary depending on their rearing environment. We compared the diversity and composition of gut microbiota in fecal samples from pigs raised in specific pathogen-free (SPF) and conventional (non-SPF) facilities. The 16S RNA metagenome sequencing results revealed that pigs raised in non-SPF facilities exhibited greater gut microbiota diversity compared to those in SPF facilities. Genera such as Streptococcus and Ruminococcus were more abundant in SPF pigs compared to non-SPF pigs, while Blautia, Bacteroides, and Roseburia were only observed in SPF pigs. Conversely, Prevotella was exclusively present in non-SPF pigs. It was predicted that SPF pigs would show higher levels of processes related to carbohydrate and nucleotide metabolism, and environmental information processing. On the other hand, energy and lipid metabolism, as well as processes associated with genetic information, cell communication, and diseases, were predicted to be more active in the gut microbiota of non-SPF pigs. This study provides insights into how the presence or absence of microorganisms, including pathogens, in pig-rearing facilities affects the composition and function of the pigs' gut microbiota. Furthermore, this serves as a reference for tracing whether xenotransplantation source pigs were maintained in a pathogen-controlled environment.


Sujet(s)
Bactéries , Fèces , Microbiome gastro-intestinal , ARN ribosomique 16S , Animaux , Suidae/microbiologie , Fèces/microbiologie , Bactéries/classification , Bactéries/génétique , Bactéries/isolement et purification , ARN ribosomique 16S/génétique , Organismes exempts d'organismes pathogènes spécifiques , Métagénome , Adaptation physiologique
2.
Microbiome ; 12(1): 116, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38943206

RÉSUMÉ

BACKGROUND: Population stratification based on interindividual variability in gut microbiota composition has revealed the existence of several ecotypes named enterotypes in humans and various animal species. Enterotypes are often associated with environmental factors including diet, but knowledge of the role of host genetics remains scarce. Moreover, enterotypes harbor functionalities likely associated with varying abilities and susceptibilities of their host. Previously, we showed that under controlled conditions, 60-day-old pig populations consistently split into two enterotypes with either Prevotella and Mitsuokella (PM enterotype) or Ruminococcus and Treponema (RT enterotype) as keystone taxa. Here, our aim was to rely on pig as a model to study the influence of host genetics to assemble enterotypes, and to provide clues on enterotype functional differences and their links with growth traits. RESULTS: We established two pig lines contrasted for abundances of the genera pairs specifying each enterotype at 60 days of age and assessed them for fecal microbiota composition and growth throughout three consecutive generations. Response to selection across three generations revealed, per line, an increase in the prevalence of the selected enterotype and in the average relative abundances of directly and indirectly selected bacterial genera. The PM enterotype was found less diverse than the RT enterotype but more efficient for piglet growth during the post-weaning period. Shotgun metagenomics revealed differentially abundant bacterial species between the two enterotypes. By using the KEGG Orthology database, we show that functions related to starch degradation and polysaccharide metabolism are enriched in the PM enterotype, whereas functions related to general nucleoside transport and peptide/nickel transport are enriched in the RT enterotype. Our results also suggest that the PM and RT enterotypes might differ in the metabolism of valine, leucin, and isoleucine, favoring their biosynthesis and degradation, respectively. CONCLUSION: We experimentally demonstrated that enterotypes are functional ecosystems that can be selected as a whole by exerting pressure on the host genetics. We also highlight that holobionts should be considered as units of selection in breeding programs. These results pave the way for a holistic use of host genetics, microbiota diversity, and enterotype functionalities to understand holobiont shaping and adaptation. Video Abstract.


Sujet(s)
Fèces , Microbiome gastro-intestinal , Animaux , Microbiome gastro-intestinal/génétique , Suidae/microbiologie , Fèces/microbiologie , Bactéries/classification , Bactéries/génétique , Métagénomique/méthodes , Prevotella/génétique , Prevotella/classification , Ruminococcus/génétique , Treponema/génétique
3.
Curr Microbiol ; 81(8): 225, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38877167

RÉSUMÉ

Linezolid resistance in Enterococcus spp. is increasingly considered critically important and a public health threat which mandates the need to understand their genomic contents and dissemination patterns. Here, we used whole-genome sequencing to characterize the resistome, virulome and mobile genetic elements of nine linezolid-resistant (LZDR) enterococci (seven optrA-E. faecalis, one poxtA-E. faecium and one optrA-E. casseliflavus) previously obtained from the nares of healthy dogs, pigs, pig farmers and tracheal samples of nestling storks in Spain. Also, the relatedness of the isolates with publicly available genomes was accessed by core-genome single nucleotide polymorphism (SNP) analysis. The optrA gene of the E. faecalis and E. casseliflavus isolates was located downstream of the fexA gene. The optrA gene in the E. casseliflavus isolate was carried in a plasmid (pURX4962), while those in the seven E. faecalis isolates were chromosomally located. The OptrA proteins were mostly variants of wild type (DP-2: Y176D/T481P; RDK: I104R/Y176D/E256K; DD-3: Y176D/G393D; and EDD: K3E/Y176D/G393D), except two that were wild type (one E. faecalis and one E. casseliflavus). The poxtA gene in the E. faecium isolate was found alone within its contig. The cfrD was upstream of ermB gene in the E. casseliflavus isolate and flanked by ISNCY and IS1216. All the LZDR enterococci carried plasmid rep genes (2-3) containing tetracycline, chloramphenicol and aminoglycoside resistance genes. All isolates except E. casseliflavus carried at least one intact prophage, of which E. faecalis-ST330 (X4957) from a pig carried the highest (n = 5). Tn6260 was associated with lnuG in E. faecalis-ST330 while Tn554 was with fexA in E. feaecalis-ST59 isolates. All except E. casseliflavus (n = 0) carried at least two metal resistance genes (MRGs), of which poxtA-carrying E. faecium-ST1739 isolate contained the most (arsA, copA, fief, ziaA, znuA, zosA, zupT, and zur). SNP-based analyses identified closely related optrA-E. faecalis isolates from a pig and a pig farmer on the same farm (SNP = 4). Moreover, optrA- carrying E. faecalis-ST32, -ST59, and -ST474 isolates from pigs were related to those previously described from humans (sick and healthy) and cattle in Spain, Belgium, and Switzerland (SNP range 43-86). These findings strongly suggest the transmission of LZDR-E. faecalis between a pig and a pig farmer and potential inter-country dissemination. These highlight the need to strengthen molecular surveillance of LZDR enterococci in all ecological niches and body parts to direct appropriate control strategies.


Sujet(s)
Antibactériens , Résistance bactérienne aux médicaments , Enterococcus , Génome bactérien , Linézolide , Phylogenèse , Animaux , Linézolide/pharmacologie , Suidae/microbiologie , Résistance bactérienne aux médicaments/génétique , Chiens , Antibactériens/pharmacologie , Enterococcus/génétique , Enterococcus/effets des médicaments et des substances chimiques , Enterococcus/isolement et purification , Enterococcus/classification , Infections bactériennes à Gram positif/microbiologie , Infections bactériennes à Gram positif/transmission , Infections bactériennes à Gram positif/médecine vétérinaire , Humains , Séquençage du génome entier , Espagne , Polymorphisme de nucléotide simple , Tests de sensibilité microbienne , Protéines bactériennes/génétique , Génomique , Plasmides/génétique
4.
J Antimicrob Chemother ; 79(8): 1900-1909, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38943539

RÉSUMÉ

OBJECTIVES: To characterize blaNDM-carrying Salmonella recovered from a pig slaughterhouse. METHODS: In this study, 46 environment samples were collected from a slaughterhouse in China, and screened for carbapenem-resistant Enterobacterales. WGS, antimicrobial susceptibility testing and conjugation experiments were carried out to identify the isolates' resistance phenotypes and genetic characteristics. The phylogenetic relatedness of the Salmonella isolates obtained in this study and Salmonella (ST34 and ST29) in GenBank was determined. RESULTS: Two ST34 Salmonella Typhimurium and one ST29 Salmonella Stanley, recovered from three environmental samples (6.52%), were positive for blaNDM-1 and blaNDM-5, respectively. The two ST34 S. Typhimurium strains exhibited a close relationship (10-36 SNPs) with two human-derived blaNDM-1-bearing isolates from China (Hong Kong and Guangxi Province) and two blaNDM-negative ST34 Salmonella strains from the UK. The blaNDM-1 genes were located on IncHI2/ST3 plasmids. The capture of blaNDM-1 by the IncHI2/ST3 plasmid seems to be due to homologous recombination mediated by circular structures, as the genetic arrangements of the blaNDM-1 gene contain two IS26 elements of the same orientation. The blaNDM-5 gene was also carried by the IncHI2/ST3 plasmid, which shares highly similar structures with other blaNDM-5-bearing IncHI2/ST3 plasmids from other sources (fish, chicken, duck, human). CONCLUSIONS: This is the first report of a blaNDM-5-carrying IncHI2/ST3 plasmid in Salmonella. The clonal spread of NDM-1-producing ST34 S. Typhimurium across human and animal-associated environments, and the widespread dissemination of epidemic blaNDM-5-carrying IncHI2/ST3 plasmids among Enterobacteriaceae in China indicate the potential of further dissemination of blaNDM among Salmonella, which poses a threat to public health.


Sujet(s)
Antibactériens , Phylogenèse , Plasmides , Salmonella typhimurium , bêta-Lactamases , Animaux , Humains , Abattoirs , Antibactériens/pharmacologie , bêta-Lactamases/génétique , Chine/épidémiologie , Conjugaison génétique , Tests de sensibilité microbienne , Plasmides/génétique , Salmonelloses animales/microbiologie , Salmonelloses animales/épidémiologie , Salmonella typhimurium/génétique , Salmonella typhimurium/effets des médicaments et des substances chimiques , Salmonella typhimurium/isolement et purification , Suidae/microbiologie , Séquençage du génome entier
5.
Methods Mol Biol ; 2815: 93-113, 2024.
Article de Anglais | MEDLINE | ID: mdl-38884913

RÉSUMÉ

Massive sequencing of a fragment of 16S rRNA gene allows the characterization of bacterial communities in different body sites: the microbiota. Nasal microbiota can be analyzed by DNA extraction from nasal swabs, amplification of the specific fragment of interest, and posterior sequencing. The raw sequences obtained need to go through a computational process to check their quality and then assign the taxonomy. Here, we will describe the complete process from sampling to get the microbial diversity of nasal microbiota in health and disease.


Sujet(s)
Microbiote , ARN ribosomique 16S , Animaux , Microbiote/génétique , Suidae/microbiologie , ARN ribosomique 16S/génétique , Nez/microbiologie , Bactéries/génétique , Bactéries/classification , Bactéries/isolement et purification , ADN bactérien/génétique , Séquençage nucléotidique à haut débit/méthodes , Maladies des porcs/microbiologie , Analyse de séquence d'ADN/méthodes
6.
Fungal Genet Biol ; 173: 103898, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38815692

RÉSUMÉ

The skin and its microbiome function to protect the host from pathogen colonization and environmental stressors. In this study, using the Wisconsin Miniature Swine™ model, we characterize the porcine skin fungal and bacterial microbiomes, identify bacterial isolates displaying antifungal activity, and use whole-genome sequencing to identify biosynthetic gene clusters encoding for secondary metabolites that may be responsible for the antagonistic effects on fungi. Through this comprehensive approach of paired microbiome sequencing with culturomics, we report the discovery of novel species of Corynebacterium and Rothia. Further, this study represents the first comprehensive evaluation of the porcine skin mycobiome and the evaluation of bacterial-fungal interactions on this surface. Several diverse bacterial isolates exhibit potent antifungal properties against opportunistic fungal pathogens in vitro. Genomic analysis of inhibitory species revealed a diverse repertoire of uncharacterized biosynthetic gene clusters suggesting a reservoir of novel chemical and biological diversity. Collectively, the porcine skin microbiome represents a potential unique source of novel antifungals.


Sujet(s)
Champignons , Microbiote , Peau , Animaux , Peau/microbiologie , Suidae/microbiologie , Microbiote/génétique , Champignons/génétique , Champignons/effets des médicaments et des substances chimiques , Antifongiques/pharmacologie , Antibiose , Mycobiome/génétique , Bactéries/génétique , Bactéries/classification , Bactéries/effets des médicaments et des substances chimiques , Bactéries/isolement et purification , Bactéries/métabolisme , Corynebacterium/génétique , Corynebacterium/effets des médicaments et des substances chimiques , Porc miniature/microbiologie , Famille multigénique , Séquençage du génome entier , Métabolisme secondaire/génétique
7.
Trop Anim Health Prod ; 56(5): 179, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38809309

RÉSUMÉ

We evaluated the effects of supplementing yeast mannan-reach-fraction on growth performance, jejunal morphology and lymphoid tissue characteristics in weaned piglets challenged with E. Coli F4. A total of 20 crossbred piglets were used. At weaning, piglets were assigned at random to one of four groups: piglets challenged and fed the basal diet supplemented with yeast mannan-rich fraction (C-MRF, n = 5); piglets challenged and fed the basal diet (C-BD, n = 5); piglets not challenged and fed the basal diet supplemented with yeast mannan-rich fraction (NC-MRF, n = 5), and piglets not challenged and fed the basal diet (NC-BD). Each dietary treatment had five replicates. On days 4, 5 and 10, piglets were orally challenged with 108 CFU/mL of E. Coli F4. C-MRF piglets had higher BW (p = 0.002; interactive effect) than C-BD piglets. C-MRF piglets had higher (p = 0.02; interactive effect) ADG in comparison with C-BD piglets. C-MRF piglets had higher (p = 0.04; interactive effect) ADFI than C-BD piglets. The diameter of lymphoid follicles was larger (p = 0.010; interactive effect) in the tonsils of C-MRF piglets than C-BD piglets. Lymphoid cells proliferation was greater in the mesenteric lymphnodes and ileum (p = 0.04 and p = 0.03, respectively) of C-MRF piglets. A reduction (p > 0.05) in E. Coli adherence in the ileum of piglets fed MRF was observed. In conclusion, the results of the present study demonstrate that dietary yeast mannan-rich fraction supplementation was effective in protecting weaned piglets against E. Coli F4 challenge.


Sujet(s)
Compléments alimentaires , Escherichia coli entérotoxigène , Mannanes , Levures , Animaux , Suidae/croissance et développement , Suidae/microbiologie , Infections à Escherichia coli/médecine vétérinaire , Maladies des porcs/microbiologie , Jéjunum/croissance et développement , Sevrage , Élevage , Tissu lymphoïde/physiologie
8.
Appl Environ Microbiol ; 90(5): e0026424, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38695519

RÉSUMÉ

The emergence of foodborne Salmonella strains carrying antimicrobial resistance (AMR) in mobile genetic elements (MGE) is a significant public health threat in a One Health context requiring continuous surveillance. Resistance to ciprofloxacin and cephalosporins is of particular concern. Since pigs are a relevant source of foodborne Salmonella for human beings, we studied transmissible AMR genes and MGE in a collection of 83 strains showing 9 different serovars and 15 patterns of multidrug resistant (MDR) previously isolated from pigs raised in the conventional breeding system of Northern Spain. All isolates were susceptible to ciprofloxacin and three isolates carried blaCMY-2 or blaCTX-M-9 genes responsible for cefotaxime resistance. Filter mating experiments showed that the two plasmids carrying blaCTX-M-9 were conjugative while that carrying blaCMY-2 was self-transmissible by transformation. Whole-genome sequencing and comparative analyses were performed on the isolates and plasmids. The IncC plasmid pSB109, carrying blaCMY-2, was similar to one found in S. Reading from cattle, indicating potential horizontal transfer between serovars and animal sources. The IncHI2 plasmids pSH102 in S. Heidelberg and pSTM45 in S. Typhimurium ST34, carrying blaCTX-M-9, shared similar backbones and two novel "complex class 1 integrons" containing different AMR and heavy metal genes. Our findings emphasize the importance of sequencing techniques to identify emerging AMR regions in conjugative and stable plasmids from livestock production. The presence of MGE carrying clinically relevant AMR genes raises public health concerns, requiring monitoring to mitigate the emergence of bacteria carrying AMR genes and subsequent spread through animals and food.IMPORTANCEThe emergence of foodborne Salmonella strains carrying antimicrobial resistance (AMR) in mobile genetic elements (MGE) is a significant public health threat in a One Health context. Since pigs are a relevant source of foodborne Salmonella for humans, in this study, we investigate different aspects of AMR in a collection of 83 Salmonella showing nine different serovars and 15 patterns of multidrug resistant (MDR) isolated from pigs raised in the conventional breeding system. Our findings emphasize the importance of sequencing techniques to identify emerging AMR regions in conjugative and stable plasmids from livestock production. The presence of MGE carrying clinically relevant AMR genes raises public health concerns, requiring monitoring to mitigate the emergence of bacteria carrying AMR genes and subsequent spread through animals and food.


Sujet(s)
Antibactériens , Multirésistance bactérienne aux médicaments , Séquences répétées dispersées , Plasmides , Salmonella , Animaux , Suidae/microbiologie , Plasmides/génétique , Salmonella/génétique , Salmonella/effets des médicaments et des substances chimiques , Salmonella/isolement et purification , Multirésistance bactérienne aux médicaments/génétique , Antibactériens/pharmacologie , Humains , Résistance aux céphalosporines/génétique , Salmonelloses animales/microbiologie , Espagne , Maladies des porcs/microbiologie , Céphalosporines/pharmacologie , Transfert horizontal de gène
9.
BMC Microbiol ; 24(1): 182, 2024 May 25.
Article de Anglais | MEDLINE | ID: mdl-38789948

RÉSUMÉ

BACKGROUND: It is vital to understand healthy gut microbiota composition throughout early life stages when environments are changing, and immunity is developing. There are limited large-scale longitudinal studies classifying healthy succession of swine microbiota. The objectives of this study were to (a) determine the microbiota composition of fecal samples collected from piglets within a few days after birth until one-week post-weaning, and (b) investigate the associations of early fecal microbiota with pig growth performance in nursery and later growing stages. Fecal samples were collected from nine cohorts of 40 pigs (n = 360) from distinct farrowing sources in Ontario and Quebec, Canada at four timepoints from birth to one-week post-weaning, with pig body weight was recorded at each fecal sampling. RESULTS: Microbiota was dominated by the phyla Firmicutes, Bacteroides and Proteobacteria. There were notable differences in genera abundance between pigs from different provinces and farming systems. Over the early life stage, the genera Bacteroides, Escherichia/Shigella, and Clostridium cluster XIVa were abundant preweaning, while Prevotella dominated post-weaning. Hierarchical clustering identified three major stages of microbiota development, each associated with distinct composition. Stage one occurs from birth to 7 days, stage two from 7 days after birth until weaning, and stage three from weaning to one-week post-weaning. Three enterotypes were identified in stage two that showed differences in growth before weaning, and in the grower production stage. Piglets with a microbiota enterotype characterized by higher abundance of Prevotella and unclassified Ruminococcaceae had lower growth performance in the pre-weaning stage, and the growing stage. CONCLUSION: These findings help identify the timing of microbiota shifts across early swine life which may be the optimal time for external intervention to shift the microbiota to a beneficial state. The project findings should help decrease antimicrobial use, increase animal welfare, and have positive economic impacts.


Sujet(s)
Bactéries , Fèces , Microbiome gastro-intestinal , Sevrage , Animaux , Fèces/microbiologie , Études longitudinales , Suidae/microbiologie , Bactéries/classification , Bactéries/génétique , Bactéries/isolement et purification , Bactéries/croissance et développement , Ontario , ARN ribosomique 16S/génétique , Québec , Animaux nouveau-nés
10.
PLoS One ; 19(5): e0302724, 2024.
Article de Anglais | MEDLINE | ID: mdl-38709788

RÉSUMÉ

The early gut microbiota composition is fundamentally important for piglet health, affecting long-term microbiome development and immunity. In this study, the gut microbiota of postparturient dams was compared with that of their offspring in three Finnish pig farms at three growth phases. The differences in fecal microbiota of three study development groups (Good, Poorly, and PrematureDeath) were analyzed at birth (initial exposure phase), weaning (transitional phase), and before slaughter (stable phase). Dam Lactobacillaceae abundance was lower than in piglets at birth. Limosilactobacillus reuteri and Lactobacillus amylovorus were dominantly expressed in dams and their offspring. Altogether 17 piglets (68%) were identified with Lactobacillaceae at the initial exposure phase, divided unevenly among the development groups: 85% of Good, 37.5% of Poorly, and 75% of PrematureDeath pigs. The development group Good was identified with the highest microbial diversity, whereas the development group PrematureDeath had the lowest diversity. After weaning, the abundance and versatility of Lactobacillaceae in piglets diminished, shifting towards the microbiome of the dam. In conclusion, the fecal microbiota of pigs tends to develop towards a similar alpha and beta diversity despite development group and rearing environment.


Sujet(s)
Fèces , Microbiome gastro-intestinal , Sevrage , Animaux , Fèces/microbiologie , Suidae/microbiologie , Suidae/croissance et développement , Femelle , Lactobacillaceae/croissance et développement , Lactobacillaceae/génétique , ARN ribosomique 16S/génétique
11.
Microbiol Spectr ; 12(6): e0408423, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38717193

RÉSUMÉ

Researchers have extensively studied the effect of oxygen on the growth and survival of bacteria. However, the impact of oxygen on bacterial community structure, particularly its ability to select for taxa within the context of a complex microbial community, is still unclear. In a 21-day microcosm experiment, we investigated the effect of aerobic exposure on the fecal community structure and succession pattern in broiler, calf, and piglet feces (n = 10 for each feces type). Bacterial diversity decreased and community structure changed rapidly in the broiler microbiome (P < 0.001), while the fecal community of calves and piglets, which have higher initial diversity, was stable after initial exposure but decreased in diversity after 3 days (P < 0.001). The response to aerobic exposure was host animal specific, but in all three animals, the change in community structure was driven by a decrease in anaerobic species, primarily belonging to Firmicutes and Bacteroidetes (except in broilers where Bacteroidetes increased), along with an increase in aerobic species belonging to Proteobacteria and Actinobacteria. Using random forest regression, we identified microbial features that predict aerobic exposure. In all three animals, host-beneficial Prevotella-related ASVs decreased after exposure, while ASVs belonging to Acinetobacter, Corynbacterium, and Tissierella were increased. The decrease of Prevotella was rapid in broilers but delayed in calves and piglets. Knowing when these pathobionts increase in abundance after aerobic exposure could inform farm sanitation practices and could be important in designing animal experiments that modulate the microbiome.IMPORTANCEThe fecal microbial community is contained within a dynamic ecosystem of interacting microbes that varies in biotic and abiotic components across different animal species. Although oxygen affects bacterial growth, its specific impact on the structure of complex communities, such as those found in feces, and how these effects vary between different animal species are poorly understood. In this study, we demonstrate that the effect of aerobic exposure on the fecal microbiota was host-animal-specific, primarily driven by a decrease in Firmicutes and Bacteroidetes, but accompanied by an increase in Actinobacteria, Proteobacteria, and other pathobionts. Interestingly, we observed that more complex communities from pig and cattle exhibited initial resilience, while a less diverse community from broilers displayed a rapid response to aerobic exposure. Our findings offer insights that can inform farm sanitation practices, as well as experimental design, sample collection, and processing protocols for microbiome studies across various animal species.


Sujet(s)
Bactéries , Poulets , Fèces , Microbiome gastro-intestinal , Animaux , Fèces/microbiologie , Poulets/microbiologie , Suidae/microbiologie , Bovins/microbiologie , Bactéries/classification , Bactéries/génétique , Bactéries/isolement et purification , Aérobiose , ARN ribosomique 16S/génétique , Bacteroidetes/génétique , Bacteroidetes/classification , Bacteroidetes/isolement et purification , Microbiote
12.
J Antimicrob Chemother ; 79(6): 1329-1336, 2024 06 03.
Article de Anglais | MEDLINE | ID: mdl-38629139

RÉSUMÉ

OBJECTIVES: To characterize and elucidate the spread of amikacin-resistant Enterobacteriaceae isolates from environmental samples on a pig farm in the UK, following the previous identification of index Salmonella isolates harbouring the rmtB gene, a 16S rRNA methylase. METHODS: Environmental samples were collected during two visits to a pig farm in the UK. Isolates were recovered using selective media (amikacin 128 mg/L) followed by real-time PCR and WGS to analyse rmtB-carrying Salmonella and Escherichia coli isolates. RESULTS: Salmonella and E. coli isolates harbouring the rmtB gene were detected at both farm visits. All Salmonella isolates were found to be monophasic S. enterica serovar Typhimurium variant Copenhagen of ST34. rmtB-harbouring E. coli isolates were found to be one of three STs: ST4089, ST1684 and ST34. Long-read sequencing identified the rmtB gene to be chromosomally located in Salmonella isolates and on IncFII-type plasmids in E. coli isolates. The results showed the rmtB gene to be flanked by IS26 elements and several resistance genes. CONCLUSIONS: We report on the occurrence of rmtB-harbouring Enterobacteriaceae on a pig farm in the UK. rmtB confers resistance to multiple aminoglycosides and this work highlights the need for surveillance to assess dissemination and risk.


Sujet(s)
Antibactériens , Escherichia coli , Fermes , Methyltransferases , Salmonella , Animaux , Suidae/microbiologie , Escherichia coli/génétique , Escherichia coli/effets des médicaments et des substances chimiques , Escherichia coli/isolement et purification , Escherichia coli/classification , Antibactériens/pharmacologie , Royaume-Uni , Salmonella/génétique , Salmonella/effets des médicaments et des substances chimiques , Salmonella/isolement et purification , Salmonella/classification , Methyltransferases/génétique , Tests de sensibilité microbienne , Amikacine/pharmacologie , Séquençage du génome entier , Plasmides/génétique , Résistance bactérienne aux médicaments/génétique , Infections à Escherichia coli/microbiologie , Infections à Escherichia coli/médecine vétérinaire , Maladies des porcs/microbiologie , Protéines Escherichia coli/génétique
13.
J Microbiol Biotechnol ; 34(5): 1101-1108, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38563109

RÉSUMÉ

Earlier studies have validated the isolation of extended-spectrum beta-lactamase-producing Salmonella (ESBL-Sal) strains from food. While poultry is recognized as a reservoir for Salmonella contamination, pertinent data regarding ESBL-Sal remains limited. Consequently, the Ministry of Food and Drug Safety has isolated Salmonella spp. from retail meat and evaluated their antibiotic susceptibility and genetic characteristics via whole-genome sequencing. To further elucidate these aspects, this study investigates the prevalence, antibiotic resistance profiles, genomic characteristics, and homology of ESBL-Sal spp. obtained from livestock-derived products in South Korean retail outlets. A total of 653 Salmonella spp. were isolated from 1,876 meat samples, including 509 beef, 503 pork, 555 chicken, and 309 duck samples. The prevalence rates of Salmonella were 0.0%, 1.4%, 17.5%, and 28.2% in the beef, pork, chicken, and duck samples, respectively. ESBL-Sal was exclusively identified in poultry meat, with a prevalence of 1.4% in the chicken samples (8/555) and 0.3% in the duck samples (1/309). All ESBL-Sal strains carried the blaCTX-M-1 gene and exhibited resistance to ampicillin, ceftiofur, ceftazidime, nalidixic acid, and tetracycline. Eight ESBL-Sal isolates were identified as S. Enteritidis with sequence type (ST) 11. The major plasmid replicons of the Enteritidis-ST11 strains were IncFIB(S) and IncFII(S), carrying antimicrobial resistance genes (ß-lactam, tetracycline, and aminoglycoside) and 166 virulence factor genes. The results of this study provide valuable insights for the surveillance and monitoring of ESBL-Sal in South Korean food chain.


Sujet(s)
Antibactériens , Poulets , Canards , Microbiologie alimentaire , Viande , Tests de sensibilité microbienne , Salmonella , bêta-Lactamases , bêta-Lactamases/génétique , Animaux , République de Corée , Salmonella/génétique , Salmonella/isolement et purification , Salmonella/enzymologie , Salmonella/effets des médicaments et des substances chimiques , Viande/microbiologie , Antibactériens/pharmacologie , Poulets/microbiologie , Canards/microbiologie , Bovins , Suidae/microbiologie , Séquençage du génome entier , Multirésistance bactérienne aux médicaments/génétique , Prévalence , Volaille/microbiologie , Plasmides/génétique
14.
Benef Microbes ; 15(2): 195-210, 2024 Jan 31.
Article de Anglais | MEDLINE | ID: mdl-38350485

RÉSUMÉ

Post-weaning diarrhoea in piglets remains an important cause of economic losses for swine producers. Feed supplementation with probiotics is one of the alternatives to antibiotics used to reduce the impact of such gastrointestinal disease. The aim of the present study was to evaluate the effect of Ligilactobacillus salivarius PS21603 supplementation on the intestinal structure and the gut microbiota composition of weaned piglets. Safety and tolerance of L. salivarius PS21603 were previously evaluated in a 28-days study using 384 weaned piglets (28 ± 2 days old and 7.5 ± 1.5 kg) divided in three treatment groups: T1: Basal diet + L. salivarius PS21603 109 cfu/day, T2: Basal diet + L. salivarius PS21603 107 cfu/day, and T3: Basal diet (control group). For the present study, 16 piglets per treatment group were randomly selected and faecal samples were collected on day 0 (weaning) and 28 of study. At the end of study, three males and three females per treatment were euthanised. Intestinal morphometric values were measured after necropsy. Faecal counts of Escherichia coli were evaluated by culture techniques, and faecal microbiota composition was assessed by high-throughput sequencing. All data were analysed and compared between treatment groups. Supplementation with L. salivarius PS21603 caused an increase in the intestine length of piglets from T1 and in the villous height:crypt ratio of piglets from T2 (P < 0.05) compared to T3 on day 28. According to the Shannon Diversity Index, microbiota diversity increased on day 28 compared to day 0, with no significant differences observed between treatments. The main changes in the relative abundance of bacteria at the phylum, family, and genus levels were observed between different sampling time points. However, piglets from T1 and T2 had lower faecal E. coli counts than T3 on day 28 (P < 0.05). Moreover, supplementation with L. salivarius PS21603 modulated gut microbiota through a more optimal composition, reducing Escherichia and increasing Bifidobacterium relative abundance in piglets from T1 (P < 0.05) from the beginning to the end of the study. Therefore, the strain L. salivarius PS21603 has shown probiotic properties to be used as feed additive in the pig industry, along with good hygiene and farm management practices, for the prevention and/or treatment of post-weaning diarrhoea in piglets.


Sujet(s)
Aliment pour animaux , Compléments alimentaires , Fèces , Microbiome gastro-intestinal , Ligilactobacillus salivarius , Probiotiques , Sevrage , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Suidae/microbiologie , Probiotiques/administration et posologie , Probiotiques/pharmacologie , Mâle , Femelle , Ligilactobacillus salivarius/physiologie , Aliment pour animaux/analyse , Fèces/microbiologie , Intestins/microbiologie , Diarrhée/médecine vétérinaire , Diarrhée/microbiologie , Diarrhée/prévention et contrôle , Maladies des porcs/microbiologie , Maladies des porcs/prévention et contrôle , Escherichia coli
15.
Curr Microbiol ; 80(3): 91, 2023 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-36725751

RÉSUMÉ

A Gram-negative, obligate anaerobic, non-motile, non-spore-forming, rod-shaped bacterial strain designated AGMB00274T was isolated from swine faeces. An 16S rRNA gene analysis indicated that strain AGMB00274T belonged to the genus Parabacteroides, with the highest similarity to Parabacteroides johnsonii (P. johnsonii) DSM 18315T (sequence similarity of 94.9%). The genome size of strain AGMB00274T was 4,308,683 bp, with a DNA G+C content of 42.5 mol%. The biochemical analysis of strain AGMB00274T showed that it was positive for gelatin hydrolysis and α-fucosidase, but negative for the acid production from D-glucose, D-mannitol, D-maltose, salicin, glycerol, D-cellobiose, D-mannose, D-melezitose, D-sorbitol, D-trehalose, and negative for α-arabinosidase, glutamic acid decarboxylase, and pyroglutamic acid arylamidase. The dominant cellular fatty acids (> 10%) of the isolate were anteiso-C15: 0 (23.2%), iso-C15: 0 (16.6%), C18: 1 ω9c (16.4%), summed feature 11 (iso-C17: 0 3-OH and/or C18: 2 DMA) (12.5%), and C16: 0 (11.3%). The major respiratory quinones of strain AGMB00274T were MK-9 (55.4%) and MK-10 (44.6%). The major polar lipid was phosphatidylethanolamine. Based on phylogenetic, genetic, physiological, and chemotaxonomic analyses, as a novel species of the genus Parabacteroides, strain AGMB00274T was proposed with the name Parabacteroides faecalis sp. nov. The type strain used was AGMB00274T (= KCTC 25286T = GDMCC 1.2742T).


Sujet(s)
Bacteroidetes , Phylogenèse , Animaux , Techniques de typage bactérien , ADN bactérien/génétique , Acides gras/composition chimique , Fèces/microbiologie , Phospholipides/composition chimique , ARN ribosomique 16S/génétique , Analyse de séquence d'ADN , Suidae/microbiologie , Vitamine K2/composition chimique , Bacteroidetes/classification , Bacteroidetes/isolement et purification
16.
Food Microbiol ; 109: 104135, 2023 Feb.
Article de Anglais | MEDLINE | ID: mdl-36309455

RÉSUMÉ

Salmonella enterica serovar Typhimurium monophasic variants (Salmonella 4,[5],12:i:-) has increased dramatically, causing human salmonellosis and colonization in pigs. With a difference to S. Typhimurium, the monophasic variants of S. Typhimurium lose the gene cassettes encoding the second phase flagellin. To establish a rapid method to detect and differentiate the two serotypes, we analyzed the published 679 genomes of S. Typhimurium and its monophasic variants and found that no Salmonella 4,[5],12:i:- strains carry both fljB and hin genes. Therefore, we established a novel multiplex PCR method using the fljB-hin region and mdh gene as target sequences to detect and differentiate both serotypes. This method can be used to specifically detect both serotypes with a detection limit for DNA concentration at 10 pg/µL. In addition, the PCR assay successfully differentiated 36 S. Typhimurium isolates from 62 isolates of monophasic variants preserved in our laboratory from 2009 to 2017, which corresponds to the whole-genome-based serotyping results. Application of the multiplex PCR method to 60 fecal samples from a pig farm identified 11.7% (7/60) of S. Typhimurium monophasic variants, which is consistent with the whole-genome-based serotyping results. The multiplex PCR assay is a rapid and precise method for the detection of S. Typhimurium monophasic variants from samples across food production chains.


Sujet(s)
Salmonella enterica , Salmonella typhimurium , Animaux , Fermes , Réaction de polymérisation en chaine multiplex , Salmonella enterica/génétique , Salmonella enterica/isolement et purification , Salmonella typhimurium/génétique , Salmonella typhimurium/isolement et purification , Sérogroupe , Suidae/microbiologie , Génome bactérien
17.
Parasit Vectors ; 15(1): 433, 2022 Nov 17.
Article de Anglais | MEDLINE | ID: mdl-36397169

RÉSUMÉ

BACKGROUND: The porcine roundworm Ascaris suum impairs feed conversion and weight gain, but its effects on intestinal microbiota remain largely unexplored. METHODS: Modulation of the intestinal microbiota was assessed in pigs that were infected once with 10,000 A. suum eggs and pigs that received a trickle infection (1000 eggs/day over 10 days), compared with a non-infected control group. Six pigs each were sacrificed per group at days 21, 35 and 49 post-infection (p.i.). Faecal samples taken weekly until slaughter and ingesta samples from different intestinal compartments were subjected to next-generation sequencing of the bacterial 16S rRNA gene. RESULTS: The results revealed marked differences between the single- and the trickle-infected group. Single infection caused a remarkable but transient decrease in microbial diversity in the caecum, which was not observed in the trickle-infected group. However, an increase in short-chain fatty acid-producing genera in the caecum on day 21 p.i., which shifted to a decrease on day 35 p.i., was common to both groups, possibly related to changes in excretory-secretory products following the parasite's final moult. Faecal microbial interaction networks were more similar between the single-infected and control group than the trickle-infected group. In addition, a lower degree of similarity over time indicated that A. suum trickle infection prevented microbiota stabilization. CONCLUSIONS: These different patterns may have important implications regarding the comparability of experimental infections with natural scenarios characterized by continuous exposure, and should be confirmed by further studies.


Sujet(s)
Ascaridiose , Ascaris suum , Microbiome gastro-intestinal , Maladies des porcs , Suidae , Animaux , Ascaridiose/médecine vétérinaire , ARN ribosomique 16S/génétique , Suidae/microbiologie , Suidae/parasitologie , Maladies des porcs/parasitologie
18.
Braz J Microbiol ; 53(4): 1799-1806, 2022 Dec.
Article de Anglais | MEDLINE | ID: mdl-35984599

RÉSUMÉ

Salmonella enterica serovar Choleraesuis (S. Choleraesuis) is a swine-adapted serovar associated to invasive infections in humans. In Brazil, data of strains of this serovar are scarce. In the present study, six S. Choleraesuis strains of animal (n = 5) and human (n = 1) origin from Brazil were screened for phenotypic antimicrobial resistance using disk-diffusion assay and using whole-genome sequencing data to search for antimicrobial resistance genes, plasmids, prophages, and Salmonella pathogenicity islands (SPIs). Its genetic relatedness was evaluated by MLST and SNP analysis. A single isolate from swine gallbladder harbored the colistin resistance gene mcr-1.1 into a IncX4 plasmid. In the six strains analyzed, resistance was found to tetracycline, nalidixic acid, ciprofloxacin, ampicillin, piperacillin, streptomycin, cefazoline, gentamycin, sulfamethoxazole-trimethoprim, and choloramphenicol, along with resistance genes aac(6')-Iaa, aac(3)-IV, aph(3'')-Ib, aph(6)-Id, aph(4)-Ia, aadA1, aph(3')-IIa, blaTEM-1A, floR, sul1, sul2, tet(B), drfA1, erm(B), mph(B), lnu(G), qacE, and gyrA point mutation Serine83 → Tyrosine and parC Threonine57 → Serine. Furthermore, IncF and IncH plasmids, ten SPIs, and seven prophage types were detected. All strains were assigned to ST145 and five belonged to a common SNP cluster of S. Choleraesuis strains from Brazil. The presence of S. Choleraesuis isolated from animals harboring relevant antimicrobial resistance profiles and virulence determinants reinforced the urge for enhanced surveillance to avoid its transmission to humans through food items.


Sujet(s)
Colistine , Salmonella enterica , Animaux , Antibactériens/pharmacologie , Brésil , Multirésistance bactérienne aux médicaments/génétique , Vésicule biliaire/microbiologie , Génomique , Tests de sensibilité microbienne , Typage par séquençage multilocus , Plasmides/génétique , Salmonella enterica/génétique , Sérogroupe , Suidae/microbiologie
19.
J Glob Antimicrob Resist ; 30: 81-87, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35667645

RÉSUMÉ

OBJECTIVES: To perform the first prospective surveillance evaluating the occurrence of genes encoding colistin resistance, fosfomycin resistance, carbapenemase, or extended-spectrum ß-lactamases (ESBLs) among Enterobacterial isolates recovered from the gut flora of pigs from Egypt. METHODS: Between February and April 2020, 81 rectal swabs were collected from pigs in a slaughterhouse, Cairo, Egypt. Samples were screened for different resistance mechanisms using SuperPolymyxin, ChromID ESBL, SuperFOS, and SuperCarba selective agar plates. Antimicrobial susceptibility testing was performed for all isolates using disk diffusion and broth microdilution techniques. PCR screening was performed for ESBLs, carbapenemases, mcr, and fosA genes. Mating-out assays, multilocus sequence typing analysis, and plasmid typing were also performed. RESULTS: A high prevalence of ESBLs, carbapenemases, fosfomycin, and colistin resistance genes was evidenced among those isolates. The predominant ESBL identified was blaCTX-M-15, followed by blaCTX-M-9. We also identified blaNDM-5 and blaOXA-244. fosA3, fosA4, and fosA6 were identified in E. coli isolates. In addition, 11 MCR-1 producers were recovered. Notably, co-occurrence of ESBL genes and mcr or fosA genes was observed. MLST analysis revealed a high clonal diversity, ruling out the dissemination of one major clone. IncFIB-type was predominantly present among ESBL and FosA producers. The blaNDM-5 gene was carried on an IncX4-type, although the blaOXA-244 gene was chromosomally located. The mcr-1 gene was carried on a diversity of plasmids (IncI2, IncX4, and IncHI2). CONCLUSION: These results raise serious public health concerns as Egyptian pig meat could serve as a reservoir for antimicrobial resistance genes (ARGs), leading to worldwide dissemination.


Sujet(s)
Protéines bactériennes , Colistine , Résistance bactérienne aux médicaments , Protéines Escherichia coli , Escherichia coli , Fosfomycine , Klebsiella pneumoniae , Polymyxines , Suidae , bêta-Lactamases , Animaux , Protéines bactériennes/génétique , Colistine/pharmacologie , Résistance bactérienne aux médicaments/génétique , Égypte , Escherichia coli/effets des médicaments et des substances chimiques , Escherichia coli/génétique , Escherichia coli/isolement et purification , Protéines Escherichia coli/génétique , Fosfomycine/pharmacologie , Klebsiella pneumoniae/effets des médicaments et des substances chimiques , Klebsiella pneumoniae/génétique , Klebsiella pneumoniae/isolement et purification , Viande/microbiologie , Tests de sensibilité microbienne , Typage par séquençage multilocus , Polymyxines/pharmacologie , Études prospectives , Suidae/microbiologie , bêta-Lactamases/génétique
20.
Nature ; 606(7913): 358-367, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35477154

RÉSUMÉ

The composition of the intestinal microbiome varies considerably between individuals and is correlated with health1. Understanding the extent to which, and how, host genetics contributes to this variation is essential yet has proved to be difficult, as few associations have been replicated, particularly in humans2. Here we study the effect of host genotype on the composition of the intestinal microbiota in a large mosaic pig population. We show that, under conditions of exacerbated genetic diversity and environmental uniformity, microbiota composition and the abundance of specific taxa are heritable. We map a quantitative trait locus affecting the abundance of Erysipelotrichaceae species and show that it is caused by a 2.3 kb deletion in the gene encoding N-acetyl-galactosaminyl-transferase that underpins the ABO blood group in humans. We show that this deletion is a ≥3.5-million-year-old trans-species polymorphism under balancing selection. We demonstrate that it decreases the concentrations of N-acetyl-galactosamine in the gut, and thereby reduces the abundance of Erysipelotrichaceae that can import and catabolize N-acetyl-galactosamine. Our results provide very strong evidence for an effect of the host genotype on the abundance of specific bacteria in the intestine combined with insights into the molecular mechanisms that underpin this association. Our data pave the way towards identifying the same effect in rural human populations.


Sujet(s)
Système ABO de groupes sanguins , Acétyl-galactosamine , Microbiome gastro-intestinal , Génotype , Suidae , Système ABO de groupes sanguins/génétique , Acétyl-galactosamine/métabolisme , Animaux , Bactéries/isolement et purification , Microbiome gastro-intestinal/génétique , N-acetylgalactosaminyltransferase/métabolisme , Locus de caractère quantitatif , Suidae/génétique , Suidae/microbiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE