Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.471
Filter
1.
Microb Drug Resist ; 30(9): 372-384, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39250785

ABSTRACT

Objective: This study aimed to evaluate antibiotic susceptibility and antimicrobial resistance trends among clinically significant anaerobes in Kuwait hospitals from 2013 to 2022, comparing these findings with data from 2002 to 2012. Methods: The study prospectively collected 2,317 anaerobic isolates from various body sites across four Kuwaiti hospitals between January 2013 and December 2022. The minimum inhibitory concentrations for 11 antianaerobic antibiotics were determined using E-test methodology. The study analyzed trends and resistance rates across two periods: 2013-2017 and 2018-2022, using statistical analysis for resistance comparison. Results: Of the 2,317 isolates, most were from wounds (42.2%), fluids (28.0%), and tissues (20.5%). Bacteroides fragilis was the most common pathogen (34.0%), followed by Prevotella bivia (13.4%). Over 90% of isolates were susceptible to imipenem, meropenem, tigecycline, and metronidazole, whereas lower susceptibility was observed for penicillin, amoxicillin-clavulanic acid, and clindamycin. Notable differences in resistance profiles since 2002 were observed, especially in amoxicillin-clavulanic acid, piperacillin, piperacillin-tazobactam, and clindamycin. Conclusion: Owing to detected resistance to all antibiotics, susceptibility testing for anaerobic isolates is recommended in severe infections to ensure effective antimicrobial therapy. Continuous surveillance is crucial for developing antibiotic policies to manage invasive anaerobic infections.


Subject(s)
Anti-Bacterial Agents , Bacteria, Anaerobic , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Bacteria, Anaerobic/drug effects , Kuwait/epidemiology , Humans , Prospective Studies , Tigecycline/pharmacology , Drug Resistance, Bacterial , Bacteroides fragilis/drug effects , Bacteroides fragilis/isolation & purification , Metronidazole/pharmacology , Metronidazole/therapeutic use
2.
Carbohydr Polym ; 346: 122661, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39245515

ABSTRACT

Raffinose family oligosaccharides (RFOs) have diverse structures and exhibit various biological activities. When using RFOs as prebiotics, their structures need to be identified. If we first knew whether an RFO was classical or non-classical, structural identification would become much easier. Here, we cloned and expressed an α-galactosidase (BF0224) from Bacteroides fragilis which showed strict specificity for hydrolyzing α-Gal-(1 â†’ 6)-Gal linkages in RFOs. BF0224 efficiently distinguished classical from non-classical RFOs by identifying the resulting hydrolyzed oligo- and mono-saccharides with HPAEC-PAD-MS. Using this strategy, we identified a non-classical RFO from Pseudostellaria heterophylla (Miquel) Pax with DP6 (termed PHO-6), as well as a classical RFO from Lycopus lucidus Turcz. with DP7 (termed LTO-7). To characterize these RFO structures, we employed four other commercial or reported α-galactosidases in combination with NMR and methylation analysis. Using this approach, we elucidated the accurate chemical structure of PHO-6 and LTO-7. Our study provides an efficient analytical approach to structurally analyze RFOs. This enzyme-based strategy also can be applied to structural analysis of other glycans.


Subject(s)
Bacteroides fragilis , Oligosaccharides , Raffinose , alpha-Galactosidase , Bacteroides fragilis/enzymology , alpha-Galactosidase/chemistry , alpha-Galactosidase/metabolism , alpha-Galactosidase/genetics , Raffinose/chemistry , Raffinose/metabolism , Oligosaccharides/chemistry , Hydrolysis
3.
J Diabetes ; 16(8): e13593, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39136533

ABSTRACT

BACKGROUND: We aimed to investigate whether alpha-galactosylceramide (α-GalCer)-producing Bacteroides fragilis could induce natural killer T (NKT) cells in nonobese diabetic (NOD) mice and reduce their diabetes incidence. METHODS: Five-week-old female NOD mice were treated orally with B. fragilis, and islet pathology and diabetes onset were monitored. Immune responses were analyzed by flow cytometry and multiplex technology. Effects of ultraviolet (UV)-killed α-GalCer-producing B. fragilis and their culture medium on invariant NKT (iNKT) cells were tested ex vivo on murine splenocytes, and the immunosuppressive capacity of splenocytes from B. fragilis-treated NOD mice were tested by adoptive transfer to nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. RESULTS: B. fragilis reduced the diabetes incidence from 69% to 33% and the percent of islets with insulitis from 40% to 7%, which doubled the serum insulin level compared with the vehicle-treated control mice. Furthermore, the early treatment reduced proinflammatory mediators in the serum, whereas the proportion of CD4+ NKT cell population was increased by 33%. B. fragilis growth media stimulated iNKT cells and anti-inflammatory M2 macrophages ex vivo in contrast to UV-killed bacteria, which had no effect, strongly indicating an α-GalCer-mediated effect. Adoptive transfer of splenocytes from B. fragilis-treated NOD mice induced a similar diabetes incidence as splenocytes from untreated NOD mice. CONCLUSIONS: B. fragilis induced iNKT cells and M2 macrophages and reduced type 1 diabetes in NOD mice. The protective effect seemed to be more centered on gut-pancreas interactions rather than a systemic immunosuppression. B. fragilis should be considered for probiotic use in individuals at risk of developing type 1 diabetes.


Subject(s)
Bacteroides fragilis , Galactosylceramides , Mice, Inbred NOD , Natural Killer T-Cells , Probiotics , Animals , Female , Galactosylceramides/pharmacology , Probiotics/therapeutic use , Probiotics/pharmacology , Mice , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 1/metabolism , Incidence , Mice, SCID
4.
Microbiologyopen ; 13(4): e1429, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39109824

ABSTRACT

The understanding of how central metabolism and fermentation pathways regulate antimicrobial susceptibility in the anaerobic pathogen Bacteroides fragilis is still incomplete. Our study reveals that B. fragilis encodes two iron-dependent, redox-sensitive regulatory pirin protein genes, pir1 and pir2. The mRNA expression of these genes increases when exposed to oxygen and during growth in iron-limiting conditions. These proteins, Pir1 and Pir2, influence the production of short-chain fatty acids and modify the susceptibility to metronidazole and amixicile, a new inhibitor of pyruvate: ferredoxin oxidoreductase in anaerobes. We have demonstrated that Pir1 and Pir2 interact directly with this oxidoreductase, as confirmed by two-hybrid system assays. Furthermore, structural analysis using AlphaFold2 predicts that Pir1 and Pir2 interact stably with several central metabolism enzymes, including the 2-ketoglutarate:ferredoxin oxidoreductases Kor1AB and Kor2CDAEBG. We used a series of metabolic mutants and electron transport chain inhibitors to demonstrate the extensive impact of bacterial metabolism on metronidazole and amixicile susceptibility. We also show that amixicile is an effective antimicrobial against B. fragilis in an experimental model of intra-abdominal infection. Our investigation led to the discovery that the kor2AEBG genes are essential for growth and have dual functions, including the formation of 2-ketoglutarate via the reverse TCA cycle. However, the metabolic activity that bypasses the function of Kor2AEBG following the addition of phospholipids or fatty acids remains undefined. Overall, our study provides new insights into the central metabolism of B. fragilis and its regulation by pirin proteins, which could be exploited for the development of new narrow-spectrum antimicrobials in the future.


Subject(s)
Anti-Bacterial Agents , Bacteroides fragilis , Metronidazole , Bacteroides fragilis/genetics , Bacteroides fragilis/drug effects , Bacteroides fragilis/enzymology , Bacteroides fragilis/metabolism , Metronidazole/pharmacology , Metronidazole/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Microbial Sensitivity Tests , Gene Expression Regulation, Bacterial
5.
Gut Microbes ; 16(1): 2377570, 2024.
Article in English | MEDLINE | ID: mdl-39034613

ABSTRACT

Recent evidence indicates that repeated antibiotic usage lowers microbial diversity and ultimately changes the gut microbiota community. However, the physiological effects of repeated - but not recent - antibiotic usage on microbiota-mediated mucosal barrier function are largely unknown. By selecting human individuals from the deeply phenotyped Estonian Microbiome Cohort (EstMB), we here utilized human-to-mouse fecal microbiota transplantation to explore long-term impacts of repeated antibiotic use on intestinal mucus function. While a healthy mucus layer protects the intestinal epithelium against infection and inflammation, using ex vivo mucus function analyses of viable colonic tissue explants, we show that microbiota from humans with a history of repeated antibiotic use causes reduced mucus growth rate and increased mucus penetrability compared to healthy controls in the transplanted mice. Moreover, shotgun metagenomic sequencing identified a significantly altered microbiota composition in the antibiotic-shaped microbial community, with known mucus-utilizing bacteria, including Akkermansia muciniphila and Bacteroides fragilis, dominating in the gut. The altered microbiota composition was further characterized by a distinct metabolite profile, which may be caused by differential mucus degradation capacity. Consequently, our proof-of-concept study suggests that long-term antibiotic use in humans can result in an altered microbial community that has reduced capacity to maintain proper mucus function in the gut.


Subject(s)
Anti-Bacterial Agents , Bacteria , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Mucus , Humans , Gastrointestinal Microbiome/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Mice , Mucus/metabolism , Mucus/microbiology , Bacteria/classification , Bacteria/genetics , Bacteria/drug effects , Bacteria/isolation & purification , Bacteria/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Male , Female , Feces/microbiology , Adult , Middle Aged , Akkermansia , Mice, Inbred C57BL , Colon/microbiology , Bacteroides fragilis/drug effects
6.
Gut Microbes ; 16(1): 2379566, 2024.
Article in English | MEDLINE | ID: mdl-39013030

ABSTRACT

Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality in premature infants with no specific treatments available. We aimed to identify the molecular mechanisms underlying NEC and investigate the therapeutic effects of Bacteroides fragilis on NEC. Clinical samples of infant feces, bile acid-targeted metabolomics, pathological staining, bioinformatics analysis, NEC rat model, and co-immunoprecipitation were used to explore the pathogenesis of NEC. Taxonomic characterization of the bile salt hydrolase (bsh) gene, enzyme activity assays, 16S rRNA sequencing, and organoids were used to explore the therapeutic effects of B. fragilis on NEC-related intestinal damage. Clinical samples, NEC rat models, and in vitro experiments revealed that total bile acid increased in the blood but decreased in feces. Moreover, the levels of FXR and other bile acid metabolism-related genes were abnormal, resulting in disordered bile acid metabolism in NEC. Taurochenodeoxycholic acid accelerated NEC pathogenesis and taurodeoxycholate alleviated NEC. B. fragilis displayed bsh genes and enzyme activity and alleviated intestinal damage by restoring gut microbiota dysbiosis and bile acid metabolism abnormalities by inhibiting the FXR-NLRP3 signaling pathway. Our results provide valuable insights into the therapeutic role of B. fragilis in NEC. Administering B. fragilis may substantially alleviate intestinal damage in NEC.


Subject(s)
Amidohydrolases , Bacteroides fragilis , Bile Acids and Salts , Enterocolitis, Necrotizing , Gastrointestinal Microbiome , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Cytoplasmic and Nuclear , Signal Transduction , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/microbiology , Enterocolitis, Necrotizing/drug therapy , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Bacteroides fragilis/metabolism , Bacteroides fragilis/genetics , Signal Transduction/drug effects , Bile Acids and Salts/metabolism , Rats , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Gastrointestinal Microbiome/drug effects , Amidohydrolases/metabolism , Amidohydrolases/genetics , Humans , Rats, Sprague-Dawley , Infant, Newborn , Disease Models, Animal , Male , Female , Probiotics/administration & dosage , Probiotics/pharmacology , Infant, Premature , Dysbiosis/microbiology
7.
Int J Antimicrob Agents ; 64(3): 107241, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38942247

ABSTRACT

OBJECTIVES: Bacteroides fragilis is the most frequent cause of anaerobic bacteraemia. Although recent data suggest a rise in antimicrobial resistance (AMR) of this and other anaerobic bacteria, surveillance remains limited due to a lack of both data availability and comparability. However, a newly introduced standardised method for antimicrobial susceptibility testing (AST) of anaerobic bacteria has made larger scale surveillance possible for the first time. The aim of this study was to investigate phenotypic AMR of Bacteroides fragilis isolates from bacteraemia across Europe in 2022. METHODS: In a multicentre approach, clinical microbiology laboratories in Europe were invited to contribute results of AST for Bacteroides fragilis blood culture isolates (including only the first isolate per patient and year). AST of a selection of four antibiotics was performed locally by participating laboratories in a prospective or retrospective manner, using the new EUCAST disc diffusion method on fastidious anaerobe agar (FAA-HB). RESULTS: A total of 16 European countries reported antimicrobial susceptibilities in 449 unique isolates of Bacteroides fragilis from blood cultures in 2022. Clindamycin demonstrated the highest resistance rates (20.9%, range 0 - 63.6%), followed by piperacillin-tazobactam (11.1%, 0-54.5%), meropenem (13.4%, 0-45.5%), and metronidazole (1.8%, 0-20.0%), all with wide variation between countries. CONCLUSION: Considering that the mean resistance rates across Europe were higher than expected for three of the four anti-anaerobic antibiotics under surveillance, both local AST of clinically relevant isolates of Bacteroides fragilis and continued surveillance on an international level is warranted.


Subject(s)
Anti-Bacterial Agents , Bacteremia , Bacteroides Infections , Bacteroides fragilis , Blood Culture , Microbial Sensitivity Tests , Bacteroides fragilis/drug effects , Bacteroides fragilis/isolation & purification , Humans , Europe , Anti-Bacterial Agents/pharmacology , Bacteroides Infections/microbiology , Bacteremia/microbiology , Retrospective Studies , Prospective Studies , Drug Resistance, Bacterial , Piperacillin, Tazobactam Drug Combination/pharmacology , Clindamycin/pharmacology , Meropenem/pharmacology , Epidemiological Monitoring
8.
J Agric Food Chem ; 72(27): 15345-15356, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38932522

ABSTRACT

The human intestinal mucus layer protects against pathogenic microorganisms and harmful substances, whereas it also provides an important colonization niche for mutualistic microbes. The main functional components of mucus are heavily glycosylated proteins, called mucins. Mucins can be cleaved and utilized by intestinal microbes. The mechanisms between intestinal microbes and the regulation of mucin glycosylation are still poorly understood. In this study, in vitro mucus was produced by HT29-MTX-E12 cells under Semi-Wet interface with Mechanical Stimulation. Cells were exposed to pasteurized nonpathogenic bacteria Akkermansia muciniphila, Ruminococcus gnavus, and Bacteroides fragilis to evaluate influence on glycosylation patterns. Following an optimized protocol, O- and N-glycans were efficiently and reproducibly released, identified, and semiquantified using MALDI-TOF-MS and PGC-LC-MS/MS. Exposure of cells to bacteria demonstrated increased diversity of sialylated O-glycans and increased abundance of high mannose N-glycans in in vitro produced mucus. Furthermore, changes in glycan ratios were observed. It is speculated that bacterial components interact with the enzymatic processes in glycan production and that pasteurized bacteria influence glycosyltransferases or genes involved. These results highlight the influence of pasteurized bacteria on glycosylation patterns, stress the intrinsic relationship between glycosylation and microbiota, and show the potential of using in vitro produced mucus to study glycosylation behavior.


Subject(s)
Gastrointestinal Microbiome , Mucus , Polysaccharides , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Glycosylation , Humans , Tandem Mass Spectrometry/methods , Mucus/microbiology , Mucus/metabolism , Mucus/chemistry , Polysaccharides/metabolism , Polysaccharides/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Mucins/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Bacteria/metabolism , Bacteria/classification , Bacteria/genetics , HT29 Cells , Chromatography, Liquid/methods , Bacteroides fragilis/metabolism , Bacteroides fragilis/chemistry , Bacteroides fragilis/physiology , Pasteurization , Akkermansia/metabolism , Liquid Chromatography-Mass Spectrometry
9.
Gut Microbes ; 16(1): 2363012, 2024.
Article in English | MEDLINE | ID: mdl-38860458

ABSTRACT

The intestinal microbiota is an important environmental factor implicated in CRC development. Intriguingly, modulation of DNA methylation by gut microbiota has been reported in preclinical models, although the relationship between tumor-infiltrating bacteria and CIMP status is currently unexplored. In this study, we investigated tumor-associated bacteria in 203 CRC tumor cases and validated the findings using The Cancer Genome Atlas datasets. We assessed the abundance of Bacteroides fragilis, Escherichia coli, Fusobacterium nucleatum, and Klebsiella pneumoniae through qPCR analysis and observed enrichment of all four bacterial species in CRC samples. Notably, except for E. coli, all exhibited significant enrichment in cases of CIMP. This enrichment was primarily driven by a subset of cases distinguished by high levels of these bacteria, which we labeled as "Superhigh". The bacterial Superhigh status showed a significant association with CIMP (odds ratio 3.1, p-value = 0.013) and with MLH1 methylation (odds ratio 4.2, p-value = 0.0025). In TCGA CRC cases (393 tumor and 45 adj. normal), bacterial taxa information was extracted from non-human whole exome sequencing reads, and the bacterial Superhigh status was similarly associated with CIMP (odds ratio 2.9, p < 0.001) and MLH1 methylation (odds ratio 3.5, p < 0.001). Finally, 16S ribosomal RNA gene sequencing revealed high enrichment of Bergeyella spp. C. concisus, and F. canifelinum in CIMP-Positive tumor cases. Our findings highlight that specific bacterial taxa may influence DNA methylation, particularly in CpG islands, and contribute to the development and progression of CIMP in colorectal cancer.


Subject(s)
Bacteria , Colorectal Neoplasms , CpG Islands , DNA Methylation , Gastrointestinal Microbiome , Humans , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/genetics , Bacteria/genetics , Bacteria/classification , Bacteria/isolation & purification , Female , Male , Middle Aged , Bacteroides fragilis/genetics , Bacteroides fragilis/isolation & purification , Aged , Phenotype
10.
Anaerobe ; 88: 102880, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38942229

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is a significant global health concern, and understanding the role of specific bacterial infections in its development and progression is of increasing interest. This cross-sectional study investigated the associations between Bacteroides fragilis (B. fragilis) and Fusobacterium nucleatum (F. nucleatum) infections and Vietnamese CRC patients. METHODS: 192 patients with either polyps or CRC at varying stages were recruited from May 2017 to December 2020. Real-time PCR assessed infection rates and bacterial loads in CRC tissues. RESULTS: B. fragilis infection was notably higher in CRC tissues (51.6 %) than polyps (9.4 %), with a fivefold higher relative load. Positive associations were found in stages II and III, indicating a fivefold increase in CRC progression risk. F. nucleatum infection rates were significantly higher in CRC tissues (55.2 %) than in polyps (10.5 %). In stage II, the infection rate exceeded that in adjacent tissues. The relative load of F. nucleatum was higher in stage III than in stages I and II. Positive F. nucleatum patients had a 3.2 times higher risk of CRC progression. CONCLUSION: These findings suggest associations between loading of F. nucleatum or/and B. fragilis with the advanced stages of CRC.


Subject(s)
Bacteroides Infections , Bacteroides fragilis , Colorectal Neoplasms , Fusobacterium Infections , Fusobacterium nucleatum , Humans , Colorectal Neoplasms/microbiology , Bacteroides fragilis/isolation & purification , Bacteroides fragilis/genetics , Fusobacterium nucleatum/isolation & purification , Fusobacterium Infections/microbiology , Fusobacterium Infections/epidemiology , Fusobacterium Infections/complications , Male , Female , Vietnam/epidemiology , Middle Aged , Cross-Sectional Studies , Bacteroides Infections/microbiology , Bacteroides Infections/epidemiology , Aged , Adult , Bacterial Load , Southeast Asian People
11.
mSystems ; 9(7): e0051624, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38934546

ABSTRACT

Bacteroides fragilis is a Gram-negative commensal bacterium commonly found in the human colon, which differentiates into two genomospecies termed divisions I and II. Through a comprehensive collection of 694 B. fragilis whole genome sequences, we identify novel features distinguishing these divisions. Our study reveals a distinct geographic distribution with division I strains predominantly found in North America and division II strains in Asia. Additionally, division II strains are more frequently associated with bloodstream infections, suggesting a distinct pathogenic potential. We report differences between the two divisions in gene abundance related to metabolism, virulence, stress response, and colonization strategies. Notably, division II strains harbor more antimicrobial resistance (AMR) genes than division I strains. These findings offer new insights into the functional roles of division I and II strains, indicating specialized niches within the intestine and potential pathogenic roles in extraintestinal sites. IMPORTANCE: Understanding the distinct functions of microbial species in the gut microbiome is crucial for deciphering their impact on human health. Classifying division II strains as Bacteroides fragilis can lead to erroneous associations, as researchers may mistakenly attribute characteristics observed in division II strains to the more extensively studied division I B. fragilis. Our findings underscore the necessity of recognizing these divisions as separate species with distinct functions. We unveil new findings of differential gene prevalence between division I and II strains in genes associated with intestinal colonization and survival strategies, potentially influencing their role as gut commensals and their pathogenicity in extraintestinal sites. Despite the significant niche overlap and colonization patterns between these groups, our study highlights the complex dynamics that govern strain distribution and behavior, emphasizing the need for a nuanced understanding of these microorganisms.


Subject(s)
Bacteroides fragilis , Genetic Variation , Genome, Bacterial , Bacteroides fragilis/genetics , Bacteroides fragilis/pathogenicity , Bacteroides fragilis/isolation & purification , Humans , Genome, Bacterial/genetics , Gastrointestinal Microbiome/genetics , Phylogeny , Bacteroides Infections/microbiology , Whole Genome Sequencing , Drug Resistance, Bacterial/genetics
12.
Microbiol Res ; 286: 127796, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38870618

ABSTRACT

The contribution of commensal microbes to human health and disease is unknown. Bacteroides fragilis (B. fragilis) is an opportunistic pathogen and a common colonizer of the human gut. Nontoxigenic B. fragilis (NTBF) and enterotoxigenic B. fragilis (ETBF) are two kinds of B. fragilis. NTBF has been shown to affect the host immune system and interact with gut microbes and pathogenic microbes. Previous studies indicated that certain strains of B. fragilis have the potential to serve as probiotics, based on their observed relationship with the immune system. However, several recent studies have shown detrimental effects on the host when beneficial gut bacteria are found in the digestive system or elsewhere. In some pathological conditions, NTBF may have adverse reactions. This paper presents a comprehensive analysis of NTBF ecology from the host-microbe perspective, encompassing molecular disease mechanisms analysis, bacteria-bacteria interaction, bacteria-host interaction, and the intricate ecological context of the gut. Our review provides much-needed insights into the precise application of NTBF.


Subject(s)
Bacteroides Infections , Bacteroides fragilis , Gastrointestinal Microbiome , Bacteroides fragilis/genetics , Bacteroides fragilis/pathogenicity , Humans , Bacteroides Infections/microbiology , Probiotics , Animals , Host Microbial Interactions , Host-Pathogen Interactions , Gastrointestinal Tract/microbiology , Symbiosis , Microbial Interactions
13.
Nat Commun ; 15(1): 3872, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719797

ABSTRACT

The gut microbiota and microglia play critical roles in Alzheimer's disease (AD), and elevated Bacteroides is correlated with cerebrospinal fluid amyloid-ß (Aß) and tau levels in AD. We hypothesize that Bacteroides contributes to AD by modulating microglia. Here we show that administering Bacteroides fragilis to APP/PS1-21 mice increases Aß plaques in females, modulates cortical amyloid processing gene expression, and down regulates phagocytosis and protein degradation microglial gene expression. We further show that administering Bacteroides fragilis to aged wild-type male and female mice suppresses microglial uptake of Aß1-42 injected into the hippocampus. Depleting murine Bacteroidota with metronidazole decreases amyloid load in aged 5xFAD mice, and activates microglial pathways related to phagocytosis, cytokine signaling, and lysosomal degradation. Taken together, our study demonstrates that members of the Bacteroidota phylum contribute to AD pathogenesis by suppressing microglia phagocytic function, which leads to impaired Aß clearance and accumulation of amyloid plaques.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Disease Models, Animal , Mice, Transgenic , Microglia , Phagocytosis , Plaque, Amyloid , Animals , Microglia/metabolism , Microglia/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/microbiology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/metabolism , Female , Mice , Male , Bacteroides fragilis/metabolism , Gastrointestinal Microbiome , Humans , Mice, Inbred C57BL , Hippocampus/metabolism , Hippocampus/pathology
14.
Anaerobe ; 88: 102863, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38718918

ABSTRACT

This paper reports a case of Bacteroides fragilis induced spondylitis. Diagnosis was confirmed through blood culture and metagenomic sequencing of pus for pathogen detection. Due to persistent lumbar pain, surgical intervention became imperative, resulting in favorable postoperative outcomes. A detailed patient history revealed a severe episode of oral ulceration two weeks before symptom onset, although a direct link to the infection remained elusive. Leveraging insights from this case, we conducted a comprehensive literature review on B. fragilis spondylitis, elucidating clinical manifestations, diagnostic methodologies, and therapeutic strategies.


Subject(s)
Bacteroides Infections , Bacteroides fragilis , Oral Ulcer , Spondylitis , Humans , Bacteroides fragilis/isolation & purification , Bacteroides fragilis/genetics , Bacteroides Infections/diagnosis , Bacteroides Infections/microbiology , Bacteroides Infections/drug therapy , Spondylitis/microbiology , Spondylitis/diagnosis , Oral Ulcer/microbiology , Oral Ulcer/diagnosis , Male , Anti-Bacterial Agents/therapeutic use , Female , Middle Aged
15.
Gut Microbes ; 16(1): 2350156, 2024.
Article in English | MEDLINE | ID: mdl-38726597

ABSTRACT

Extensive research has explored the role of gut microbiota in colorectal cancer (CRC). Nonetheless, metatranscriptomic studies investigating the in situ functional implications of host-microbe interactions in CRC are scarce. Therefore, we characterized the influence of CRC core pathogens and biofilms on the tumor microenvironment (TME) in 40 CRC, paired normal, and healthy tissue biopsies using fluorescence in situ hybridization (FISH) and dual-RNA sequencing. FISH revealed that Fusobacterium spp. was associated with increased bacterial biomass and inflammatory response in CRC samples. Dual-RNA sequencing demonstrated increased expression of pro-inflammatory cytokines, defensins, matrix-metalloproteases, and immunomodulatory factors in CRC samples with high bacterial activity. In addition, bacterial activity correlated with the infiltration of several immune cell subtypes, including M2 macrophages and regulatory T-cells in CRC samples. Specifically, Bacteroides fragilis and Fusobacterium nucleatum correlated with the infiltration of neutrophils and CD4+ T-cells, respectively. The collective bacterial activity/biomass appeared to exert a more significant influence on the TME than core pathogens, underscoring the intricate interplay between gut microbiota and CRC. These results emphasize how biofilms and core pathogens shape the immune phenotype and TME in CRC while highlighting the need to extend the bacterial scope beyond CRC pathogens to advance our understanding and identify treatment targets.


Subject(s)
Biofilms , Colorectal Neoplasms , Gastrointestinal Microbiome , Tumor Microenvironment , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Humans , Biofilms/growth & development , Tumor Microenvironment/immunology , Male , Female , Bacteria/classification , Bacteria/genetics , Bacteria/immunology , Middle Aged , In Situ Hybridization, Fluorescence , Aged , Fusobacterium nucleatum/immunology , Cytokines/metabolism , Macrophages/immunology , Macrophages/microbiology , Phenotype , Bacteroides fragilis/immunology , Bacteroides fragilis/physiology , Bacteroides fragilis/genetics
16.
Biochem J ; 481(12): 793-804, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38785320

ABSTRACT

CRISPR-Cas systems confer adaptive immunity in prokaryotes, facilitating the recognition and destruction of invasive nucleic acids. Type III CRISPR systems comprise large, multisubunit ribonucleoprotein complexes with a catalytic Cas10 subunit. When activated by the detection of foreign RNA, Cas10 generates nucleotide signalling molecules that elicit an immune response by activating ancillary effector proteins. Among these systems, the Bacteroides fragilis type III CRISPR system was recently shown to produce a novel signal molecule, SAM-AMP, by conjugating ATP and SAM. SAM-AMP regulates a membrane effector of the CorA family to provide immunity. Here, we focus on NYN, a ribonuclease encoded within this system, probing its potential involvement in crRNA maturation. Structural modelling and in vitro ribonuclease assays reveal that NYN displays robust sequence-nonspecific, Mn2+-dependent ssRNA-cleavage activity. Our findings suggest a role for NYN in trimming crRNA intermediates into mature crRNAs, which is necessary for type III CRISPR antiviral defence. This study sheds light on the functional relevance of CRISPR-associated NYN proteins and highlights the complexity of CRISPR-mediated defence strategies in bacteria.


Subject(s)
CRISPR-Cas Systems , RNA, Bacterial , Ribonucleases , RNA, Bacterial/metabolism , RNA, Bacterial/genetics , Ribonucleases/metabolism , Ribonucleases/genetics , Bacteroides fragilis/genetics , Bacteroides fragilis/enzymology , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , RNA Processing, Post-Transcriptional , Models, Molecular
17.
J Antimicrob Chemother ; 79(7): 1683-1687, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38814812

ABSTRACT

BACKGROUND: Bacteroides fragilis, an anaerobic gut bacterium and opportunistic pathogen, comprises two genetically divergent groups (or divisions) at the species level. Differences exist both in the core and accessory genomes and the beta-lactamase genes, with the cephalosporinase gene cepA represented only in division I and the carbapenemase gene cfiA only in division II. METHODS: Multidrug resistance in a clinical B. fragilis strain was examined by whole-genome sequencing. RESULTS: Strain CNM20200260 carried the antimicrobial resistance genes cepA, cfiA2, ant(6'), erm(F), mef(En2), est(T), tet(Q) and cat(A), along with 82-Phe mutation in gyrA (together with 47 amino acid changes in gyrA/B and parC/parE). bexA/B and other efflux pump genes were also observed. None of the detected insertion sequences was located upstream of cfiA2. The genome-based taxonomy coefficients (average nucleotide identity, DNA-DNA hybridization similarity and difference in genomic G + C%) with respect to genomes of the strains of B. fragilis division II and the novel species Bacteroides hominis (both cfiA-positive) met the criteria for CNM20200260 to belong to either species (>95%, >70% and <1%, respectively). No such similarity was seen with type strain NCTC 9343 or the representative genome FDAARGOS 1225 of B. fragilis (division I, cfiA-negative). Strain CNM20200260 harboured four out of nine Kyoto Encyclopedia of Genes and Genomes orthologues defined for division I and one of two defined for division II. CONCLUSIONS: This is the first description of the co-occurrence of cepA and cfiA in a Bacteroides strain, confirming the complexity of the taxonomy of this species.


Subject(s)
Bacterial Proteins , Bacteroides Infections , Bacteroides fragilis , Cephalosporinase , beta-Lactamases , Bacteroides fragilis/genetics , Bacteroides fragilis/enzymology , Bacteroides fragilis/isolation & purification , Bacteroides fragilis/classification , beta-Lactamases/genetics , Bacterial Proteins/genetics , Humans , Cephalosporinase/genetics , Bacteroides Infections/microbiology , Whole Genome Sequencing , Drug Resistance, Multiple, Bacterial/genetics , Anti-Bacterial Agents/pharmacology , Genome, Bacterial , Microbial Sensitivity Tests , Sequence Analysis, DNA
18.
Cell Mol Gastroenterol Hepatol ; 18(2): 101350, 2024.
Article in English | MEDLINE | ID: mdl-38704148

ABSTRACT

BACKGROUND & AIMS: Gut bacterial sphingolipids, primarily produced by Bacteroidetes, have dual roles as bacterial virulence factors and regulators of the host mucosal immune system, including regulatory T cells and invariant natural killer T cells. Patients with inflammatory bowel disease display altered sphingolipids profiles in fecal samples. However, how bacterial sphingolipids modulate mucosal homeostasis and regulate intestinal inflammation remains unclear. METHODS: We used dextran sodium sulfate (DSS)-induced colitis in mice monocolonized with Bacteroides fragilis strains expressing or lacking sphingolipids to assess the influence of bacterial sphingolipids on intestinal inflammation using transcriptional, protein, and cellular analyses. Colonic explant and organoid were used to study the function of bacterial sphingolipids. Host mucosal immune cells and cytokines were profiled and characterized using flow cytometry, enzyme-linked immunosorbent assay, and Western blot, and cytokine function in vivo was investigated by monoclonal antibody injection. RESULTS: B fragilis sphingolipids exacerbated intestinal inflammation. Mice monocolonized with B fragilis lacking sphingolipids exhibited less severe DSS-induced colitis. This amelioration of colitis was associated with increased production of interleukin (IL)-22 by ILC3. Mice colonized with B fragilis lacking sphingolipids following DSS treatment showed enhanced epithelial STAT3 activity, intestinal cell proliferation, and antimicrobial peptide production. Protection against DSS colitis associated with B fragilis lacking sphingolipids was reversed on IL22 blockade. Furthermore, bacterial sphingolipids restricted epithelial IL18 production following DSS treatment and interfered with IL22 production by a subset of ILC3 cells expressing both IL18R and major histocompatibility complex class II. CONCLUSIONS: B fragilis-derived sphingolipids exacerbate mucosal inflammation by impeding epithelial IL18 expression and concomitantly suppressing the production of IL22 by ILC3 cells.


Subject(s)
Bacteroides fragilis , Colitis , Dextran Sulfate , Interleukin-22 , Interleukins , Sphingolipids , Animals , Sphingolipids/metabolism , Interleukins/metabolism , Mice , Colitis/immunology , Colitis/pathology , Colitis/chemically induced , Colitis/microbiology , Colitis/metabolism , Dextran Sulfate/toxicity , Bacteroides fragilis/immunology , Disease Models, Animal , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , STAT3 Transcription Factor/metabolism , Mice, Inbred C57BL
19.
Anal Chem ; 96(22): 9151-9158, 2024 06 04.
Article in English | MEDLINE | ID: mdl-38758019

ABSTRACT

Lipopolysaccharides (LPS) and lipooligosaccharides (LOS) are located in the outer membrane of Gram-negative bacteria and are comprised of three distinctive parts: lipid A, core oligosaccharide (OS), and O-antigen. The structure of each region influences bacterial stability, toxicity, and pathogenesis. Here, we highlight the use of targeted activated-electron photodetachment (a-EPD) tandem mass spectrometry to characterize LPS and LOS from two crucial players in the human gut microbiota, Escherichia coli Nissle and Bacteroides fragilis. a-EPD is a hybrid activation method that uses ultraviolet photoirradiation to generate charge-reduced radical ions followed by collisional activation to produce informative fragmentation patterns. We benchmark the a-EPD method for top-down characterization of triacyl LOS from E. coli R2, then focus on characterization of LPS from E. coli Nissle and B. fragilis. Notably, a-EPD affords extensive fragmentation throughout the backbone of the core OS and O-antigen regions of LPS from E. coli Nissle. This hybrid approach facilitated the elucidation of structural details for LPS from B. fragilis, revealing a putative hexuronic acid (HexA) conjugated to lipid A.


Subject(s)
Escherichia coli , Lipopolysaccharides , Lipopolysaccharides/chemistry , Escherichia coli/chemistry , Bacteroides fragilis/chemistry , Electrons , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL