Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41.181
Filter
1.
Cancer Med ; 13(17): e70180, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39234654

ABSTRACT

BACKGROUND: Gut bacteria are related to colorectal cancer (CRC) and its clinicopathologic characteristics. OBJECTIVE: To develop gut bacterial subtypes and explore potential microbial targets for CRC. METHODS: Stool samples from 914 volunteers (376 CRCs, 363 advanced adenomas, and 175 normal controls) were included for 16S rRNA sequencing. Unsupervised learning was used to generate gut microbial subtypes. Gut bacterial community composition and clustering effects were plotted. Differences of gut bacterial abundance were analyzed. Then, the association of CRC-associated bacteria with subtypes and the association of gut bacteria with clinical information were assessed. The CatBoost models based on gut differential bacteria were constructed to identify the diseases including CRC and advanced adenoma (AA). RESULTS: Four gut microbial subtypes (A, B, C, D) were finally obtained via unsupervised learning. The characteristic bacteria of each subtype were Escherichia-Shigella in subtype A, Streptococcus in subtype B, Blautia in subtype C, and Bacteroides in subtype D. Clinical information (e.g., free fatty acids and total cholesterol) and CRC pathological information (e.g., tumor depth) varied among gut microbial subtypes. Bacilli, Lactobacillales, etc., were positively correlated with subtype B. Positive correlation of Blautia, Lachnospiraceae, etc., with subtype C and negative correlation of Coriobacteriia, Coriobacteriales, etc., with subtype D were found. Finally, the predictive ability of CatBoost models for CRC identification was improved based on gut microbial subtypes. CONCLUSION: Gut microbial subtypes provide characteristic gut bacteria and are expected to contribute to the diagnosis of CRC.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , RNA, Ribosomal, 16S , Humans , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Male , Female , RNA, Ribosomal, 16S/genetics , Middle Aged , Feces/microbiology , Adenoma/microbiology , Adenoma/pathology , Aged , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Case-Control Studies
2.
Gut Microbes ; 16(1): 2394249, 2024.
Article in English | MEDLINE | ID: mdl-39224018

ABSTRACT

Inflammatory bowel diseases (IBD) etiology is multifactorial. Luminal microRNAs (miRNAs) have been suspected to play a role in the promotion of chronic inflammation, but the extent to which fecal miRNAs are interacting with the intestinal ecosystem in a way that contribute to diseases, including IBD, remains unknown. Here, fecal let-7b and miR-21 were found elevated, associated with inflammation, and correlating with multiple bacteria in IBD patients and IL-10-/- mice, model of spontaneous colitis. Using an in vitro microbiota modeling system, we revealed that these two miRNAs can directly modify the composition and function of complex human microbiota, increasing their proinflammatory potential. In vivo investigations revealed that luminal increase of let-7b drastically alters the intestinal microbiota and enhances macrophages' associated proinflammatory cytokines (TNF, IL-6, and IL-1ß). Such proinflammatory effects are resilient and dependent on the bacterial presence. Moreover, we identified that besides impairing the intestinal barrier function, miR-21 increases myeloperoxidase and antimicrobial peptides secretion, causing intestinal dysbiosis. More importantly, in vivo inhibition of let-7b and miR-21 with anti-miRNAs significantly improved the intestinal mucosal barrier function and promoted a healthier host-microbiota interaction in the intestinal lining, which altogether conferred protection against colitis. In summary, we provide evidence of the functional significance of fecal miRNAs in host-microbiota communication, highlighting their therapeutic potential in intestinal inflammation and dysbiosis-related conditions, such as IBD.


Subject(s)
Colitis , Feces , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Humans , Feces/microbiology , Mice , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Colitis/microbiology , Colitis/chemically induced , Colitis/genetics , Inflammation/microbiology , Inflammation/metabolism , Dysbiosis/microbiology , Mice, Inbred C57BL , Female , Mice, Knockout , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Male , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Cytokines/metabolism , Macrophages/immunology , Macrophages/microbiology , Macrophages/metabolism , Disease Models, Animal , Interleukin-10/genetics , Interleukin-10/metabolism
3.
Sci Rep ; 14(1): 20607, 2024 09 04.
Article in English | MEDLINE | ID: mdl-39232075

ABSTRACT

Biofilm formation and toxin production are some of the virulence factors of Clostridioides difficile (C. difficile), which causes hospital-acquired C. difficile infection (HA-CDI). This work investigated the prevalence and distribution of different strains recovered from HA-CDI patients hospitalized in 4 medical centres across Israel, and characterized strains' virulence factors and antibiotic susceptibility. One-hundred and eighty-eight faecal samples were collected. C. difficile 's toxins were detected by the CerTest Clostridium difficile GDH + Toxin A + B combo card test kit. Toxin loci PaLoc and PaCdt were detected by whole-genome sequencing (WGS). Multi-locus sequence typing (MLST) was performed to classify strains. Biofilm production was assessed by crystal violet. Antibiotic susceptibility was determined using Etest. Fidaxomicin susceptibility was tested via agar dilution. Sequence type (ST) 42 was the most (13.8%) common strain. All strains harboured the 2 toxins genes; 6.9% had the binary toxin. Most isolates were susceptible to metronidazole (98.9%) and vancomycin (99.5%). Eleven (5.85%) isolates were fidaxomicin-resistant. Biofilm production capacity was associated with ST (p < 0.001). In conclusion, a broad variety of C. difficile strains circulate in Israel's medical centres. Further studies are needed to explore the differences and their contribution to HA-CDI epidemiology.


Subject(s)
Anti-Bacterial Agents , Biofilms , Clostridioides difficile , Clostridium Infections , Cross Infection , Microbial Sensitivity Tests , Virulence Factors , Clostridioides difficile/genetics , Clostridioides difficile/drug effects , Clostridioides difficile/isolation & purification , Clostridioides difficile/pathogenicity , Humans , Israel/epidemiology , Clostridium Infections/microbiology , Clostridium Infections/epidemiology , Anti-Bacterial Agents/pharmacology , Virulence Factors/genetics , Male , Female , Biofilms/drug effects , Biofilms/growth & development , Cross Infection/microbiology , Cross Infection/epidemiology , Aged , Middle Aged , Multilocus Sequence Typing , Adult , Aged, 80 and over , Whole Genome Sequencing , Feces/microbiology
4.
Sci Rep ; 14(1): 21711, 2024 09 17.
Article in English | MEDLINE | ID: mdl-39289419

ABSTRACT

Following bowel surgery, infectious complications, including anastomotic leak (AL), remain major sources of morbidity and mortality. Bowel preparation is often administered with the assumption that gut decontamination reduces post-surgical complications. In this study, we tested this hypothesis using a murine model of colon surgery. The mice were fed either regular chow or a high-fat, high-sugar Western diet. The day before surgery, the mice received one of four interventions: water (control), mechanical bowel preparation (MBP), oral antibiotics (OA), or both MBP and OA. We found no differences in the rates of AL among the experimental groups, and diet did not appear to affect the outcomes. Exploratory analyses showed changes in the gut microbiome consistent with the different treatments, but investigations of fecal short-chain fatty acids and RNA sequencing of colonic tissue did not reveal specific effects of the treatments or the presence of AL. However, we did identify bacterial genera that may be causally associated with AL and developed a predictive index from stool samples as a marker for the presence of AL. Future research is needed to identify and validate a microbial predictive tool and to uncover the microbial-driven mechanisms that lead to AL.


Subject(s)
Anastomotic Leak , Gastrointestinal Microbiome , Animals , Anastomotic Leak/etiology , Anastomotic Leak/microbiology , Anastomotic Leak/prevention & control , Gastrointestinal Microbiome/drug effects , Mice , Feces/microbiology , Colon/microbiology , Colon/surgery , Male , Mice, Inbred C57BL , Anti-Bacterial Agents/pharmacology , Fatty Acids, Volatile/metabolism , Fatty Acids, Volatile/analysis , Disease Models, Animal
5.
BMC Microbiol ; 24(1): 350, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39289612

ABSTRACT

Diarrheal diseases remain the leading cause of high mortality among the infants, particularly in the developing countries; Probiotic intervention for diarrhea has been an ongoing novel approach to diarrheal prevention and treatment. This study aims to characterize immunogenic and probiotic properties of lactic acid bacteria (LAB) isolated from human breast milk and neonates' faeces. The LAB isolates from 16 mothers' breast milk and 13 infants' faeces were screened and identified by 16 S rRNA gene partial sequencing. Their antimicrobial activities against 5 strains of diarrheagenic Escherichia coli were tested. Organic acids production was quantified by HPLC, and antibiotic resistance pattern were determined by VITEK®. Autoaggregation, co-aggregation and hydrophobicity properties were assessed by UV spectrophotometry and immunomodulatory effect was determined in mouse model. Ninety-three LAB of five genera were identified. The most abundant species was Lactiplantibacillus plantarum with inhibition zones ranged from 8.0 to 25.0 ± 1 mm. Lacticaseibacillus rhamnosus A012 had 76.8 mg/mL lactic acid, (the highest concentration), was susceptible to all antibiotics tested. L. plantarum A011 and L. rhamnosus A012 were highly resistance to gastrointestinal conditions. L. rhamnosus A012 produced hydrophobicity of 25.01% (n-hexadecane), 15.4% (xylene) and its autoaggregation was 32.52%. L. rhamnosus A012 and L. plantarum A011 exert immunomodulatory effects on the cyclophosphamide-treated mice by upregulating anti-inflammatory cytokine and downregulating proinflammatory cytokines. Lactobacillus sp. demonstrated good probiotic and immunomodulatory properties. Further works are ongoing on the practical use of the strains.


Subject(s)
Diarrhea , Escherichia coli , Feces , Lactobacillales , Milk, Human , Probiotics , Probiotics/pharmacology , Humans , Feces/microbiology , Animals , Female , Milk, Human/microbiology , Milk, Human/immunology , Mice , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/immunology , Lactobacillales/isolation & purification , Lactobacillales/physiology , Lactobacillales/classification , Diarrhea/microbiology , Diarrhea/prevention & control , Escherichia coli Infections/microbiology , Escherichia coli Infections/prevention & control , Escherichia coli Infections/veterinary , Infant , RNA, Ribosomal, 16S/genetics , Anti-Bacterial Agents/pharmacology , Infant, Newborn , Adult , Microbial Sensitivity Tests
6.
Integr Cancer Ther ; 23: 15347354241280390, 2024.
Article in English | MEDLINE | ID: mdl-39313964

ABSTRACT

BACKGROUND: In patients with metastatic colorectal cancer (mCRC), Quxie Cap-sule (QX)-a combination of conventional therapy (including chemotherapy, targeted therapy or supportive care)-has shown a significant overall survival benefit compared with placebo and might have the property of dual effects of antitumor and immunity enhancement, both mediated by the microbiome. In preclinical models, QX has also shown activity against colorectal cancer. This study aimed to describe how the aforementioned effects of QX look after when focusing on the patients in third or above line setting. METHODS: A Simon's Minimax two-stage phase II design was used in this study, which enrolled mCRC patients who progressed after second-line treatment. Patients received conventional therapy plus QX until disease progression or unacceptable toxicity. Before and after 1-month intervention, we collected patients' stool samples for microbiome analysis by 16s rRNA sequencing approaches. And the microbiome analysis before and after 1-month intervention was done through bioinformation analysis platform. RESULTS: Fifteen patients were enrolled and gut microbiome were analyzed from 7 of 10 patients that with PFS over 3.7 months. Microbiome community analysis on genus level showed that the proportion of Lachnospiraceae_UCG-001 (0.04% vs 1.06%, P = .02249) significantly increased after conventional therapy plus QX while the proportion of Alistipes (2.96% vs 1.35%, P = .03461), Flavonifractor (0.04% vs 0.02%, P = .02249), Bifidobacterium (6.11% vs 1.14%, P = .02249) and Butyricimonas (0.24% vs 0.11%, P = .03603) significantly decreased after intervention . LEfSe analysis showed that after intervention, samples were highly related with unclassified-f-lachnospiraceae, Eubacterium and Lachnospiraceae_UCG-001. CONCLUSIONS: Decrease of gut bacteria with potential roles in carcinogenesis of colorectal cancer and increase in the abundance of gut anticancer bacteria such as Lachnospiraceae may partly explain how conventional therapy combined with QX can influence carcinogenesis and tumor progression in colon cancer. TRIAL REGISTRATION: Chinese Clinical Trial Registry (ChiCTR2100053874).


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Male , Female , Middle Aged , Aged , Adult , Neoplasm Metastasis , Capsules , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/pharmacology , Feces/microbiology
7.
Sci Rep ; 14(1): 21746, 2024 09 18.
Article in English | MEDLINE | ID: mdl-39294269

ABSTRACT

The aim of this study was to investigate the vertical transfer of microbiota from dams to the offspring. We studied a pair of 20 dams and its offspring. Maternal sources (colostrum, feces and vaginal secretion) and newborn fecal samples were analyzed using 16S rDNA amplicon sequencing on days 1, 3, 7, 14 and 28. Overall, newborns were maintained healthy and did not receive antimicrobial therapy. The Source Tracker analysis indicated that the newborn fecal microbiota was similar to colostrum and vaginal secretion from day 1 up to 7. However, an unknown source (probably from the environment) showed a gradual increase in its similarity with fecal samples from calves measured from day 3 to 28. The most abundant bacteria groups on meconium (day 1) and calf fecal samples on day 3 were Escherichia-Shigella and Clostridium, respectively. On day 7, the predominant genus were Bifidobacterium and Lactobacillus, while Fusobacterium was the most abundant genus on day 14, coinciding with the diarrhea peak. Faecalibacterium showed a gradual increase throughout the neonatal period. Maternal sources contribute to the neonatal microbiota, however other unknown sources (probably environment) had a strong influence on development of the gut microbiota later in the neonate period.


Subject(s)
Animals, Newborn , Colostrum , Feces , Gastrointestinal Microbiome , Animals , Gastrointestinal Microbiome/genetics , Cattle , Female , Feces/microbiology , Colostrum/microbiology , RNA, Ribosomal, 16S/genetics , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Pregnancy , Vagina/microbiology , Meconium/microbiology
8.
Appl Microbiol Biotechnol ; 108(1): 469, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39298023

ABSTRACT

Prior research has indicated that the gut-lung-axis can be influenced by the intestinal microbiota, thereby impacting lung immunity. Rifaximin is a broad-spectrum antibacterial drug that can maintain the homeostasis of intestinal microflora. In this study, we established an influenza A virus (IAV)-infected mice model with or without rifaximin supplementation to investigate whether rifaximin could ameliorate lung injury induced by IAV and explore the molecular mechanism involved. Our results showed that IAV caused significant weight loss and disrupted the structure of the lung and intestine. The analysis results of 16S rRNA and metabolomics indicated a notable reduction in the levels of probiotics Lachnoclostridium, Ruminococcaceae_UCG-013, and tryptophan metabolites in the fecal samples of mice infected with IAV. In contrast, supplementation with 50 mg/kg rifaximin reversed these changes, including promoting the repair of the lung barrier and increasing the abundance of Muribaculum, Papillibacter and tryptophan-related metabolites content in the feces. Additionally, rifaximin treatment increased ILC3 cell numbers, IL-22 level, and the expression of RORγ and STAT-3 protein in the lung. Furthermore, our findings demonstrated that the administration of rifaximin can mitigate damage to the intestinal barrier while enhancing the expression of AHR, IDO-1, and tight junction proteins in the small intestine. Overall, our results provided that rifaximin alleviated the imbalance in gut microbiota homeostasis induced by IAV infection and promoted the production of tryptophan-related metabolites. Tryptophan functions as a signal to facilitate the activation and movement of ILC3 cells from the intestine to the lung through the AHR/STAT3/IL-22 pathway, thereby aiding in the restoration of the barrier. KEY POINTS: • Rifaximin ameliorated IAV infection-caused lung barrier injury and induced ILC3 cell activation. • Rifaximin alleviated IAV-induced gut dysbiosis and recovered tryptophan metabolism. • Tryptophan mediates rifaximin-induced ILC3 cell activation via the AHR/STAT3/IL-22 pathway.


Subject(s)
Gastrointestinal Microbiome , Influenza A virus , Lung , Orthomyxoviridae Infections , Rifaximin , Animals , Gastrointestinal Microbiome/drug effects , Rifaximin/therapeutic use , Mice , Lung/microbiology , Lung/drug effects , Orthomyxoviridae Infections/drug therapy , Influenza A virus/drug effects , Disease Models, Animal , RNA, Ribosomal, 16S/genetics , Interleukins/metabolism , Interleukins/genetics , Interleukin-22 , Mice, Inbred C57BL , Anti-Bacterial Agents/pharmacology , STAT3 Transcription Factor/metabolism , Feces/microbiology , Tryptophan/metabolism , Lung Injury/drug therapy , Probiotics/administration & dosage , Probiotics/pharmacology
9.
Sci Rep ; 14(1): 21924, 2024 09 20.
Article in English | MEDLINE | ID: mdl-39300177

ABSTRACT

Emerging research on the microbiome highlights the significant role of gut health in the development of kidney stones, indicating that an imbalance in gut bacteria or dysbiosis can influence the formation of stones by altering oxalate metabolism and urinary metabolite profiles. In particular, the overabundance of specific bacteria such as Enterococcus and Oxalobacter spp., which are known to affect oxalate absorption, is observed in patients with urolithiasis. This study investigates the effects of gut dysbiosis on urolithiasis through fecal microbiota transplantation (FMT) from patients to rats and its impact on urinary mineral excretion and stone formation. Fecal samples from eight patients with calcium oxalate stones and ten healthy volunteers were collected to assess the gut microbiome. These samples were then transplanted to antibiotic-pretreated Wistar rats for a duration of four weeks. After transplantation, we evaluated changes in the fecal gut microbiome profile, urinary mineral excretion rates, and expression levels of intestinal zonula occluden-1 (ZO-1), SLC26A6 and renal NF-κB. In humans, patients with urolithiasis exhibited increased urinary calcium and oxalate levels, along with decreased citrate excretion and increased urinary supersaturation index. The fecal microbiota showed a notable abundance of Bacteroidota. In rodents, urolithiasis-FMT rats showed urinary disturbances similar to patients, including elevated pH, oxalate level, and supersaturation index, despite negative renal pathology. In addition, a slight elevation in the expression of renal NF-κB, a significant intestinal SLC26A6, and a reduction in ZO-1 expression were observed. The gut microbiome of urolithiasis-FMT rats showed an increased abundance of Bacteroidota, particularly Muribaculaceae, compared to their healthy FMT counterparts. In conclusion, the consistent overabundance of Bacteroidota in both urolithiasis patients and urolithiasis-FMT rats is related to altered intestinal barrier function, hyperoxaluria, and renal inflammation. These findings suggest that gut dysbiosis, characterized by an overgrowth of Bacteroidota, plays a crucial role in the pathogenesis of calcium oxalate urolithiasis, underscoring the potential of targeting the gut microbiota as a therapeutic strategy.


Subject(s)
Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Kidney Calculi , Rats, Wistar , Animals , Kidney Calculi/microbiology , Kidney Calculi/metabolism , Kidney Calculi/therapy , Humans , Rats , Male , Dysbiosis/microbiology , Disease Models, Animal , Feces/microbiology , Female , Adult , Middle Aged
10.
Microbiome ; 12(1): 178, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300575

ABSTRACT

BACKGROUND: Microbial pdu and cob-cbi-hem gene clusters encode the key enzyme glycerol/diol dehydratase (PduCDE), which mediates the transformation of dietary nutrients glycerol and 1,2-propanediol (1,2-PD) to a variety of metabolites, and enzymes for cobalamin synthesis, a co-factor and shared good of microbial communities. It was the aim of this study to relate pdu as a multipurpose functional trait to environmental conditions and microbial community composition. We collected fecal samples from wild animal species living in captivity with different gut physiology and diet (n = 55, in total 104 samples), determined occurrence and diversity of pdu and cob-cbi-hem using a novel approach combining metagenomics with quantification of metabolic and genetic biomarkers, and conducted in vitro fermentations to test for trait-based activity. RESULTS: Fecal levels of the glycerol transformation product 1,3-propanediol (1,3-PD) were higher in hindgut than foregut fermenters. Gene-based analyses indicated that pduC harboring taxa are common feature of captive wild animal fecal microbiota that occur more frequently and at higher abundance in hindgut fermenters. Phylogenetic analysis of genomes reconstructed from metagenomic sequences identified captive wild animal fecal microbiota as taxonomically rich with a total of 4150 species and > 1800 novel species but pointed at only 56 species that at least partially harbored pdu and cbi-cob-hem. While taxonomic diversity was highest in fecal samples of foregut-fermenting herbivores, higher pduC abundance and higher diversity of pdu/cbi-cob-hem related to higher potential for glycerol and 1,2-PD utilization of the less diverse microbiota of hindgut-fermenting carnivores in vitro. CONCLUSION: Our approach combining metabolite and gene biomarker analysis with metagenomics and phenotypic characterization identified Pdu as a common function of fecal microbiota of captive wild animals shared by few taxa and stratified the potential of fecal microbiota for glycerol/1,2-PD utilization and cobalamin synthesis depending on diet and physiology of the host. This trait-based study suggests that the ability to utilize glycerol/1,2-PD is a key function of hindgut-fermenting carnivores, which does not relate to overall community diversity but links to the potential for cobalamin formation. Video Abstract.


Subject(s)
Feces , Fermentation , Gastrointestinal Microbiome , Glycerol , Metagenomics , Animals , Feces/microbiology , Glycerol/metabolism , Metagenomics/methods , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Propylene Glycols/metabolism , Vitamin B 12/metabolism , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/enzymology , Phylogeny , Animals, Wild/microbiology
11.
PLoS One ; 19(9): e0310648, 2024.
Article in English | MEDLINE | ID: mdl-39302919

ABSTRACT

Ruminant livestock are major contributors to anthropogenic methane emissions in the United States and worldwide. Enteric methane is generated by methanogenic archaea residing in ruminant digestive tracts. Information on when methanogens colonize the gut and when they begin to interact with bacteria during the early phases of the ruminant life cycle is less explored. The objectives of this study were (i) to investigate the composition of the methanogenic archaeal community at birth and through the weaning transition and (ii) to determine if and when the methanogenic archaea begin to interact with bacteria in the lower gut of neonatal dairy calves. Ten female Holstein calves (approximately 45kg birth weight) were enrolled in the study. Fecal samples were collected every two weeks (Wk 2, 4, 6, 8, 10, and 12) between birth and weaning and analyzed for methanogenic archaeal diversity via 16S rRNA amplicon sequencing and quantitative real-time PCR (RT-qPCR). Estimates of alpha diversity (Observed species, and Shannon diversity index) and beta diversity (weighted and unweighted UniFrac distances) showed significant differences (P < 0.05) between archaeal communities across timepoints. Both 16S rRNA amplicon sequencing and RT-qPCR analyses revealed Methanobrevibacter was the most prevalent genus at Wk2, Wk4, and Wk6, whereas Methanosphaera gradually increased with time and was most abundant at Wk10 and Wk12. Correlation analysis revealed that Methanobrevibacter and Methanosphaera were inversely correlated with each other and formed distinct cohorts with specific bacterial lineages similar to those reported in the mature rumen, thus revealing that these associations are established during the preweaning period. Therefore, the preweaning period presents a window of opportunity to interfere with early-life methanogenic colonization with the ultimate goal of reducing enteric methane emissions without perturbing ruminal function later in the life of dairy cattle.


Subject(s)
Methane , RNA, Ribosomal, 16S , Weaning , Animals , Cattle/microbiology , Female , Methane/metabolism , RNA, Ribosomal, 16S/genetics , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Gastrointestinal Microbiome , Feces/microbiology , Archaea/genetics , Archaea/classification , Animals, Newborn/microbiology
12.
FASEB J ; 38(18): e70065, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39305117

ABSTRACT

One in six people are projected to be 65 years or older by 2050. As the population ages, better treatments for injuries that disproportionately impact the aged population will be needed. Clinical studies show that people aged 65 and older experience higher rates of morbidity and mortality after burn injury, including a greater incidence of pulmonary complications when compared to younger burn injured adults, which we and others believe is mediated, in part, by inflammation originating in the intestines. Herein, we use our clinically relevant model of scald burn injury in young and aged mice to determine whether cohousing aged mice with young mice or giving aged mice oral gavage of fecal material from young mice is sufficient to alter the microbiome of the aged mice and protect them from inflammation in the ileum and the lungs. Aged burn injured mice have less DNA expression of Bacteroidetes in the feces and an unhealthy Firmicutes/Bacteroidetes ratio. Both Bacteroidetes and the ratio of these two phyla are restored in aged burn injured by prior cohousing for a month with younger mice but not fecal transfer from young mice. This shift in the microbiome coincides with heightened expression of danger-associated molecular patterns (DAMP), and pro-inflammatory cytokine interleukin-6 (il6) in the ileum and lung of aged, burn injured mice, and heightened antimicrobial peptide camp in the lung. Cohousing reverses DAMP expression in the ileum and lung, and cathelicidin-related antimicrobial peptide protein (camp) in the lung, while fecal transfer heightened DAMPs while reducing camp in the lung, and also increased IL-6 protein in the lungs. These results highlight the importance of the intestinal microbiome in mediating inflammation within the gut-lung axis, giving insights into potential future treatments in the clinic.


Subject(s)
Burns , Gastrointestinal Microbiome , Inflammation , Animals , Burns/microbiology , Mice , Inflammation/microbiology , Mice, Inbred C57BL , Male , Aging , Feces/microbiology , Lung/microbiology , Lung/metabolism , Lung/pathology , Fecal Microbiota Transplantation , Bacteroidetes , Ileum/microbiology , Ileum/metabolism
13.
BMC Infect Dis ; 24(1): 928, 2024 Sep 08.
Article in English | MEDLINE | ID: mdl-39245731

ABSTRACT

INTRODUCTION: Diarrhoea is a major public health concern in developing countries, usually exacerbated by poor water, sanitation and hygiene but its aetiology is under-studied, particularly away from capital cities. We identified diarrhoeagenic Escherichia coli (DEC) from stools collected in Ile-Ife and Ilesa, Osun state, Nigeria and determined their antibiotic resistance profiles. METHODS: Stool samples from 167 children with diarrhoea and 334 controls under the age of 5 years were cultured for Escherichia coli and Salmonella. Bacterial isolates were identified biochemically and DEC were identified by PCR. Antimicrobial susceptibility testing was by modified Kirby-Bauer disc diffusion method in accordance with the CLSI guidelines. Data were analyzed using Chi-square and Fisher's exact tests. RESULT: Diarrhoea infection is significantly high among children under 12 months (p = 0.002), caregivers without at least primary school education (p = 0.006), breastfeeding for under 6 months (p˂0.001), and caregivers who were siblings (p = 0.004). DEC was detected in 69(41.3%) cases but only 86(25.7%) controls (p < 0.001) and more commonly recovered during the wet season (p < 0.001). Enterotoxigenic E. coli (p = 0.031), enteropathogenic E. coli (p = 0.031) and Shiga-toxin-producing E. coli (p = 0.044) were recovered more commonly from cases than controls. DEC from patients with diarrhoea were commonly resistant to sulphonamides (91.3%), trimethoprim (82.6%), and ampicillin (78.3%) but were largely susceptible to quinolones and carbapenems (97.1%). CONCLUSION: Enteropathogenic, enterotoxigenic and Shiga toxin-producing E. coli are associated with diarrhoea in our setting, and show considerable resistance to first-line antimicrobials. Risk factors for DEC diarrhoea include infancy, inadequate breastfeeding and caregivers with education below primary school.


Subject(s)
Anti-Bacterial Agents , Diarrhea , Escherichia coli Infections , Escherichia coli , Humans , Nigeria/epidemiology , Diarrhea/microbiology , Diarrhea/epidemiology , Infant , Female , Child, Preschool , Male , Escherichia coli Infections/microbiology , Escherichia coli Infections/epidemiology , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/isolation & purification , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Feces/microbiology , Microbial Sensitivity Tests , Infant, Newborn , Risk Factors , Drug Resistance, Bacterial
14.
BMC Microbiol ; 24(1): 357, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39304799

ABSTRACT

BACKGROUND: Antibiotic-resistant Salmonella is one of the main public health concerns in the world. Isolation of Salmonella in abattoirs has been considered the core source of infection in the community from meat. Still, there is limited information on the contamination rate of cattle carcasses. OBJECTIVE: This study aimed to document the occurrence and antimicrobial susceptibility profile of Salmonella species recovered from cattle carcass and abattoir personnel at Dessie, municipality abattoir, Northeast Ethiopia: METHODS: A total of 336 carcass swabs of abdomen, neck, and hind limb from cattle carcasses and 24 stool samples were collected from abattoir personnel using a systematic sampling method from February to April 2019. The collected samples were transported using Cary-Blair transport media and cultivated on Selenite cysteine F-broth, Brilliant green agar, and Xylose-lysine deoxycholate agar plates to isolate Salmonella species. Gram stain, colony morphology, and biochemical tests were performed to identify the isolated bacteria. An antimicrobial susceptibility test for Salmonella was performed using the Kirby-Bauer Disc Diffusion method. Descriptive statistics; both bivariable and multivariable logistic regression analysis was performed using SPSS version 25 software. P-value < 0.05 at 95% CI was considered statistically significant. RESULTS: The prevalence of salmonella species was 8%(27/336) from all samples.'The prevalence of Salmonella isolates in cattle carcass and abattoir personnel was 8%(25/312) and 8.3%(2/24) respectively. The antimicrobial test showed that Salmonella species were 100% resistant to ampicillin, 59.3% to trimethoprim-sulfamethoxazole, 59.3% to tetracycline, and 55.6% to amoxicillin/clavulanate. From the total antimicrobial tested bacteria, 81.5%(22/27) were resistant to three and above classes of antibiotics (drug classes). Unwashed knives, carcasses, and hands of butchers during slaughtering were significantly associated (p < 0.05) with Salmonella found in carcasses. CONCLUSIONS: Salmonella isolation rates from cattle carcasses were high, with the bacteria showing notable resistance to most tested antibiotics. Poor hygiene practices, unsanitized equipment, and unhygienic beef processing were contributing factors.


Subject(s)
Abattoirs , Anti-Bacterial Agents , Microbial Sensitivity Tests , Salmonella , Animals , Cattle , Ethiopia , Salmonella/drug effects , Salmonella/isolation & purification , Salmonella/classification , Anti-Bacterial Agents/pharmacology , Humans , Feces/microbiology , Meat/microbiology
15.
BMC Microbiol ; 24(1): 359, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39304810

ABSTRACT

BACKGROUND: Studies have found dysbiosis of the gut microbiota in individuals infected with the hepatitis B virus (HBV). Tenofovir dipivoxil (TDF) is one of the preferred oral antiviral drugs used for the treatment of chronic hepatitis B (CHB), but the extent to which TDF is able to affect the gut microbiota and inflammatory factors of a patient remains largely unexplored. In this study, we collected stool samples from HBV patients prior to medication and from CHB patients treated with TDF. RESULTS: The gut microbiota and inflammatory factors were assessed in 42 healthy subjects (HC group), 109 HBV-infected subjects, including 48 CHB patients who were not medicated with nucleoside analogue drugs (No-NAs group), and 61 CHB patients who were medicated with TDF (TDF group). 16 S rRNA sequencing revealed that TDF treatment caused significant changes in the gut microbiota of HBV-infected individuals; however, the gut microbiota of HBV-infected individuals did not fully recover to a pre-dysbiosis state. The relative abundance of Bacteroidota gradually decreased from the HC group to the No-NAs and TDF groups. The relative abundance of Fusobacteriota was significantly higher in the No-NAs group than in the HC group. At the genus level, Dialister, Eubacterium_hallii_group, Halomonas, Collinsella, Sphingomonas, Xanthomonadaceae_unclassified, and Rhizobiaceae_unclassified were overrepresented; while the abundance of Bacteroides and Fusobacterium decreased significantly in the No-NAs and TDF groups. CONCLUSIONS: This study showed that TDF treatment significantly improved the regulation of the gut microbiota and aided in dysbiosis recovery. We did not observe significant improvement in serum inflammatory factor concentrations, which may be related to the relatively short duration of TDF administration in this study.


Subject(s)
Antiviral Agents , Bacteria , Dysbiosis , Feces , Gastrointestinal Microbiome , Hepatitis B, Chronic , Tenofovir , Humans , Dysbiosis/microbiology , Gastrointestinal Microbiome/drug effects , Tenofovir/therapeutic use , Tenofovir/administration & dosage , Male , Female , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/virology , Hepatitis B, Chronic/microbiology , Adult , Middle Aged , Antiviral Agents/therapeutic use , Antiviral Agents/administration & dosage , Bacteria/classification , Bacteria/drug effects , Bacteria/genetics , Bacteria/isolation & purification , Feces/microbiology , Feces/virology , RNA, Ribosomal, 16S/genetics , Hepatitis B virus/genetics , Hepatitis B virus/drug effects
16.
Sci Total Environ ; 953: 176153, 2024 Nov 25.
Article in English | MEDLINE | ID: mdl-39260480

ABSTRACT

Microplastics (MPs) can persist in the environment and human body. Murine studies showed that exposure to MPs could cause metabolic dysregulation, contributing metabolic dysfunction-associated steatotic liver disease (MASLD) or steatohepatitis (MASH). However, research on the role of MPs in humans is limited. Thus, we aimed to assess links between human fecal MPs and liver histology, gene expression, immune cells and intestinal microbiota (IM). We included 6 lean healthy liver donors and 6 normal liver (obese) and 11 MASH patients. Overall, pre-BSx, we observed no significant differences in fecal MPs between groups. However, fecal MP fibers and total MPs positively correlated with portal and total macrophages and total killer T cells while total fecal MPs were positively correlated with natural killer cells. Additionally, 19 genes related to immune system and apoptosis correlated with fecal MPs at baseline. Fecal MP fibers correlated positively with fecal Bifidobacterium and negatively with Lachnospiraceae. Patients with MASH (n = 11) were re-assessed 12-months post-bariatric surgery (BSx) and we found that those with persistent disease (n = 4) had higher fecal MP fragments than those with normalized liver histology (n = 7). At 12-month post-BSx, MP fragments positively correlated with helper T cells and total MPs positively correlated with natural killer T cells and B cells. Our study is the first to look at 1) the role of MPs in MASH and its association with IM, immune cells and hepatic gene expression and 2) look at the role of MPs longitudinally in MASH persistence following BSx. Future research should further explore this relationship.


Subject(s)
Feces , Microplastics , Humans , Feces/microbiology , Feces/chemistry , Male , Gastrointestinal Microbiome , Female , Middle Aged , Fatty Liver , Adult , Liver/pathology
17.
Front Cell Infect Microbiol ; 14: 1444031, 2024.
Article in English | MEDLINE | ID: mdl-39282498

ABSTRACT

Tigecycline is a last-resort drug used to treat serious infections caused by multidrug-resistant bacteria. tet(X4) is a recently discovered plasmid-mediated tigecycline resistance gene that confers high-level resistance to tigecycline and other tetracyclines. Since the first discovery of tet(X4) in 2019, it has spread rapidly worldwide, and as a consequence, tigecycline has become increasingly ineffective in the clinical treatment of multidrug-resistant infections. In this study, we identified and analyzed tet(X4)-positive Escherichia coli isolates from duck farms in Hunan Province, China. In total, 976 samples were collected from nine duck farms. Antimicrobial susceptibility testing and whole-genome sequencing (WGS) were performed to establish the phenotypes and genotypes of tet(X4)-positive isolates. In addition, the genomic characteristics and transferability of tet(X4) were determined based on bioinformatics analysis and conjugation. We accordingly detected an E. coli strain harboring tet(X4) and seven other resistance genes in duck feces. Multi-locus sequence typing analysis revealed that this isolate belonged to a new clone, and subsequent genetic analysis indicated that tet(X4) was carried in a 4608-bp circular intermediate, flanked by ISVsa3-ORF2-abh elements. Moreover, it exhibited transferability to E. coli C600 with a frequency of 10-5. The detection of tet(X4)-harboring E, coli strains on duck farms enhances our understanding of tigecycline resistance dynamics. The transferable nature of the circular intermediate of tet(X4) contributing to the spread of tigecycline resistance genes poses a substantial threat to healthcare. Consequently, vigilant monitoring and proactive measures are necessary to prevent their spread.


Subject(s)
Anti-Bacterial Agents , Ducks , Escherichia coli Infections , Escherichia coli , Farms , Tigecycline , Whole Genome Sequencing , Animals , Anti-Bacterial Agents/pharmacology , China , Drug Resistance, Bacterial/genetics , Drug Resistance, Multiple, Bacterial/genetics , Ducks/microbiology , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/isolation & purification , Escherichia coli Infections/veterinary , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Feces/microbiology , Gene Transfer, Horizontal , Genotype , Microbial Sensitivity Tests , Multilocus Sequence Typing , Plasmids/genetics , Poultry Diseases/microbiology , Tigecycline/pharmacology
18.
Parasit Vectors ; 17(1): 387, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39267171

ABSTRACT

Soil-transmitted helminth (STH) infections account for a significant global health burden, necessitating mass drug administration with benzimidazole-class anthelmintics, such as albendazole (ALB), for morbidity control. However, ALB efficacy shows substantial variability, presenting challenges for achieving consistent treatment outcomes. We have explored the potential impact of the baseline gut microbiota on ALB efficacy in hookworm-infected individuals through microbiota profiling and machine learning (ML) techniques. Our investigation included 89 stool samples collected from hookworm-infected individuals that were analyzed by microscopy and quantitative PCR (qPCR). Of these, 44 were negative by microscopy for STH infection using the Kato-Katz method and qPCR 21 days after treatment, which entails a cure rate of 49.4%. Microbiota characterization was based on amplicon sequencing of the V3-V4 16S ribosomal RNA gene region. Alpha and beta diversity analyses revealed no significant differences between participants who were cured and those who were not cured, suggesting that baseline microbiota diversity does not influence ALB treatment outcomes. Furthermore, differential abundance analysis at the phylum, family and genus levels yielded no statistically significant associations between bacterial communities and ALB efficacy. Utilizing supervised ML models failed to predict treatment response accurately. Our investigation did not provide conclusive insights into the relationship between gut microbiota and ALB efficacy. However, the results highlight the need for future research to incorporate longitudinal studies that monitor changes in the gut microbiota related to the infection and the cure with ALB, as well as functional metagenomics to better understand the interaction of the microbiome with the drug, and its role, if there is any, in modulating anthelmintic treatment outcomes in STH infections. Interdisciplinary approaches integrating microbiology, pharmacology, genetics and data science will be pivotal in advancing our understanding of STH infections and optimizing treatment strategies globally.


Subject(s)
Albendazole , Anthelmintics , Feces , Gastrointestinal Microbiome , Hookworm Infections , Albendazole/therapeutic use , Albendazole/pharmacology , Albendazole/administration & dosage , Humans , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Anthelmintics/therapeutic use , Anthelmintics/administration & dosage , Hookworm Infections/drug therapy , Feces/parasitology , Feces/microbiology , Female , Male , RNA, Ribosomal, 16S/genetics , Adult , Treatment Outcome , Animals , Young Adult , Middle Aged , Ancylostomatoidea/drug effects , Ancylostomatoidea/genetics , Adolescent , Child
19.
Pol J Microbiol ; 73(3): 403-410, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39268956

ABSTRACT

Vancomycin-resistant Enterococcus faecium (VRE) has been detected in Türkiye. Only limited information is available on its dissemination in the central regions of the country. This study describes the first epidemiological characterization of VRE clinical isolates detected in patients in a hospital in the province of Aksaray. In this one-year study conducted between 2021 and 2022, stool samples from intensive care unit patients were screened for VRE using the phenotypic E-test method, and the antibiotic sensitivity test was analyzed by using the VITEK® 2 system. A molecular assay for confirmation of species level was carried out by 16S rRNA gene-based sequencing and testing for antibiotic resistance (vanA or vanB) and virulence factor-encoding genes (esp, asa1, and hyl). Further, genotypic characterization was determined by macro-restriction fragment pattern analysis (MRFPA) of genomic DNA digested with SmaI restriction enzyme. Of the total 350 Enterococcus positive patients from different hospital intensive care units, 22 (6.3%) were positive for VRE using the phenotypic E-test method. All isolates showed resistance to ampicillin, ciprofloxacin, vancomycin, and teicoplanin and positive amplification for the vanA gene. However, none of the isolates was positive for the vanB gene. The most prevalent virulence gene was esp. The results indicate that the isolates are persistent in the hospital environment and subsequently transmitted to hospitalized patients, thus representing challenges to an outbreak and infection control. These study results would also help formulate more effective strategies to reduce the transmission and propagation of VRE contamination in various hospital settings.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Enterococcus faecium , Genotype , Gram-Positive Bacterial Infections , Intensive Care Units , Microbial Sensitivity Tests , Vancomycin-Resistant Enterococci , Humans , Enterococcus faecium/genetics , Enterococcus faecium/drug effects , Enterococcus faecium/isolation & purification , Vancomycin-Resistant Enterococci/genetics , Vancomycin-Resistant Enterococci/isolation & purification , Vancomycin-Resistant Enterococci/drug effects , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/epidemiology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Virulence Factors/genetics , Vancomycin/pharmacology , Feces/microbiology , RNA, Ribosomal, 16S/genetics , Phenotype , Male , Female , Vancomycin Resistance/genetics , Middle Aged
20.
Syst Appl Microbiol ; 47(5): 126545, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39241699

ABSTRACT

This study provides an emended description of Acinetobacter faecalis, a species previously described based on a single isolate (YIM 103518T) from elephant feces in China. Our emended description is based on 15 novel isolates conspecific with the A. faecalis type strain, obtained from eight cattle farms in the Czech Republic. The A. faecalis strains have relatively small genomes (≈2.5-2.7 Mbp), with a GC content of 36.3-36.7 mol%. Core genome-based phylogenetic analysis showed that the 15 strains, together with the type strain of A. faecalis, form a distinct and internally coherent phylogroup within the genus. Pairwise genomic ANIb values for the 16 A. faecalis strains were 97.32-99.04 %, while ANIb values between the genomes of the 16 strains and those of the other Acinetobacter spp. were ≤ 86.2 %. Analysis of whole-cell MALDI-TOF mass spectra supported the distinctness and cohesiveness of the taxon. The A. faecalis strains could be differentiated from the other validly named Acinetobacter spp. by the absence of hemolytic activity along with their ability to grow at 37 °C and on L-aspartate, ethanol, and L-glutamate but not at 41 °C or on adipate or 2,3-butanediol. Reduced susceptibility to sulfamethoxazole, trimethoprim and/or streptomycin was shown in eight strains, along with the presence of corresponding antibiotic resistance genes. In conclusion, this study provides a comprehensive description of A. faecalis and demonstrates its occurrence in cattle feces. Though the ecological role of A. faecalis remains unknown, our results show its ability to acquire antibiotic resistance genes, likely as an adaptation to antibiotic selection pressure in livestock farms.


Subject(s)
Anti-Bacterial Agents , Feces , Phylogeny , Animals , Cattle/microbiology , Feces/microbiology , Anti-Bacterial Agents/pharmacology , Genome, Bacterial/genetics , Czech Republic , Acinetobacter/genetics , Acinetobacter/classification , Acinetobacter/isolation & purification , DNA, Bacterial/genetics , Microbial Sensitivity Tests , Base Composition , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Bacterial Typing Techniques
SELECTION OF CITATIONS
SEARCH DETAIL