Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
1.
Int J Biol Macromol ; 277(Pt 1): 133726, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39084973

ABSTRACT

Epidemiological and preclinical studies have indicated a factual association between gut microbiota dysbiosis and high incidence of colitis. Dietary polysaccharides can specifically shift the composition of gut microbiome response to colitis. Here we validated the preventive role of polysaccharides from Pericarpium Citri Reticulatae 'Chachiensis' (PCRCP), a well-known traditional Chinese medicine, in colitis induced by dextrose sodium sulfate (DSS) in both rats and mice. We found that treatment with PCRCP not only significantly reduced DSS-induced colitis via down-regulating colonic inflammatory signaling pathways including PI3K-Akt, NLRs and NF-κB, but also enhanced colonic barrier integrity in rats. These protective activities of PCRCP against DSS-induced injuries in rats were in part due to the modulation of the gut microbiota revealed by both broad-spectrum antibiotic (ABX)-deleted bacterial and non-oral treatments. Furthermore, the improvement of PCRCP on colitis was impaired by intestinal neomycin-sensitive bacteria in DSS-exposed mice. Specifically, in vivo and in vitro treatment with PCRCP led to a highly sensible enrichment in the gut commensal Parabacteroides goldsteinii. Administration of Parabacteroides goldsteinii significantly alleviated typical symptoms of colitis and suppressed the activation of PI3K-Akt-involved inflammatory response in DSS-exposed mice. The anti-colitic effects of Parabacteroides goldsteinii were abolished after the activation of PI3K-Akt signaling pathway by lipopolysaccharide treatment in mice exposed to DSS. This study provides new insights into an anti-colitic mechanism driven by PCRCP and highlights the potential prebiotic of Parabacteroides goldsteinii for the prevention of ulcerative colitis.


Subject(s)
Colitis , Lipopolysaccharides , Phosphatidylinositol 3-Kinases , Polysaccharides , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Polysaccharides/pharmacology , Polysaccharides/chemistry , Mice , Signal Transduction/drug effects , Rats , Bacteroidetes/drug effects , Male , Gastrointestinal Microbiome/drug effects , Dextran Sulfate , Citrus/chemistry , Disease Models, Animal
2.
BMC Microbiol ; 24(1): 283, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39085808

ABSTRACT

BACKGROUND: The guts of mammals are home to trillions of microbes, forming a complex and dynamic ecosystem. Gut microbiota is an important biological barrier for maintaining immune homeostasis. Recently, the use of antibiotics to clear gut microbiota has gained popularity as a low cost and easy-to-use alternative to germ-free animals. However, the effect of the duration of the antibiotic cocktail on the gut microbiome is unclear, and more importantly, the effect of dramatic changes in the gut microbiota on intestinal tissue morphology and local immune response is rarely reported. RESULTS: We observed a significant reduction in fecal microbiota species and abundance after 1 week of exposure to an antibiotic cocktail, gavage twice daily by intragastric administration. In terms of composition, Bacteroidetes and Firmicutes were replaced by Proteobacteria. Extending antibiotic exposure to 2-3 weeks did not significantly improve the overall efficiency of microbiotal consumption. No significant histomorphological changes were observed in the first 2 weeks of antibiotic cocktail exposure, but the expression of inflammatory mediators in intestinal tissue was increased after 3 weeks of antibiotic cocktail exposure. Mendelian randomization analysis showed that Actinobacteria had a significant causal association with the increase of IL-1ß (OR = 1.65, 95% CI = 1.23 to 2.21, P = 0.007) and TNF-α (OR = 1.81, 95% CI = 1.26 to 2.61, P = 0.001). CONCLUSIONS: Our data suggest that treatment with an antibiotic cocktail lasting 1 week is sufficient to induce a significant reduction in gut microbes. 3 weeks of antibiotic exposure can lead to the colonization of persistant microbiota and cause changes in intestinal tissue and local immune responses.


Subject(s)
Anti-Bacterial Agents , Feces , Gastrointestinal Microbiome , Anti-Bacterial Agents/pharmacology , Animals , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Interleukin-1beta/genetics , Mice , Bacteria/drug effects , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Mice, Inbred C57BL , Bacteroidetes/drug effects , Firmicutes/drug effects
3.
FASEB J ; 38(8): e23603, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38648368

ABSTRACT

Recent evidence suggests that chronic exposure to opioid analgesics such as morphine disrupts the intestinal epithelial layer and causes intestinal dysbiosis. Depleting gut bacteria can preclude the development of tolerance to opioid-induced antinociception, suggesting an important role of the gut-brain axis in mediating opioid effects. The mechanism underlying opioid-induced dysbiosis, however, remains unclear. Host-produced antimicrobial peptides (AMPs) are critical for the integrity of the intestinal epithelial barrier as they prevent the pathogenesis of the enteric microbiota. Here, we report that chronic morphine or fentanyl exposure reduces the antimicrobial activity in the ileum, resulting in changes in the composition of bacteria. Fecal samples from morphine-treated mice had increased levels of Akkermansia muciniphila with a shift in the abundance ratio of Firmicutes and Bacteroidetes. Fecal microbial transplant (FMT) from morphine-naïve mice or oral supplementation with butyrate restored (a) the antimicrobial activity, (b) the expression of the antimicrobial peptide, Reg3γ, (c) prevented the increase in intestinal permeability and (d) prevented the development of antinociceptive tolerance in morphine-dependent mice. Improved epithelial barrier function with FMT or butyrate prevented the enrichment of the mucin-degrading A. muciniphila in morphine-dependent mice. These data implicate impairment of the antimicrobial activity of the intestinal epithelium as a mechanism by which opioids disrupt the microbiota-gut-brain axis.


Subject(s)
Analgesics, Opioid , Dysbiosis , Fentanyl , Gastrointestinal Microbiome , Intestinal Mucosa , Mice, Inbred C57BL , Morphine , Animals , Morphine/pharmacology , Mice , Dysbiosis/chemically induced , Dysbiosis/microbiology , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Male , Fentanyl/pharmacology , Analgesics, Opioid/pharmacology , Brain-Gut Axis/drug effects , Fecal Microbiota Transplantation , Pancreatitis-Associated Proteins/metabolism , Akkermansia/drug effects , Antimicrobial Peptides/pharmacology , Bacteroidetes/drug effects
4.
Cell ; 185(3): 513-529.e21, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35120663

ABSTRACT

The human gut microbiota resides within a diverse chemical environment challenging our ability to understand the forces shaping this ecosystem. Here, we reveal that fitness of the Bacteroidales, the dominant order of bacteria in the human gut, is an emergent property of glycans and one specific metabolite, butyrate. Distinct sugars serve as strain-variable fitness switches activating context-dependent inhibitory functions of butyrate. Differential fitness effects of butyrate within the Bacteroides are mediated by species-level variation in Acyl-CoA thioesterase activity and nucleotide polymorphisms regulating an Acyl-CoA transferase. Using in vivo multi-omic profiles, we demonstrate Bacteroides fitness in the human gut is associated together, but not independently, with Acyl-CoA transferase expression and butyrate. Our data reveal that each strain of the Bacteroides exists within a unique fitness landscape based on the interaction of chemical components unpredictable by the effect of each part alone mediated by flexibility in the core genome.


Subject(s)
Gastrointestinal Microbiome , Metabolome , Polysaccharides/metabolism , Acyl Coenzyme A/metabolism , Amino Acid Sequence , Amino Acids, Branched-Chain/metabolism , Bacteroidetes/drug effects , Bacteroidetes/genetics , Bacteroidetes/growth & development , Butyrates/chemistry , Butyrates/pharmacology , Coenzyme A-Transferases/chemistry , Coenzyme A-Transferases/metabolism , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Genetic Variation/drug effects , Hydrogen-Ion Concentration , Metabolome/drug effects , Metabolome/genetics , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , Species Specificity , Stress, Physiological/drug effects , Stress, Physiological/genetics , Transcription, Genetic/drug effects
5.
Food Funct ; 13(5): 2952-2965, 2022 Mar 07.
Article in English | MEDLINE | ID: mdl-35191911

ABSTRACT

Green tea polyphenols (GTP) play an important role in shaping the gut microbiome, comprising a range of densely colonizing microorganisms, including bacteriophages. Previous studies focused on the effect of GTP on the bacteria in the gut microbiota. However, little is known about the role of GTP in the bacteriophage composition of healthy intestines. In this study, SPF male C57BL/6J mice were divided into a polyphenol-free diet group and a tea polyphenol diet group where drinking water was supplemented with 0.1% GTP for 28 days. The ultra-deep metagenomic sequencing of virus-like particle preparations and bacterial 16S rRNA sequencing were performed on mouse stool samples. Changes in the gut bacteriome, bacteriophages, and bacterial-bacteriophage correlations were then compared between the groups. The results revealed an abundance of Firmicutes, a significant decrease in Bacteroidetes, and a significant increase in the ratio of F/B after GTP exposure. The GTP altered the abundance (relative abundance > 1.00%) of Bifidobacterium (regulation rate of 89.78% and the abundance up-regulated by 0.89%) and Akkermansia (regulation rate of 99.70% and the abundance down-regulated by 1.77%). The abundance of Faecalibaculum (regulation rate of 60.17%) increased by 24.38% following GTP treatment. The GTP also altered the abundance of Salmonella phage (regulation rate of 98.64% and the abundance up-regulated by 3.16%) and that of Gordonia_phage_Yakult (regulation rate of 99.99% and the abundance down-regulated by 5.44%). It significantly increased the intestine's lytic phages and reduced the temperate phages by 29.22%. The dominant microorganisms (relative abundance >1.00%) of Bifidobacterium and Dubosiella had a significantly negative relationship with the Faecalibacterium phage and a significantly positive relationship with the Lactobacillus prophage. Exposure to GTP positively promoted changes in the gut bacteriophage community and interaction network in the microbial community of the SPF mice. These findings highlight the importance of "profitable" bacteriophage-bacteria relationships and reveal a potential mechanism of GTP towards the regulation of intestinal flora via intestinal phage communities.


Subject(s)
Antioxidants/pharmacology , Polyphenols/pharmacology , Tea , Animals , Antioxidants/chemistry , Bacteroidetes/drug effects , Gastrointestinal Microbiome/drug effects , Male , Mice , Mice, Inbred C57BL , Polyphenols/chemistry , Specific Pathogen-Free Organisms
6.
mBio ; 12(5): e0228521, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34517753

ABSTRACT

Bacteroidetocins are a family of antibacterial peptide toxins that are produced by and target members of the phylum Bacteroidetes. To date, 19 bacteroidetocins have been identified, and four have been tested and shown to kill diverse Bacteroidales species (M. J. Coyne, N. Béchon, L. M. Matano, V. L. McEneany, et al., Nat Commun 10:3460, 2019, https://doi.org/10.1038/s41467-019-11494-1). Here, we identify the target and likely mechanism of action of the bacteroidetocins. We selected seven spontaneous mutants of four different genera, all resistant to bacteroidetocin A (Bd-A) and found that all contained mutations in a single gene, bamA. Construction of three of these bamA mutants in the wild-type (WT) strains confirmed they confer resistance to Bd-A as well as to other bacteroidetocins. We identified an aspartate residue of BamA at the beginning of exterior loop 3 (eL3) that, when altered, renders strains resistant to Bd-A. Analysis of a panel of diverse Bacteroidales strains showed a correlation between the presence of this aspartate residue and Bd-A sensitivity. Fluorescence microscopy and transmission electron microscopy (TEM) analysis of Bd-A-treated cells showed cellular morphological changes consistent with a BamA defect. Transcriptomic analysis of Bd-A-treated cells revealed gene expression changes indicative of cell envelope stress. Studies in mice revealed that bacteroidetocin-resistant mutants are outcompeted by their WT strain in vivo. Analyses of longitudinal human gut isolates showed that bamA mutations leading to bacteroidetocin resistance do not become fixed in the human gut, even in bacteroidetocin-producing strains and nonproducing coresident strains. Together, these data lend further support to the applicability of the bacteroidetocins as therapeutic peptides in the treatment of maladies involving Bacteroidales species. IMPORTANCE The bacteroidetocins are a newly discovered class of bacteriocins specific to Bacteroidetes with a spectrum of targets extending from symbiotic gut Bacteroides, Parabacteroides, and Prevotella species to pathogenic oral and vaginal Prevotella species. We previously showed that one such bacteroidetocin, Bd-A, is active at nanomolar concentrations, is water soluble, and is bactericidal, all desirable features in a therapeutic antibacterial peptide. Here, we identify the target of several of the bacteroidetocins as the essential outer membrane protein BamA. Although mutations in bamA can be selected in bacteria grown in vitro, we show both in a mouse model and in human gut ecosystems that bamA mutants leading to Bd-A resistance are fitness attenuated and are not selected. These features further support the potential usefulness of the bacteroidetocins as therapeutics for maladies associated with pathogenic Prevotella species, such as recurrent bacterial vaginosis, for which there are few effective treatments.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/metabolism , Bacteriocins/pharmacology , Bacteroidetes/drug effects , Amino Acid Motifs , Amino Acid Sequence , Animals , Bacterial Outer Membrane/chemistry , Bacterial Outer Membrane/drug effects , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacteroidetes/chemistry , Bacteroidetes/genetics , Bacteroidetes/physiology , Drug Resistance, Bacterial , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/microbiology , Gram-Negative Bacterial Infections/microbiology , Humans , Mice , Sequence Alignment , Symbiosis
7.
Microbiologyopen ; 10(4): e1231, 2021 08.
Article in English | MEDLINE | ID: mdl-34459542

ABSTRACT

Marine biofouling imposes serious environmental and economic impacts on marine applications, especially in the shipping industry. To combat biofouling, protective coatings are applied on vessel hulls which are divided into two major groups: biocidal and non-toxic fouling release. The current study aimed to explore the effect of coating type on microbial biofilm community profiles to better understand the differences between the communities developed on fouling control biocidal antifouling and biocidal-free coatings. Biocidal (Intersmooth® 7460HS SPC), fouling release (Intersleek® 900), and inert surfaces were deployed in the marine environment for 4 months, and the biofilms that developed on these surfaces were investigated using Illumina NGS sequencing, targeting the prokaryotic 16S rRNA gene. The results confirmed differences in the community profiles between coating types. The biocidal coating supported communities dominated by Alphaproteobacteria (Loktanella, Sphingorhabdus, Erythrobacter) and Bacteroidetes (Gilvibacter), while other taxa, such as Portibacter and Sva0996 marine group, proliferated on the fouling-release surface. Knowledge of these marine biofilm components on fouling control coatings will serve as a guide for future investigations of marine microfouling as well as informing the coatings industry of potential microbial targets for robust coating formulations.


Subject(s)
Alphaproteobacteria/growth & development , Bacteroidetes/growth & development , Biofilms/growth & development , Biofouling/prevention & control , Disinfectants/pharmacology , Alphaproteobacteria/drug effects , Alphaproteobacteria/genetics , Bacteroidetes/drug effects , Bacteroidetes/genetics , Biofilms/drug effects , Computational Biology , High-Throughput Nucleotide Sequencing , Microbiota/drug effects , Seawater/microbiology
9.
Gut Microbes ; 13(1): 1922241, 2021.
Article in English | MEDLINE | ID: mdl-34196581

ABSTRACT

Parabacteroides distasonis is the type strain for the genus Parabacteroides, a group of gram-negative anaerobic bacteria that commonly colonize the gastrointestinal tract of numerous species. First isolated in the 1930s from a clinical specimen as Bacteroides distasonis, the strain was re-classified to form the new genus Parabacteroides in 2006. Currently, the genus consists of 15 species, 10 of which are listed as 'validly named' (P. acidifaciens, P. chartae, P. chinchillae, P. chongii, P. distasonis, P. faecis, P. goldsteinii, P. gordonii, P. johnsonii, and P. merdae) and 5 'not validly named' (P. bouchesdurhonensis, P. massiliensis, P. pacaensis, P. provencensis, and P. timonensis) by the List of Prokaryotic names with Standing in Nomenclature. The Parabacteroides genus has been associated with reports of both beneficial and pathogenic effects in human health. Herein, we review the literature on the history, ecology, diseases, antimicrobial resistance, and genetics of this bacterium, illustrating the effects of P. distasonis on human and animal health.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteroidetes/drug effects , Bacteroidetes/isolation & purification , Drug Resistance, Bacterial , Gastrointestinal Microbiome , Gram-Negative Bacterial Infections/microbiology , Animals , Bacteroidetes/genetics , Bacteroidetes/physiology , Humans , Phylogeny , Probiotics/chemistry , Probiotics/isolation & purification
10.
BMC Microbiol ; 21(1): 166, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34082715

ABSTRACT

Antibiotic exposure during pregnancy will adversely affect the growth of offspring; however, this remains controversial and the mechanism is poorly understood. To study this phenomenon, we added ceftriaxone sodium to the drinking water of pregnant rats and continuously monitored the body weight of their offspring. The results showed that compared with the control group, the offspring exposed to antibiotics during pregnancy had a higher body weight up to 3 weeks old but had a lower body weight at 6 weeks old. To determine the role of the gut microbiota and its metabolites in the growth of offspring, we collected feces for sequencing and further established that the experimental group has a different composition ratio of dominant bacteria at 6 week old, among which S24-7 correlated negatively with body weight and the metabolites that correlated with body weight-related unique flora were L-Valine, L-Leucine, Glutaric acid, N-Acetyl-L-glutamate, and 5-Methylcytosine. To further explore how they affect the growth of offspring, we submitted these data to Kyoto Encyclopedia of Genes and Genomes website for relevant pathway analysis. The results showed that compared with the control, the following metabolic pathways changed significantly: Valine, leucine, and isoleucine biosynthesis; Protein digestion and absorption; and Mineral absorption. Therefore, we believe that our findings support the conclusion that ceftriaxone sodium exposure in pregnancy has a long-lasting adverse effect on the growth of offspring because of an imbalance of gut microbiota, especially S24-7, via different metabolic pathways.


Subject(s)
Anti-Bacterial Agents/adverse effects , Bacteroidetes/drug effects , Body Weight/drug effects , Ceftriaxone/adverse effects , Gastrointestinal Microbiome/drug effects , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects/microbiology , Prenatal Exposure Delayed Effects/physiopathology , Animals , Bacteria/classification , Bacteria/drug effects , Bacteria/genetics , Bacteria/isolation & purification , Bacteroidetes/classification , Bacteroidetes/genetics , Bacteroidetes/isolation & purification , Female , Humans , Male , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/genetics , Rats , Rats, Sprague-Dawley
12.
Int J Biol Macromol ; 167: 1308-1318, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33202270

ABSTRACT

Flammulina velutipes polysaccharides (FVP) can improve gut health through gut microbiota and metabolism regulation. In this study, the 28-days fed experiment was used to investigate gut microbime and metabolic profiling induced by FVP. After treatment, intestinal tissue section showed the higher villus height and villus height/crypt depth (V/C) value in FVP-treated group. The 16 s rRNA gene sequencing revealed microbiota composition alteration caused by FVP, as the Firmicutes phylum increased while Bacteroidetes phylum slightly decreased. The metabolic profiling was detected by LC/MS and results showed 56 and 99 compounds were dramatically changed after FVP treatment in positive and negative ion mode, respectively. Annotation in Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways displayed the adjustment of energy metabolism, amino acid metabolism, nucleotide metabolism and other related basic pathways after FVP treatment. Our study suggested that FVP can be developed as a dietary supplement for intestine health promotion.


Subject(s)
Dietary Carbohydrates/pharmacology , Flammulina/chemistry , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/drug effects , Metabolic Networks and Pathways/drug effects , Polysaccharides/pharmacology , Amino Acids/metabolism , Animals , Bacteroidetes/drug effects , Body Weight/drug effects , Chromatography, Liquid , Energy Metabolism/drug effects , Firmicutes/drug effects , Intestinal Mucosa/anatomy & histology , Intestinal Mucosa/cytology , Jejunum/cytology , Jejunum/drug effects , Male , Mass Spectrometry , Metabolomics , Mice , Mice, Inbred C57BL , Nucleotides/metabolism , Polysaccharides/chemistry , RNA, Ribosomal, 16S/genetics
13.
Anaerobe ; 66: 102283, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33022383

ABSTRACT

Alistipes spp is a genus of Gram-negative anaerobic rods involved in very few human diseases. We report the first case of abdominal infection due to Alistipes onderdonkii in a 58-year-old man with a history of chronic obstructive pulmonary disease. He presented with abdominal pain and general malaise after retrogastric drainage for a pancreatitis episode a few days earlier. After the diagnosis of diffuse peritonitis with perforation and necrotizing pancreatic collection, abundant pancreatic fluid was drained and yielded the isolation of A. onderdonkii in pure culture. Resistance to penicillin and moxifloxacin was documented for this strain. Treatment with metronidazole was prescribed, and the patient was discharged after improvement of his general condition.


Subject(s)
Bacteroidetes/classification , Intraabdominal Infections/diagnosis , Intraabdominal Infections/microbiology , Anaerobiosis , Anti-Bacterial Agents/therapeutic use , Bacteroidetes/drug effects , Bacteroidetes/genetics , DNA, Bacterial , Humans , Intraabdominal Infections/drug therapy , Male , Microbial Sensitivity Tests , Middle Aged , Peritonitis/diagnosis , Peritonitis/microbiology , RNA, Ribosomal, 16S , Treatment Outcome
14.
PLoS One ; 15(10): e0241338, 2020.
Article in English | MEDLINE | ID: mdl-33125401

ABSTRACT

BACKGROUND: The study was conducted to investigate the effects of metformin treatment on the human gut microbiome's taxonomic and functional profile in the Latvian population, and to evaluate the correlation of these changes with therapeutic efficacy and tolerance. METHODS: In this longitudinal observational study, stool samples for shotgun metagenomic sequencing-based analysis were collected in two cohorts. The first cohort included 35 healthy nondiabetic individuals (metformin dose 2x850mg/day) at three time-points during metformin administration. The second cohort was composed of 50 newly-diagnosed type 2 diabetes patients (metformin dose-determined by an endocrinologist) at two concordant times. Patients were defined as Responders if their HbA1c levels during three months of metformin therapy had decreased by ≥12.6 mmol/mol (1%), while in Non-responders HbA1c were decreased by <12.6 mmol/mol (1%). RESULTS: Metformin reduced the alpha diversity of microbiota in healthy controls (p = 0.02) but not in T2D patients. At the species level, reduction in the abundance of Clostridium bartlettii and Barnesiella intestinihominis, as well as an increase in the abundance of Parabacteroides distasonis and Oscillibacter unclassified overlapped between both study groups. A large number of group-specific changes in taxonomic and functional profiles was observed. We identified an increased abundance of Prevotella copri (FDR = 0.01) in the Non-Responders subgroup, and enrichment of Enterococcus faecium, Lactococcus lactis, Odoribacter, and Dialister at baseline in the Responders group. Various taxonomic units were associated with the observed incidence of side effects in both cohorts. CONCLUSIONS: Metformin effects are different in T2D patients and healthy individuals. Therapy induced changes in the composition of gut microbiome revealed possible mediators of observed short-term therapeutic effects. The baseline composition of the gut microbiome may influence metformin therapy efficacy and tolerance in T2D patients and could be used as a powerful prediction tool.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/microbiology , Gastrointestinal Microbiome/physiology , Metformin/therapeutic use , Adult , Bacteroidetes/drug effects , Female , Humans , Lactococcus lactis/drug effects , Longitudinal Studies , Male , Microbiota/drug effects , Prevotella/drug effects , Young Adult
15.
Article in English | MEDLINE | ID: mdl-32998606

ABSTRACT

Given the toxicity and widespread occurrence of hexavalent chromium [Cr(VI)] in aquatic environments, we investigated the feasibility of a down-flow hanging sponge (DHS) biofilm reactor for the enrichment of microbial communities capable of Cr(VI) removal. In the present study, a laboratory-scale DHS reactor fed with a molasses-based medium containing Cr(VI) was operated for 112 days for the investigation. The enrichment of Cr(VI)-removing microbial communities was evaluated based on water quality and prokaryotic community analyses. Once the DHS reactor began to operate, high average volumetric Cr(VI) removal rates of 1.21-1.45 mg L-sponge-1 h-1 were confirmed under varying influent Cr(VI) concentrations (approximately 20-40 mg L-1). 16S rRNA gene amplicon sequencing analysis suggested the presence of phylogenetically diverse prokaryotic lineages, including phyla that contain well-known Cr(VI)-reducing bacteria (e.g., Bacteroidetes, Firmicutes, and Proteobacteria) in the polyurethane sponge media of the DHS reactor. Therefore, our findings indicate that DHS reactors have great potential for the enrichment of Cr(VI)-removing microbial communities.


Subject(s)
Biofilms/growth & development , Bioreactors/microbiology , Chromium/analysis , Microbiota/drug effects , Water Pollutants, Chemical/analysis , Water Purification/methods , Bacteroidetes/drug effects , Firmicutes/drug effects , Polyurethanes/chemistry , Proteobacteria/drug effects , RNA, Ribosomal, 16S/genetics
16.
Gut Microbes ; 12(1): 1795492, 2020 11 09.
Article in English | MEDLINE | ID: mdl-32991820

ABSTRACT

INTRODUCTION: Antimicrobial drugs are known to have effects on the human gut microbiota. We studied the long-term temporal relationship between several antimicrobial drug groups and the composition of the human gut microbiota determined in feces samples. METHODS: Feces samples were obtained from a community-dwelling cohort of middle-aged and elderly individuals (Rotterdam Study). Bacterial DNA was isolated and sequenced using V3/V4 16 S ribosomal RNA sequencing (Illumina MiSeq). The time between the last prescription of several antimicrobial drug groups and the day of sampling was categorized into 0-12, 12-24, 24-48 and >48 months. The effects of the antimicrobial drug groups on the Shannon alpha-diversity (diversity), the Bray-Curtis beta-diversity (community structure), the Firmicutes/Bacteroidetes (F/B) ratio and individual genera were determined. RESULTS: We studied the gut microbiota of 1413 individuals (57.5% female, median age 62.6 years). The alpha-diversity was significantly lower up to 4 years after prescriptions of macrolides and lincosamides. It was also lower in the first year after the use of beta-lactams. The community structure (beta-diversity) of the microbiota was significantly different up to 4 years for macrolides and lincosamides, the first year for beta-lactams and at least the first year for quinolones. For the F/B ratio, drugs with a high anaerobic activity shifted the ratio toward Firmicutes in the first year whereas other antimicrobial drugs shifted the ratio toward Bacteroidetes. CONCLUSION: Use of antimicrobial drugs is associated with a shift in the composition of the gut microbiota.These effects differ in strength and duration, depending on the antimicrobial drug group used. These findings should be considered when prescribing antimicrobial drugs.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Gastrointestinal Microbiome/drug effects , Aged , Anti-Bacterial Agents/pharmacology , Bacteroidetes/drug effects , Bacteroidetes/isolation & purification , Biodiversity , Cohort Studies , Feces/microbiology , Female , Firmicutes/drug effects , Firmicutes/isolation & purification , Humans , Male , Middle Aged
17.
Sci Rep ; 10(1): 15054, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32929122

ABSTRACT

This study examined the genetic mutation and toxicant exposure in producing gut microbiota alteration and neurotoxicity. Homozygous α-synuclein mutant (SNCA) mice that overexpress human A53T protein and littermate wild-type mice received a single injection of LPS (2 mg/kg) or a selective norepinephrine depleting toxin DSP-4 (50 mg/kg), then the motor activity, dopaminergic neuron loss, colon gene expression and gut microbiome were examined 13 months later. LPS and DSP-4 decreased rotarod and wirehang activity, reduced dopaminergic neurons in substantia nigra pars compacta (SNpc), and SNCA mice were more vulnerable. SNCA mice had 1,000-fold higher human SNCA mRNA expression in the gut, and twofold higher gut expression of NADPH oxidase (NOX2) and translocator protein (TSPO). LPS further increased expression of TSPO and IL-6 in SNCA mice. Both LPS and DSP-4 caused microbiome alterations, and SNCA mice were more susceptible. The altered colon microbiome approximated clinical findings in PD patients, characterized by increased abundance of Verrucomicrobiaceae, and decreased abundance of Prevotellaceae, as evidenced by qPCR with 16S rRNA primers. The Firmicutes/Bacteroidetes ratio was increased by LPS in SNCA mice. This study demonstrated a critical role of α-synuclein and toxins interactions in producing gut microbiota disruption, aberrant gut pro-inflammatory gene expression, and dopaminergic neuron loss.


Subject(s)
Benzylamines/toxicity , Gastrointestinal Microbiome/drug effects , Lipopolysaccharides/toxicity , Neurotoxins/toxicity , alpha-Synuclein/genetics , Animals , Bacteroidetes/drug effects , Benzylamines/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Motor Activity , Mutation, Missense , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , Neurotoxins/pharmacology , Norepinephrine/metabolism , Receptors, GABA/genetics , Receptors, GABA/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Verrucomicrobia/drug effects , alpha-Synuclein/metabolism
18.
Carbohydr Polym ; 248: 116780, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32919569

ABSTRACT

In this study, the beneficial effects of a homogalacturonan(HG)-type pectic polysaccharide from Ficus pumila L. fruits (FPLP) in obese mice were investigated. The 17-week FPLP treatment effectively attenuated obesity, as mainly demonstrated by the reductions of body weight, serum total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) levels in high-fat diet (HFD)-induced obese mice. The decreased Firmicutes to Bacteroidetes abundance ratio, enriched Akkermansia, and reduced Blautia abundance suggested that FPLP ameliorated the HFD-induced gut dysbiosis. FPLP also influenced the levels of metabolites altered upon HFD feeding, including increases in myristoleic acid and pentadecanoic acid levels. The correlation studies indicated that FPLP ameliorated HFD-induced rise in TC and LDL-C levels through regulating gut microbial community and their associated metabolites. In conclusion, this study extends our understanding of the relationships among gut microbiota (Akkermansia and Blautia), metabolites (myristoleic acid and pentadecanoic acid), HG-type pectin and its TC- and LDL-C- lowering functions.


Subject(s)
Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/drug effects , Obesity/prevention & control , Pectins/pharmacology , Polysaccharides/pharmacology , Akkermansia/drug effects , Animals , Bacteroidetes/drug effects , Body Weight/drug effects , Diet, High-Fat/adverse effects , Dysbiosis/etiology , Dysbiosis/prevention & control , Ficus/chemistry , Firmicutes/drug effects , Fruit/chemistry , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Male , Mice, Inbred C57BL , Obesity/etiology , Pectins/administration & dosage , Polysaccharides/administration & dosage , Population Dynamics
19.
Aquat Toxicol ; 227: 105591, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32853898

ABSTRACT

The effects of allelopathy and the potential harm of several isolated allelochemicals have been studied in detail. Microorganisms in the phycosphere play an important role in algal growth, decay and nutrient cycling. However, it is unknown and often neglected whether allelochemicals affect the phycosphere. The present study selected a phenolic acid protocatechuic acid (PA) - previously shown to be an allelochemical. We studied PA at a half maximal effective concentration of 0.20 mM (30 mg L-1) against Scrippsiella trochoidea to assess the effect of PA on its phycosphere in an acute time period (48 h). The results showed that: 1) OTUs (operational taxonomic units) in the treatment groups (31.4 ± 0.55) exceeded those of the control groups (28.2 ± 1.30) and the Shannon and Simpson indices were lower than the control groups (3.31 ± 0.08 and 0.84 ± 0.02, 3.45 ± 0.09 and 0.88 ± 0.01); 2) Gammaproteobacteria predominated in the treatment groups (44.71 ± 2.13 %) while Alphaproteobacteria dominated in the controls (67.17 ± 3.87 %); 3) Gammaproteobacteria and Alphaproteobacteria were important biomarkers in the treatment and control groups respectively (LDA > 4.0). PA improved the relative abundance of Alteromonas significantly and decreased the one of Rhodobacteraceae. PICRUSt analysis showed that the decrease of Rhodobacterceae was closely related with the decline of most functional genes in metabolism such as amino acid, carbohydrate, xenobiotics, cofactors and vitamins metabolism after PA-treated.


Subject(s)
Allelopathy/drug effects , Dinoflagellida/drug effects , Harmful Algal Bloom/drug effects , Hydroxybenzoates/pharmacology , Microbiota/drug effects , Pheromones/pharmacology , Allelopathy/genetics , Bacteroidetes/drug effects , Bacteroidetes/genetics , Bacteroidetes/isolation & purification , Dinoflagellida/genetics , Dinoflagellida/growth & development , Microbiota/genetics , Proteobacteria/drug effects , Proteobacteria/genetics , Proteobacteria/isolation & purification
20.
Int J Biol Macromol ; 163: 1393-1402, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32755709

ABSTRACT

In this study, the effect of Ziziphus jujuba Mill var. spinosa seeds (ZSS) polysaccharides on the bioavailability of spinosin in mice and its molecular mechanism were investigated. After continuously fed with ZSS polysaccharides 100 mg/kg·d-1 for 28 consecutive days, the C57BL/6 mice absorbed spinosin at an obvious lower level compared with the control group. The expression levels of P-gp, MRP2 and Occludin in the colon were significantly increased. ZSS polysaccharides significantly regulated the composition of the gut microbiota, reducing the abundance of Bacteroidetes, and increasing the richness of Firmicutes and Verrucomicrobia. Moreover, ZSS polysaccharides can significantly regulate the expression levels of tight junction proteins and efflux transporters in Caco-2 cells. However, the gut microbiota culture supernatant showed no obvious biological activity in this regard. Furthermore, histopathological analysis revealed ZSS polysaccharides can alleviate TNBS-induced colitis, reduced inflammatory cell infiltration in mice. This immune regulation was related to the NF-κB and MAPK pathways in RAW264.7 cells.


Subject(s)
Colitis/prevention & control , Flavonoids/metabolism , Polysaccharides/pharmacology , Seeds/chemistry , Ziziphus/chemistry , Animals , Bacteroidetes/drug effects , Biological Availability , Caco-2 Cells , Cell Line, Tumor , Colitis/microbiology , Firmicutes/drug effects , Gastrointestinal Microbiome/drug effects , Humans , Inflammation/microbiology , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Verrucomicrobia/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL