Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 144
1.
Proc Natl Acad Sci U S A ; 119(26): e2122364119, 2022 06 28.
Article En | MEDLINE | ID: mdl-35727971

Solar-driven bioelectrosynthesis represents a promising approach for converting abundant resources into value-added chemicals with renewable energy. Microorganisms powered by electrochemical reducing equivalents assimilate CO2, H2O, and N2 building blocks. However, products from autotrophic whole-cell biocatalysts are limited. Furthermore, biocatalysts tasked with N2 reduction are constrained by simultaneous energy-intensive autotrophy. To overcome these challenges, we designed a biohybrid coculture for tandem and tunable CO2 and N2 fixation to value-added products, allowing the different species to distribute bioconversion steps and reduce the individual metabolic burden. This consortium involves acetogen Sporomusa ovata, which reduces CO2 to acetate, and diazotrophic Rhodopseudomonas palustris, which uses the acetate both to fuel N2 fixation and for the generation of a biopolyester. We demonstrate that the coculture platform provides a robust ecosystem for continuous CO2 and N2 fixation, and its outputs are directed by substrate gas composition. Moreover, we show the ability to support the coculture on a high-surface area silicon nanowire cathodic platform. The biohybrid coculture achieved peak faradaic efficiencies of 100, 19.1, and 6.3% for acetate, nitrogen in biomass, and ammonia, respectively, while maintaining product tunability. Finally, we established full solar to chemical conversion driven by a photovoltaic device, resulting in solar to chemical efficiencies of 1.78, 0.51, and 0.08% for acetate, nitrogenous biomass, and ammonia, correspondingly. Ultimately, our work demonstrates the ability to employ and electrochemically manipulate bacterial communities on demand to expand the suite of CO2 and N2 bioelectrosynthesis products.


Carbon Dioxide , Firmicutes , Nitrogen Fixation , Photosynthesis , Rhodopseudomonas , Acetates/metabolism , Ammonia , Carbon Dioxide/metabolism , Coculture Techniques , Ecosystem , Firmicutes/growth & development , Firmicutes/metabolism , Nitrogen/metabolism , Rhodopseudomonas/growth & development , Rhodopseudomonas/metabolism
2.
Sci Rep ; 12(1): 3140, 2022 02 24.
Article En | MEDLINE | ID: mdl-35210542

Childhood obesity is a risk factor for numerous health conditions. A critical factor in the etiology of obesity appears to be the gut microbiota, which is the microbial community that resides in the human gut. The ratio of the phyla Firmicutes and Bacteroidetes (F/B) and gut bacterial genera that produce short-chain fatty acids (SCFA) have been suggested to contribute to obesity. The current study investigated (1) whether differences in F/B ratio can be observed in infancy and childhood in relation to zBMI in healthy children, and (2) whether an innovative proxy measure adds evidence to a relationship between SCFA producers and the etiology of obesity. Stool samples were collected at five time points, and zBMI was assessed at eight time points throughout the first 12 years of life. Our confirmatory analyses with Bayesian multilevel models showed no relationship between the F/B ratio and zBMI. Also, a proxy measure constructed from known SCFA producers was unrelated to zBMI throughout the first 12 years of life. Exploratory analyses using multilevel and random forest models suggest that the relative abundances of Firmicutes and Bacteroidetes were independently negatively associated with zBMI from infancy through childhood, and the SCFA producing genera Subdoligranulum and Alistipes were negatively related to future BMI in childhood.


Bacteroidetes , Body Mass Index , Child Development , Fatty Acids, Volatile/metabolism , Firmicutes , Gastrointestinal Microbiome , Pediatric Obesity/microbiology , Bacteroidetes/classification , Bacteroidetes/growth & development , Child , Child, Preschool , Female , Firmicutes/classification , Firmicutes/growth & development , Humans , Infant , Infant, Newborn , Longitudinal Studies , Male
3.
mBio ; 12(6): e0220621, 2021 12 21.
Article En | MEDLINE | ID: mdl-34749527

Xyloglucan utilization by Ruminiclostridium cellulolyticum was formerly shown to imply the uptake of large xylogluco-oligosaccharides, followed by cytosolic depolymerization into glucose, galactose, xylose, and cellobiose. This raises the question of how the anaerobic bacterium manages the simultaneous presence of multiple sugars. Using genetic and biochemical approaches targeting the corresponding metabolic pathways, we observed that, surprisingly, all sugars are catabolized, collectively, but glucose consumption is prioritized. Most selected enzymes display unusual features, especially the GTP-dependent hexokinase of glycolysis, which appeared reversible and crucial for xyloglucan utilization. In contrast, mutant strains lacking either galactokinase, cellobiose-phosphorylase, or xylulokinase still catabolize xyloglucan but display variably altered growth. Furthermore, the xylogluco-oligosaccharide depolymerization process appeared connected to the downstream pathways through an intricate network of competitive and noncompetitive inhibitions. Altogether, our data indicate that xyloglucan utilization by R. cellulolyticum relies on an energy-saving central carbon metabolism deviating from current bacterial models, which efficiently prevents carbon overflow. IMPORTANCE The study of the decomposition of recalcitrant plant biomass is of great interest as the limiting step of terrestrial carbon cycle and to produce plant-derived valuable chemicals and energy. While extracellular cellulose degradation and catabolism have been studied in detail, few publications describe the complete metabolism of hemicelluloses and, to date, the published models are limited to the extracellular degradation and sequential entry of simple sugars. Here, we describe how the model anaerobic bacterium Ruminiclostridium cellulolyticum deals with the synchronous intracellular release of glucose, galactose, xylose, and cellobiose upon cytosolic depolymerization of imported xyloglucan oligosaccharides. The described novel metabolic strategy involves the simultaneous activity of different metabolic pathways coupled to a network of inhibitions controlling the carbon flux and is distinct from the ubiquitously observed sequential uptake and metabolism of carbohydrates known as the diauxic shift. Our results highlight the diversity of cellular responses related to a complex environment.


Firmicutes/metabolism , Glucans/metabolism , Xylans/metabolism , Anaerobiosis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cellobiose/metabolism , Firmicutes/genetics , Firmicutes/growth & development , Glucose/metabolism , Hexokinase/genetics , Hexokinase/metabolism , Metabolic Networks and Pathways , Polysaccharides/metabolism
4.
Food Funct ; 12(19): 9456-9465, 2021 Oct 04.
Article En | MEDLINE | ID: mdl-34606528

Exopolysaccharides (EPSs) derived from Lactobacilli have important physiological effects and are commonly used as new prebiotics. We identified and studied a new Lactobacillus strain, YY-112, isolated from waxberry (Myrica rubra). This strain, identified as Lactobacillus pentosus, tolerates acids, bile salts, and artificial digestive fluids. The EPS derived from this strain weighed 5.9 × 104 Da and contained glucose, mannose, glucosamine, galactose, and rhamnose at 62.69 : 85.85 : 2.46 : 2.92 : 1.00 molar ratios. We found that the EPS from this strain increased the ratio of Bacteroidetes to Firmicutes and decreased the relative abundance of Proteobacteria, especially Escherichia-Shigella, when added to a simulated gastrointestinal system in vitro. After analysing the short-chain fatty acids, we found that this EPS promoted the production of acetic acid, propionic acid, and butyric acid, and reduced the ratio of acetic acid to propionic acid. We conclude that Lactobacillus pentosus YY-112 is a potential probiotic strain with EPS that is beneficial for the intestinal microbiota and short-chain fatty acid production.


Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Lactobacillus pentosus/metabolism , Polysaccharides, Bacterial , Prebiotics , Bacteroidetes/growth & development , Cheek , Digestion , Fatty Acids, Volatile/metabolism , Fermentation , Firmicutes/growth & development , Gastrointestinal Tract/metabolism , Humans , In Vitro Techniques , Lactobacillus pentosus/classification , Phylogeny , Polysaccharides, Bacterial/biosynthesis , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/isolation & purification , Polysaccharides, Bacterial/metabolism , Proteobacteria/growth & development
5.
Biomolecules ; 11(8)2021 08 04.
Article En | MEDLINE | ID: mdl-34439821

Irritable bowel syndrome is not a life-threatening disease, yet it significantly affects the quality of life and contributes to economic loss. It is estimated that even up to 45% of the world's population can suffer from the disease. The first attempts to diagnose irritable bowel syndrome were made at the end of the 19th century; however, establishing appropriate diagnostic criteria and treatment methods is still ongoing. To date, little is known about the etiology of irritable bowel syndrome; however, growing attention is drawn to the intestinal microbiota as a factor in the disease development. For this reason, researchers have conducted many studies on therapies that modulate the microbiota, among which probiotics, prebiotics, and synbiotics are widely studied. To date, most studies have examined probiotics; however, there are also several studies demonstrating the efficacy of prebiotics and synbiotics. The aim of this review was to summarize findings on the usefulness of probiotics, prebiotics, and synbiotics in the treatment of irritable bowel syndrome.


Dysbiosis/diet therapy , Gastrointestinal Microbiome/drug effects , Irritable Bowel Syndrome/diet therapy , Prebiotics/administration & dosage , Probiotics/administration & dosage , Synbiotics/administration & dosage , Actinobacteria/genetics , Actinobacteria/growth & development , Actinobacteria/isolation & purification , Anti-Bacterial Agents/adverse effects , Bacteroidetes/genetics , Bacteroidetes/growth & development , Bacteroidetes/isolation & purification , Clinical Trials as Topic , Dysbiosis/etiology , Dysbiosis/microbiology , Dysbiosis/pathology , Firmicutes/genetics , Firmicutes/growth & development , Firmicutes/isolation & purification , Fusobacteria/genetics , Fusobacteria/growth & development , Fusobacteria/isolation & purification , Gastrointestinal Microbiome/genetics , Humans , Irritable Bowel Syndrome/microbiology , Irritable Bowel Syndrome/pathology , Proteobacteria/genetics , Proteobacteria/growth & development , Proteobacteria/isolation & purification , Quality of Life
6.
Genome Biol ; 22(1): 204, 2021 08 05.
Article En | MEDLINE | ID: mdl-34348764

BACKGROUND: Human-to-human transmission of symbiotic, anaerobic bacteria is a fundamental evolutionary adaptation essential for membership of the human gut microbiota. However, despite its importance, the genomic and biological adaptations underpinning symbiont transmission remain poorly understood. The Firmicutes are a dominant phylum within the intestinal microbiota that are capable of producing resistant endospores that maintain viability within the environment and germinate within the intestine to facilitate transmission. However, the impact of host transmission on the evolutionary and adaptive processes within the intestinal microbiota remains unknown. RESULTS: We analyze 1358 genomes of Firmicutes bacteria derived from host and environment-associated habitats. Characterization of genomes as spore-forming based on the presence of sporulation-predictive genes reveals multiple losses of sporulation in many distinct lineages. Loss of sporulation in gut Firmicutes is associated with features of host-adaptation such as genome reduction and specialized metabolic capabilities. Consistent with these data, analysis of 9966 gut metagenomes from adults around the world demonstrates that bacteria now incapable of sporulation are more abundant within individuals but less prevalent in the human population compared to spore-forming bacteria. CONCLUSIONS: Our results suggest host adaptation in gut Firmicutes is an evolutionary trade-off between transmission range and colonization abundance. We reveal host transmission as an underappreciated process that shapes the evolution, assembly, and functions of gut Firmicutes.


Firmicutes/genetics , Gastrointestinal Microbiome/genetics , Genome, Bacterial , Host Adaptation/genetics , Microbiota/genetics , Spores, Bacterial/genetics , Symbiosis/genetics , Anaerobiosis/genetics , Biological Evolution , Firmicutes/growth & development , Humans , Metagenome , Spores, Bacterial/growth & development
7.
Food Funct ; 12(18): 8635-8646, 2021 Sep 20.
Article En | MEDLINE | ID: mdl-34346464

Ulcerative colitis (UC) is a chronic lifetime disorder with a high incidence worldwide. A functional food-based method to prevent UC would be a good option for disease control. G. lemaneiformis oligosaccharides (GLOs) should have potent benefits for the gastrointestinal tract, based on in vitro fermentation assessed in our previous study. This study evaluated the therapeutic potential of GLOs in UC, as well as their possible mechanisms of action. The administration of GLOs was able to reduce the severity of dextran sulphate sodium-induced colitis by protecting mice from weight loss, reductions in colon length, inflammatory infiltration, and colon damage. Gut microbiota composition analysis showed that at the phylum level, GLOs could restore the composition of Bacteroidetes and decrease the level of Firmicutes. Consistently, it increased the contents of beneficial microbial metabolites and short-chain fatty acids in the mouse colitis model. In conclusion, GLOs could comprise a promising functional food strategy to alleviate UC symptoms.


Colitis, Ulcerative/diet therapy , Gastrointestinal Microbiome , Gracilaria/chemistry , Oligosaccharides/administration & dosage , Oligosaccharides/chemistry , Animals , Animals, Outbred Strains , Bacteroidetes/growth & development , Carbohydrate Conformation , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Colon/immunology , Colon/metabolism , Colon/pathology , Cytokines/metabolism , Dextran Sulfate , Disease Models, Animal , Fatty Acids, Volatile/metabolism , Firmicutes/growth & development , Functional Food , Intestines/metabolism , Intestines/microbiology , Male , Mice
8.
Science ; 373(6551): 181-186, 2021 07 09.
Article En | MEDLINE | ID: mdl-34244407

Relatives have more similar gut microbiomes than nonrelatives, but the degree to which this similarity results from shared genotypes versus shared environments has been controversial. Here, we leveraged 16,234 gut microbiome profiles, collected over 14 years from 585 wild baboons, to reveal that host genetic effects on the gut microbiome are nearly universal. Controlling for diet, age, and socioecological variation, 97% of microbiome phenotypes were significantly heritable, including several reported as heritable in humans. Heritability was typically low (mean = 0.068) but was systematically greater in the dry season, with low diet diversity, and in older hosts. We show that longitudinal profiles and large sample sizes are crucial to quantifying microbiome heritability, and indicate scope for selection on microbiome characteristics as a host phenotype.


Bacteria/classification , Environment , Gastrointestinal Microbiome/genetics , Papio/microbiology , Actinobacteria/classification , Actinobacteria/genetics , Actinobacteria/growth & development , Actinobacteria/isolation & purification , Aging , Animals , Bacteria/genetics , Bacteria/growth & development , Bacteria/isolation & purification , Bacteroidetes/classification , Bacteroidetes/genetics , Bacteroidetes/growth & development , Bacteroidetes/isolation & purification , Diet , Feces/microbiology , Female , Firmicutes/classification , Firmicutes/genetics , Firmicutes/growth & development , Firmicutes/isolation & purification , Genotype , Humans , Male , Papio/genetics , Phenotype , Seasons , Social Behavior
9.
BMC Microbiol ; 21(1): 165, 2021 06 03.
Article En | MEDLINE | ID: mdl-34082713

BACKGROUND: Lactobacillus rhamnosus GG (LGG) is the most widely used probiotic, but the mechanisms underlying its beneficial effects remain unresolved. Previous studies typically inoculated LGG in hosts with established gut microbiota, limiting the understanding of specific impacts of LGG on host due to numerous interactions among LGG, commensal microbes, and the host. There has been a scarcity of studies that used gnotobiotic animals to elucidate LGG-host interaction, in particular for gaining specific insights about how it modifies the metabolome. To evaluate whether LGG affects the metabolite output of pathobionts, we inoculated with LGG gnotobiotic mice containing Propionibacterium acnes, Turicibacter sanguinis, and Staphylococcus aureus (PTS). RESULTS: 16S rRNA sequencing of fecal samples by Ion Torrent and MinION platforms showed colonization of germ-free mice by PTS or by PTS plus LGG (LTS). Although the body weights and feeding rates of mice remained similar between PTS and LTS groups, co-associating LGG with PTS led to a pronounced reduction in abundance of P. acnes in the gut. Addition of LGG or its secretome inhibited P. acnes growth in culture. After optimizing procedures for fecal metabolite extraction and metabolomic liquid chromatography-mass spectrometry analysis, unsupervised and supervised multivariate analyses revealed a distinct separation among fecal metabolites of PTS, LTS, and germ-free groups. Variables-important-in-projection scores showed that LGG colonization robustly diminished guanine, ornitihine, and sorbitol while significantly elevating acetylated amino acids, ribitol, indolelactic acid, and histamine. In addition, carnitine, betaine, and glutamate increased while thymidine, quinic acid and biotin were reduced in both PTS and LTS groups. Furthermore, LGG association reduced intestinal mucosal expression levels of inflammatory cytokines, such as IL-1α, IL-1ß and TNF-α. CONCLUSIONS: LGG co-association had a negative impact on colonization of P. acnes, and markedly altered the metabolic output and inflammatory response elicited by pathobionts.


Gram-Positive Bacterial Infections/microbiology , Lacticaseibacillus rhamnosus/metabolism , Probiotics/administration & dosage , Animals , Cytokines/genetics , Cytokines/metabolism , Female , Firmicutes/growth & development , Firmicutes/physiology , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Gram-Positive Bacterial Infections/genetics , Gram-Positive Bacterial Infections/metabolism , Humans , Lacticaseibacillus rhamnosus/genetics , Male , Mice , Mice, Inbred C57BL , Propionibacterium acnes/growth & development , Propionibacterium acnes/physiology , Staphylococcus aureus/growth & development , Staphylococcus aureus/physiology
10.
Food Funct ; 12(9): 3954-3964, 2021 May 11.
Article En | MEDLINE | ID: mdl-33977937

The therapeutic effects of water extract of ginseng (WEG) on exercise-induced fatigue (EF) have been reported in several previous studies, but the molecular mechanisms involved remain unexplored. In this study, the anti-EF effects of WEG were studied, and the potential mechanisms were discussed. We characterized the chemical components of WEG by ultra-high performance liquid chromatography-tandem triple quadrupole mass spectrometry (UHPLC-QqQ-MS/MS) and high performance liquid chromatography coupled with evaporative light scattering detection (HPLC-ELSD), and then examined the anti-EF effects of WEG on a rat model of weight-loaded swimming with a focus on endogenous metabolism and gut microbiota. WEG contains abundant (90.15%, w/w) saccharides and ginsenosides with structurally diverse glycosyls. WEG taken orally showed strong anti-EF effects by ameliorating energy metabolism abnormality, oxidative stress, lipid peroxidation, inflammatory response, disorders in the metabolism of bile acid, amino acid, fatty acid and lipid, as well as the gut microbiota dysbiosis. Given that gut microbiota is significantly associated with energy expenditure, systemic inflammation and host metabolism, these findings suggest a potential central role of the gut microbiota in mediating the anti-EF effect of WEG. That is, the saccharides and ginsenosides in WEG serve as energy substrates for specific intestinal bacteria, thereby beneficially regulating the gut microbiota, and the reshaped gut microbial ecosystem then triggers several molecular and cellular signaling pathways (e.g. butyrate or TGR5 signals) to achieve the therapeutic effects on EF. The outcomes highlighted here enable deeper insight into how WEG overcomes EF.


Fatigue/drug therapy , Gastrointestinal Microbiome/drug effects , Panax , Physical Exertion , Plant Extracts/pharmacology , Amino Acids/metabolism , Animals , Bacteroidetes/classification , Bacteroidetes/growth & development , Bacteroidetes/isolation & purification , Bile Acids and Salts/metabolism , Dysbiosis , Fatigue/etiology , Fatty Acids/metabolism , Firmicutes/classification , Firmicutes/growth & development , Firmicutes/isolation & purification , Lipid Metabolism , Male , Metabolome , Phytotherapy , Plant Extracts/chemistry , Plant Extracts/therapeutic use , Rats , Rats, Sprague-Dawley , Swimming
11.
PLoS One ; 16(4): e0250354, 2021.
Article En | MEDLINE | ID: mdl-33872333

Constipation is a common condition that affects individuals of all ages, and prolonged constipation needs to be prevented to avoid potential complications and reduce the additional stress on individuals with pre-medical conditions. This study aimed to evaluate the effects of heat-inactivated Lactobacillus plantarum (HLp-nF1) on loperamide-induced constipation in rats. Constipation-induced male rats were treated orally with low to high doses of HLp-nF1 and an anti-constipation medication Dulcolax for five weeks. Study has 8 groups, control group; loperamide-treated group; Dulcolax-treated group; treatment with 3.2 × 1010, 8 × 1010 and 1.6 × 1011, cells/mL HLp-nF1; Loperamide + Dulcolax treated group. HLp-nF1 treated rats showed improvements in fecal pellet number, weight, water content, intestinal transit length, and contractility compared to the constipation-induced rats. Also, an increase in the intestine mucosal layer thickness and the number of mucin-producing crypt epithelial cells were observed in HLp-nF1-treated groups. Further, the levels of inflammatory cytokines levels were significantly downregulated by treatment with HLp-nF1 and Dulcolax. Notably, the metagenomics sequencing analysis demonstrated a similar genus pattern to the pre-preparation group and control with HLp-nF1 treatment. In conclusion, the administration of >3.2 × 1010 cells/mL HLp-nF1 has a positive impact on the constipated rats overall health.


Constipation/therapy , Gastrointestinal Transit/drug effects , Intestinal Mucosa/drug effects , Lactobacillus plantarum/physiology , Laxatives/pharmacology , Metagenome , Actinobacteria/genetics , Actinobacteria/growth & development , Actinobacteria/isolation & purification , Animals , Bacteroidetes/genetics , Bacteroidetes/growth & development , Bacteroidetes/isolation & purification , Bisacodyl/pharmacology , Constipation/chemically induced , Constipation/microbiology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Feces/microbiology , Firmicutes/genetics , Firmicutes/growth & development , Firmicutes/isolation & purification , Gastrointestinal Transit/physiology , Gene Expression/drug effects , Hot Temperature , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Intestinal Mucosa/microbiology , Loperamide/adverse effects , Male , Microbial Viability , Proteobacteria/genetics , Proteobacteria/growth & development , Proteobacteria/isolation & purification , RNA, Ribosomal, 16S/genetics , Rats , Rats, Sprague-Dawley , Treatment Outcome , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Verrucomicrobia/genetics , Verrucomicrobia/growth & development , Verrucomicrobia/isolation & purification
12.
PLoS One ; 16(4): e0250344, 2021.
Article En | MEDLINE | ID: mdl-33872339

Canine acute hemorrhagic diarrhea syndrome (AHDS) has been associated in some studies with Clostridioides perfringens overgrowth and toxin-mediated necrosis of the intestinal mucosa. We aimed to determine the effect of a single fecal microbiota transplantation (FMT) on clinical scores and fecal microbiomes of 1 and 7 dogs with AHDS from New Zealand and South Africa. We hypothesized that FMT would improve AHDS clinical scores and increase microbiota alpha-diversity and short-chain fatty acid (SCFA)-producing microbial communities' abundances in dogs with AHDS after FMT. We sequenced the V3-V4 region of the 16S-rRNA gene in the feces of AHDS FMT-recipients and sham-treated control dogs, and their healthy donors at admission, discharge, and 30 days post-discharge. There were no significant differences in median AHDS clinical scores between FMT-recipients and sham-treated controls at admission or discharge (P = 0.22, P = 0.41). At admission, the Shannon diversity index (SDI) was lower in AHDS dogs than healthy donors (P = 0.002). The SDI did not change from admission to 30 days in sham-treated dogs yet increased in FMT-recipients from admission to discharge (P = 0.04) to levels not different than donors (P = 0.33) but significantly higher than sham-treated controls (P = 0.002). At 30 days, the SDI did not differ between FMT recipients, sham-treated controls, and donors (P = 0.88). Principal coordinate analysis of the Bray-Curtis index separated post-FMT and donor dogs from pre-FMT and sham-treated dogs (P = 0.009) because of increased SCFA-producing genera's abundances after FMT. A single co-abundance subnetwork contained many of the same OTUs found to be differentially abundant in FMT-recipients, and the abundance of this module was increased in FMT-recipients at discharge and 30 days, compared to sham-treated controls. We conclude in this small pilot study FMT did not have any clinical benefit. A single FMT procedure has the potential to increase bacterial communities of SCFA-producing genera important for intestinal health up to 30 days post-FMT.


Clostridioides/pathogenicity , Clostridium Infections/therapy , Diarrhea/therapy , Fecal Microbiota Transplantation/veterinary , Feces/microbiology , Gastrointestinal Hemorrhage/therapy , Gastrointestinal Microbiome/physiology , Actinobacteria/genetics , Actinobacteria/growth & development , Actinobacteria/isolation & purification , Animals , Bacteroidetes/genetics , Bacteroidetes/growth & development , Bacteroidetes/isolation & purification , Clostridioides/genetics , Clostridioides/growth & development , Clostridium Infections/microbiology , Clostridium Infections/pathology , Clostridium Infections/veterinary , Diarrhea/microbiology , Diarrhea/pathology , Diarrhea/veterinary , Dogs , Fatty Acids, Volatile/biosynthesis , Female , Firmicutes/genetics , Firmicutes/growth & development , Firmicutes/isolation & purification , Fusobacteria/genetics , Fusobacteria/growth & development , Fusobacteria/isolation & purification , Gastrointestinal Hemorrhage/microbiology , Gastrointestinal Hemorrhage/pathology , Gastrointestinal Hemorrhage/veterinary , Intestinal Mucosa/microbiology , Male , New Zealand , Pilot Projects , Prospective Studies , Proteobacteria/genetics , Proteobacteria/growth & development , Proteobacteria/isolation & purification , RNA, Ribosomal, 16S/genetics , South Africa
13.
PLoS One ; 16(4): e0250423, 2021.
Article En | MEDLINE | ID: mdl-33914799

The tight association between malnutrition and gut microbiota (GM) dysbiosis enables microbiota-targeting intervention to be a promising strategy. Thus, we used a malnourished pig model to investigate the host response and GM alterations under different diet supplementation strategies. Pigs at age of 4 weeks were fed with pure maize diet to induce malnutrition symptoms, and followed by continuous feeding with maize (Maize, n = 8) or re-feeding using either corn-soy-blend (CSB+, n = 10) or millet-soy-blend based (MSB+, n = 10) supplementary food for 3 weeks. Meanwhile, 8 pigs were fed on a standard formulated ration as control (Ref). The effect of nutritional supplementation was assessed by the growth status, blood chemistry, gastrointestinal pathology, mucosal microbiota composition and colon production of short-chain fatty acids. Compared with purely maize-fed pigs, both CSB+ and MSB+ elevated the concentrations of total protein and globulin in blood. These pigs still showed most malnutrition symptoms after the food intervention period. MSB+ had superior influence on the GM development, exhibiting better performance in both structural and functional aspects. MSB+ pigs were colonized by less Proteobacteria but more Bacteroidetes, Firmicutes and Lachnospira spp. Pearson's correlation analysis indicated a strong correlation between the abundance of mucosal e.g., Faecalibacterium and Lachnospira spp. and body weight, crown-rump length and total serum protein. In conclusion, the malnutrition symptoms were accompanied by an aberrant GM, and millet-based nutritional supplementation showed promising potentials to restore the reduced GM diversity implicated in pig malnutrition.


Animal Feed/analysis , Diet/methods , Dysbiosis/diet therapy , Gastrointestinal Microbiome/physiology , Malnutrition/diet therapy , Millets/chemistry , Animals , Bacteroidetes/genetics , Bacteroidetes/growth & development , Bacteroidetes/isolation & purification , Biodiversity , Blood Proteins/agonists , Blood Proteins/metabolism , Body Weight , Clostridiales/genetics , Clostridiales/growth & development , Clostridiales/isolation & purification , Dysbiosis/microbiology , Dysbiosis/pathology , Faecalibacterium/genetics , Faecalibacterium/growth & development , Faecalibacterium/isolation & purification , Fatty Acids, Volatile/biosynthesis , Female , Firmicutes/genetics , Firmicutes/growth & development , Firmicutes/isolation & purification , Malnutrition/microbiology , Malnutrition/pathology , Proteobacteria/genetics , Proteobacteria/growth & development , Proteobacteria/isolation & purification , RNA, Ribosomal, 16S/genetics , Glycine max/chemistry , Swine , Verrucomicrobia/genetics , Verrucomicrobia/growth & development , Verrucomicrobia/isolation & purification , Zea mays/chemistry
14.
Nutrients ; 13(3)2021 Mar 10.
Article En | MEDLINE | ID: mdl-33801901

Trilobatin was identified as the primary bioactive component in the Lithocarpus polystachyus Rehd (LPR) leaves. This study explored the antiobesity effect of trilobatin from LPR leaves and its influence on gut microbiota in obese rats. Results showed that trilobatin could significantly reduce body and liver weight gain induced by a high-fat diet, and the accumulation of perirenal fat, epididymal fat, and brown fat of SD (Male Sprague-Dawley) obese rats in a dose-independent manner. Short-chain fatty acids (SCFAs) concentrations increased, especially the concentration of butyrate. Trilobatin supplementation could significantly increase the relative abundance of Lactobacillus, Prevotella, CF231, Bacteroides, and Oscillospira, and decrease greatly the abundance of Blautia, Allobaculum, Phascolarctobacterium, and Coprococcus, resulting in an increase of the ratio of Bacteroidetes to Firmicutes (except the genera of Lactobacillus and Oscillospira). The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway predicted by the Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) indicated the different relative metabolic pathways after trilobatin supplementation. This study may reveal the contribution of gut microbiota to the antiobesity effect of trilobatin from LPR leaves and predict the potential regulatory mechanism for obesity induced by a high-fat diet.


Anti-Obesity Agents/pharmacology , Diet, High-Fat , Dietary Supplements , Flavonoids/pharmacology , Gastrointestinal Microbiome/drug effects , Obesity/microbiology , Polyphenols/pharmacology , Animals , Anti-Obesity Agents/administration & dosage , Bacteroidetes/classification , Bacteroidetes/growth & development , Body Weight/drug effects , Fagaceae/chemistry , Fatty Acids, Volatile/analysis , Firmicutes/classification , Firmicutes/growth & development , Flavonoids/administration & dosage , Liver/drug effects , Male , Metabolic Networks and Pathways/drug effects , Obesity/etiology , Obesity/metabolism , Organ Size/drug effects , Plant Leaves/chemistry , Polyphenols/administration & dosage , Rats , Rats, Sprague-Dawley
15.
Biosci Biotechnol Biochem ; 85(5): 1235-1242, 2021 Apr 24.
Article En | MEDLINE | ID: mdl-33674867

The role of Fusobacterium nucleatum, often associated with intestinal diseases, in the remission of dextran sulfate sodium (DSS)-induced colitis was investigated. Female mice were divided into groups DC (DSS control) and DF (DSS + F. nucleatum). F. nucleatum (1.0 × 1010 cfu/mouse/day) in phosphate-buffered saline (PBS) was orally given to DF, while DC had PBS only. All mice had DSS in drinking water. In Experiment 1, mice underwent 2 inflammation phases, an in-between recovery phase and had their disease activity indices (DAI) calculated. Experiment 2 was similarly conducted, except that mice were dissected 3 days postrecovery, and had blood and colonic mucosal samples collected. In Experiment 1, DF had significantly (P < .05) higher DAI than DC, during the recovery and 2nd inflammation phases. In Experiment 2, genus Bacteroides was significantly (P < .05) higher and family Lachnospiraceae significantly lower in cecal mucosa-associated microbiota of DF than in that of DC. We concluded that F. nucleatum can impede colitis remission.


Colitis/microbiology , Colon/microbiology , Fusobacterium nucleatum/pathogenicity , Intestinal Mucosa/microbiology , Actinobacteria/genetics , Actinobacteria/growth & development , Actinobacteria/isolation & purification , Animals , Bacteroidetes/genetics , Bacteroidetes/growth & development , Bacteroidetes/isolation & purification , Colitis/chemically induced , Colitis/pathology , Colon/pathology , Convalescence , Dextran Sulfate/administration & dosage , Disease Models, Animal , Female , Firmicutes/genetics , Firmicutes/growth & development , Firmicutes/isolation & purification , Fusobacterium nucleatum/growth & development , Fusobacterium nucleatum/isolation & purification , Gastrointestinal Microbiome/genetics , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Permeability , Proteobacteria/genetics , Proteobacteria/growth & development , Proteobacteria/isolation & purification , RNA, Bacterial/genetics
16.
Commun Biol ; 4(1): 307, 2021 03 08.
Article En | MEDLINE | ID: mdl-33686191

The deep biosphere contains members from all three domains of life along with viruses. Here we investigate the deep terrestrial virosphere by sequencing community nucleic acids from three groundwaters of contrasting chemistries, origins, and ages. These viromes constitute a highly unique community compared to other environmental viromes and sequenced viral isolates. Viral host prediction suggests that many of the viruses are associated with Firmicutes and Patescibacteria, a superphylum lacking previously described active viruses. RNA transcript-based activity implies viral predation in the shallower marine water-fed groundwater, while the deeper and more oligotrophic waters appear to be in 'metabolic standby'. Viral encoded antibiotic production and resistance systems suggest competition and antagonistic interactions. The data demonstrate a viral community with a wide range of predicted hosts that mediates nutrient recycling to support a higher microbial turnover than previously anticipated. This suggests the presence of 'kill-the-winner' oscillations creating slow motion 'boom and burst' cycles.


Groundwater/virology , Virome , Virus Replication , Viruses/growth & development , Firmicutes/growth & development , Firmicutes/virology , Groundwater/microbiology , Host-Pathogen Interactions , Metagenomics , Population Density , Time Factors , Viruses/genetics , Viruses/metabolism , Water Microbiology
17.
Poult Sci ; 100(2): 810-819, 2021 Feb.
Article En | MEDLINE | ID: mdl-33518135

This study determined the effects of dietary supplementation of rhamnolipids (RLS) on the growth performance, gut morphology, immune function, intestinal volatile fatty acid, and microflora community in Linnan yellow broilers. A total of 480 1-day-old broiler chicks were randomly assigned to groups for supplementation with one of the following for 56 d: no supplement (control), 30 mg/kg bacitracin (ANT), 500 mg/kg RLS, or 1,000 mg/kg RLS (RLS2). The RLS2 diet was found to improve the final BW and ADG on day 56. The RLS diet reduced jejunal crypt depth, increased jejunal villus length, and increased serum IgA, IgM, IgY, IL-1ß, IL-6, and tumor necrosis factor-alpha (TNF-α) levels. The RLS broilers had higher cecum concentrations of acetic acid, propionic acid, butyrate, isobutyric acid, valerate, and isovalerate. High-throughput sequencing indicated that RLS affected microbial quantity and diversity in the cecum. Bacterial richness was higher in the RLS broilers than the ANT broilers. The RLS broilers had higher relative abundances of Megasphaera hypermegale and Lachnospiraceae bacterium 19gly4 on day 28 and Clostridium spiroforme and Alistipes obesi on day 56. These results suggest that RLS supplementation improves growth performance, benefits the intestinal villus morphology, regulates host immune function, and raises intestinal volatile fatty acid content and the relative abundance of the gut microbiota in broiler chickens.


Animal Feed , Chickens , Glycolipids/administration & dosage , Intestines/physiology , Animal Feed/analysis , Animals , Bacteroidetes/growth & development , Chickens/classification , Chickens/growth & development , Chickens/physiology , Diet/veterinary , Dietary Supplements , Firmicutes/growth & development , Intestines/growth & development , Intestines/microbiology , Random Allocation
18.
Medicine (Baltimore) ; 100(7): e24845, 2021 Feb 19.
Article En | MEDLINE | ID: mdl-33607855

ABSTRACT: Despite the establishment of the links between ulcerative colitis (UC) and depression, between UC and gut microbiota, few correlations between depression and gut microbiota have yet been demonstrated especially in ulcerative colitis patients. The objective of our study was therefore to determine whether the comorbidity of depressive disorder in ulcerative colitis patients correlate with alterations in the gut microbiota and to identify the specific microbiota signatures associated with depression.Between March 2017 and February 2018, 31 healthy volunteers, 31 UC patients without depression, and 31 UC patients with depression from Longhua Hospital were enrolled. Clinical data and fecal samples were collected for each patient. Fecal bacteria were identified using 16 s rRNA sequencing. We compared microbial composition among the 3 groups using bioinformatic analysis.Patients with UC with depression had higher disease severity (P < .05). The UC without depression group had moderate reduction of microbial abundance and uniformity compared to the control group. The UC with depression group had the lowest microbial abundance. With regard to the vital bacteria in the microbiota-gut-brain axis, patients with UC and depression had the lowest abundance of Firmicutes, Clostridia, and Clostridiales but the highest abundance of Proteobacteria, Gammaproteobacteria, and Bacilli.The presence of depression in UC patients presented significant differences in the composition of gut microbiota compared with UC patients without depression, with increased abundance of Firmicutes and reduced abundance of Proteobacteria.


Colitis, Ulcerative/microbiology , Depression/microbiology , Feces/microbiology , Gastrointestinal Microbiome/genetics , Adult , Bacteria/classification , Bacteria/genetics , Bacteria/growth & development , Case-Control Studies , Clostridiales/growth & development , Colitis, Ulcerative/psychology , Comorbidity , Computational Biology/methods , Depression/complications , Female , Firmicutes/growth & development , Gammaproteobacteria/growth & development , Humans , Male , Middle Aged , Pilot Projects , Proteobacteria/growth & development , RNA, Ribosomal, 16S/genetics , Severity of Illness Index
19.
Sci Rep ; 11(1): 1421, 2021 01 14.
Article En | MEDLINE | ID: mdl-33446825

Gut microbiota composition can modulate neuroendocrine function, inflammation, and cellular and immunological responses against different pathogens, including viruses. Zika virus (ZIKV) can infect adult immunocompetent individuals and trigger brain damage and antiviral responses. However, it is not known whether ZIKV infection could impact the gut microbiome from adult immunocompetent mice. Here, we investigated modifications induced by ZIKV infection in the gut microbiome of immunocompetent C57BL/6J mice. Adult C57BL/6J mice were infected with ZIKV and the gut microbiota composition was analyzed by next-generation sequencing of the V4 hypervariable region present in the bacterial 16S rDNA gene. Our data showed that ZIKV infection triggered a significant decrease in the bacteria belonging to Actinobacteria and Firmicutes phyla, and increased Deferribacteres and Spirochaetes phyla components compared to uninfected mice. Interestingly, ZIKV infection triggered a significant increase in the abundance of bacteria from the Spirochaetaceae family in the gut microbiota. Lastly, we demonstrated that modulation of microbiota induced by ZIKV infection may lead to intestinal epithelium damage and intense leukocyte recruitment to the intestinal mucosa. Taken together, our data demonstrate that ZIKV infection can impact the gut microbiota composition and colon tissue homeostasis in adult immunocompetent mice.


Firmicutes , Gastrointestinal Microbiome , Intestinal Mucosa , Spirochaetaceae , Zika Virus Infection , Zika Virus/metabolism , Animals , Firmicutes/classification , Firmicutes/growth & development , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/virology , Mice , Spirochaetaceae/classification , Spirochaetaceae/growth & development , Zika Virus Infection/metabolism , Zika Virus Infection/microbiology
20.
Nutr Res ; 85: 47-59, 2021 01.
Article En | MEDLINE | ID: mdl-33444970

Regular almond consumption has been shown to improve body weight management, lipid profile and blood glucose control. We hypothesized that almond consumption would alter fecal microbiota composition, including increased abundance and activity of potentially beneficial bacterial taxa in adults who are overweight and obese with elevated fasting blood glucose. A total of 69 adults who were overweight or obese with an elevated plasma glucose (age: 60.8 ± 7.4, BMI ≥27 kg/m2, fasting plasma glucose ≥5.6 to <7.0 mmol/L) were randomized to daily consumption of either 2 servings of almonds (AS:56 g/day) or an isocaloric, high carbohydrate biscuit snack for 8 weeks. AS but not biscuit snack experienced significant changes in microbiota composition (P= .011) and increases in bacterial richness, evenness, and diversity (P< .01). Increases in both the relative and absolute abundance of operational taxonomic units in the Ruminococcaceae family, including Ruminiclostridium (false discovery rate P = .002), Ruminococcaceae NK4A214 (P = .002) and Ruminococcaceae UCG-003 (P = .002) were the principal drivers of microbiota-level changes. No changes in fecal short chain fatty acid levels, or in the carriage of the gene encoding butyryl-CoA:acetate CoA-transferase (an enzyme involved in butyrate synthesis) occurred. Almond consumption was not associated with reduced gut permeability, but fecal pH (P= .0006) and moisture content (P = .027) decreased significantly in AS when compared to BS. Regular almond consumption increased the abundance of potentially beneficial ruminococci in the fecal microbiota in individuals with elevated blood glucose. However, fecal short-chain fatty acid levels remained unaltered and the capacity for such microbiological effects to precipitate host benefit is not known.


Blood Glucose/analysis , Feces/chemistry , Firmicutes/classification , Gastrointestinal Microbiome , Nuts , Obesity , Overweight , Prunus dulcis , Bacteria/classification , Bacteria/growth & development , Eating , Fatty Acids, Volatile/analysis , Feces/microbiology , Female , Firmicutes/growth & development , Humans , Hydrogen-Ion Concentration , Male , Middle Aged , Obesity/blood , Obesity/microbiology , Overweight/blood , Overweight/microbiology
...