Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cancer Epidemiol Biomarkers Prev ; 32(2): 281-286, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36512731

ABSTRACT

BACKGROUND: Mechanistic data indicate the benefit of short-chain fatty acids (SCFA) produced by gut microbial fermentation of fiber on colorectal cancer, but direct epidemiologic evidence is limited. A recent study identified SNPs for two SCFA traits (fecal propionate and butyrate-producing microbiome pathway PWY-5022) in Europeans and showed metabolic benefits. METHODS: We conducted a two-sample Mendelian randomization analysis of the genetic instruments for the two SCFA traits (three SNPs for fecal propionate and nine for PWY-5022) in relation to colorectal cancer risk in three large European genetic consortia of 58,131 colorectal cancer cases and 67,347 controls. We estimated the risk of overall colorectal cancer and conducted subgroup analyses by sex, age, and anatomic subsites of colorectal cancer. RESULTS: We did not observe strong evidence for an association of the genetic predictors for fecal propionate levels and the abundance of PWY-5022 with the risk of overall colorectal cancer, colorectal cancer by sex, or early-onset colorectal cancer (diagnosed at <50 years), with no evidence of heterogeneity or pleiotropy. When assessed by tumor subsites, we found weak evidence for an association between PWY-5022 and risk of rectal cancer (OR per 1-SD, 0.95; 95% confidence intervals, 0.91-0.99; P = 0.03) but it did not surpass multiple testing of subgroup analysis. CONCLUSIONS: Genetic instruments for fecal propionate levels and the abundance of PWY-5022 were not associated with colorectal cancer risk. IMPACT: Fecal propionate and PWY-5022 may not have a substantial influence on colorectal cancer risk. Future research is warranted to comprehensively investigate the effects of SCFA-producing bacteria and SCFAs on colorectal cancer risk.


Subject(s)
Butyrates , Colorectal Neoplasms , Feces , Gastrointestinal Microbiome , Propionates , Humans , Butyrates/analysis , Butyrates/metabolism , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Fatty Acids, Volatile/analysis , Fatty Acids, Volatile/genetics , Fatty Acids, Volatile/metabolism , Feces/chemistry , Feces/microbiology , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Mendelian Randomization Analysis , Propionates/analysis , Propionates/metabolism , Risk , Europe/epidemiology
2.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166266, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34481869

ABSTRACT

Sexual dimorphism exists in the onset and development of type 1 diabetes (T1D), but its potential pathological mechanism is poorly understood. In the present study, we examined sex-specific changes in the gut microbiome and host metabolome of T1D mice via 16S rRNA gene sequencing and nuclear magnetic resonance (NMR)-based metabolomics approach, and aimed to investigate potential mechanism of the gut microbiota-host metabolic interaction in the sexual dimorphism of T1D. Our results demonstrate that female mice had a greater shift in the gut microbiota than male mice during the development of T1D; however, host metabolome was more susceptible to T1D in male mice. The correlation network analysis indicates that T1D-induced host metabolic changes may be regulated by the gut microbiota in a sex-specific manner, mainly involving short-chain fatty acids (SCFAs) metabolism, energy metabolism, amino acid metabolism, and choline metabolism. Therefore, our study suggests that sex-dependent "gut microbiota-host metabolism axis" may be implicated in the sexual dimorphism of T1D, and the link between microbes and metabolites might contribute to the prevention and treatment of T1D.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Fatty Acids, Volatile/genetics , Gastrointestinal Microbiome/genetics , Metabolome/genetics , Animals , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Fatty Acids, Volatile/metabolism , Female , Humans , Magnetic Resonance Spectroscopy , Male , Metabolomics , Mice , Mice, Inbred NOD/genetics , Mice, Inbred NOD/metabolism , Sex Characteristics
3.
Int J Mol Sci ; 22(11)2021 Jun 01.
Article in English | MEDLINE | ID: mdl-34206021

ABSTRACT

Apolipoprotein A-I (ApoA-I) is the major protein of high density lipoprotein (HDL) particles and has a crucial role in reverse cholesterol transport (RCT). It has been postulated that elevating production of de novo ApoA-I might translate into the formation of new functional HDL particles that could lower cardiovascular disease (CVD) risk via RCT. During inflammation, serum ApoA-I concentrations are reduced, which contributes to the development of dysfunctional HDL particles as Serum Amyloid A (SAA) overtakes the position of ApoA-I within the HDL particles. Therefore, instead of elevating serum HDL cholesterol concentrations, rescuing lower serum ApoA-I concentrations could be beneficial in both normal and inflamed conditions. Several nutritional compounds, amongst others short chain fatty acids (SCFAs), have shown their capacity to modulate hepatic lipoprotein metabolism. In this review we provide an overview of HDL and more specific ApoA-I metabolism, SCFAs physiology and the current knowledge regarding the influence of SCFAs on ApoA-I expression and synthesis in human liver cells. We conclude that the current evidence regarding the effect of SCFAs on ApoA-I transcription and secretion is promising, however there is a need to investigate which dietary fibres could lead to increased SCFAs formation and consequent elevated ApoA-I concentrations.


Subject(s)
Apolipoprotein A-I/genetics , Fatty Acids, Volatile/metabolism , Inflammation/genetics , Liver/metabolism , Apolipoprotein A-I/metabolism , Cholesterol/genetics , Cholesterol/metabolism , Fatty Acids, Volatile/genetics , Humans , Inflammation/metabolism , Inflammation/pathology , Serum Amyloid A Protein/genetics , Serum Amyloid A Protein/metabolism
4.
Sci Rep ; 11(1): 5244, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33664396

ABSTRACT

Altered composition of gut bacteria and changes to the production of their bioactive metabolites, the short-chain fatty acids (SCFAs), have been implicated in the development of multiple sclerosis (MS). However, the immunomodulatory actions of SCFAs and intermediaries in their ability to influence MS pathogenesis are uncertain. In this study, levels of serum SCFAs were correlated with immune cell abundance and phenotype as well as with other relevant serum factors in blood samples taken at first presentation of Clinically Isolated Syndrome (CIS; an early form of MS) or MS and compared to healthy controls. There was a small but significant reduction in propionate levels in the serum of patients with CIS or MS compared with healthy controls. The frequencies of circulating T follicular regulatory cells and T follicular helper cells were significantly positively correlated with serum levels of propionate. Levels of butyrate associated positively with frequencies of IL-10-producing B-cells and negatively with frequencies of class-switched memory B-cells. TNF production by polyclonally-activated B-cells correlated negatively with acetate levels. Levels of serum SCFAs associated with changes in circulating immune cells and biomarkers implicated in the development of MS.


Subject(s)
Fatty Acids, Volatile/blood , Interleukin-10/genetics , Multiple Sclerosis/genetics , T-Lymphocytes, Regulatory/immunology , Adult , Fatty Acids, Volatile/genetics , Female , Healthy Volunteers , Humans , Interleukin-10/immunology , Male , Memory B Cells/immunology , Memory B Cells/microbiology , Middle Aged , Multiple Sclerosis/blood , Multiple Sclerosis/microbiology , Multiple Sclerosis/pathology , Propionates/blood , T Follicular Helper Cells/immunology , T Follicular Helper Cells/microbiology , T-Lymphocytes, Regulatory/microbiology
5.
Int J Mol Sci ; 21(23)2020 Nov 25.
Article in English | MEDLINE | ID: mdl-33255677

ABSTRACT

The gut microbiota could influence the pathophysiology of age-related sarcopenia through multiple mechanisms implying modulation of chronic inflammation and anabolic resistance. The aim of this study was to compare the fecal microbiota composition and functionality, assessed by shotgun metagenomics sequencing, between two groups of elderly outpatients, differing only for the presence of primary sarcopenia. Five sarcopenic elderly subjects and twelve non-sarcopenic controls, classified according to lower limb function and bioimpedance-derived skeletal muscle index, provided a stool sample, which was analyzed with shotgun metagenomics approaches, to determine the overall microbiota composition, the representation of bacteria at the species level, and the prediction of bacterial genes involved in functional metabolic pathways. Sarcopenic subjects displayed different fecal microbiota compositions at the species level, with significant depletion of two species known for their metabolic capacity of producing short-chain fatty acids (SCFAs), Faecalibacterium prausnitzii and Roseburia inulinivorans, and of Alistipes shahii. Additionally, their fecal metagenome had different representation of genes belonging to 108 metabolic pathways, namely, depletion of genes involved in SCFA synthesis, carotenoid and isoflavone biotransformation, and amino acid interconversion. These results support the hypothesis of an association between microbiota and sarcopenia, indicating novel possible mediators, whose clinical relevance should be investigated in future studies.


Subject(s)
Aging/genetics , Gastrointestinal Microbiome/genetics , Metagenome/genetics , Sarcopenia/genetics , Aged , Aged, 80 and over , Aging/pathology , Bacteroidetes/genetics , Clostridiales/genetics , Faecalibacterium prausnitzii/genetics , Fatty Acids, Volatile/biosynthesis , Fatty Acids, Volatile/genetics , Feces/microbiology , Female , Humans , Inflammation/genetics , Inflammation/microbiology , Inflammation/pathology , Male , Metabolic Networks and Pathways , Metagenomics/methods , Muscle, Skeletal/microbiology , Muscle, Skeletal/physiopathology , Sarcopenia/microbiology , Sarcopenia/physiopathology
6.
Molecules ; 25(20)2020 Oct 17.
Article in English | MEDLINE | ID: mdl-33080809

ABSTRACT

Fermented alcoholic drinks' contribution to the gut microbiota composition is mostly unknown. However, intestinal microorganisms can use compounds present in beer. This work explored the associations between moderate consumption of beer, microbiota composition, and short chain fatty acid (SCFA) profile. Seventy eight subjects were selected from a 261 healthy adult cohort on the basis of their alcohol consumption pattern. Two groups were compared: (1) abstainers or occasional consumption (ABS) (n = 44; <1.5 alcohol g/day), and (2) beer consumption ≥70% of total alcohol (BEER) (n = 34; 200 to 600 mL 5% vol. beer/day; <15 mL 13% vol. wine/day; <15 mL 40% vol. spirits/day). Gut microbiota composition (16S rRNA gene sequencing) and SCFA concentration were analyzed in fecal samples. No differences were found in α and ß diversity between groups. The relative abundance of gut bacteria showed that Clostridiaceae was lower (p = 0.009), while Blautia and Pseudobutyrivibrio were higher (p = 0.044 and p = 0.037, respectively) in BEER versus ABS. In addition, Alkaliphilus, in men, showed lower abundance in BEER than in ABS (p = 0.025). Butyric acid was higher in BEER than in ABS (p = 0.032), and correlated with Pseudobutyrivibrio abundance. In conclusion, the changes observed in a few taxa, and the higher butyric acid concentration in consumers versus non-consumers of beer, suggest a potentially beneficial effect of moderate beer consumption on intestinal health.


Subject(s)
Alcoholic Beverages/microbiology , Beer/microbiology , Fatty Acids, Volatile/genetics , Gastrointestinal Microbiome/genetics , Adult , Alcoholic Beverages/adverse effects , Butyric Acid/chemistry , Butyric Acid/metabolism , Fatty Acids, Volatile/metabolism , Female , Humans , Male , Polyphenols/chemistry , RNA, Ribosomal, 16S/genetics
7.
Sci Rep ; 10(1): 15101, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32934296

ABSTRACT

In light of recent host-microbial association studies, a consensus is evolving that species composition of the gastrointestinal microbiota is a polygenic trait governed by interactions between host genetic factors and the environment. Here, we investigated the effect of host genetic factors in shaping the bacterial species composition in the rumen by performing a genome-wide association study. Using a common set of 61,974 single-nucleotide polymorphisms found in cattle genomes (n = 586) and corresponding rumen bacterial community composition, we identified operational taxonomic units (OTUs), Families and Phyla with high heritability. The top associations (1-Mb windows) were located on 7 chromosomes. These regions were associated with the rumen microbiota in multiple ways; some (chromosome 19; position 3.0-4.0 Mb) are associated with closely related taxa (Prevotellaceae, Paraprevotellaceae, and RF16), some (chromosome 27; position 3.0-4.0 Mb) are associated with distantly related taxa (Prevotellaceae, Fibrobacteraceae, RF16, RFP12, S24-7, Lentisphaerae, and Tenericutes) and others (chromosome 23; position 0.0-1.0) associated with both related and unrelated taxa. The annotated genes associated with identified genomic regions suggest the associations observed are directed toward selective absorption of volatile fatty acids from the rumen to increase energy availability to the host. This study demonstrates that host genetics affects rumen bacterial community composition.


Subject(s)
Bacteria/genetics , Gastrointestinal Microbiome/genetics , Microbiota/genetics , Rumen/microbiology , Animal Feed/microbiology , Animals , Cattle , Fatty Acids, Volatile/genetics , Genome-Wide Association Study
8.
PLoS One ; 15(3): e0230200, 2020.
Article in English | MEDLINE | ID: mdl-32214324

ABSTRACT

Statins, a class of drugs that can effectively remove cholesterol from serum, are used to regulate plasma total cholesterol and reduce the risk of cardiovascular diseases, but it is still unclear whether the drug are modulated by gut microbiota or the structures of gut microbiota are shaped by statins. We investigated the interactions between statins and the human gut microbiota during the in vitro fermentation process by 16S rRNA gene sequencing, gas chromatography (GC), and high-performance liquid chromatography (HPLC) analyses. The presence of fluvastatin (FLU2) specifically promoted the growth of Escherichia/Shigella, Ruminococcaceae UCG 014, and Sutterella. However, the composition of the gut bacterial microbiota remained relatively static in samples treated with rosuvastatin (ROS), simvastatin (SIM), and atorvastatin (ATO). The PICRUSt program predicted moderate differences in the functional categories related to the biosynthesis of other secondary metabolites, cellular processes and signaling, and signal transduction in the FLU2 fermentation samples. Our study revealed substantial variation in the structure and function of microbiomes from the FLU2-treated samples. In addition, short-chain fatty acids (SCFAs) were also significantly decreased in FLU2-treated samples compared with the samples treated with other stains. Statins can be degraded by the human gut microbiota in vitro, and the degradation rate was approximately 7%-30% and 19%-48% after fermentation was allowed to proceed for 24 h and 48 h, respectively. Generally, FLU2 could largely shape the composition and function of human gut microbiota, which resulted in changes in the production of SCFAs. In turn, all statins could be degraded or modified by the gut microbiota. Our study paves the way for elucidating statin-gut microbiota interactions in vitro towards the improvement of the host health and personalized medicine.


Subject(s)
Bacteria/drug effects , Gastrointestinal Microbiome/drug effects , Microbiota/drug effects , Adolescent , Adult , Bacteria/genetics , Cardiovascular Diseases/drug therapy , Fatty Acids, Volatile/genetics , Feces/microbiology , Female , Fermentation/drug effects , Fermentation/genetics , Gastrointestinal Microbiome/genetics , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Male , Microbiota/genetics , RNA, Ribosomal, 16S/genetics , Young Adult
9.
Nat Genet ; 51(4): 600-605, 2019 04.
Article in English | MEDLINE | ID: mdl-30778224

ABSTRACT

Microbiome-wide association studies on large population cohorts have highlighted associations between the gut microbiome and complex traits, including type 2 diabetes (T2D) and obesity1. However, the causal relationships remain largely unresolved. We leveraged information from 952 normoglycemic individuals for whom genome-wide genotyping, gut metagenomic sequence and fecal short-chain fatty acid (SCFA) levels were available2, then combined this information with genome-wide-association summary statistics for 17 metabolic and anthropometric traits. Using bidirectional Mendelian randomization (MR) analyses to assess causality3, we found that the host-genetic-driven increase in gut production of the SCFA butyrate was associated with improved insulin response after an oral glucose-tolerance test (P = 9.8 × 10-5), whereas abnormalities in the production or absorption of another SCFA, propionate, were causally related to an increased risk of T2D (P = 0.004). These data provide evidence of a causal effect of the gut microbiome on metabolic traits and support the use of MR as a means to elucidate causal relationships from microbiome-wide association findings.


Subject(s)
Fatty Acids, Volatile/genetics , Gastrointestinal Microbiome/genetics , Metabolic Diseases/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Diabetes Mellitus, Type 2/genetics , Female , Genotype , Glucose Tolerance Test/methods , Humans , Male , Middle Aged , Netherlands , Young Adult
10.
Med Vet Entomol ; 32(3): 358-364, 2018 09.
Article in English | MEDLINE | ID: mdl-29858503

ABSTRACT

Physically disturbed Triatoma infestans (Hemiptera: Reduviidae) adults, as well as adults of other Chagas' disease vectors, secrete a mix of volatile organic compounds (VOCs) with alarm and possible sexual and defence functions. The aim of the present research was to test whether infection with the entomopathogenic fungus Beauveria bassiana (Ascomycota: Hypocreales: Clavicipitaceae) has an effect on VOC secretion in disturbed T. infestans and on the expression of two genes (Ti-brnq and Ti-bckdc) potentially involved in VOC biosynthesis. The volatiles released by insects at different time periods after fungal treatment were identified and their relative amounts measured. Isobutyric acid was the most abundant volatile found in both healthy and fungus-infected insects and underwent no significant relative changes through the infection process. The secretion of propionic acid, however, was significantly higher at 1-4 days post-infection (d.p. i.) compared with that in controls. A slight induction of both Ti-brnq and Ti-bckdc genes was found by real-time polymerase chain reaction at 4 d.p. i., with expression values reaching up to three-fold those in controls. The early stages of fungal infection seem to affect the composition of the alarm pheromone by changing the expression pattern of both genes analysed. These results help to elucidate the impact of fungal infections on the chemical ecology of triatomine bugs.


Subject(s)
Beauveria/physiology , Fatty Acids, Volatile/metabolism , Insect Proteins/genetics , Triatoma/metabolism , Triatoma/microbiology , Animals , Fatty Acids, Volatile/genetics , Insect Proteins/metabolism , Insect Vectors/genetics , Insect Vectors/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Triatoma/genetics
11.
PLoS One ; 13(5): e0196961, 2018.
Article in English | MEDLINE | ID: mdl-29742146

ABSTRACT

Stressor-exposure has been shown to exacerbate inflammation and change the composition of the gastrointestinal microbiota; however stressor-induced effects on microbiota-derived metabolites and their receptors are unknown. Thus, bacterial-produced short chain fatty acids (SCFAs), as well as microbial community composition, were assessed in the colons of mice exposed to stress during infection with Citrobacter rodentium. Mice were exposed to overnight restraint on 7 consecutive nights, or left undisturbed as a control. After the first exposure of restraint, mice were orally challenged with C. rodentium or with vehicle. Microbial community composition was assessed using 16S rRNA gene sequencing and SCFA levels measured using gas chromatography-mass spectrometry (GC-MS). Pathogen levels and colonic inflammation were also assessed 6 days post-infection. Results demonstrated that the microbial community structure and SCFA production were significantly affected by both stressor exposure and C. rodentium-infection. Exposure to prolonged restraint in the absence of infection significantly reduced SCFAs (acetic acid, butyric acid, and propionic acid). Multiple bacterial taxa were affected by stressor exposure, with the relative abundance of Lactobacillus being significantly reduced and directly correlated with propionic acid. Lactobacillus abundances were inversely correlated with colonic inflammation, supporting the contention that Lactobacillus helps to regulate mucosal inflammatory responses. Our data indicates that restraint stressor can have significant effects on pathogen-induced colonic inflammation and suggest that stressor-induced changes in the microbiota, microbial-produced SCFAs and their receptors may be involved.


Subject(s)
Enterobacteriaceae Infections/microbiology , Gastrointestinal Microbiome/genetics , Inflammation/microbiology , Lactobacillus/genetics , Animals , Citrobacter rodentium/pathogenicity , Colon/microbiology , Colon/pathology , Enterobacteriaceae Infections/genetics , Fatty Acids, Volatile/biosynthesis , Fatty Acids, Volatile/genetics , Gastrointestinal Microbiome/physiology , Inflammation/genetics , Intestinal Mucosa/microbiology , Lactobacillus/physiology , Mice , Microbiota/genetics , Microbiota/physiology , RNA, Ribosomal, 16S/genetics , Restraint, Physical/methods
12.
Lipids ; 53(1): 77-84, 2018 01.
Article in English | MEDLINE | ID: mdl-29488641

ABSTRACT

Short-chain fatty acids (SCFA) such as acetic acid, propionic acid, and butyric acid are produced by fermentation by gut microbiota. In this paper, we investigate the effects of SCFA on 3T3-L1 cells and the underlying molecular mechanisms. The cells were treated with acetic acid, propionic acid, or butyric acid when cells were induced to differentiate into adipocytes. MTT assay was employed to detect the viability of 3T3-L1 cells. Oil Red O staining was used to visualize the lipid content in 3T3-L1 cells. A triglyceride assay kit was used to detect the triacylglycerol content in 3T3-L1 cells. qRT-PCR and Western blot were used to evaluate the expression of metabolic enzymes. MTT results showed that safe concentrations of acetic acid, propionic acid, and butyric acid were less than 6.4, 3.2, and 0.8 mM, respectively. Oil Red O staining and triacylglycerols detection results showed that treatment with acetic acid, propionic acid, and butyric acid accelerated the 3T3-L1 adipocyte differentiation. qRT-PCR and Western blot results showed that the expressions of lipoprotein lipase (LPL), adipocyte fatty acid binding protein 4 (FABP4), fatty acid transporter protein 4 (FATP4), and fatty acid synthase (FAS) were significantly increased by acetic acid, propionic acid, and butyric acid treatment during adipose differentiation (p < 0.05). In conclusion, SCFA promoted lipid accumulation by modulating the expression of enzymes of fatty acid metabolism.


Subject(s)
Fatty Acid Synthases/genetics , Fatty Acid Transport Proteins/genetics , Fatty Acid-Binding Proteins/genetics , Lipoprotein Lipase/genetics , 3T3-L1 Cells , Acetic Acid/chemistry , Acetic Acid/metabolism , Adipocytes/metabolism , Adipogenesis/genetics , Animals , Butyric Acid/chemistry , Butyric Acid/metabolism , Cell Differentiation/genetics , Fatty Acid Synthases/metabolism , Fatty Acids, Volatile/genetics , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/genetics , Gene Expression Regulation, Enzymologic , Lipid Metabolism/genetics , Mice , Propionates/chemistry , Propionates/metabolism , Triglycerides/metabolism
13.
Sci Rep ; 7(1): 15876, 2017 Nov 20.
Article in English | MEDLINE | ID: mdl-29158587

ABSTRACT

Male Tsumura Suzuki obese diabetes (TSOD) mice spontaneously develop obesity and obesity-related metabolic syndrome. Gut dysbiosis, an imbalance of gut microbiota, has been implicated in the pathogenesis of metabolic syndrome, but its mechanisms are unknown. Short-chain fatty acids (SCFAs) are the main fermentation products of gut microbiota and a link between the gut microbiota and the host's physiology. Here, we investigated a correlation among gut dysbiosis, SCFAs, and metabolic syndrome in TSOD mice. We detected enriched levels of Gram-positive bacteria and corresponding decreases in Gram-negative bacteria in 24-wk-old metabolic syndrome-affected TSOD mice compared with age-matched controls. The abundance of Bacteroidetes species decreased, the abundance of Firmicutes species increased, and nine genera of bacteria were altered in 24-wk-old TSOD mice. The total plasma SCFA level was significantly lower in the TSOD mice than in controls. The major plasma SCFA-acetate-decreased in TSOD mice, whereas propionate and butyrate increased. TSOD mice had no minor SCFAs (valerate and hexanoate) but normal mice did. We thus concluded that gut dysbiosis and consequent disruptions in plasma SCFA profiles occurred in metabolic syndrome-affected TSOD mice. We also propose that the TSOD mouse is a useful model to study gut dysbiosis, SCFAs, and metabolic syndrome.


Subject(s)
Diabetes Mellitus/genetics , Fatty Acids, Volatile/blood , Gastrointestinal Microbiome/genetics , Metabolic Syndrome/genetics , Animals , Bacteroidetes/genetics , Bacteroidetes/metabolism , Diabetes Mellitus/microbiology , Diabetes Mellitus/pathology , Disease Models, Animal , Dysbiosis/blood , Dysbiosis/genetics , Dysbiosis/microbiology , Fatty Acids, Volatile/genetics , Humans , Metabolic Syndrome/blood , Metabolic Syndrome/microbiology , Mice , Mice, Obese , Obesity/blood , Obesity/genetics , Obesity/microbiology
14.
Sci Rep ; 7(1): 2816, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28588227

ABSTRACT

To better understand the function role of the melon CmLOX18 gene in the biosynthesis of C6 volatiles during fruit ripening, we biochemically characterized CmLOX18 and identified its subcellular localization in transgenic tomato plants. Heterologous expression in yeast cells showed that the molecular weight of the CmLOX18 protein was identical to that predicted, and that this enzyme possesseed lipoxygenase activity. Linoleic acid was demonstrated to be the preferred substrate for the purified recombinant CmLOX18 protein, which exhibited optimal catalytic activity at pH 4.5 and 30 °C. Chromatogram analysis of the reaction product indicated that the CmLOX18 protein exhibited positional specificity, as evidenced by its release of only a C-13 oxidized product. Subcellular localization analysis by transient expression in Arabidopsis protoplasts showed that CmLOX18 was localized to non-chloroplast organelles. When the CmLOX18 gene was transgenically expressed in tomato via Agrobacterium tumefaciens-mediated transformation, it was shown to enhance expression levels of the tomato hydroperoxide lyase gene LeHPL, whereas the expression levels of six TomLox genes were little changed. Furthermore, transgenic tomato fruits exhibited increases in the content of the C6 volatiles, namely hexanal, (Z)-3-hexanal, and (Z)-3-hexen-1-ol, indicating that CmLOX18 probably plays an important role in the synthesis of C6 compounds in fruits.


Subject(s)
Cucurbitaceae/genetics , Fatty Acids, Volatile/biosynthesis , Fruit/genetics , Lipoxygenase/genetics , Agrobacterium tumefaciens/genetics , Aldehyde-Lyases/genetics , Arabidopsis/genetics , Chloroplasts/genetics , Cucurbitaceae/enzymology , Cucurbitaceae/growth & development , Cytochrome P-450 Enzyme System/genetics , Fatty Acids, Volatile/genetics , Fruit/growth & development , Fruit/metabolism , Gene Expression Regulation, Plant , Linoleic Acid/genetics , Linoleic Acid/metabolism , Lipoxygenase/metabolism , Solanum lycopersicum/enzymology , Solanum lycopersicum/genetics , Plants, Genetically Modified
15.
mBio ; 8(3)2017 05 09.
Article in English | MEDLINE | ID: mdl-28487428

ABSTRACT

Campylobacter jejuni promotes commensalism in the intestinal tracts of avian hosts and diarrheal disease in humans, yet components of intestinal environments recognized as spatial cues specific for different intestinal regions by the bacterium to initiate interactions in either host are mostly unknown. By analyzing a C. jejuni acetogenesis mutant defective in converting acetyl coenzyme A (Ac-CoA) to acetate and commensal colonization of young chicks, we discovered evidence for in vivo microbiota-derived short-chain fatty acids (SCFAs) and organic acids as cues recognized by C. jejuni that modulate expression of determinants required for commensalism. We identified a set of C. jejuni genes encoding catabolic enzymes and transport systems for amino acids required for in vivo growth whose expression was modulated by SCFAs. Transcription of these genes was reduced in the acetogenesis mutant but was restored upon supplementation with physiological concentrations of the SCFAs acetate and butyrate present in the lower intestinal tracts of avian and human hosts. Conversely, the organic acid lactate, which is abundant in the upper intestinal tract where C. jejuni colonizes less efficiently, reduced expression of these genes. We propose that microbiota-generated SCFAs and lactate are cues for C. jejuni to discriminate between different intestinal regions. Spatial gradients of these metabolites likely allow C. jejuni to locate preferred niches in the lower intestinal tract and induce expression of factors required for intestinal growth and commensal colonization. Our findings provide insights into the types of cues C. jejuni monitors in the avian host for commensalism and likely in humans to promote diarrheal disease.IMPORTANCECampylobacter jejuni is a commensal of the intestinal tracts of avian species and other animals and a leading cause of diarrheal disease in humans. The types of cues sensed by C. jejuni to influence responses to promote commensalism or infection are largely lacking. By analyzing a C. jejuni acetogenesis mutant, we discovered a set of genes whose expression is modulated by lactate and short-chain fatty acids produced by the microbiota in the intestinal tract. These genes include those encoding catabolic enzymes and transport systems for amino acids that are required by C. jejuni for in vivo growth and intestinal colonization. We propose that gradients of these microbiota-generated metabolites are cues for spatial discrimination between areas of the intestines so that the bacterium can locate niches in the lower intestinal tract for optimal growth for commensalism in avian species and possibly infection of human hosts leading to diarrheal disease.


Subject(s)
Campylobacter jejuni/physiology , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/physiology , Symbiosis , Acetates/metabolism , Acetates/pharmacology , Acetyl Coenzyme A/metabolism , Animals , Butyrates/pharmacology , Campylobacter Infections/microbiology , Campylobacter jejuni/drug effects , Campylobacter jejuni/genetics , Campylobacter jejuni/pathogenicity , Chickens/microbiology , Fatty Acids, Volatile/biosynthesis , Fatty Acids, Volatile/genetics , Fatty Acids, Volatile/pharmacology , Gene Expression Regulation, Bacterial , Humans , Intestines/microbiology , Lactates/metabolism , Symbiosis/genetics , Virulence/genetics
16.
Nutrients ; 9(4)2017 Mar 27.
Article in English | MEDLINE | ID: mdl-28346392

ABSTRACT

We have demonstrated that polyphenol-rich sorghum bran diets alter fecal microbiota; however, little is known regarding their effect on colon inflammation. Our aim was to characterize the effect of sorghum bran diets on intestinal homeostasis during dextran sodium sulfate (DSS)-induced colitis. Male Sprague-Dawley rats (N = 20/diet) were provided diets containing 6% fiber from cellulose, or Black (3-deoxyanthocyanins), Sumac (condensed tannins) or Hi Tannin Black (both) sorghum bran. Colitis was induced (N = 10/diet) with three separate 48-h exposures to 3% DSS, and feces were collected. On Day 82, animals were euthanized and the colon resected. Only discrete mucosal lesions, with no diarrhea or bloody stools, were observed in DSS rats. Only bran diets upregulated proliferation and Tff3, Tgfß and short chain fatty acids (SCFA) transporter expression after a DSS challenge. DSS did not significantly affect fecal SCFA concentrations. Bran diets alone upregulated repair mechanisms and SCFA transporter expression, which suggests these polyphenol-rich sorghum brans may suppress some consequences of colitis.


Subject(s)
Colitis/diet therapy , Diet , Dietary Fiber/administration & dosage , Sorghum/chemistry , Animals , Apoptosis , Cell Proliferation , Colitis/chemically induced , Dextran Sulfate , Disease Models, Animal , Edible Grain/chemistry , Epithelial Cells/metabolism , Fatty Acids, Volatile/genetics , Fatty Acids, Volatile/metabolism , Feces/chemistry , Intestinal Mucosa/metabolism , Male , NF-kappa B/genetics , NF-kappa B/metabolism , Polyphenols/administration & dosage , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Trefoil Factor-3/genetics , Trefoil Factor-3/metabolism
17.
J Nutr Biochem ; 26(1): 91-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25459886

ABSTRACT

We have previously shown that high-protein (HP) diet ingestion causes marked changes in the luminal environment of the colonic epithelium. This study aimed to evaluate the impact of such modifications on small intestinal and colonic mucosa, two segments with different transit time and physiological functions. Rats were fed with either normal protein (NP; 14% protein) or HP (53% protein) isocaloric diet for 2 weeks, and parameters related to intestinal mucous-secreting cells and to several innate/adaptive immune characteristics (myeloperoxidase activity, cytokine and epithelial TLR expression, proportion of immune cells in gut-associated lymphoid tissues) were measured in the ileum and colon. In ileum from HP animals, we observed hyperplasia of mucus-producing cells concomitant with an increased expression of Muc2 at both gene and protein levels, reduction of mucosal myeloperoxidase activity, down-regulation of Tlr4 gene expression in enterocytes and down-regulation of mucosal Th cytokines associated with CD4+ lymphocyte reduction in mesenteric lymph nodes. These changes coincided with an increased amount of acetate in the ileal luminal content. In colon, HP diet ingestion resulted in a lower number of goblet cells at the epithelial surface but increased goblet cell number in colonic crypts together with an increased Muc3 and a slight reduction of Il-6 gene expression. Our data suggest that HP diet modifies the goblet cell distribution in colon and, in ileum, increases goblet cell activity and decreases parameters related to basal gut inflammatory status. The impact of HP diet on intestinal mucosa in terms of beneficial or deleterious effects is discussed.


Subject(s)
Colon/drug effects , Dietary Proteins/administration & dosage , Goblet Cells/drug effects , Ileum/drug effects , Acetates/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Colon/metabolism , Diet , Down-Regulation , Enterocytes/drug effects , Enterocytes/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fatty Acids, Volatile/genetics , Fatty Acids, Volatile/metabolism , Ileum/metabolism , Immunoglobulin A/genetics , Immunoglobulin A/metabolism , Immunohistochemistry , Interleukin-10/metabolism , Interleukin-13/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Mucin-3/genetics , Mucin-3/metabolism , Rats , Rats, Wistar , Tight Junctions/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Transforming Growth Factor beta1/metabolism , Up-Regulation
18.
Domest Anim Endocrinol ; 47: 108-18.e1, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24315753

ABSTRACT

Free fatty acid receptor 2 (FFA2, also called GPR43) is reported to play a critical role in mediating the actions of short-chain fatty acids (SCFAs) in humans and mice. However, little is known about the structure, functionality, and tissue expression of FFA2 in other mammalian species, including pigs. In the present study, the full-length cDNAs of FFA2 (pFFA2) and a novel FFA2-like gene (named pFFA2L) were cloned from pig intestines by reverse transcription PCR. Both cloned pFFA2 and pFFA2L are predicted to encode 2 receptors of remarkable structural similarity and share high amino acid sequence identities with FFA2 from other mammalian species. Interestingly, the novel FFA2L could also be identified in 9 other mammalian species, suggesting that FFA2L was likely duplicated from FFA2 in the last common ancestor of these species. With the use of a pGL4-SRE-luciferase reporter assay, we demonstrated that pFFA2 expressed in human embryonic kidney 293 cells could be activated by acetate, propionate, and butyrate equipotently, whereas pFFA2L could be activated only by acetate and propionate, indicating that both pFFA2 and pFFA2L are functional receptors for SCFAs with nonidentical pharmacologic properties. Reverse transcription PCR found that pFFA2 mRNA was widely expressed in nearly all tissues examined, including adipose tissue and gastrointestinal (GI) tract, whereas pFFA2L expression was mainly restricted to the GI tract. Taken together, our findings raise a novel concept that the actions of SCFAs are likely mediated by 2 FFA2s (FFA2 and FFA2L) in target tissues of some mammalian species, such as the GI tract of pigs.


Subject(s)
Fatty Acids, Volatile/metabolism , Gene Duplication/genetics , Gene Expression Regulation/physiology , Receptors, G-Protein-Coupled/metabolism , Swine/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , Fatty Acids, Volatile/genetics , Gene Duplication/physiology , HEK293 Cells , Humans , Molecular Sequence Data , Phylogeny , Receptors, G-Protein-Coupled/genetics , Swine/genetics
19.
J Vet Sci ; 14(4): 387-93, 2013.
Article in English | MEDLINE | ID: mdl-23820222

ABSTRACT

Prebiotics modulate microbial composition and ensure a healthy gastrointestinal tract environment that can prevent colon cancer development. These natural dietary compounds are therefore potential chemopreventive agents. Thirty Sprague-Dawley rats (4 months old) were experimentally treated with procarcinogen dimethylhydrazine to induce colon cancer development. The rats were randomly assigned to three groups: a control group (CG), a group treated with dimethylhydrazine (DMH), and a group given DMH and inulin, a prebiotic (DMH+PRE). The effects of inulin on the activities of bacterial glycolytic enzymes, short-chain fatty acids, coliform and lactobacilli counts, cytokine levels, and cyclooxygenase-2 (COX-2) and transcription nuclear factor kappa beta (NFκB) immunoreactivity were measured. Inulin significantly decreased coliform counts (p < 0.01), increased lactobacilli counts (p < 0.001), and decreased the activity of ß-glucuronidase (p < 0.01). Butyric and propionic concentrations were decreased in the DMH group. Inulin increased its concentration that had been reduced by DMH. Inulin decreased the numbers of COX-2- and NFκB-positive cells in the tunica mucosae and tela submucosae of the colon. The expression of IL-2, TNFα, and IL-10 was also diminished. This 28-week study showed that dietary intake of inulin prevents preneoplastic changes and inflammation that promote colon cancer development.


Subject(s)
Colonic Neoplasms/drug therapy , Inulin/metabolism , Prebiotics/analysis , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Colon/enzymology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/metabolism , Colony Count, Microbial , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokines/blood , Cytokines/genetics , Diet , Dietary Supplements/analysis , Dimethylhydrazines/toxicity , Enterobacteriaceae/drug effects , Enterobacteriaceae/physiology , Fatty Acids, Volatile/genetics , Fatty Acids, Volatile/metabolism , Female , Gene Expression Regulation/drug effects , Inulin/administration & dosage , Lactobacillaceae/drug effects , Lactobacillaceae/physiology , Male , NF-kappa B/genetics , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley
20.
Article in English | WPRIM (Western Pacific) | ID: wpr-197115

ABSTRACT

Prebiotics modulate microbial composition and ensure a healthy gastrointestinal tract environment that can prevent colon cancer development. These natural dietary compounds are therefore potential chemopreventive agents. Thirty Sprague-Dawley rats (4 months old) were experimentally treated with procarcinogen dimethylhydrazine to induce colon cancer development. The rats were randomly assigned to three groups: a control group (CG), a group treated with dimethylhydrazine (DMH), and a group given DMH and inulin, a prebiotic (DMH+PRE). The effects of inulin on the activities of bacterial glycolytic enzymes, short-chain fatty acids, coliform and lactobacilli counts, cytokine levels, and cyclooxygenase-2 (COX-2) and transcription nuclear factor kappa beta (NFkappaB) immunoreactivity were measured. Inulin significantly decreased coliform counts (p < 0.01), increased lactobacilli counts (p < 0.001), and decreased the activity of beta-glucuronidase (p < 0.01). Butyric and propionic concentrations were decreased in the DMH group. Inulin increased its concentration that had been reduced by DMH. Inulin decreased the numbers of COX-2- and NFkappaB-positive cells in the tunica mucosae and tela submucosae of the colon. The expression of IL-2, TNFalpha, and IL-10 was also diminished. This 28-week study showed that dietary intake of inulin prevents preneoplastic changes and inflammation that promote colon cancer development.


Subject(s)
Animals , Female , Male , Rats , Bacterial Proteins/genetics , Colon/enzymology , Colonic Neoplasms/chemically induced , Colony Count, Microbial , Cyclooxygenase 2/genetics , Cytokines/blood , Diet , Dietary Supplements/analysis , Dimethylhydrazines/toxicity , Enterobacteriaceae/drug effects , Fatty Acids, Volatile/genetics , Gene Expression Regulation/drug effects , Inulin/administration & dosage , Lactobacillaceae/drug effects , NF-kappa B/genetics , Prebiotics/analysis , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL