Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
J Nutr Biochem ; 101: 108926, 2022 03.
Article in English | MEDLINE | ID: mdl-34848335

ABSTRACT

Short-chain fatty acids (SCFAs) play a pivotal role in maintaining intestinal homeostasis. We aimed to investigate the effects of SCFA supplementation on gut inflammation and microbiota composition in a murine colitis model. Mice were fed with sodium butyrate or a mixture of SCFAs in the drinking water for 2 weeks, followed by 2% dextran sulfate sodium (DSS) for 7 d. After euthanasia, mouse colons were extracted to examine histological findings. Flow cytometry of the mouse colon tissues was performed to assess T cell differentiation. Changes in gut microbiota were assessed by high-throughput sequencing of the mouse feces. There were no significant differences in weight change, colonic length, or histologic inflammation score between the DSS, butyrate, and SCFA mix groups. However, flow cytometry revealed that both the expression of CD4+Foxp3+ regulatory T cells and of IL-17-producing T cells were increased in the butyrate and SCFA mix groups. Microbial compositions of the butyrate and SCFA mix groups were significantly different from those of the control and DSS groups in principal coordinate analysis. Relative abundances of the phyla Verrucomicrobia and Proteobacteria, species Akkermansia muciniphila and Escherichia fergusonii were increased in the butyrate and SCFA mix groups. Genera Roseburia and Lactobacillus showed a negative correlation with the degree of colitis, whereas genera Escherichia and Mucispirillum showed a positive correlation. SCFA supplementation did not result in a significant reduction in colon inflammation, but it promoted both regulatory T cell and IL-17-producing T cell expression, and increased both protective and aggressive gut microbiota.


Subject(s)
Butyrates/administration & dosage , Dietary Supplements , Fatty Acids, Volatile/administration & dosage , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Animals , Cell Differentiation , Colitis/immunology , Colitis/microbiology , Colitis/pathology , Colon/pathology , Disease Models, Animal , Feces/microbiology , Female , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Inflammatory Bowel Diseases/immunology , Mice , Mice, Inbred C57BL , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology
2.
Fish Shellfish Immunol ; 120: 560-568, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34958920

ABSTRACT

To evaluate the effects of dietary short chain fatty acids (SCFAs) on the intestinal health and innate immunity in crucian carp, a six-week feeding trial was carried out with following treatments: basal diet (BD), basal diet supplementation with 1% sodium acetate (BDSA), basal diet supplementation with 1% sodium propionate (BDSP) and basal diet supplementation with 1% sodium butyrate (BDSB). The results showed dietary BDSA, BDSP and BDSB could protect the host against oxidative stress by improving the activity of certain antioxidative enzymes (T-SOD, GSH-Px and CAT). Additionally, dietary SCFAs could enhance mucosal and humoral immune responses by improving certain innate immune parameters in serum and skin mucus productions (IgM, ACH50 and T-SOD). Furthermore, dietary BDSA and BDSP could up-regulate the expression of immune related genes (TNF-α, TGF-ß and IL-8) and tight junction protein genes (occludin and ZO-1). Dietary BDSB could also elevate the expression of IL-8, TGF-ß, ZO-1 and Occludin in the midgut. Although dietary differences of SCFAs didn't alter the α-diversity of the intestinal flora, they altered the core microbiota. Finally, the challenge trial showed that dietary basal diet supplementation with SCFAs could protect zebrafish against Aeromonas hydrophila. These results suggest that dietary SCFAs could improve innate immunity, modulate gut microbiota and increase disease resistance in the host, which indicated the potential of SCFAs as immunostimulants in aquaculture.


Subject(s)
Diet , Disease Resistance , Fatty Acids, Volatile , Fish Diseases , Gastrointestinal Microbiome , Zebrafish , Aeromonas hydrophila , Animal Feed/analysis , Animals , Antioxidants , Diet/veterinary , Dietary Supplements/analysis , Fatty Acids, Volatile/administration & dosage , Fish Diseases/immunology , Fish Diseases/microbiology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/veterinary , Interleukin-8 , Occludin , Superoxide Dismutase , Transforming Growth Factor beta , Zebrafish/immunology , Zebrafish/microbiology
3.
Clin Nutr ; 40(6): 3842-3851, 2021 06.
Article in English | MEDLINE | ID: mdl-34130031

ABSTRACT

BACKGROUND AND AIMS: Anastomotic leak (AL) is a major complication in colorectal surgery. Recent evidence suggests that the gut microbiota may affect healing and may cause or prevent AL. Butyrate is a beneficial short-chain fatty acid (SCFA) that is produced as a result of bacterial fermentation of dietary oligosaccharides and has been described as beneficial in the maintenance of colonic health. To assess the impact of oligosaccharides on colonic anastomotic healing in mice, we propose to modulate the microbiota with oligosaccharides to increase butyrate production via enhancement of butyrate-producing bacteria and, consequently, improve anastomotic healing in mice. METHODS: Animal experiments were conducted in mice that were subjected to diets supplemented with inulin, galacto-oligosaccharides (GOS) or cellulose, as a control, for two weeks before undergoing a surgical colonic anastomosis. Macroscopic and histological assessment of the anastomosis was performed. Extent of epithelial proliferation was assessed by Ki-67 immunohistochemistry. Gelatin zymography was used to evaluate the extent of matrix metalloproteinase (MMP) hydrolytic activity. RESULTS: Inulin and GOS diets were associated with increased butyrate production and better anastomotic healing. Histological analysis revealed an enhanced mucosal continuity, and this was associated with an increased re-epithelialization of the wound as determined by increased epithelial proliferation. Collagen concentration in peri-anastomotic tissue was higher with inulin and GOS diets and MMP activity, a marker of collagen degradation, was lower with both oligosaccharides. Inulin and GOS diets were further associated with lower bacterial translocation. CONCLUSIONS: Dietary supplementation with inulin and GOS may improve anastomotic healing and reinforce the gut barrier in mice.


Subject(s)
Anastomotic Leak/prevention & control , Colonic Diseases/surgery , Fatty Acids, Volatile/administration & dosage , Inulin/administration & dosage , Animals , Dietary Supplements , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Perioperative Period , Postoperative Complications/prevention & control , Treatment Outcome , Wound Healing
4.
Nutrients ; 13(3)2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33801984

ABSTRACT

Short-chain fatty acids (SCFAs) are microbial metabolites, mainly generated by the action of gut microbiota on dietary fibers. Acetate, propionate, and butyrate are the three main SCFAs produced typically in a 60:20:20 molar ratio in the colon. Acetate, propionate, and butyrate, when given individually as supplements, have shown a protective role in obesity and hyperglycemia; however, the sex-specific effects of a mixture of SCFAs, when given in 60:20:20 ratio, on the regulation of lipid metabolism and lipid profile are not known. Male and female Long-Evans rats were given a mixture of SCFAs (acetate, propionate, and butyrate; molar ratio 60:20:20) each day for seven days intraperitoneally; plasma and hepatic lipids, gene expression, and lipidomics profile were analyzed. SCFAs significantly decreased plasma and hepatic triglycerides and cholesterol in males, whereas the fatty acyl composition of cholesteryl esters, triglycerides, and phospholipids was modulated in females. SCFAs decreased the mRNA expression of hepatic acetyl-CoA carboxylase-1 in both males and females. Our findings demonstrate for the first time that SCFAs (60:20:20) improved plasma and hepatic lipid levels and fatty acyl composition in a manner that may provide cardio-protective and anti-inflammatory effects in both sexes, via independent mechanisms.


Subject(s)
Fatty Acids, Volatile/administration & dosage , Lipid Metabolism , Animals , Blood Glucose/analysis , Body Weight/drug effects , Cholesterol/metabolism , Cholesterol Esters/blood , Eating/drug effects , Fatty Acids, Nonesterified/metabolism , Female , Injections, Intraperitoneal , Liver/metabolism , Male , Rats , Rats, Long-Evans , Sex Characteristics , Triglycerides/blood , Triglycerides/metabolism
5.
J Virol ; 95(4)2021 01 28.
Article in English | MEDLINE | ID: mdl-33208449

ABSTRACT

This report evaluates a dietary manipulation approach to suppress the severity of ocular infections caused by herpes simplex virus infection. The virus causes chronic damage to the cornea that results from a T-cell-orchestrated inflammatory reaction to the infection. Lesion severity can be limited if cells with regulatory activity predominate over proinflammatory T cells and nonlymphoid inflammatory cells. In this report, we show that this outcome can be achieved by including the short-chain fatty acid (SCFA) salt sodium propionate (SP) in the drinking water. Animals given the SP supplement developed significantly fewer ocular lesions than those receiving no supplement. Corneas and lymphoid organs contained fewer CD4 Th1 and Th17 T cells, neutrophils, and macrophages than those of controls, but a higher frequency of regulatory T cells (Treg) was present. The inclusion of SP in cultures to induce CD4 T cell subsets in vitro reduced the magnitude of Th1 and Th17 responses but expanded Treg induction. Dietary manipulation was an effective approach to limit the severity of viral immuno-inflammatory lesions and may be worth exploring as a means to reduce the impact of herpetic lesions in humans.IMPORTANCE Herpetic lesions are a significant problem, and they are difficult to control with therapeutics. Our studies show that the severity of herpetic lesions in a mouse model can be diminished by changing the diet to include increased levels of SCFA, which act to inhibit the involvement of inflammatory T cells. We suggest that changing the diet to include higher levels of SCFA might be a useful approach to reducing the impact of recurrent herpetic lesions in humans.


Subject(s)
Cornea , Dietary Supplements , Fatty Acids, Volatile/administration & dosage , Keratitis, Herpetic/diet therapy , Propionates/administration & dosage , Animals , Cells, Cultured , Cornea/immunology , Cornea/virology , Herpesvirus 1, Human/immunology , Keratitis, Herpetic/immunology , Keratitis, Herpetic/virology , Macrophages/cytology , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/cytology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Regulatory/cytology
6.
PLoS One ; 15(6): e0232831, 2020.
Article in English | MEDLINE | ID: mdl-32497096

ABSTRACT

The burden of enteric pathogens in poultry is growing after the ban of antibiotic use in animal production. Organic acids gained attention as a possible alternative to antibiotics due to their antimicrobial activities, improved nutrient metabolism and performance. The current study was conducted to evaluate the effectiveness of organic acid blend on broilers cecal microbiota, histomorphometric measurements, and short-chain fatty acid production in Salmonella enterica serovar Typhimurium challenge model. Birds were divided into four treatments, including a negative control, positive control challenged with S. Typhimurium, group supplemented with an organic acid blend, and birds supplemented with organic acid blend and Salmonella challenged. Results illustrate significant differences in feed conversion ratios and production efficiency factor between treatment groups, however, the influence of organic acid supplement was marginal. Organic acid blend significantly increased cecal acetic and butyric acids concentrations when compared to unsupplemented groups and resulted in minor alterations of intestinal bacterial communities.


Subject(s)
Acetates/metabolism , Animal Feed , Butyrates/metabolism , Chickens/microbiology , Dietary Supplements , Fatty Acids/pharmacology , Gastrointestinal Microbiome/drug effects , Poultry Diseases/therapy , Salmonella Infections, Animal/therapy , Salmonella typhimurium/drug effects , Animals , Cecum/microbiology , Chickens/metabolism , Fatty Acids/administration & dosage , Fatty Acids, Volatile/administration & dosage , Fatty Acids, Volatile/pharmacology , Ileum/metabolism , Ileum/ultrastructure , Mannans/administration & dosage , Microvilli/ultrastructure , Poultry Diseases/microbiology , Poultry Diseases/prevention & control , Random Allocation , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/prevention & control , Salmonella typhimurium/isolation & purification , Salmonella typhimurium/metabolism
7.
J Therm Biol ; 89: 102520, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32364974

ABSTRACT

The present study was conducted to investigate the effects of four dietary fat types and two environmental temperatures on the hepatic mitochondrial energetic in male broilers exposed to heat stress. The birds were kept in two separate rooms at 24 °C or 36 °C from 32 to 42 d of age with four experimental groups in each room. The birds fed on the diets supplemented containing rich sources of long-chain saturated fatty acids (beef tallow), middle-length-chain saturated FA (coconut oil), monounsaturated FA (olive oil), or polyunsaturated FA (soybean oil) for ten days. At 36 °C, the highest body weight and lowest feed conversion ratio were recorded in the birds fed on the diets supplemented with coconut oil or beef tallow. Temperature and fat type significantly affected the activities of the mitochondrial electron transport chain complexes (P < 0.01). There was a significant interaction between the temperature and fat type (P < 0.01). Generally, electron transport chain complexes I-V enzymatic activities were decreased at 36 °C. The coconut oil-fed birds showed the highest complex I activity at both temperatures. The beef tallow-fed broilers showed the lowest complex II activity at 24 °C. In birds exposed to 36 °C, complex II activity was higher for birds fed saturated coconut oil or beef tallow than those feeding the unsaturated olive oil or soybean oil-supplemented diets. At 24 °C, the highest and lowest complex III activities were recorded for the coconut oil- and beef tallow-supplemented diets, respectively. At 36 °C, the activity of complex III was coconut oil > beef tallow > olive oil > soybean oil. At 24 °C, complex IV activity was highest in coconut oil- or soybean oil-fed broilers; and at 36 °C, complex IV showed the lowest activity in soybean oil-fed birds. The highest complex IV activity was observed in coconut oil-fed chickens followed by olive oil-fed and beef tallow-fed birds, respectively. At 24 or 36 °C, the highest and lowest complex V activity was observed in coconut oil-fed and soybean oil-fed chickens, respectively. ATP concentration and mitochondrial membrane potential were in the order of coconut oil > beef tallow > olive oil > soybean oil at both temperatures. Temperature and fat type significantly affected the avANT mRNA concentration. Exposure of broilers to 36 °C generally decreased the mRNA expression of avANT, with beef tallow- or coconut oil-supplemented birds showing a lower avANT mRNA expression than those receiving olive oil- or soybean oil-supplemented diets. These findings provide further information on the use of fat sources in the diet of heat stressed-broilers.


Subject(s)
Chickens/metabolism , Energy Metabolism , Fatty Acids, Volatile/pharmacology , Heat-Shock Response , Mitochondria, Liver/metabolism , Plant Oils/pharmacology , Adenosine Triphosphate/metabolism , Animals , Avian Proteins/metabolism , Dietary Supplements , Electron Transport Chain Complex Proteins/metabolism , Fatty Acids, Volatile/administration & dosage , Liver/drug effects , Liver/metabolism , Mitochondria, Liver/drug effects , Plant Oils/administration & dosage
8.
Prev Vet Med ; 178: 104983, 2020 May.
Article in English | MEDLINE | ID: mdl-32289616

ABSTRACT

In veal and dairy beef production systems, Holstein bull calves experience many stressors and excessive pathogen exposure, necessitating the use of antimicrobials for welfare and production reasons. The aim of this randomized clinical trial was to explore the effects of esterified fatty acids used as feed supplement on health, production and immune variables in veal calves. Different glycerol-esters of fatty acids were used: short chain fatty acid (SCFA)-based glycerol-mono- (C4) and tributyrate (C4), and medium chain fatty acid (MCFA)-based glycerol-monocaprylate/monocaprinate (C8/C10) and glycerol-monolaurate (C12) in two different doses. One hundred sixty eight calves (2-to 4-week-old) were randomly assigned to 6 treatment groups; tributyrate (0.5 g/animal/day); monobutyrate (1 g/animal/day); low C8/C10 (7 g/animal/day) and high C8/C10 (10 g/animal/day); low C12 (4 g/animal/day) and high C12 (6 g/animal/day) and a control group (CON). Duration of in-feed supplementation was 14 weeks. Average daily gain, bodyweight at 14 weeks on feed and slaughter weight were determined. Health monitoring consisted of clinical signs and repeated thoracic ultrasonography. After 4, 8 and 12 weeks of supplementation, the function of neutrophils, monocytes and peripheral blood mononuclear cells (PBMCs) was evaluated ex vivo by measuring reactive oxygen species (ROS) production by neutrophils and monocytes, proliferation of and cytokine release by PBMCs. Study power was based upon ROS production by neutrophils and treatment groups were too limited to detect significant differences in growth and health variables. Glycerol-ester supplementation resulted in different effects on immune cell function, depending on the type and dose of the glycerol-ester as well as duration of supplementation. Our main findings were increased secretion of interleukin IL-17A by PBMCs at 4 weeks of feed supplementation in high C8/C10 (P< 0.01), low C12 (P < 0.01) and monobutyrate (P< 0.01) groups, combined with decreased ROS production in neutrophils (P < 0.001) and monocytes (P < 0.05) in the high C8/C10 and monocytes (P < 0.05) in low C12 groups compared to the control animals. After 12 weeks on feed, ROS production by neutrophils (P < 0.001) and monocytes (P < 0.01) of monobutyrate and by monocytes (P < 0.01) of tributyrate groups was decreased compared to control calves. In summary, supplementation of glycerol-esters of MCFAs resulted in immune-modulatory effects, which did not manifest themselves in improved health and growth of calves under the conditions and limitations of this study. Especially doses of high C8/C10 and low C12 show potential to promote an early, robust pro-inflammatory response with diminished ROS production. This might be beneficial for clearance of pathogens in young calves in periods of stress and high pathogen load.


Subject(s)
Body Weight/drug effects , Cattle/physiology , Cytokines/metabolism , Esters/metabolism , Fatty Acids, Volatile/metabolism , Glycerol/metabolism , Reactive Oxygen Species/metabolism , Animal Feed/analysis , Animals , Cattle/growth & development , Cattle/immunology , Diet/veterinary , Dietary Supplements/analysis , Esters/administration & dosage , Fatty Acids, Volatile/administration & dosage , Glycerol/administration & dosage , Male , Weight Gain/drug effects
9.
J Neurosci ; 40(5): 1162-1173, 2020 01 29.
Article in English | MEDLINE | ID: mdl-31889008

ABSTRACT

Recovery after stroke is a multicellular process encompassing neurons, resident immune cells, and brain-invading cells. Stroke alters the gut microbiome, which in turn has considerable impact on stroke outcome. However, the mechanisms underlying gut-brain interaction and implications for long-term recovery are largely elusive. Here, we tested the hypothesis that short-chain fatty acids (SCFAs), key bioactive microbial metabolites, are the missing link along the gut-brain axis and might be able to modulate recovery after experimental stroke. SCFA supplementation in the drinking water of male mice significantly improved recovery of affected limb motor function. Using in vivo wide-field calcium imaging, we observed that SCFAs induced altered contralesional cortex connectivity. This was associated with SCFA-dependent changes in spine and synapse densities. RNA sequencing of the forebrain cortex indicated a potential involvement of microglial cells in contributing to the structural and functional remodeling. Further analyses confirmed a substantial impact of SCFAs on microglial activation, which depended on the recruitment of T cells to the infarcted brain. Our findings identified that microbiota-derived SCFAs modulate poststroke recovery via effects on systemic and brain resident immune cells.SIGNIFICANCE STATEMENT Previous studies have shown a bidirectional communication along the gut-brain axis after stroke. Stroke alters the gut microbiota composition, and in turn, microbiota dysbiosis has a substantial impact on stroke outcome by modulating the immune response. However, until now, the mediators derived from the gut microbiome affecting the gut-immune-brain axis and the molecular mechanisms involved in this process were unknown. Here, we demonstrate that short-chain fatty acids, fermentation products of the gut microbiome, are potent and proregenerative modulators of poststroke neuronal plasticity at various structural levels. We identified that this effect was mediated via circulating lymphocytes on microglial activation. These results identify short-chain fatty acids as a missing link along the gut-brain axis and as a potential therapeutic to improve recovery after stroke.


Subject(s)
Brain/drug effects , Brain/immunology , Fatty Acids, Volatile/administration & dosage , Stroke/immunology , Animals , Brain/metabolism , Female , Lymphocytes/drug effects , Lymphocytes/immunology , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Recovery of Function/drug effects , Stroke/metabolism , Transcriptome/drug effects
10.
Animal ; 14(6): 1176-1183, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31840620

ABSTRACT

The combined addition of branched-chain volatile fatty acids (BCVFAs) and folic acid (FA) could improve growth performance and nutrient utilization by stimulating ruminal microbial growth and enzyme activity. This study was conducted to evaluate the effects of BCVFA and FA addition on growth performance, ruminal fermentation, nutrient digestibility, microbial enzyme activity, microflora and excretion of urinary purine derivatives (PDs) in calves. Thirty-six Chinese Holstein weaned calves (60 ± 5.4 days of age and 107 ± 4.7 kg of BW) were assigned to one of four groups in a randomized block design. Treatments were control (without additives), FA (with 10 mg FA/kg dietary DM), BCVFA (with 5 g BCVFA/kg dietary DM) and the combined addition of FA and BCVFA (10 mg/kg DM of FA and 5 g/kg DM of BCVFA). Supplements were hand-mixed into the top one-third of total mixed ration. Dietary concentrate to maize silage ratio was 50 : 50 on a DM basis. Dietary BCVFA or FA addition did not affect dry matter intake but increased average daily gain (ADG) and feed conversion efficiency. Ruminal pH and ammonia N were lower, and total volatile fatty acids (VFAs) concentration was higher for BCVFA or FA addition than for control. Dietary BCVFA or FA addition did not affect acetate proportion but decreased propionate proportion and increased acetate to propionate ratio. Total tract digestibility of DM, organic matter, CP and NDF was higher for BCVFA or FA addition than for control. Dietary BCVFA or FA addition increased activity of carboxymethyl cellulase and cellobiase, population of total bacteria, fungi, Ruminococcus albus, R. flavefaciens, Fibrobacter succinogenes and Prevotella ruminicola as well as total PD excretion. Ruminal xylanase, pectinase and protease activity and Butyrivibrio fibrisolvens population were increased by BCVFA addition, whereas population of protozoa and methanogens was increased by FA addition. The BCVFA × FA interaction was significant for acetate to propionate ratio, cellobiase activity and total PD excretion, and these variables increased more with FA addition in diet without BCVFA than in diet with BCVFA. The data indicated that supplementation with BCVFA or FA increased ADG, nutrient digestibility, ruminal total VFA concentration and microbial protein synthesis by stimulating ruminal microbial growth and enzyme activity in calves.


Subject(s)
Cattle/physiology , Cellulase/metabolism , Dietary Supplements/analysis , Fatty Acids, Volatile/administration & dosage , Folic Acid/administration & dosage , Gastrointestinal Microbiome/drug effects , Ammonia/metabolism , Animals , Cattle/growth & development , Cattle/microbiology , Diet/veterinary , Digestion/drug effects , Female , Fermentation , Nutrients/metabolism , Rumen/metabolism , Rumen/microbiology , Silage/analysis , Weaning
11.
Microbiome ; 7(1): 98, 2019 06 29.
Article in English | MEDLINE | ID: mdl-31255176

ABSTRACT

BACKGROUND: Western-style diets arouse neuroinflammation and impair emotional and cognitive behavior in humans and animals. Our previous study showed that a high-fructose diet caused the hippocampal neuroinflammatory response and neuronal loss in animals, but the underlying mechanisms remained elusive. Here, alterations in the gut microbiota and intestinal epithelial barrier were investigated as the causes of hippocampal neuroinflammation induced by high-fructose diet. RESULTS: A high-fructose diet caused the hippocampal neuroinflammatory response, reactive gliosis, and neuronal loss in C57BL/6N mice. Depletion of the gut microbiota using broad-spectrum antibiotics suppressed the hippocampal neuroinflammatory response in fructose-fed mice, but these animals still exhibited neuronal loss. Gut microbiota compositional alteration, short-chain fatty acids (SCFAs) reduction, intestinal epithelial barrier impairment, NOD-like receptor family pyrin domain-containing 6 (NLRP6) inflammasome dysfunction, high levels of serum endotoxin, and FITC-dextran were observed in fructose-fed mice. Of note, SCFAs, as well as pioglitazone (a selective peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist), shaped the gut microbiota and ameliorated intestinal epithelial barrier impairment and NLRP6 inflammasome dysfunction in fructose-fed mice. Moreover, SCFAs-mediated NLRP6 inflammasome activation was inhibited by histamine (a bacterial metabolite) in ex vivo colonic explants and suppressed in murine CT26 colon carcinoma cells transfected with NLRP6 siRNA. However, pioglitazone and GW9662 (a PPAR-γ antagonist) exerted no impact on SCFAs-mediated NLRP6 inflammasome activation in ex vivo colonic explants, suggesting that SCFAs may stimulate NLRP6 inflammasome independently of PPAR-γ activation. SCFAs and pioglitazone prevented fructose-induced hippocampal neuroinflammatory response and neuronal loss in mice. Additionally, SCFAs activated colonic NLRP6 inflammasome and increased DCX+ newborn neurons in the hippocampal DG of control mice. CONCLUSIONS: Our findings reveal that gut dysbiosis is a critical factor for a high-fructose diet-induced hippocampal neuroinflammation in C57BL/6N mice possibly mediated by impairing intestinal epithelial barrier. Mechanistically, the defective colonic NLRP6 inflammasome is responsible for intestinal epithelial barrier impairment. SCFAs can stimulate NLRP6 inflammasome and ameliorate the impairment of intestinal epithelial barrier, resulting in the protection against a high-fructose diet-induced hippocampal neuroinflammation and neuronal loss. This study addresses a gap in the understanding of neuronal injury associated with Western-style diets. A new intervention strategy for reducing the risk of neurodegenerative diseases through SCFAs supplementation or dietary fiber consumption is emphasized.


Subject(s)
Dysbiosis/chemically induced , Fatty Acids, Volatile/administration & dosage , Fructose/adverse effects , Hippocampus/drug effects , Inflammation/chemically induced , Animals , Doublecortin Protein , Gastrointestinal Microbiome , Hippocampus/pathology , Inflammasomes , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Neuroimmunomodulation/drug effects , Pioglitazone/administration & dosage
12.
FEMS Microbiol Lett ; 366(13)2019 07 01.
Article in English | MEDLINE | ID: mdl-31295342

ABSTRACT

Butyrate, a key metabolite fermented by gut microbiota mainly from undigested carbohydrates such as dietary fibers is widely used as feed additive. However, mechanisms of its contributions in maintaining host health are relatively poorly revealed. The aim of this study was to investigate how butyrate impacts gut microbiota and immunity response in high-fat diet-fed mice. Gut microbial analysis exhibited that butyrate intervention increased short-chain fatty acids (SCFAs)-producing bacteria and decreased pathogenic bacteria, such as endotoxin-secreting bacteria. Our result also demonstrated that butyrate intervention enhanced fecal SCFAs concentrations, and inhibited endotoxin levels in feces and serum. Correlation analysis indicated positive relation between endotoxin level and Desulfovibrionaceae abundance. Furthermore, butyrate intervention inhibited expressions of IL-1ß, IL-6 and MCP1/CCL2 in liver, as well as TLR4 in adipose tissue. Apart from inhibiting expressions of proinflammatory cytokines, butyrate exerted anti-inflammation effect through selectively modulating gut microbiota, such as increasing SCFAs-producing bacteria and decreasing endotoxin-secreting bacteria, as well as via regulating levels of microbiota-dependent metabolites and components, such as SCFAs and endotoxin.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Butyrates/administration & dosage , Diet, High-Fat , Dietary Supplements , Gastrointestinal Microbiome/drug effects , Animals , Computational Biology/methods , Disease Models, Animal , Endotoxins/blood , Fatty Acids, Volatile/administration & dosage , Fatty Acids, Volatile/metabolism , Feces/chemistry , Inflammation/drug therapy , Inflammation/etiology , Inflammation/pathology , Mice , Models, Biological , RNA, Ribosomal, 16S
13.
Hormones (Athens) ; 18(3): 245-250, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30840230

ABSTRACT

Study of the interactions between the gut microbiota and brain-gut axis represents a very appealing approach to increasing our knowledge about the mechanisms leading to obesity and obesity-related diseases. The aim of this review is to focus on the effects of short-chain fatty acids (SCFAs), which are the main products of gut microbial fermentation from non-digestible carbohydrates in the colon, on the gut-brain axis. Evidence is accumulating regarding the role of SCFAs in the fine-tuning of the gut-brain axis, a feedback system which is vital not only for the proper maintenance of gastrointestinal and metabolic functions, but also for the regulation of food intake and energy expenditure. SCFAs are thought to play a key role in increasing the host capacity to harvest excess energy from the diet. SCFAs, however, can exert their effects on the host metabolism via multiple complementary pathways. Metabolic, inflammatory, and neural pathways can be regulated by SCFAs, which can act by sensing nutritional status, thereby maintaining body energy homeostasis. SCFA production from prebiotic consumption is the rationale for targeting intestinal mechanisms to increase energy expenditure and thereby reduce obesity risk.


Subject(s)
Dysbiosis/complications , Dysbiosis/diet therapy , Fatty Acids, Volatile/administration & dosage , Gastrointestinal Microbiome/physiology , Obesity/etiology , Diet , Dietary Supplements , Dysbiosis/epidemiology , Energy Metabolism/physiology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Humans , Obesity/microbiology , Obesity/prevention & control , Prebiotics/administration & dosage
14.
Sci Rep ; 8(1): 1395, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29362450

ABSTRACT

Microbiota-derived short-chain fatty acids (SCFAs) and organic acids produced by the fermentation of non-digestible fibre can communicate from the microbiome to host tissues and modulate homeostasis in mammals. The microbiome has circadian rhythmicity and helps the host circadian clock function. We investigated the effect of SCFA or fibre-containing diets on circadian clock phase adjustment in mouse peripheral tissues (liver, kidney, and submandibular gland). Initially, caecal SCFA concentrations, particularly acetate and butyrate, induced significant day-night differences at high concentrations during the active period, which were correlated with lower caecal pH. By monitoring luciferase activity correlated with the clock gene Period2 in vivo, we found that oral administration of mixed SCFA (acetate, butyrate, and propionate) and an organic acid (lactate), or single administration of each SCFA or lactate for three days, caused phase changes in the peripheral clocks with stimulation timing dependency. However, this effect was not detected in cultured fibroblasts or cultured liver slices with SCFA applied to the culture medium, suggesting SCFA-induced indirect modulation of circadian clocks in vivo. Finally, cellobiose-containing diets facilitated SCFA production and refeeding-induced peripheral clock entrainment. SCFA oral gavage and prebiotic supplementation can facilitate peripheral clock adjustment, suggesting prebiotics as novel therapeutic candidates for misalignment.


Subject(s)
Bacteria/metabolism , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Fatty Acids, Volatile/administration & dosage , Gene Expression Regulation/drug effects , Animals , Circadian Clocks , Fatty Acids, Volatile/metabolism , Fatty Acids, Volatile/pharmacology , Fermentation , Gastrointestinal Microbiome , Kidney , Liver , Mice , Submandibular Gland
15.
Mucosal Immunol ; 11(3): 785-795, 2018 05.
Article in English | MEDLINE | ID: mdl-29067994

ABSTRACT

The mammalian gastrointestinal tract harbors a microbial community with metabolic activity critical for host health, including metabolites that can modulate effector functions of immune cells. Mice treated with vancomycin have an altered microbiome and metabolite profile, exhibit exacerbated T helper type 2 cell (Th2) responses, and are more susceptible to allergic lung inflammation. Here we show that dietary supplementation with short-chain fatty acids (SCFAs) ameliorates this enhanced asthma susceptibility by modulating the activity of T cells and dendritic cells (DCs). Dysbiotic mice treated with SCFAs have fewer interleukin-4 (IL4)-producing CD4+ T cells and decreased levels of circulating immunoglobulin E (IgE). In addition, DCs exposed to SCFAs activate T cells less robustly, are less motile in response to CCL19 in vitro, and exhibit a dampened ability to transport inhaled allergens to lung draining nodes. Our data thus demonstrate that gut dysbiosis can exacerbate allergic lung inflammation through both T cell- and DC-dependent mechanisms that are inhibited by SCFAs.


Subject(s)
Asthma/immunology , Dendritic Cells/immunology , Dysbiosis/immunology , Fatty Acids, Volatile/administration & dosage , Hypersensitivity/immunology , Pneumonia/immunology , Th2 Cells/immunology , Allergens/immunology , Animals , Antigen Presentation , Asthma/prevention & control , Chemokine CCL19/metabolism , Dietary Supplements , Dysbiosis/prevention & control , Gastrointestinal Microbiome/immunology , Hypersensitivity/prevention & control , Interleukin-4/genetics , Interleukin-4/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/immunology , Pneumonia/prevention & control , Vancomycin/administration & dosage
16.
Am J Clin Nutr ; 106(3): 930-945, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28793992

ABSTRACT

BACKGROUND: Prebiotic soluble fibers are fermented by beneficial bacteria in the colon to produce short-chain fatty acids (SCFAs), which are proposed to have systemic anti-inflammatory effects. OBJECTIVE: This review examines the effect of SCFAs, prebiotics, and pre- and probiotic combinations (synbiotics) on systemic inflammation. DESIGN: Relevant English language studies from 1947 to May 2017 were identified with the use of online databases. Studies were considered eligible if they examined the effects of SCFAs, prebiotics, or synbiotics; were delivered orally, intravenously, or per rectum; were on biomarkers of systemic inflammation in humans; and performed meta-analysis where possible. RESULTS: Sixty-eight studies were included. Fourteen of 29 prebiotic studies and 13 of 26 synbiotic studies reported a significant decrease in ≥1 marker of systemic inflammation. Eight studies compared prebiotic and synbiotic supplementation, 2 of which reported a decrease in inflammation with synbiotics only, with 1 reporting a greater anti-inflammatory effect with synbiotics than with prebiotics alone. Meta-analyses indicated that prebiotics reduce C-reactive protein (CRP) [standardized mean difference (SMD): -0.60; 95% CI: -0.98, -0.23], and synbiotics reduce CRP (SMD: -0.40; 95% CI: -0.73, -0.06) and tumor necrosis factor-α (SMD -0.90; 95% CI: -1.50, -0.30). CONCLUSIONS: There is significant heterogeneity of outcomes in studies examining the effect of prebiotics and synbiotics on systemic inflammation. Approximately 50% of included studies reported a decrease in ≥1 inflammatory biomarker. The inconsistency in reported outcomes may be due to heterogeneity in study design, supplement formulation, dosage, duration, and subject population. Nonetheless, meta-analyses provide evidence to support the systemic anti-inflammatory effects of prebiotic and synbiotic supplementation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Fatty Acids, Volatile/pharmacology , Inflammation/prevention & control , Prebiotics , Synbiotics , C-Reactive Protein/analysis , Dietary Supplements , Fatty Acids, Volatile/administration & dosage , Humans
17.
Lipids Health Dis ; 16(1): 96, 2017 May 22.
Article in English | MEDLINE | ID: mdl-28532421

ABSTRACT

BACKGROUND: Rodent models may guide investigations towards identifying either environmental neuro-toxicants or drugs with neuro-therapeutic effects. This work aims to study the therapeutic effects of bee pollen on brain glutamate excitotoxicity and the impaired glutamine-glutamate- gamma amino butyric acid (GABA) circuit induced by propionic acid (PPA), a short chain fatty acid, in rat pups. METHODS: Twenty-four young male Western Albino rats 3-4 weeks of age, and 45-60 g body weight were enrolled in the present study. They were grouped into four equal groups: Group 1, the control received phosphate buffered saline at the same time of PPA adminstration; Group 2, received 750 mg/kg body weight divided into 3 equal daily doses and served as acute neurotoxic dose of PPA; Group 3, received 750 mg/kg body weight divided in 10 equal doses of 75 mg/kg body weight/day, and served as the sub-acute group; and Group 4, the therapeutic group, was treated with bee pollen (50 mg/kg body weight) for 30 days after acute PPA intoxication. GABA, glutamate and glutamine were measured in the brain homogenates of the four groups. RESULTS: The results showed that PPA caused multiple signs of excitotoxicity, as measured by the elevation of glutamate and the glutamate/glutamine ratio and the decrease of GABA, glutamine and the GABA/glutamate ratio. Bee pollen was effective in counteracting the neurotoxic effects of PPA to a certain extent. CONCLUSION: In conclusion, bee pollen demonstrates ameliorating effects on glutamate excitotoxicity and the impaired glutamine-glutamate-GABA circuit as two etiological mechanisms in PPA-induced neurotoxicity.


Subject(s)
Fatty Acids, Volatile/toxicity , Glutamic Acid/metabolism , Pollen/chemistry , Propionates/toxicity , Animals , Bees , Fatty Acids, Volatile/administration & dosage , Male , Propionates/administration & dosage , Rats , gamma-Aminobutyric Acid/metabolism
18.
PLoS One ; 12(2): e0173032, 2017.
Article in English | MEDLINE | ID: mdl-28235016

ABSTRACT

Autoimmune diseases are influenced by both genetic and environmental factors. The gut environment has attracted much attention as an essential component that modulates immune responses, and therefore immune-mediated disorders, such as autoimmune diseases. Growing evidence suggests that microbiota and their metabolites are critical factors for immune modulation. Recently, we reported that the microbiome in patients with multiple sclerosis, an autoimmune disease targeting the myelin sheath of the central nervous system, is characterized by a reduction of bacteria belonging to Clostridia clusters IV and XIVa, which are potent producers of short-chain fatty acids (SCFAs) by fermentation of indigestible carbohydrates. In the present study, we investigated the role of SCFAs in the regulation of inflammation. We demonstrated that oral administration of SCFAs ameliorated the disease severity of systemic autoimmune inflammatory conditions mediated by lymphocytes such as experimental autoimmune encephalitis and collagen-induced arthritis. Amelioration of disease was associated with a reduction of Th1 cells and an increase in regulatory T cells. In contrast, SCFAs contributed to the exaggeration of K/BxN serum transfer arthritis, representing the effector phase of inflammation in rheumatoid arthritis. An increased understanding of the effect of microbiota metabolites will lead to the effective treatment and prevention of systemic inflammatory disorders.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Arthritis, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Fatty Acids, Volatile/administration & dosage , Administration, Oral , Animals , Arthritis, Experimental/immunology , Drug Evaluation, Preclinical , Encephalomyelitis, Autoimmune, Experimental/immunology , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells/drug effects , Th1 Cells/immunology
19.
Diabetes ; 64(7): 2398-408, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25695945

ABSTRACT

Short-chain fatty acids (SCFAs) are the main products of dietary fiber fermentation and are believed to drive the fiber-related prevention of the metabolic syndrome. Here we show that dietary SCFAs induce a peroxisome proliferator-activated receptor-γ (PPARγ)-dependent switch from lipid synthesis to utilization. Dietary SCFA supplementation prevented and reversed high-fat diet-induced metabolic abnormalities in mice by decreasing PPARγ expression and activity. This increased the expression of mitochondrial uncoupling protein 2 and raised the AMP-to-ATP ratio, thereby stimulating oxidative metabolism in liver and adipose tissue via AMPK. The SCFA-induced reduction in body weight and stimulation of insulin sensitivity were absent in mice with adipose-specific disruption of PPARγ. Similarly, SCFA-induced reduction of hepatic steatosis was absent in mice lacking hepatic PPARγ. These results demonstrate that adipose and hepatic PPARγ are critical mediators of the beneficial effects of SCFAs on the metabolic syndrome, with clearly distinct and complementary roles. Our findings indicate that SCFAs may be used therapeutically as cheap and selective PPARγ modulators.


Subject(s)
Adipose Tissue/metabolism , Fatty Acids, Volatile/administration & dosage , Lipogenesis , Obesity/prevention & control , PPAR gamma/physiology , AMP-Activated Protein Kinases/physiology , Adenosine Triphosphate/metabolism , Animals , Diet, High-Fat , Fatty Acids, Volatile/pharmacology , Insulin Resistance , Ion Channels/physiology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/physiology , Oxidation-Reduction , Uncoupling Protein 2
20.
J Dairy Sci ; 96(4): 2366-2373, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23415518

ABSTRACT

Milk fat synthesis might be promoted by the dietary addition of long-chain fatty acids (LCFA) or short- and medium-chain fatty acids (SMCFA). This study evaluated unprotected lipid supplementation with different ratios of SMCFA to LCFA, which had equivalent fatty acid (FA) proportions (by weight) to those in milk, on milk fat production and milk FA composition. Thirty-six Holstein cows (183±46 d in milk) were divided into 3 treatments according to a randomized block design. Cows in 3 treatments received supplements of 80 g/d of SMCFA mixture and 320 g/d of LCFA mixture (ratio of SMCFA to LCFA was 20:80); 400 g/d of butterfat (ratio of SMCFA to LCFA was 40:60); or 240 g/d of SMCFA mixture and 160 g/d of LCFA mixture (ratio of SMCFA to LCFA was 60:40). The FA compositions of the SMCFA mixture and the LCFA mixture were similar to the de novo synthesized FA (except C4:0) and preformed FA (except trans FA) found in the butterfat, respectively. Fatty acid supplements and butterfat were consumed by cows daily before the morning feeding during the 8-wk experimental period. Dry matter intake and milk yield were not different among the treatments. The milk fat percentage and total SMCFA concentration in milk fat tended to increase linearly and the proportion of milk total solids increased linearly with increasing ratios of SMCFA to LCFA in the supplements, whereas milk fat yield was not changed. We suggest that increasing ratios of SMCFA to LCFA in diets has the potential to improve milk fat synthesis.


Subject(s)
Cattle/metabolism , Fatty Acids/administration & dosage , Fatty Acids/analysis , Lipids/biosynthesis , Milk/chemistry , Animal Nutritional Physiological Phenomena , Animals , Diet/veterinary , Dietary Supplements , Fats/analysis , Fatty Acids/chemistry , Fatty Acids, Volatile/administration & dosage , Female , Lactation
SELECTION OF CITATIONS
SEARCH DETAIL