Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Nutr Food Res ; 65(15): e2000986, 2021 08.
Article in English | MEDLINE | ID: mdl-33974360

ABSTRACT

SCOPE: Studies based on DHA/EPA supplementation in animal models of inflammatory bowel disease (IBD) reveal controversial results. It is speculated that different forms of DHA may explain the controversial results. Therefore, the effects of DHA-enriched phospholipids (DHA-PL) and DHA-enriched triglyceride (DHA-TG) on IBD are compared. METHODS AND RESULTS: Male C57BL6/J mice are given DHA-PL and DHA-TG for 14 consecutive days, and receive ad libitum a 3.0% dextran sodium sulfate solution on the eighth day to establish IBD model. The results show that both DHA-PL and DHA-TG can reverse the colitis pathological process by decreasing the disease activity indexes (DAI), raising the colon length, suppressing the intestinal permeability, suppressing the oxidative stress, down-regulating pro-inflammatory factors, up-regulating anti-inflammatory factor in colon tissues. DHA-PL and DHA-TG also regulate the composition of gut microbiota via decreasing of the abundance Bacteroidetes and Firmicutes, and DHA-TG increases the abundance of Odoribacter. Importantly, DHA-PL and DHA-TG obviously attenuate the activation of microglia. CONCLUSIONS: DHA-PL shows outstanding advantages in regulating oxidative stress, inflammatory responses, and intestinal barrier permeability. The current research indicates that the existence of DHA affects the improvement, DHA in phospholipid form could be a more effective choice for nutritional intervention to prevent and treat colitis.


Subject(s)
Colitis/diet therapy , Encephalitis/diet therapy , Gastrointestinal Microbiome/drug effects , Oxidative Stress/drug effects , Phospholipids/pharmacology , Administration, Oral , Animals , Colitis/chemically induced , Colitis/microbiology , Colitis/pathology , Cytokines/metabolism , Dextran Sulfate/toxicity , Dietary Supplements , Docosahexaenoic Acids/pharmacology , Encephalitis/etiology , Fatty Acids/analysis , Fatty Acids/chemistry , Fatty Acids, Omega-3/chemistry , Fatty Acids, Omega-3/pharmacology , Gastrointestinal Microbiome/genetics , Male , Mice, Inbred C57BL , Phospholipids/administration & dosage
2.
Food Funct ; 12(9): 3831-3841, 2021 May 11.
Article in English | MEDLINE | ID: mdl-33977958

ABSTRACT

The aim of the present study was to determine the inhibitory effects and the potential underlying mechanisms of a novel Pleurotus eryngii ß-type glycosidic polysaccharide (WPEP) on colitis. To achieve this, sixty CD-1 (ICR) mice were divided into six groups including healthy and colitic mice treated with or without WPEP at two different doses (n = 10). The results showed that WPEP displayed a significant inhibitory effect on colitis as indicated by the lowered disease activity index in the treated colitic mice compared to the untreated colitic mice (2.78 ± 0.50 to 1.80 ± 0.17). A decrease in pro-inflammatory cytokine concentrations and pro-inflammatory protein expressions and an increase in the colon length (9.31 ± 0.59 cm to 10.89 ± 1.20 cm) along with histological improvements were also observed in the treated colitic mice compared to the untreated colitic mice in the present study. Flow cytometry and western blotting analysis revealed that these anti-colitis effects were associated with decreased accumulation of CD45+ immune cells, CD45 + F4/80+ macrophages and CD45 + Gr1+ neutrophils. Moreover, the 16s rRNA sequencing analysis of the gut microbiota revealed that WPEP partially reversed gut microbiota dysbiosis in the colitic mice including the decreased abundance of Akkermansia muciniphila (35.80 ± 9.10% to 18.24 ± 6.23%) and Clostridium cocleatum (2.34 ± 1.78% to 0.011 ± 0.003%) and the increased abundance of Bifidobacterium pseudolongum (3.48 ± 2.72% to 9.65 ± 3.74%), Lactobacillus reuteri (0.007 ± 0.002% to 0.21 ± 0.12%), Lactobacillus salivarius (1.23 ± 0.87% to 2.22 ± 1.53%) and Ruminococcus bromii (0.009 ± 0.001% to 3.83 ± 1.98%). In summary, our results demonstrated that WPEP could be utilized as a functional food component in colitis management as well as a potential prebiotic agent to improve inflammation-related disorders.


Subject(s)
Colitis/diet therapy , Colon , Dietary Supplements , Glycosides/administration & dosage , Pleurotus/chemistry , Animals , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Colon/immunology , Colon/metabolism , Colon/pathology , Cytokines/metabolism , Dextran Sulfate , Disease Models, Animal , Gastrointestinal Microbiome , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred ICR , Proteins/metabolism
3.
Mol Nutr Food Res ; 65(8): e2000922, 2021 04.
Article in English | MEDLINE | ID: mdl-33629501

ABSTRACT

SCOPE: The availability of studies related to the effects of natural macronutrients on inflammatory bowel disease (IBD) remain relatively limited. This study investigates whether and to what extent the consumption of five different native starches alleviate the clinical symptoms and dysbiosis of gut microbiota associated with colitis. METHODS AND RESULTS: Using dextran sodium sulfate (DSS)-induced mouse model of colitis, the potential effects of native potato starch (PS), pea starch (PEAS), corn starch (CS), Chinese yam starch (CYS), and red sorghum starch (RSS) on the clinical manifestations and dysbiosis of gut microbiota are studied. Compared to CS and RSS, the consumption of PEAS, PS, and CYS significantly diminishes clinical enteritis symptoms, including reduced disease activity index, and the alleviated degree of colonic histological damage. Furthermore, the analysis of gut microbiota reveals the significant prebiotic characteristics of PEAS, PS and CYS, as indicated by the maintenance of gut microbiota hemostasis and the inhibition of typically pathogenic bacteria, including Escherichia coli and Helicobacter hepaticus. CONCLUSION: Starches from potato, pea, and Chinese yam alleviate colitis symptoms in a mouse model, and also show significant prebiotic characteristics. These findings suggest a cost-effective and convenient dietary strategy for the management of IBD.


Subject(s)
Colitis/diet therapy , Gastrointestinal Microbiome/physiology , Prebiotics , Starch/pharmacology , Animals , Colitis/chemically induced , Colitis/microbiology , Colitis/pathology , Dextran Sulfate , Dioscorea/chemistry , Disease Models, Animal , Fatty Acids, Volatile/metabolism , Inflammatory Bowel Diseases/diet therapy , Male , Mice, Inbred C57BL , Pisum sativum/chemistry , Solanum tuberosum/chemistry , Sorghum/chemistry , Tumor Necrosis Factor-alpha/metabolism , Zea mays/chemistry
4.
Br J Nutr ; 125(1): 50-61, 2021 01 14.
Article in English | MEDLINE | ID: mdl-32792032

ABSTRACT

Dietary choline, which is converted to phosphatidylcholine (PC) in intestinal enterocytes, may benefit inflammatory bowel disease patients who typically have reduced intestinal choline and PC. The present study investigated the effect of dietary choline supplementation on colitis severity and intestinal mucosal homoeostasis using a Citrobacter rodentium-induced colitis model. C57BL/6J mice were fed three isoenergetic diets differing in choline level: choline-deficient (CD), choline-sufficient (CS) and choline-excess (CE) for 3 weeks prior to infection with C. rodentium. The effect of dietary choline levels on the gut microbiota was also characterised in the absence of infection using 16S rRNA gene amplicon sequencing. At 7 d following infection, the levels of C. rodentium in CD mice were significantly greater than that in CS or CE groups (P < 0·05). CD mice exhibited greater damage to the surface epithelium and goblet cell loss than the CS or CE mice, which was consistent with elevated pro-inflammatory cytokine and chemokine levels in the colon. In addition, CD group exhibited decreased concentrations of PC in the colon after C. rodentium infection, although the decrease was not observed in the absence of challenge. Select genera, including Allobaculum and Turicibacter, were enriched in response to dietary choline deficiency; however, there was minimal impact on the total bacterial abundance or the overall structure of the gut microbiota. Our results suggest that insufficient dietary choline intake aggravates the severity of colitis and demonstrates an essential role of choline in maintaining intestinal homoeostasis.


Subject(s)
Choline/pharmacology , Colitis/diet therapy , Diet/adverse effects , Dietary Supplements , Gastrointestinal Microbiome/drug effects , Animals , Chemokines/metabolism , Citrobacter rodentium , Colitis/etiology , Colitis/microbiology , Colon/metabolism , Cytokines/metabolism , Disease Models, Animal , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S/analysis , Severity of Illness Index
5.
NPJ Biofilms Microbiomes ; 6(1): 44, 2020 10 28.
Article in English | MEDLINE | ID: mdl-33116127

ABSTRACT

In this study, we show that calcium pectinate beads (CPB) allow the formation of 20 µm spherical microcolonies of the probiotic bacteria Lacticaseibacillus paracasei (formerly designated as Lactobacillus paracasei) ATCC334 with a high cell density, reaching more than 10 log (CFU/g). The bacteria within these microcolonies are well structured and adhere to a three-dimensional network made of calcium-pectinate through the synthesis of extracellular polymeric substances (EPS) and thus display a biofilm-like phenotype, an attractive property for their use as probiotics. During bacterial development in the CPB, a coalescence phenomenon arises between neighboring microcolonies accompanied by their peripheral spatialization within the bead. Moreover, the cells of L. paracasei ATCC334 encased in these pectinate beads exhibit increased resistance to acidic stress (pH 1.5), osmotic stress (4.5 M NaCl), the freeze-drying process and combined stresses, simulating the harsh conditions encountered in the gastrointestinal (GI) tract. In vivo, the oral administration of CPB-formulated L. paracasei ATCC334 in mice demonstrated that biofilm-like microcolonies are successfully released from the CPB matrix in the colonic environment. In addition, these CPB-formulated probiotic bacteria display the ability to reduce the severity of a DSS-induced colitis mouse model, with a decrease in colonic mucosal injuries, less inflammation, and reduced weight loss compared to DSS control mice. To conclude, this work paves the way for a new form of probiotic administration in the form of biofilm-like microcolonies with enhanced functionalities.


Subject(s)
Biofilms/growth & development , Colitis/diet therapy , Lacticaseibacillus paracasei/physiology , Pectins/chemistry , Probiotics/administration & dosage , Animals , Capsules , Colitis/chemically induced , Dextran Sulfate/adverse effects , Disease Models, Animal , Drug Compounding , Extracellular Polymeric Substance Matrix/metabolism , Freeze Drying , Male , Mice , Osmotic Pressure , Probiotics/pharmacology , Treatment Outcome
6.
Phytomedicine ; 77: 153212, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32711288

ABSTRACT

BACKGROUND: Peppermint oil and caraway oil are established remedies in gastroenterological medicine because of their spasmolytic/analgesic effects. PURPOSE: We investigated whether Menthacarin, a combination of both oils, exerted anti-inflammatory effects in a dextran sodium sulphate (DSS, 2%) murine model of colitis. STUDY DESIGN AND METHODS: C57BL/6 mice were orally administered Menthacarin in doses of 10, 30, 60, and 120 µg/g body weight (BW), and control mice received 0.2% agar, 10 µl/g BW, during 8 days of DSS-induced colitis. Colitis was monitored by BW measurements and colonoscopies. Colons of euthanised mice were excised for histological staining and ELISA measurements of the cytokines TNFα, IL-6, IL-10, IL-1ß, and TGF-ß. RESULTS: Menthacarin-treated mice compared to controls showed improved macroscopical and microscopical parameters and lower BW loss during the course of colitis. Menthacarin changed the colonic cytokine profile towards a regulatory/anti-inflammatory phenotype. CONCLUSION: Menthacarin attenuates experimental colitis and may be a promising add-on therapy for the treatment of IBD.


Subject(s)
Colitis/diet therapy , Colon/drug effects , Plant Oils/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Body Weight/drug effects , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Cytokines/metabolism , Dextran Sulfate/toxicity , Male , Mentha piperita , Mice, Inbred C57BL
7.
Molecules ; 25(10)2020 May 14.
Article in English | MEDLINE | ID: mdl-32422882

ABSTRACT

Vitamin D is widely known to regulate bone health, but there is increasing evidence that it may also ameliorate colitis through inflammation, cell proliferation and apoptosis, and the microbiota. The purpose of this review is to systematically examine the mechanisms by which vitamin D reduces colitis. PubMed and Web of Science were searched for articles published between 2008 and 2019 using key words such as "vitamin D," "colitis," "inflammatory bowel disease," "inflammation," "apoptosis," "cell proliferation," and "gut bacteria". Retrieved articles were further narrowed and it was determined whether their title and abstracts contained terminology pertaining to vitamin D in relation to colitis in human clinical trials, animal studies, and cell culture/biopsy studies, as well as selecting the best match sorting option in relation to the research question. In total, 30 studies met the established criteria. Studies consistently reported results showing that vitamin D supplementation can downregulate inflammatory pathways of COX-2, TNF-α, NF-κB, and MAPK, modify cell kinetics, and alter gut microbiome, all of which contribute to an improved state of colitis. Although vitamin D and vitamin D analogs have demonstrated positive effects against colitis, more randomized, controlled human clinical trials are needed to determine the value of vitamin D as a therapeutic agent in the treatment of colitis.


Subject(s)
Colitis/diet therapy , Dietary Supplements , Gene Expression Regulation/drug effects , Inflammatory Bowel Diseases/diet therapy , Vitamin D/administration & dosage , Animals , Clinical Trials as Topic , Colitis/genetics , Colitis/microbiology , Colitis/pathology , Colon/drug effects , Colon/microbiology , Colon/pathology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Epithelial Cells/pathology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Humans , Inflammation , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Mice , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
Food Funct ; 11(4): 3459-3470, 2020 Apr 30.
Article in English | MEDLINE | ID: mdl-32239008

ABSTRACT

Tea consumption has been found to be associated with low incidence of inflammatory bowel disease in Asian countries. However, there is very limited knowledge of such potential protection and its underlying mechanism. Ripened Pu-erh tea (RPT) belongs to the variety of microbial fermented tea, but its function regarding anti-inflammation remains unclear. In the present study, we investigated the effects of RPT on dextran sulfate sodium (DSS)-induced colitis in mice. The results demonstrated that RPT significantly relieved the loss of body weight, disease severity and shortening of colon length, and remarkably inhibited the secretion of pro-inflammatory cytokines by lessening the infiltration of inflammatory cells. Furthermore, we found that RPT suppressed the activation of the NF-κB pathway and down-regulated the expression of HIF-1α. Thus, it was concluded that RPT attenuated the progress of colitis via suppressing the HIF-1α/NF-κB signaling pathways thus reducing inflammation. This suggests that RPT may be a potential anti-inflammatory nutraceutical for the prevention and treatment of colonic colitis.


Subject(s)
Colitis/diet therapy , Plant Extracts , Tea , Animals , Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate , Disease Models, Animal , Down-Regulation , Female , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Signal Transduction
9.
Mol Nutr Food Res ; 64(6): e1900873, 2020 03.
Article in English | MEDLINE | ID: mdl-31945799

ABSTRACT

SCOPE: Marine-derived n-3 PUFAs may ameliorate inflammation associated with inflammatory bowel diseases. Plant-derived n-3 PUFAs are thought to be inferior owing to shorter chain lengths. The aim of this study is to compare the impact of plant- and fish-derived PUFAs on murine colitis. METHODS AND RESULTS: C57BL/6 mice are fed high fat (36% kcal) diets with either 2.5% w/w sunflower oil (SO), flaxseed oil (FSO), ahiflower oil (AO), or fish oil (FO). After 4 weeks, mice are orogastrically challenged with Citrobacter rodentium (108 CFU) or sham gavaged. Fecal shedding is assayed at 2, 7, 10, and 14 days post infection (PI), and fecal microbiota at 14 days PI. Colonic inflammation and lipid mediators are measured. Supplementation regulates intestinal inflammation with crypt lengths being 66, 73, and 62 ±17 µm shorter (compared to SO) for FSO, AO, and FO respectively, p < 0.01. FSO blunts pathogen shedding at the peak of infection and FSO and AO both enhance fecal microbial diversity. FO attenuates levels of lipoxin and leukotriene B4 while plant oils increase pro-resolving mediator concentrations including D, E, and T-series resolvins. CONCLUSION: Plant and fish n-3 PUFAs attenuate colitis-induced inflammation while exhibiting characteristic pro-resolving lipid mediator metabolomes. Plant oils additionally promote microbial diversity.


Subject(s)
Citrobacter rodentium/pathogenicity , Colitis/diet therapy , Fatty Acids, Omega-3/pharmacology , Fish Oils/pharmacology , Plant Oils/pharmacology , Animals , Bacterial Shedding/drug effects , Colitis/microbiology , Colitis/pathology , Colon/drug effects , Colon/metabolism , Dietary Supplements , Enterobacteriaceae Infections/diet therapy , Inflammation Mediators/metabolism , Linseed Oil/chemistry , Linseed Oil/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Sunflower Oil/pharmacology
10.
Sci Rep ; 9(1): 14661, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31601878

ABSTRACT

Extracellular vesicles (EVs) are involved in cell-to-cell communication and modulation of numerous physiological and pathological processes. EVs are found in large quantities in milk and contain several inflammation- and immunity-modulating proteins and microRNAs, through which they exert beneficial effects in several inflammatory disease models. Here, we investigated the effects of two EV subsets, concentrated from commercial cow's milk, on a murine model of colitis induced with dextran sodium sulfate (DSS). P35K EVs, isolated by ultracentrifugation at 35,000 g, and P100K EVs, isolated at 100,000 g, were previously characterized and administered by gavage to healthy and DSS-treated mice. P35K EVs and, to a lesser extent, P100K EVs improved several outcomes associated to DSS-induced colitis, modulated the gut microbiota, restored intestinal impermeability and replenished mucin secretion. Also, P35K EVs modulated innate immunity, while P100K EVs decreased inflammation through the downregulation of colitis-associated microRNAs, especially miR-125b, associated with a higher expression of the NFκB inhibitor TNFAIP3 (A20). These results suggest that different milk EV subsets may improve colitis outcomes through different, and possibly complementary, mechanisms. Further unveiling of these mechanisms might offer new opportunities for improving the life of patients with colitis and be of importance for milk processing, infant milk formulation and general public health.


Subject(s)
Colitis/diet therapy , Dietary Supplements , Extracellular Vesicles/immunology , Intestinal Mucosa/immunology , Milk/cytology , Animals , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Gastrointestinal Microbiome/immunology , Humans , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Milk/immunology , Mucins/metabolism , Ultracentrifugation
11.
Molecules ; 24(19)2019 Oct 05.
Article in English | MEDLINE | ID: mdl-31590413

ABSTRACT

BACKGROUND: Inflammatory bowel diseases are an important health problem. Therefore, the aim of the present study was to compare the impact of isolated oat beta-glucan fractions of low and high molecular weight, taken as dietary supplementation, on inflammatory markers in the colitis model. METHODS: Two groups of Sprague-Dawley rats-control and with experimentally induced colitis-were subsequently divided into three subgroups and fed over 21 days feed supplemented with 1% of low (ßGl) or high (ßGh) molecular weight oat beta-glucan fraction or feed without supplementation. The level of colon inflammatory markers, cytokines, and their receptors' genes expressions and immune cells numbers were measured by ELISA, RT-PCR, and by flow cytometry methods, respectively. RESULTS: The results showed moderate inflammation affecting the colon mucosa and submucosa, with significant changes in the number of lymphocytes in the colon tissue, elevated cytokines and eicosanoid levels, as well as disruption of the main cytokine and chemokine cell signaling pathways in colitis rats. Beta-glucans supplementation caused a reverse in the percentage of lymphocytes with stronger effects of ßGh and reduction of the levels of the inflammatory markers, and improvement of cytokine and chemokine signaling pathways with stronger effects of ßGl supplementation. CONCLUSIONS: The results indicate the therapeutic effect of dietary oat beta-glucan supplementation in the colitis in evident relation to the molecular weight of polymer.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Avena/chemistry , Colitis/diet therapy , Trinitrobenzenesulfonic Acid/adverse effects , beta-Glucans/administration & dosage , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Cytokines/genetics , Cytokines/metabolism , Dietary Supplements , Disease Models, Animal , Gene Expression Regulation/drug effects , Lymphocyte Count , Male , Molecular Weight , Rats , Rats, Sprague-Dawley , beta-Glucans/chemistry , beta-Glucans/pharmacology
12.
Food Funct ; 10(10): 6331-6341, 2019 Oct 16.
Article in English | MEDLINE | ID: mdl-31524900

ABSTRACT

Increased consumption of fruits may decrease the risk of chronic inflammatory diseases including inflammatory bowel disease (IBD). Gut microbiota dysbiosis plays an important etiological role in IBD. However, the mechanisms of action underlying the anti-inflammatory effects of dietary cranberry (Vaccinium macrocarpon) in the colon and its role on gut microbiota were unclear. In this study, we determined the anti-inflammatory efficacy of whole cranberry in a mouse model of dextran sodium sulfate (DSS)-induced colitis, as well as its effects on the structure of gut microbiota. The results showed that dietary cranberry significantly decreased the severity of colitis in DSS-treated mice, evidenced by increased colon length, and decreased disease activity and histologic score of colitis in DSS-treated mice compared to the positive control group (p < 0.05). Moreover, the colonic levels of pro-inflammatory cytokine (IL-1ß, IL-6 and TNF-α) were significantly reduced by cranberry supplementation (p < 0.05). Analysis of the relative abundance of fecal microbiota in phylum and genus levels revealed that DSS treatment significantly altered the microbial structure of fecal microbiota in mice. α diversity was significantly decreased in the DSS group, compared to the healthy control group. But, cranberry treatment significantly improved DSS-induced decline in α-diversity. Moreover, cranberry treatment partially reversed the change of gut microbiota in colitic mice by increasing the abundance of potential beneficial bacteria, for example, Lactobacillus and Bifidobacterium, and decreasing the abundance of potential harmful bacteria, such as Sutterella and Bilophila. Overall, our results for the first time demonstrated that modification of gut microbiota by dietary whole cranberry might contribute to its inhibitory effects against the development of colitis in DSS-treated mice.


Subject(s)
Colitis/diet therapy , Dysbiosis/diet therapy , Gastrointestinal Microbiome/drug effects , Vaccinium macrocarpon/metabolism , Animals , Colitis/immunology , Colitis/microbiology , Colon/immunology , Colon/microbiology , Dextran Sulfate/adverse effects , Dysbiosis/chemically induced , Dysbiosis/genetics , Dysbiosis/immunology , Fruit/chemistry , Fruit/metabolism , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Male , Mice , Sulfates/adverse effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vaccinium macrocarpon/chemistry
13.
Gut Microbes ; 10(6): 654-662, 2019.
Article in English | MEDLINE | ID: mdl-31062653

ABSTRACT

Vitamin B12 is a critical nutrient for humans as well as microbes. Due to saturable uptake, high dose oral B12 supplements are largely unabsorbed and reach the distal gut where they are available to interact with the microbiota. The aim of this study was to determine if oral B12 supplementation in mice alters 1) the concentration of B12 and related corrinoids in the distal gut, 2) the fecal microbiome, 3) short chain fatty acids (SCFA), and 4) susceptibility to experimental colitis. C57BL/6 mice (up to 24 animals/group) were supplemented with oral 3.94 µg/ml cyanocobalamin (B12), a dose selected to approximate a single 5 mg supplement for a human. Active vitamin B12 (cobalamin), and four B12-analogues ([ADE]CN-Cba, [2Me-ADE]CN-Cba, [2MeS-ADE]CN-Cba, CN-Cbi) were analyzed in cecal and fecal contents using liquid chromatography/mass spectrometry (LC/MS), in parallel with evaluation of fecal microbiota, cecal SCFA, and susceptibility to dextran sodium sulfate (DSS) colitis. At baseline, active B12 was a minor constituent of overall cecal (0.86%) and fecal (0.44%) corrinoid. Oral B12 supplementation increased active B12 at distal sites by >130-fold (cecal B12 increased from 0.08 to 10.60 ng/mg, fecal B12 increased from 0.06 to 7.81 ng/ml) and reduced microbe-derived fecal corrinoid analogues ([ADE]CN-Cba, [2Me-ADE]CN-Cba, [2MeS-ADE]CN-Cba). Oral B12 had no effect on cecal SCFA. Microbial diversity was unaffected by this intervention, however a selective decrease in Bacteroides was observed with B12 treatment. Lastly, no difference in markers of DSS-induced colitis were detected with B12 treatment.


Subject(s)
Bacteroides/drug effects , Corrinoids/analysis , Dietary Supplements/analysis , Vitamin B 12/administration & dosage , Vitamin B Complex/administration & dosage , Administration, Oral , Animals , Bacteroides/growth & development , Cecum/chemistry , Colitis/chemically induced , Colitis/diet therapy , Dextran Sulfate/toxicity , Fatty Acids, Volatile/analysis , Feces/chemistry , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Mice, Inbred C57BL , Vitamin B 12/pharmacology , Vitamin B Complex/pharmacology
14.
Nutr Cancer ; 71(2): 199-206, 2019.
Article in English | MEDLINE | ID: mdl-30862196

ABSTRACT

Ulcerative colitis is an unremitting and lifelong inflammatory bowel disease that is increasing in prevalence worldwide. Patients display various clinical symptoms such as abdominal pain, diarrhea and fatigue. The etiology of ulcerative colitis remains unknown and the current pharmaceutical treatments are variably effective and not curative, highlighting the need for improved therapeutic approaches. Furthermore, patients with ulcerative colitis are at an increased risk of developing colorectal cancer. Some naturally sourced agents, named nutraceuticals, have been identified to possess anti-inflammatory and antioxidant properties. Of particular interest is Emu Oil, grape seed extract and Japanese Kampo medicine. Previously, Emu Oil has protected and repaired intestinal damage in models of gastrointestinal diseases including colitis and colitis-associated colorectal cancer. Additionally, grape seed extract possesses anticancer properties in vitro. Moreover, Kampo medicine, composed of herbal ingredients, is widely used in Japan for the treatment of various medical conditions and has demonstrated efficacy in targeting cancer cells in vitro. Nutraceuticals in combination have not yet been widely investigated in a setting of colitis-associated colorectal cancer. Investigation into the efficacy of Emu Oil combined with other nutraceuticals, including grape seed extract and Kampo medicine, is warranted as they may provide a novel approach to conventional colitis and colorectal cancer management.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colitis/complications , Colitis/diet therapy , Colorectal Neoplasms/diet therapy , Dietary Supplements , Grape Seed Extract/therapeutic use , Medicine, Kampo/methods , Oils/therapeutic use , Antioxidants/therapeutic use , Colorectal Neoplasms/etiology , Humans
15.
Front Immunol ; 10: 2979, 2019.
Article in English | MEDLINE | ID: mdl-31921214

ABSTRACT

Dietary fiber, with intake of soluble fibers in particular, has been reported to lower the risk for developing inflammatory bowel diseases (IBD). This is at least partly attributable to the fermentation of dietary fiber by the colonic microbiota to produce short chain fatty acids. Pectin, a widely consumed soluble fiber, is known to exert a protective effect in murine models of IBD, but the underlying mechanism remains elusive. Apart from having a prebiotic effect, it has been suggested that pectin direct influences host cells by modulating the inflammatory response in a manner dependent on its neutral sugar side chains. Here we examined the effect of the side chain content of pectin on the pathogenesis of experimental colitis in mice. Male C57BL/6 mice were fed a pectin-free diet, or a diet supplemented with characteristically high (5% orange pectin) or low (5% citrus pectin) side chain content for 10-14 days, and then administered 2,4,6-trinitrobenzene sulfonic acid or dextran sulfate sodium to induce colitis. We found that the clinical symptoms and tissue damage in the colon were ameliorated in mice that were pre-fed with orange pectin, but not in those pre-fed with citrus pectin. Although the population of CD4+Foxp+ regulatory T cells and CD4+RORγt+ inflammatory T cells in the colon were comparable between citrus and orange pectin-fed mice, colonic interleukin (IL)-1ß and IL-6 levels in orange pectin-fed mice were significantly decreased. The fecal concentration of propionic acid in orange pectin-fed mice was slightly but significantly higher than that in control and citrus pectin-fed mice but the cecal concentration of propionic acid after the induction of TNBS colitis was comparable between orange and citrus pectin-fed mice. Furthermore, the protective effect of orange pectin against colitis was observed even in mice treated with antibiotics. IL-6 production from RAW264.7 cells stimulated with the toll-like receptor agonist Pam3CSK4 or lipopolysaccharide was suppressed by pre-treatment with orange pectin in vitro. Taken together, these results suggest that the side chains of pectin not only augment prebiotic effects but also directly regulate IL-6 production from intestinal host cells in a microbiota-independent fashion to attenuate colitis.


Subject(s)
Colitis/diet therapy , Colitis/metabolism , Dietary Fiber , Pectins/administration & dosage , Sugars/metabolism , Animals , Colitis/etiology , Colitis/pathology , Cytokines/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Toll-Like Receptors/metabolism
16.
PLoS One ; 13(10): e0202460, 2018.
Article in English | MEDLINE | ID: mdl-30308045

ABSTRACT

Inflammatory bowel disease (IBD) is associated with anaemia and oral iron replacement to correct this can be problematic, intensifying inflammation and tissue damage. The intestinal microbiota also plays a key role in the pathogenesis of IBD, and iron supplementation likely influences gut bacterial diversity in patients with IBD. Here, we assessed the impact of dietary iron, using chow diets containing either 100, 200 or 400 ppm, fed ad libitum to adult female C57BL/6 mice in the presence or absence of colitis induced using dextran sulfate sodium (DSS), on (i) clinical and histological severity of acute DSS-induced colitis, and (ii) faecal microbial diversity, as assessed by sequencing the V4 region of 16S rRNA. Increasing or decreasing dietary iron concentration from the standard 200 ppm exacerbated both clinical and histological severity of DSS-induced colitis. DSS-treated mice provided only half the standard levels of iron ad libitum (i.e. chow containing 100 ppm iron) lost more body weight than those receiving double the amount of standard iron (i.e. 400 ppm); p<0.01. Faecal calprotectin levels were significantly increased in the presence of colitis in those consuming 100 ppm iron at day 8 (5.94-fold) versus day-10 group (4.14-fold) (p<0.05), and for the 400 ppm day-8 group (8.17-fold) versus day-10 group (4.44-fold) (p<0.001). In the presence of colitis, dietary iron at 400 ppm resulted in a significant reduction in faecal abundance of Firmicutes and Bacteroidetes, and increase of Proteobacteria, changes which were not observed with lower dietary intake of iron at 100 ppm. Overall, altering dietary iron intake exacerbated DSS-induced colitis; increasing the iron content of the diet also led to changes in intestinal bacteria diversity and composition after colitis was induced with DSS.


Subject(s)
Anemia/drug therapy , Colitis/diet therapy , Inflammatory Bowel Diseases/drug therapy , Iron, Dietary/administration & dosage , Iron/metabolism , Administration, Oral , Anemia/microbiology , Anemia/pathology , Animals , Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Humans , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Mice , RNA, Ribosomal, 16S/genetics
17.
J Nutr Biochem ; 62: 50-58, 2018 12.
Article in English | MEDLINE | ID: mdl-30245183

ABSTRACT

We used water-soluble Chitosan obtained by Maillard reaction with glucosamine to microencapsulate soy genistein (Ge) and preserve its biological activity for oral administration. Release of Ge was pH dependent with a super Case II mechanism at pH 1.2 and an anomalous transport with non-Fickian kinetics at pH 6.8. Microencapsulated Ge retained its antioxidant properties in vitro and its daily administration to mice attenuated clinical signs of acute colitis, limited inflammatory reaction and reduced oxidative stress and tissue injury as well. Remarkably, after feeding microencapsulated Ge the production of IL-10 in colonic tissue was restored to levels of untreated controls. According to statistical multivariate analysis, this cytokine was the parameter with the highest influence on the inflammatory/oxidative status. Microencapsulation of Ge with derivatized Chitosan becomes an interesting alternative to develop therapeutic approaches for oxidative inflammatory diseases; our findings suggest that the soy isoflavone could be incorporated into any functional food for application in intestinal inflammation.


Subject(s)
Antioxidants/administration & dosage , Colitis/diet therapy , Genistein/administration & dosage , Administration, Oral , Animals , Antioxidants/chemistry , Antioxidants/pharmacology , Chitosan/chemistry , Colitis/chemically induced , Colitis/metabolism , Cytokines/metabolism , Dietary Supplements , Disease Models, Animal , Drug Compounding/methods , Female , Genistein/chemistry , Genistein/pharmacology , Interleukin-10/metabolism , Mice, Inbred C57BL , Oxidative Stress/drug effects , Solubility , Glycine max/chemistry
18.
J Med Food ; 21(10): 1070-1074, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30183449

ABSTRACT

We investigated the preventive effects of Ramyeon made from brown rice-sorghum noodles and doenjang-bamboo salt soup (BS+DB) on colitis induced by dextran sulfate sodium (DSS) in C57BL/6 mice. Noodles were prepared with 10% brown rice and 10% sorghum powders added to wheat flour and potato powder, and soup was made using starter fermented (SF) doenjang powder (32%) and bamboo salt (13.5%). The experimental animals were divided into five groups: Normal, Control, BS (brown rice and sorghum noodles)+DB (doenjang and bamboo salt soup) (BS+DB Ramyeon), W (white flour noodles, commercial one)+DB, W+dC (W+doenjang commercial soup), and W + D (W+SF doenjang powder [100%]). The BS+DB and W + D groups showed significant reduction of DSS-induced colitis symptoms (P < .05). Doenjang soup (100%) (W+D) also showed a strong anticolitic effect even though the noodles were prepared with W. Histological observation of the colon revealed that BS+DB Ramyeon markedly alleviated colitis development in mice. Serum protein and mRNA levels of proinflammatory cytokines (TNF-α, IL-1ß, and IL-6) were significantly suppressed in colon tissue of the BS+DB group compared with those of the W+DB and W+dC groups. BS+DB Ramyeon also reduced colon mRNA levels of inducible nitric oxide synthase and cyclooxygenase-2 compared with those of other groups (P < .05). Our results show that modification of noodle ingredients using brown rice and sorghum as well as alteration of soup composition using doenjang and bamboo salt improved the health benefits of Ramyeon.


Subject(s)
Colitis/diet therapy , Functional Food/analysis , Animals , Colitis/genetics , Colitis/immunology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Flour/analysis , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Oryza/chemistry , Oryza/metabolism , Sodium Chloride, Dietary/analysis , Sodium Chloride, Dietary/metabolism , Solanum tuberosum/chemistry , Solanum tuberosum/metabolism , Sorghum/chemistry , Sorghum/metabolism , Triticum/chemistry , Triticum/metabolism
20.
J Nutr ; 148(5): 667-674, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29897487

ABSTRACT

BACKGROUND: Ulcerative colitis causes recurring intestinal mucosal injury and sustained inflammation, increasing the likelihood of colorectal cancer (CRC) development. Dietary red raspberry (RB) is a rich source of phytonutrients known to have anti-inflammatory activity; however, the role of RB on CRC prevention in chronic colitis has not been examined. OBJECTIVE: This study examined the effects of dietary RB supplementation on inflammation, epithelium repair, and oncogenic signaling in dextran sulfate sodium (DSS)-induced chronic colitis in mice. METHODS: Six-week-old male C57BL/6J mice were fed a control or RB (5% of dry feed weight; n = 12/group) diet for 10 wk. Starting from the fourth week, mice were administered 2 repeated cycles of 1% DSS (7-d DSS treatment plus 14-d recovery) and were monitored daily for disease activity index (DAI) score. Colonic tissues were collected at the end of the study for histochemical, immunohistochemical, and biochemical analysis of inflammation, differentiation and proliferation markers. RESULTS: RB supplementation reduced the DAI score and histologic damage (by 38.9%; P ≤ 0.01), expression of inflammatory mediators (by 20-70%; P ≤ 0.01), infiltration of CD4 T cells (by 50%; P ≤ 0.05), and α4ß7 integrin and related adhesion molecules (by 33.3%; P ≤ 0.01). Furthermore, RB supplementation facilitated epithelium repair, as evidenced by enhanced goblet cell density, expression of transcription factors including Kruppel-like factor 4 (Klf4) and Hairy and enhancer of split 1 (Hes1), terminal differentiation markers, mucin 2 (Muc2), and intestinal alkaline phosphatase (by 20-200%; P ≤ 0.01). Conversely, proliferating cell nuclear antigen (by 70%; P ≤ 0.01), ß-catenin, and signal transducer and activator of transcription 3 (STAT3) signaling (by 19-33%; P ≤ 0.05) were reduced by RB supplementation. In addition, RB supplementation enhanced p53 stability (by 53%) and reduced oncogenic gene expression (by 50-60%). CONCLUSION: RB supplementation reduced DAI score and the risk of CRC development during recurring colitis in mice, suggesting that RB is a possible dietary supplement for patients with ulcerative colitis and related gut inflammatory diseases.


Subject(s)
Colitis/diet therapy , Colorectal Neoplasms/prevention & control , Dietary Supplements , Rubus , Animals , Colitis/chemically induced , Colitis/complications , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/etiology , Dextran Sulfate/toxicity , Disease Models, Animal , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Kruppel-Like Factor 4 , Male , Mice , Mice, Inbred C57BL , Risk Factors , STAT3 Transcription Factor/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL