Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nutrients ; 16(4)2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38398817

ABSTRACT

Obesity and its complications constitute a main threat to global human health. The purpose of this investigation was to explore the influences of Clostridium tyrobutyricum (Ct) on lipid metabolism, intestinal barrier function, and intestinal microbiome in obese mice induced by a high-fat diet (HFD). After establishing the obesity model, 107 CFU/mL and 108 CFU/mL C. tyrobutyricum were used to intervene in HFD-fed mice by gavage for six weeks, and indexes related to obesity were measured. In the liver of HFD-fed mice, the results revealed that C. tyrobutyricum reduced liver weight and the levels of triglyceride (TG), total cholesterol (TC), and nonesterified fatty acid (NEFA), along with decreasing red lipid droplets and fat vacuoles. After C. tyrobutyricum intervention, the mRNA expression of peroxisome proliferator-activated receptor-γ (PPARγ) was downregulated, and AMP-activated protein kinase (AMPK), peroxisome proliferator-activated receptor-α (PPARα), adipose triglyceride lipase (ATGL), and hormone-sensitive lipase (HSL) were upregulated in the liver. Additionally, C. tyrobutyricum alleviated intestinal morphology injury caused by HFD, decreased the expression of tumor necrosis factor-α (TNF-α), interleukin 6 (IL-6), and IL-1ß in the colon, and upregulated tight junction protein expression. In addition, 16S rRNA sequencing revealed that C. tyrobutyricum increases the diversity of intestinal microbiota. Overall, C. tyrobutyricum improved HFD-induced lipid metabolism disorders, preserved the intestinal barrier's integrity, and modulated the structure of the intestinal microbiome. These findings provide a novel insight into the role of C. tyrobutyricum as a probiotic in regulating lipid metabolism.


Subject(s)
Clostridium tyrobutyricum , Gastrointestinal Microbiome , Humans , Animals , Mice , Diet, High-Fat/adverse effects , Lipid Metabolism , Gastrointestinal Microbiome/physiology , Mice, Obese , Intestinal Barrier Function , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Liver/metabolism , Obesity/metabolism , Mice, Inbred C57BL
2.
Food Res Int ; 155: 111003, 2022 05.
Article in English | MEDLINE | ID: mdl-35400418

ABSTRACT

Present study was conducted to comprehensively investigate the protective effects of galactooligosaccharides (GOS, 100%) against LPS-induced intestinal barrier damages, and the regulatory effect for intestinal microbes. Results showed that GOS intervention restored villi (jejunum and ileum) integrity, which were atrophic and broken in LPS-challenged mice. Electron microscopy, western blotting and immunofluorescence analysis exhibited that mice administrated with GOS showed higher expression of tight junction, which was confirmed in IPEC-J2 cells model. Meanwhile, the GOS increased the secretion of mucin and SIgA, as well as it alleviated inflammatory response caused by LPS in NF-κB dependent way. Administration of GOS could also increase the relative abundances of several specific friendly bacteria, and enhance the adaptability of intestinal microbiota. Collectively, these results indicated the potential of GOS for protecting intestine from injuries caused by stress as LPS challenge.


Subject(s)
Gastrointestinal Microbiome , Animals , Cell Line , Intestines , Lipopolysaccharides , Mice , Protective Agents
3.
Front Immunol ; 12: 758227, 2021.
Article in English | MEDLINE | ID: mdl-34917080

ABSTRACT

Probiotics are clinically used for diarrhea and inflammatory bowel diseases in both humans and animals. Previous studies have shown that Clostridium tyrobutyricum (Ct) protects against intestinal dysfunction, while its regulatory function in the gut needs further investigation and the related mechanisms are still not fully elucidated. This study aims to further verify the protective function of Ct and reveal its underlying mechanisms in alleviating diarrhea and intestinal inflammation. Ct inhibited LPS-induced diarrhea and intestinal inflammation in the ileum. IL-22 was identified and the protective role of Ct in the ileum presented an IL-22-dependent manner according to the transcriptomic analysis and in vivo interference mice experiments. The flow cytometric analysis of immune cells in the ileum showed that Ct enhanced the proportions of Th17 cells in response to LPS. The results of in situ hybridization further verified that Ct triggered Th17 cells to produce IL-22, which combined with IL-22RA1 expressed in the epithelial cells. Moreover, Ct was unable to enhance the levels of short-chain fatty acids (SCFAs) in the ileum, suggesting that the protective role of Ct in the ileum was independent of SCFAs. This study uncovered the role of Ct in alleviating diarrhea and inflammation with the mechanism of stimulating Th17 cells in the lamina propria to produce IL-22, highlighting its potential application as a probiotic for diarrhea and inflammation in the ileum.


Subject(s)
Clostridium tyrobutyricum/physiology , Diarrhea/prevention & control , Ileum/immunology , Probiotics , Th17 Cells/metabolism , Animals , Bacterial Translocation , Epithelial Cells/physiology , Ileum/metabolism , Interleukins/biosynthesis , Interleukins/genetics , Intestinal Mucosa/pathology , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Models, Immunological , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Interleukin-22
4.
Food Chem ; 362: 130195, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34082294

ABSTRACT

The aim of this study was to optimize and compare the production of galactooligosaccharides (GOSs) by free and cotton cloth-immobilized Aspergillus oryzae ß-galactosidase, and perform economical evaluation of production of GOSs (100%) between them. Using the response surface method, the optimal reaction time (3.9 h), initial lactose concentration (57.13%), and enzyme to lactose ratio (44.81 U/g) were obtained for the free enzyme, which provided a GOSs yield of 32.62%. For the immobilized enzyme, the optimal yield of GOSs (32.48%) was obtained under reaction time (3.09 h), initial lactose concentration (52.74%), and temperature (50.0 ℃). And it showed desirable reusability during five successive enzymatic reactions. The recovery rate of GOSs (100%) is 65% using silica gel filtration chromatography. The economical evaluation showed almost no difference in the manufacturing cost for the GOSs (100%) between these two systems, and that the recovery rate had a great impact on the cost.


Subject(s)
Aspergillus oryzae/enzymology , Enzymes, Immobilized/chemistry , Oligosaccharides/biosynthesis , beta-Galactosidase/chemistry , Chromatography, Gel , Costs and Cost Analysis , Enzymes, Immobilized/metabolism , Lactose/chemistry , Lactose/metabolism , Oligosaccharides/chemistry , Oligosaccharides/economics , Oligosaccharides/isolation & purification , Silica Gel , Spectroscopy, Fourier Transform Infrared , Temperature , beta-Galactosidase/metabolism
5.
Mol Nutr Food Res ; 65(14): e2001213, 2021 07.
Article in English | MEDLINE | ID: mdl-34021704

ABSTRACT

SCOPE: This study aims to roundly investigate whether Clostridium tyrobutyricum (Ct) alleviates inflammation via regulating immune cells in the small intestines. METHODS AND RESULTS: Mice are pre-treated with different concentrations of Ct follow by LPS stimulation. Ct maintains the mice body weight under inflammation. In response to LPS, 107 CFU mL-1 Ct decreases the mRNA expression of inflammatory cytokines in the duodenum, while 108 CFU mL-1 Ct reduces inflammatory cytokines expression in both duodenum and ileum and protected intestinal morphology. The small intestinal immune cells are analyzed using flow cytometry. Ct decreases the numbers of macrophages and mast cells in the intestines in response to LPS. In the duodenum, Ct enhances dentritic cells (DCs), regulatory T cells (Tregs), and T helper cell 17 (Th17) proportions. Ct decreases DCs and Tregs proportions, while enhances Th17 numbers in the ileum. The underlying mechanism of Ct in preventing inflammation may rely on the physiological immune cell composition of the intestines. In response to LPS, Ct may mainly stimulate Tregs via activating DCs in the duodenum while trigger Th17 cells in the ileum, thereby maintaining the intestinal homeostasis. CONCLUSION: Ct alleviates the LPS-induce inflammation via regulating different immune cell types in the small intestines, highlighting that Ct is a potential prophylactic probiotic in intestinal diseases.


Subject(s)
Clostridium tyrobutyricum , Inflammatory Bowel Diseases/therapy , Intestine, Small/immunology , Probiotics , Animals , Cytokines/metabolism , Homeostasis , Macrophages/immunology , Male , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Th17 Cells/immunology
6.
Food Funct ; 12(4): 1569-1579, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33459741

ABSTRACT

Galactooligosaccharides (GOS) have been identified as beneficial prebiotics for animals and human beings. Most studies have focused on the effect of GOS on the hindgut populated with abundant microbes. However, few research studies have been conducted on the small intestine, and many results are inconsistent due to the purity of GOS, commonly mixed with monosaccharides or lactose. Therefore, pure GOS with definite structures were prepared and used in the present study to evaluate their effects on intestinal barrier function, inflammatory responses and short-chain fatty acids (SCFAs) produced in the colon of mice challenged with lipopolysaccharide (LPS). The results of 1H and 13C nuclear magnetic resonance spectral analyses indicated that the main structures of GOS with a degree of polymerization of 3 (trisaccharide) and 4 (tetrasaccharide) are [ß-Gal-(1 → 6)-ß-Gal(1 → 4)-ß-Glc] and [ß-Gal-(1 → 6)-ß-Gal-(1 → 6)-ß-Gal-(1 → 4)-ß-Glc], respectively. The results of an in vivo study in mice showed that intragastric administration of 0.5 g per kg BW GOS attenuated intestinal barrier damage and inflammatory responses induced by LPS in the jejunum and ileum, as indicated by increasing villus height and villus-to-crypt ratio, up-regulated intestinal tight junction (ZO-1, occludin, and claudin-1) gene expression, and down-regulated pro-inflammatory cytokines such as IL-1ß, IL-6, IFN-γ, and TNF-α gene expression. Nevertheless, the protective effects of GOS on the intestinal barrier are independent of glucagon-like peptide 2. In addition, 0.5 g per kg BW GOS administration promoted the recovery of colonic acetate, propionate, butyrate, and total SCFA production reduced by LPS challenge. The obtained results provide practical evidence that pure GOS can act as protective agents for intestinal health.


Subject(s)
Inflammation/metabolism , Intestinal Mucosa , Oligosaccharides , Prebiotics , Animals , Body Weight/drug effects , Colon/metabolism , Cytokines/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiopathology , Lipopolysaccharides/adverse effects , Male , Mice , Mice, Inbred C57BL , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Tight Junctions/metabolism
7.
Nutrients ; 13(1)2021 Jan 13.
Article in English | MEDLINE | ID: mdl-33451114

ABSTRACT

This study aimed to investigate the effects of Clostridium tyrobutyricum (C. tyrobutyricum) on colonic immunity and the role of IL-22 in the protective function of C. tyrobutyricum. Mice were supplemented with 108 CFU/mL C. tyrobutyricum daily for 20 days, followed by injecting with LPS for 24 h. In vivo interference of IL-22 via injecting with an adeno-associated virus was conducted to elucidate the role of IL-22 in C. tyrobutyricum attenuating colonic inflammation. The results showed that C. tyrobutyricum decreased the mRNA expression of IL-6 and IL-1ß. C. tyrobutyricum enhanced the mRNA expression of IL-22 and the expression of MUC2 in the colon. The in vivo interference results showed that C. tyrobutyricum enhanced the mRNA expression of IL-6 and IL-1ß while decreased the expression of MUC2 after knocking down IL-22. The flow cytometric analysis showed that C. tyrobutyricum decreased the proportions of macrophages, DCs, and mast cells and effectively regulated the proportion of Th17 cells, indicating that C. tyrobutyricum may stimulate the expression of IL-22 via regulating Th17 cells. Our study concluded that C. tyrobutyricum protected against LPS-induced colonic barrier dysfunction and inflammation via IL-22 signaling, suggesting that C. tyrobutyricum could be a potential probiotic in regulating colonic health.


Subject(s)
Clostridium tyrobutyricum/physiology , Colitis/etiology , Colitis/metabolism , Interleukins/metabolism , Lipopolysaccharides/adverse effects , Signal Transduction , Animals , Colitis/prevention & control , Dendritic Cells , Disease Models, Animal , Immunomodulation , Intestinal Mucosa/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Knockout , Probiotics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Interleukin-22
8.
Biol Trace Elem Res ; 199(12): 4666-4674, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33512662

ABSTRACT

The present study was conducted to investigate the effects of supranutritional selenium nanoparticles (SeNPs) on immune and antioxidant capacity in rats. Forty male Sprague-Dawley (SD) rats were randomly divided into four groups and given intragastric administration of SeNPs at doses of 0, 0.2, 0.4, and 0.8 mg Se/kg BW, respectively, for 2 weeks. Serum immune parameters, serum and organic tissues (liver, heart, kidney) antioxidant indices, and liver mRNA expression of glutathione peroxidase 1 (GPx1) and glutathione peroxidase 4 (GPx4) were examined. The results showed that supranutritional doses of 0.4 and 0.8 mg Se/kg BW SeNPs promoted the immune responses in serum. SeNPs administration improved antioxidant capacity in the liver and kidney, and the best improvement on antioxidant capacity was found in the kidney. Furthermore, intragastric administration of SeNPs upregulated mRNA expression of GPx1 and GPx4 in the liver. The results obtained indicated that SeNPs administration at supranutritional levels had beneficial effects on immune and antioxidant capacity and supplemental SeNPs at dose of 0.4 mg Se/kg BW exhibited the best response in SD rats.


Subject(s)
Nanoparticles , Selenium , Animals , Antioxidants , Liver , Male , Rats , Rats, Sprague-Dawley , Selenium/pharmacology
9.
Oxid Med Cell Longev ; 2020: 7680276, 2020.
Article in English | MEDLINE | ID: mdl-32922654

ABSTRACT

The effects of selenium nanoparticles (SeNPs) on the antioxidant capacity in Sprague-Dawley (SD) rats were investigated. The rats were given intragastric administration of an SeNP suspension at doses of 0, 2, 4, and 8 mg Se/kg BW for two weeks. The antioxidant capacity in serum and organic tissues (liver, heart, and kidney) and the gene expression levels of glutathione peroxidase 1 (GPX1) and glutathione peroxidase 4 (GPX4) in the liver were measured. Buffalo rat liver (BRL) cell lines were further constructed to explore the cytotoxicity mechanism induced by SeNPs through the determination of antioxidant capacity; cell activity; apoptosis; and Caspase-3, Caspase-8, and Caspase-9 family activities. The results showed that SeNP administration over 4.0 mg Se/kg BW decreased the antioxidant capacities in the serum, liver, and heart and downregulated mRNA expression of GPX1 and GPX4 in the liver. The BRL cell line experiments showed that treatment with over 24 µM SeNPs decreased the viability of the cells and damaged the antioxidant capacity. Flow cytometry analysis showed that decreased cell viability induced by SeNPs is mainly due to apoptosis, rather than cell necrosis. Caspase-3 and Caspase-8 activities were also increased when BRL cells were treated with 24 µM and 48 µM SeNPs. Taken together, a nonlethal level of SeNPs could impair the antioxidant capacity in serum and organic tissues of rats, and the liver is the most sensitive to the toxicity of SeNPs. A pharmacological dose of SeNPs could lead to cytotoxicity and induce cell death through apoptosis and extrinsic pathways contributing to SeNP-induced apoptosis in BRL cells.


Subject(s)
Hepatocytes/pathology , Liver/pathology , Metal Nanoparticles/chemistry , Selenium/pharmacology , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Apoptosis/drug effects , Caspases/metabolism , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Hepatocytes/drug effects , Hepatocytes/ultrastructure , Liver/drug effects , Liver/ultrastructure , Male , Metal Nanoparticles/ultrastructure , Organ Specificity/drug effects , Oxidation-Reduction , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Glutathione Peroxidase GPX1
10.
Theriogenology ; 114: 70-80, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-29602134

ABSTRACT

In this study, we investigated the effects of Selenium (Se) on the proliferation of and steroidogenesis in goat luteinized granulosa cells (LGCs) and elucidated the mechanisms underlying these effects. Our results showed that proliferating cell nuclear antigen (PCNA), Akt, and phosphoinositide 3-kinase (PI3K) were expressed mainly in ovarian oocytes and granulosa cells (GCs). We observed that 5 ng/mL Se significantly stimulated LGC proliferation, which could be attributed to increases in PCNA, cyclin-dependent kinase 1 (CDK1), phosphorylated adenosine monophosphate-activated protein kinase (p-AMPK; Thr172), and phosphorylated Akt (p-Akt; Ser473) and decreases in p21 (P < 0.05). Se treatment also significantly increased estradiol (E2) production, which could be, at least partially, due to increased levels of 3ß-hydroxysteroid dehydrogenase(3ß-HSD), steroidogenic acute regulatory protein (StAR), p-Akt (Ser473), and cyclic adenosine monophosphate (cAMP) (P < 0.05); however, follicle-stimulating hormone (FSH) significantly enhanced the production of E2, progesterone (P4) and cAMP (P < 0.05). Moreover, Se treatment stimulated proliferation and the synthesis of E2 and cAMP in the presence of FSH (P < 0.05). Additionally, the expression of antioxidant-related genes [glutathione peroxidase (GSH-Px) and superoxide dismutase 2 (SOD2)] and the activity of GSH-Px and SOD were progressively elevated by Se treatment (P < 0.05). These data suggested that Se plays an important role in the proliferation of and steroidogenesis in LGC by activating the PI3K/Akt and AMPK pathways, thereby increasing the expression of its downstream cell-cycle- and steroid-synthesis-related genes, as well as regulating cellular oxidative stress.


Subject(s)
Cell Proliferation/drug effects , Goats , Granulosa Cells/drug effects , Selenium/pharmacology , Adenylate Kinase/genetics , Adenylate Kinase/metabolism , Animals , Antioxidants/metabolism , Cells, Cultured , Female , Follicle Stimulating Hormone/administration & dosage , Follicle Stimulating Hormone/pharmacology , Gene Expression Regulation/drug effects , Granulosa Cells/physiology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...