Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Int J Mol Sci ; 24(18)2023 Sep 09.
Article in English | MEDLINE | ID: mdl-37762180

ABSTRACT

Obesity and metabolic comorbidities are associated with gut permeability. While high-fructose and Western-style diet (WSD) disrupt intestinal barrier function, oral administration of human α-defensin 5 (HD5) and ß-defensin 2 (hBD2) is believed to improve intestinal integrity and metabolic disorders. Eighty-four male C57BL/6J mice were fed a WSD or a control diet (CD) ± fructose (F) for 18 weeks. In week 13, mice were randomly divided into three intervention groups, receiving defensin fragment HD51-9, full-length hBD2, or bovine serum albumin (BSA)-control for six weeks. Subsequently, parameters of hepatic steatosis, glucose metabolism, and gut barrier function were assessed. WSDF increased body weight and hepatic steatosis (p < 0.01) compared to CD-fed mice, whereas peptide intervention decreased liver fat (p < 0.05) and number of hepatic lipid droplets (p < 0.01) compared to BSA-control. In addition, both peptides attenuated glucose intolerance by reducing blood glucose curves in WSDF-fed mice. Evaluation of gut barrier function revealed that HD51-9 and hBD2 improve intestinal integrity by upregulating tight junction and mucin expression. Moreover, peptide treatment restored ileal host defense peptides (HDP) expression, likely by modulating the Wnt, Myd88, p38, and Jak/STAT pathways. These findings strongly suggest that α- and ß-defensin treatment improve hepatic steatosis, glucose metabolism, and gut barrier function.

2.
Nutrients ; 15(2)2023 Jan 11.
Article in English | MEDLINE | ID: mdl-36678247

ABSTRACT

Vitamin A and D deficiencies are associated with immune modulatory effects and intestinal barrier impairment. However, the underlying mechanisms remain unclear. C57BL/6J mice were fed either a diet lacking in vitamin A (VAd), vitamin D (VDd) or a control diet (CD) for 12 weeks. Gut barrier function, antimicrobial peptide (AMP) defense and regulatory pathways were assessed. VAd mice compared to CD mice showed a reduced villus length in the ileum (p < 0.01) and decreased crypt depth in the colon (p < 0.05). In both VAd- and VDd-fed mice, ileal α-defensin 5 (p < 0.05/p < 0.0001 for VAd/VDd) and lysozyme protein levels (p < 0.001/p < 0.0001) were decreased. Moreover, mRNA expression of lysozyme (p < 0.05/p < 0.05) and total cryptdins (p < 0.001/p < 0.01) were reduced compared to controls. Furthermore, matrix metalloproteinase-7 (Mmp7) mRNA (p < 0.0001/p < 0.001) as well as components of the Wnt signaling pathway were decreased. VAd- and VDd-fed mice, compared to control mice, exhibited increased expression of pro-inflammatory markers and ß-defensins in the colon. Organoid cell culture confirmed that vitamins A and D regulate AMP expression, likely through the Jak/STAT5 signaling pathway. In conclusion, our data show that vitamin A and D regulate intestinal antimicrobial peptide defense through Wnt and STAT5 signaling pathways.


Subject(s)
Muramidase , Vitamin A , Animals , Mice , STAT5 Transcription Factor , Mice, Inbred C57BL , Vitamins , Diet , Signal Transduction , RNA, Messenger/genetics , Antimicrobial Peptides
3.
Front Immunol ; 12: 678360, 2021.
Article in English | MEDLINE | ID: mdl-34177920

ABSTRACT

Defects in the mucosal barrier have been associated with metabolic diseases such as obesity and non-alcoholic fatty liver disease (NAFLD). Mice fed a Western-style diet (WSD) develop obesity and are characterized by a diet-induced intestinal barrier dysfunction, bacterial endotoxin translocation and subsequent liver steatosis. To examine whether inulin or sodium butyrate could improve gut barrier dysfunction, C57BL/6 mice were fed a control diet or a WSD ± fructose supplemented with either 10% inulin or 5% sodium butyrate for 12 weeks respectively. Inulin and sodium butyrate attenuated hepatosteatitis in the WSD-induced obesity mouse model by reducing weight gain, liver weight, plasma and hepatic triglyceride level. Furthermore, supplementation with inulin or sodium butyrate induced expression of Paneth cell α-defensins and matrix metalloproteinase-7 (MMP7), which was impaired by the WSD and particularly the fructose-added WSD. Effects on antimicrobial peptide function in the ileum were accompanied by induction of ß-defensin-1 and tight junction genes in the colon resulting in improved intestinal permeability and endotoxemia. Organoid culture of small intestinal crypts revealed that the short chain fatty acids (SCFA) butyrate, propionate and acetate, fermentation products of inulin, induce Paneth cell α-defensin expression in vitro, and that histone deacetylation and STAT3 might play a role in butyrate-mediated induction of α-defensins. In summary, inulin and sodium butyrate attenuate diet-induced barrier dysfunction and induce expression of Paneth cell antimicrobials. The administration of prebiotic fiber or sodium butyrate could be an interesting therapeutic approach to improve diet-induced obesity.


Subject(s)
Butyric Acid/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Inulin/administration & dosage , Obesity/metabolism , Pore Forming Cytotoxic Proteins/biosynthesis , Prebiotics/administration & dosage , Animal Feed , Animals , Biomarkers , Dietary Supplements , Disease Models, Animal , Female , Gastrointestinal Microbiome/drug effects , Liver/drug effects , Liver/metabolism , Mice , Obesity/drug therapy , Obesity/etiology , Permeability , Tight Junctions/metabolism
4.
Int J Med Microbiol ; 311(4): 151499, 2021 May.
Article in English | MEDLINE | ID: mdl-33864957

ABSTRACT

The gut barrier has been recognized as being of relevance in the pathogenesis of multiple different diseases ranging from inflammatory bowel disease, irritable bowel syndrome, inflammatory joint disease, fatty liver disease, and cardiometabolic disorders. The regulation of the gut barrier is, however, poorly understood. Especially, the role of food components such as sugars and complex carbohydrates has been discussed controversially in this respect. More recently, the intestinal microbiota has been proposed as an important regulator of the gut barrier. Whether the microbiota affects the barrier by its own, or whether food components such as carbohydrates mediate their effects through alterations of the microbiota composition or its metabolites, is still not clear. In this review, we will summarize the current knowledge on this topic derived from both animal and human studies and discuss data for possible clinical impact.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Microbiota , Animals , Carbohydrates , Diet , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...