Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.697
Filter
1.
Sci Rep ; 14(1): 20367, 2024 09 02.
Article in English | MEDLINE | ID: mdl-39223174

ABSTRACT

This study elucidated the unique pathological features of tissue healing by magnamosis and revealed the changes in landmark molecule expression levels related to collagen synthesis and tissue hypoxia. Forty-eight male Sprague-Dawley rats were divided into the magnamosis and suture anastomosis groups, and gastrojejunal anastomosis surgery was performed. Rats were dissected at 6, 24, and 48 h and 5, 6, 8, 10, and 12 days postoperatively. Hematoxylin, eosin, and Masson's trichrome staining were used to evaluate granulation tissue proliferation and collagen synthesis density at the anastomosis site. Immunohistochemistry was used to measure TGF-ß1 and HIF-1α expression levels. Magnamosis significantly shortened the operation time, resulting in weaker postoperative abdominal adhesions (P < 0.0001). Histopathological results showed a significantly lower granulation area in the magnamosis group than in the suture anastomosis group (P = 0.0388), with no significant difference in the density of collagen synthesis (P = 0.3631). Immunohistochemistry results indicated that the magnamosis group had significantly lower proportions of TGF-ß1-positive cells at 24 (P = 0.0052) and 48 h (P = 0.0385) postoperatively and HIF-1α-positive cells at 24 (P = 0.0402) and 48 h postoperatively (P = 0.0005). In a rat model of gastrojejunal anastomosis, magnamosis leads to improved tissue healing at the gastrojejunal anastomosis, associated with downregulated expression levels of TGF-ß1 and HIF-1α.


Subject(s)
Anastomosis, Surgical , Hypoxia-Inducible Factor 1, alpha Subunit , Rats, Sprague-Dawley , Transforming Growth Factor beta1 , Wound Healing , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Transforming Growth Factor beta1/metabolism , Male , Rats , Jejunum/surgery , Jejunum/metabolism , Down-Regulation , Collagen/metabolism , Stomach/surgery , Stomach/pathology
2.
J Diabetes Res ; 2024: 5544296, 2024.
Article in English | MEDLINE | ID: mdl-39263491

ABSTRACT

Aim: The aim of the study is to identify the regulatory role of intestinal sweet taste receptors (STRs) and glucose transporters (SGLT1, GLUT2) and gut peptide secretion in duodenal-jejunal bypass (DJB)-ameliorated glycemic control in Type 2 diabetes. Materials and Methods: DJB and sham surgeries were performed in streptozotocin-induced diabetic male rats. The blood GLP-1 and GLP-2 levels were evaluated under feeding and fasting conditions. The expression of STRs (T1R2, T1R3), sweet taste signaling effector (Gα-gustducin), SGLT1, and GLUT2 was detected in the intestinal alimentary limb (A limb), biliopancreatic limb (BP limb), and common limb (C limb). The effects of STR inhibition on glucose control were measured with lactisole. Results: Glucose tolerance was improved in DJB-operated rats compared with the sham group, similar to that of normal control rats, without significant differences in food intake and body weight. The plasma GLP-1 levels of DJB rats were increased under diet-fed condition, and GLP-2 levels were increased after fasting. The villus height and crypt depth were significantly increased in the A limb of DJB-operated rats. In addition, GLP-1 expression was restored in enterocytes. The expression of T1R2, Gα-gustducin, and SGLT1 was elevated in the A limb after DJB, while GLUT2 was downregulated in the A, BP, and C limbs. The localization of GLUT2 was normalized in the three intestinal limbs after DJB. However, the beneficial effects of DJB on glucose control were abolished in the presence of lactisole in vivo. Conclusion: DJB ameliorates glycemic control probably by restoring STR-mediated glucose sensing and absorption with the responses of GLP-1 and GLP-2 to carbohydrate.


Subject(s)
Blood Glucose , Diabetes Mellitus, Experimental , Duodenum , Glucagon-Like Peptide 1 , Glucose Transporter Type 2 , Jejunum , Receptors, G-Protein-Coupled , Sodium-Glucose Transporter 1 , Animals , Male , Sodium-Glucose Transporter 1/metabolism , Glucose Transporter Type 2/metabolism , Jejunum/surgery , Jejunum/metabolism , Duodenum/surgery , Duodenum/metabolism , Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Experimental/metabolism , Rats , Receptors, G-Protein-Coupled/metabolism , Blood Glucose/metabolism , Glucagon-Like Peptide 1/metabolism , Intestinal Absorption , Glucagon-Like Peptide 2/metabolism , Glucose/metabolism , Rats, Sprague-Dawley , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/surgery , Transducin/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/surgery
3.
Anim Biotechnol ; 35(1): 2391520, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39222080

ABSTRACT

MicroRNAs (miRNAs) were identified to be involved in various biological functions by regulating the degradation or suppressing the translation of their downstream target genes. Recent studies have identified miR-29a play a key role in functions of mammal cell differentiation, proliferation, apoptosis, and signal transduction. However, the underlying functions for miR-29a in jejunal epithelial cells biological function still to be investigated. In order to explore the yak jejunal epithelial cells proliferation and barrier dysfunction with over expression of miR-29a gene, three 0-day-old Pamir male yaks were randomly selected and slaughtered in present study, and the jejunal epithelial cells were isolated and cultured to determine yak jejunal epithelial cells proliferation and protein composition on differential expression of miR-29a gene in Pamir plateau. Here, we demonstrated that the overexpression of miR-29a gene could inhibit the proliferation of Pamir yaks jejunum epithelial cells, and contribute to the apoptosis of Pamir yaks jejunal epithelial cells with some extent. A total of 133 differentially expressed proteins were identified in different expression of miR-29a groups by label-free Mass Spectrometry (MS), which could be concluded to two predominant themes: cell proliferation and inflammatory response. Interestingly, GPR41, as a bridge protein, was contacted two predominant themes to involved in Pamir Yaks jejunal mechanical barrier PPI network, and the target proteins displayed strong mutual interactions in the complex PPI network. Overall, our study suggested that the over-expression miR-29a inhibited the jejunal epithelial cells proliferation and the expressions of specific proteins, which damaged jejunal barrier function to slow down the intestine structure and function advanced mature development during young livestock period for influence the enhanced performance of production efficiency.


Subject(s)
Apoptosis , Cell Proliferation , Epithelial Cells , Jejunum , MicroRNAs , Animals , Cattle/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Epithelial Cells/physiology , Epithelial Cells/metabolism , Apoptosis/genetics , Apoptosis/physiology , Jejunum/cytology , Jejunum/metabolism , Cell Proliferation/genetics , Male
4.
Molecules ; 29(16)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39202934

ABSTRACT

This study reports the synthesis of 2-thioxo-1,3-dithiol-carboxamides (TDTCAs) under mild conditions at room temperature using HBTU as a coupling agent, which significantly improved amide bond formation. The synthesized compounds were characterized using several analytical techniques, including 1H and 13C NMR spectroscopy, and HRMS, confirming their intended structures and structural integrity. A DFT computational study at the B3LYP/6-31G(d,p) level was conducted on the four synthesized compounds to compare their electronic properties and molecular structures. The results showed that these compounds demonstrated antispasmodic effects on jejunum contractions. Molecular docking revealed that compounds c and d displayed the highest docking scores on potassium and voltage-gated calcium channels and adrenergic receptors. In summary, compounds c and d exhibit antispasmodic effects, potentially blocking alpha-adrenergic receptors and calcium channels, thus providing a scientific basis for their potential use in treating gastrointestinal disorders.


Subject(s)
Molecular Docking Simulation , Parasympatholytics , Parasympatholytics/pharmacology , Parasympatholytics/chemistry , Parasympatholytics/chemical synthesis , Animals , Density Functional Theory , Drug Design , Molecular Structure , Structure-Activity Relationship , Jejunum/metabolism , Jejunum/drug effects
5.
Sci Rep ; 14(1): 19235, 2024 08 20.
Article in English | MEDLINE | ID: mdl-39164530

ABSTRACT

This study aimed to determine the effects of Saccharomyces yeast postbiotics on cell turnover, immune responses, and oxidative stress in the jejunal mucosa of pigs. Thirty-two newly weaned pigs at 6.05 ± 0.24 kg were assigned to two dietary treatments based on a randomized complete block design. The treatments were control group receiving a basal diet and a group supplemented with Saccharomyces yeast postbiotics (175 g/ton diet) in the basal diet. After 35 d of the study, pigs were euthanized and jejunal mucosa were collected to assess immune status, oxidative stress, barrier markers, cell proliferation, and apoptosis. Saccharomyces yeast postbiotics reduced (P < 0.05) the fecal score from d 3 to d 7 and tended to increase the gene expression of interferon-γ (IFN-γ) (P = 0.071) and mammalian/mechanistic target of rapamycin (mTOR) (P = 0.080), decrease the gene expression of B-cell lymphoma 2-associated X protein 1 (BAX1) (P < 0.05), tended to decrease the gene expression of serum and glucocorticoid-induced protein kinase 1 (SGK1) (P = 0.066), increased (P < 0.05) cell proliferation in the crypts, and tended to increase the villus height (P = 0.078) and crypt depth (P = 0.052) in the jejunum. In conclusion, the supplementation of Saccharomyces yeast postbiotics in nursery diets reduced diarrhea within the first week after weaning and provided protection to the villi in the jejunum by enhancing the immune responses of nursery pigs, promoting crypt cell proliferation, and reducing the expression of genes associated with apoptosis without affecting inflammatory and oxidative stress status in the jejunum of the nursery pigs.


Subject(s)
Intestinal Mucosa , Jejunum , Oxidative Stress , Saccharomyces cerevisiae , Animals , Oxidative Stress/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Swine , Jejunum/metabolism , Jejunum/immunology , Jejunum/drug effects , Jejunum/microbiology , Probiotics/administration & dosage , Probiotics/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Weaning , Animal Feed
6.
Eur J Pharmacol ; 981: 176881, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39127300

ABSTRACT

BACKGROUND: Increasing epidemiologic studies have shown a positive correlation between obesity and chronic diarrhea. Nevertheless, the precise etiology remains uncertain. METHODS: We performed a comprehensive proteomics analysis utilizing the data-independent acquisition (DIA) technique on jejunal tissues from patients with obesity and chronic diarrhea (OD, n = 33), obese patients (OB, n = 10), and healthy controls (n = 8). Differentially expressed proteins (DEPs) in OD vs. control and OD vs. OB comparisons were subjected to pathway enrichment and protein-protein interaction (PPI) network analysis. Machine learning algorithms were adopted on overlapping DEPs in both comparisons. The candidate protein was further validated using Western blot, immunohistochemistry (IHC), and in vitro experiments. RESULTS: We identified 189 and 228 DEPs in OD vs. control and OD vs. OB comparisons, respectively. DEPs in both comparisons were co-enriched in extracellular matrix (ECM) organization. Downregulated DEPs were associated with tight junction and ECM-receptor interaction in OD vs. control and OD vs. OB comparisons, respectively. Machine learning algorithms selected 3 proteins from 14 overlapping DEPs in both comparisons, among which collagen alpha-1(III) chain (COL3A1) was identified as a core protein in PPI networks. Western blot and IHC verified the expression of COL3A1. Moreover, the tight junction-related proteins decreased after the knockdown of COL3A1 in Caco2 intestinal cells upon PA challenge, consistent with the proteomics results. CONCLUSIONS: We generated in-depth profiling of a proteomic dataset from samples of OD patients and provided unique insights into disease pathogenesis. COL3A1 was involved in the crosstalk between obesity and intestinal homeostasis via the ECM-receptor interaction pathway.


Subject(s)
Collagen Type III , Diarrhea , Machine Learning , Obesity , Protein Interaction Maps , Proteomics , Humans , Proteomics/methods , Obesity/metabolism , Diarrhea/metabolism , Male , Female , Collagen Type III/metabolism , Adult , Middle Aged , Caco-2 Cells , Jejunum/metabolism , Case-Control Studies
7.
Toxicol Lett ; 400: 16-23, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39096942

ABSTRACT

Diacylglycerol O-acyltransferase 1 (DGAT1) is a key enzyme for fat absorption step in the enterocytes. We previously reported that DGAT1 inhibition increased plasma alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities in corn oil-loaded rats via protein kinase C (PKC) activation. In the present study, we investigated the mechanism with respect to the morphology and permeability of the small intestine, focusing on PKC function, and found that shortening of the intestinal villi and a decrease in the number of tdT-mediated dUTP-biotin nick-end labeling-positive cells in the tips of the villi were observed in the jejunum of DGAT1 inhibitor-treated rats loaded with corn oil. These results suggested that the tips of the villi were shed into the intestinal lumen. Next, fluorescein isothiocyanate-dextran, 110 kDa (FD-110) was administered intraduodenally to DGAT1 inhibitor-treated rats loaded with corn oil and we found that plasma FD-110 concentrations increased, indicating that the intestinal permeability to molecules with a molecular weight of approximately 110,000 (e.g., ALT and AST) increased. Taken together, the present results suggested that DGAT1 inhibitor-treatment in combination with corn oil causes ALT and AST to leak from the enterocytes into the blood by shedding the tips of the intestinal villi and increasing intestinal permeability.


Subject(s)
Alanine Transaminase , Aspartate Aminotransferases , Corn Oil , Diacylglycerol O-Acyltransferase , Intestinal Mucosa , Permeability , Animals , Alanine Transaminase/blood , Male , Aspartate Aminotransferases/blood , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Diacylglycerol O-Acyltransferase/metabolism , Permeability/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Rats , Dextrans , Protein Kinase C/metabolism , Protein Kinase C/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Fluorescein-5-isothiocyanate/analogs & derivatives , Jejunum/drug effects , Jejunum/metabolism , Intestinal Absorption/drug effects , Rats, Sprague-Dawley , Rats, Wistar , Intestinal Barrier Function
8.
Microb Pathog ; 195: 106852, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39147213

ABSTRACT

The purpose of this study was to evaluate the ability of Bacillus subtilis JATP3 to stimulate immune response and improve intestinal health in piglets during the critical weaning period. Twelve 28-day-old weaned piglets were randomly divided into two groups. One group was fed a basal diet, while the other group was fed a basal diet supplemented with B. subtilis JATP3 (1 × 109 CFU/mL; 10 mL) for 28 days. The results revealed a significant increase in the intestinal villus gland ratio of weaned piglets following the inclusion of B. subtilis JATP3 (P < 0.05). Inclusion of a probiotic supplement improve the intestinal flora of jejunum and ileum of weaned piglets. Metabolomics analysis demonstrated a notable rise in citalopram levels in the jejunum and ileum, along with elevated levels of isobutyric acid and isocitric acid in the ileum. The results of correlation analysis show that indicated a positive correlation between citalopram and microbial changes. Furthermore, the probiotic-treated group exhibited a significant upregulation in the relative expression of Claudin, Zonula Occludens 1 (ZO-1), and Interleukin 10 (IL-10) in the jejunum and ileum, while displaying a noteworthy reduction in the relative expression of Interleukin 1ß (IL-1ß). Overall, these findings suggest that B. subtilis JATP3 can safeguard intestinal health by modulating the structure of the intestinal microbiota and their metabolites, wherein citalopram might be a key component contributing to the therapeutic effects of B. subtilis JATP3.


Subject(s)
Bacillus subtilis , Citalopram , Gastrointestinal Microbiome , Ileum , Jejunum , Probiotics , Weaning , Animals , Gastrointestinal Microbiome/drug effects , Bacillus subtilis/metabolism , Swine , Probiotics/administration & dosage , Probiotics/pharmacology , Ileum/microbiology , Ileum/immunology , Citalopram/pharmacology , Jejunum/microbiology , Jejunum/immunology , Jejunum/metabolism , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Metabolomics , Intestinal Mucosa/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Zonula Occludens-1 Protein/metabolism , Dietary Supplements
9.
PLoS One ; 19(8): e0308983, 2024.
Article in English | MEDLINE | ID: mdl-39146343

ABSTRACT

We previously demonstrated that postruminal casein infusion and exogenous glucagon-like peptide 2 (GLP-2) administration independently stimulated growth and carbohydrase activity of the pancreas and jejunal mucosa in cattle. The objective of the current study was to profile the jejunal mucosal transcriptome of cattle using next-generation RNA sequencing in response to postruminal casein infusion and exogenous GLP-2. Twenty-four Holstein steers [250 ± 23.1 kg body weight (BW)] received a continuous abomasal infusion of 3.94 g raw corn starch/kg of BW combined with either 0 or 1.30 g casein/kg of BW for 7 d. Steers received subcutaneous injections at 0800 and 2000 h to provide either 0 or 100 µg GLP-2/kg of BW per day. At the end of the 7-d treatment period, steers were slaughtered for collection of the jejunal mucosa. Total RNA was extracted from jejunal mucosal tissue, strand-specific cDNA libraries were prepared, and RNA sequencing was conducted to generate 150-bp paired-end reads at a depth of 40 M reads per sample. Differentially expressed genes (DEG), KEGG pathway enrichment, and gene ontology enrichment were determined based on the FDR-corrected P-value (padj). Exogenous GLP-2 administration upregulated (padj < 0.05) 667 genes and downregulated 1,101 genes of the jejunal mucosa. Sphingolipid metabolism, bile secretion, adherens junction, and galactose metabolism were among the top KEGG pathways enriched with upregulated DEG (padj < 0.05) in response to exogenous GLP-2 administration. The top gene ontologies enriched with upregulated DEG (padj < 0.05) in response to exogenous GLP-2 administration included nutrient metabolic processes, brush border and bicellular tight junction assembly, and enzyme and transporter activities. Exogenous GLP-2 administration increased or tended to increase (padj < 0.10) brush border carbohydrase (MGAM, LCT, TREH), hexose transporter (SLC5A1, SLC2A2), and associated transcription factor (HNF1, GATA4, KAT2B) mRNA expression of the jejunal mucosa. Gene ontologies and KEGG pathways that were downregulated (padj < 0.05) in response to exogenous GLP-2 were related to genetic information processing. Postruminal casein infusion downregulated (padj < 0.05) 7 jejunal mucosal genes that collectively did not result in enriched KEGG pathways or gene ontologies. This study highlights some of the transcriptional mechanisms associated with increased growth, starch assimilation capacity, and barrier function of the jejunal mucosa in response to exogenous GLP-2 administration.


Subject(s)
Caseins , Glucagon-Like Peptide 2 , Intestinal Mucosa , Jejunum , Transcriptome , Animals , Cattle , Glucagon-Like Peptide 2/administration & dosage , Glucagon-Like Peptide 2/pharmacology , Jejunum/metabolism , Jejunum/drug effects , Caseins/genetics , Caseins/administration & dosage , Transcriptome/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Male , Abomasum/drug effects , Abomasum/metabolism , Gene Expression Profiling
10.
Nutrients ; 16(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39125449

ABSTRACT

We aimed to characterize the anti-obesity and anti-atherosclerosis effects of Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032 using high-fat diet (HFD)-fed obese C57BL/6 mice. We divided the mice into control (CON), HFD, HFD with 108 CFU/kg/day probiotics (HFD + KL, HY7301:KY1032 = 1:1), and HFD with 109 CFU/kg/day probiotics (HFD + KH, HY7301:KY1032 = 1:1) groups and fed/treated them during 7 weeks. The body mass, brown adipose tissue (BAT), inguinal white adipose tissue (iWAT), and epididymal white adipose tissue (eWAT) masses and the total cholesterol and triglyceride concentrations were remarkably lower in probiotic-treated groups than in the HFD group in a dose-dependent manner. In addition, the expression of uncoupling protein 1 in the BAT, iWAT, and eWAT was significantly higher in probiotic-treated HFD mice than in the HFD mice, as demonstrated by immunofluorescence staining and Western blotting. We also measured the expression of cholesterol transport genes in the liver and jejunum and found that the expression of those encoding liver-X-receptor α, ATP-binding cassette transporters G5 and G8, and cholesterol 7α-hydroxylase were significantly higher in the HFD + KH mice than in the HFD mice. Thus, a Lactobacillus HY7601 and KY1032 mixture with 109 CFU/kg/day concentration can assist with body weight regulation through the management of lipid metabolism and thermogenesis.


Subject(s)
Cholesterol , Diet, High-Fat , Energy Metabolism , Lactobacillus , Mice, Inbred C57BL , Probiotics , Animals , Diet, High-Fat/adverse effects , Probiotics/pharmacology , Probiotics/administration & dosage , Cholesterol/metabolism , Cholesterol/blood , Energy Metabolism/drug effects , Male , Mice , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Obesity/metabolism , Obesity/microbiology , Adipose Tissue, White/metabolism , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Adipose Tissue/metabolism , Liver/metabolism , Lactobacillus plantarum , Jejunum/metabolism , Jejunum/drug effects , Jejunum/microbiology
11.
Poult Sci ; 103(9): 104046, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39033572

ABSTRACT

Carnosine is a physiologically important molecule in normal human body functions. Chicken meat is an excellent source of carnosine; especially slow-growing Korat chicken (KR) females have a high carnosine content in their meat. The carnosine content of chicken meat can be increased by dietary supplementation of ß-alanine (ßA) and L-histidine (L-His). Our objective was to reveal the pathways and genes through jejunal transcriptomic profiling related to ßA and L-His absorption and transportation. We collected whole jejunum samples from 5 control and 5 experimental KR chicken, fed with 1% ßA and 0.5% L-His supplementation. A total of 407 differentially expressed genes (P < 0.05, log2 fold change ≥2) were identified, 272 of which were down-regulated and 135 up-regulated in the group with dietary supplementation compared to the control group. Based on the integrated analysis of the protein-protein interaction network and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway maps, 87 gene ontology terms were identified and 6 KEGG pathways were significantly (P < 0.05) enriched in the jejunum. The analyses revealed 6 key genes, KCND3, OPRM1, CCK, GCG, TRH, and GABBR2, that are related to neuroactive ligand-receptor interaction and the calcium signaling pathway. These findings give insight regarding the molecular mechanism related to carnosine precursor absorption and transportation in the jejunum and help to identify useful molecular markers for improving the carnosine content in slow-growing KR chicken meat.


Subject(s)
Animal Feed , Carnosine , Chickens , Gene Expression Profiling , Jejunum , Animals , Chickens/genetics , Chickens/growth & development , Chickens/metabolism , Carnosine/metabolism , Jejunum/metabolism , Gene Expression Profiling/veterinary , Animal Feed/analysis , Female , Dietary Supplements/analysis , Diet/veterinary , Transcriptome , beta-Alanine/metabolism , Histidine/metabolism , Avian Proteins/genetics , Avian Proteins/metabolism
12.
PLoS One ; 19(7): e0307411, 2024.
Article in English | MEDLINE | ID: mdl-39024309

ABSTRACT

The microbiological environment and their corresponding secreted metabolite spectrum are an essential modulator of the enterocyte function, effecting the whole organism. Intestinal porcine jejunal epithelial cell line (IPEC-J2) is an established in vitro model for differentiation of enterocytes in different cell culture models. An improved oxygen supply seems to be the main reason for differentiation in an air-liquid-interface culture, but this has not yet been conclusively clarified. In this context, the nutrition of the cell and its influence on the metabolism is also of crucial importance. The interest in short-chain fatty acids (SCFAs) has grown steadily in recent years due to their clinical relevance in certain diseases such as multiple sclerosis and other inflammatory diseases, but not much is known of FFAR2 and FFAR3 (free fatty acid receptor 2 and 3) in pigs. We want to address the questions: 1. about the distribution of FFAR2 and FFAR3 in vivo and in vitro in sus scrofa 2. whether there is an influence of propionic acid, glucose content and cultivation on metabolism of enterocytes? The morphological analysis of FFAR2 and FFAR3 in vivo was investigated through immunostaining of frozen sections of the porcine gut segments jejunum, ileum and colon. Both receptors are expressed along the gut and were found in the smooth muscle cells of the tunica muscularis and lamina muscularis mucosae. Furthermore, a high expression of FFAR2 and a low expression of FFAR3 in the enteric nerve system was also observed in jejunum, ileum and colon of sus scrofa. In addition, FFAR2 and FFAR3 within the vessels was investigated. FFAR3 showed a strong expression on endothelial cells of veins and lymphatic vessels but was not detectable on arteries. Furthermore, we demonstrate for the first time, FFAR2 and FFAR3 in IPEC-J2 cells on RNA- and protein level, as well as with confocal microscopy. In addition, ENO1 and NDUFA4 were investigated on RNA-level in IPEC-J2 cells as 2 important genes, which play an essential role in metabolism. Here, NDUFA4 is detected in the model animal sus scrofa as well as in the porcine cell line IPEC-J2. A potential impact of propionic acid and/or glucose and/or cultivation method on the metabolism of the cells was tested with the Seahorse analyzer. Here, a significant higher ECAR was observed in the SMC than in the OCR. In summary, we were able to show that the cultivation system appears to have a greater influence than the medium composition or nutrition of the cells. However, this can be modulated by incubation time or combination of different SCFAs.


Subject(s)
Glucose , Propionates , Animals , Propionates/metabolism , Glucose/metabolism , Swine , Cell Line , Cell Culture Techniques/methods , Metabolome , Receptors, G-Protein-Coupled/metabolism , Jejunum/metabolism , Jejunum/cytology , Enterocytes/metabolism , Intestinal Mucosa/metabolism
13.
J Agric Food Chem ; 72(29): 16287-16297, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38986018

ABSTRACT

Variances in the biological functions of astaxanthin geometric isomers (i.e., all-E, Z) are related to their intestinal absorption, but the mechanism of isomer absorption mediated by transporters remains unclear. Here, models of in vitro cell overexpression, in situ intestinal perfusion, and in vivo mouse inhibition were employed to investigate the impact of cluster of differentiation 36 (CD36) on the absorption of astaxanthin isomers. Cells overexpressing CD36 notably enhanced the uptake of Z-astaxanthin, particularly the 9-Z-isomer (47.76%). The absorption rate and permeability of Z-astaxanthin surpassed that of the all-E-isomer by the in situ model. Furthermore, the addition of the CD36-specific inhibitor sulfo-N-succinimidyl oleate significantly reduced the absorption of Z-astaxanthin in the mouse duodenum and jejunum, especially the 9-Z-isomer (57.66%). Molecular docking and surface plasmon resonance techniques further validated that 9-Z-astaxanthin binds to more amino acids of CD36 with higher affinity and in a fast-binding, fast-dissociating mode, thus favoring transport. Our findings elucidate, for the first time, the mechanism of the CD36-mediated transmembrane transport of astaxanthin geometric isomers.


Subject(s)
CD36 Antigens , Intestinal Absorption , Molecular Docking Simulation , Xanthophylls , Xanthophylls/metabolism , Xanthophylls/chemistry , Animals , CD36 Antigens/metabolism , CD36 Antigens/genetics , Mice , Intestinal Absorption/drug effects , Male , Humans , Isomerism , Mice, Inbred C57BL , Jejunum/metabolism , Protein Binding
14.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39000048

ABSTRACT

Bisphenols are dangerous endocrine disruptors that pollute the environment. Due to their chemical properties, they are globally used to produce plastics. Structural similarities to oestrogen allow bisphenols to bind to oestrogen receptors and affect internal body systems. Most commonly used in the plastic industry is bisphenol A (BPA), which also has negative effects on the nervous, immune, endocrine, and cardiovascular systems. A popular analogue of BPA-bisphenol S (BPS) also seems to have harmful effects similar to BPA on living organisms. Therefore, with the use of double immunofluorescence labelling, this study aimed to compare the effect of BPA and BPS on the enteric nervous system (ENS) in mouse jejunum. The study showed that both studied toxins impact the number of nerve cells immunoreactive to substance P (SP), galanin (GAL), vasoactive intestinal polypeptide (VIP), the neuronal isoform of nitric oxide synthase (nNOS), and vesicular acetylcholine transporter (VAChT). The observed changes were similar in the case of both tested bisphenols. However, the influence of BPA showed stronger changes in neurochemical coding. The results also showed that long-term exposure to BPS significantly affects the ENS.


Subject(s)
Benzhydryl Compounds , Enteric Nervous System , Jejunum , Phenols , Sulfones , Animals , Phenols/toxicity , Benzhydryl Compounds/toxicity , Mice , Jejunum/drug effects , Jejunum/metabolism , Enteric Nervous System/drug effects , Enteric Nervous System/metabolism , Sulfones/pharmacology , Sulfones/toxicity , Substance P/metabolism , Vasoactive Intestinal Peptide/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism , Male , Galanin/metabolism , Endocrine Disruptors/toxicity , Endocrine Disruptors/pharmacology , Nitric Oxide Synthase Type I/metabolism
15.
Nutrients ; 16(14)2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39064711

ABSTRACT

Plastics are present in almost every aspect of our lives. Polyethylene terephthalate (PET) is commonly used in the food industry. Microparticles can contaminate food and drinks, posing a threat to consumers. The presented study aims to determine the effect of microparticles of PET on the population of neurons positive for selected neurotransmitters in the enteric nervous system of the jejunum and histological structure. An amount of 15 pigs were divided into three groups (control, receiving 0.1 g, and 1 g/day/animal orally). After 28 days, fragments of the jejunum were collected for immunofluorescence and histological examination. The obtained results show that histological changes (injury of the apical parts of the villi, accumulations of cellular debris and mucus, eosinophil infiltration, and hyperaemia) were more pronounced in pigs receiving a higher dose of microparticles. The effect on neuronal nitric oxide synthase-, and substance P-positive neurons, depends on the examined plexus and the dose of microparticles. An increase in the percentage of galanin-positive neurons and a decrease in cocaine and amphetamine-regulated transcript-, vesicular acetylcholine transporter-, and vasoactive intestinal peptide-positive neurons do not show such relationships. The present study shows that microparticles can potentially have neurotoxic and pro-inflammatory effects, but there is a need for further research to determine the mechanism of this process and possible further effects.


Subject(s)
Jejunum , Microplastics , Neurons , Animals , Jejunum/drug effects , Jejunum/metabolism , Swine , Microplastics/toxicity , Neurons/drug effects , Neurons/metabolism , Enteric Nervous System/drug effects , Enteric Nervous System/metabolism , Substance P/metabolism , Vasoactive Intestinal Peptide/metabolism , Polyethylene Terephthalates , Nitric Oxide Synthase Type I/metabolism , Galanin/metabolism , Neuronal Plasticity/drug effects , Administration, Oral , Neurotransmitter Agents/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism , Male , Nerve Tissue Proteins
16.
J Virol ; 98(8): e0103924, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39012142

ABSTRACT

In maintaining organismal homeostasis, gut immunity plays a crucial role. The coordination between the microbiota and the immune system through bidirectional interactions regulates the impact of microorganisms on the host. Our research focused on understanding the relationships between substantial changes in jejunal intestinal flora and metabolites and intestinal immunity during porcine epidemic diarrhea virus (PEDV) infection in piglets. We discovered that Lactobacillus rhamnosus GG (LGG) could effectively prevent PEDV infection in piglets. Further investigation revealed that LGG metabolites interact with type 3 innate lymphoid cells (ILC3s) in the jejunum of piglets through the aryl hydrocarbon receptor (AhR). This interaction promotes the activation of ILC3s and the production of interleukin-22 (IL-22). Subsequently, IL-22 facilitates the proliferation of IPEC-J2 cells and activates the STAT3 signaling pathway, thereby preventing PEDV infection. Moreover, the AhR receptor influences various cell types within organoids, including intestinal stem cells (ISCs), Paneth cells, and enterocytes, to promote their growth and development, suggesting that AhR has a broad impact on intestinal health. In conclusion, our study demonstrated the ability of LGG to modulate intestinal immunity and effectively prevent PEDV infection in piglets. These findings highlight the potential application of LGG as a preventive measure against viral infections in livestock.IMPORTANCEWe observed high expression of the AhR receptor on pig and human ILC3s, although its expression was negligible in mouse ILC3s. ILC3s are closely related to the gut microbiota, particularly the secretion of IL-22 stimulated by microbial signals, which plays a crucial regulatory role in intestinal immunity. In our study, we found that metabolites produced by beneficial gut bacteria interact with ILC3s through AhR, thereby maintaining intestinal immune homeostasis in pigs. Moreover, LGG feeding can enhance the activation of ILC3s and promote IL-22 secretion in the intestines of piglets, ultimately preventing PEDV infection.


Subject(s)
Coronavirus Infections , Immunity, Innate , Interleukin-22 , Interleukins , Lymphocytes , Porcine epidemic diarrhea virus , Receptors, Aryl Hydrocarbon , Animals , Receptors, Aryl Hydrocarbon/metabolism , Swine , Interleukins/metabolism , Porcine epidemic diarrhea virus/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Infections/metabolism , Gastrointestinal Microbiome/immunology , Swine Diseases/immunology , Swine Diseases/virology , Swine Diseases/prevention & control , Swine Diseases/microbiology , Jejunum/immunology , Jejunum/metabolism , Signal Transduction , Ligands , Intestines/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism
17.
J Virol ; 98(7): e0202023, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38884472

ABSTRACT

Human noroviruses (HuNoVs) are a diverse group of RNA viruses that cause endemic and pandemic acute viral gastroenteritis. Previously, we reported that many HuNoV strains require bile or bile acid (BA) to infect human jejunal intestinal enteroid cultures. BA was not essential for the replication of a pandemic-causing GII.4 HuNoV strain. We found the hydrophobic BA glycochenodeoxycholic acid (GCDCA) promotes the replication of the BA-dependent strain GII.3 in jejunal enteroids. Furthermore, we found that inhibition of the G-protein-coupled BA receptor, sphingosine-1-phosphate receptor 2 (S1PR2), by JTE-013, reduced GII.3 infection dose-dependently and inhibited GII.3 cellular uptake in enteroids. Herein, we sought to determine whether S1PR2 is required for other BA-dependent HuNoV strains, the BA-independent GII.4, and whether S1PR2 is required for BA-dependent HuNoV infection in HIEs from other small intestinal segments. We found a second S1PR2 inhibitor, GLPG2938, reduces GII.3 infection dose-dependently, and an S1PR2 agonist (CYM-5520) enhances GII.3 replication in the absence of GCDCA. GII.3 replication also is abrogated in the presence of JTE-013 and CYM-5520. JTE-013 inhibition of S1PR2 in jejunal HIEs reduces GI.1, GII.3, and GII.17 (BA-dependent) but not GII.4 Sydney (BA-independent) infection, providing additional evidence of strain-specific differences in HuNoV infection. Finally, GII.3 infection of duodenal, jejunal, and ileal lines derived from the same individual is reduced with S1PR2 inhibition, indicating a common mechanism of BA-dependent infection among multiple segments of the small intestine. Our results support a model where BA-dependent HuNoVs exploit BA effects on S1PR2 to infect the entire small intestine.IMPORTANCEHuman noroviruses (HuNoVs) are important viral human pathogens that cause both outbreaks and sporadic gastroenteritis. These viruses are diverse, and many strains are capable of infecting humans. Our previous studies have identified strain-specific requirements for hydrophobic bile acids (BAs) to infect intestinal epithelial cells. Moreover, we identified a BA receptor, sphingosine-1-phosphate receptor 2 (S1PR2), required for infection by a BA-dependent strain. To better understand how various HuNoV strains enter and infect the small intestine and the role of S1PR2 in HuNoV infection, we evaluated infection by additional HuNoV strains using an expanded repertoire of intestinal enteroid cell lines. We found that multiple BA-dependent strains, but not a BA-independent strain, all require S1PR2 for infection. In addition, BA-dependent infection requires S1PR2 in multiple segments of the small intestine. Together, these results indicate that S1PR2 has value as a potential therapeutic target for BA-dependent HuNoV infection.


Subject(s)
Bile Acids and Salts , Norovirus , Sphingosine-1-Phosphate Receptors , Virus Replication , Humans , Norovirus/drug effects , Norovirus/physiology , Norovirus/genetics , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Virus Replication/drug effects , Bile Acids and Salts/pharmacology , Bile Acids and Salts/metabolism , Caliciviridae Infections/virology , Caliciviridae Infections/metabolism , Pyridines/pharmacology , Gastroenteritis/virology , Jejunum/virology , Jejunum/metabolism , Organoids/virology , Organoids/metabolism , Pyrazoles
18.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38928261

ABSTRACT

Consumption of a high-fat diet (HFD) has been suggested as a contributing factor behind increased intestinal permeability in obesity, leading to increased plasma levels of microbial endotoxins and, thereby, increased systemic inflammation. We and others have shown that HFD can induce jejunal expression of the ketogenic rate-limiting enzyme mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase (HMGCS). HMGCS is activated via the free fatty acid binding nuclear receptor PPAR-α, and it is a key enzyme in ketone body synthesis that was earlier believed to be expressed exclusively in the liver. The function of intestinal ketogenesis is unknown but has been described in suckling rats and mice pups, possibly in order to allow large molecules, such as immunoglobulins, to pass over the intestinal barrier. Therefore, we hypothesized that ketone bodies could regulate intestinal barrier function, e.g., via regulation of tight junction proteins. The primary aim was to compare the effects of HFD that can induce intestinal ketogenesis to an equicaloric carbohydrate diet on inflammatory responses, nutrition sensing, and intestinal permeability in human jejunal mucosa. Fifteen healthy volunteers receiving a 2-week HFD diet compared to a high-carbohydrate diet were compared. Blood samples and mixed meal tests were performed at the end of each dietary period to examine inflammation markers and postprandial endotoxemia. Jejunal biopsies were assessed for protein expression using Western blotting, immunohistochemistry, and morphometric characteristics of tight junctions by electron microscopy. Functional analyses of permeability and ketogenesis were performed in Caco-2 cells, mice, and human enteroids. Ussing chambers were used to analyze permeability. CRP and ALP values were within normal ranges and postprandial endotoxemia levels were low and did not differ between the two diets. The PPARα receptor was ketone body-dependently reduced after HFD. None of the tight junction proteins studied, nor the basal electrical parameters, were different between the two diets. However, the ketone body inhibitor hymeglusin increased resistance in mucosal biopsies. In addition, the tight junction protein claudin-3 was increased by ketone inhibition in human enteroids. The ketone body ß-Hydroxybutyrate (ßHB) did not, however, change the mucosal transition of the large-size molecular FD4-probe or LPS in Caco-2 and mouse experiments. We found that PPARα expression was inhibited by the ketone body ßHB. As PPARα regulates HMGCS expression, the ketone bodies thus exert negative feedback signaling on their own production. Furthermore, ketone bodies were involved in the regulation of permeability on intestinal mucosal cells in vitro and ex vivo. We were not, however, able to reproduce these effects on intestinal permeability in vivo in humans when comparing two weeks of high-fat with high-carbohydrate diet in healthy volunteers. Further, neither the expression of inflammation markers nor the aggregate tight junction proteins were changed. Thus, it seems that not only HFD but also other factors are needed to permit increased intestinal permeability in vivo. This indicates that the healthy gut can adapt to extremes of macro-nutrients and increased levels of intestinally produced ketone bodies, at least during a shorter dietary challenge.


Subject(s)
Diet, High-Fat , Intestinal Mucosa , Jejunum , Ketone Bodies , Permeability , Humans , Male , Intestinal Mucosa/metabolism , Diet, High-Fat/adverse effects , Ketone Bodies/metabolism , Adult , Jejunum/metabolism , Hydroxymethylglutaryl-CoA Synthase/metabolism , Hydroxymethylglutaryl-CoA Synthase/genetics , Female , Animals , Mice , Claudin-3/metabolism
19.
World J Gastroenterol ; 30(21): 2777-2792, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38899329

ABSTRACT

BACKGROUND: Obesity is associated with a significantly increased risk for chronic diarrhea, which has been proposed as Linghu's obesity-diarrhea syndrome (ODS); however, its molecular mechanisms are largely unknown. AIM: To reveal the transcriptomic changes in the jejunum involved in ODS. METHODS: In a cohort of 6 ODS patients (JOD group), 6 obese people without diarrhea (JO group), and 6 healthy controls (JC group), high-throughput sequencing and bioinformatics analyses were performed to identify jejunal mucosal mRNA expression alterations and dysfunctional biological processes. In another cohort of 16 ODS patients (SOD group), 16 obese people without diarrhea (SO group), and 16 healthy controls (SC group), serum diamine oxidase (DAO) and D-lactate (D-LA) concentrations were detected to assess changes in intestinal barrier function. RESULTS: The gene expression profiles of jejunal mucosa in the JO and JC groups were similar, with only 1 differentially expressed gene (DEG). The gene expression profile of the JOD group was significantly changed, with 411 DEGs compared with the JO group and 211 DEGs compared with the JC group, 129 of which overlapped. The enrichment analysis of these DEGs showed that the biological processes such as digestion, absorption, and transport of nutrients (especially lipids) tended to be up-regulated in the JOD group, while the biological processes such as rRNA processing, mitochondrial translation, antimicrobial humoral response, DNA replication, and DNA repair tended to be down-regulated in the JOD group. Eight DEGs (CDT1, NHP2, EXOSC5, EPN3, NME1, REG3A, PLA2G2A, and PRSS2) may play a key regulatory role in the pathological process of ODS, and their expression levels were significantly decreased in ODS patients (P < 0.001). In the second cohort, compared with healthy controls, the levels of serum intestinal barrier function markers (DAO and D-LA) were significantly increased in all obese individuals (P < 0.01), but were higher in the SOD group than in the SO group (P < 0.001). CONCLUSION: Compared with healthy controls and obese individuals without diarrhea, patients with Linghu's ODS had extensive transcriptomic changes in the jejunal mucosa, likely affecting intestinal barrier function and thus contributing to the obesity and chronic diarrhea phenotypes.


Subject(s)
Diarrhea , Gene Expression Profiling , Intestinal Mucosa , Jejunum , Obesity , Transcriptome , Humans , Jejunum/metabolism , Male , Pilot Projects , Female , Diarrhea/genetics , Diarrhea/etiology , Diarrhea/metabolism , Adult , Intestinal Mucosa/metabolism , Obesity/genetics , Obesity/complications , Middle Aged , Gene Expression Profiling/methods , Case-Control Studies , Syndrome , Amine Oxidase (Copper-Containing)/genetics , Amine Oxidase (Copper-Containing)/blood , Amine Oxidase (Copper-Containing)/metabolism , Computational Biology , Lactic Acid/blood , Lactic Acid/metabolism , Chronic Disease
20.
Int J Pharm ; 661: 124353, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38909926

ABSTRACT

Labrafac™ MC60 (glycerol monocaprylocaprate) is a lipid-based excipient used in oral formulations as a solubiliser. Due to the high proportions of established permeability enhancers, caprylate (C8) and caprate (C10), in Labrafac™ MC60, we hypothesised that it might behave as an intestinal permeation enhancer. We therefore evaluated this using two paracellular markers (ex vivo) and insulin (in vivo) as model molecules. Ex vivo studies were conducted in isolated muscle-stripped rat colonic mucosae mounted in Ussing chambers. Apical addition of Labrafac™ MC60 (8, 12, and 16 mg/ml) enhanced the apparent permeability coefficients (Papp) of [14C] mannitol and FITC-dextran 4 kDa (FD4) across colonic mucosae. Similar effects were observed in isolated jejunal mucosae, but at higher concentrations (40 mg/ml). The enhancing capacity of Labrafac™ MC60 was transient due to reversibility of reductions in transepithelial electrical resistance (TEER) upon wash-out and effects on fluxes were molecular weight-dependent (MW) as suggested by fluxes of a set of high MW FITC-dextrans. The permeability enhancing effects of Labrafac™ MC60 ex vivo were maintained in the presence of simulated intestinal fluids, FaSSIF and FaSSCoF, in both jejunal and colonic mucosae, respectively. Following intra-intestinal regional instillations to rats, the relative bioavailability of 50 IU/kg insulin ad-mixed with Labrafac™ MC60 was 5 % in jejunum (40 mg/ml) and 6 % in colon (8 mg/ml). When Labrafac™ MC60 was combined with PEG-60 hydrogenated castor oil (1 % v/v), this further increased the bioavailability of insulin to 8 % in jejunum. Absorption enhancement was also maintained in the presence of FaSSIF in jejunal instillations. Histology after 120 min exposure to Labrafac™ MC60 in vivo for both jejunum and colon was similar to untreated control. Labrafac™ MC60 therefore acts as a non-damaging intestinal permeation enhancer for macromolecules and can be considered as another excipient in screening programmes to develop orally administered macromolecules.


Subject(s)
Dextrans , Excipients , Fluorescein-5-isothiocyanate , Glycerides , Intestinal Absorption , Intestinal Mucosa , Permeability , Animals , Male , Intestinal Absorption/drug effects , Dextrans/pharmacokinetics , Dextrans/administration & dosage , Excipients/chemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Glycerides/chemistry , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/pharmacokinetics , Fluorescein-5-isothiocyanate/administration & dosage , Insulin , Rats , Mannitol , Rats, Wistar , Colon/metabolism , Colon/drug effects , Jejunum/metabolism , Jejunum/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL