Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 497
Filter
1.
Nat Commun ; 15(1): 4764, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834561

ABSTRACT

Bacteriophage are sophisticated cellular parasites that can not only parasitize bacteria but are increasingly recognized for their direct interactions with mammalian hosts. Phage adherence to mucus is known to mediate enhanced antimicrobial effects in vitro. However, little is known about the therapeutic efficacy of mucus-adherent phages in vivo. Here, using a combination of in vitro gastrointestinal cell lines, a gut-on-a-chip microfluidic model, and an in vivo murine gut model, we demonstrated that a E. coli phage, øPNJ-6, provided enhanced gastrointestinal persistence and antimicrobial effects. øPNJ-6 bound fucose residues, of the gut secreted glycoprotein MUC2, through domain 1 of its Hoc protein, which led to increased intestinal mucus production that was suggestive of a positive feedback loop mediated by the mucus-adherent phage. These findings extend the Bacteriophage Adherence to Mucus model into phage therapy, demonstrating that øPNJ-6 displays enhanced persistence within the murine gut, leading to targeted depletion of intestinal pathogenic bacteria.


Subject(s)
Escherichia coli Infections , Escherichia coli , Intestinal Mucosa , Mucin-2 , Animals , Escherichia coli/virology , Mice , Intestinal Mucosa/microbiology , Intestinal Mucosa/virology , Mucin-2/metabolism , Humans , Escherichia coli Infections/microbiology , Escherichia coli Infections/therapy , Phage Therapy/methods , Bacterial Adhesion , Female , Mucus/metabolism , Mucus/virology , Coliphages/physiology , Fucose/metabolism , Mice, Inbred C57BL
2.
Food Res Int ; 187: 114343, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763636

ABSTRACT

Human breast milk promotes maturation of the infant gastrointestinal barrier, including the promotion of mucus production. In the quest to produce next generation infant milk formula (IMF), we have produced IMF by membrane filtration (MEM-IMF). With a higher quantity of native whey protein, MEM-IMF more closely mimics human breast milk than IMF produced using conventional heat treatment (HT-IMF). After a 4-week dietary intervention in young pigs, animals fed a MEM-IMF diet had a higher number of goblet cells, acidic mucus and mucin-2 in the jejunum compared to pigs fed HT-IMF (P < 0.05). In the duodenum, MEM-IMF fed pigs had increased trypsin activity in the gut lumen, increased mRNA transcript levels of claudin 1 in the mucosal scrapings and increased lactase activity in brush border membrane vesicles than those pigs fed HT-IMF (P < 0.05). In conclusion, MEM-IMF is superior to HT-IMF in the promotion of mucus production in the young gut.


Subject(s)
Filtration , Infant Formula , Mucus , Animals , Infant Formula/chemistry , Mucus/metabolism , Swine , Whey Proteins/metabolism , Intestine, Small/metabolism , Trypsin/metabolism , Humans , Goblet Cells/metabolism , Claudin-1/metabolism , Claudin-1/genetics , Lactase/metabolism , Lactase/genetics , Mucin-2/metabolism , Mucin-2/genetics , Intestinal Mucosa/metabolism , Duodenum/metabolism , Jejunum/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , Milk Proteins/metabolism , Milk Proteins/analysis
3.
Int J Mol Sci ; 25(10)2024 May 07.
Article in English | MEDLINE | ID: mdl-38791132

ABSTRACT

Inflammatory bowel disease (IBD) is a multifactorial disease involving the interaction of the gut microbiota, genes, host immunity, and environmental factors. Dysbiosis in IBD is associated with pathobiont proliferation, so targeted antibiotic therapy is a rational strategy. When restoring the microbiota with probiotics, it is necessary to take into account the mutual influence of co-cultivated microorganisms, as the microbiota is a dynamic community of species that mediates homeostasis and physiological processes in the intestine. The aim of our study was to investigate the recovery efficacy of two potential probiotic bacteria, L. johnsonii and E. faecalis, in Muc2-/- mice with impaired mucosal layer. Two approaches were used to determine the efficacy of probiotic supplementation in mice with dysbiosis caused by mucin-2 deficiency: bacterial seeding on selective media and real-time PCR analysis. The recovery time and the type of probiotic bacteria relocated affected only the number of E. faecalis. A significant positive correlation was found between colony-forming unit (CFU) and the amount of E. faecalis DNA in the group that was replanted with probiotic E. faecalis. As for L. johnsonii, it could be restored to its original level even without any additional bacteria supplementation after two weeks. Interestingly, the treatment of mice with L. johnsonii caused a decrease in the amount of E. faecalis. Furthermore, either L. johnsonii or E. faecalis treatment eliminated protozoan overgrowth caused by antibiotic administration.


Subject(s)
Anti-Bacterial Agents , Dysbiosis , Enterococcus faecalis , Lactobacillus johnsonii , Probiotics , Animals , Enterococcus faecalis/drug effects , Mice , Anti-Bacterial Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Disease Models, Animal , Mucin-2/metabolism , Mucin-2/genetics , Inflammatory Bowel Diseases/microbiology , Mice, Knockout
4.
Cell Rep ; 43(5): 114207, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38733585

ABSTRACT

The MUC2 mucin protects the colonic epithelium by a two-layered mucus with an inner attached bacteria-free layer and an outer layer harboring commensal bacteria. CysD domains are 100 amino-acid-long sequences containing 10 cysteines that separate highly O-glycosylated proline, threonine, serine (PTS) regions in mucins. The structure of the second CysD, CysD2, of MUC2 is now solved by nuclear magnetic resonance. CysD2 shows a stable stalk region predicted to be partly covered by adjacent O-glycans attached to neighboring PTS sequences, whereas the CysD2 tip with three flexible loops is suggested to be well exposed. It shows transient dimer interactions at acidic pH, weakened at physiological pH. This transient interaction can be stabilized in vitro and in vivo by transglutaminase 3-catalyzed isopeptide bonds, preferring a specific glutamine residue on one flexible loop. This covalent dimer is modeled suggesting that CysD domains act as connecting hubs for covalent stabilization of mucins to form a protective mucus.


Subject(s)
Mucin-2 , Protein Domains , Transglutaminases , Mucin-2/metabolism , Mucin-2/chemistry , Humans , Transglutaminases/metabolism , Transglutaminases/chemistry , Models, Molecular , Cysteine/metabolism , Cysteine/chemistry , Amino Acid Sequence , Protein Multimerization , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/metabolism
5.
J Agric Food Chem ; 72(19): 10923-10935, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38691832

ABSTRACT

This study aimed to explore the ameliorative effects and potential mechanisms of Huangshan Umbilicaria esculenta polysaccharide (UEP) in dextran sulfate sodium-induced acute ulcerative colitis (UC) and UC secondary liver injury (SLI). Results showed that UEP could ameliorate both colon and liver pathologic injuries, upregulate mouse intestinal tight junction proteins (TJs) and MUC2 expression, and reduce LPS exposure, thereby attenuating the effects of the gut-liver axis. Importantly, UEP significantly downregulated the secretion levels of TNF-α, IL-1ß, and IL-6 through inhibition of the NF-κB pathway and activated the Nrf2 signaling pathway to increase the expression levels of SOD and GSH-Px. In vitro, UEP inhibited the LPS-induced phosphorylation of NF-κB P65 and promoted nuclear translocation of Nrf2 in RAW264.7 cells. These results revealed that UEP ameliorated UC and SLI through NF-κB and Nrf2-mediated inflammation and oxidative stress. The study first investigated the anticolitis effect of UEP, suggesting its potential for the treatment of colitis and colitis-associated liver disease.


Subject(s)
Colitis , Dextran Sulfate , NF-E2-Related Factor 2 , NF-kappa B , Polysaccharides , Animals , Mice , Polysaccharides/pharmacology , Polysaccharides/chemistry , Polysaccharides/administration & dosage , Dextran Sulfate/adverse effects , Male , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Humans , Colitis/drug therapy , Colitis/chemically induced , Colitis/metabolism , RAW 264.7 Cells , NF-kappa B/metabolism , NF-kappa B/genetics , Mice, Inbred C57BL , Protective Agents/pharmacology , Protective Agents/administration & dosage , Protective Agents/chemistry , Liver/drug effects , Liver/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/immunology , Oxidative Stress/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-6/immunology , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/chemically induced , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/metabolism , Mucin-2/genetics , Mucin-2/metabolism
6.
Ecotoxicol Environ Saf ; 279: 116458, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38759536

ABSTRACT

Heavy metals interact with each other in a coexisting manner to produce complex combined toxicity to organisms. At present, the toxic effects of chronic co-exposure to heavy metals hexavalent chromium [Cr(VI)] and divalent nickel [Ni(II)] on organisms are seldom studied and the related mechanisms are poorly understood. In this study, we explored the mechanism of the colon injury in mice caused by chronic exposure to Cr or/and Ni. The results showed that, compared with the control group, Cr or/and Ni chronic exposure affected the body weight of mice, and led to infiltration of inflammatory cells in the colon, decreased the number of goblet cells, fusion of intracellular mucus particles and damaged cell structure of intestinal epithelial. In the Cr or/and Ni exposure group, the activity of nitric oxide synthase (iNOS) increased, the expression levels of MUC2 were significantly down-regulated, and those of ZO-1 and Occludin were significantly up-regulated. Interestingly, factorial analysis revealed an interaction between Cr and Ni, which was manifested as antagonistic effects on iNOS activity, ZO-1 and MUC2 mRNA expression levels. Transcriptome sequencing further revealed that the expression of genes-related to inflammation, intestinal mucus and tight junctions changed obviously. Moreover, the relative contents of Cr(VI) and Ni(II) in the Cr, Ni and Cr+Ni groups all changed with in-vitro gastrointestinal (IVG)digestion, especially in the Cr+Ni group. Our results indicated that the chronic exposure to Cr or/and Ni can lead to damage to the mice colon, and the relative content changes of Cr(VI) and Ni(II) might be the main reason for the antagonistic effect of Cr+Ni exposure on the colon damage.


Subject(s)
Chromium , Colon , Mucin-2 , Nickel , Animals , Chromium/toxicity , Nickel/toxicity , Mice , Colon/drug effects , Colon/pathology , Mucin-2/genetics , Mucin-2/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type II/genetics , Gene Expression Profiling , Male , Digestion/drug effects , Zonula Occludens-1 Protein/metabolism , Zonula Occludens-1 Protein/genetics , Transcriptome/drug effects , Occludin/metabolism , Occludin/genetics , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology
7.
Nutrients ; 16(8)2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38674791

ABSTRACT

Sleep deprivation (SD) leads to impaired intestinal barrier function and intestinal flora disorder, especially a reduction in the abundance of the next generation of probiotic Faecalibacterium prausnitzii (F. prausnitzii). However, it remains largely unclear whether F. prausnitzii can ameliorate SD-induced intestinal barrier damage. A 72 h SD mouse model was used in this research, with or without the addition of F. prausnitzii. The findings indicated that pre-colonization with F. prausnitzii could protect against tissue damage from SD, enhance goblet cell count and MUC2 levels in the colon, boost tight-junction protein expression, decrease macrophage infiltration, suppress pro-inflammatory cytokine expression, and reduce apoptosis. We found that the presence of F. prausnitzii helped to balance the gut microbiota in SD mice by reducing harmful bacteria like Klebsiella and Staphylococcus, while increasing beneficial bacteria such as Akkermansia. Ion chromatography analysis revealed that F. prausnitzii pretreatment increased the fecal butyrate level in SD mice. Overall, these results suggested that incorporating F. prausnitzii could help reduce gut damage caused by SD, potentially by enhancing the intestinal barrier and balancing gut microflora. This provides a foundation for utilizing probiotics to protect against intestinal illnesses.


Subject(s)
Dysbiosis , Faecalibacterium prausnitzii , Gastrointestinal Microbiome , Intestinal Mucosa , Probiotics , Sleep Deprivation , Animals , Sleep Deprivation/complications , Mice , Probiotics/pharmacology , Probiotics/administration & dosage , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Feces/microbiology , Mice, Inbred C57BL , Dietary Supplements , Disease Models, Animal , Mucin-2/metabolism , Butyrates/metabolism , Colon/microbiology , Colon/metabolism
8.
Nutrients ; 16(8)2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38674854

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) serotype O157:H7 is a commonly encountered foodborne pathogen that can cause hemorrhagic enteritis and lead to hemolytic uremic syndrome (HUS) in severe cases. Bifidobacterium is a beneficial bacterium that naturally exists in the human gut and plays a vital role in maintaining a healthy balance in the gut microbiota. This study investigated the protective effects of B. longum K5 in a mouse model of EHEC O157:H7 infection. The results indicated that pretreatment with B. longum K5 mitigated the clinical symptoms of EHEC O157:H7 infection and attenuated the increase in myeloperoxidase (MPO) activity in the colon of the mice. In comparison to the model group, elevated serum D-lactic acid concentrations and diamine oxidase (DAO) levels were prevented in the K5-EHEC group of mice. The reduced mRNA expression of tight junction proteins (ZO-1, Occludin, and Claudin-1) and mucin MUC2, as well as the elevated expression of virulence factors Stx1A and Stx2A, was alleviated in the colon of both the K5-PBS and K5-EHEC groups. Additionally, the increase in the inflammatory cytokine levels of TNF-α and IL-1ß was inhibited and the production of IL-4 and IL-10 was promoted in the K5-EHEC group compared with the model group. B. longum K5 significantly prevented the reduction in the abundance and diversity of mouse gut microorganisms induced by EHEC O157:H7 infection, including blocking the decrease in the relative abundance of Roseburia, Lactobacillus, and Oscillibacter. Meanwhile, the intervention with B. longum K5 promoted the production of acetic acid and butyric acid in the gut. This study provides insights into the use of B. longum K5 for developing probiotic formulations to prevent intestinal diseases caused by pathogenic bacterial infections.


Subject(s)
Bifidobacterium longum , Colon , Escherichia coli Infections , Escherichia coli O157 , Gastrointestinal Microbiome , Probiotics , Animals , Mice , Probiotics/pharmacology , Escherichia coli Infections/prevention & control , Escherichia coli Infections/microbiology , Colon/microbiology , Colon/metabolism , Disease Models, Animal , Mucin-2/metabolism , Cytokines/metabolism , Peroxidase/metabolism , Amine Oxidase (Copper-Containing)/metabolism
9.
Food Funct ; 15(9): 5118-5131, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38682277

ABSTRACT

This study investigated the impact of in vivo available colon-mango (poly)phenols on stress-induced impairment of intestinal barrier function. Caco-2/HT29-MTX cells were incubated with six extracts of ileal fluid collected pre- and 4-8 h post-mango consumption before being subjected to inflammatory stress. (Poly)phenols in ileal fluids were analysed by UHPLC-HR-MS. Epithelial barrier function was monitored by measurement of trans-epithelial electrical resistance (TEER) and the production of selected inflammatory markers (interleukin-8 (IL-8) and nitric oxide (NO)) and the major mucin of the mucosal layer (MUC2). Post-mango intake ileal fluids contained principally benzoic acids, hydroxybenzenes and galloyl derivatives. There was a high interindividual variability in the levels of these compounds, which was reflected by the degree of variability in the protective effects of individual ileal extracts on inflammatory changes in the treated cell cultures. The 24 h treatment with non-cytotoxic doses of extracts of 4-8 h post-mango intake ileal fluid significantly reduced the TEER decrease in monolayers treated with the inflammatory cytomix. This effect was not associated with changes in IL-8 expression and secretion or claudine-7 expression. The mango derived-ileal fluid extract (IFE) also mitigated cytomix-dependent nitrite secretion, as a proxy of NO production, and the MUC2 reduction observed upon the inflammatory challenge. These insights shed light on the potential protective effect of mango (poly)phenols on the intestinal barrier exposed to inflammatory conditions.


Subject(s)
Interleukin-8 , Intestinal Mucosa , Mangifera , Mucin-2 , Humans , Mangifera/chemistry , Caco-2 Cells , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Interleukin-8/metabolism , Mucin-2/metabolism , HT29 Cells , Polyphenols/pharmacology , Colon/drug effects , Colon/metabolism , Nitric Oxide/metabolism , Plant Extracts/pharmacology , Plant Extracts/chemistry , Inflammation/drug therapy , Intestinal Barrier Function
10.
Int J Biol Macromol ; 267(Pt 2): 131434, 2024 May.
Article in English | MEDLINE | ID: mdl-38614182

ABSTRACT

The gastrointestinal (GI) tract's mucus layer serves as a critical barrier and a mediator in drug nanoparticle delivery. The mucus layer's diverse molecular structures and spatial complexity complicates the mechanistic study of the diffusion dynamics of particulate materials. In response, we developed a bi-component coarse-grained mucus model, specifically tailored for the colorectal cancer environment, that contained the two most abundant glycoproteins in GI mucus: Muc2 and Muc5AC. This model demonstrated the effects of molecular composition and concentration on mucus pore size, a key determinant in the permeability of nanoparticles. Using this computational model, we investigated the diffusion rate of polyethylene glycol (PEG) coated nanoparticles, a widely used muco-penetrating nanoparticle. We validated our model with experimentally characterized mucus pore sizes and the diffusional coefficients of PEG-coated nanoparticles in the mucus collected from cultured human colorectal goblet cells. Machine learning fingerprints were then employed to provide a mechanistic understanding of nanoparticle diffusional behavior. We found that larger nanoparticles tended to be trapped in mucus over longer durations but exhibited more ballistic diffusion over shorter time spans. Through these discoveries, our model provides a promising platform to study pharmacokinetics in the GI mucus layer.


Subject(s)
Mucus , Nanoparticles , Polyethylene Glycols , Humans , Nanoparticles/chemistry , Diffusion , Polyethylene Glycols/chemistry , Mucus/metabolism , Mucus/chemistry , Mucin-2/metabolism , Mucin-2/chemistry , Mucin 5AC/metabolism , Mucin 5AC/chemistry , Intestinal Mucosa/metabolism , Gastrointestinal Tract/metabolism , Goblet Cells/metabolism , Models, Biological
11.
Phytomedicine ; 129: 155541, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38579640

ABSTRACT

BACKGROUND: Diarrheal irritable bowel syndrome (IBS-D), characterized primarily by the presence of diarrhea and abdominal pain, is a clinical manifestation resulting from a multitude of causative factors. Furthermore, Sishen Wan (SSW) has demonstrated efficacy in treating IBS-D. Nevertheless, its mechanism of action remains unclear. METHODS: A model of IBS-D was induced by a diet containing 45 % lactose and chronic unpredictable mild stress. Additionally, the impact of SSW was assessed by measuring body weight, visceral sensitivity, defecation parameters, intestinal transport velocity, intestinal neurotransmitter levels, immunohistochemistry, and transmission electron microscopy analysis. Immunofluorescent staining was used to detect the expression of Mucin 2 (MUC2) and Occludin in the colon. Western blotting was used to detect changes in proteins related to tight junction (TJ), autophagy, and endoplasmic reticulum (ER) stress in the colon. Finally, 16S rRNA amplicon sequencing was used to monitor the alteration of gut microbiota after SSW treatment. RESULTS: Our study revealed that SSW administration resulted in reduced visceral sensitivity, improved defecation parameters, decreased intestinal transport velocity, and reduced intestinal permeability in IBS-D mice. Furthermore, SSW promotes the secretion of colonic mucus by enhancing autophagy and inhibiting ER stress. SSW treatment caused remodeling of the gut microbiome by increasing the abundance of Blautia, Muribaculum and Ruminococcus torques group. CONCLUSION: SSW can improve intestinal barrier function by promoting autophagy and inhibiting ER stress, thus exerting a therapeutic effect on IBS-D.


Subject(s)
Diarrhea , Disease Models, Animal , Drugs, Chinese Herbal , Endoplasmic Reticulum Stress , Gastrointestinal Microbiome , Intestinal Mucosa , Irritable Bowel Syndrome , Irritable Bowel Syndrome/drug therapy , Animals , Endoplasmic Reticulum Stress/drug effects , Diarrhea/drug therapy , Drugs, Chinese Herbal/pharmacology , Mice , Gastrointestinal Microbiome/drug effects , Male , Intestinal Mucosa/drug effects , Mucin-2/metabolism , Colon/drug effects , Autophagy/drug effects , Permeability/drug effects , Occludin/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism , Mice, Inbred C57BL , Intestinal Barrier Function
12.
Chem Biol Interact ; 395: 111014, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38648921

ABSTRACT

There is an increasing appreciation that colonic barrier function is closely related to the development and progression of colitis. The mucus layer is a crucial component of the colonic barrier, responsible for preventing harmful bacteria from invading the intestinal epithelium and causing inflammation. Furthermore, a defective mucus barrier is also a significant characteristic of ulcerative colitis (UC). Biochanin A (BCA), an isoflavonoid, has garnered increasing interest due to its significant biological activities. However, the impact of BCA on UC has not been reported yet. In this study, we used a dextran sodium sulfate (DSS)-induced ulcerative colitis model and the Muc2 deficient (Muc2-/-) mice spontaneous colitis model to explore the mechanisms of BCA in the treatment of UC. Here, we verified that DSS-induced UC was observably attenuated and spontaneous colitis in Muc2-/- mice was relieved by BCA. Treatment with BCA improved colitis-related symptoms and reduced intestinal permeability by upregulating the levels of goblet cells and tight junction (TJ) proteins. In addition, we confirmed that BCA promotes autophagy through the AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR)/Unc-51-like kinase 1 (ULK1) pathway, thereby alleviating DSS-induced UC. In addition, the administration of BCA was able to reduce apoptosis and promote proliferation by suppressing Cleaved Caspase-3 (Cleaved Cas-3) expression, and increasing PCNA and Ki67 levels. Further research revealed that BCA treatment ameliorated spontaneous colitis and alleviated epithelial damage in Muc2-/- mice by restoring the intestinal barrier and promoting autophagy. Our results demonstrated that BCA alleviated UC by enhancing intestinal barrier function and promoting autophagy. These findings indicate that BCA may be a novel treatment alternative for UC.


Subject(s)
Colitis, Ulcerative , Colon , Dextran Sulfate , Genistein , Mucin-2 , Animals , Mucin-2/metabolism , Mucin-2/genetics , Dextran Sulfate/toxicity , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , Colitis, Ulcerative/metabolism , Genistein/pharmacology , Genistein/therapeutic use , Mice , Colon/pathology , Colon/drug effects , Colon/metabolism , Autophagy/drug effects , Mice, Inbred C57BL , Disease Models, Animal , Mice, Knockout , Apoptosis/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , AMP-Activated Protein Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism
13.
Sci Rep ; 14(1): 6954, 2024 03 23.
Article in English | MEDLINE | ID: mdl-38521809

ABSTRACT

Mucin protein glycosylation is important in determining biological properties of mucus gels, which form protective barriers at mucosal surfaces of the body such as the intestine. Ecological factors including: age, sex, and diet can change mucus barrier properties by modulating mucin glycosylation. However, as our understanding stems from controlled laboratory studies in house mice, the combined influence of ecological factors on mucin glycosylation in real-world contexts remains limited. In this study, we used histological staining with 'Alcian Blue, Periodic Acid, Schiff's' and 'High-Iron diamine' to assess the acidic nature of mucins stored within goblet cells of the intestine, in a wild mouse population (Mus musculus). Using statistical models, we identified sex as among the most influential ecological factors determining the acidity of intestinal mucin glycans in wild mice. Our data from wild mice and experiments using laboratory mice suggest estrogen signalling associates with an increase in the relative abundance of sialylated mucins. Thus, estrogen signalling may underpin sex differences observed in the colonic mucus of wild and laboratory mice. These findings highlight the significant influence of ecological parameters on mucosal barrier sites and the complementary role of wild populations in augmenting standard laboratory studies in the advancement of mucus biology.


Subject(s)
Colon , Mucins , Mice , Female , Male , Animals , Mucins/metabolism , Colon/pathology , Goblet Cells/metabolism , Intestines , Estrogens/metabolism , Mucin-2/metabolism , Intestinal Mucosa/metabolism
14.
Genomics ; 116(3): 110809, 2024 May.
Article in English | MEDLINE | ID: mdl-38492821

ABSTRACT

Colorectal cancer (CRC) is the third most common cancer and the prevalence rate of CRC is increasing in the China. In this study, whole-exome sequencing (WES) was performed on primary tissues of 47 CRC Chinese patients including 22 metastatic and 25 non-metastatic patients. By comparison with data from western colorectal cancer patients in the Cancer Genome Atlas (TCGA), we identified a number of genes that are more likely to be mutated in Chinese colorectal cancer patients, such as MUC12, MUC12, MUC2, MUC4, HYDIN and KMT2C. Interestingly, MUC family genes including MUC12, MUC2 and MUC4, have mutation rates of >20%, while the mutation frequency was extremely low in western colorectal cancer patients, which were <3% in TCGA and 0% in Memorial Sloan Kettering Cancer Center (MSKCC). We detected metastasis-specific mutated genes including TCF7L2, MST1L, HRNR and SMAD4, while MUC4, NEB, FLG and RFPL4A alteration is more prevalent in the non-metastasis group. Further analysis reveals mutation genes in metastasis group are more focus in the Wnt and Hippo signaling pathway. APC, SMAD4 and TCF7L2 accounted for the major genetic abnormalities in this pathway. In conclusion, this study identified the unique characteristics of gene mutations in Chinese patients with colorectal cancer, and is a valuable reference for personalized treatment in Chinese CRC patients.


Subject(s)
Colorectal Neoplasms , Exome Sequencing , Mutation , Neoplasm Metastasis , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Male , Female , Middle Aged , Aged , Adult , Hippo Signaling Pathway , Mucin-4/genetics , Mucin-4/metabolism , China , Mucin-2/genetics , Mucin-2/metabolism , Smad4 Protein/genetics , Smad4 Protein/metabolism , Filaggrin Proteins , Protein Serine-Threonine Kinases/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Wnt Signaling Pathway , Asian People/genetics , East Asian People , DNA-Binding Proteins
15.
Cell Biochem Funct ; 42(2): e3989, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38500386

ABSTRACT

Colorectal mucinous adenocarcinoma (MAC) is one of the most lethal histological types of colorectal cancer, and its mechanism of development is not well understood. In this study, we aimed to clarify the molecular characteristics of MAC via in silico analysis using The Cancer Genome Atlas database. The expression of genes on chromosome 20q (Chr20q) was negatively associated with the expression of MUC2, which is a key molecule that can be used to distinguish between MAC and nonmucinous adenocarcinoma (NMAC). This was consistent with a significant difference in copy number alteration of Chr20q between the two histological types. We further identified 475 differentially expressed genes (DEGs) between MAC and NMAC, and some of the Chr20q genes among the DEGs are considered to be pivotal genes used to define MAC. Both in vitro and in vivo analysis showed that simultaneous knockdown of POFUT1 and PLAGL2, both of which are located on Chr20q, promoted MUC2 expression. Moreover, these genes were highly expressed in NMAC but not in MAC according to the results of immunohistological studies using human samples. In conclusion, POFUT1 and PLAGL2 are considered to be important for defining MAC, and these genes are associated with MUC2 expression.


Subject(s)
Adenocarcinoma, Mucinous , Adenocarcinoma , Colorectal Neoplasms , Humans , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/pathology , Colorectal Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Mucin-2/genetics , Mucin-2/metabolism , RNA-Binding Proteins/genetics , Transcription Factors/genetics
16.
Vet Immunol Immunopathol ; 271: 110740, 2024 May.
Article in English | MEDLINE | ID: mdl-38537313

ABSTRACT

Intestinal mucus barrier disruption may occur with chronic inflammatory enteropathies. The lack of studies evaluating mucus health in dogs with chronic colitis arises from inherent challenges with assessment of the intestinal mucus layer. It is therefore unknown if reduced goblet cell (GBC) numbers and/or mucin 2 (MUC2) expression, which are responsible for mucus production and secretion, correlate with inflammation severity in dogs with granulomatous colitis (GC) or lymphocytic-plasmacytic colitis (LPC). It is undetermined if Ki-67 immunoreactivity, which has been evaluated in dogs with small intestinal inflammation, similarly correlates to histologic severity in GC and LPC. Study objectives included comparing Ki-67 immunoreactivity, GBC population and MUC2 expression in dogs with GC, LPC and non-inflamed colon; and exploring the use of ribonucleic acid (RNAscope®) in-situ hybridization (ISH) to evaluate MUC2 expression in canine colon. Formalin-fixed endoscopic colonic biopsies were obtained from 48 dogs over an eight-year period. A blinded pathologist reviewed all biopsies. Dogs were classified into the GC (n=19), LPC (n=19) or no colitis (NC) (n=10) group based on final histopathological diagnosis. Ki-67 immunohistochemistry, Alcian-Blue/PAS staining to highlight GBCs, and RNAscope® ISH using customized canine MUC2-targeted probes were performed. At least five microscopic fields per dog were selected to measure Ki-67 labelling index (KI67%), GBC staining percentage (GBC%) and MUC2 expression (MUC2%) using image analysis software. Spearman's correlation coefficients were used to determine associations between World Small Animal Veterinary Association histologic score (WHS) and measured variables. Linear regression models were used to compare relationships between WHS with KI67%, GBC%, and MUC2%; and between GBC% and MUC2%. Median WHS was highest in dogs with GC. Median KI67% normalised to WHS was highest in the NC group (6.69%; range, 1.70-23.60%). Median GBC% did not correlate with colonic inflammation overall. Median MUC2% normalised to WHS in the NC group (10.02%; range, 3.05-39.09%) was two- and three-fold higher than in the GC and LPC groups respectively. With increased colonic inflammation, despite minimal changes in GBC% overall, MUC2 expression markedly declined in the LPC group (-27.4%; 95%-CI, -49.8, 5.9%) and mildly declined in the GC and NC groups. Granulomatous colitis and LPC likely involve different pathways regulating MUC2 expression. Decreased MUC2 gene expression is observed in dogs with chronic colitis compared to dogs without colonic signs. Changes in MUC2 expression appear influenced by GBC activity rather than quantity in GC and LPC.


Subject(s)
Colitis , Dog Diseases , Goblet Cells , Ki-67 Antigen , Mucin-2 , Animals , Dogs , Mucin-2/genetics , Mucin-2/metabolism , Goblet Cells/pathology , Goblet Cells/metabolism , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Dog Diseases/metabolism , Dog Diseases/genetics , Dog Diseases/immunology , Colitis/veterinary , Colitis/pathology , Female , Male , Colon/pathology , Granuloma/veterinary , Granuloma/pathology , Immunohistochemistry/veterinary
17.
Int J Biol Macromol ; 266(Pt 1): 131232, 2024 May.
Article in English | MEDLINE | ID: mdl-38554896

ABSTRACT

Inflammatory bowel diseases (IBD) are chronic inflammatory conditions characterized by disruptions in the colonic mucus barrier and gut microbiota. In this study, a novel soluble polysaccharide obtained from Boletus aereus (BAP) through water extraction was examined for its structure. The protective effects of BAP on colitis were investigated using a DSS-induced mice model. BAP was found to promote the expression of intestinal mucosal and tight junction proteins, restore the compromised mucus barrier, and suppress the activation of inflammatory signaling. Moreover, BAP reshape the gut microbiota and had a positive impact on the composition of the gut microbiota by reducing inflammation-related microbes. Additionally, BAP decreased cytokine levels through the MANF-BATF2 signaling pathway. Correlation analysis revealed that MANF was negatively correlated with the DAI and the level of cytokines. Furthermore, the depletion of gut microbiota using antibiotic partially inhabited the effect of BAP on the activation of MANF and Muc2, indicating the role of gut microbiota in its protective effect against colitis. In conclusion, BAP had an obvious activation on MANF under gut inflammation. This provides new insights into the prospective use of BAP as a functional food to enhance intestinal health.


Subject(s)
Colitis , Dextran Sulfate , Gastrointestinal Microbiome , Mucin-2 , Signal Transduction , Animals , Gastrointestinal Microbiome/drug effects , Mucin-2/metabolism , Mucin-2/genetics , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Mice , Signal Transduction/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Disease Models, Animal , Polysaccharides/pharmacology , Polysaccharides/chemistry , Cytokines/metabolism , Basidiomycota/chemistry , Male , Fungal Polysaccharides/pharmacology , Fungal Polysaccharides/chemistry
18.
Am J Pathol ; 194(6): 975-988, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38423356

ABSTRACT

Radiation-induced enteritis, a significant concern in abdominal radiation therapy, is associated closely with gut microbiota dysbiosis. The mucus layer plays a pivotal role in preventing the translocation of commensal and pathogenic microbes. Although significant expression of REGγ in intestinal epithelial cells is well established, its role in modulating the mucus layer and gut microbiota remains unknown. The current study revealed notable changes in gut microorganisms and metabolites in irradiated mice lacking REGγ, as compared to wild-type mice. Concomitant with gut microbiota dysbiosis, REGγ deficiency facilitated the infiltration of neutrophils and macrophages, thereby exacerbating intestinal inflammation after irradiation. Furthermore, fluorescence in situ hybridization assays unveiled an augmented proximity of bacteria to intestinal epithelial cells in REGγ knockout mice after irradiation. Mechanistically, deficiency of REGγ led to diminished goblet cell populations and reduced expression of key goblet cell markers, Muc2 and Tff3, observed in both murine models, minigut organoid systems and human intestinal goblet cells, indicating the intrinsic role of REGγ within goblet cells. Interestingly, although administration of broad-spectrum antibiotics did not alter the goblet cell numbers or mucin 2 (MUC2) secretion, it effectively attenuated inflammation levels in the ileum of irradiated REGγ absent mice, bringing them down to the wild-type levels. Collectively, these findings highlight the contribution of REGγ in counteracting radiation-triggered microbial imbalances and cell-autonomous regulation of mucin secretion.


Subject(s)
Enteritis , Gastrointestinal Microbiome , Goblet Cells , Homeostasis , Mice, Knockout , Animals , Enteritis/microbiology , Enteritis/metabolism , Enteritis/pathology , Mice , Goblet Cells/pathology , Goblet Cells/metabolism , Humans , Pancreatitis-Associated Proteins/metabolism , Mucin-2/metabolism , Dysbiosis/microbiology , Dysbiosis/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Trefoil Factor-3/metabolism , Mice, Inbred C57BL , Radiation Injuries/metabolism , Radiation Injuries/microbiology , Radiation Injuries/pathology , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/microbiology
19.
J Biol Chem ; 300(3): 105675, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38272223

ABSTRACT

The O-glycoprotein Mucin-2 (MUC2) forms the protective colon mucus layer. While animal models have demonstrated the importance of Muc2, few studies have explored human MUC2 in similar depth. Recent studies have revealed that secreted MUC2 is bound to human feces. We hypothesized human fecal MUC2 (HF-MUC2) was accessible for purification and downstream structural and functional characterization. We tested this via histologic and quantitative imaging on human fecal sections; extraction from feces for proteomic and O-glycomic characterization; and functional studies via growth and metabolic assays in vitro. Quantitative imaging of solid fecal sections showed a continuous mucus layer of varying thickness along human fecal sections with barrier functions intact. Lectin profiling showed HF-MUC2 bound several lectins but was weak to absent for Ulex europaeus 1 (α1,2 fucose-binding) and Sambucus nigra agglutinin (α2,6 sialic acid-binding), and did not have obvious b1/b2 barrier layers. HF-MUC2 separated by electrophoresis showed high molecular weight glycoprotein bands (∼1-2 MDa). Proteomics and Western analysis confirmed the enrichment of MUC2 and potential MUC2-associated proteins in HF-MUC2 extracts. MUC2 O-glycomics revealed diverse fucosylation, moderate sialylation, and little sulfation versus porcine colonic MUC2 and murine fecal Muc2. O-glycans were functional and supported the growth of Bacteroides thetaiotaomicron (B. theta) and short-chain fatty acid (SCFA) production in vitro. MUC2 could be similarly analyzed from inflammatory bowel disease stools, which displayed an altered glycomic profile and differential growth and SCFA production by B. theta versus healthy samples. These studies describe a new non-invasive platform for human MUC2 characterization in health and disease.


Subject(s)
Colon , Feces , Proteomics , Animals , Humans , Mice , Colon/metabolism , Glycoproteins/metabolism , Intestinal Mucosa/metabolism , Mucin-2/genetics , Mucin-2/metabolism , Mucus/metabolism , Swine , Male , Mice, Inbred C57BL , Gastrointestinal Microbiome
20.
Microbiol Res ; 281: 127599, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38219635

ABSTRACT

The colonic mucus layer plays a critical role in maintaining the integrity of the colonic mucosal barrier, serving as the primary defense against colonic microorganisms. Predominantly composed of mucin 2 (MUC2), a glycosylation-rich protein, the mucus layer forms a gel-like coating that covers the colonic epithelium surface. This layer provides a habitat for intestinal microorganisms, which can utilize mucin glycans present in the mucus layer as a sustainable source of nutrients. Additionally, metabolites produced by the microbiota during the metabolism of mucus glycans have a profound impact on host health. Under normal conditions, the production and consumption of mucus maintain a dynamic balance. However, several studies have demonstrated that certain factors, such as dietary fiber deficiency, can enhance the metabolism of mucus glycans by gut bacteria, thereby disturbing this balance and weakening the mucus barrier function of the mucus layer. To better understand the occurrence and development of colon-related diseases, it is crucial to investigate the complex metabolic patterns of mucus glycosylation by intestinal microorganisms. Our objective was to comprehensively review these patterns in order to clarify the effects of mucus layer glycan metabolism by intestinal microorganisms on the host.


Subject(s)
Ants , Intestinal Mucosa , Animals , Intestinal Mucosa/metabolism , Mucus/metabolism , Mucin-2/metabolism , Mucins/metabolism , Colon/metabolism , Colon/microbiology , Polysaccharides/metabolism , Ants/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...