Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 745
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732126

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) is a critical public health concern due to its role in severe gastrointestinal illnesses in humans, including hemorrhagic colitis and the life-threatening hemolytic uremic syndrome. While highly pathogenic to humans, cattle, the main reservoir for EHEC, often remain asymptomatic carriers, complicating efforts to control its spread. Our study introduces a novel method to investigate EHEC using organoid-derived monolayers from adult bovine ileum and rectum. These polarized epithelial monolayers were exposed to EHEC for four hours, allowing us to perform comparative analyses between the ileal and rectal tissues. Our findings mirrored in vivo observations, showing a higher colonization rate in the rectum compared with the ileum (44.0% vs. 16.5%, p < 0.05). Both tissues exhibited an inflammatory response with increased expression levels of TNF-a (p < 0.05) and a more pronounced increase of IL-8 in the rectum (p < 0.01). Additionally, the impact of EHEC on the mucus barrier varied across these gastrointestinal regions. Innovative visualization techniques helped us study the ultrastructure of mucus, revealing a net-like mucin glycoprotein organization. While further cellular differentiation could enhance model accuracy, our research significantly deepens understanding of EHEC pathogenesis in cattle and informs strategies for the preventative measures and therapeutic interventions.


Subject(s)
Enterohemorrhagic Escherichia coli , Ileum , Organoids , Rectum , Animals , Cattle , Ileum/microbiology , Ileum/metabolism , Ileum/ultrastructure , Rectum/microbiology , Enterohemorrhagic Escherichia coli/pathogenicity , Organoids/metabolism , Organoids/microbiology , Mucus/metabolism , Escherichia coli Infections/microbiology , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure
2.
Gut ; 71(3): 487-496, 2022 03.
Article in English | MEDLINE | ID: mdl-33963042

ABSTRACT

OBJECTIVE: Although immunoglobulin A (IgA) is abundantly expressed in the gut and known to be an important component of mucosal barriers against luminal pathogens, its precise function remains unclear. Therefore, we tried to elucidate the effect of IgA on gut homeostasis maintenance and its mechanism. DESIGN: We generated various IgA mutant mouse lines using the CRISPR/Cas9 genome editing system. Then, we evaluated the effect on the small intestinal homeostasis, pathology, intestinal microbiota, cytokine production, and immune cell activation using intravital imaging. RESULTS: We obtained two lines, with one that contained a <50 base pair deletion in the cytoplasmic region of the IgA allele (IgA tail-mutant; IgAtm/tm) and the other that lacked the most constant region of the IgH α chain, which resulted in the deficiency of IgA production (IgA-/-). IgA-/- exhibited spontaneous inflammation in the ileum but not the other parts of the gastrointestinal tract. Associated with this, there were significantly increased lamina propria CD4+ T cells, elevated productions of IFN-γ and IL-17, increased ileal segmented filamentous bacteria and skewed intestinal microflora composition. Intravital imaging using Ca2+ biosensor showed that IgA-/- had elevated Ca2+ signalling in Peyer's patch B cells. On the other hand, IgAtm/tm seemed to be normal, suggesting that the IgA cytoplasmic tail is dispensable for the prevention of the intestinal disorder. CONCLUSION: IgA plays an important role in the mucosal homeostasis associated with the regulation of intestinal microbiota and protection against mucosal inflammation especially in the ileum.


Subject(s)
Ileitis/etiology , Ileum/pathology , Immunoglobulin A/physiology , Animals , B-Lymphocytes/physiology , Cytokines/metabolism , Disease Models, Animal , Female , Gastrointestinal Microbiome , Homeostasis , Ileitis/metabolism , Ileitis/pathology , Ileum/metabolism , Ileum/ultrastructure , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Intravital Microscopy , Male , Mice , Mice, Mutant Strains , T-Lymphocytes/physiology
3.
Cell Tissue Res ; 385(3): 697-711, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33961127

ABSTRACT

Several types of macrophages have been reported in the intestinal mucosa, but their histological localization remains ambiguous. Here, we obtained detailed information about ultrastructural and phenotypical diversity of macrophage-like cells (MLCs) in the rat ileal mucosa using immunofluorescent analysis and serial block-face scanning electron microscopy (SBF-SEM). The results revealed that the cells immunopositive for CD68, the pan-macrophage marker, included CD163-CD4+, CD163+CD4+, and CD163-CD4- cells in the lamina propria (LP) of the intestinal villus and around the crypt. CD68+CD4+CD163- cells seemed to be preferentially localized in the intestinal villus, whereas CD68+CD163+CD4+ cells were frequently localized around the crypt. SBF-SEM analysis identified three types of MLCs in the ileal mucosa, which were tentatively named types I-III MLC based on aspects of the 3D-ultrastructure, such as the localization, quantity of lysosomes, endoplasmic reticulum, and exoplasm. Type I and II MLCs were localized in the villous LP, while type III MLCs were localized around the crypt, although type II MLCs were a minor population. All three MLC types extended their cellular processes into the epithelium, with type I MLCs showing the greatest abundance of extended processes. Type I MLCs in the upper portion of the intestinal villus showed a higher level of attachment to intraepithelial lymphocytes (IELs) compared to type III MLCs around the crypt. These findings suggest that macrophages of the rat ileal mucosa differed by region along the longitudinal axis of the villous tip-crypt from the perspective of ultrastructure, cellular composition, localization, and interactions with IELs.


Subject(s)
Ileum/ultrastructure , Macrophages/ultrastructure , Animals , Rats , Rats, Wistar
5.
Nutrients ; 13(1)2020 Dec 25.
Article in English | MEDLINE | ID: mdl-33375592

ABSTRACT

Dietary fiber intake during pregnancy may improve offspring intestinal development. The aim of this study was to evaluate the effect of maternal high fiber intake during late gestation on intestinal morphology, microbiota, and intestinal proteome of newborn piglets. Sixteen sows were randomly allocated into two groups receiving the control diet (CD) and high-fiber diet (HFD) from day 90 of gestation to farrowing. Newborn piglets were selected from each litter, named as CON and Fiber group, respectively. Maternal high fiber intake did not markedly improve the birth weight, but increased the body length, the ileal crypt depth and colonic acetate level. In addition, maternal high fiber intake increased the -diversity indices (Observed species, Simpson, and ACE), and the abundance of Acidobacteria and Bacteroidetes at phylum level, significantly increased the abundance of Bradyrhizobium and Phyllobacterium at genus level in the colon of newborn piglets. Moreover, maternal high fiber intake markedly altered the ileal proteome, increasing the abundances of proteins associated with oxidative status, energy metabolism, and immune and inflammatory responses, and decreasing abundances of proteins related to cellular apoptosis, cell structure, and motility. These findings indicated that maternal high fiber intake could alter intestinal morphology, along with the altered intestinal microbiota composition and proteome of offspring.


Subject(s)
Animals, Newborn/anatomy & histology , Animals, Newborn/physiology , Dietary Fiber/administration & dosage , Gastrointestinal Microbiome/physiology , Intestines/embryology , Proteome/physiology , Actinobacteria , Animals , Animals, Newborn/microbiology , Bacteria/classification , Bacteroidetes , Colon/chemistry , Colon/microbiology , Fatty Acids, Volatile/analysis , Female , Ileum/metabolism , Ileum/ultrastructure , Intestines/physiology , Maternal Nutritional Physiological Phenomena/physiology , Models, Animal , Pregnancy , Proteobacteria , Sus scrofa
6.
Int J Mol Sci ; 22(1)2020 Dec 22.
Article in English | MEDLINE | ID: mdl-33374948

ABSTRACT

Consumer products manufactured with antimicrobial silver nanoparticles (AgNPs) may affect the gastrointestinal (GI) system. The human GI-tract is complex and there are physiological and anatomical differences between human and animal models that limit comparisons between species. Thus, assessment of AgNP toxicity on the human GI-tract may require tools that allow for the examination of subtle changes in inflammatory markers and indicators of epithelial perturbation. Fresh tissues were excised from the GI-tract of human male and female subjects to evaluate the effects of AgNPs on the GI-system. The purpose of this study was to perform an assessment on the ability of the ex vivo model to evaluate changes in levels of pro-/anti-inflammatory cytokines/chemokines and mRNA expression of intestinal permeability related genes induced by AgNPs in ileal tissues. The ex vivo model preserved the structural and biological functions of the in-situ organ. Analysis of cytokine expression data indicated that intestinal tissue of male and female subjects responded differently to AgNP treatment, with male samples showing significantly elevated Granulocyte-macrophage colony-stimulating factor (GM-CSF) after treatment with 10 nm and 20 nm AgNPs for 2 h and significantly elevated RANTES after treatment with 20 nm AgNPs for 24 h. In contrast, tissues of female showed no significant effects of AgNP treatment at 2 h and significantly decreased RANTES (20 nm), TNF-α (10 nm), and IFN-γ (10 nm) at 24 h. Smaller size AgNPs (10 nm) perturbed more permeability-related genes in samples of male subjects, than in samples from female subjects. In contrast, exposure to 20 nm AgNPs resulted in upregulation of a greater number of genes in female-derived samples (36 genes) than in male-derived samples (8 genes). The ex vivo tissue model can distinguish sex dependent effects of AgNP and could serve as a translational non-animal model to assess the impacts of xenobiotics on human intestinal mucosa.


Subject(s)
Cytokines/metabolism , Epithelial Cells/drug effects , Intestinal Mucosa/drug effects , Metal Nanoparticles/administration & dosage , RNA, Messenger/genetics , Silver/administration & dosage , Epithelial Cells/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Ileum/drug effects , Ileum/metabolism , Ileum/ultrastructure , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Male , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Particle Size , Permeability/drug effects , RNA, Messenger/metabolism , Sex Factors
7.
Sci Rep ; 10(1): 11507, 2020 07 13.
Article in English | MEDLINE | ID: mdl-32661347

ABSTRACT

Gut hyperpermeability can be caused by either apoptosis of the intestinal epithelium or altered status, permeability or porosity of tight junctions. This project aims to elucidate these mechanisms in the early phase of sepsis. Eighteen male wild type mice were randomized to two groups. All mice received one single gavage of fluorescein isothiocyanate (FITC) dextran 30 min before intervention. One group (n = 10) underwent cecal ligation and puncture to induce sepsis. The other group (n = 8) was sham operated. Septic animals exhibited significantly increased permeability for FITC 8 h post-operatively. Significantly increased serum interleukin-6, tumor-necrosis-factor-alpha and interleukin-1-beta confirmed sepsis. Septic animals showed significant bowel wall inflammation of ileum and colon samples. PCR revealed significantly increased expression of claudin-2 and decreased expressions of claudin-4, tight-junction-protein-1 and occludin-1 resembling increased permeability of tight junctions. However, these alterations could not be confirmed at the protein level. Light microscopy revealed significant dilatation of intercellular spaces at the basal sections of intestinal epithelial cells (IEC) in septic animals confirmed by increased intercellular spaces at the level of tight junctions and adherens junctions in electron microscopy (TEM). In small angle X-ray scattering no increase in number or size of nanopores could be shown in the bowel wall. HOECHST staining and PCR of ileum samples for apoptosis markers proofed no relevant differences in intestinal epithelial cell apoptosis between the groups. Intestinal hyperpermeability in septic animals was most likely caused by alterations of the intercellular contacts and not by apoptosis or increased size/number of nanopores of intestinal epithelial cells in this murine model of early sepsis.


Subject(s)
Epithelial Cells/ultrastructure , Intestines/ultrastructure , Sepsis/pathology , Tight Junctions/ultrastructure , Animals , Apoptosis/genetics , Cecum/pathology , Cecum/ultrastructure , Colon/pathology , Colon/ultrastructure , Disease Models, Animal , Epithelial Cells/pathology , Humans , Ileum/pathology , Ileum/ultrastructure , Intestinal Mucosa/pathology , Intestinal Mucosa/ultrastructure , Intestines/pathology , Mice , Permeability , Sepsis/metabolism , Tight Junctions/pathology
8.
Biochem Biophys Res Commun ; 529(2): 289-295, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32703425

ABSTRACT

Long-term high-fat feeding (HF) induces intestinal mucosal barrier damage. However, the mechanism for this remains unclear. HF can elevate the intestinal and circulating bile acid (BA) levels, especially deoxycholic acid (DCA). We hypothesize that BAs elevated by HF regulate intestinal stem cell (ISC) function, which may contribute to mucosal barrier injury in the ileum of mice. In this study, we showed that 2 weeks of HF resulted in a shortening of intestinal villi and a decrease in the tight junction (TJ) protein occludin in the ileum of mice, accompanied by an increase in circulating BA levels. Importantly, 2 weeks of HF also reduced ileal ISCs and goblet cells and decreased the proliferation function of ISCs and their ability to differentiate into goblet cells. Endoplasmic reticulum (ER) stress was found to be involved in the process of ISC damage. All these alterations were reversed by cofeeding with the bile acid binder cholestyramine. In addition, the in vitro studies also confirmed a cytotoxic effect of DCA at a high concentration on ISCs and goblet cells. In conclusion, these data suggested that high levels of BAs induced by HF could impair ISC function by triggering ER stress, resulting in the disruption of the intestinal mucosal barrier.


Subject(s)
Bile Acids and Salts/metabolism , Diet, High-Fat/adverse effects , Endoplasmic Reticulum Stress , Ileum/cytology , Intestinal Mucosa/metabolism , Stem Cells/cytology , Animals , Cell Proliferation , Ileum/ultrastructure , Intestinal Mucosa/ultrastructure , Male , Mice, Inbred C57BL , Stem Cells/metabolism
9.
J Vet Med Sci ; 82(7): 990-999, 2020 Jul 31.
Article in English | MEDLINE | ID: mdl-32493889

ABSTRACT

The comprehensive targets of innervation in the intestinal mucosa are unknown, partly because of the diversity of cell types and the complexity of the neural circuits. Herein, we investigated the comprehensive targets of neural connectivity and analyzed the precise characteristics of their contact structures in the mucosa of rat ileum. We examined target cells of neural connections and the characteristics of their contact structures by serial block-face scanning electron microscopy at four portions of the rat ileal mucosa: the apical and basal portions in the villi, and the lateral and basal portions around/in the crypts. Nerve fibers were in contact with several types of fibroblast-like cells (FBLCs), macrophage-like cells, eosinophils, lymphocyte-like cells, and other types of cells. The nerve fibers almost always ran more inside of lamina propria than subepithelial FBLC, and thus contacts with epithelial cells were very scarce. The contact structures of the nerve fibers were usually contained synaptic vesicle-like structures, and we classified them into patterns based on the number of nerve fiber contacting the target cells at one site, the maximum diameter of the contact structures, and the relationship between nerve fibers and nerve bundles. The contact structures for each type of cells occasionally dug into the cellular bodies of the target cells. We revealed the comprehensive targets of neural connectivity based on the characteristics of contact structures, and identified FBLCs, immunocompetent cells, and eosinophils as the candidate targets for innervation in the rat ileal mucosa.


Subject(s)
Ileum/innervation , Intestinal Mucosa/innervation , Nerve Fibers/ultrastructure , Animals , Fibroblasts/ultrastructure , Ileum/cytology , Ileum/ultrastructure , Intestinal Mucosa/cytology , Intestinal Mucosa/ultrastructure , Male , Microscopy, Electron, Scanning , Rats, Wistar
10.
PLoS One ; 15(6): e0232831, 2020.
Article in English | MEDLINE | ID: mdl-32497096

ABSTRACT

The burden of enteric pathogens in poultry is growing after the ban of antibiotic use in animal production. Organic acids gained attention as a possible alternative to antibiotics due to their antimicrobial activities, improved nutrient metabolism and performance. The current study was conducted to evaluate the effectiveness of organic acid blend on broilers cecal microbiota, histomorphometric measurements, and short-chain fatty acid production in Salmonella enterica serovar Typhimurium challenge model. Birds were divided into four treatments, including a negative control, positive control challenged with S. Typhimurium, group supplemented with an organic acid blend, and birds supplemented with organic acid blend and Salmonella challenged. Results illustrate significant differences in feed conversion ratios and production efficiency factor between treatment groups, however, the influence of organic acid supplement was marginal. Organic acid blend significantly increased cecal acetic and butyric acids concentrations when compared to unsupplemented groups and resulted in minor alterations of intestinal bacterial communities.


Subject(s)
Acetates/metabolism , Animal Feed , Butyrates/metabolism , Chickens/microbiology , Dietary Supplements , Fatty Acids/pharmacology , Gastrointestinal Microbiome/drug effects , Poultry Diseases/therapy , Salmonella Infections, Animal/therapy , Salmonella typhimurium/drug effects , Animals , Cecum/microbiology , Chickens/metabolism , Fatty Acids/administration & dosage , Fatty Acids, Volatile/administration & dosage , Fatty Acids, Volatile/pharmacology , Ileum/metabolism , Ileum/ultrastructure , Mannans/administration & dosage , Microvilli/ultrastructure , Poultry Diseases/microbiology , Poultry Diseases/prevention & control , Random Allocation , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/prevention & control , Salmonella typhimurium/isolation & purification , Salmonella typhimurium/metabolism
11.
Science ; 369(6499): 50-54, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32358202

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can cause coronavirus disease 2019 (COVID-19), an influenza-like disease that is primarily thought to infect the lungs with transmission through the respiratory route. However, clinical evidence suggests that the intestine may present another viral target organ. Indeed, the SARS-CoV-2 receptor angiotensin-converting enzyme 2 (ACE2) is highly expressed on differentiated enterocytes. In human small intestinal organoids (hSIOs), enterocytes were readily infected by SARS-CoV and SARS-CoV-2, as demonstrated by confocal and electron microscopy. Enterocytes produced infectious viral particles, whereas messenger RNA expression analysis of hSIOs revealed induction of a generic viral response program. Therefore, the intestinal epithelium supports SARS-CoV-2 replication, and hSIOs serve as an experimental model for coronavirus infection and biology.


Subject(s)
Betacoronavirus/physiology , Enterocytes/virology , Ileum/virology , Virus Replication , Angiotensin-Converting Enzyme 2 , Betacoronavirus/ultrastructure , Cell Culture Techniques , Cell Differentiation , Cell Lineage , Cell Proliferation , Culture Media , Enterocytes/metabolism , Enterocytes/ultrastructure , Gene Expression , Humans , Ileum/metabolism , Ileum/ultrastructure , Lung/virology , Male , Organoids , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Respiratory Mucosa/virology , Severe acute respiratory syndrome-related coronavirus/physiology , SARS-CoV-2
12.
Sci Rep ; 10(1): 6692, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32317678

ABSTRACT

Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease of incompletely understood pathophysiology predominantly affecting premature infants. While NEC is associated with microbial invasion of intestinal tissues, and mucus modulates interactions between microbes and underlying tissues, variations in mucus barrier properties with NEC-associated risk factors have not been investigated. This study explored differences in mucus composition (total protein, DNA, mucin content, sialic acid, and immunoregulatory proteins), as well as structural and transport properties, assessed by tracking of particles and bacteria (E. coli and E. cloacae) with developmental age and exposure to NEC stressors in Sprague Dawley rats. Early developmental age (5 day old) was characterized by a more permeable mucus layer relative to 21 day old pups, suggesting immaturity may contribute to exposure of the epithelium to microbes. Exposure to NEC stressors was associated with reduced mucus permeability, which may aid in survival. Feeding with breastmilk as opposed to formula reduces incidence of NEC. Thus, NEC-stressed (N-S) rat pups were orally dosed with breastmilk components lysozyme (N-S-LYS) or docosahexaenoic acid (N-S-DHA). N-S-LYS and N-S-DHA pups had a less permeable mucus barrier relative to N-S pups, which suggests the potential of these factors to strengthen the mucus barrier and thus protect against disease.


Subject(s)
Aging/pathology , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/therapeutic use , Enterocolitis, Necrotizing/drug therapy , Mucus/metabolism , Muramidase/administration & dosage , Muramidase/therapeutic use , Stress, Physiological , Administration, Oral , Animals , DNA/metabolism , Docosahexaenoic Acids/pharmacology , Enterobacter cloacae/physiology , Enterocolitis, Necrotizing/microbiology , Escherichia coli/physiology , Fucose/metabolism , Ileum/pathology , Ileum/ultrastructure , Immunoglobulin G/metabolism , Mucins/metabolism , Mucus/drug effects , Muramidase/pharmacology , N-Acetylneuraminic Acid/metabolism , Permeability , Polyethylene Glycols/chemistry , Rats, Sprague-Dawley , Stress, Physiological/drug effects
13.
Poult Sci ; 99(2): 1135-1149, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32036965

ABSTRACT

Salmonella and Campylobacter are considered major public health burdens worldwide, and poultry are known to be one of the main reservoirs for these zoonotic pathogens. This study was conducted to evaluate the effect of a commercial probiotic or direct-fed microbial (DFM) Calsporin (CSP), and prebiotic or mannan oligosaccharide (MOS) (IMW50) on ultrastructural changes and the villous integrity of intestinal mucosa in turkey poults challenged with Salmonella and Campylobacter. A 21-day battery cage study was conducted using 4 dietary treatments including a basal diet (corn and soybean-based) nonsupplemented and uninfected as a negative control (NC); basal diet supplemented with 0.05% DFM (CSP); basal diet supplemented with 0.05% MOS (IMW50); and basal diet supplemented with 0.05% mixture of DFM and MOS at equal proportions. Female large white turkey poults aged 336 days were obtained from a local commercial hatchery and randomly distributed in electrically heated battery cages with 12 treatments of 4 replicates per treatment containing 7 poults per pen. The first 16 pens were not infected with bacteria, poults in pens 17-32 were orally challenged at day 7 with 105 cfu Salmonella Heidelberg, and the poults in pens 33-48 were orally challenged at day 7 with 105 cfu Campylobacter jejuni. Feed and water were provided ad libitum throughout the study. At day 21, ileal tissue samples from 1 bird per cage were collected for intestinal integrity and ultrastructural examination by scanning and electron microscopy. DFM and MOS supplementation was effective in both challenged and nonchallenged (not infected with Salmonella and Campylobacter) birds. Goblet cells and mucus were increased, with the presence of large numbers of segmented filamentous bacteria in DFM- and MOS-supplemented groups compared with birds in control treatments. The number and size of villi were reduced in poults exposed to Salmonella and Campylobacter. Results show that CSP and IMW50 provide protection of ileal mucosal integrity in poults exposed to Salmonella or Campylobacter.


Subject(s)
Campylobacter Infections/veterinary , Poultry Diseases/prevention & control , Prebiotics , Probiotics/pharmacology , Salmonella Infections, Animal/prevention & control , Turkeys , Animal Feed/analysis , Animals , Campylobacter/physiology , Campylobacter Infections/microbiology , Campylobacter Infections/prevention & control , Diet/veterinary , Dietary Supplements/analysis , Ileum/drug effects , Ileum/ultrastructure , Intestinal Mucosa/drug effects , Mannans/pharmacology , Microscopy, Electron, Scanning/veterinary , Microscopy, Electron, Transmission/veterinary , Oligosaccharides/pharmacology , Random Allocation , Saccharomyces cerevisiae/chemistry , Salmonella/physiology , Salmonella Infections, Animal/microbiology
14.
Microb Ecol ; 79(4): 1021-1033, 2020 May.
Article in English | MEDLINE | ID: mdl-31728601

ABSTRACT

Segmented filamentous bacteria (SFB) are well known for their functions in the immunoregulation of hosts including the promotion of Th17 cell differentiation, B cell maturation, and immune system development. However, most analyses of SFB have focused on animal models, and thus, investigation of SFB prevalence in humans and their roles in human immunoregulation and health is needed. Although little is known overall of SFB prevalence in humans, they are characteristically abundant in animals during weaning. In this study, SFB-like bacteria were detected in ileal lavage samples from human children that were aged between 1 to 17 years old by scanning electron microscopy (SEM) analysis, and their insertion into the mucosa was also observed. In addition, the expression of SFB flagellin at the human bacterial interface was observed by immunohistochemistry (IHC) and western blot. Moreover, two pairs of primers specific for SFB, but targeting different genes, were used to detect SFB in human intestinal lavage samples. These analyses indicated that SFB were present in over 50% of patient ileal samples independent of age. High-throughput gene sequencing indicated that different SFB strains were detected among samples. Between nine and 23 SFB flagellin gene operational taxonomic units were identified. In addition to evaluating the prevalence of SFB in human samples, correlations between SFB presence and chief complaints of clinical symptoms were evaluated, as well as the relationship between SFB and patient serum immunoglobulin concentrations. SFB prevalence was significantly higher in hematochezia patients (68%) than in abdominal pain (56.10%) and diarrhea (43.75%) patients. Furthermore, the concentrations of serum IgA, IgM, and IgE, were similar between SFB-positive and SFB-negative patient groups, although IgG concentrations were significantly higher in the SFB-negative group.


Subject(s)
Bacteria/isolation & purification , Gastrointestinal Microbiome , Ileum/microbiology , Adolescent , Bacteria/classification , Child , Child, Preschool , China , Female , Flagellin/analysis , Humans , Ileum/ultrastructure , Infant , Male , Microscopy, Electron, Scanning
15.
Sci Transl Med ; 11(517)2019 11 06.
Article in English | MEDLINE | ID: mdl-31694928

ABSTRACT

Cryptosporidium is an intestinal pathogen that causes severe but self-limiting diarrhea in healthy humans, yet it can turn into a life-threatening, unrelenting infection in immunocompromised patients and young children. Severe diarrhea is recognized as the leading cause of mortality for children below 5 years of age in developing countries. The only approved treatment against cryptosporidiosis, nitazoxanide, has limited efficacy in the most vulnerable patient populations, including malnourished children, and is ineffective in immunocompromised individuals. Here, we investigate inhibition of the parasitic cleavage and polyadenylation specificity factor 3 (CPSF3) as a strategy to control Cryptosporidium infection. We show that the oxaborole AN3661 selectively blocked Cryptosporidium growth in human HCT-8 cells, and oral treatment with AN3661 reduced intestinal parasite burden in both immunocompromised and neonatal mouse models of infection with greater efficacy than nitazoxanide. Furthermore, we present crystal structures of recombinantly produced Cryptosporidium CPSF3, revealing a mechanism of action whereby the mRNA processing activity of this enzyme is efficiently blocked by the binding of the oxaborole group at the metal-dependent catalytic center. Our data provide insights that may help accelerate the development of next-generation anti-Cryptosporidium therapeutics.


Subject(s)
Cleavage And Polyadenylation Specificity Factor/metabolism , Cryptosporidiosis/genetics , Cryptosporidiosis/parasitology , Cryptosporidium/genetics , Metals/chemistry , RNA Precursors/genetics , RNA Processing, Post-Transcriptional , Animals , Antiparasitic Agents/chemistry , Antiparasitic Agents/pharmacology , Cell Line, Tumor , Cleavage And Polyadenylation Specificity Factor/chemistry , Crystallization , Humans , Ileum/parasitology , Ileum/ultrastructure , Mice, Inbred C57BL , Models, Molecular , Recombinant Proteins/metabolism
16.
Biomed Pharmacother ; 118: 109393, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31545258

ABSTRACT

OBJECTIVE: Diabetes mellitus is associated with gut microbiota disturbance and intestinal mucosal injuries. This study investigated the influence of propolis on the gut microbiota and intestinal mucosa in rats with diabetes. METHODS: Sprague-Dawley (SD) rats were randomly assigned to the control group, model group, and three propolis groups (supplemented with 80, 160, and 240 mg/kg·bw propolis, respectively). A high-fat diet combined with a streptozotocin (STZ) abdominal injection were used to induce diabetes in the rats. After 4 weeks, the intestinal histopathological analysis of the ileum was observed by transmission electron microscopy. The fasting blood glucose (FBG), plasma insulin, glucose tolerance (OGTT) and glycosylated hemoglobin (HbA1c) levels were measured. The expression of tight junction (TJ) proteins in the ileum was measured using western blotting. The molecular ecology of the fecal gut microbiota was analyzed by 16S rDNA high-throughput sequencing. The contents of the short-chain fatty acids (SCFAs) in feces were measured using high-performance liquid chromatography (HPLC). RESULTS: After propolis treatment, compared to the model group, FBG and HbA1c levels declined, while the glucose tolerance and insulin sensitivity index (ISI) increased. The levels of TJ proteins in the ileum increased in the propolis groups. The tight junctions and gap junctions of the intestinal epithelium were also improved in the propolis groups. The contents of the feces acetic acid, propionic acid and butyrate were increased in the propolis groups. 16S rDNA high-throughput sequencing revealed that the composition of the gut microbiota of rats in the propolis supplement group was significantly improved. CONCLUSIONS: Compared to the model group, propolis exerted hypoglycemic effects in diabetic rats, and it repaired intestinal mucosal damage, benefited the communities of the gut microbiota and increased SCFA levels in diabetic rats.


Subject(s)
Diabetes Mellitus, Experimental/microbiology , Diabetes Mellitus, Experimental/physiopathology , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiopathology , Propolis/pharmacology , Animals , Biodiversity , Blood Glucose/metabolism , Body Weight/drug effects , Drinking/drug effects , Fasting/blood , Fatty Acids/metabolism , Feces/chemistry , Ileum/drug effects , Ileum/pathology , Ileum/ultrastructure , Insulin/blood , Intestinal Mucosa/drug effects , Male , Metabolic Networks and Pathways/drug effects , Phylogeny , Rats, Sprague-Dawley
17.
Acta Histochem ; 121(6): 665-679, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31202513

ABSTRACT

The aim of this study was to analyze the effect of ischemia and reperfusion injury (IS) on enteric glial cells (EGCs) and neurons immunoreactive for the P2X7 receptor. Intestinal ischemia was induced by obstructing blood flow in the ileal vessels for 35 min. Afterwards, the vessels were reperfused for 14 days. Tissues were prepared for immunohistochemical labeling of P2X7 receptor, HuC/D (Hu) (pan-neuronal marker) and S100ß (glial marker); HuC/D (Hu) and glial fibrillary acidic protein (GFAP, glial marker)/DAPI (nuclear marker); or S100ß and GFAP/DAPI. Qualitative and quantitative analyses of colocalization, density, profile area and cell proliferation were performed via fluorescence and confocal laser scanning microscopy. The quantitative analyses revealed that a) neurons and EGCs were immunoreactive for P2X7 receptor; b) the P2X7 receptor immunoreactive cells and Hu immunoreactive neurons were reduced after 0 h and 14 days of reperfusion; c) the S100ß and GFAP immunoreactive EGCs were increased; d) the profile area of S100ß immunoreactive EGCs was increased by IS; e) few GFAP immunoreactive proliferated at 14 days of reperfusion; f) distinct populations of glial cells can be discerned: S100ß+/GFAP+ cells, S100ß+/GFAP- cells and S100ß-/GFAP + cells; g) histological analysis revealed less alterations in the epithelium cells in the IS groups and h) myeloperoxidase reaction revealed increased of the neutrophils in the lamina propria in the IS groups. This study showed that IS is associated with significant neuronal loss, increase of glial cells and altered purinergic receptor expression and that these changes may contribute to intestinal disorders.


Subject(s)
Ileum/metabolism , Neuroglia/metabolism , Receptors, Purinergic P2X7/metabolism , Reperfusion Injury/metabolism , Animals , Ileum/ultrastructure , Male , Neuroglia/ultrastructure , Rats , Rats, Wistar , Reperfusion Injury/pathology
18.
J Vet Med Sci ; 81(3): 454-465, 2019 Mar 30.
Article in English | MEDLINE | ID: mdl-30700677

ABSTRACT

Serial block-face scanning electron microscopy (SBF-SEM) is useful for three-dimensional observation of tissues or cells at high-resolution. In this study, SBF-SEM was used to three-dimensionally analyze the characteristics of fibroblast-like cells (FBLCs) in the rat ileal lamina propria (LP). The results revealed that FBLCs in LP could be divided into four types, tentatively named FBLC type I-IV, based on the external cellular appearance, abundance or shape of each organelle, detailed distribution in the LP and relationship with surrounding cells. FBLC-I and -II localized around the intestinal crypt (InC), FBLC-III localized from the lateral portion of InC to the apical portion of the intestinal villus (InV), and FBLC-IV localized in the InV. FBLC-I, -II and -III, but not FBLC-IV, localized beneath the epithelium. FBLC-II possessed thin lamellar-shaped endoplasmic reticula, whereas FBLC-I possessed expanded endoplasmic reticula that occasionally showed a spherical shape. FBLC-III and -IV possessed a cytoplasmic region with high-electron density and no organelles immediately beneath the cellular membrane; this region was found at the epithelial sides in FBLC-III and scattered in FBLC-IV. FBLC-IV were in constant close proximity to villous myocytes throughout the InV and also in contact with FBLC-III especially in the apical portion of the InV. FBLC-I, -II and -IV, but not -III, were constantly found to be in contact with various immunocompetent cells in the LP. Three-dimensional analysis using SBF-SEM indicates that four types of FBLC localized in the rat ileal LP.


Subject(s)
Fibroblasts/ultrastructure , Ileum/ultrastructure , Intestinal Mucosa/ultrastructure , Animals , Ileum/cytology , Intestinal Mucosa/cytology , Male , Microscopy, Electron, Scanning/methods , Organelles/ultrastructure , Rats , Rats, Wistar
19.
J Cell Mol Med ; 22(11): 5617-5628, 2018 11.
Article in English | MEDLINE | ID: mdl-30188001

ABSTRACT

Intestinal mesenchymal cells deposit extracellular matrix in fibrotic Crohn's disease (CD). The contribution of epithelial to mesenchymal transition (EMT) to the mesenchymal cell pool in CD fibrosis remains obscure. The miR-200 family regulates fibrosis-related EMT in organs other than the gut. E-cadherin, cytokeratin-18 and vimentin expression was assessed using immunohistochemistry on paired strictured (SCD) and non-strictured (NSCD) ileal CD resections and correlated with fibrosis grade. MiR-200 expression was measured in paired SCD and NSCD tissue compartments using laser capture microdissection and RT-qPCR. Serum miR-200 expression was also measured in healthy controls and CD patients with stricturing and non-stricturing phenotypes. Extra-epithelial cytokeratin-18 staining and vimentin-positive epithelial staining were significantly greater in SCD samples (P = 0.04 and P = 0.03, respectively). Cytokeratin-18 staining correlated positively with subserosal fibrosis (P < 0.001). Four miR-200 family members were down-regulated in fresh SCD samples (miR-141, P = 0.002; miR-200a, P = 0.002; miR-200c, P = 0.001; miR-429; P = 0.004); miR-200 down-regulation in SCD tissue was localised to the epithelium (P = 0.001-0.015). The miR-200 target ZEB1 was up-regulated in SCD samples (P = 0.035). No difference in serum expression between patient groups was observed. Together, these observations suggest the presence of EMT in CD strictures and implicate the miR-200 family as regulators. Functional studies to prove this relationship are now warranted.


Subject(s)
Antigens, CD/genetics , Cadherins/genetics , Crohn Disease/genetics , Fibrosis/genetics , MicroRNAs/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Adult , Crohn Disease/pathology , Crohn Disease/surgery , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Female , Fibrosis/pathology , Fibrosis/surgery , Gene Expression Regulation/genetics , Humans , Ileum/pathology , Ileum/ultrastructure , Keratin-18/genetics , Male , Vimentin/genetics
20.
Kaohsiung J Med Sci ; 34(3): 134-141, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29475460

ABSTRACT

The intestinal mucosal barrier (IMB) enables the intestine to provide adequate containment of luminal microorganisms and molecules while preserving the ability to absorb nutrients. In this study, we explored the effect of brain-derived neurotrophic factor (BDNF) on IMB function and gut microbiota in mice. BDNF gene knock-out mice (the BDNF+/- group) and wild-type mice (the BDNF+/+ group) were selected. The gut microbiota of these mice was analyzed by denaturing gradient gel electrophoresis (DGGE) assay. The ultrastructure of the ileum and the colonic epithelium obtained from decapitated mice were observed by transmission electron microscopy. The protein expression of epithelial tight junction proteins, zonula occludens-1 (ZO-1) and occludin was detected by immunohistochemistry staining. The protein expression of claudin-1 and claudin-2 was determined by Western blotting. The DGGE band patterns of gut microbiota in the BDNF+/- group were significantly different from that in the BDNF+/+ group, which indicated that the BDNF expression alters the gut microbiota in mice. Compared with the BDNF+/+ group, the BDNF+/- group presented no significant difference in the ultrastructure of ileal epithelium; however, a significant difference was observed in the colonic epithelial barrier, manifested by decreased microvilli, widening intercellular space and bacterial invasion. Compared with the BDNF+/+ group, the expression of ZO-1 and occludin in the BDNF+/- group was significantly decreased. The expression of claudin-1 in the BDNF+/- group was significantly reduced, while the expression of claudin-2 was elevated. These findings indicate that BDNF preserves IMB function and modulates gut microbiota in mice.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Colon/microbiology , Gastrointestinal Microbiome/physiology , Ileum/microbiology , Intestinal Mucosa/microbiology , Animals , Brain-Derived Neurotrophic Factor/deficiency , Claudin-1/genetics , Claudin-1/metabolism , Claudin-2/genetics , Claudin-2/metabolism , Colon/metabolism , Colon/ultrastructure , Gene Expression Regulation , Ileum/metabolism , Ileum/ultrastructure , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Male , Mice , Mice, Knockout , Microvilli/metabolism , Microvilli/microbiology , Microvilli/ultrastructure , Occludin/genetics , Occludin/metabolism , Tight Junctions/metabolism , Tight Junctions/microbiology , Tight Junctions/ultrastructure , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...