Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; 10(27): e2300164, 2023 09.
Article in English | MEDLINE | ID: mdl-37525340

ABSTRACT

Several stomach diseases are attributed to the dysregulation of physiological function of gastric mucosal barrier by pathogens. Gastric organoids are a promising tool to develop treatment strategies for gastric infections. However, their functional features of in vivo gastric mucosal barrier and host-microbe interactions are limited due to the lack of physiological stimuli. Herein, a human stomach micro-physiological system (hsMPS) with physiologically relevant gastric mucosal defense system is described based on the combination of organoid and MPS technology. A fluid flow enhanced epithelial-mesenchymal interaction in the hsMPS enables functional maturation of gastric epithelial cells, which allows for the recreation of mesh-like mucus layer containing high level of mucus protective peptides and well-developed epithelial junctional complexes. Furthermore, gastroprotection mechanisms against Helicobacter pylori (H. pylori) are successfully demonstrated in this system. Therefore, hsMPS represents a new in vitro tool for research where gastric mucosal defense mechanism is pivotal for developing therapeutic strategies.


Subject(s)
Mucous Membrane , Stomach , Humans , Epithelial Cells , Organoids , Defense Mechanisms
2.
Gastroenterology ; 164(4): 550-566, 2023 04.
Article in English | MEDLINE | ID: mdl-36587707

ABSTRACT

BACKGROUND & AIMS: Infection with Helicobacter pylori strongly affects global health by causing chronic gastritis, ulcer disease, and gastric cancer. Although extensive research into the strong immune response against this persistently colonizing bacterium exists, the specific role of CD8+ T cells remains elusive. METHODS: We comprehensively characterize gastric H pylori-specific CD8+ T-cell responses in mice and humans by flow cytometry, RNA-sequencing, immunohistochemistry, and ChipCytometry, applying functional analyses including T-cell depletion, H pylori eradication, and ex vivo restimulation. RESULTS: We define CD8+ T-cell populations bearing a tissue-resident memory (TRM) phenotype, which infiltrate the gastric mucosa shortly after infection and mediate pathogen control by executing antigen-specific effector properties. These induced CD8+ tissue-resident memory T cells (TRM cells) show a skewed T-cell receptor beta chain usage and are mostly specific for cytotoxin-associated gene A, the distinctive oncoprotein injected by H pylori into host cells. As the infection progresses, we observe a loss of the TRM phenotype and replacement of CD8+ by CD4+ T cells, indicating a shift in the immune response during the chronic infection phase. CONCLUSIONS: Our results point toward a hitherto unknown role of CD8+ T-cell response in this bacterial infection, which may have important clinical implications for treatment and vaccination strategies against H pylori.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Humans , Animals , Mice , CD8-Positive T-Lymphocytes , CD4-Positive T-Lymphocytes , Stomach , Gastric Mucosa/microbiology , Helicobacter Infections/microbiology , Antigens, Bacterial , Bacterial Proteins
3.
EMBO Rep ; 24(2): e54925, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36440604

ABSTRACT

Vault RNAs (vtRNAs) are small noncoding RNAs and highly expressed in many eukaryotes. Here, we identified vtRNA2-1 as a novel regulator of the intestinal barrier via interaction with RNA-binding protein HuR. Intestinal mucosal tissues from patients with inflammatory bowel diseases and from mice with colitis or sepsis express increased levels of vtRNAs relative to controls. Ectopically expressed vtRNA2-1 decreases the levels of intercellular junction (IJ) proteins claudin 1, occludin, and E-cadherin and causes intestinal epithelial barrier dysfunction in vitro, whereas vtRNA2-1 silencing promotes barrier function. Increased vtRNA2-1 also decreases IJs in intestinal organoid, inhibits epithelial renewal, and causes Paneth cell defects ex vivo. Elevating the levels of tissue vtRNA2-1 in the intestinal mucosa increases the vulnerability of the gut barrier to septic stress in mice. vtRNA2-1 interacts with HuR and prevents HuR binding to claudin 1 and occludin mRNAs, thus decreasing their translation. These results indicate that vtRNA2-1 impairs intestinal barrier function by repressing HuR-facilitated translation of claudin 1 and occludin.


Subject(s)
Colitis , MicroRNAs , Paneth Cells , Animals , Mice , Claudin-1/genetics , Claudin-1/metabolism , Colitis/genetics , Colitis/metabolism , Intestinal Mucosa/metabolism , Occludin/metabolism , MicroRNAs/metabolism
4.
Nutrients ; 16(1)2023 Dec 23.
Article in English | MEDLINE | ID: mdl-38201885

ABSTRACT

Cinnamomum cassia (cassia) is a tropical aromatic evergreen tree of the Lauraceae family well known for its fragrance and spicy flavor and widely used in Asian traditional medicine. It has recently garnered attention for its diverse potential health benefits, including anti-inflammatory, anti-cancer, and anti-diabetic properties. However, the gastroprotective effect of C. cassia, particularly against ethanol-induced gastric damage, remains unclear. We investigated the potential gastroprotective property of C. cassia and the underlying mechanisms of action in a rat model of ethanol-induced gastric injury. To assess its effectiveness, rats were fed C. cassia for a 14-day period prior to inducing gastric damage by oral administration of ethanol. Our results indicated that pre-treatment with C. cassia mitigated ethanol-induced gastric mucosal lesions and bleeding. Reduced gastric acid secretion and expression of acid secretion-linked receptors were also observed. Additionally, pretreatment with C. cassia led to decreased levels of inflammatory factors, including TNF-α, p-p65, and IκBα. Notably, C. cassia upregulated the expressions of HO1 and HSP90, with particular emphasis on the enhanced expression of PAS and MUC, the crucial gastric mucosa defense molecules. These findings suggest that C. cassia has protective effects on the gastric mucosa and can effectively reduce oxidative stress and inflammation.


Subject(s)
Cinnamomum aromaticum , Animals , Rats , Gastric Mucosa , Stomach , Administration, Oral , Ethanol/toxicity
5.
Fish Shellfish Immunol ; 131: 349-357, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36241003

ABSTRACT

Mucins are highly glycosylated proteins that make up the mucus covering internal and external surfaces of fish. Mucin O-glycans regulate pathogen quorum sensing, growth, virulence and attachment to the host. Knowledge on this mucosal defense system can enable alternative treatments to diseases posing a threat to productivity and welfare in aquaculture. Here, we characterize the rainbow trout (Oncorhynchus mykiss) gill, skin, pyloric ceca and distal intestinal mucin O-glycosylation and compare it to known teleost O-glycomes. We identified 54 O-glycans, consisting of up to nine monosaccharide residues. Skin glycans were most acidic, shortest on average and consisted mainly of NeuAcα2-6GalNAc. Glycans from the gills were less acidic with predominantly core 1 and 2 glycans, whereas glycans from pyloric ceca and distal intestine expressed an increased number of core 5 glycans, distinctly decorated with NeuAcα2-8NeuAc- like epitopes. When compared to Atlantic salmon and Arctic charr, trends on the core distribution, average size and overall acidity remained similar, although the epitopes varied. Rainbow trout mucins from gill and intestine bound A. salmonicida and A. hydrophila more efficiently than skin mucins. This is in line with a model where skin mucins with small glycans limit bacterial adhesion to the fish surface whereas the complex intestinal mucin glycans aid in trapping and removing pathogens from the epithelial surface.


Subject(s)
Mucins , Oncorhynchus mykiss , Animals , Mucins/metabolism , Glycosylation , Oncorhynchus mykiss/metabolism , Intestines , Polysaccharides/metabolism
7.
Fish Shellfish Immunol ; 122: 181-190, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35077869

ABSTRACT

Mucus, whereof the highly glycosylated mucins are a major component, protects the epithelial mucosal surfaces. The aim of this study was to characterize the rainbow trout (Oncorhynchus mykiss) gastrointestinal mucus barrier function, mucin production, glycosylation and response to lipopolysaccharide. Both gastric and intestinal mucus was thick and impenetrable to bacteria-sized beads ex vivo. The secreted mucus covering the gastric epithelium predominantly contained sialylated mucins. Plume-like structures emerging from the gastric pits were both sialylated and fucosylated, indicating heterogeneity in gastric mucus secreted by the surface mucus cells and gland secretory cells, whereas intestinal mucus appeared more homogenous. In vivo metabolic mucin labelling revealed regional differences in mucin production and basal to apical transport, while lipopolysaccharide stimulation increased the rate of mucin production and basal to apical transport in both stomach and intestine. Using mass spectrometry, 34 mucin O-glycans were identified, with ∼70% of the relative abundance being sialylated, ∼40% di-sialylated and 20-25% fucosylated. No effects of lipopolysaccharide treatment were apparent regarding O-glycan repertoires, relative abundance of components, size distribution or core structures. Thus, the mucus production and organization differ between epithelial sites but provide a barrier to bacteria in both stomach and intestine. Furthermore, mucin production and basal to apical transport was stimulated by lipopolysaccharide in all regions, suggesting a mechanism to combat infections.


Subject(s)
Mucins , Oncorhynchus mykiss , Animals , Glycosylation , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Mucins/metabolism , Mucus/metabolism , Oncorhynchus mykiss/metabolism
8.
Biomed Pharmacother ; 144: 112258, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34614465

ABSTRACT

Portal hypertensive gastropathy (PHG) is a complication of cirrhotic or noncirrhotic portal hypertension. PHG is very important in the clinic because it can cause acute or even massive blood loss, and its treatment efficacy and prognosis are poor. Currently, the incidence of PHG in patients with cirrhosis is 20-80%, but its pathogenesis is complicated and poorly understood. Studies have shown that portal hypertension can cause changes in gastric mucosal microcirculation hemodynamics, leading to changes in gastric mucosal histology and function and thereby weakening the mucosal defense barrier. However, no specific drug treatment plans are currently available. This article reviews the current literature to further our understanding of the mechanism underlying PHG and the relationship between PHG and the posterior mucosal defense barrier and to explore new therapeutic targets.


Subject(s)
Endothelial Cells/metabolism , Gastric Mucosa/blood supply , Hemodynamics , Hypertension, Portal/metabolism , Microcirculation , Splanchnic Circulation , Stomach Diseases/metabolism , Animals , Antioxidants/therapeutic use , Apoptosis , Endothelial Cells/drug effects , Endothelial Cells/pathology , Gastrointestinal Agents/therapeutic use , Humans , Hypertension, Portal/drug therapy , Hypertension, Portal/pathology , Hypertension, Portal/physiopathology , Oxidative Stress , Stomach Diseases/drug therapy , Stomach Diseases/pathology , Stomach Diseases/physiopathology
9.
Front Pediatr ; 9: 624045, 2021.
Article in English | MEDLINE | ID: mdl-33681101

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic relapsing-remitting immune-mediated disorder affecting the gut. It is common in Westernized regions and is increasing in incidence in developing countries. At a molecular level, intrinsic deficiencies in epithelial integrity, mucosal barrier function, and mechanisms of immune response and resolution contribute to the development of IBD. Traditionally two platforms have been utilized for disease modeling of IBD; in-vitro monolayer cell culture and in-vivo animal models. Both models have limitations, including cost, lack of representative cell types, lack of complexity of cellular interactions in a living organism, and xenogeneity. Organoids, three-dimensional cellular structures which recapitulate the basic architecture and functional processes of the organ of origin, hold potential as a third platform with which to investigate the pathogenesis and molecular defects which give rise to IBD. Organoids retain the genetic and transcriptomic profile of the tissue of origin over time and unlike monolayer cell culture can be induced to differentiate into most adult intestinal cell types. They may be used to model intestinal host-microbe interactions occurring at the mucosal barrier, are amenable to genetic manipulation and can be co-cultured with other cell lines of interest. Bioengineering approaches may be applied to render a more faithful representation of the intestinal epithelial niche. In this review, we outline the concept of intestinal organoids, discuss the advantages and disadvantages of the platform comparative to alternative models, and describe the translational applications of organoids in IBD.

10.
Proc Natl Acad Sci U S A ; 117(47): 29862-29871, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33172989

ABSTRACT

Organelle remodeling is critical for cellular homeostasis, but host factors that control organelle function during microbial infection remain largely uncharacterized. Here, a genome-scale CRISPR/Cas9 screen in intestinal epithelial cells with the prototypical intracellular bacterial pathogen Salmonella led us to discover that type I IFN (IFN-I) remodels lysosomes. Even in the absence of infection, IFN-I signaling modified the localization, acidification, protease activity, and proteomic profile of lysosomes. Proteomic and genetic analyses revealed that multiple IFN-I-stimulated genes including IFITM3, SLC15A3, and CNP contribute to lysosome acidification. IFN-I-dependent lysosome acidification was associated with elevated intracellular Salmonella virulence gene expression, rupture of the Salmonella-containing vacuole, and host cell death. Moreover, IFN-I signaling promoted in vivo Salmonella pathogenesis in the intestinal epithelium where Salmonella initiates infection, indicating that IFN-I signaling can modify innate defense in the epithelial compartment. We propose that IFN-I control of lysosome function broadly impacts host defense against diverse viral and microbial pathogens.


Subject(s)
Epithelial Cells/immunology , Interferon Type I/metabolism , Intestinal Mucosa/immunology , Lysosomes/metabolism , Salmonella Infections/immunology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Cas Systems/genetics , Disease Models, Animal , Epithelial Cells/chemistry , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation, Bacterial/immunology , HT29 Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Hydrogen-Ion Concentration , Immunity, Innate , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Lysosomes/chemistry , Lysosomes/immunology , Mice , Mice, Knockout , Necroptosis/immunology , Peptide Hydrolases/metabolism , Proteomics , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Salmonella Infections/microbiology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Signal Transduction/immunology , Virulence/immunology , Virulence Factors/genetics , Virulence Factors/metabolism
11.
Front Immunol ; 11: 2054, 2020.
Article in English | MEDLINE | ID: mdl-33013869

ABSTRACT

Throughout the gastrointestinal (GI) tract, a distinct mucus layer composed of highly glycosylated proteins called mucins plays an essential role in providing lubrication for the passage of food, participating in cell signaling pathways and protecting the host epithelium from commensal microorganisms and invading pathogens, as well as toxins and other environmental irritants. These mucins can be broadly classified into either secreted gel-forming mucins, those that provide the structural backbone for the mucus barrier, or transmembrane mucins, those that form the glycocalyx layer covering the underlying epithelial cells. Goblet cells dispersed among the intestinal epithelial cells are chiefly responsible for the synthesis and secretion of mucins within the gut and are heavily influenced by interactions with the immune system. Evidence from both clinical and animal studies have indicated that several GI conditions, including inflammatory bowel disease (IBD), colorectal cancer, and numerous enteric infections are accompanied by considerable changes in mucin quality and quantity. These changes include, but are not limited to, impaired goblet cell function, synthesis dysregulation, and altered post-translational modifications. The current review aims to highlight the structural and functional features as well as the production and immunological regulation of mucins and the impact these key elements have within the context of barrier function and host defense in intestinal inflammation.


Subject(s)
Gastrointestinal Diseases/immunology , Goblet Cells/physiology , Inflammation/immunology , Intestinal Mucosa/metabolism , Mucins/metabolism , Animals , Humans , Immunity, Mucosal , Models, Animal
12.
Microorganisms ; 8(9)2020 Sep 02.
Article in English | MEDLINE | ID: mdl-32887435

ABSTRACT

Innate lymphoid cells (ILCs) are a recently discovered type of innate immune lymphocyte. They include three different groups classified by the nature of the transcription factors required for their development and by the cytokines they produce. ILCs mainly reside in tissues close to the mucosal barrier such as the respiratory and gastrointestinal tracts. Due to their close proximity to the mucosal surface, ILCs are exposed to a variety of both commensal and pathogenic bacteria. Under non-pathological conditions, ILCs have been shown to be important regulators for the maintenance of tissue homeostasis by mutual interactions with the microbiome. Besides these important functions at homeostasis, several studies have also provided emerging evidence that ILCs contribute to defense against pathogenic bacterial infection by responding rapidly to the pathogens as well as orchestrating other immune cells. In this review, we summarize recent advances in our understanding of the interactions of ILCs and bacteria, with special focus on the function of the different ILC subsets in bacterial infections.

13.
Mol Cell Biol ; 40(6)2020 02 27.
Article in English | MEDLINE | ID: mdl-31932481

ABSTRACT

Intestinal epithelial autophagy is crucial for host defense against invasive pathogens, and defects in this process occur frequently in patients with inflammatory bowel disease (IBD) and other mucosal disorders, but the exact mechanism that activates autophagy is poorly defined. Here, we investigated the role of RNA-binding protein HuR (human antigen R) in the posttranscriptional control of autophagy-related genes (ATGs) in the intestinal epithelium. We found that targeted deletion of HuR in intestinal epithelial cells (IECs) specifically decreased the levels of ATG16L1 in the intestinal mucosa. Intestinal mucosa from patients with IBD exhibited reduced levels of both HuR and ATG16L1. HuR directly interacted with Atg16l1 mRNA via its 3' untranslated region and enhanced ATG16L1 translation, without affecting Atg16l1 mRNA stability. Circular RNA circPABPN1 blocked HuR binding to Atg16l1 mRNA and lowered ATG16L1 production. HuR silencing in cultured IECs also prevented rapamycin-induced autophagy, which was abolished by overexpressing ATG16L1. These findings indicate that HuR regulates autophagy by modulating ATG16L1 translation via interaction with circPABPN1 in the intestinal epithelium.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy/genetics , ELAV-Like Protein 1/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/metabolism , Poly(A)-Binding Protein I/metabolism , 3' Untranslated Regions/genetics , Animals , Autophagy/physiology , Caco-2 Cells , Cell Line, Tumor , ELAV-Like Protein 1/genetics , Gene Expression Regulation/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Biosynthesis/genetics
14.
Cell Mol Gastroenterol Hepatol ; 9(4): 611-625, 2020.
Article in English | MEDLINE | ID: mdl-31862317

ABSTRACT

BACKGROUND & AIMS: The protective intestinal mucosal barrier consists of multiple elements including mucus and epithelial layers and immune defense; nonetheless, barrier dysfunction is common in various disorders. The imprinted and developmentally regulated long noncoding RNA H19 is involved in many cell processes and diseases. Here, we investigated the role of H19 in regulating Paneth and goblet cells and autophagy, and its impact on intestinal barrier dysfunction induced by septic stress. METHODS: Studies were conducted in H19-deficient (H19-/-) mice, mucosal tissues from patients with sepsis, primary enterocytes, and Caco-2 cells. Septic stress was induced by cecal ligation and puncture (CLP), and gut permeability was detected by tracer fluorescein isothiocyanate-dextran assays. The function of Paneth and goblet cells was examined by immunostaining for lysozyme and mucin 2, respectively, and autophagy was examined by microtubule-associated proteins 1A/1B light chain 3 II immunostaining and Western blot analysis. Intestinal organoids were isolated from H19-/- and control littermate mice and treated with lipopolysaccharide (LPS). RESULTS: Intestinal mucosal tissues in mice 24 hours after exposure to CLP and in patients with sepsis showed high H19 levels, associated with intestinal barrier dysfunction. Targeted deletion of the H19 gene in mice enhanced the function of Paneth and goblet cells and promoted autophagy in the small intestinal mucosa. Knockout of H19 protected Paneth and goblet cells against septic stress, preserved autophagy activation, and promoted gut barrier function after exposure to CLP. Compared with organoids from control littermate mice, intestinal organoids isolated from H19-/- mice had increased numbers of lysozyme- and mucin 2-positive cells and showed increased tolerance to LPS. Conversely, ectopic overexpression of H19 in cultured intestinal epithelial cells prevented rapamycin-induced autophagy and abolished the rapamycin-induced protection of the epithelial barrier against LPS. CONCLUSIONS: In investigations of mice, human tissues, primary organoids, and intestinal epithelial cells, we found that increased H19 inhibited the function of Paneth and goblet cells and suppressed autophagy, thus potentially contributing to barrier dysfunction in intestinal pathologies.


Subject(s)
Autophagy/genetics , Goblet Cells/pathology , Paneth Cells/pathology , RNA, Long Noncoding/metabolism , Sepsis/pathology , Animals , Autophagy/immunology , Caco-2 Cells , Disease Models, Animal , Female , Goblet Cells/immunology , Humans , Intestine, Small/cytology , Intestine, Small/immunology , Intestine, Small/pathology , Male , Mice , Mice, Knockout , Organoids , Paneth Cells/immunology , Permeability , RNA, Long Noncoding/genetics , Sepsis/immunology
15.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1016338

ABSTRACT

The defense and repair function of the gastrointestinal mucosa is of great significance for protecting the integrity of mucosa. A large number of studies have shown that the defense and repair system of gastrointestinal mucosa is a three-dimensional network system, and gut hormones participate in the whole process, thereby playing an important role for diseases related to mucosal injury. This article reviewed the roles of gut hormones in mucosal defense and repair.

16.
Curr Gastroenterol Rep ; 21(8): 35, 2019 Jul 10.
Article in English | MEDLINE | ID: mdl-31289927

ABSTRACT

PURPOSE OF REVIEW: Short-chain fatty acids (SCFAs), the main bacterial fermentation products in the hindgut of hindgut fermenters, are also present in the foregut lumen. We discuss the impact of SCFAs in the duodenal defense mechanisms and in the gastrointestinal (GI) pathogenesis. RECENT FINDINGS: Luminal SCFAs augment the duodenal mucosal defenses via release of serotonin (5-HT) and glucagon-like peptide-2 (GLP-2) from enteroendocrine cells. Released GLP-2 protects the small intestinal mucosa from nonsteroidal anti-inflammatory drug-induced enteropathy. SCFAs are also rapidly absorbed via SCFA transporters and interact with afferent and myenteric nerves. Excessive SCFA signals with 5-HT3 receptor overactivation may be implicated in the pathogenesis of irritable bowel syndrome symptoms. SCFA production exhibits diurnal rhythms with host physiological responses, suggesting that oral SCFA treatment may adjust the GI clocks. SCFAs are not only a source of energy but also signaling molecules for the local regulation of the GI tract and systemic regulation via release of gut hormones. Targeting SCFA signals may be a novel therapeutic for GI diseases and metabolic syndrome.


Subject(s)
Duodenum/metabolism , Fatty Acids, Volatile/physiology , Gastrointestinal Diseases/metabolism , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Circadian Rhythm/physiology , Duodenum/microbiology , Gastrointestinal Diseases/chemically induced , Gastrointestinal Microbiome/physiology , Humans , Intestinal Mucosa/metabolism
17.
Gastroenterology ; 157(3): 731-743, 2019 09.
Article in English | MEDLINE | ID: mdl-31103627

ABSTRACT

BACKGROUND & AIMS: Paneth cells secrete antimicrobial proteins including lysozyme via secretory autophagy as part of the mucosal protective response. The ELAV like RNA-binding protein 1 (ELAVL1, also called HuR) regulates stability and translation of messenger RNAs (mRNAs) and many aspects of mucosal physiology. We studied the posttranscriptional mechanisms by which HuR regulates Paneth cell function. METHODS: Intestinal mucosal tissues were collected from mice with intestinal epithelium (IE)-specific disruption of HuR (IE-HuR-/-), HuRfl/fl-Cre- mice (controls), and patients with inflammatory bowel diseases and analyzed by histology and immunohistochemistry. Paneth cell functions were determined by lysozyme-immunostaining assays. We isolated primary enterocytes from IE-HuR-/- and control mice and derived intestinal organoids. HuR and the chaperone CNPY3 were overexpressed from transgenes in intestinal epithelial cells (IECs) or knocked down with small interfering RNAs. We performed RNA pulldown assays to investigate interactions between HuR and its target mRNAs. RESULTS: Intestinal tissues from IE-HuR-/- mice had reduced numbers of Paneth cells, and Paneth cells had fewer lysozyme granules per cell, compared with tissues from control mice, but there were no effects on Goblet cells or enterocytes. Intestinal mucosa from patients with inflammatory bowel diseases had reduced levels of HuR and fewer Paneth cells. IE-HuR-/- mice did not have the apical distribution of TLR2 in the intestinal mucosa as observed in control mice. IECs from IE-HuR-/- mice expressed lower levels of CNPY3. Intestinal organoids from IE-HuR-/- mice were smaller and contained fewer buds compared with those generated from controls, and had fewer lysozyme-positive cells. In IECs, knockdown of HuR decreased levels of the autophagy proteins LC3-I and LC3-II, compared with control cells, and prevented rapamycin-induced autophagy. We found HuR to interact directly with the Cnpy3 mRNA coding region and increase levels of CNPY3 by increasing the stability and translation of Cnpy3 mRNA. CNPY3 bound TLR2, and cells with knockdown of CNPY3 or HuR lost membrane localization of TLR2, but increased cytoplasmic levels of TLR2. CONCLUSIONS: In studies of mice, IECs, and human tissues, we found HuR to increase expression of CNPY3 at the posttranscriptional level. CNPY3 is required for membrane localization of TLR2 and Paneth cell function.


Subject(s)
Cell Membrane/metabolism , ELAV-Like Protein 1/metabolism , Intestine, Small/metabolism , Molecular Chaperones/metabolism , Paneth Cells/metabolism , RNA Processing, Post-Transcriptional , Toll-Like Receptor 2/metabolism , Animals , Case-Control Studies , Cells, Cultured , ELAV-Like Protein 1/deficiency , ELAV-Like Protein 1/genetics , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intestine, Small/pathology , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/genetics , Paneth Cells/pathology , Protein Transport , Signal Transduction , Up-Regulation
18.
Int J Mol Sci ; 20(1)2018 Dec 21.
Article in English | MEDLINE | ID: mdl-30577574

ABSTRACT

l-Tryptophan (Trp) is known to play an important role in the health of the large intestine. However, a role of dietary Trp in the small-intestinal mucosal barrier and microbiota remains poorly understood. The present study was conducted with weaned piglets to address this issue. Postweaning piglets were fed for 4 weeks a corn- and soybean meal-based diet supplemented with 0 (Control), 0.1, 0.2, or 0.4% Trp. The small-intestinal microbiota and serum amino acids were analyzed by bacterial 16S rRNA gene-based high-throughput sequencing methods and high-performance liquid chromatography, respectively. The mRNA levels for genes involved in host defense and the abundances of tight-junction proteins in jejunum and duodenum were measured by real time-PCR and Western blot techniques, respectively. The concentrations of Trp in the serum of Trp-supplemented piglets increased in a dose-dependent manner. Compared with the control group, dietary supplementation with 0.2⁻0.4% Trp reduced the abundances of Clostridium sensu stricto and Streptococcus in the jejunum, increased the abundances of Lactobacillus and Clostridium XI (two species of bacteria that can metabolize Trp) in the jejunum, and augmented the concentrations of secretory immunoglobulin A (sIgA) as well as mRNA levels for porcine ß-defensins 2 and 3 in jejunal tissues. Moreover, dietary Trp supplementation activated the mammalian target of rapamycin signaling and increased the abundances of tight-junction proteins (zonula occludens (ZO)-1, ZO-3, and claudin-1) in jejunum and duodenum. We suggested that Trp-metabolizing bacteria in the small intestine of weaned pigs primarily mediated the beneficial effects of dietary Trp on its mucosal integrity, health, and function.


Subject(s)
Dietary Supplements , Intestinal Mucosa/metabolism , Tryptophan/metabolism , Amino Acids/blood , Animals , Animals, Newborn , Biodiversity , Gastrointestinal Microbiome , Gene Expression , Immunoglobulin A, Secretory/biosynthesis , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Permeability , Signal Transduction , Swine , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism , Tryptophan/pharmacology , Weaning , beta-Defensins/genetics , beta-Defensins/metabolism
19.
Dig Dis Sci ; 63(10): 2582-2592, 2018 10.
Article in English | MEDLINE | ID: mdl-29876779

ABSTRACT

AIM: The present study investigates the role of innate and adaptive immune system of intestinal mucosal barrier function in cirrhosis. METHODS: Forty patients with decompensated (n = 40, group A), 27 with compensated cirrhosis (n = 27, group B), and 27 controls (n = 27, group C) were subjected to duodenal biopsy. Expression of α-defensins 5 and 6 at the intestinal crypts was evaluated by immunohistochemistry and immunofluorescence. Serum endotoxin, intestinal T-intraepithelial, and lamina propria B-lymphocytes were quantified. RESULTS: Cirrhotic patients presented higher endotoxin concentrations (p < 0.0001) and diminished HD5 and HD6 expression compared to healthy controls (p = 0.000287, p = 0.000314, respectively). The diminished HD5 and HD6 expressions were also apparent among the decompensated patients compared to compensated group (p = 0.025, p = 0.041, respectively). HD5 and HD6 expressions were correlated with endotoxin levels (r = -0.790, p < 0.0001, r = - 0.777, p < 0.0001, respectively). Although intraepithelial T-lymphocytes were decreased in group A compared to group C (p = 0.002), no notable alterations between groups B and C were observed. The B-lymphocytic infiltrate did not differ among the investigated groups. CONCLUSIONS: These data demonstrate that decreased expression of antimicrobial peptides may be considered as a potential pathophysiological mechanism of intestinal barrier dysfunction in liver cirrhosis, while remodeling of gut-associated lymphoid tissue as an acquired immune response to bio-pathogens remains an open field to illuminate.


Subject(s)
Immunity, Mucosal , Liver Cirrhosis/immunology , Paneth Cells/metabolism , alpha-Defensins/metabolism , Endotoxins/blood , Female , Humans , Liver Cirrhosis/metabolism , Lymphocytes , Lymphoid Tissue/cytology , Male , Middle Aged , Prospective Studies
20.
Curr Pharm Des ; 24(18): 1936-1946, 2018.
Article in English | MEDLINE | ID: mdl-29766781

ABSTRACT

Gastrointestinal (GI) mucosal integrity is based on the balance of aggressive and protective mechanisms. Mucosal damage may occur when the injurious factors become dominant or the mucosal defensive processes are impaired. The main target of the therapy against GI mucosal injury is the reduction of aggressive factors, however, the therapeutic possibilities for stimulation of mucosal defensive processes are rather limited. This overview focuses on the gastric and intestinal mucosal protective mechanisms and discusses the main targets that increase protective processes and increase the mucosal resistance to injurious stimuli at pre-epithelial, epithelial and sub-epithelial levels.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Gastrointestinal Tract/drug effects , Protective Agents/pharmacology , Gastrointestinal Tract/immunology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...