Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Nutrients ; 13(6)2021 Jun 16.
Article in English | MEDLINE | ID: mdl-34208504

ABSTRACT

The soybean allergen Gly m 4 is known to cause severe allergic reactions including anaphylaxis, unlike other Bet v 1 homologues, which induce mainly local allergic reactions. In the present study, we aimed to investigate whether the food Bet v 1 homologue Gly m 4 can be a sensitizer of the immune system. Susceptibility to gastrointestinal digestion was assessed in vitro. Transport through intestinal epithelium was estimated using the Caco-2 monolayer. Cytokine response of different immunocompetent cells was evaluated by using Caco-2/Immune cells co-culture model. Absolute levels of 48 cytokines were measured by multiplex xMAP technology. It was shown that Gly m 4 can cross the epithelial barrier with a moderate rate and then induce production of IL-4 by mature dendritic cells in vitro. Although Gly m 4 was shown to be susceptible to gastrointestinal enzymes, some of its proteolytic fragments can selectively cross the epithelial barrier and induce production of Th2-polarizing IL-5, IL-10, and IL-13, which may point at the presence of the T-cell epitope among the crossed fragments. Our current data indicate that Gly m 4 can potentially be a sensitizer of the immune system, and intercommunication between immunocompetent and epithelial cells may play a key role in the sensitization process.


Subject(s)
Antigens, Plant/pharmacology , Food Hypersensitivity/therapy , Immunization/methods , Antigens, Plant/immunology , Caco-2 Cells/drug effects , Caco-2 Cells/immunology , Chemokines/metabolism , Coculture Techniques , Cytokines/metabolism , Escherichia coli/metabolism , Food Hypersensitivity/immunology , Gas Chromatography-Mass Spectrometry , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Models, Biological , Organisms, Genetically Modified , THP-1 Cells/drug effects , THP-1 Cells/immunology
2.
Article in English | MEDLINE | ID: mdl-31475120

ABSTRACT

Enterococcus faecium is a clinically important pathogen associated with opportunistic infection and multi-drug resistance. E. faecium has been shown to produce membrane vesicles (MVs), but MV production by E. faecium under antibiotic stress conditions and the pathogenic traits thereof have yet to be determined. This study investigated the production of MVs in E. faecium ATCC 700221 cultured with sub-minimum inhibitory concentrations (MICs) of vancomycin or linezolid and determined their pathologic effects on colon epithelial Caco-2 cells. E. faecium ATCC 700221 cultured with 1/2 MIC of vancomycin or linezolid produced 3.0 and 1.5 times more MV proteins than bacteria cultured without antibiotics, respectively. Totals of 438, 461, and 513 proteins were identified in MVs from E. faecium cultured in brain heart infusion broth (MVs/BHI), BHI broth with 1/2 MIC of vancomycin (MVs/VAN), or BHI broth with 1/2 MIC of linezolid (MVs/LIN), respectively. Intact MVs/BHI induced cytotoxicity and the expression of pro-inflammatory cytokine and chemokine genes in Caco-2 cells in a dose-dependent manner, but proteinase K-treated MVs significantly suppressed these pro-inflammatory responses. MVs/LIN were more cytotoxic toward Caco-2 cells than MVs/BHI and MVs/VAN, whereas MVs/VAN stimulated more pro-inflammatory cytokine gene expression in Caco-2 cells than MVs/BHI and MVs/LIN. Overall results indicated that antibiotics modulate the biogenesis and proteomes of MVs in E. faecium at subinhibitory concentrations. MVs produced by E. faecium cultured under antibiotic stress conditions induce strong host cell responses that may contribute to the pathogenesis E. faecium.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enterococcus faecium/drug effects , Enterococcus faecium/metabolism , Epithelial Cells/metabolism , Extracellular Vesicles/metabolism , Gram-Positive Bacterial Infections/immunology , Bacterial Proteins , Caco-2 Cells/immunology , Caco-2 Cells/metabolism , Cell Survival , Chemokines/genetics , Chemokines/metabolism , Drug Resistance, Bacterial , Enterococcus faecium/pathogenicity , Gram-Positive Bacterial Infections/microbiology , Host-Pathogen Interactions , Humans , Microbial Sensitivity Tests , Proteome/drug effects , Proteome/metabolism , Stress, Physiological , Vancomycin/pharmacology , Virulence/drug effects , Virulence Factors/metabolism
3.
Microb Pathog ; 133: 103554, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31121271

ABSTRACT

We have previously shown that Listeria monocytogenes, a causative agent of listeriosis, can produce membrane vesicles (MVs) during in vitro culture. The aim of this study was to investigate the ability of MVs from L. monocytogenes cultured with or without salt stress to induce cytotoxicity and pro-inflammatory responses in colon epithelial Caco-2 cells. MVs were purified from wild-type L. monocytogenes 10403S strain and an isogenic ΔsigB mutant strain. MVs from both wild-type and ΔsigB mutant strains increased viability of Caco-2 cells regardless of salt stress. Both MVs from wild-type and ΔsigB mutant strains stimulated expression of pro-inflammatory cytokine and chemokine genes in Caco-2 cells. Expression levels of pro-inflammatory cytokine genes in cells treated with MVs from bacteria cultured without salt stress were significantly higher than those in cells treated with MVs from bacteria cultured with salt stress. However, expression levels of chemokine genes in cells treated with MVs from bacteria cultured with salt stress were significantly higher than those in cells treated with MVs from bacteria cultured without salt stress. In addition, expression levels of interleukin (IL)-1ß and IL-8 genes were partially inhibited by either lysozyme-treated MVs or ethylenediaminetetraacetic acid-treated MVs compared to those after treatment with intact MVs. Our results suggest that salt stress can affect the production of L. monocytogenes MVs, thus causing different pro-inflammatory responses in host cells.


Subject(s)
Bacterial Proteins/immunology , Caco-2 Cells/immunology , Epithelial Cells/immunology , Listeria monocytogenes/metabolism , Salt Stress/physiology , Bacterial Proteins/genetics , Cell Survival , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Sigma Factor/genetics , Sigma Factor/immunology , Stress, Physiological/physiology
4.
Microb Pathog ; 120: 79-84, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29715536

ABSTRACT

Listeria monocytogenes expresses various virulence factors enabling the invasion and multiplying in host cells, and together induces cytokines transcription. In order to explore the relationship between virulence factors of L. monocytogenes wild-type EGD-e and cellular response in human colonic epithelial cell line(Caco-2), we constructed mutant strains with in-frame deletions of critical virulence genes of inlA, inlB, hly, actA and virulence regulatory factor prfA from EGD-e, respectively. Compared with EGD-e, mutant strains showed significantly decreased invasion and apoptosis in Caco-2 cells. However, mutant strains were capable to evoke cytokines transcription of interleukin-8 (IL-8), mononuclear chemoattractant protein-1 (MCP-1), tumor necrosis factor-a (TNF-a), interleukin-1ß (IL-1ß), interleukin-6 (IL-6) and CXCL-2 production in Caco-2 cells. Interestingly, EGD-e Δhly-infected Caco-2 cells showed a significant decrease of IL-6, IL-8 and MCP-1 transcription compared with EGD-e at 1 h post-infection. Simultaneously, EGD-e ΔinlB-infected cells showed a decrease in IL-6 transcription, while EGD-e ΔactA-infected cells reflected a decrease in MCP-1 transcription. Virulence genes play a role in inflammatory transcription, but the interaction between pathogenic bacteria and the host cells predominates in inflammatory transcription. Overall, the data showed cellular response of Caco-2 cells infected with EGD-e mutant strains.


Subject(s)
Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Listeria monocytogenes/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Virulence Factors/adverse effects , Apoptosis/drug effects , Bacterial Proteins/adverse effects , Bacterial Toxins/adverse effects , Caco-2 Cells/immunology , Chemokine CCL2/metabolism , Chemokine CXCL2/metabolism , Cytokines/metabolism , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology , Humans , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Listeria monocytogenes/pathogenicity , Membrane Proteins/adverse effects , Peptide Termination Factors/adverse effects , Tumor Necrosis Factor-alpha/metabolism , Virulence/genetics , Virulence Factors/genetics
5.
J Med Chem ; 60(12): 5193-5208, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28541707

ABSTRACT

PI3Kδ plays an important role controlling immune cell function and has therefore been identified as a potential target for the treatment of immunological disorders. This article highlights our work toward the identification of a potent, selective, and efficacious PI3Kδ inhibitor. Through careful SAR, the successful replacement of a polar pyrazole group by a simple chloro or trifluoromethyl group led to improved Caco-2 permeability, reduced Caco-2 efflux, reduced hERG PC activity, and increased selectivity profile while maintaining potency in the CD69 hWB assay. The optimization of the aryl substitution then identified a 4'-CN group that improved the human/rodent correlation in microsomal metabolic stability. Our lead molecule is very potent in PK/PD assays and highly efficacious in a mouse collagen-induced arthritis model.


Subject(s)
Arthritis, Experimental/drug therapy , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Structure-Activity Relationship , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Caco-2 Cells/drug effects , Caco-2 Cells/immunology , Dogs , ERG1 Potassium Channel/metabolism , Enzyme Inhibitors/chemistry , Female , Humans , Immune System Diseases/drug therapy , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Lectins, C-Type/metabolism , Male , Mice, Inbred BALB C , Pyrazoles/chemistry , Pyrazoles/metabolism , Pyrazoles/pharmacology , Rabbits
6.
BMC Microbiol ; 17(1): 66, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28288575

ABSTRACT

BACKGROUND: Probiotic bacteria are known to modulate host immune responses against various pathogens. Recently, extracellular vesicles (EVs) have emerged as potentially important mediators of host-pathogen interactions. In this study, we explored the role of L. plantarum derived EVs in modulating host responses to vancomycin-resistant Enterococcus faecium (VRE) using both Caenorhabditis elegans and human cells. RESULTS: Our previous work has shown that probiotic conditioning C. elegans with L. acidophilus NCFM prolongs the survival of nematodes exposed to VRE. Similarly, L. plantarum WCFS1 derived extracellular vesicles (LDEVs) also significantly protected the worms against VRE infection. To dissect the molecular mechanisms of this EV-induced protection, we found that treatment of C. elegans with LDEVs significantly increased the transcription of host defense genes, cpr-1 and clec-60. Both cpr-1 and clec-60 have been previously reported to have protective roles against bacterial infections. Incubating human colon-derived Caco-2 cells with fluorescent dye-labeled LDEVs confirmed that LDEVs could be transported into the mammalian cells. Furthermore, LDEV uptake was associated with significant upregulation of CTSB, a human homologous gene of cpr-1, and REG3G, a human gene that has similar functions to clec-60. CONCLUSIONS: We have found that EVs produced from L. plantarum WCFS1 up-regulate the expression of host defense genes and provide protective effects on hosts. Using probiotic-derived EVs instead of probiotic bacteria themselves, this study provides a new direction to treat antimicrobial resistant pathogens, such as VRE.


Subject(s)
Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Host-Pathogen Interactions/immunology , Lactobacillus/metabolism , Probiotics/therapeutic use , Vancomycin-Resistant Enterococci/immunology , Vancomycin-Resistant Enterococci/pathogenicity , Animals , Caco-2 Cells/immunology , Caco-2 Cells/microbiology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/immunology , Caenorhabditis elegans/microbiology , Cell Survival , Extracellular Vesicles/ultrastructure , Gene Expression Regulation , Gram-Positive Bacterial Infections/microbiology , Humans , Lactobacillus plantarum/metabolism , Microscopy, Electron
7.
PLoS One ; 11(1): e0147630, 2016.
Article in English | MEDLINE | ID: mdl-26799658

ABSTRACT

Poultry is known to be a major reservoir of Salmonella. The use of lactic acid bacteria has become one of successful strategies to control Salmonella in poultry. The purpose of this study was to select lactic acid bacteria strains by their in vitro immunomodulatory properties for potential use as probiotics against Salmonella infection in broiler chicks. Among 101 isolated lactic acid bacteria strains, 13 strains effectively survived under acidic (pH 2.5) and bile salt (ranging from 0.1% to 1.0%) conditions, effectively inhibited growth of 6 pathogens, and adhered to Caco-2 cells. However, their in vitro immunomodulatory activities differed significantly. Finally, three strains with higher in vitro immunomodulatory properties (Lactobacillus plantarum PZ01, Lactobacillus salivarius JM32 and Pediococcus acidilactici JH231) and three strains with lower in vitro immunomodulatory activities (Enterococcus faecium JS11, Lactobacillus salivarius JK22 and Lactobacillus salivarius JM2A1) were compared for their inhibitory effects on Salmonella adhesion and invasion to Caco-2 cells in vitro and their antimicrobial effects in vivo. The former three strains inhibited Salmonella adhesion and invasion to Caco-2 cells in vitro, reduced the number of Salmonella in intestinal content, spleen and liver, reduced the levels of lipopolysaccharide-induced TNF-α factor (LITAF), IL-1ß, IL-6 and IL-12 in serum and increased the level of IL-10 in serum during a challenge study in vivo more efficiently than the latter three strains. These results suggest that in vitro immunomodulatory activities could be used as additional parameters to select more effective probiotics as feed supplements for poultry.


Subject(s)
Poultry Diseases/prevention & control , Probiotics/therapeutic use , Salmonella Infections, Animal/prevention & control , Animals , Caco-2 Cells/immunology , Chickens/immunology , Chickens/microbiology , Enterococcus faecium/immunology , Female , Humans , In Vitro Techniques , Interleukins/blood , Lactobacillus/immunology , Lactobacillus plantarum/immunology , Pediococcus/immunology , Poultry Diseases/microbiology , Tumor Necrosis Factor-alpha/blood
8.
BMC Microbiol ; 15: 172, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26303932

ABSTRACT

BACKGROUND: Breastfed human infants are predominantly colonized by bifidobacteria that thrive on human milk oligosaccharides (HMO). Two predominant species of bifidobacteria in infant feces are Bifidobacterium breve (B. breve) and Bifidobacterium longum subsp. infantis (B. infantis), both of which include avid HMO-consumer strains. Our laboratory has previously shown that B. infantis, when grown on HMO, increases adhesion to intestinal cells and increases the expression of the anti-inflammatory cytokine interleukin-10. The purpose of the current study was to investigate the effects of carbon source-glucose, lactose, or HMO-on the ability of B. breve and B. infantis to adhere to and affect the transcription of intestinal epithelial cells on a genome-wide basis. RESULTS: HMO-grown B. infantis had higher percent binding to Caco-2 cell monolayers compared to B. infantis grown on glucose or lactose. B. breve had low adhesive ability regardless of carbon source. Despite differential binding ability, both HMO-grown strains significantly differentially affected the Caco-2 transcriptome compared to their glucose or lactose grown controls. HMO-grown B. breve and B. infantis both downregulated genes in Caco-2 cells associated with chemokine activity. CONCLUSION: The choice of carbon source affects the interaction of bifidobacteria with intestinal epithelial cells. HMO-grown bifidobacteria reduce markers of inflammation, compared to glucose or lactose-grown bifidobacteria. In the future, the design of preventative or therapeutic probiotic supplements may need to include appropriately chosen prebiotics.


Subject(s)
Bacterial Adhesion , Bifidobacterium/immunology , Bifidobacterium/physiology , Caco-2 Cells/immunology , Caco-2 Cells/microbiology , Milk, Human/chemistry , Oligosaccharides/metabolism , Bifidobacterium/growth & development , Bifidobacterium/metabolism , Gene Expression Profiling , Glucose/metabolism , Humans , Lactose/metabolism , Molecular Sequence Data , Sequence Analysis, DNA
9.
J Biochem ; 153(4): 347-54, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23316083

ABSTRACT

Allergenic potential of food proteins is associated with stability to gastric and pancreatic digestive enzymes. However, much attention has not been focused on intracellular digestion of protein antigens during the passage through intestinal epithelia. We report here the degradation and survival of a bis-phosphorylated protein, ovalbumin (OVA), in the course of passage through Caco-2 cell monolayers cultured on porous membrane. SDS-PAGE in combination with phosphoprotein staining showed that OVA, which had passed through the cell layers, was almost intact in its polypeptide chain but partly dephosphorylated. By contrast, quantitative analysis using ELISA indicated that complete dephosphorylation in advance by an alkaline phosphatase markedly reduced the OVA passage. The reduced passage was restored in the presence of cathepsin inhibitors, leupeptin and pepstatin-A. Moreover, the complete dephosphorylation increased susceptibility of OVA to in vitro digestion with cathepsin B, which cleaved near an OVA phosphorylation site, Ser345. The susceptibility of OVA to lysosomal proteases may affect its passage through the intestinal epithelia, leading to determination of allergic sensitization and elicitation in egg allergy.


Subject(s)
Caco-2 Cells/immunology , Intestinal Mucosa/metabolism , Ovalbumin/immunology , Ovalbumin/metabolism , Alkaline Phosphatase , Animals , Antigens/metabolism , Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Cathepsins/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Female , Humans , Intestinal Mucosa/immunology , Leupeptins/pharmacology , Mice , Mice, Inbred Strains , Pepstatins/pharmacology , Phosphorylation/drug effects , Protease Inhibitors/pharmacology
10.
Am J Gastroenterol ; 106(11): 2029-40, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21788990

ABSTRACT

OBJECTIVES: High-mobility group box 1 (HMGB1) is a nuclear protein with functions in the regulation of transcription. In inflammatory conditions, HMGB1 is actively secreted from immune cells in the extracellular matrix, where it behaves as a proinflammatory cytokine. The aim of the present study was to investigate the role of HMGB1 in pediatric inflammatory bowel disease (IBD). METHODS: We analyzed the stools of 19 children with Crohn's disease (CD), 21 with ulcerative colitis (UC), and 13 controls. The gene/protein expression levels of HMGB1 were assessed in bioptic specimens of all children using real-time PCR and western blot assay. Finally, intracellular localization of the protein was analyzed by western blot, after separation of nuclear and cytoplasmic extracts, and by immunohistochemistry. RESULTS: HMGB1 protein levels were significantly increased (P<0.001) in the stools of patients, but were undetectable in the controls; fecal HMGB1 correlated well with fecal calprotectin levels (r: 0.77 in CD, r: 0.70 in UC; P<0.01); and mRNA and protein expression were unchanged in inflamed bioptic tissues compared with controls. However, by separately analyzing the nuclear and cytoplasmic fraction, we detected the cytoplasmic HMGB1 expression to be significantly enhanced (P<0.01) in the inflamed tissues of the patients. In addition, HMGB1 was significantly detected in 16 patients with inactive disease, whose endoscopic scores showed persisting inflammation, suggesting that it may be a sensitive marker of mucosal inflammation, although the disease is clinically inactive. CONCLUSIONS: It was shown for the first time in our study that HMGB1 is secreted by human inflamed intestinal tissues and abundantly found in the stools of IBD patients. Hence, it can be considered as a novel marker for intestinal inflammation. We can also suggest that the presence of HMGB1 in large amounts in the fecal stream of IBD patients is mainly due to active secretion of the protein stored in the nucleus rather than a "de novo" synthesis.


Subject(s)
Colitis, Ulcerative/metabolism , Crohn Disease/metabolism , Feces/chemistry , HMGB1 Protein/metabolism , Intestinal Mucosa/metabolism , Active Transport, Cell Nucleus , Adolescent , Biomarkers/analysis , Biomarkers/metabolism , Biopsy , Caco-2 Cells/immunology , Caco-2 Cells/metabolism , Cell Nucleus/metabolism , Child , Child, Preschool , Colitis, Ulcerative/pathology , Crohn Disease/pathology , Female , HMGB1 Protein/analysis , HMGB1 Protein/genetics , Humans , Infant , Interferon-alpha/immunology , Interferon-gamma/immunology , Interleukin-8/genetics , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Cell Host Microbe ; 8(5): 433-44, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-21075354

ABSTRACT

Actin-based motility is used by various pathogens for dissemination within and between cells. Yet host factors restricting this process have not been identified. Septins are GTP-binding proteins that assemble as filaments and are essential for cell division. However, their role during interphase has remained elusive. Here, we report that septin assemblies are recruited to different bacteria that polymerize actin. We observed that intracytosolic Shigella either become compartmentalized in septin cage-like structures or form actin tails. Inactivation of septin caging increases the number of Shigella with actin tails and enhances cell-to-cell spread. TNF-α, a host cytokine produced upon Shigella infection, stimulates septin caging and restricts actin tail formation and cell-to-cell spread. Finally, we show that septin cages entrap bacteria targeted to autophagy. Together, these results reveal an unsuspected mechanism of host defense that restricts dissemination of invasive pathogens.


Subject(s)
Cervix Uteri/microbiology , Colon/microbiology , Cytosol/microbiology , Host-Pathogen Interactions , Septins/metabolism , Shigella flexneri/pathogenicity , Actins/metabolism , Caco-2 Cells/immunology , Caco-2 Cells/microbiology , Caco-2 Cells/ultrastructure , Cervix Uteri/cytology , Colon/cytology , Female , HeLa Cells/immunology , HeLa Cells/microbiology , HeLa Cells/ultrastructure , Humans , Shigella flexneri/metabolism , Tumor Necrosis Factor-alpha/metabolism
12.
Inflamm Res ; 59(10): 847-54, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20383790

ABSTRACT

OBJECTIVE: We investigated the inhibitory effects of quercetin and kaempferol treatment on the suppression of immunoglobulin E (IgE)-mediated allergic responses in relation to intestinal epithelium barrier function in RBL-2H3 and Caco-2 cells. METHODS: RBL-2H3 cells as a model of intestinal mucosa mast cells were treated with flavonols followed by IgE-anti-dinitrophenyl sensitization. The extent of degranulation and the release of pro-inflammatory cytokines were measured. Caco-2 cells were stimulated with interleukin (IL)-4 or IgE-allergen with or without flavonol pretreatment and changes in the expression of CD23 mRNA and mitogen-activated protein kinase (MAPK), and chemokine release were determined. RESULTS: Flavonols inhibited the secretion of allergic mediators in RBL-2H3 cells and suppressed the CD23 mRNA expression and p38 MAPK activation in IL-4 stimulated Caco-2 cells. Flavonols also suppressed IgE-OVA induced extra signal-regulated protein kinase (ERK) activation and chemokine release. CONCLUSIONS: Quercetin and kaempferol effectively suppressed the development of IgE-mediated allergic inflammation of intestinal cell models.


Subject(s)
Antioxidants/pharmacology , Caco-2 Cells , Hypersensitivity/immunology , Immunoglobulin E/immunology , Inflammation/immunology , Kaempferols/pharmacology , Quercetin/pharmacology , Animals , Antioxidants/chemistry , Caco-2 Cells/drug effects , Caco-2 Cells/immunology , Chemokine CCL20/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Food Hypersensitivity/immunology , Humans , Interleukin-8/immunology , Intestinal Mucosa/cytology , Kaempferols/chemistry , Kaempferols/immunology , Molecular Structure , Quercetin/chemistry , Quercetin/immunology , Receptors, IgE/genetics , Receptors, IgE/immunology , p38 Mitogen-Activated Protein Kinases/immunology
13.
J Membr Biol ; 229(1): 11-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19418087

ABSTRACT

Late endocytic compartments, containing MHC class II molecules in antigen presenting cells, fuse to each other in order to deliver antigens to these molecules. We have shown previously that fusion of late endocytic compartments takes place also in hybridomas. Therefore, we investigate here whether the level of fused late endocytic compartments affects the immunostimulatory capacity of hybridomas obtained by the electrofusion of dendritic and tumor cells. The level of fused late endocytic compartments in a single hybridoma cell was assessed and samples of electrofused cells were then cocultured with autologous T cells, resulting in the priming of naïve T cells. To test the immunostimulatory capacity of hybridoma cells, T-cell-induced cytotoxicity of tumor cells was assayed. The results demonstrate that in vitro cytotoxic T cell responses are enhanced if a higher percentage of fused late endocytic compartments is present in the cell population of electrofused hybridoma cells.


Subject(s)
Caco-2 Cells/immunology , Dendritic Cells/immunology , Endocytosis/immunology , Hybridomas/immunology , Immunization/methods , Humans
14.
Gastroenterology ; 132(4): 1245-53, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17408665

ABSTRACT

BACKGROUND & AIMS: Tissue transglutaminase (tTG) autoantibodies are markers of celiac disease, and the enzyme is required for several crucial biological processes. The aim of this study was to determine whether these autoantibodies are involved in the pathogenesis of the mucosal lesion typical of celiac disease. METHODS: Using rhodamine-conjugated phalloidin staining, we evaluated whether tTG antibodies, both commercially available and cloned from patients with celiac disease, cause cytoskeletal changes in Caco-2, MCF7, and NIH 3T3 cells. We monitored cell levels of bromodeoxyuridine incorporation to determine whether tTG autoantibodies are able to induce NIH 3T3 fibroblasts and epithelial mucosal cells into S phase. RESULTS: Treatment with tTG antibodies caused a dose-dependent increase of membrane ruffling in Caco-2, MCF7, and NIH 3T3 cells. It also dose-dependently induced G(0)-synchronized NIH 3T3 fibroblasts into S phase but did not affect the rate of apoptosis. Similarly, tTG antibodies induced S-phase entry of epithelial cells in cultured intestinal biopsy specimens from patients with celiac disease. They did not affect biopsy specimens from patients without celiac disease. CONCLUSIONS: Our results suggest that tTG autoantibodies per se, by interacting with the extracellular membrane-bound transglutaminase, may play an important role in epithelial cell proliferation in celiac disease.


Subject(s)
Antibody Formation/drug effects , Autoantibodies/therapeutic use , Celiac Disease/drug therapy , Cell Proliferation/drug effects , Epithelial Cells/pathology , Intestinal Mucosa/pathology , Transglutaminases/immunology , Actins/drug effects , Actins/metabolism , Animals , Apoptosis/immunology , Biopsy , Bromodeoxyuridine , Caco-2 Cells/drug effects , Caco-2 Cells/immunology , Caco-2 Cells/pathology , Celiac Disease/immunology , Celiac Disease/pathology , Cell Cycle/drug effects , Cell Cycle/immunology , Epithelial Cells/drug effects , Epithelial Cells/immunology , GTP-Binding Proteins , Humans , In Situ Nick-End Labeling , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Mice , NIH 3T3 Cells/drug effects , NIH 3T3 Cells/immunology , NIH 3T3 Cells/pathology , Protein Glutamine gamma Glutamyltransferase 2 , Recombinant Proteins
15.
Lab Invest ; 85(7): 932-41, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15880135

ABSTRACT

Anomalies in the regulation and function of integrins have been implicated in the etiology of various pathologic conditions, including inflammatory disorders such as irritable bowel disease. Several classes of cell surface glycoproteins such as CD98 have been shown to play roles in integrins-mediated events. Here, we investigated the role of CD98 in intestinal inflammation using both in vivo and in vitro approaches. We found that in Caco2-BBE monolayers and colonic tissues, expression of CD98 was upregulated by the proinflammatory cytokine, interferon gamma (INF gamma). Furthermore, CD98 was highly upregulated in colonic tissues from mice with active colitis induced by dextran sodium sulfate (DSS), but not in DSS-treated INF gamma -/- mice. Administration of an anti-CD98 antibody worsened DSS-induced colitis in mice but had no effect on untreated control mice. Finally, we used Caco2-BBE cell monolayers to model intestinal epithelial wound healing, and found that activation of epithelial CD98 in DSS-treated monolayers inhibited monolayer reconstitution, but had no affect on untreated control monolayers. Our data collectively indicate that (i) CD98 upregulation is mediated by INF gamma during intestinal inflammation and (ii) activation of epithelial CD98 protein aggravates intestinal inflammation by reducing intestinal epithelial reconstitution. Overall, our data suggest that epithelial CD98 plays an important role in the perpetuation of intestinal inflammation.


Subject(s)
Caco-2 Cells/metabolism , Colitis/metabolism , Colon/metabolism , Fusion Regulatory Protein-1/biosynthesis , Intestinal Mucosa/metabolism , Up-Regulation , Animals , Antibodies, Blocking/pharmacology , Caco-2 Cells/drug effects , Caco-2 Cells/immunology , Cell Adhesion/drug effects , Colitis/chemically induced , Colitis/pathology , Colon/drug effects , Colon/pathology , Dextran Sulfate/pharmacology , Disease Models, Animal , Drug Therapy, Combination , Female , Fluorescent Antibody Technique, Indirect , Fusion Regulatory Protein-1/genetics , Fusion Regulatory Protein-1/immunology , Humans , Injections, Intraperitoneal , Interferon-gamma/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Recombinant Proteins , Wound Healing/drug effects
16.
FEMS Immunol Med Microbiol ; 41(3): 227-35, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15196572

ABSTRACT

Intestinal epithelial cells (IEC) are important in initiation and regulation of immune responses against numerous foreign substances including food, microorganisms and their metabolites in the intestine. Since the responses of IEC against yeasts have not yet been well understood, we investigated the effects of Candida albicans, Saccharomyces cerevisiae, and their cell wall components on interleukin-8 (IL-8) secretion by the IEC-like Caco-2 cells. Live cells of both yeast species stimulated Caco-2 cells to produce IL-8 only in the presence of butyric acid, which is a metabolite produced by intestinal bacteria. S. cerevisiae zymosan and glucan also enhanced IL-8 secretion. Treatment of Caco-2 cells with butyric acid increased the expression of mRNAs coding for Toll-like receptor 1 (TLR1), TLR6 and dectin-1, which recognize zymosan. C. albicans induced more IL-8 secretion and also decreased transepithelial electrical resistance more rapidly than S. cerevisiae. These results suggest that both yeasts in the intestine stimulate the host's mucosal immune systems by interacting with IEC.


Subject(s)
Butyric Acid/pharmacology , Candida albicans/pathogenicity , Interleukin-8/biosynthesis , Intestines/drug effects , Intestines/immunology , Saccharomyces cerevisiae/pathogenicity , Caco-2 Cells/immunology , Caco-2 Cells/microbiology , Colony Count, Microbial , Humans , Intestines/cytology , Intestines/microbiology , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
Parasitology ; 123(Pt 3): 225-33, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11578086

ABSTRACT

The initial attachment of Toxoplasma tachyzoites to the target host cell is an important event in the life-cycle of the parasite and a critical stage in infection. Previous studies have shown that polyclonal antibodies directed against the major surface antigen of Toxoplasma gondii (SAG1) inhibit the infection of enterocyte cell lines. Here, we demonstrate that antibodies raised against a central peptide (V41T) of SAG1 and the SAGI protein itself are able to inhibit the infection of various cell lines by the tachyzoites. Antibodies directed against SAG1 peptides were used to define a site on the SAGI antigen that interacts with the host cell. The epitope carried by V41T was identified on the tachyzoite surface by immunofluorescence. The peptide sequence seems to be conserved in all the members of the SAGI Related Sequence family (SRS). Using undifferentiated and differentiated Caco-2 cells, we found that tachyzoites enter preferentially via the basolateral side of the cell. These findings highlight the role of the SRS family members in the mediation of host cell invasion.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Enterocytes/parasitology , Protozoan Proteins/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Amino Acid Sequence , Animals , Caco-2 Cells/immunology , Caco-2 Cells/parasitology , Cells, Cultured , Enterocytes/immunology , Epitopes , Humans , Mice , Mice, Inbred CBA , Microscopy, Fluorescence , Molecular Sequence Data , Protozoan Proteins/physiology , Rats , Toxoplasma/physiology , Toxoplasmosis/parasitology
18.
Gut ; 49(1): 47-55, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11413110

ABSTRACT

BACKGROUND: Escherichia coli is part of the normal gastrointestinal microflora which exerts a barrier effect against enteropathogens. Several E coli strains develop a protective effect against other Enterobacteriaceae. AIMS: Two E coli strains, EM0, a human faecal strain, and JM105 K-12 were tested for their ability to prevent in vivo and in vitro infection by Salmonella typhimurium C5. METHODS: Inhibition of C5 cell invasion by E coli was investigated in vitro using Caco-2/TC7 cells. The protective effect of E coli was examined in vivo in germfree or conventional C3H/He/Oujco mice orally infected by the lethal strain C5. RESULTS: EMO expresses haemolysin and cytotoxic necrotising factor in vitro. In vitro, the two strains did not prevent the growth of C5 by secreted microcins or modified cell invasion of C5. In vivo, establishment of EM0 or JM105 in the gut of germfree mice resulted in a significant increase in the number of surviving mice: 11/12 and 9/12, respectively, at 58 days after infection (2x10(6)/mouse) versus 0/12 in control germfree group at 13 days after infection. Colonisation level and translocation rate of C5 were significantly reduced during the three days after infection. In contrast, no reduction in faecal C5 excretion was observed in C5 infected conventional mice (1x10(8)/mouse) receiving the EM0 or JM105 cultures daily. CONCLUSIONS: Establishment of E coli strains, which do not display antimicrobial activity, protects germfree mice against infection and delays the establishment of C5 in the gut. Possible mechanisms of defence are discussed.


Subject(s)
Antibiosis/physiology , Escherichia coli Infections/immunology , Escherichia coli Proteins , Germ-Free Life/immunology , Salmonella Infections/immunology , Salmonella typhimurium/pathogenicity , Analysis of Variance , Animals , Bacterial Toxins/analysis , Caco-2 Cells/immunology , Cells, Cultured , Cytotoxins/analysis , Female , Hemolysin Proteins/analysis , Humans , Mice , Mice, Inbred C3H , Polymerase Chain Reaction/methods
19.
J Immunol ; 166(12): 7290-9, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11390479

ABSTRACT

It is becoming increasingly clear that epigenetic silencing of gene transcription plays a critical role in the regulation of gene expression in many biological processes. Tight regulation of immunomodulatory substances that are important for the initiation of the inflammatory cascade, such as chemoattractive cytokines, is essential to prevent initiation of unrestrained immune activation. Using the Caco-2 intestinal cell line as a model, we reveal two distinctly different mechanisms by which the gene for the neutrophil chemoattractive cytokine IL-8 is silenced. Nuclear run-on studies, as well as stably transfected reporter and marked minigene constructs, demonstrate that cellular differentiation inhibits immune-activated transcription of the IL-8 gene, a mechanism that is dependent on histone deacetylase activity. Unexpectedly, this silencing mechanism does not involve previously described regulatory elements in the IL-8 promoter but rather cis-acting regions located at a distance from the IL-8 gene locus. Genomic elements distant to the immediate IL-8 locus are also required to silence aberrant basal transcriptional activity of the IL-8 promoter in the absence of immune activation. However, in this case, silencing occurs in a histone deacetylase-independent fashion. These findings were confirmed in transgenic mice in which, in the absence of these elements, aberrant IL-8 gene activity was present primarily in the intestinal tract. Epigenetic silencing of cytokine gene transcription through distant genomic elements is an important level of gene regulation that may be relevant to the pathogenesis of immunologic disease states.


Subject(s)
Caco-2 Cells/immunology , Caco-2 Cells/metabolism , Interleukin-8/antagonists & inhibitors , Interleukin-8/genetics , Transcription, Genetic/immunology , Animals , Base Composition/genetics , Base Composition/immunology , Caco-2 Cells/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Clone Cells , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Female , Genes, Immediate-Early/immunology , Genetic Markers , Histone Deacetylases/physiology , Humans , Interleukin-1/physiology , Interleukin-8/biosynthesis , Interleukin-8/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic/immunology , Transcriptional Activation/genetics , Transcriptional Activation/immunology , Transfection , Transgenes/immunology
20.
Br J Nutr ; 85(4): 483-90, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11348563

ABSTRACT

The trend towards an increased consumption of minimally processed plant food results in a higher intake of non-nutritive compounds such as lectins. Lectins are typically globular proteins that are resistant to digestion in the gastrointestinal tract. They affect the integrity of the intestinal epithelium and the absorption of dietary antigens, and induce the release of allergic mediators from mast cells in vitro. Based on this information we have studied whether dietary wheat germ agglutinin (WGA) could be involved in triggering food allergies. Brown Norway rats were immunized intraperitoneally using ovalbumin (OVA; 10 microg/rat) and 10 d later treated for five consecutive days with WGA (10 mg/rat per d) administered intragastrically. Rats were then orally challenged with OVA (100 microg/rat) 1 h after the last WGA application, and blood was collected 4 h later. Immunological responses (anti-OVA immunoglobulins E and G, rat mast cell protease II, interferon-gamma and lymphocyte proliferation) were measured and lymphocyte subpopulations were determined. In immunized rats WGA treatment resulted in increased serum rat mast cell protease II concentrations (pre-challenge 0.26 (SE 0.08) microg/ml, post-challenge 0.49 (SE 0.09) microg/ml; P < 0.01) 4 h after the OVA challenge. After 5 d serum concentrations of anti-OVA immunoglobulin E were significantly increased only in the immunized controls (absorbance at 405 nm on days 14 and 19 was 0.09 (SE 0.008) and 0.24 (SE 0.046) respectively; P = 0.02), while in WGA-treated rats no significant increase was seen (0.08 (SE 0.004) and 0.15 (SE 0.037 respectively; P = 0.14). CD4+ : CD8+ T lymphocytes in the spleen was significantly increased at this time (OVA 1.1 (SD 0.2), 1.4 (sd 0.1), P < 0.05). The treatment did not impair the proliferation and interferon-gamma production of mesenteric lymphocytes. In conclusion, these data suggest that high dietary intake of lectins such as WGA may affect the allergic response towards oral antigens in the gut-associated lymphoid tissue.


Subject(s)
Diet , Food Hypersensitivity/immunology , Immune Tolerance/immunology , Ovalbumin/immunology , Wheat Germ Agglutinins/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Caco-2 Cells/immunology , Cell Culture Techniques , Humans , Immunoglobulin E/biosynthesis , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Male , Mast Cells/enzymology , Mast Cells/immunology , Mesentery/immunology , Metalloendopeptidases/blood , Rats , Rats, Inbred BN
SELECTION OF CITATIONS
SEARCH DETAIL
...