Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Gastroenterology ; 141(3): 959-71, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21699778

ABSTRACT

BACKGROUND & AIMS: Matrix metalloproteases (MMPs) mediate pathogenesis of chronic intestinal inflammation. We characterized the role of the gelatinase (GelE), a metalloprotease from Enterococcus faecalis, in the development of colitis in mice. METHODS: Germ-free, interleukin-10-deficient (IL-10(-/-)) mice were monoassociated with the colitogenic E faecalis strain OG1RF and isogenic, GelE-mutant strains. Barrier function was determined by measuring E-cadherin expression, transepithelial electrical resistance (TER), and translocation of permeability markers in colonic epithelial cells and colon segments from IL-10(-/-) and TNF(ΔARE/Wt) mice. GelE specificity was shown with the MMP inhibitor marimastat. RESULTS: Histologic analysis (score 0-4) of E faecalis monoassociated IL-10(-/-) mice revealed a significant reduction in colonic tissue inflammation in the absence of bacteria-derived GelE. We identified cleavage sites for GelE in the sequence of recombinant mouse E-cadherin, indicating that it might be degraded by GelE. Experiments with Ussing chambers and purified GelE revealed the loss of barrier function and extracellular E-cadherin in mice susceptible to intestinal inflammation (IL-10(-/-) and TNF(ΔARE/Wt) mice) before inflammation developed. Colonic epithelial cells had reduced TER and increased translocation of permeability markers after stimulation with GelE from OG1RF or strains of E faecalis isolated from patients with Crohn's disease and ulcerative colitis. CONCLUSIONS: The metalloprotease GelE, produced by commensal strains of E faecalis, contributes to development of chronic intestinal inflammation in mice that are susceptible to intestinal inflammation (IL-10(-/-) and TNF(ΔARE/Wt) mice) by impairing epithelial barrier integrity.


Subject(s)
Cell Membrane Permeability/physiology , Colitis/physiopathology , Enterococcus faecalis/metabolism , Gelatinases/metabolism , Gram-Positive Bacterial Infections/physiopathology , Intestinal Mucosa/metabolism , Metalloproteases/metabolism , Animals , CD4-Positive T-Lymphocytes/physiology , Cadherins/metabolism , Colitis/etiology , Colitis/metabolism , Disease Models, Animal , Gram-Positive Bacterial Infections/complications , Gram-Positive Bacterial Infections/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Intestinal Mucosa/cytology , Mice , Mice, Knockout , Mice, Mutant Strains , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
2.
Mol Nutr Food Res ; 55(5): 703-13, 2011 May.
Article in English | MEDLINE | ID: mdl-21254393

ABSTRACT

SCOPE: IL-10-deficient (IL-10(-/-) ) mice are susceptible to the development of chronic intestinal inflammation in response to the colonization with commensal Enterococcus faecalis isolates. The aim of this study was to characterize the impact of a probiotic E. faecalis strain in germ-free, wild-type (WT), and disease-susceptible IL-10(-/-) mice. METHODS AND RESULTS: The probiotic E. faecalis and the colitogenic control strain OG1RF induced IL-6 and IFN-γ inducible protein-10 secretion in the murine intestinal epithelial cell line Mode K. Epithelial cell activation involved nuclear factor κ B, p38 and extracellular signal-regulated kinase 1/2-dependent pathways. Mouse embryonic fibroblasts from WT and toll-like receptor-2-deficient (TLR-2(-/-) ) mice confirmed that both E. faecalis strains trigger pro-inflammatory responses via the pattern recognition receptor TLR-2. Monoassociation of germ-free IL-10(-/-) mice with the probiotic E. faecalis strain revealed pro-inflammatory epithelial cell activation and colonic tissue pathology. The non-pathogenic nature of E. faecalis was confirmed in monoassociated WT mice. 2-DE and MALDI-TOF MS identified the ER stress chaperone Hspa5 (glucose-regulated protein 78) and 3-mercaptopyruvate sulfurtransferase as key targets in the epithelium from IL-10(-/-) and TLR-2(-/-) mice. CONCLUSION: This study shows the potential of probiotic bacteria to initiate pro-inflammatory responses in the disease-susceptible but not the normal host.


Subject(s)
Colitis/etiology , Enterococcus faecalis/physiology , Probiotics/pharmacology , Animals , Cells, Cultured , Chemokine CXCL10/biosynthesis , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Germ-Free Life , Heat-Shock Proteins/physiology , Interleukin-6/biosynthesis , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C3H , NF-kappa B/physiology , Toll-Like Receptor 2/physiology
3.
Int J Cardiol ; 149(2): 221-226, 2011 Jun 02.
Article in English | MEDLINE | ID: mdl-20207035

ABSTRACT

BACKGROUND: The radial artery (RA) is known as an atherosclerosis-prone vessel in contrast to the atherosclerosis-resistant internal thoracic artery (ITA). The purpose of the present study was to compare the gene expression profile of these arteries from the same patient in order to identify genes involved in atherogenesis or intimal hyperplasia. METHODS: Paired specimens of RA and ITA (n=6) were analyzed by histomorphometry and whole genome microarray. The microarray data underwent pathway analysis to identify biological networks. Laser microdissection (LMD) was used to identify the cellular expression of candidate genes in the intimal or medial layer of the ITA and RA. RESULTS: Histomorphometric analyses revealed a significantly higher degree of intimal hyperplasia in the RA compared to the ITA. 552 genes were differentially expressed in the ITA and RA. qRT-PCR confirmed a significant up-regulation of six anti-apoptotic genes. p21 (11.8-fold, p=0.011), CCL2 (5.4-fold, p=0.034), SOCS3 (7.2-fold, p=0.002), IER3 (4.1-fold, p=0.048), MCL-1 (2.6-fold, p=0.025) and IL-6 (17.8-fold, p=0.046) were up-regulated in the ITA. LMD confirmed that cells of the intimal layer of the ITA consistently expressed higher levels of all six candidate genes than those of the RA. CONCLUSIONS: Microarray analysis and qRT-PCR identified significantly up-regulated genes in the ITA involved in an anti-apoptotic network. LMD revealed a higher expression of all anti-apoptotic genes in the intimal area of the ITA. These genes may play an important role in protecting the intima of the ITA from developing hyperplasia and atherosclerosis.


Subject(s)
Apoptosis/genetics , Gene Regulatory Networks/physiology , Mammary Arteries/physiology , Up-Regulation/genetics , Aged , Aged, 80 and over , Apoptosis/physiology , Coronary Artery Bypass/methods , Humans , Microarray Analysis/methods , Middle Aged , Up-Regulation/physiology
4.
J Clin Gastroenterol ; 44 Suppl 1: S10-5, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20562631

ABSTRACT

BACKGROUND AND AIM: Inflammatory bowel diseases (IBD) are immune-mediated chronic diseases that are characterized by an overreaction of the intestinal immune system to the intestinal microbiota. VSL#3, a mixture of 8 different lactic acid bacteria, is a clinically relevant probiotic compound in the context of IBD, but the bacterial structures and molecular mechanisms underlying the observed protective effects are largely unknown. The intestinal epithelium plays a very important role in the maintenance of the intestinal homeostasis, as the intestinal epithelial cells (IEC) are capable of sensing, processing, and reacting upon signals from the luminal microbiota and the intestinal immune system. This immune regulatory function of the IEC is lost in IBD owing to dysregulated activation of the IEC. Thus, the aim of this study was to reveal protective mechanisms of VSL#3 on IEC function. RESULTS: In vitro, VSL#3 was found to selectively inhibit activation-induced secretion of the T-cell chemokine interferon-inducible protein (IP)-10 in IEC. Cell wall-associated proteins of VSL#3-derived Lactobacillus casei (L. casei) were identified to be the active anti-inflammatory component of VSL#3. Mechanistically, L. casei did not impair initial IP-10 protein production, but induced posttranslational degradation of IP-10 in IEC. Feeding studies in tumor necrosis factor (TNF)Delta ARE/+ mice, a mouse model for experimental ileitis, revealed that neither VSL#3 nor L. casei is capable of reducing ileal inflammation. Even preweaning feeding of VSL#3 did not prevent the development of severe ileitis in TNF Delta ARE/+ mice. In contrast, VSL#3 feeding studies in IL-10-/- mice, a model for experimental colitis, revealed that VSL#3 has local, intestinal compartment-specific protective effects on the development of inflammation. Reduced histopathologic inflammation in the cecum of IL-10-/- mice after VSL#3 treatment was found to correlate with reduced levels of IP-10 protein in primary cecal epithelial cells. CONCLUSION AND OUTLOOK: These results suggest that the inhibitory effect of VSL#3-derived L. casei on IP-10 secretion in IEC is an important probiotic mechanism that contributes to the anti-inflammatory effects of VSL#3 in specific subsets of patients with IBD. An important future aim is the identification of the active probiotic protein, which could serve as a basis for the development of new efficient therapies in the context of IBD.


Subject(s)
Bacterial Proteins/metabolism , Chemokines/metabolism , Epithelial Cells/microbiology , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/therapy , Intestinal Mucosa/microbiology , Lacticaseibacillus casei/metabolism , Probiotics , Protein Processing, Post-Translational , Animals , Chemokine CXCL10/metabolism , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/pathology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Interleukin-10/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Time Factors , Tumor Necrosis Factors/metabolism
5.
PLoS One ; 4(2): e4365, 2009.
Article in English | MEDLINE | ID: mdl-19197385

ABSTRACT

BACKGROUND: Clinical and experimental studies suggest that the probiotic mixture VSL#3 has protective activities in the context of inflammatory bowel disease (IBD). The aim of the study was to reveal bacterial strain-specific molecular mechanisms underlying the anti-inflammatory potential of VSL#3 in intestinal epithelial cells (IEC). METHODOLOGY/PRINCIPAL FINDINGS: VSL#3 inhibited TNF-induced secretion of the T-cell chemokine interferon-inducible protein (IP-10) in Mode-K cells. Lactobacillus casei (L. casei) cell surface proteins were identified as active anti-inflammatory components of VSL#3. Interestingly, L. casei failed to block TNF-induced IP-10 promoter activity or IP-10 gene transcription at the mRNA expression level but completely inhibited IP-10 protein secretion as well as IP-10-mediated T-cell transmigration. Kinetic studies, pulse-chase experiments and the use of a pharmacological inhibitor for the export machinery (brefeldin A) showed that L. casei did not impair initial IP-10 production but decreased intracellular IP-10 protein stability as a result of blocked IP-10 secretion. Although L. casei induced IP-10 ubiquitination, the inhibition of proteasomal or lysosomal degradation did not prevent the loss of intracellular IP-10. Most important for the mechanistic understanding, the inhibition of vesicular trafficking by 3-methyladenine (3-MA) inhibited IP-10 but not IL-6 expression, mimicking the inhibitory effects of L. casei. These findings suggest that L. casei impairs vesicular pathways important for the secretion of IP-10, followed by subsequent degradation of the proinflammatory chemokine. Feeding studies in TNF(DeltaARE) and IL-10(-/-) mice revealed a compartimentalized protection of VSL#3 on the development of cecal but not on ileal or colonic inflammation. Consistent with reduced tissue pathology in IL-10(-/-) mice, IP-10 protein expression was reduced in primary epithelial cells. CONCLUSIONS/SIGNIFICANCE: We demonstrate segment specific effects of probiotic intervention that correlate with reduced IP-10 protein expression in the native epithelium. Furthermore, we revealed post-translational degradation of IP-10 protein in IEC to be the molecular mechanism underlying the anti-inflammatory effect.


Subject(s)
Chemokine CXCL10/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Inflammation/microbiology , Intestines/cytology , Lacticaseibacillus casei/metabolism , Protein Processing, Post-Translational , Animals , Autophagy/drug effects , Bacterial Proteins/metabolism , Chemotaxis/drug effects , Chronic Disease , Colitis/microbiology , Colitis/pathology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Lacticaseibacillus casei/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Organ Specificity/drug effects , Probiotics/pharmacology , Protein Processing, Post-Translational/drug effects , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology
6.
J Proteome Res ; 8(3): 1183-92, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19166300

ABSTRACT

Monoassociation of germfree Interleukin 10 gene deficient (IL-10-/-) 129SvEv but not wild-type mice with Enterococcus faecalis induces severe chronic colitis. Bacterial strain-specific effects on the development of chronic intestinal inflammation are not understood. We investigated the molecular mechanisms of E. faecalis OG1RF (human clinical isolate, colitogenic) and E. faecalis ms2 (endogenous isolate from an IL-10-/- mouse) in initiating chronic experimental colitis using IL-10-/- mice. Monoassociation of IL-10-/- mice for 14 weeks revealed significant differences in colonic inflammation (3.6 +/- 0.2 and 2.4 +/- 0.6 for OG1RF and ms2, respectively) (n = 5 mice in each group) (histological scoring (0-4)). Consistent with the tissue pathology, gene expression of the pro-inflammatory chemokine interferon-gamma inducible protein-10 (IP-10) was significantly higher in intestinal epithelial cells (IEC) derived from E. faecalis OG1RF monoassociated IL-10-/- mice. We further compared the differentially E. faecalis induced colitis on the epithelial level by 2D-SDS PAGE coupled with MALDI-TOF MS. Proteome analysis identified 13 proteins which were differentially regulated during disease progression in the epithelium of E. faecalis-monoassociated IL-10-/- mice. Regulation of Alix/AIP1 protein expression and ERK1/2 phosphorylation was validated in primary IEC and epithelial cell lines, suggesting a protective role for Alix/AIP1 in the process of disease progression. Alix/AIP1 protein expression was further characterized in epithelial cell lines using siRNA-mediated knock-down. Our study demonstrates E. faecalis strain-specific induction of colitis in IL-10-/- mice after 14 weeks of monoassociation. Our study suggests that Alix/AIP1 protein expression and ERK1/2 activation are decreased in severe colitis.


Subject(s)
Calcium-Binding Proteins/metabolism , Colitis/metabolism , Enterococcus faecalis/physiology , Intestinal Mucosa/metabolism , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 3/biosynthesis , Animals , Cells, Cultured , Chronic Disease , Colitis/genetics , Colitis/microbiology , Disease Models, Animal , Enzyme Activation , Epithelial Cells/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Intestinal Mucosa/microbiology , Mice , Species Specificity
7.
J Nutr ; 138(9): 1684-91, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18716170

ABSTRACT

Monoassociations of germ-free animals with colitogenic and probiotic bacterial strains trigger intestinal epithelial cell (IEC) activation and host-derived feedback mechanisms. To characterize the impact of a single nonpathogenic bacterial strain on the intestinal epithelium in the presence of an established microbiota, we inoculated reconstituted Lacotobacillus-free (RLF) mice at 8 wk of age with Lactobacillus reuteri 100-23. Primary IEC from the small intestine of L. reuteri-inoculated and control RLF mice were isolated 2, 6, and 21 d after inoculation followed by gene expression analysis (real-time PCR; Affymetrix microarrays) as well as 2-dimensional-gel electrophoreses (2D SDS-PAGE) and peptide mass fingerprinting via matrix-assisted laser desorption/ionization time of flight MS. At d 6, gene expression of proinflammatory cytokines and chemokines including interleukin (IL)-1alpha, IL-6, interferon-gamma-inducible protein 10, and macrophage inflammatory protein 2 was transiently induced, whereas gene expression levels of regulatory proteins A20 and Toll-interacting protein decreased. In addition, 8 target proteins with changes in the steady-state protein expression levels were identified at d 2 and 6 of L. reuteri colonization. Consistent with the absence of histopathology, L. reuteri-induced activation of primary IEC returned to control levels by d 21 after inoculation of RLF mice. The capability of L. reuteri 100-23 to directly trigger epithelial cell activation was confirmed in small IEC cultures using the murine cell line Mode-K. These results clearly indicate that the intestinal epithelium is reactive toward environmental changes induced by the commensal bacterial strain L. reuteri even in the presence of an already-established microbiota. The induction of transient IEC activation may help to maintain mucosal homeostasis.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/microbiology , Intestinal Mucosa/cytology , Limosilactobacillus reuteri/physiology , Animals , Cell Line , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Gene Expression Profiling , Intestinal Mucosa/microbiology , Mice , Mice, Inbred BALB C , RNA, Messenger/metabolism , Specific Pathogen-Free Organisms
8.
Immunology ; 115(4): 441-50, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16011513

ABSTRACT

Bifidobacteria comprise a dominant microbial population group in the human intestinal tract with purported beneficial health effects on the host. In this study, we characterized the molecular mechanisms for the initial interaction of probiotic Bifidobacterium lactis strain BB12 with native and intestinal epithelial cell (IEC) lines. We showed that B. lactis-monoassociated Fisher F344 rats transiently induce phosphorylation/activation of the NF-kappaB transcriptionally active subunit RelA and the mitogen-activated protein kinase (MAPK) p38 in native IEC at day 5 after initial bacterial colonization. In addition, Interleukin 6 (IL-6) gene expression was significantly increased at day 5, demonstrating the physiological relevance of transient transcription factor activation in IEC. In contrast, Bacteroides vulgatus-monoassociated Fisher rats revealed RelA but not p38 MAPK phosphorylation and failed to trigger significant IL-6 gene expression in native IEC. Moreover, we demonstrated that B. lactis triggers NF-kappaB RelA and p38 MAPK phosphorylation in IEC lines. Adenoviral delivery of mutant IKK-beta (Ad5dnIKKbeta) and inhibition of the p38 MAPK pathway through the pharmacological inhibitor SB203580 significantly blocked B. lactis-induced IL-6 gene expression in IEC, suggesting that B. lactis triggers NF-kappaB and MAPK signaling to induce gene expression in the intestinal epithelium. Regarding the mechanisms of bacteria epithelial cell cross-talk, B. lactis-induced IL-6 gene expression was completely inhibited in TLR2 deficient mouse embryogenic fibroblasts (MEF TLR2-/-) as well as TLR2DeltaTIR transfected Mode-K cells. In conclusion, we demonstrated that probiotic bacteria transiently trigger innate signal transduction and pro-inflammatory gene expression in the intestinal epithelium at early stages of bacterial colonization.


Subject(s)
Bifidobacterium/immunology , Epithelial Cells/immunology , Animals , Cell Line , Cysteine Endopeptidases , Enzyme Inhibitors/immunology , Gene Expression/immunology , Imidazoles/immunology , Inflammation/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Intestines/immunology , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/immunology , NF-kappa B/immunology , Nuclear Proteins , Phosphorylation , Probiotics , Proteins/immunology , Pyridines/immunology , Rats , Rats, Inbred F344 , Receptors, Cell Surface/immunology , Signal Transduction/immunology , Toll-Like Receptor 2 , Toll-Like Receptors , Transcription Factor RelA , Tumor Necrosis Factor alpha-Induced Protein 3 , p38 Mitogen-Activated Protein Kinases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...