Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
EMBO Mol Med ; 14(6): e14121, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35491615

ABSTRACT

The gut has a specific vascular barrier that controls trafficking of antigens and microbiota into the bloodstream. However, the molecular mechanisms regulating the maintenance of this vascular barrier remain elusive. Here, we identified Caspase-8 as a pro-survival factor in mature intestinal endothelial cells that is required to actively maintain vascular homeostasis in the small intestine in an organ-specific manner. In particular, we find that deletion of Caspase-8 in endothelial cells results in small intestinal hemorrhages and bowel inflammation, while all other organs remained unaffected. We also show that Caspase-8 seems to be particularly needed in lymphatic endothelial cells to maintain gut homeostasis. Our work demonstrates that endothelial cell dysfunction, leading to the breakdown of the gut-vascular barrier, is an active driver of chronic small intestinal inflammation, highlighting the role of the intestinal vasculature as a safeguard of organ function.


Subject(s)
Caspase 8 , Endothelial Cells , Intestinal Mucosa , Animals , Caspase 8/metabolism , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Enteritis/enzymology , Enteritis/pathology , Homeostasis , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/enzymology , Intestine, Small/pathology , Mice
2.
Int J Mol Sci ; 22(14)2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34299188

ABSTRACT

Na-K-ATPase provides a favorable transcellular Na gradient required for the functioning of Na-dependent nutrient transporters in intestinal epithelial cells. The primary metabolite for enterocytes is glutamine, which is absorbed via Na-glutamine co-transporter (SN2; SLC38A5) in intestinal crypt cells. SN2 activity is stimulated during chronic intestinal inflammation, at least in part, secondarily to the stimulation of Na-K-ATPase activity. Leukotriene D4 (LTD4) is known to be elevated in the mucosa during chronic enteritis, but the way in which it may regulate Na-K-ATPase is not known. In an in vitro model of rat intestinal epithelial cells (IEC-18), Na-K-ATPase activity was significantly stimulated by LTD4. As LTD4 mediates its action via Ca-dependent protein kinase C (PKC), Ca levels were measured and were found to be increased. Phorbol 12-myristate 13-acetate (PMA), an activator of PKC, also mediated stimulation of Na-K-ATPase like LTD4, while BAPTA-AM (Ca chelator) and calphostin-C (Cal-C; PKC inhibitor) prevented the stimulation of Na-K-ATPase activity. LTD4 caused a significant increase in mRNA and plasma membrane protein expression of Na-K-ATPase α1 and ß1 subunits, which was prevented by calphostin-C. These data demonstrate that LTD4 stimulates Na-K-ATPase in intestinal crypt cells secondarily to the transcriptional increase of Na-K-ATPase α1 and ß1 subunits, mediated via the Ca-activated PKC pathway.


Subject(s)
Calcium/metabolism , Enteritis/enzymology , Epithelial Cells/enzymology , Intestines/enzymology , Leukotriene D4/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Cell Survival/physiology , Cells, Cultured , Enteritis/drug therapy , Enteritis/pathology , Enzyme Activation , Epithelial Cells/drug effects , Intestines/drug effects , Protein Kinase C/metabolism , Rats
3.
Vet Pathol ; 55(1): 98-107, 2018 01.
Article in English | MEDLINE | ID: mdl-29254476

ABSTRACT

Molecular clonality analysis of T-cell receptor (TCR) genes for diagnosing T-cell lymphoma is widely used in veterinary medicine. However, differentiating chronic enteritis (CE) from intestinal lymphoma is challenging because of the incompatibility between histopathologic and clonality analysis results. On the basis of findings that canine intestinal T-cell lymphoma and celiac disease share some common features, we conducted serologic examinations in combination with histopathologic and T-cell receptor clonality analyses in 48 dogs diagnosed with either CE or intestinal lymphoma. Immunoglobulin A (IgA) and immunoglobulin G (IgG) antibodies against gliadin and tissue transglutaminase (tTG) were quantitatively measured using ELISA. The conditions were classified according to the histopathologic diagnosis, clonality analysis, and combined histopathologic/clonality analysis. Histopathologic analysis showed that dogs with intestinal lymphoma were likely to have high levels of serum IgA antibodies against gliadin and tTG, and serum IgG antibodies against tTG. No correlation between the diagnosed groups and control group was observed in the results of the clonality analysis and histopathologic/clonality analysis. It is interesting that dogs with intestinal lymphoma had a higher serum IgA titer against gliadin and tTG than did dogs with CE. These results suggest an association between repetitive inflammatory stimulation by gliadin peptides and subsequent intestinal lymphoma in dogs.


Subject(s)
Dog Diseases/immunology , Enteritis/veterinary , GTP-Binding Proteins/immunology , Gliadin/immunology , Immunoglobulin A/immunology , Intestinal Neoplasms/veterinary , Lymphoma, T-Cell/veterinary , Transglutaminases/immunology , Animals , Blotting, Western/veterinary , Chronic Disease/veterinary , Diagnosis, Differential , Dog Diseases/diagnosis , Dog Diseases/enzymology , Dog Diseases/pathology , Dogs , Enteritis/enzymology , Enteritis/immunology , Enteritis/pathology , Enzyme-Linked Immunosorbent Assay/veterinary , Female , Immunoglobulin G/immunology , Intestinal Neoplasms/enzymology , Intestinal Neoplasms/immunology , Intestinal Neoplasms/pathology , Lymphoma, T-Cell/diagnosis , Lymphoma, T-Cell/enzymology , Lymphoma, T-Cell/immunology , Male , Microscopy, Fluorescence/veterinary , Polymerase Chain Reaction/veterinary , Protein Glutamine gamma Glutamyltransferase 2
4.
Am J Vet Res ; 77(7): 721-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27347825

ABSTRACT

OBJECTIVE To compare expression, activity, and fecal concentration of intestinal alkaline phosphatase (IAP) between healthy dogs and dogs with chronic enteropathy (CE). ANIMALS 9 healthy university-owned Beagles and 109 healthy client-owned dogs (controls) and 28 dogs with CE (cases). PROCEDURES Cases were defined as dogs with persistent (> 3 weeks) gastrointestinal signs that failed to respond to antimicrobials and anti-inflammatory doses of prednisolone or dietary trials, did not have mechanical gastrointestinal abnormalities as determined by abdominal radiography and ultrasonography, and had a diagnosis of lymphoplasmacytic enteritis or eosinophilic gastroenteritis on histologic examination of biopsy specimens. Duodenal and colonic mucosa biopsy specimens were obtained from the 9 university-owned Beagles and all cases for histologic examination and determination of IAP expression (by real-time quantitative PCR assay) and activity (by enzyme histochemical analysis). Fecal samples were obtained from all dogs for determination of fecal IAP concentration by a quantitative enzyme reaction assay. RESULTS For dogs evaluated, IAP expression and activity were localized at the luminal side of epithelial cells in the mucosa and intestinal crypts, although both were greater in the duodenum than in the colon. Active IAP was detected in the feces of all dogs. Intestinal alkaline phosphatase expression and activity were lower for cases than for controls, and fecal IAP concentration for dogs with moderate and severe CE was lower than that for dogs with mild CE. CONCLUSIONS AND CLINICAL RELEVANCE Results indicated that dogs with CE had impaired IAP expression and activity. Additional research is necessary to elucidate the role of IAP in the pathogenesis of CE.


Subject(s)
Alkaline Phosphatase/metabolism , Dog Diseases/enzymology , Enteritis/veterinary , Eosinophilia/veterinary , Gastritis/veterinary , Alkaline Phosphatase/biosynthesis , Animals , Colon/enzymology , Colon/pathology , Dog Diseases/pathology , Dogs , Duodenum/enzymology , Duodenum/pathology , Enteritis/enzymology , Enteritis/pathology , Eosinophilia/enzymology , Eosinophilia/pathology , Feces/enzymology , Female , Gastritis/enzymology , Gastritis/pathology , Male , Real-Time Polymerase Chain Reaction
5.
Jpn J Antibiot ; 69(5): 343-346, 2016 Oct.
Article in English | MEDLINE | ID: mdl-30226947

ABSTRACT

This clinical case report concerns a pediatric patient with acute enteritis caused by multi-drug resistant Salmonella enterica serovar Blockley (Salmonella Blockley). A 3-year-old boy presented to our emergency room with a 5-day history of fever, abdominal pain, and bloody diarrhea. Stool culture tested positive for a Salmonella species, while the blood culture was negative. The patient was successfully treated with an oral antibiotic regimen of fosfomycin. The stool isolate was found to be resistant to multiple drugs, including cefpodoxime, cefotaxime, ceftazidime, and aztreonam, and was confirmed to be a CTX-M-15 extended-spectrum ß-lactamase (ESBL)-producing strain of Salmonella Blockley. This is the first report of a pediatric patient in Japan with acute enteritis caused by a CTX-M-15 ESBL- producing strain of Salmonella Blockley.


Subject(s)
Enteritis/mortality , Salmonella Infections , beta-Lactamases/biosynthesis , Acute Disease , Child, Preschool , Drug Resistance, Multiple, Bacterial , Enteritis/enzymology , Humans , Male , Salmonella
6.
World J Gastroenterol ; 21(35): 10072-9, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26401072

ABSTRACT

AIM: To investigate local corticosterone production and angiotensin-I converting enzyme (ACE) protein expression and their interaction in healthy and inflamed intestine. METHODS: Acute intestinal inflammation was induced to six weeks old male Balb/c mice by administration of either 3% or 5% dextran sodium sulfate (DSS) in drinking water for 7 d (n = 12 in each group). Healthy controls (n = 12) were given tap water. Corticosterone production and ACE protein shedding were measured from ex vivo incubates of the small and large intestine using EIA and ELISA, respectively. Morphological changes of the intestinal wall were assessed in hematoxylin-eosin stained tissue preparations of jejunum and distal colon. Effects of angiotensin II, captopril and metyrapone on corticosterone production was assessed by incubating pieces of small intestine of healthy mice in the presence of 0.1, 1 or 10 µmol/L angiotensin II, 1, 10 or 100 µmol/L captopril or 1, 10 or 100 µmol/L metyrapone solutions and measuring corticosterone released to the incubation buffer after 90 min (n = 5 in each group). RESULTS: Both concentrations of DSS induced inflammation and morphological changes in large intestines but not in small intestines. Changes were observed as distortions of the crypt structure, mucosal erosion, immune cell infiltration to the mucosa and submucosal edema. Ex vivo corticosterone production (2.9 ± 1.0 ng/mL vs 2.0 ± 0.8 ng/mL, P = 0.034) and ACE shedding (269.2 ± 97.1 ng/mL vs 175.7 ± 52.2 ng/mL, P = 0.016) were increased in small intestines in 3% DSS group compared to the controls. In large intestine, corticosterone production was increased compared to the controls in both 3% DSS (229 ± 81 pg/mL vs 158 ± 30 pg/mL, P = 0.017) and 5% DSS groups (366 ± 163 pg/mL vs 158 ± 30 pg/mL, P = 0.002). Large intestine ACE shedding was increased in 5% DSS group (41.5 ± 9.0 ng/mL vs 20.9 ± 5.2 ng/mL, P = 0.034). Angiotensin II treatment augmented corticosterone production in small intestine at concentration of 10 µmol/L (0.97 ± 0.21 ng/mg protein vs 0.40 ± 0.09 ng/mg protein, P = 0.036). CONCLUSION: Intestinal ACE shedding is increased by DSS-induced intestinal inflammation and parallels local corticosterone production. ACE product angiotensin II stimulates corticosterone formation in healthy intestine.


Subject(s)
Colitis/enzymology , Colon/enzymology , Corticosterone/metabolism , Enteritis/enzymology , Intestinal Mucosa/enzymology , Jejunal Diseases/enzymology , Jejunum/enzymology , Peptidyl-Dipeptidase A/metabolism , Angiotensin II/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Captopril/pharmacology , Colitis/chemically induced , Colitis/pathology , Colon/drug effects , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Dose-Response Relationship, Drug , Enteritis/chemically induced , Enteritis/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Jejunal Diseases/chemically induced , Jejunal Diseases/pathology , Jejunum/drug effects , Jejunum/pathology , Male , Mice, Inbred BALB C , Pyridines/pharmacology
7.
Dig Dis Sci ; 60(11): 3264-70, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26138652

ABSTRACT

BACKGROUND AND AIMS: PepT1 can transport bacterial oligopeptide products and induce intestinal inflammation. Our aim was to investigate the mechanism of the small intestine injury induced by bacterial oligopeptide product muramyl dipeptide (MDP) which is transported by PepT1. METHODS: We perfused the jejunum with a solution with or without MDP, or with a solution of MDP + Gly-Gly and explored the degree of inflammation to determine the role of PepT1-Nod2 signaling pathway in small intestine mucosa. RESULTS: MDP perfusion induced inflammatory cell accumulation and intestinal damage, accompanied by an increase in mucosal Nod2 and Rip2 transcript expression. NFκB activity and inflammatory cytokine expression, including serum levels of TNF-α, IL-1ß, and IL-6, increased in the MDP group compared to the controls; these effects were reversed by perfusion of the nutritional dipeptide Gly-Gly. CONCLUSION: MDP can be transported through PepT1, causing inflammatory damage in the rat small intestine. Nod2-Rip2-NFκB signaling involved in the small intestinal inflammatory injury caused by MDP which is transported through PepT1.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/toxicity , Enteritis/chemically induced , Intestinal Mucosa/drug effects , Jejunum/drug effects , Nod2 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Symporters/metabolism , Acetylmuramyl-Alanyl-Isoglutamine/metabolism , Animals , Cytokines/metabolism , Enteritis/enzymology , Enteritis/pathology , Glycylglycine/pharmacology , Inflammation Mediators/metabolism , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Jejunum/enzymology , Jejunum/pathology , Male , NF-kappa B/metabolism , Peptide Transporter 1 , Rats, Sprague-Dawley , Signal Transduction/drug effects
9.
Eur Rev Med Pharmacol Sci ; 18(3): 344-51, 2014.
Article in English | MEDLINE | ID: mdl-24563433

ABSTRACT

AIM: Enterobacterial translocation into the gut mucosa is the first step required for activation of neutrophils and inducible nitric oxide synthase (iNOS), involved in the pathogenesis of indomethacin-induced intestinal lesions. Rifaximin may limit NSAID-associated intestinal damage by decreasing the bacterial load. We aimed to study the effect of rifaximin on indomethacin-induced intestinal damage in guinea-pigs. MATERIALS AND METHODS: Twenty-four guinea pigs, equally divided in four interventional groups (A-D), received indomethacin, given orally once daily (30 mg/kg) for three consecutive days. In groups B, C, D different doses of rifaximin (50 mg/kg, 100 mg/kg and 200 mg/kg) were given orally two hours before indometachin administration. Semi-quantitative grades were measure for gross findings, degenerative lesions, neutrophils and eosinophils infiltrates and iNOS immunopositivity. Statistical comparisons used Mann Whitney Test, with a Bonferroni correction for alpha (p ≤ 0.016). RESULTS: Statistical analysis of graded gross findings, microscopic degenerative lesions, endothelium damage and iNOS immunopositivity found no difference between A and B groups. Significant fewer gross findings (U = 3, p = 0.015), microscopic degenerative lesions (U = 2, p = 0.008) and lower grades for iNOS immunopositivity (U = 0, p = 0.002) were found in group C compared with group A. In group D, significant lower grades for iNOS immunopositivity were obtained (U = 0, p = 0.002) compared with group A and fewer degenerative lesions without reaching statistical significance (U = 4, p = 0.026). CONCLUSIONS: 100 mg/kg of rifaximin proved efficient in preventing gut degenerative lesions induced by indomethacin in a guinea pig model, the iNOS activity being significantly decreased.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Enteritis/drug therapy , Gastrointestinal Agents/therapeutic use , Indomethacin/adverse effects , Rifamycins/therapeutic use , Animals , Enteritis/enzymology , Enteritis/microbiology , Enteritis/pathology , Female , Gastrointestinal Agents/administration & dosage , Guinea Pigs , Immunohistochemistry , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Neutrophil Infiltration/drug effects , Nitric Oxide Synthase Type II/biosynthesis , Rifamycins/administration & dosage , Rifaximin
10.
Gut ; 63(1): 96-104, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23268370

ABSTRACT

OBJECTIVE: Cryptogenic multifocal ulcerating stenosing enteritis (CMUSE) is an extremely rare, but devastating, disease of unknown aetiology. We investigated the genetic basis of this autosomal recessive condition in a pair of affected siblings who have 40-year histories of catastrophic gastrointestinal and extraintestinal disease. DESIGN: Genome-wide single-nucleotide polymorphism homozygosity mapping in the two affected family members combined with whole-exome sequencing of one affected sibling. This was followed by confirmatory Sanger sequencing of the likely disease-causing sequence variant and functional studies in affected and unaffected family members. RESULTS: Insertion/deletion variation analysis revealed the presence of a homozygous 4 bp deletion (g.155574_77delGTAA) in the PLA2G4A gene, located in the splice donor site directly after exon 17 (the penultimate exon) of the gene in both affected siblings. This introduces a frameshift of 10 amino acids before a premature stop codon (p.V707fsX10), which is predicted to result in the loss of 43 amino acids (residues 707-749) at the C-terminus of cytosolic phospholipase A2-α (cPLA(2)α). cPLA(2)α protein expression was undetectable in the gut of both siblings, with platelet aggregation and thromboxane A(2) production, as functional assays for cPLA(2)α activity, grossly impaired. CONCLUSIONS: We have identified mutations in PLA2G4A as a cause of CMUSE in two affected siblings. Further studies are needed to determine if mutations in this gene are also responsible for disease of a similar phenotype in other cases.


Subject(s)
Enteritis/genetics , Group IV Phospholipases A2/genetics , Homozygote , Peptic Ulcer/genetics , Sequence Deletion , Adult , Biomarkers/metabolism , Blotting, Western , Case-Control Studies , Codon, Nonsense , Enteritis/complications , Enteritis/enzymology , Female , Fluorescent Antibody Technique , Frameshift Mutation , Genetic Markers , Group IV Phospholipases A2/metabolism , Humans , Intestinal Obstruction/etiology , Male , Peptic Ulcer/complications , Peptic Ulcer/enzymology , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Siblings
11.
J Gastroenterol Hepatol ; 28(4): 632-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23216607

ABSTRACT

BACKGROUND AND AIM: Although non-steroidal anti-inflammatory drugs can induce intestinal injury, the mechanisms are not fully understood, and treatment has yet to be established. Heme oxygenase-1 (HO-1) has recently gained attention for anti-inflammatory and cytoprotective effects. This study aimed to investigate the effects of hemin, an HO-1 inducer, on indomethacin-induced enteritis in mice. METHODS: Enteritis was induced by single subcutaneous administration of indomethacin (10 mg/kg) in male C57BL/6 mice. Hemin (30 mg/kg) was administered by intraperitoneal administration 6 h before indomethacin administration. Mice were randomly divided into four groups: (i) sham + vehicle; (ii) sham + hemin; (iii) indomethacin + vehicle; or (iv) indomethacin + hemin. Enteritis was evaluated by measuring ulcerative lesions. Myeloperoxidase activity was measured as an index of neutrophil accumulation. The mRNA expression of inflammatory cytokines and chemokines, such as tumor necrosis factor-α, monocyte chemoattractant protein-1, macrophage inflammatory protein-1α, and keratinocyte chemoattractant, were analyzed by real-time polymerase chain reaction. RESULTS: The area of ulcerative lesions, myeloperoxidase activity, and mRNA expression of inflammatory cytokines and chemokines were significantly increased in mice administrated with indomethacin compared with vehicle-treated sham mice. Development of intestinal lesions, increased levels of myeloperoxidase activities, and mRNA expressions of inflammatory cytokines and chemokines were significantly suppressed in mice treated with hemin compared with vehicle-treated mice. Protective effects of hemin were reversed by co-administration of tin protoporphyrin, an HO-1 inhibitor. CONCLUSIONS: Induction of HO-1 by hemin inhibits indomethacin-induced intestinal injury through upregulation of HO-1. Pharmacological induction of HO-1 may offer a novel therapeutic strategy to prevent indomethacin-induced small intestinal injury.


Subject(s)
Enteritis/prevention & control , Heme Oxygenase-1/metabolism , Hemin/therapeutic use , Intestine, Small/drug effects , Animals , Blotting, Western , Chemokines/genetics , Cytokines/genetics , DNA Primers/chemistry , Disease Models, Animal , Enteritis/chemically induced , Enteritis/enzymology , Enteritis/pathology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Heme Oxygenase-1/antagonists & inhibitors , Hemin/administration & dosage , Immunohistochemistry , Indomethacin/toxicity , Male , Metalloporphyrins/pharmacology , Mice , Mice, Inbred C57BL , Protoporphyrins/pharmacology , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction
12.
Nature ; 477(7364): 330-4, 2011 Jul 31.
Article in English | MEDLINE | ID: mdl-21804564

ABSTRACT

Intestinal immune homeostasis depends on a tightly regulated cross talk between commensal bacteria, mucosal immune cells and intestinal epithelial cells (IECs). Epithelial barrier disruption is considered to be a potential cause of inflammatory bowel disease; however, the mechanisms regulating intestinal epithelial integrity are poorly understood. Here we show that mice with IEC-specific knockout of FADD (FADD(IEC-KO)), an adaptor protein required for death-receptor-induced apoptosis, spontaneously developed epithelial cell necrosis, loss of Paneth cells, enteritis and severe erosive colitis. Genetic deficiency in RIP3, a critical regulator of programmed necrosis, prevented the development of spontaneous pathology in both the small intestine and colon of FADD(IEC-KO) mice, demonstrating that intestinal inflammation is triggered by RIP3-dependent death of FADD-deficient IECs. Epithelial-specific inhibition of CYLD, a deubiquitinase that regulates cellular necrosis, prevented colitis development in FADD(IEC-KO) but not in NEMO(IEC-KO) mice, showing that different mechanisms mediated death of colonic epithelial cells in these two models. In FADD(IEC-KO) mice, TNF deficiency ameliorated colon inflammation, whereas MYD88 deficiency and also elimination of the microbiota prevented colon inflammation, indicating that bacteria-mediated Toll-like-receptor signalling drives colitis by inducing the expression of TNF and other cytokines. However, neither CYLD, TNF or MYD88 deficiency nor elimination of the microbiota could prevent Paneth cell loss and enteritis in FADD(IEC-KO) mice, showing that different mechanisms drive RIP3-dependent necrosis of FADD-deficient IECs in the small and large bowel. Therefore, by inhibiting RIP3-mediated IEC necrosis, FADD preserves epithelial barrier integrity and antibacterial defence, maintains homeostasis and prevents chronic intestinal inflammation. Collectively, these results show that mechanisms preventing RIP3-mediated epithelial cell death are critical for the maintenance of intestinal homeostasis and indicate that programmed necrosis of IECs might be implicated in the pathogenesis of inflammatory bowel disease, in which Paneth cell and barrier defects are thought to contribute to intestinal inflammation.


Subject(s)
Colitis/pathology , Colon/pathology , Enteritis/pathology , Epithelial Cells/pathology , Fas-Associated Death Domain Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Chronic Disease , Colitis/enzymology , Colitis/metabolism , Colon/enzymology , Colon/metabolism , Cysteine Endopeptidases/metabolism , Deubiquitinating Enzyme CYLD , Enteritis/enzymology , Enteritis/metabolism , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Fas-Associated Death Domain Protein/deficiency , Inflammatory Bowel Diseases/enzymology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/metabolism , Metagenome/physiology , Mice , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/metabolism , Necrosis , Paneth Cells/pathology , Signal Transduction , Tumor Necrosis Factors/deficiency
13.
Am J Physiol Gastrointest Liver Physiol ; 300(6): G976-87, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21350188

ABSTRACT

Wound healing of the gastrointestinal mucosa is essential for the maintenance of gut homeostasis and integrity. Enteric glial cells play a major role in regulating intestinal barrier function, but their role in mucosal barrier repair remains unknown. The impact of conditional ablation of enteric glia on dextran sodium sulfate (DSS)-induced mucosal damage and on healing of diclofenac-induced mucosal ulcerations was evaluated in vivo in GFAP-HSVtk transgenic mice. A mechanically induced model of intestinal wound healing was developed to study glial-induced epithelial restitution. Glial-epithelial signaling mechanisms were analyzed by using pharmacological inhibitors, neutralizing antibodies, and genetically engineered intestinal epithelial cells. Enteric glial cells were shown to be abundant in the gut mucosa, where they associate closely with intestinal epithelial cells as a distinct cell population from myofibroblasts. Conditional ablation of enteric glia worsened mucosal damage after DSS treatment and significantly delayed mucosal wound healing following diclofenac-induced small intestinal enteropathy in transgenic mice. Enteric glial cells enhanced epithelial restitution and cell spreading in vitro. These enhanced repair processes were reproduced by use of glial-conditioned media, and soluble proEGF was identified as a secreted glial mediator leading to consecutive activation of epidermal growth factor receptor and focal adhesion kinase signaling pathways in intestinal epithelial cells. Our study shows that enteric glia represent a functionally important cellular component of the intestinal epithelial barrier microenvironment and that the disruption of this cellular network attenuates the mucosal healing process.


Subject(s)
Enteritis/enzymology , Epidermal Growth Factor/metabolism , Focal Adhesion Kinase 1/metabolism , Intestinal Mucosa/enzymology , Intestine, Small/enzymology , Neuroglia/enzymology , Peptic Ulcer/enzymology , Protein Precursors/metabolism , Wound Healing , Analysis of Variance , Animals , Caco-2 Cells , Cell Shape , Coculture Techniques , Culture Media, Conditioned/metabolism , Dextran Sulfate , Diclofenac , Disease Models, Animal , Enteritis/chemically induced , Enteritis/genetics , Enteritis/pathology , Epithelial Cells/enzymology , Epithelial Cells/pathology , ErbB Receptors/metabolism , Focal Adhesion Kinase 1/genetics , Glial Fibrillary Acidic Protein , Humans , Intestinal Mucosa/pathology , Intestine, Small/pathology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroglia/pathology , Paracrine Communication , Peptic Ulcer/chemically induced , Peptic Ulcer/genetics , Peptic Ulcer/pathology , Phosphorylation , RNA Interference , Rats , Signal Transduction , Simplexvirus/enzymology , Simplexvirus/genetics , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Time Factors , Transfection
14.
J Immunol ; 186(1): 423-31, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21131421

ABSTRACT

The injection of Clostridium difficile toxin A into the ileal loops caused fluid accumulation with the destruction of intestinal epithelial structure and the recruitment of neutrophils and macrophages. Concomitantly, intraileal gene expression of CX3CL1/fractalkine (FKN) and its receptor, CX3CR1, was enhanced. When treated with toxin A in a similar manner, CX3CR1-deficient (CX3CR1(-/-)) mice exhibited exaggerated fluid accumulation, histopathological alterations, and neutrophil recruitment, but not macrophage infiltration. Mice reconstituted with CX3CR1(-/-) mouse-derived bone marrow cells exhibited exacerbated toxin A-induced enteritis, indicating that the lack of the CX3CR1 gene for hematopoietic cells aggravated toxin A-induced enteritis. A heme oxygenase-1 (HO-1) inhibitor, tin-protoporphyrin-IX, markedly increased fluid accumulation in toxin A-treated wild-type mice, indicating the protective roles of HO-1 in this situation. HO-1 expression was detected mainly in F4/80-positive cells expressing CX3CR1, and CX3CR1(-/-) mice failed to increase HO-1 expression after toxin A treatment. Moreover, CX3CL1/FKN induced HO-1 gene expression by isolated lamina propria-derived macrophages or a mouse macrophage cell line, RAW264.7, through the activation of the ERK signal pathway. Thus, CX3CL1/FKN could induce CX3CR1-expressing macrophages to express HO-1, thereby ameliorating toxin A-induced enteritis.


Subject(s)
Bacterial Toxins/toxicity , Enteritis/immunology , Enteritis/prevention & control , Enterotoxins/toxicity , Gene Expression Regulation, Enzymologic/immunology , Heme Oxygenase-1/biosynthesis , MAP Kinase Signaling System/immunology , Receptors, Chemokine/physiology , Animals , Bacterial Toxins/administration & dosage , Bacterial Toxins/antagonists & inhibitors , CX3C Chemokine Receptor 1 , Cell Line , Chemokine CX3CL1/biosynthesis , Chemokine CX3CL1/genetics , Clostridioides difficile/immunology , Dose-Response Relationship, Immunologic , Enteritis/enzymology , Enterotoxins/administration & dosage , Enterotoxins/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/therapeutic use , MAP Kinase Signaling System/genetics , Macrophages/enzymology , Macrophages/immunology , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/enzymology , Mucous Membrane/immunology , Mucous Membrane/microbiology , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Up-Regulation/genetics , Up-Regulation/immunology
15.
PLoS One ; 5(9): e12969, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20885979

ABSTRACT

BACKGROUND: Intestinal mucosa barrier (IMB) dysfunction results in many notorious diseases for which there are currently few effective treatments. We studied curcumin's protective effect on IMB and examined its mechanism by using methotrexate (MTX) induced rat enteritis model and lipopolysaccharide (LPS) treated cell death model. METHODOLOGY/PRINCIPAL FINDINGS: Curcumin was intragastrically administrated from the first day, models were made for 7 days. Cells were treated with curcumin for 30 min before exposure to LPS. Rat intestinal mucosa was collected for evaluation of pathological changes. We detected the activities of D-lactate and diamine oxidase (DAO) according to previous research and measured the levels of myeloperoxidase (MPO) and superoxide dismutase (SOD) by colorimetric method. Intercellular adhesion molecule-1 (ICAM-1), tumor necrosis factor α (TNF-α) and interleukin 1ß (IL-1ß) were determined by RT-PCR and IL-10 production was determined by ELISA. We found Curcumin decreased the levels of D-lactate, DAO, MPO, ICAM-1, IL-1ß and TNF-α, but increased the levels of IL-10 and SOD in rat models. We further confirmed mitogen-activated protein kinase phosphatase-1 (MKP-1) was activated but phospho-p38 was inhibited by curcumin by western blot assay. Finally, NF-κB translocation was monitored by immunofluorescent staining. We showed that curcumin repressed I-κB and interfered with the translocation of NF-κB into nucleus. CONCLUSIONS/SIGNIFICANCE: The effect of curcumin is mediated by the MKP-1-dependent inactivation of p38 and inhibition of NF-κB-mediated transcription. Curcumin, with anti-inflammatory and anti-oxidant activities may be used as an effective reagent for protecting intestinal mucosa barrier and other related intestinal diseases.


Subject(s)
Curcumin/administration & dosage , Dual Specificity Phosphatase 1/metabolism , Enteritis/drug therapy , Intestinal Mucosa/drug effects , NF-kappa B/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Disease Models, Animal , Dual Specificity Phosphatase 1/genetics , Enteritis/enzymology , Enteritis/genetics , Enteritis/metabolism , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Mice , NF-kappa B/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases/genetics
16.
Br J Surg ; 96(5): 552-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19358174

ABSTRACT

BACKGROUND: Green tea has been shown to repair fasting-induced mucosal damage in rat intestine. The aim of this study was to elucidate the underlying mechanism. METHODS: Five groups of rats were used. Group 1 had free access to chow diet and water, and those in group 2 were fasted for 3 days. Animals in group 3 were fasted for 3 days, then were allowed drinking water for a further 7 days. Groups 4 and 5 were fasted for 3 days, then given drinking water containing green tea or vitamin E respectively for 7 days. Blood was collected for estimation of total plasma antioxidants, and jejunal samples were used for immunohistochemical analysis of superoxide dismutase (SOD), catalase and glutathione peroxidase (GPx), and for estimation of myeloperoxidase (MPO) activity. RESULTS: Use of green tea was associated with a significant increase in total plasma antioxidants (P < 0.001), and mucosal SOD (P < 0.001), catalase (P = 0.006) and GPx (P = 0.017), but a significant decrease in MPO activity (P < 0.001). Vitamin E produced similar changes, but the effects were smaller. CONCLUSION: Green tea reverses the fasting-induced damage to the intestinal mucosa by its antioxidant and anti-inflammatory effect.


Subject(s)
Antioxidants/metabolism , Enteritis/drug therapy , Fasting/metabolism , Jejunal Diseases/drug therapy , Peroxidase/metabolism , Plant Preparations/pharmacology , Tea/physiology , Animals , Catalase/metabolism , Enteritis/enzymology , Glutathione Peroxidase/metabolism , Immunohistochemistry , Intestinal Mucosa/enzymology , Jejunal Diseases/enzymology , Jejunum/enzymology , Male , Oxidative Stress , Phytotherapy , Random Allocation , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Vitamin E/pharmacology
17.
Inflamm Bowel Dis ; 15(9): 1391-6, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19322906

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) is a tryptophan catabolizing enzyme that has a number of immunoregulatory effects. It is expressed at high levels in the gastrointestinal tract, particularly in the small intestine, and has been implicated in the control of intestinal inflammation. However, its precise role in intestinal immunity is not well understood. This review will summarize the current state of knowledge about IDO function, particularly as it pertains to inflammatory responses in the gut.


Subject(s)
Enteritis/enzymology , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Intestinal Mucosa/immunology , Intestine, Small/enzymology , Humans , Immunity , T-Lymphocyte Subsets/immunology
18.
Life Sci ; 84(17-18): 615-21, 2009 Apr 24.
Article in English | MEDLINE | ID: mdl-26324992

ABSTRACT

AIMS: We established a new model of ischemic enteritis in rats and evaluated its usefulness for screening prophylactic drugs. MAIN METHODS: Male SD rats were used after 18 h of fasting. Under ether anesthesia, the superior mesenteric artery (SMA) was exposed, and a calibrated stenosis was produced by placing a needle on a blood vessel, ligating both the vessel and needle, and then removing the needle from the ligature. KEY FINDINGS: The stenosis caused severe damage on the anti-mesenteric side of the small intestine within 3 days; the severity of the damage increased with the gauge of a needle. No damage occurred in the small intestine following the stenosis with a needle of less than 21 gauge. Multiple hemorrhagic lesions occurred at an incidence of 100% when a 23-gauge needle was used. The development of enteritis was accompanied by enterobacterial invasion in the mucosa, with an up- regulation of inducible nitric oxide synthase (iNOS) expression and nitric oxide (NO) production. The ischemia-induced enteritis was significantly prevented by repeated treatment with aminoguanidine (a selective iNOS inhibitor), L-NAME (a nonselective NOS inhibitor), ampicillin, and aztreonam (a gram- negative bacterium antibiotic), but not vancomycin (a gram-positive bacterium antibiotic). SIGNIFICANCE: These results showed that a novel model of ischemic enteritis is induced in rats by stenosis of the SMA, this model may be useful for screening drugs against ischemic enteritis, and gram-negative bacteria as well as iNOS/NO are involved in the pathogenesis of enteritis in this model.


Subject(s)
Disease Models, Animal , Enteritis , Enzyme Inhibitors/pharmacology , Intestinal Mucosa , Ischemia , Mesenteric Arteries , Nitric Oxide Synthase Type II , Animals , Constriction, Pathologic , Enteritis/drug therapy , Enteritis/enzymology , Enteritis/pathology , Gene Expression Regulation, Enzymologic/drug effects , Intestinal Mucosa/blood supply , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Ischemia/drug therapy , Ischemia/enzymology , Ischemia/pathology , Male , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/biosynthesis , Rats , Up-Regulation/drug effects
19.
Dig Dis Sci ; 53(1): 123-32, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17503181

ABSTRACT

Nitric oxide has been implicated in the pathogenic mechanism of inflammatory bowel disease states. We evaluated indomethacin-induced enteropathy in rats, in relation to the expression of the inducible isoform of NO synthase (iNOS) using aminosalicylic acid (5-ASA), its isomer 4-ASA (10 or 50 mg/kg/day, po), and dexamethasone, an iNOS transcription inhibitor (3 mg/kg/day, sc). Enteropathy was induced by indomethacin (7.5 mg/kg/day, sc) for two days and the small intestine was examined for lesions over the next 14 days. Indomethacin-induced small-intestinal ulcer size, mucosal myeloperoxidase activity, iNOS expression and serum nitrite/nitrate levels were maximally increased by day 4 and gradually decreased by day 14. Treatment with 5-ASA, but not 4-ASA, decreased indomethacin-induced ulcer length, myeloperoxidase activity, serum nitrite/nitrate levels and iNOS expression at day 4. Dexamethasone had a greater effect than 5-ASA in reducing myeloperoxidase activity and ulcer length by 26 and 32%, respectively. Dexamethasone also reduced serum nitrate/nitrite and iNOS expression to their basal levels. In conclusion, inhibition of iNOS expression by 5-ASA appears to be associated with diminished intestinal ulceration in indomethacin-induced enteropathy.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Enteritis/drug therapy , Ileitis/drug therapy , Jejunal Diseases/drug therapy , Mesalamine/therapeutic use , Nitric Oxide Synthase Type II/biosynthesis , Transcription, Genetic/drug effects , Animals , Biomarkers , Blotting, Western , Cyclooxygenase Inhibitors/toxicity , Disease Models, Animal , Disease Progression , Electrophoresis, Polyacrylamide Gel , Enteritis/chemically induced , Enteritis/enzymology , Ileitis/chemically induced , Ileitis/enzymology , Indomethacin/toxicity , Jejunal Diseases/chemically induced , Jejunal Diseases/enzymology , Male , Rats , Rats, Sprague-Dawley , Severity of Illness Index
20.
Scand J Gastroenterol ; 41(11): 1320-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17060126

ABSTRACT

OBJECTIVE: Tumor necrosis factor (TNF)-alpha-converting enzyme (TACE), which has been purified, regulates maturity of TNF-alpha. Matrix metalloproteinases (MMPs) play a key role in various inflammatory conditions. The incidence of intestinal damage has increased, but the mechanism and treatment have not been well understood. The purpose of this study was to investigate the roles of TACE and MMP in indomethacin (Indo)-induced intestinal damage as well as the therapeutic effects of TACE inhibitor and selective MMP inhibitor (sMMPi) on this intestinal damage in rats. MATERIAL AND METHODS: In the first experiment, serial changes in intestinal ulcers and the production of MMP were investigated. In the second experiment, we assessed the effect of three TACE and/or MMP inhibitors and the production of TNF-alpha, TACE, MMP-3, -9 and tissue inhibitor of MMP (TIMP)-1. The rats were divided into five groups: a control group, and four groups that received Indo alone, Indo plus TACE inhibitor (GM6001), Indo plus a selective MMP-3 inhibitor and Indo plus an MMP-9/13 inhibitor, respectively. RESULTS: MMP-3 was overexpressed at 24 h after Indo administration, when intestinal injury was most prominent macroscopically and microscopically. GM6001 significantly decreased ulcer severity and suppressed MMP-3 in a dose-dependent fashion. The selective MMP-3 inhibitor dose-dependently ameliorated intestinal damage to the same degree as GM6001, but the MMP-9 inhibitor had no effect on the injury. CONCLUSIONS: MMP-3 inhibition ameliorates intestinal damage without apparently affecting either TNF-alpha or TACE production and the dose-response curve suggests that the beneficial effect of the so-called TACE inhibitor is actually mainly mediated via MMP-3 inhibition rather than TNF-alpha inhibition.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Enteritis/drug therapy , Ileitis/drug therapy , Jejunal Diseases/drug therapy , Matrix Metalloproteinase 3/biosynthesis , ADAM17 Protein , Animals , Blotting, Western , Disease Models, Animal , Enteritis/chemically induced , Enteritis/enzymology , Enzyme-Linked Immunosorbent Assay , Ileitis/chemically induced , Ileitis/enzymology , Jejunal Diseases/chemically induced , Jejunal Diseases/enzymology , Male , Random Allocation , Rats , Rats, Wistar , Treatment Outcome , Tumor Necrosis Factor-alpha/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...