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










Publication year range
1.
Gastroenterology ; 166(1): 103-116.e9, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37716376

ABSTRACT

BACKGROUND & AIMS: CXADR-like membrane protein (CLMP) is structurally related to coxsackie and adenovirus receptor. Pathogenic variants in CLMP gene have been associated with congenital short bowel syndrome, implying a role for CLMP in intestinal development. However, the contribution of CLMP to regulating gut development and homeostasis is unknown. METHODS: In this study, we investigated CLMP function in the colonic epithelium using complementary in vivo and in vitro approaches, including mice with inducible intestinal epithelial cell (IEC)-specific deletion of CLMP (ClmpΔIEC), intestinal organoids, IECs with overexpression, or loss of CLMP and RNA sequencing data from individuals with colorectal cancer. RESULTS: Loss of CLMP enhanced IEC proliferation and, conversely, CLMP overexpression reduced proliferation. Xenograft experiments revealed increased tumor growth in mice implanted with CLMP-deficient colonic tumor cells, and poor engraftment was observed with CLMP-overexpressing cells. ClmpΔIEC mice showed exacerbated tumor burden in an azoxymethane and dextran sulfate sodium-induced colonic tumorigenesis model, and CLMP expression was reduced in human colorectal cancer samples. Mechanistic studies revealed that CLMP-dependent regulation of IEC proliferation is linked to signaling through mTOR-Akt-ß-catenin pathways. CONCLUSIONS: These results reveal novel insights into CLMP function in the colonic epithelium, highlighting an important role in regulating IEC proliferation, suggesting tumor suppressive function in colon cancer.


Subject(s)
Colitis , Colonic Neoplasms , Animals , Humans , Mice , Cell Proliferation , Colitis/chemically induced , Colitis/metabolism , Colonic Neoplasms/pathology , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Epithelial Cells/pathology , Intestinal Mucosa/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism
2.
FASEB J ; 33(12): 13632-13643, 2019 12.
Article in English | MEDLINE | ID: mdl-31585047

ABSTRACT

Mucosal wound repair is coordinated by dynamic crosstalk between endogenous and exogenous mediators and specific receptors on epithelial cells and infiltrating immune cells. One class of such receptor-ligand pairs involves formyl peptide receptors (FPRs) that have been shown to influence inflammatory response and repair. Here we explored the role of murine Fpr2/3, an ortholog of human FPR2/receptor for lipoxin A4 (ALX), in orchestrating intestinal mucosal repair. Compared with wild-type (WT) mice, Fpr2/3-/- mice exhibited delayed recovery from acute experimental colitis and perturbed repair after biopsy-induced colonic mucosal injury. Decreased numbers of infiltrating monocytes were observed in healing wounds from Fpr2/3-/- mice compared with WT animals. Bone marrow transplant experiments revealed that Fpr2/3-/- monocytes showed a competitive disadvantage when infiltrating colonic wounds. Moreover, Fpr2/3-/- monocytes were defective in chemotactic responses to the chemokine CC chemokine ligand (CCL)20, which is up-regulated during early phases of inflammation. Analysis of Fpr2/3-/- monocytes revealed altered expression of the CCL20 receptor CC chemokine receptor (CCR)6, suggesting that Fpr2/3 regulates CCL20-CCR6-mediated monocyte chemotaxis to sites of mucosal injury in the gut. These findings demonstrate an important contribution of Fpr2/3 in facilitating monocyte recruitment to sites of mucosal injury to influence wound repair.-Birkl, D., O'Leary, M. N., Quiros, M., Azcutia, V., Schaller, M., Reed, M., Nishio, H., Keeney, J., Neish, A. S., Lukacs, N. W., Parkos, C. A., Nusrat, A. Formyl peptide receptor 2 regulates monocyte recruitment to promote intestinal mucosal wound repair.


Subject(s)
Cell Movement , Inflammation/therapy , Intestinal Mucosa/physiology , Monocytes/metabolism , Receptors, Formyl Peptide/physiology , Wound Healing , Animals , Bone Marrow Transplantation , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Dextran Sulfate/toxicity , Inflammation/etiology , Inflammation/pathology , Intestinal Mucosa/injuries , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Receptors, CCR6/genetics , Receptors, CCR6/metabolism
3.
Front Immunol ; 10: 486, 2019.
Article in English | MEDLINE | ID: mdl-30936879

ABSTRACT

Inflammatory Bowel Disease (IBD) is a multi-factorial chronic inflammation of the gastrointestinal tract prognostically linked to CD8+ T-cells, but little is known about their mechanism of activation during initiation of colitis. Here, Grb2-associated binding 2/3 adaptor protein double knockout mice (Gab2/3-/-) were generated. Gab2/3-/- mice, but not single knockout mice, developed spontaneous colitis. To analyze the cellular mechanism, reciprocal bone marrow (BM) transplantation demonstrated a Gab2/3-/- hematopoietic disease-initiating process. Adoptive transfer showed individual roles for macrophages and T-cells in promoting colitis development in vivo. In spontaneous disease, intestinal intraepithelial CD8+ but much fewer CD4+, T-cells from Gab2/3-/- mice with rectal prolapse were more proliferative. To analyze the molecular mechanism, reduced PI3-kinase/Akt/mTORC1 was observed in macrophages and T-cells, with interleukin (IL)-2 stimulated T-cells showing increased pSTAT5. These results illustrate the importance of Gab2/3 collectively in signaling responses required to control macrophage and CD8+ T-cell activation and suppress chronic colitis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , CD8-Positive T-Lymphocytes/immunology , Colitis/immunology , Inflammatory Bowel Diseases/immunology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/transplantation , Colitis/pathology , Disease Models, Animal , Intraepithelial Lymphocytes/immunology , Lipocalin-2/analysis , Lymphocyte Activation , Macrophage Activation , Macrophages/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Radiation Chimera , Rectal Prolapse/etiology , Rectal Prolapse/immunology , Rectal Prolapse/pathology , Signal Transduction , TOR Serine-Threonine Kinases/physiology
4.
J Clin Invest ; 127(9): 3510-3520, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28783045

ABSTRACT

In response to injury, epithelial cells migrate and proliferate to cover denuded mucosal surfaces and repair the barrier defect. This process is orchestrated by dynamic crosstalk between immune cells and the epithelium; however, the mechanisms involved remain incompletely understood. Here, we report that IL-10 was rapidly induced following intestinal mucosal injury and was required for optimal intestinal mucosal wound closure. Conditional deletion of IL-10 specifically in CD11c-expressing cells in vivo implicated macrophages as a critical innate immune contributor to IL-10-induced wound closure. Consistent with these findings, wound closure in T cell- and B cell-deficient Rag1-/- mice was unimpaired, demonstrating that adaptive immune cells are not absolutely required for this process. Further, following mucosal injury, macrophage-derived IL-10 resulted in epithelial cAMP response element-binding protein (CREB) activation and subsequent synthesis and secretion of the pro-repair WNT1-inducible signaling protein 1 (WISP-1). WISP-1 induced epithelial cell proliferation and wound closure by activating epithelial pro-proliferative pathways. These findings define the involvement of macrophages in regulating an IL-10/CREB/WISP-1 signaling axis, with broad implications in linking innate immune activation to mucosal wound repair.


Subject(s)
CCN Intercellular Signaling Proteins/metabolism , Interleukin-10/metabolism , Macrophages/metabolism , Proto-Oncogene Proteins/metabolism , Animals , CD11 Antigens/metabolism , Cell Proliferation , Colon/pathology , Cyclic AMP Response Element-Binding Protein/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Deletion , Gene Expression Regulation , Humans , Inflammation , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Wound Healing
5.
Proc Natl Acad Sci U S A ; 113(51): 14787-14792, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27930307

ABSTRACT

Inflammatory bowel disease (IBD) results from aberrant immune stimulation against a dysbiotic mucosal but relatively preserved luminal microbiota and preferentially affects males in early onset disease. However, factors contributing to sex-specific risk and the pattern of dysbiosis are largely unexplored. Core 1 ß3GalT-specific molecular chaperone (Cosmc), which encodes an X-linked chaperone important for glycocalyx formation, was recently identified as an IBD risk factor by genome-wide association study. We deleted Cosmc in mouse intestinal epithelial cells (IECs) and found marked reduction of microbiota diversity in progression from the proximal to the distal gut mucosa, but not in the overlying lumen, as seen in IBD. This loss of diversity coincided with local emergence of a proinflammatory pathobiont and distal gut restricted pathology. Mechanistically, we found that Cosmc regulates host genes, bacterial ligands, and nutrient availability to control microbiota biogeography. Loss of one Cosmc allele in males (IEC-Cosmc-/y) resulted in a compromised mucus layer, spontaneous microbe-dependent inflammation, and enhanced experimental colitis; however, females with loss of one allele and mosaic deletion of Cosmc in 50% of crypts (IEC-Cosmc+/-) were protected from spontaneous inflammation and partially protected from experimental colitis, likely due to lateral migration of normal mucin glycocalyx from WT cells over KO crypts. These studies functionally validate Cosmc as an IBD risk factor and implicate it in regulating the spatial pattern of dysbiosis and sex bias in IBD.


Subject(s)
Gastrointestinal Microbiome , Genes, X-Linked , Inflammatory Bowel Diseases/genetics , Molecular Chaperones/genetics , Sex Factors , Alleles , Animals , Colitis/microbiology , Female , Gene Deletion , Genetic Linkage , Genome-Wide Association Study , Glycocalyx , Inflammation , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mosaicism , Risk Factors , X Chromosome
6.
Nat Microbiol ; 1: 15021, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-27571978

ABSTRACT

The mammalian intestine houses a complex microbial community, which influences normal epithelial growth and development, and is integral to the repair of damaged intestinal mucosa(1-3). Restitution of injured mucosa involves the recruitment of immune cells, epithelial migration and proliferation(4,5). Although microenvironmental alterations have been described in wound healing(6), a role for extrinsic influences, such as members of the microbiota, has not been reported. Here, we show that a distinct subpopulation of the normal mucosal-associated gut microbiota expands and preferentially colonizes sites of damaged murine mucosa in response to local environmental cues. Our results demonstrate that formyl peptide receptor 1 (FPR1) and neutrophilic NADPH oxidase (NOX2) are required for the rapid depletion of microenvironmental oxygen and compensatory responses, resulting in a dramatic enrichment of an anaerobic bacterial consortium. Furthermore, the dominant member of this wound-mucosa-associated microbiota, Akkermansia muciniphila (an anaerobic, mucinophilic gut symbiont(7,8)), stimulated proliferation and migration of enterocytes adjacent to the colonic wounds in a process involving FPR1 and intestinal epithelial-cell-specific NOX1-dependent redox signalling. These findings thus demonstrate how wound microenvironments induce the rapid emergence of 'probiont' species that contribute to enhanced repair of mucosal wounds. Such microorganisms could be exploited as potential therapeutics.


Subject(s)
Bacteria, Anaerobic/growth & development , Gastrointestinal Microbiome , Intestinal Mucosa/injuries , Intestinal Mucosa/microbiology , Wounds and Injuries/microbiology , Anaerobiosis , Animals , Cell Movement , Cell Proliferation , Enterocytes/physiology , Mice , NADPH Oxidase 1/metabolism , NADPH Oxidase 2/metabolism , Receptors, Formyl Peptide/metabolism
7.
J Immunol ; 196(1): 34-8, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26590314

ABSTRACT

IL-1 family members are central mediators of host defense. In this article, we show that the novel IL-1 family member IL-36γ was expressed during experimental colitis and human inflammatory bowel disease. Germ-free mice failed to induce IL-36γ in response to dextran sodium sulfate (DSS)-induced damage, suggesting that gut microbiota are involved in its induction. Surprisingly, IL-36R-deficient (Il1rl2(-/-)) mice exhibited defective recovery following DSS-induced damage and impaired closure of colonic mucosal biopsy wounds, which coincided with impaired neutrophil accumulation in the wound bed. Failure of Il1rl2(-/-) mice to recover from DSS-induced damage was associated with a profound reduction in IL-22 expression, particularly by colonic neutrophils. Defective recovery of Il1rl2(-/-) mice could be rescued by an aryl hydrocarbon receptor agonist, which was sufficient to restore IL-22 expression and promote full recovery from DSS-induced damage. These findings implicate the IL-36/IL-36R axis in the resolution of intestinal mucosal wounds.


Subject(s)
Colitis/immunology , Interleukin-1/biosynthesis , Interleukins/biosynthesis , Receptors, Interleukin/immunology , Wound Healing/immunology , Animals , Colitis/chemically induced , Colitis/microbiology , Colon/immunology , Colon/injuries , Dextran Sulfate , Helicobacter hepaticus/pathogenicity , Humans , Inflammation/immunology , Inflammation/pathology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/metabolism , Receptors, Aryl Hydrocarbon/agonists , Receptors, Interleukin/genetics , Wound Healing/genetics , Interleukin-22
8.
Mol Biol Cell ; 26(18): 3165-77, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26224314

ABSTRACT

Desmosomal cadherins mediate intercellular adhesion and control epithelial homeostasis. Recent studies show that proteinases play an important role in the pathobiology of cancer by targeting epithelial intercellular junction proteins such as cadherins. Here we describe the proinflammatory cytokine-induced activation of matrix metalloproteinase 9 and a disintegrin and metalloproteinase domain-containing protein 10, which promote the shedding of desmosomal cadherin desmoglein-2 (Dsg2) ectodomains in intestinal epithelial cells. Epithelial exposure to Dsg2 ectodomains compromises intercellular adhesion by promoting the relocalization of endogenous Dsg2 and E-cadherin from the plasma membrane while also promoting proliferation by activation of human epidermal growth factor receptor 2/3 signaling. Cadherin ectodomains were detected in the inflamed intestinal mucosa of mice with colitis and patients with ulcerative colitis. Taken together, our findings reveal a novel response pathway in which inflammation-induced modification of columnar epithelial cell cadherins decreases intercellular adhesion while enhancing cellular proliferation, which may serve as a compensatory mechanism to promote repair.


Subject(s)
Desmoglein 2/metabolism , Intestinal Mucosa/metabolism , Animals , CHO Cells , Cell Adhesion/physiology , Cell Line , Cricetulus , Cytokines/metabolism , Disintegrins/metabolism , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Intestinal Mucosa/pathology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL
9.
J Clin Invest ; 125(3): 1215-27, 2015 Mar 02.
Article in English | MEDLINE | ID: mdl-25664854

ABSTRACT

Epithelial restitution is an essential process that is required to repair barrier function at mucosal surfaces following injury. Prolonged breaches in epithelial barrier function result in inflammation and further damage; therefore, a better understanding of the epithelial restitution process has potential for improving the development of therapeutics. In this work, we demonstrate that endogenous annexin A1 (ANXA1) is released as a component of extracellular vesicles (EVs) derived from intestinal epithelial cells, and these ANXA1-containing EVs activate wound repair circuits. Compared with healthy controls, patients with active inflammatory bowel disease had elevated levels of secreted ANXA1-containing EVs in sera, indicating that ANXA1-containing EVs are systemically distributed in response to the inflammatory process and could potentially serve as a biomarker of intestinal mucosal inflammation. Local intestinal delivery of an exogenous ANXA1 mimetic peptide (Ac2-26) encapsulated within targeted polymeric nanoparticles (Ac2-26 Col IV NPs) accelerated healing of murine colonic wounds after biopsy-induced injury. Moreover, one-time systemic administration of Ac2-26 Col IV NPs accelerated recovery following experimentally induced colitis. Together, our results suggest that local delivery of proresolving peptides encapsulated within nanoparticles may represent a potential therapeutic strategy for clinical situations characterized by chronic mucosal injury, such as is seen in patients with IBD.


Subject(s)
Annexin A1/physiology , Exosomes/physiology , Intestinal Mucosa/physiopathology , Animals , Annexin A1/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Cell Line , Colitis/blood , Colitis/physiopathology , Humans , Intestinal Mucosa/drug effects , Mice, Knockout , Nanoparticles , Peptides/administration & dosage , Wound Healing
10.
Bioorg Med Chem ; 22(11): 3021-9, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24767819

ABSTRACT

Interleukin-12 (IL-12) and IL-23 are proinflammatory cytokines and therapeutic targets for inflammatory and autoimmune diseases, including inflammatory bowel diseases, psoriasis, rheumatoid arthritis, and multiple sclerosis. We describe the discovery of APY0201, a unique small molecular IL-12/23 production inhibitor, from activated macrophages and monocytes, and demonstrate ameliorated inflammation in an experimental model of colitis. Through a chemical proteomics approach using a highly sensitive direct nanoflow LC-MS/MS system and bait compounds equipped with the FLAG epitope associated regulator of PIKfyve (ArPIKfyve) was detected. Further study identified its associated protein phosphoinositide kinase, FYVE finger-containing (PIKfyve), as the target protein of APY0201, which was characterized as a potent, highly selective, ATP-competitive PIKfyve inhibitor that interrupts the conversion of phosphatidylinositol 3-phosphate (PtdIns3P) to PtdIns(3,5)P2. These results elucidate the function of PIKfyve kinase in the IL-12/23 production pathway and in IL-12/23-driven inflammatory disease pathologies to provide a compelling rationale for targeting PIKfyve kinase in inflammatory and autoimmune diseases.


Subject(s)
Interleukin-12/antagonists & inhibitors , Interleukin-23/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Small Molecule Libraries/pharmacology , Animals , Cell Line , Colitis/drug therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Discovery , Female , Humans , Inflammation/drug therapy , Interleukin-10/deficiency , Leukocytes, Mononuclear/drug effects , Macrophages/drug effects , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , Models, Molecular , Molecular Structure , Pyrazoles/chemistry , Pyrimidines/chemistry , Small Molecule Libraries/chemistry , Structure-Activity Relationship
11.
Med Sci Monit ; 18(1): BR9-15, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22207112

ABSTRACT

BACKGROUND: Reflux esophagitis is caused mainly by excessive exposure of the mucosa to gastric contents. In the present study, we examined the effect of several amino acids on acid reflux esophagitis in rats. MATERIAL/METHODS: After 18 h of fasting, acid reflux esophagitis was induced by ligating both the pylorus and the transitional region between the forestomach and the corpus under ether anesthesia, and the animals were killed 4 h later. The severity of esophagitis was reduced by the oral administration of omeprazole, a proton pump inhibitor, or pepstatin, a specific pepsin inhibitor. RESULTS: The development of esophageal lesions was dose-dependently prevented by L-arginine and glycine, given intragastrically (i.g.) after the ligation, with complete inhibition obtained at 250 mg/kg and 750 mg/kg, respectively, and these effects were not influenced by the prior s.c. administration of indomethacin or L-NAME. By contrast, both L-alanine and L-glutamine given i.g. after the ligation aggravated these lesions in a dose-dependent manner. These amino acids had no effect on acid secretion but increased the pH of the gastric contents to 1.8~2.3 due to their buffering action. CONCLUSIONS: The results confirmed an essential role for acid and pepsin in the pathogenesis of acid reflux esophagitis in the rat model and further suggested that various amino acids affect the severity of esophagitis in different ways, due to yet unidentified mechanisms; L-alanine and L-glutamine exert a deleterious effect on the esophagitis, while L-arginine and glycine are highly protective, independent of endogenous prostaglandins and nitric oxide.


Subject(s)
Arginine/therapeutic use , Esophagitis, Peptic/prevention & control , Glycine/therapeutic use , Administration, Oral , Animals , Arginine/administration & dosage , Dose-Response Relationship, Drug , Gastrointestinal Contents/chemistry , Glycine/administration & dosage , Hydrogen-Ion Concentration , Rats
12.
Am J Physiol Gastrointest Liver Physiol ; 297(3): G506-13, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19589943

ABSTRACT

Inflammatory responses triggered by activation of the lipopolysaccharide (LPS)/Toll-like receptor (TLR) 4 signaling pathway are a key mechanism in nonsteroidal anti-inflammatory drug-induced enteropathy. The aim of this study was to investigate the probiotic effect of Lactobacillus casei strain Shirota (LcS) on indomethacin-induced small intestinal injury. Rats pretreated with viable LcS or heat-killed LcS once or once daily for a week were administered indomethacin by gavage to induce injury. Anti-inflammatory effects of L-lactic acid (1-15 mM) were evaluated in vitro by use of THP-1 cells. One-week treatment with viable LcS prevented indomethacin-induced intestinal injury with increase in the concentration of lactic acid in small intestinal content and inhibited increases in myeloperoxidase activity and expression of mRNA for tumor necrosis factor-alpha (TNF-alpha) while affecting neither TLR4 expression nor the number of gram-negative bacteria in intestinal content, whereas neither heat-killed LcS nor a single dose of viable LcS inhibited intestinal injury. Prevention of this injury was also observed in rats given l-lactic acid in drinking water. Both L-lactic acid and LcS culture supernatant containing 10 mM lactic acid inhibited NF-kappaB activation and increases in TNF-alpha mRNA expression and TNF-alpha protein secretion in THP-1 cells treated with LPS. Western blot analyses showed that both L-lactic acid and LcS culture supernatants suppressed phosphorylation and degradation of I-kappaB-alpha induced by LPS without affecting expression of TLR4. These findings suggest that LcS exhibits a prophylactic effect on indomethacin-induced enteropathy by suppressing the LPS/TLR4 signaling pathway and that this probiotic effect of LcS may be mediated by L-lactic acid.


Subject(s)
Intestinal Diseases/prevention & control , Intestine, Small/microbiology , Lactic Acid/metabolism , Lacticaseibacillus casei/metabolism , Probiotics , Animals , Cell Line , Disease Models, Animal , Humans , I-kappa B Proteins/metabolism , Indomethacin , Intestinal Diseases/chemically induced , Intestinal Diseases/metabolism , Intestinal Diseases/microbiology , Intestine, Small/metabolism , Intestine, Small/pathology , Lactic Acid/administration & dosage , Lacticaseibacillus casei/isolation & purification , Male , Monocytes/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Peroxidase/metabolism , Phosphorylation , RNA, Messenger/metabolism , Rats , Rats, Wistar , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
13.
Life Sci ; 84(5-6): 172-80, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-19070625

ABSTRACT

AIMS: We examined the effect of a prostacyclin (PGI(2)) analog iloprost on histamine-induced acid secretion and investigated how endogenous PGI(2) mediated the decreased secretory response in the damaged stomach after exposure to taurocholate (TC). MAIN METHODS: Male C57BL/6 mice, both wild-type (WT) and IP receptor knockout (IP-KO) animals, were used after 18 h of fasting. Under urethane anesthesia, the abdomen was incised, and an acute fistula was provided in the stomach. KEY FINDINGS: Acid secretion in WT and IP-KO mice was similarly and dose-dependently increased by histamine. Iloprost decreased the histamine-stimulated secretion in WT but not IP-KO mice. The inhibitory effect of iloprost in WT mice was totally abrogated by the prior administration of CYN154806, a selective somatostatin SST2 receptor antagonist. On the other hand, the acid secretion in WT mice was decreased after exposure of the stomach to 20 mM TC for 20 min, with an increase in mucosal PGI(2) content, but the decrease was significantly less marked in IP-KO mice. The decreased acid response to TC in WT mice was totally prevented by the prior administration of CYN154806 as well as indomethacin. Somatostatin contents in the stomach were reduced after the administration of iloprost or the mucosal exposure to TC, while the blood levels increased. SIGNIFICANCE: Somatostatin/SST2 receptors are involved in the decreased acid response of the damaged stomach, in addition to PGI(2)/IP receptors. It is assumed that PGI(2) releases somatostatin from D cells, which in turn decreases acid secretion via the activation of SST2 receptors.


Subject(s)
Gastric Acid/metabolism , Receptors, Prostaglandin/physiology , Receptors, Somatostatin/physiology , Stomach Diseases/metabolism , 6-Ketoprostaglandin F1 alpha/metabolism , Animals , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Histamine/pharmacology , Iloprost/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligopeptides/pharmacology , Receptors, Epoprostenol , Receptors, Prostaglandin/genetics , Receptors, Somatostatin/antagonists & inhibitors , Receptors, Somatostatin/metabolism , Somatostatin/analysis , Somatostatin/blood , Stomach Diseases/chemically induced , Stomach Diseases/prevention & control , Taurocholic Acid
14.
Life Sci ; 81(16): 1309-16, 2007 Sep 29.
Article in English | MEDLINE | ID: mdl-17920637

ABSTRACT

NSAIDs damage the small intestine as well as the stomach as adverse effects. We previously reported that the gastric ulcerogenic response to NSAIDs was markedly increased in arthritic rats. The present study was designed to examine the intestinal ulcerogenic property of indomethacin in adjuvant-induced arthritic rats in comparison with normal animals. Arthritis was induced in male Dark Agouti rats by injection of Freund's complete adjuvant into the right hindfoot. Two weeks later, indomethacin was given orally and the intestine was examined for lesions at several time points after indomethacin. Indomethacin produced intestinal lesions in both normal and arthritic rats, but in the latter, the ulcerogenic response occurred much earlier and the severity was markedly enhanced. Aminoguanidine, an inhibitor of iNOS, significantly suppressed the damage, yet the efficacy differed in normal and arthritic rats, depending on the dose schedule; the effect of post-administration (6 h after) was greater than that of pre-administration (0.5 h before) in normal rats, whereas that of post-administration was less than that of pre-administration in arthritic rats. The expression of iNOS and TLR4 in the intestine was enhanced in arthritic rats as compared with normal rats. These results suggest that the intestinal ulcerogenic response to indomethacin is markedly aggravated in arthritic rats. Notably, the onset of the ulceration was much earlier in arthritic rats than normal rats. These phenomena may be accounted for by the upregulation of iNOS/NO through the increased expression of TLR4 in the small intestine of arthritic rats.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Arthritis, Experimental , Intestine, Small/drug effects , Peptic Ulcer , Toll-Like Receptor 4/biosynthesis , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/complications , Arthritis, Experimental/metabolism , Freund's Adjuvant , Gene Expression/drug effects , Immunohistochemistry , Intestine, Small/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Peptic Ulcer/chemically induced , Peptic Ulcer/etiology , Peptic Ulcer/metabolism , Peroxidase/metabolism , Rats , Rats, Inbred Strains , Time Factors , Toll-Like Receptor 4/genetics , Up-Regulation
15.
Dig Dis Sci ; 52(9): 2095-103, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17429720

ABSTRACT

We examined the effect of cyclooxygenase (COX) inhibitors on dextran sulfate sodium (DSS)-induced ulcerative colitis in rats and investigated the role of COX isozymes in the pathogenesis of this model. Experimental colitis was induced by treatment with 2.5% DSS in drinking water for 6 days. Indomethacin (a nonselective COX inhibitor), SC-560 (a selective COX-1 inhibitor), or celecoxib (a selective COX-2 inhibitor) was given PO twice daily for 6 days, during the first 3 or last 3 days of the experimental period. Daily treatment with 2.5% DSS for 6 days caused damage to the colon, with a decrease in body weight gain and colon length as well as an increase of myeloperoxidase (MPO) activity. All COX inhibitors given for 6 days significantly worsened the severity of DSS-induced colonic damage with increased MPO activity. The aggravation was also observed by SC-560 given for the first 3 days or by celecoxib given for the last 3 days. The expression of COX-2 mRNA in the colon was upregulated on day 3 during DSS treatment, with significant increase of prostaglandin E(2) PGE(2) production. The PGE(2) content on day 3 during DSS treatment was inhibited by both indomethacin and SC-560, but not by celecoxib; on day 6 it was suppressed by both indomethacin and celecoxib, but not SC-560. These results suggest that endogenous prostaglandins (PGs) afford protection against colonic ulceration, yet the COX isozyme responsible for the production of PGs differs depending on the stage of ulceration; COX-1 in the early stage and COX-2 in the late stage.


Subject(s)
Colitis, Ulcerative/drug therapy , Cyclooxygenase 1/drug effects , Cyclooxygenase 2/drug effects , Cyclooxygenase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , Animals , Cardiovascular Diseases , Celecoxib , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/metabolism , Colon/drug effects , Colon/metabolism , Colon/pathology , Cyclooxygenase 2/genetics , Dextran Sulfate/toxicity , Dinoprostone/biosynthesis , Disease Models, Animal , Gene Expression/drug effects , Indomethacin/therapeutic use , Male , Peroxidase/metabolism , Plasma Substitutes/toxicity , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Spectrophotometry , Treatment Outcome
16.
J Pharmacol Sci ; 103(1): 40-7, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17202747

ABSTRACT

We examined the effect of tacrolimus (FK506), an immunosuppressive drug, on indomethacin-induced small intestinal ulceration in rats. Animals were given indomethacin (10 mg/kg, s.c.), killed 24 h later, and myeloperoxidase (MPO) activity and thiobarbituric acid reactants (TBARS) were evaluated in intestinal lesions. Tacrolimus (0.3 - 3 mg/kg) was administered p.o. twice 0.5 h before and 6 h after indomethacin injection. The expression of inducible nitric oxide synthase (iNOS) mRNA was determined by a TaqMan real-time RT-PCR, while the activity of nuclear factor (NF)-kappaB DNA-binding was analyzed by electrophoresis mobility shift assays (EMSA) 6 h after indomethacin treatment. Indomethacin provoked severe hemorrhagic lesions in the small intestine, mainly in the jejunum and ileum, accompanied with increases in MPO activity and TBARS. Oral administration of tacrolimus reduced the severity of indomethacin-induced intestinal lesions in a dose-dependent manner. The increases in MPO activity and TBARS were also significantly attenuated by tacrolimus. The expression of iNOS mRNA was markedly enhanced when examined 6 h after indomethacin administration, and this response was counteracted by tacrolimus. Indomethacin also activated NF-kappaB in a tacrolimus-preventable manner. These results suggest that tacrolimus prevents indomethacin-induced small intestinal ulceration in the rat. This effect may be due to inhibition of iNOS induction through suppression of NF-kappaB activation.


Subject(s)
Immunosuppressive Agents/pharmacology , Indomethacin/antagonists & inhibitors , Intestinal Diseases/prevention & control , Nitric Oxide Synthase Type II/antagonists & inhibitors , Tacrolimus/pharmacology , Ulcer/prevention & control , Animals , Intestinal Diseases/chemically induced , Lipid Peroxidation/drug effects , Male , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Peroxidase/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/genetics , Ulcer/chemically induced
17.
Digestion ; 74(2): 91-100, 2006.
Article in English | MEDLINE | ID: mdl-17143008

ABSTRACT

We examined the effects of various cyclooxygenase (COX) inhibitors on the healing of colonic lesions induced by dextran sulfate sodium (DSS) in the rat. Colonic lesions were induced by 2.5% DSS in the drinking water for 7 days, and then the animals were fed with tap water for subsequent 7 days. Indomethacin (a nonselective COX inhibitor), SC-560 (a selective COX-1 inhibitor), or rofecoxib (a selective COX-2 inhibitor) was given orally twice daily after termination of the DSS treatment. DSS treatment caused severe colonic lesions with a decrease in body weight gain and colon length as well as an increase in myeloperoxidase activity and thiobarbituric acid reactant levels. The severity of colitis gradually reduced, with an improvement of morphological and histological alterations. Daily administration of indomethacin and rofecoxib significantly delayed the healing of colitis with deleterious influences on histological restitution as well as mucosal inflammation, while SC-560 had no effect. Although COX-1 mRNA was expressed in the colon without much alteration during the test period, the expression of COX-2 was upregulated with a peak on day 3 and decreased thereafter. The mucosal prostaglandin E2 content in the colon showed a biphasic change, in parallel with that of the COX-2 expression. The increased prostaglandin E2 production in the injured mucosa was attenuated by indomethacin and rofecoxib, but not by SC-560. These results suggest that endogenous prostaglandins produced by COX-2 play an important role in the healing of DSS-induced colonic lesions. Caution should be paid to the use of selective COX-2 inhibitors as well as nonsteroidal anti-inflammatory drugs in patients with colitis.


Subject(s)
Colitis/pathology , Cyclooxygenase 2 Inhibitors/adverse effects , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/physiology , Dinoprostone/antagonists & inhibitors , Animals , Colitis/chemically induced , Colitis/enzymology , Colon/chemistry , Colon/drug effects , Colon/pathology , Cyclooxygenase 1/drug effects , Cyclooxygenase 1/genetics , Cyclooxygenase 1/physiology , Cyclooxygenase 2/genetics , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Dextran Sulfate/toxicity , Dinoprostone/analysis , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Male , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Wistar
18.
Life Sci ; 79(16): 1523-30, 2006 Sep 13.
Article in English | MEDLINE | ID: mdl-16730029

ABSTRACT

Nitric oxide (NO) plays a role in regulating the mucosal integrity of the stomach. However, its part in the mucosal defense of the inflamed stomach remains unclear. In the present study, we examined the effects of various NO synthase (NOS) inhibitors on gastric ulcerogenic and acid secretory responses following daily exposure of the stomach to iodoacetamide and investigated the role of each NOS isozyme in gastric protection from subchronic mucosal irritation. Gastric mucosal irritation was induced in rats by addition of 0.1% iodoacetamide to drinking water, and the gastric mucosa was examined on the 6th day. L-NAME (a nonselective NOS inhibitor: 20 mg/kg) or aminoguanidine (a selective iNOS inhibitor: 20 mg/kg) was given s.c. twice 24 h and 3 h before the termination of iodoacetamide treatment. Giving iodoacetamide in drinking water for 5 days produced minimal damage in the stomach with an increase in myeloperoxidase (MPO) activity and lipid peroxidation. Iodoacetamide treatment up-regulated the expression of iNOS mRNA and NO production in the stomach, without affecting nNOS expression. Both L-NAME and aminoguanidine markedly aggravated gastric lesions induced by iodoacetamide treatment, with a further enhancement in MPO activity and lipid peroxidation. Basal acid secretion as determined in pylorous-ligated stomachs was decreased following iodoacetamide treatment, but the response was significantly restored by both L-NAME and aminoguanidine. These results suggest that endogenous NO derived from both cNOS and iNOS is involved in mucosal defense of the inflamed stomach, partly by decreasing acid secretion, and contributes to maintaining mucosal integrity under such conditions.


Subject(s)
Gastric Mucosa/metabolism , Gastritis/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Animals , Body Weight/drug effects , Enzyme Inhibitors , Gastric Acid/metabolism , Gastric Mucosa/enzymology , Gastric Mucosa/pathology , Gastritis/chemically induced , Gastritis/enzymology , Guanidines/pharmacology , Iodoacetamide/toxicity , Lipid Peroxidation/drug effects , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley
19.
Dig Dis Sci ; 51(2): 303-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16534673

ABSTRACT

Pepsin, a protease activated by gastric acid, is a component of the refluxate, yet the role of pepsin in the pathogenesis of reflux esophagitis has not been well studied. In the present study, we examined the effect of pepstatin, a specific inhibitor of pepsin, on acid reflux esophagitis. Acid reflux esophagitis was induced in rats by ligating both the pylorus and the forestomach for 3 or 4 hr. Pepstatin, ecabet Na (the anti-ulcer drug), and L-glutamine were administered intragastrically after the ligation. Pepstatin or ecabet Na, given intragastrically, significantly prevented esophageal lesions, even though they did not affect basal acid secretion in pylorus-ligated rats. Pepstatin significantly inhibited pepsin activity in vivo and in vitro, while ecabet Na inhibited this activity in vitro. By contrast, L-glutamine given intragastrically aggravated the lesions in a dose-dependent manner, but even in the presence of L-glutamine the development of esophageal lesions was totally prevented by coadministration of pepstatin or ecabet Na. L-Glutamine increased the pH of gastric contents to approximately 2.0, the optimal pH for the proteolytic activity of pepsin in vitro. In addition, intragastric administration of exogenous pepsin worsened the severity of esophageal damage. These results suggest that pepstatin is highly effective against acid reflux esophagitis, without influencing acid secretion, while L-glutamine aggravated these lesions by increasing the pepsin activity by shifting the intraluminal pH to the optimal pH range for proteolytic action. It is assumed that pepsin plays a major pathogenic role in the development of acid reflux esophagitis.


Subject(s)
Esophagitis, Peptic/etiology , Esophagus/drug effects , Glutamine/pharmacology , Pepsin A/physiology , Pepstatins/pharmacology , Protease Inhibitors/pharmacology , Animals , Anti-Ulcer Agents/therapeutic use , Disease Models, Animal , Esophagitis, Peptic/pathology , Esophagitis, Peptic/prevention & control , Esophagus/pathology , Male , Rats , Rats, Sprague-Dawley
20.
Digestion ; 70(4): 240-9, 2004.
Article in English | MEDLINE | ID: mdl-15687726

ABSTRACT

BACKGROUND/AIM: Rebamipide, an anti-ulcer drug, has various actions including radical scavenging and mucus-stimulating as well as anti-inflammatory effects, and exhibits both mucosal protective and healing promoting actions in the stomach. In the present study, we examined the effect of rebamipide on an animal model of colitis induced by dextran sulfate sodium (DSS). METHODS: Experimental colitis was induced in rats by daily treatment with 3% DSS in drinking water for 7 days. Rebamipide (3-30 mg/kg), 5-aminosalicylic acid (5-ASA: 150 mg/kg) or metronidazole (10 and 30 mg/kg) was administered intra-rectally once daily for 6 days. The ulceration area, colon length, and mucosal myeloperoxidase (MPO) activity as well as thiobarbituric acid-reactive substance (TBARS) were measured on the 7th day after the onset of DSS treatment. The effects of rebamipide on the secretion of mucus in the colon was also examined. RESULTS: DSS treatment caused severe lesions in the colon, accompanied by an increase in MPO activity and TBARS as well as a decrease in body weight gain and colon length. Repeated administration of rebamipide dose-dependently suppressed the colon lesions and improved the pathological changes induced by DSS treatment. Rebamipide significantly increased the mucus contents in the colon. Both 5-ASA and metronidazole also reduced the severity of DSS-induced lesions. CONCLUSION: These results suggest that intra-rectal administration of rebamipide is effective against DSS-induced colitis. The protective effect of rebamipide may be attributable to both the radical scavenging action and the increase in the production of mucus in the colon, the latter presumably suppressing the process of intestinal bacterial infiltration.


Subject(s)
Alanine/analogs & derivatives , Alanine/administration & dosage , Alanine/pharmacology , Anti-Ulcer Agents/administration & dosage , Anti-Ulcer Agents/pharmacology , Colitis/prevention & control , Quinolones/administration & dosage , Quinolones/pharmacology , Administration, Rectal , Animals , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Colitis/chemically induced , Colitis/veterinary , Dextran Sulfate/administration & dosage , Dextran Sulfate/adverse effects , Disease Models, Animal , Male , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...