Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 117(15): 8431-8436, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32220957

ABSTRACT

Intestinal barrier dysfunction, which leads to translocation of bacteria or toxic bacterial products from the gut into bloodstream and results in systemic inflammation, is a key pathogenic factor in many human diseases. However, the molecular mechanisms leading to intestinal barrier defects are not well understood, and there are currently no available therapeutic approaches to target intestinal barrier function. Here we show that soluble epoxide hydrolase (sEH) is an endogenous regulator of obesity-induced intestinal barrier dysfunction. We find that sEH is overexpressed in the colons of obese mice. In addition, pharmacologic inhibition or genetic ablation of sEH abolishes obesity-induced gut leakage, translocation of endotoxin lipopolysaccharide or bacteria, and bacterial invasion-induced adipose inflammation. Furthermore, systematic treatment with sEH-produced lipid metabolites, dihydroxyeicosatrienoic acids, induces bacterial translocation and colonic inflammation in mice. The actions of sEH are mediated by gut bacteria-dependent mechanisms, since inhibition or genetic ablation of sEH fails to attenuate obesity-induced gut leakage and adipose inflammation in mice lacking gut bacteria. Overall, these results support that sEH is a potential therapeutic target for obesity-induced intestinal barrier dysfunction, and that sEH inhibitors, which have been evaluated in human clinical trials targeting other human disorders, could be promising agents for prevention and/or treatment.


Subject(s)
Bacterial Translocation , Epoxide Hydrolases/immunology , Intestinal Diseases/enzymology , Intestines/enzymology , Obesity/complications , Adipose Tissue/immunology , Animals , Bacteria/genetics , Bacteria/isolation & purification , Bacterial Physiological Phenomena , Epoxide Hydrolases/genetics , Gastrointestinal Microbiome , Humans , Intestinal Diseases/etiology , Intestinal Diseases/immunology , Intestinal Diseases/microbiology , Intestines/immunology , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/enzymology , Obesity/genetics
2.
J Surg Res ; 243: 363-370, 2019 11.
Article in English | MEDLINE | ID: mdl-31277013

ABSTRACT

BACKGROUND: Intestinal ischemia/reperfusion (I/R) is a grave condition related to high morbidity and mortality. Autophagy, which can induce a new cell death named type II programmed cell death, has been reported in some intestinal diseases, but little is known in I/R-induced intestinal injury. In this study, we aimed to explore the role of autophagy in intestinal injury induced by I/R and its potential mechanisms. MATERIALS AND METHODS: The rats pretreated with rapamycin or 3-methyladenine had intestinal I/R injury. After reperfusion, intestinal injury was measured by Chiu's score, intestinal mucosal wet-to-dry ratio, and lactic acid level. Intestinal mucosal oxidative stress level was measured by malondialdehyde and superoxide dismutase. Autophagosome, LC3, and p62 were detected to evaluate autophagy level. Mammalian target of rapamycin (mTOR) was detected to explore potential mechanism. RESULTS: Chiu's score, intestinal mucosal wet-to-dry ratio, lactic acid level, malondialdehyde level, autophagosomes, and LC3-II/LC3-I were significantly increased, and superoxide dismutase level and expression of p62 were significantly decreased in intestinal mucosa after intestinal ischemia/reperfusion. Pretreatment with rapamycin significantly aggravated intestinal injury evidenced by increased Chiu's score, intestinal mucosal wet-to-dry ratio and lactic acid level, increased autophagy level evidenced by increased autophagosomes and LC3-II/LC3-I and decreased expression of p62, and downregulated expression of p-mTOR/mTOR. On the contrary, pretreatment with 3-methyladenine significantly attenuated intestinal injury and autophagy level and upregulated expression of p-mTOR/mTOR. CONCLUSIONS: In summary, autophagy was significantly enhanced in intestinal mucosa after intestinal ischemia/reperfusion, and inhibition of autophagy attenuated intestinal injury induced by I/R through activating mTOR signaling.


Subject(s)
Adenine/analogs & derivatives , Autophagy/drug effects , Intestinal Diseases/prevention & control , Reperfusion Injury/prevention & control , Adenine/pharmacology , Adenine/therapeutic use , Animals , Drug Evaluation, Preclinical , Intestinal Diseases/enzymology , Intestinal Diseases/etiology , Intestinal Diseases/pathology , Intestinal Mucosa/enzymology , Intestinal Mucosa/ultrastructure , Male , Malondialdehyde/metabolism , Random Allocation , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Reperfusion Injury/etiology , Reperfusion Injury/pathology , Sirolimus , Superoxide Dismutase/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
3.
J Surg Res ; 219: 266-278, 2017 11.
Article in English | MEDLINE | ID: mdl-29078893

ABSTRACT

BACKGROUND: The pathophysiological role of pancreatic digestive hydrolases in intestinal ischemia-reperfusion (I/R) injury is still not clear. Here, we studied whether ischemia-induced injury to the small intestine can be explained by the autodigestion hypothesis. MATERIALS AND METHODS: Mesenteric I/R was induced in rats by superior mesenteric artery occlusion (90 min) and reopening (120 min). Thirty minutes before superior mesenteric artery occlusion, aprotinin (14.7 mg/kg), orlistat (5 mg/kg), and their combination or α1-proteinase inhibitor (60 mg/kg) were injected into the lumen of the small intestine. Systemic and vital parameters, intestinal microcirculation, and mucosal barrier function were monitored during the observation phase; markers of small intestinal injury, as well as trypsin-, chymotrypsin-, elastase-, and lipase-like activities in intestinal effluates were assessed at the end. RESULTS: The pattern of small intestinal injury correlated inversely with the local alterations in microvascular tissue perfusion and corresponded with the intestinal distribution of trypsin-like activity. Aprotinin almost completely inhibited trypsin-like activity (P < 0.05) and significantly reduced intestinal tissue injury. Combined with orlistat, it also increased the postischemic blood pressure (P < 0.05) but not the intestinal barrier function. Macroscopic as well as the histologic alterations were decreased by α1-proteinase inhibitor, which significantly improved postischemic blood pressure (P < 0.05). CONCLUSIONS: The I/R-induced pattern of small intestinal injury is likely to result from both local differences in tissue ischemia and the digestive activity of migrated pancreatic trypsin. Therefore, administration of aprotinin and orlistat into ischemic small intestines may be a therapeutic option in patients with a poor diagnosis.


Subject(s)
Intestinal Diseases/enzymology , Intestine, Small/enzymology , Reperfusion Injury/enzymology , Trypsin/metabolism , Animals , Aprotinin/therapeutic use , Drug Evaluation, Preclinical , Intestinal Diseases/drug therapy , Intestine, Small/blood supply , Lactones/therapeutic use , Orlistat , Rats , Reperfusion Injury/drug therapy , Splanchnic Circulation , Trypsin Inhibitors/therapeutic use
4.
J Pediatr Gastroenterol Nutr ; 64(5): 777-782, 2017 05.
Article in English | MEDLINE | ID: mdl-27482764

ABSTRACT

OBJECTIVES: Data on factors affecting absorptive function in children with intestinal failure (IF) are sparse. We evaluated duodenal disaccharidase activities and inflammation in relation to parenteral nutrition (PN) and intestinal resection in pediatric onset IF. METHODS: Disaccharidase (maltase, sucrase, and lactase) activities and histologic inflammation were evaluated from duodenal biopsies in 58 patients during PN (n = 23) or full enteral nutrition (n = 40) and in 43 matched controls. The first and the last postresection biopsies were analyzed separately after 4.3 (1.2-9.7) years and 6.5 (2.3-12.4) years, respectively. RESULTS: During PN, maltase and sucrase activities were 1.6-fold lower and mucosal inflammation more frequent (22% vs 3%) when compared to matched controls (P < 0.05 for both). In patients on full enteral nutrition, activities of maltase and sucrase were significantly higher than that in patients receiving PN and comparable to those of matched controls. Postresection time correlated positively (r = 0.448 and r = 0.369) and percentage length of the remaining small intestine inversely (r = -0.337 and r = -0.407) with maltase and sucrase activity in patients on full enteral nutrition (P < 0.05 for all), whereas proportional length of remaining colon correlated positively with maltase and lactase activity (r = 0.424-0.544, P < 0.05) in patients receiving PN. CONCLUSIONS: In children with IF, PN dependency associated with decreased duodenal maltase and sucrase activities and mucosal inflammation, which may disturb intestinal absorptive function. Localization and extent of intestinal resection and post-resection time correlated with duodenal disaccharidase activities.


Subject(s)
Disaccharidases/metabolism , Duodenum/enzymology , Intestinal Absorption , Intestinal Diseases/therapy , Intestinal Mucosa/enzymology , Parenteral Nutrition, Total , Biomarkers/metabolism , Biopsy , Case-Control Studies , Child , Child, Preschool , Combined Modality Therapy , Duodenum/pathology , Duodenum/surgery , Female , Humans , Infant , Inflammation/pathology , Intestinal Diseases/enzymology , Intestinal Diseases/pathology , Intestinal Mucosa/pathology , Male , Retrospective Studies , Withholding Treatment
5.
J Pharmacol Exp Ther ; 348(1): 86-95, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24135073

ABSTRACT

Nonsteroidal anti-inflammatory drugs (NSAIDs) can induce intestinal mucosal damage, but the underlying mechanisms remain poorly understood. The present study investigated the effects of celecoxib, etoricoxib, indomethacin, and diclofenac on small bowel integrity in rats. Male rats were treated orally with test drugs for 14 days. Animals were processed for assessment of blood hemoglobin levels and hepatic mitochondrial functions, microscopic evaluation of small intestinal damage, Western blot analysis of cyclooxygenase-1 and -2 (COX-1, COX-2) expression, and assay of malondialdehyde (MDA), myeloperoxidase (MPO), and prostaglandin E2 (PGE2) levels in small intestine. Indomethacin and diclofenac decreased blood hemoglobin levels, whereas etoricoxib and celecoxib were without effects. Celecoxib caused a lower degree of intestinal damage in comparison with the other test drugs. Indomethacin and diclofenac, but not etoricoxib or celecoxib, reduced intestinal PGE2 levels. Test drugs did not modify intestinal COX-1 expression, although they enhanced COX-2, with the exception of celecoxib, which downregulated COX-2. Indomethacin, diclofenac, and etoricoxib altered mitochondrial respiratory parameters, although celecoxib was without effects. Indomethacin or diclofenac increased MDA and MPO levels in both jejunum and ileum. In the jejunum, etoricoxib or celecoxib did not modify such parameters, whereas in the ileum, etoricoxib, but not celecoxib, increased both MDA and MPO levels. These findings suggest that nonselective NSAIDs and etoricoxib can induce enteropathy through a topic action, whereas celecoxib lacks relevant detrimental actions. The selectivity profile of COX-1/COX-2 inhibition by test drugs and the related effects on prostaglandin production do not appear to play a major role in the pathogenesis of enteropathy.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Cyclooxygenase 2 Inhibitors/toxicity , Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cyclooxygenase 2 Inhibitors/chemistry , Intestinal Diseases/chemically induced , Intestinal Diseases/enzymology , Intestinal Diseases/pathology , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Intestine, Small/enzymology , Intestine, Small/pathology , Male , Rats , Rats, Wistar
6.
J Pediatr Gastroenterol Nutr ; 59(2): 264-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24625969

ABSTRACT

OBJECTIVES: Butyrate is well known to induce apoptosis in differentiating intestinal epithelial cells. The present study was designed to examine the role of p38 mitogen-activated protein kinase (MAPK) in butyrate-induced intestinal barrier impairment. METHODS: The intestinal barrier was determined by measuring the transepithelial electrical resistance (TER) in a Caco-2 cell monolayer model. The permeability was determined by measuring transepithelial passage of fluorescein isothiocyanate-conjugated inulin (inulin-FITC). The morphology of the monolayers was examined with scanning electron microscopy. The apoptosis status was determined by annexin V-FITC labeling and flow cytometry. The activity of p38 MAPK was determined by the phosphorylation status of p38 with Western blotting. RESULTS: Butyrate at 5 mM increases the apoptosis rate of Caco-2 cells and induces impairment of intestinal barrier functions as determined by decreased TER and increased inulin-FITC permeability. Butyrate treatment activates p38 MAPK in a concentration- and time-dependent manner. SB203580, a specific p38 inhibitor, inhibits butyrate-induced Caco-2 cell apoptosis. Treatment of SB203580 significantly attenuates the butyrate-induced impairment of barrier functions in the Caco-2 cell monolayer model. CONCLUSIONS: p38 MAPK can be activated by butyrate and is involved in the butyrate-induced apoptosis and impairment of intestinal barrier function. Inhibition of p38 MAPK can significantly attenuate butyrate-induced intestinal barrier dysfunction.


Subject(s)
Apoptosis , Butyrates/adverse effects , Intestinal Absorption , Intestinal Diseases/enzymology , Intestinal Mucosa/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism , Annexin A5/metabolism , Apoptosis/drug effects , Caco-2 Cells , Electric Impedance , Enzyme Inhibitors/pharmacology , Fluorescein-5-isothiocyanate/metabolism , Humans , Imidazoles/pharmacology , Intestinal Absorption/drug effects , Intestinal Diseases/metabolism , Intestinal Mucosa/metabolism , Inulin/metabolism , Permeability , Phosphorylation , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
7.
Scand J Clin Lab Invest ; 74(5): 447-53, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24734782

ABSTRACT

OBJECTIVE: The purpose of this study in humans was to examine the influence of the gastrointestinal tract and liver on the serum concentrations of cystatin C. METHODS: Eighteen healthy volunteers and 28 patients suspected of having chronic intestinal ischemia underwent catheterization of the abdominal aorta and the central hepatic vein. Blood samples were taken simultaneously from the abdominal aorta and the central hepatic vein 60, 90 and 120 minutes after the start of the investigation. After the first blood sample, a standard liquid meal was ingested. Measurement of splanchnic blood flow was performed using the Fick principle with constant infusion of (99m)Tc-Bridatec. Angiography was performed at the end of the investigation. RESULTS: The splanchnic blood flow increased significantly postprandially in the healthy volunteers and in the patients with normal angiography by 0.613-0.698 L/min and increased non- significantly in the patients with abnormal angiography (n = 5) by 0.135 L/min on average. ANOVA and the Bonferroni's multiple comparison test showed no significant difference between the means of cystatin C, creatinine or urea in the samples taken 60, 90 and 120 minutes after the start of the investigation in the abdominal aorta and the hepatic vein in the healthy volunteers or in the patients suspected of chronic intestinal ischemia with normal angiography. CONCLUSION: There was no indication of hepatic elimination of cystatin C, creatinine or urea. The serum concentrations of cystatin C, creatinine and urea in the central hepatic vein and the abdominal aorta were independent of the splanchnic blood flow.


Subject(s)
Cystatin C/metabolism , Intestinal Diseases/blood , Intestines/blood supply , Ischemia/blood , Adult , Aged , Aged, 80 and over , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Biomarkers/blood , Case-Control Studies , Creatinine/blood , Female , Humans , Intestinal Diseases/enzymology , Ischemia/enzymology , L-Lactate Dehydrogenase/blood , Liver/blood supply , Liver/enzymology , Male , Middle Aged , Regional Blood Flow , Splanchnic Circulation , Urea/blood
8.
Zhongguo Zhong Yao Za Zhi ; 39(12): 2318-21, 2014 Jun.
Article in Zh | MEDLINE | ID: mdl-25244767

ABSTRACT

OBJECTIVE: To discuss the effect of matrine on nitric oxide (NO) and asymmetric methylarginine (ADMA) metabolism pathways in serum and tissues of mice with lipopolysaccharide (LPS) -induced intestine tissue inflammation. METHOD: Kunming mice were randomly divided into five groups: the normal control group, the LPS group and matrine (80, 40, 20 mg x kg(-1) x d(-1)) groups. The mice were intragastrically administered with drugs for 3 d (distilled water of the same volume for the normal control group and the LPS group). One hour after the last intragastrical administration, normal saline or LPS (1 mg x kg(-1)) were intraperitoneally injected. Twelve hours later, serum and tissues were collected to determine NO and ADMA levels and observe the pathological changes of intestinal tissues. The Western blot method was adopted to detect the protein expressions of arginine methyltransferases 1 (PRMT1) and dimethylarginine dimethylaminohydrolase 2 (DDAH2) in intestinal tissues. RESULT: Compared with the model group, matrine (80, 40, 20 mg x kg(-1) x d(-1)) groups showed lower NO content in serum and tissues, higher ADMA level in serum and increased PRMT1 expression in intestinal tissues, but without effect on DDAH2 expression. CONCLUSION: Matrine could inhibit LPS-induced intestine tissue inflammation in mice. Its action mechanism is related to the decreased NO content in serum and tissues and increased ADMA level in serum and PRMT1 expression in intestinal tissues.


Subject(s)
Alkaloids/administration & dosage , Arginine/analogs & derivatives , Intestinal Diseases/drug therapy , Intestines/immunology , Lipopolysaccharides/adverse effects , Nitric Oxide/metabolism , Quinolizines/administration & dosage , Animals , Arginine/blood , Arginine/metabolism , Humans , Inflammation , Intestinal Diseases/enzymology , Intestinal Diseases/immunology , Intestinal Diseases/metabolism , Intestinal Mucosa/metabolism , Intestines/drug effects , Intestines/enzymology , Male , Mice , Nitric Oxide/blood , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Matrines
9.
Curr Opin Gastroenterol ; 29(2): 146-52, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23283180

ABSTRACT

PURPOSE OF REVIEW: Several gastrointestinal diseases including the inflammatory bowel diseases (IBDs) and malignancy are associated with elevated expression of indoleamine 2,3 dioxygenase-1 (IDO1). IDO1 initiates tryptophan catabolism along a pathway that generates several bioactive kynurenine-based metabolites. Promotion of T-cell-mediated tolerance and antimicrobial effects are among the variety of functions attributed to IDO1 activity. Recent advances addressing the diverse implications of gut-associated IDO1 expression are herein reviewed. RECENT FINDINGS: In active IBD, IDO1 is highly expressed both in the cells of the lamina propria and epithelium. Experimental models demonstrate that IDO1 promotes gut immune homeostasis by limiting inflammatory responses and protecting the epithelium. In human colon cancer, high expression of IDO1 by the neoplastic epithelium correlates with poor prognosis. The serum kynurenine : tryptophan ratio is elevated in both active Crohn's disease and in colon cancer, suggesting this measurement may prove useful as a disease biomarker. IDO1 inhibitors have moved to clinical trials providing new hope as immunotherapy for advanced malignancy. SUMMARY: IDO1 activity significantly shapes gastrointestinal disease pathophysiology and severity. Measures of IDO1 activity may be useful as a disease biomarker. Manipulation of IDO1 activity has great potential as a treatment for both inflammatory and malignancy associated gastrointestinal disease.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Intestinal Diseases/enzymology , Animals , Biomarkers/blood , Biomarkers, Tumor/metabolism , Colitis/enzymology , Colonic Neoplasms/diagnosis , Gastrointestinal Diseases/diagnosis , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Inflammatory Bowel Diseases/enzymology , Mice
10.
J Pharmacol Exp Ther ; 341(2): 447-54, 2012 May.
Article in English | MEDLINE | ID: mdl-22328575

ABSTRACT

Small intestinal mucosal injury is a frequent adverse effect caused by nonsteroidal anti-inflammatory drugs (NSAIDs). The underlying mechanisms are not completely understood, but topical (luminal) effects have been implicated. Many carboxylic acid-containing NSAIDs, including diclofenac (DCF), are metabolized to acyl glucuronides (AGs), and/or ether glucuronides after ring hydroxylation, and exported into the biliary tree. In the gut, these conjugates are cleaved by bacterial ß-glucuronidase, releasing the potentially harmful aglycone. We first confirmed that DCF-AG was an excellent substrate for purified Escherichia coli ß-D-glucuronidase. Using a previously characterized novel bacteria-specific ß-glucuronidase inhibitor (Inhibitor-1), we then found that the enzymatic hydrolysis of DCF-AG in vitro was inhibited concentration dependently (IC50 ∼164 nM). We next hypothesized that pharmacologic inhibition of bacterial ß-glucuronidase would reduce exposure of enterocytes to the aglycone and, as a result, alleviate enteropathy. C57BL/6J mice were administered an ulcerogenic dose of DCF (60 mg/kg i.p.) with or without oral pretreatment with Inhibitor-1 (10 µg per mouse, b.i.d.). Whereas DCF alone caused the formation of numerous large ulcers in the distal parts of the small intestine and increased (2-fold) the intestinal permeability to fluorescein isothiocyanate-dextran, Inhibitor-1 cotreatment significantly alleviated mucosal injury and reduced all parameters of enteropathy. Pharmacokinetic profiling of DCF plasma levels in mice revealed that Inhibitor-1 coadministration did not significantly alter the C(max), half-life, or area under the plasma concentration versus time curve of DCF. Thus, highly selective pharmacologic targeting of luminal bacterial ß-D-glucuronidase by a novel class of small-molecule inhibitors protects against DCF-induced enteropathy without altering systemic drug exposure.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Diclofenac/toxicity , Glucuronidase/antagonists & inhibitors , Intestinal Diseases/chemically induced , Intestinal Diseases/enzymology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Diclofenac/pharmacokinetics , Enterocytes/drug effects , Enterocytes/enzymology , Enterocytes/metabolism , Escherichia coli/enzymology , Escherichia coli/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/metabolism , Glucuronidase/metabolism , Glycoproteins/pharmacology , Intestinal Diseases/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Intestine, Small/drug effects , Intestine, Small/enzymology , Intestine, Small/metabolism , Male , Mice , Mice, Inbred C57BL , Ulcer/chemically induced , Ulcer/enzymology , Ulcer/metabolism
11.
Am J Pathol ; 179(1): 180-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21640975

ABSTRACT

Intestinal fibrosis is a serious complication of Crohn's disease (CD) that can lead to stricture formation, which requires surgery. Mechanisms underlying intestinal fibrosis remain elusive because of a lack of suitable mouse models. Herein, we describe a spontaneous mouse model of intestinal inflammation with fibrosis and the profibrotic role of arginase I. The Src homology 2 domain-containing inositol polyphosphate 5'-phosphatase-deficient (SHIP(-/-)) mice developed spontaneous discontinuous intestinal inflammation restricted to the distal ileum starting at the age of 4 weeks. Mice developed several key features resembling CD, including inflammation and fibrosis. Inflammation was characterized by abundant infiltrating Gr-1-positive immune cells, granuloma-like immune cell aggregates that contained multinucleated giant cells, and a mixed type 2 and type 17 helper T-cell cytokine profile. Fibrosis was characterized by a thickened ileal muscle layer, collagen deposition, and increased fibroblasts at the sites of collagen deposition. SHIP(-/-) ilea had increased arginase activity and arginase I expression that was inversely proportional to nitrotyrosine staining. SHIP(-/-) mice were treated with the arginase inhibitor S-(2-boronoethyl)-l-cysteine, and changes in the disease phenotype were measured. Arginase inhibition did not affect the number of immune cell infiltrates in the SHIP(-/-) mouse ilea; rather, it reduced collagen deposition and muscle hyperplasia. These findings suggest that arginase activity is a potential target to limit intestinal fibrosis in patients with CD.


Subject(s)
Arginase/metabolism , Fibrosis/enzymology , Fibrosis/pathology , Inflammation/pathology , Intestinal Diseases/pathology , Phosphoric Monoester Hydrolases/physiology , Animals , Arginase/antagonists & inhibitors , Boronic Acids/pharmacology , Cytokines/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/etiology , Hyperplasia/metabolism , Hyperplasia/pathology , Ileum/metabolism , Ileum/pathology , Immunoenzyme Techniques , Inflammation/enzymology , Inflammation/etiology , Inositol Polyphosphate 5-Phosphatases , Intestinal Diseases/enzymology , Intestinal Diseases/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Receptors, Chemokine/metabolism , Th17 Cells
12.
J Surg Res ; 173(2): 332-40, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21316707

ABSTRACT

BACKGROUND: Gut-derived endotoxin and pathogenic bacteria have been proposed to be an important causative factor of morbidity and death during heatstroke. However, the molecular changes underlying heat stress-induced small intestine lesions have not yet been well characterized. MATERIALS AND METHODS: A heatstroke model was established in mice, and the thermal response and pathologic changes in the small intestine were examined during heat stress. Proteins extracted from the small intestine of the heated and control mice were separated by two-dimensional (2D) gel electrophoresis, and different protein spots were further identified by peptide mass fingerprint. Targeted proteins were further verified by Western blot and immunohistochemistry analysis. RESULTS: Pathologic changes in the small intestine during heat stress were found to be substantial. Using 2 D gel proteomics we identified 14 proteins that were regulated differentially in the small intestine of the mice subjected to heat stress. These 14 identified proteins, seven were down-regulated and the other seven were up-regulated, appeared to be involved in metabolism, chaperone, cell skeleton, defense, signal transduction, DNA repair, and recombination. Using Western blot and immunohistochemical analysis, we further confirmed that down-regulated expression of intestinal fructose 1,6-bisphosphatase (FBP) correlated to the severity of small intestine lesions during heat stress and cooling treatment. CONCLUSIONS: Our results identified 14 differentially expressed proteins, which may contribute to the understanding of molecular mechanisms underlying intestinal injury during heatstroke. Furthermore, intestinal FBP, one of the seven down-regulated proteins, may function as a potential marker for prognosis of gut dysfunction.


Subject(s)
Fructose-Bisphosphatase/metabolism , Heat Stroke/complications , Intestinal Diseases/etiology , Animals , Blotting, Western , Body Temperature , Electrophoresis, Gel, Two-Dimensional , Heat Stroke/enzymology , Heat Stroke/pathology , Heat Stroke/therapy , Immunohistochemistry , Intestinal Diseases/enzymology , Intestinal Diseases/pathology , Intestines/enzymology , Intestines/pathology , Male , Mass Spectrometry , Mice , Mice, Inbred BALB C , Proteome/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
13.
Anesthesiology ; 114(2): 363-73, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21245730

ABSTRACT

BACKGROUND: Renal ischemia-reperfusion injury (IRI) is a major cause of acute kidney injury and often leads to multiorgan dysfunction and systemic inflammation. Volatile anesthetics have potent antiinflammatory effects. We aimed to determine whether the representative volatile anesthetic isoflurane protects against acute kidney injury-induced liver and intestinal injury and to determine the mechanisms involved in this protection. METHODS: Mice were anesthetized with pentobarbital and subjected to 30 min of left renal ischemia after right nephrectomy, followed by exposure to 4 h of equianesthetic doses of pentobarbital or isoflurane. Five hours after renal IRI, plasma creatinine and alanine aminotransferase concentrations were measured. Liver and intestine tissues were analyzed for proinflammatory messenger RNA (mRNA) concentrations, histologic features, sphingosine kinase-1 (SK1) immunoblotting, SK1 activity, and sphingosine-1-phosphate concentrations. RESULTS: Renal IRI with pentobarbital led to severe renal, hepatic, and intestinal injury with focused periportal hepatocyte vacuolization; small-intestinal apoptosis; and proinflammatory mRNA up-regulation. Isoflurane protected against renal IRI and reduced hepatic and intestinal injury via induction of small-intestinal crypt SK1 mRNA, protein and enzyme activity, and increased sphingosine-1-phosphate. We confirmed the importance of SK1 because mice treated with a selective SK inhibitor or mice deficient in the SK1 enzyme were not protected against hepatic and intestinal dysfunction with isoflurane. CONCLUSIONS: Isoflurane protects against multiorgan injury after renal IRI via induction of the SK1/sphingosine-1-phosphate pathway. Our findings may help to unravel the cellular signaling pathways of volatile anesthetic-mediated hepatic and intestinal protection and may lead to new therapeutic applications of volatile anesthetics during the perioperative period.


Subject(s)
Acute Kidney Injury/prevention & control , Intestinal Diseases/prevention & control , Isoflurane/pharmacology , Liver Diseases/prevention & control , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Reperfusion Injury/prevention & control , Acute Kidney Injury/complications , Acute Kidney Injury/enzymology , Anesthetics, Inhalation/pharmacology , Animals , Enzyme Activation , Intestinal Diseases/enzymology , Intestinal Diseases/etiology , Intestine, Small/drug effects , Intestine, Small/enzymology , Kidney/drug effects , Kidney/enzymology , Liver/drug effects , Liver/enzymology , Liver Diseases/enzymology , Liver Diseases/etiology , Male , Mice , Mice, Inbred C57BL , Phosphotransferases (Alcohol Group Acceptor)/drug effects , Reperfusion Injury/enzymology , Up-Regulation/drug effects
14.
Biomed Pharmacother ; 134: 111130, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33348309

ABSTRACT

OBJECTIVE: Dimethyl fumarate (DMFU), a known Nrf2 activator, has proven its positive effect in different organs against ischemia/reperfusion (Is/Re) injury. Nevertheless, its possible impact to modulate intestinal Is/Re-induced injury has not been previously demonstrated before. Hence, this study aimed to investigate DMFU mechanistic maneuver against intestinal Is/Re. METHODS: To accomplish this goal, Wistar rats were allocated into four groups; Sham-operated (SOP), intestinal Is/Re (1 h/6 h), and 14 days pre-treated DMFU (15 and 25 mg/kg/day, p.o). RESULTS: The mechanistic maneuver divulged that DMFU safeguarded the intestine partly via amplifying the expression/content of Nrf2 along with enhancing its downstream, HO-1 expression/content. In addition, DMFU lessened GSK-3ß expression/content accompanied by enriching ß-catenin expression/content. The antioxidant action was affirmed by enhancing total antioxidant capacity, besides reducing MDA, iNOS, and its by-product, NOx. The DMFU action entailed anti-inflammatory character manifested by down-regulation of expression/content NF-κB with subsequent rebating the contents of TNF-α, IL-1ß, and P-selectin, as well as MPO activity. Moreover, DMFU had anti-apoptotic nature demonstrated through enriching Bcl-2 level and diminishing that of caspase-3. CONCLUSION: DMFU purveyed tenable novel protective mechanisms and mitigated events associated with intestinal Is/Re mischief either in the lower or the high dose partly by amending of oxidative stress and inflammation through the modulation of Nrf2/HO-1, GSK-3ß, and Wnt/ß-catenin pathways.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dimethyl Fumarate/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Intestinal Diseases/prevention & control , Intestines/drug effects , NF-E2-Related Factor 2/metabolism , Reperfusion Injury/prevention & control , Wnt Signaling Pathway/drug effects , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/genetics , Heme Oxygenase (Decyclizing)/genetics , Intestinal Diseases/enzymology , Intestinal Diseases/genetics , Intestinal Diseases/pathology , Intestines/enzymology , Intestines/pathology , Male , NF-E2-Related Factor 2/genetics , Nitrosative Stress/drug effects , Rats, Wistar , Reperfusion Injury/enzymology , Reperfusion Injury/genetics , Reperfusion Injury/pathology
15.
J Surg Res ; 163(1): 102-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20605598

ABSTRACT

BACKGROUND: Administration of L-nil, a selective inhibitor of inducible nitric oxide synthase (iNOS), improves ileus in an animal model of resuscitation induced intestinal edema. The purpose of this study was to elucidate the iNOS/nitric oxide (NO) signal transduction pathway in intestinal edema. MATERIALS AND METHODS: Male Sprague Dawley rats were divided into two groups; CONTROL and RESUS+VH (edema, 80 cc/kg normal saline (resuscitation) with mesenteric venous hypertension). iNOS mRNA and protein, iNOS activity, NO tissue levels, soluble guanylyl cyclase (sGC) expression, and cyclic guanosine monophosphate (cGMP) levels were measured. As a functional endpoint, we evaluated intestinal contractile strength and frequency in L-nil treated animals. RESULTS: Edema was associated with increased iNOS mRNA and protein expression without subsequent increases in iNOS activity or tissue NO levels. There was no significant change in sGC expression or increase in cGMP induced by edema. Administration of L-nil did not decrease edema development or preserve contractile strength, but increased contractile frequency. CONCLUSION: Hydrostatic intestinal edema is not associated with increased iNOS activity or tissue NO levels. Administration of L-nil in edema increases intestinal contractile frequency. This may represent a potential mechanism for the amelioration of ileus seen with the administration of L-nil.


Subject(s)
Cyclic GMP/metabolism , Edema/metabolism , Gastrointestinal Motility , Intestinal Diseases/enzymology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Animals , Guanylate Cyclase/metabolism , Hydrostatic Pressure , Immunohistochemistry , Lysine/analogs & derivatives , Male , Nitric Oxide Synthase Type II/antagonists & inhibitors , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
16.
Article in Russian | MEDLINE | ID: mdl-21061579

ABSTRACT

AIM: To study several parameters of local non-specific resistance during development of experimental dysbiosis caused by administration of wide spectrum antibiotic. MATERIALS AND METHODS: Objects of the study were colonocytes and coprofiltrates from 120 outbred mice developing experimental dysbiosis, in which activity of antioxidant enzymes (superoxid dismutase [SOD] and catalase) and level of one of the final product of lipid peroxidation--malondialdehyde (MDA). RESULTS: Decrease of SOD and catalase activity and significant increase of MDA level during development of experimental dysbiosis were established, which were associated with decreased activity of oxygen-dependent bactericidal systems of colonocytes and changes in composition of colon microbiocenosis. CONCLUSION: It was suggested that changes in parameters of local non-specific resistance could be one of mechanisms, which cause alteration of microbiocenosis composition during colon dysbiosis.


Subject(s)
Anti-Bacterial Agents/adverse effects , Catalase/metabolism , Colon/enzymology , Colon/microbiology , Intestinal Diseases/enzymology , Intestinal Diseases/microbiology , Superoxide Dismutase/metabolism , Animals , Malondialdehyde/metabolism , Mice
17.
Transplantation ; 104(12): 2507-2515, 2020 12.
Article in English | MEDLINE | ID: mdl-32639407

ABSTRACT

BACKGROUND: Prolyl hydroxylase inhibitors (PHI) promote stabilization of hypoxia-inducible factor-1 alpha and affect signaling cascades of inflammation and cell death. Their beneficial use in experimental models of ulcerative colitis and lung allograft rejection led us to test the effect of the PHI dimethyl oxalyl glycine (DMOG) in the pathophysiology of graft versus host disease (GVHD). METHODS: Acute GVHD was induced in lethally irradiated BALB/c mice. DMOG was administered intraperitoneally on alternate days for the first 2-weeks posttransplant, and then twice a week till day +50, while controls received vehicle only. Animals were monitored for clinical GVHD and analyzed at day +7 and at day +50. RESULTS: DMOG treatment of allogeneic recipients improved survival by day +50, which was associated with decreased early gut injury and serum tumor necrosis factor-α compared with allogeneic controls. DMOG treatment of allogeneic recipients resulted in increased hypoxia-inducible factor-1 alpha expression and reduced apoptosis in the terminal ileum via Fas-associated protein with death domain protein repression along with decreased T-cell infiltration. Reduced pathology in colon after DMOG treatment associates with intestinal epithelium integrity and reduced damage caused by diminished recruitment of neutrophils. CONCLUSIONS: Taken together, we show protective effects of DMOG on early gut GVHD and improved survival in a model of allogeneic hematopoietic cell transplantation, providing the rationale for further evaluation of PHIs, in the prevention and treatment of acute GVHD.


Subject(s)
Amino Acids, Dicarboxylic/pharmacology , Colon/drug effects , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Ileum/drug effects , Intestinal Diseases/prevention & control , Prolyl Hydroxylases/metabolism , Prolyl-Hydroxylase Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Colon/enzymology , Colon/immunology , Colon/pathology , Graft vs Host Disease/enzymology , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ileum/enzymology , Ileum/immunology , Ileum/pathology , Intestinal Diseases/enzymology , Intestinal Diseases/immunology , Intestinal Diseases/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Time Factors , Transplantation, Homologous/adverse effects , Treatment Outcome , Whole-Body Irradiation
18.
Int Immunopharmacol ; 89(Pt A): 107014, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33039959

ABSTRACT

Acute lung injury (ALI) is a life-threatening disorder with high rates of morbidity and mortality. Up to now, there are still no effective drugs for its therapies due to the complexity of its etiology and pathogenesis. In this present study, we investigated the protective effect of Nervilifordin F (NF) on ALI induced by intestinal ischemia/reperfusion (II/R) and its related mechanism. Firstly, the ALI model rats were induced through II/R, and treated with NF. Then, the pathological and cytokine level changes in the lung tissue of ALI rats were evaluated by hematoxylin and eosin and enzyme-linked immunosorbent assay (ELISA). The related genes expression level of mammalian target of rapamycin (mTOR) pathway and inflammasome were measured by real-time quantitative polymerase chain reaction (RT-qPCR), western blot and immunohistochemistry. Finally, the NF-protein complexes were predicted by SYBYL-X 2.0. The results indicated that NF can significant reduces the levels of tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6 and IL-1ß, and inhibits the expression of inflammasome related genes (such as toll-like receptor 4 (TLR4), p65, NOD-like receptor protein 3 (NLRP3) and Caspase 1), thereby reduce inflammation in II/R-induced ALI rats. Moreover, NF can activate the expression of FK506 binding protein 25 (FKBP25) and down-regulate the expression of mTOR and p70 ribosomal protein S6 kinase 1 (p70S6K). In addition, molecular docking results showed that NF can be combined well with p70S6K, TLR4, mTOR and NLRP3, which further verified the inhibitory effect of NF on ALI inflammation. Therefore, the findings indicate that NF can alleviates II/R-induced inflammation of ALI rats by inhibiting inflammasome related genes and mTOR pathway, which expected to use as a potential drug for the treatment of ALI.


Subject(s)
Acute Lung Injury/prevention & control , Anti-Inflammatory Agents/pharmacology , Inflammasomes/metabolism , Intestinal Diseases/drug therapy , Lung/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reperfusion Injury/drug therapy , TOR Serine-Threonine Kinases/metabolism , Acute Lung Injury/enzymology , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation , Inflammasomes/genetics , Intestinal Diseases/complications , Intestinal Diseases/enzymology , Intestinal Diseases/pathology , Lung/enzymology , Lung/pathology , Male , Molecular Docking Simulation , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Rats, Sprague-Dawley , Reperfusion Injury/complications , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
19.
Sci Prog ; 92(Pt 3-4): 241-87, 2009.
Article in English | MEDLINE | ID: mdl-19960866

ABSTRACT

A staggering 4000 million people cannot digest lactose, the sugar in milk, properly. All mammals, apart from white Northern Europeans and few tribes in Africa and Asia, lose most of their lactase, the enzyme that cleaves lactose into galactose and glucose, after weaning. Lactose intolerance causes gut and a range of systemic symptoms, though the threshold to lactose varies considerably between ethnic groups and individuals within a group. The molecular basis of inherited hypolactasia has yet to be identified, though two polymorphisms in the introns of a helicase upstream from the lactase gene correlate closely with hypolactasia, and thus lactose intolerance. The symptoms of lactose intolerance are caused by gases and toxins produced by anaerobic bacteria in the large intestine. Bacterial toxins may play a key role in several other diseases, such as diabetes, rheumatoid arthritis, multiple sclerosis and some cancers. The problem of lactose intolerance has been exacerbated because of the addition of products containing lactose to various foods and drinks without being on the label. Lactose intolerance fits exactly the illness that Charles Darwin suffered from for over 40 years, and yet was never diagnosed. Darwin missed something else--the key to our own evolution--the Rubicon some 300 million years ago that produced lactose and lactase in sufficient amounts to be susceptible to natural selection.


Subject(s)
Intestinal Diseases/metabolism , Lactase/metabolism , Lactose Intolerance/metabolism , Lactose/metabolism , Carbohydrate Sequence , Humans , Intestinal Diseases/enzymology , Intestinal Diseases/genetics , Lactase/genetics , Lactose Intolerance/enzymology , Lactose Intolerance/genetics , Molecular Sequence Data
20.
Dig Dis Sci ; 54(4): 738-44, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18683050

ABSTRACT

AIM: Intestinal ischemia reperfusion (IR) causes tissue injury in two ways, starting a pro-inflammatory cascade and oxidative stress. The aim of this study was to investigate the possible protective effects of caffeic acid phenethyl ester (CAPE), which has antioxidant and anti-inflammatory properties, against intestinal IR injury in rats. MATERIALS AND METHODS: Forty male Wistar-Albino rats were divided into five groups: Sham, IR, IR plus ethanol (vehicle), IR plus 10 mg/kg (IR + 10CAPE), and 30 mg/kg CAPE (IR + 30CAPE) at the 30-min ischemic period. Intestines were exteriorized and the superior mesenteric artery was occluded for 45-min ischemia and then the clamp was removed for 120-min reperfusion. After the experiment, the intestines were removed for biochemical and light microscopic examinations. Additionally, blood samples were taken for plasma TNF-alpha measurement. RESULTS: The TBARS levels of the IR and IR + Ethanol groups were higher than the Sham group (P < 0.05). Both CAPE treatments decreased TBARS levels in comparison with the IR group (P < 0.05). In both CAPE-treated groups, while the superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activities were increased compared to all other groups, which was similarly the case for the CAT activity compared to the Sham and IR + Ethanol groups (P < 0.05). There were no significant differences between GSH levels of all study groups. The TNF-alpha levels of the IR and IR + Ethanol groups were non-significantly increased compared to the Sham group (P > 0.05). The TNF-alpha levels of 10 and 30 mg/kg CAPE groups were non-significantly decreased compared to the IR group (P > 0.05). The tissue MPO activities of the IR and IR + Ethanol groups were higher than the Sham group (P < 0.05). The MPO activities of the IR + 10CAPE and IR + 30CAPE groups were not significantly different from the Sham group (P > 0.05). There was necrosis of mucosa in the IR and IR + Ethanol groups in light microscopic evaluations. Those changes were significantly reversed by 30 mg/kg CAPE treatment. CONCLUSION: The intestinal IR injury may be reversed by anti-inflammatory and antioxidant actions of the CAPE. However, 30 mg/kg CAPE treatment may be more efficient in preventing intestinal IR injury in rats.


Subject(s)
Antioxidants/metabolism , Caffeic Acids/therapeutic use , Intestinal Diseases/drug therapy , Reperfusion Injury/drug therapy , Animals , Catalase/metabolism , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Intestinal Diseases/enzymology , Intestinal Diseases/pathology , Intestinal Mucosa/pathology , Jejunum/pathology , Lipid Peroxidation , Male , Malondialdehyde/metabolism , Peroxidase/metabolism , Phenylethyl Alcohol/analogs & derivatives , Rats , Rats, Wistar , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/blood
SELECTION OF CITATIONS
SEARCH DETAIL