Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 177
1.
Biomed Pharmacother ; 134: 111130, 2021 Feb.
Article En | MEDLINE | ID: mdl-33348309

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.


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
2.
Int Immunopharmacol ; 89(Pt A): 107014, 2020 Dec.
Article En | MEDLINE | ID: mdl-33039959

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.


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
3.
Transplantation ; 104(12): 2507-2515, 2020 12.
Article En | MEDLINE | ID: mdl-32639407

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.


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
4.
Proc Natl Acad Sci U S A ; 117(15): 8431-8436, 2020 04 14.
Article En | MEDLINE | ID: mdl-32220957

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.


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
5.
J Surg Res ; 243: 363-370, 2019 11.
Article En | MEDLINE | ID: mdl-31277013

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.


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
6.
Sci Rep ; 9(1): 8742, 2019 06 19.
Article En | MEDLINE | ID: mdl-31217465

With increased climate change pressures likely to influence harmful algal blooms, exposure to microcystin, a known hepatotoxin and a byproduct of cyanobacterial blooms can be a risk factor for NAFLD associated comorbidities. Using both in vivo and in vitro experiments we show that microcystin exposure in NAFLD mice cause rapid alteration of gut microbiome, rise in bacterial genus known for mediating gut inflammation and lactate production. Changes in the microbiome were strongly associated with inflammatory pathology in the intestine, gut leaching, tight junction protein alterations and increased oxidative tyrosyl radicals. Increased lactate producing bacteria from the altered microbiome was associated with increased NOX-2, an NADPH oxidase isoform. Activationof NOX2 caused inflammasome activation as shown by NLRP3/ASCII and NLRP3/Casp-1 colocalizations in these cells while use of mice lacking a crucial NOX2 component attenuated inflammatory pathology and redox changes. Mechanistically, NOX2 mediated peroxynitrite species were primary to inflammasome activation and release of inflammatory mediators. Thus, in conclusion, microcystin exposure in NAFLD could significantly alter intestinal pathology especially by the effects on microbiome and resultant redox status thus advancing our understanding of the co-existence of NAFLD-linked inflammatory bowel disease phenotypes in the clinic.


Environmental Exposure/adverse effects , Gastrointestinal Microbiome/drug effects , Intestinal Diseases , Microcystins/administration & dosage , NADPH Oxidase 2/metabolism , Non-alcoholic Fatty Liver Disease , Animals , Disease Models, Animal , Inflammation/chemically induced , Inflammation/enzymology , Inflammation/microbiology , Inflammation/pathology , Intestinal Diseases/chemically induced , Intestinal Diseases/enzymology , Intestinal Diseases/microbiology , Intestinal Diseases/pathology , Male , Mice , Mice, Knockout , Microcystins/pharmacology , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/microbiology , Non-alcoholic Fatty Liver Disease/pathology
7.
J Int AIDS Soc ; 21(7): e25144, 2018 07.
Article En | MEDLINE | ID: mdl-29987877

INTRODUCTION: Combined anti-retroviral therapy (cART) transformed HIV-1 from a deadly disease into a chronic infection, but does not cure HIV infection. It also does not fully restore HIV-induced gut damage unless administered extremely early after infection. Additional biomarkers are needed to evaluate the capacity of therapies aimed at HIV remission/cure to restore HIV-induced intestinal immune damage and limit chronic inflammation. Herein, we aimed to identify a systemic surrogate marker whose levels would reflect gut immune damage such as intestinal Th17 cell loss starting from primary HIV-1 infection. METHODS: Biomarker discovery approaches were performed in four independent cohorts, covering HIV-1 primary and chronic infection in 496 naïve or cART-treated patients (Amsterdam cohort (ACS), ANRS PRIMO, COPANA and CODEX cohorts). The concentration and activity of soluble Dipeptidylpeptidase 4 (sDPP4) were quantified in the blood from these patients, including pre- and post-infection samples in the ACS cohort. For quantification of DPP4 in the gut, we utilized two non-human primate models, representing pathogenic (macaque) and non-pathogenic (African green monkey) SIV infection. Four gut compartments were analysed in each animal model (ileum, jejunum, colon and rectum) for quantification of DPP4, RORC and TBX21 gene expression in sorted CD4+ cells. To analyse if sDPP4 levels increase when Th17 cells were restored, we quantified sDPP4 in plasma from SIV-infected macaques treated with IL-21. RESULTS: We showed that sDPP4 levels were strongly decreased in primary HIV-1 infection. Strikingly, sDPP4 levels in primary HIV-1 infection predicted time to AIDS. They were not increased by cART in chronic HIV-1 infection (median 36 months on cART). In the gut of SIV-infected non-human primates, DPP4 mRNA was higher in CD4+ than CD4- leucocytes. DPP4 specifically correlated with RORC expression, a Th17 marker, in CD4+ cells from the intestine. We further demonstrated that sDPP4 activity levels were increased in animals treated with IL-21 and that this increase was associated with restoration of the Th17 compartment and reduced inflammation. Furthermore, DPP4 mRNA levels in small intestine CD4+ cells positively correlated with circulating DPP4 activity. CONCLUSION: These data provide evidence that blood sDPP4 levels could be useful as a correlate for HIV-induced intestinal damage.


CD4-Positive T-Lymphocytes/immunology , Dipeptidyl Peptidase 4/blood , HIV Infections/enzymology , Intestinal Diseases/virology , Adult , Animals , Biomarkers , CD4 Lymphocyte Count , Chlorocebus aethiops , HIV Infections/complications , HIV Infections/drug therapy , HIV-1/immunology , Humans , Interleukins , Intestinal Diseases/enzymology , Intestinal Diseases/immunology , Intestinal Diseases/pathology , Macaca , Male , Nuclear Receptor Subfamily 1, Group F, Member 3 , Simian Acquired Immunodeficiency Syndrome/blood , Th17 Cells/immunology
8.
Acta Vet Scand ; 60(1): 16, 2018 Mar 12.
Article En | MEDLINE | ID: mdl-29530095

BACKGROUND: Matrix metalloproteinases (MMPs) 2 and 9 are zinc- and calcium-dependent endopeptidases involved in the breakdown and reconstitution of extracellular matrix under both physiological and pathological conditions. Mucosal MMP-2 and -9 activities have been reported to be upregulated in the intestine of humans with inflammatory bowel disease (IBD), and in animal models of IBD. However, their involvement in the pathogenesis of canine chronic enteropathies (CE) is unknown. This study investigated mucosal pro- and active MMP-2 and -9 activities in dogs with CE and healthy dogs using gelatin zymography, and also to determine the association of their activities in dogs with CE with the canine IBD activity index (CIBDAI), histopathologic findings, the clinical outcome, and hypoalbuminemia. Intestinal mucosal samples from duodenum, ileum, colon, and cecum were collected from 40 dogs with CE and 18 healthy Beagle dogs. RESULTS: In dogs with CE, the number of samples positive for mucosal pro- and active MMP-2 was significantly higher in the duodenum (P < 0.0001 and P = 0.011, respectively), ileum (P = 0.002 and P = 0.018, respectively), and colon (P < 0.0001 and P = 0.002, respectively), compared with healthy controls. Mucosal pro-MMP-9-positive samples in the duodenum and colon were significantly more frequent in dogs with CE than in healthy dogs (P = 0.0004 and P = 0.001, respectively). Despite the presence of mucosal samples positive for active MMP-9 in the intestinal segments of dogs with CE, the difference compared to healthy controls did not reach statistical significance. None of the intestinal mucosal samples in healthy dogs showed gelatinolytic activity corresponding to the control bands of active MMP-2 and -9. Mucosal active MMP-9 activities displayed a significant positive association with the severity of neutrophil infiltration in the duodenum (P = 00.040), eosinophils in the cecum (P = 00.037), and the CIBDAI score for ileum samples (P = 0.023). There was no significant association of pro- and active MMP-2 and -9 levels with the clinical outcome or hypoalbuminemia. CONCLUSIONS: This study is the first to demonstrate upregulation of mucosal pro- and active MMP-2 and pro-MMP-9 in the intestine of dogs with CE compared to healthy dogs. The results provide supporting evidence for the possible involvement of MMP-2 and -9 in the pathogenesis of canine CE.


Dog Diseases/enzymology , Intestinal Diseases/veterinary , Intestinal Mucosa/enzymology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Animals , Chronic Disease , Dogs , Intestinal Diseases/enzymology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Up-Regulation
9.
Acta Cir Bras ; 32(11): 935-948, 2017 Nov.
Article En | MEDLINE | ID: mdl-29236798

PURPOSE: To investigate the expression of nitric oxide synthase (NOS) and apoptosis associated with ischemic preconditioning (IPC) and pentoxifylline (PTX) in intestinal ischemia (I) and reperfusion (R) injury. METHODS: Thirty male rats were assigned to 5 groups: (CG), no clamping of the superior mesenteric artery (90 minutes); (IR-SS) saline + ischemia (30 minutes) + reperfusion (60 minutes); (IR-PTX) PTX + ischemia (30 minutes) + reperfusion (60 minutes); (IPC-IR-SS) 5 minutes of ischemia + 5 minutes of reperfusion (IPC) + saline + I(30 minutes)+R(60 minutes); and (IPC-IR-PTX) IPC + PTX + I(30 minutes)+ R(60 minutes). RESULTS: The application of IPC and PTX showed a significantly lower immunohistochemistry reaction for active caspase-3 (P<0.05) compared to IR+SS. The number of cells immunoreactive to BCL-2 was higher in the IR-PTX group (P>0.05). The NOS-2 expression (qRTPCR) in the IR-PTX group (P<0.05) was higher than the values for the IPC+IR-SS and IPC-IR-PTX groups. The NOS-3 expression was significantly upper in the IPC-IR-PTX group than in the CG (P<0.05), the IR-SS (P<0.05) and the IR-PTX (P<0.05) groups. CONCLUSIONS: The BCL-2 and active caspase-3 showed beneficial effects on PTX and IPC. The expression of NOS-2 and NOS-3 in the IPC and IPC-PTX groups showed no synergistic effect.


Apoptosis/drug effects , Intestinal Diseases/prevention & control , Intestines/blood supply , Ischemic Preconditioning , Nitric Oxide Synthase/metabolism , Pentoxifylline/therapeutic use , Animals , Apoptosis/physiology , Disease Models, Animal , Humans , Immunohistochemistry , Intestinal Diseases/enzymology , Intestines/pathology , Male , RNA, Messenger/analysis , Rats , Rats, Wistar , Vasodilator Agents/therapeutic use
10.
Acta cir. bras ; 32(11): 935-948, Nov. 2017. graf
Article En | LILACS | ID: biblio-886187

Abstract Purpose: To investigate the expression of nitric oxide synthase (NOS) and apoptosis associated with ischemic preconditioning (IPC) and pentoxifylline (PTX) in intestinal ischemia (I) and reperfusion (R) injury. Methods: Thirty male rats were assigned to 5 groups: (CG), no clamping of the superior mesenteric artery (90 minutes); (IR-SS) saline + ischemia (30 minutes) + reperfusion (60 minutes); (IR-PTX) PTX + ischemia (30 minutes) + reperfusion (60 minutes); (IPC-IR-SS) 5 minutes of ischemia + 5 minutes of reperfusion (IPC) + saline + I(30 minutes)+R(60 minutes); and (IPC-IR-PTX) IPC + PTX + I(30 minutes)+ R(60 minutes). Results: The application of IPC and PTX showed a significantly lower immunohistochemistry reaction for active caspase-3 (P<0.05) compared to IR+SS. The number of cells immunoreactive to BCL-2 was higher in the IR-PTX group (P>0.05). The NOS-2 expression (qRTPCR) in the IR-PTX group (P<0.05) was higher than the values for the IPC+IR-SS and IPC-IR-PTX groups. The NOS-3 expression was significantly upper in the IPC-IR-PTX group than in the CG (P<0.05), the IR-SS (P<0.05) and the IR-PTX (P<0.05) groups. Conclusions: The BCL-2 and active caspase-3 showed beneficial effects on PTX and IPC. The expression of NOS-2 and NOS-3 in the IPC and IPC-PTX groups showed no synergistic effect.


Humans , Animals , Male , Rats , Pentoxifylline/therapeutic use , Apoptosis/drug effects , Nitric Oxide Synthase/metabolism , Ischemic Preconditioning , Intestinal Diseases/prevention & control , Intestines/blood supply , Vasodilator Agents/therapeutic use , RNA, Messenger/analysis , Immunohistochemistry , Rats, Wistar , Apoptosis/physiology , Disease Models, Animal , Intestinal Diseases/enzymology , Intestines/pathology
11.
J Surg Res ; 219: 266-278, 2017 11.
Article En | MEDLINE | ID: mdl-29078893

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.


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
12.
Open Biol ; 7(8)2017 08.
Article En | MEDLINE | ID: mdl-28835570

Adenosine monophosphate (AMP)-activated protein kinase (AMPK), a highly conserved energy sensor, has a crucial role in cardiovascular, neurodegenerative and inflammatory diseases, as well as in cancer and metabolic disorders. Accumulating studies have demonstrated that AMPK activation enhances paracellular junctions, nutrient transporters, autophagy and apoptosis, and suppresses inflammation and carcinogenesis in the intestine, indicating an essential role of AMPK in intestinal health. AMPK inactivation is an aetiological factor in intestinal dysfunctions. This review summarizes the favourable outcomes of AMPK activation on intestinal health, and discusses AMPK as a potential therapeutic target for intestinal diseases.


AMP-Activated Protein Kinases/metabolism , Enzyme Activation , Intestinal Diseases/enzymology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Autophagy , Humans , Intestinal Absorption , Intestinal Diseases/drug therapy , Signal Transduction/drug effects
13.
J Pediatr Gastroenterol Nutr ; 64(5): 777-782, 2017 05.
Article En | MEDLINE | ID: mdl-27482764

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.


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
14.
Toxins (Basel) ; 8(11)2016 11 19.
Article En | MEDLINE | ID: mdl-27869757

Clostridium perfringens is a major cause of histotoxic and intestinal infections of humans and other animals. This Gram-positive anaerobic bacterium can produce up to three sialidases named NanH, NanI, and NanJ. The role of sialidases in histotoxic infections, such as gas gangrene (clostridial myonecrosis), remains equivocal. However, recent in vitro studies suggest that NanI may contribute to intestinal virulence by upregulating production of some toxins associated with intestinal infection, increasing the binding and activity of some of those toxins, and enhancing adherence of C. perfringens to intestinal cells. Possible contributions of NanI to intestinal colonization are further supported by observations that the C. perfringens strains causing acute food poisoning in humans often lack the nanI gene, while other C. perfringens strains causing chronic intestinal infections in humans usually carry a nanI gene. Certain sialidase inhibitors have been shown to block NanI activity and reduce C. perfringens adherence to cultured enterocyte-like cells, opening the possibility that sialidase inhibitors could be useful therapeutics against C. perfringens intestinal infections. These initial in vitro observations should be tested for their in vivo significance using animal models of intestinal infections.


Bacterial Proteins , Bacterial Toxins , Clostridium perfringens/enzymology , Neuraminidase , Animals , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/genetics , Clostridium Infections/drug therapy , Clostridium Infections/enzymology , Clostridium perfringens/genetics , Clostridium perfringens/pathogenicity , Gene Expression Regulation, Fungal , Humans , Intestinal Diseases/drug therapy , Intestinal Diseases/enzymology , Neuraminidase/antagonists & inhibitors , Neuraminidase/genetics , Virulence
15.
Shock ; 45(4): 359-66, 2016 Apr.
Article En | MEDLINE | ID: mdl-26263437

Intestinal ischemia-reperfusion (I/R) occurs in various clinical situations and causes local and remote organ injury, especially in the lungs, leading to significant morbidity and mortality. The maintenance of mitochondrial biogenesis is essential for cell survival and is regulated in part by sirtuin 1 (SIRT1), an energy-sensing enzyme. We hypothesized that SIRT1 activation with SRT1720 would reduce local and remote organ injury after intestinal I/R. Intestinal I/R was induced by the occlusion of the superior mesenteric artery of adult male C57BL/6 mice for 45 min, followed by reperfusion for 4 h. SRT1720 or vehicle was injected intravenously at the time of reperfusion. Blood, small intestine, and lung tissues were collected for analysis. The SRT1720 treatment of I/R mice resulted in a 57% increase in protein levels of succinate dehydrogenase, an index of mitochondrial mass, and a 120% increase in messenger RNA levels of mitochondrial transcription factor A, a marker for mitochondrial biogenesis. The microscopic architecture and apoptosis of the gut tissue was improved in the SRT1720-treated I/R mice. SRT1720 decreased intestinal messenger RNA levels of tumor necrosis factor-α by 60% and inducible nitric oxide synthase to baseline after I/R. Systemic inflammation, as determined by serum interleukin-6, was reduced in treated mice. Lung injury, as measured by histological architecture and myeloperoxidase activity, and lung apoptosis were also improved after the SRT1720 treatment. SRT1720 preserved mitochondrial biogenesis and mass, leading to inhibition of inflammation and oxidative stress, thereby protecting against intestinal I/R-induced injury. Thus, the SIRT1-mediated pathway is a promising target for the treatment of intestinal I/R injury.


Enzyme Activators/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Intestinal Diseases/prevention & control , Intestines/enzymology , Reperfusion Injury/prevention & control , Sirtuin 1/metabolism , Animals , Intestinal Diseases/enzymology , Intestinal Diseases/pathology , Intestines/blood supply , Intestines/pathology , Male , Mice , Reperfusion Injury/enzymology , Reperfusion Injury/pathology
16.
Zhongguo Zhong Yao Za Zhi ; 39(12): 2318-21, 2014 Jun.
Article Zh | MEDLINE | ID: mdl-25244767

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.


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
17.
PLoS One ; 9(6): e99134, 2014.
Article En | MEDLINE | ID: mdl-24927128

Pre-induction of heme oxygenase (HO)-1, which is regarded as an effective method of "organ preconditioning", exerts beneficial effects during hemorrhagic shock (HS). However, the available HO-1 inducers exhibit disadvantages such as toxicity or complex technical requirements. Therefore, a safe and convenient HO-1 inducer would be promising and could be exploited in the treatment of foreseeable hemorrhaging, such as prior to major surgery. Here we investigated the effect of vitamin C (VitC), a common antioxidant, on intestinal HO-1 expression and examined whether VitC pretreatment prevented HS related intestinal tissue injuries after HO-1 induction. First, we conducted an in vitro study and found that HO-1 expression in rat intestinal epithelial cells (IEC-6) was induced by non-toxic VitC in a time and concentration dependent manner, and the mechanism was related to the activation of extracellular signal-regulated kinase 1/2 (ERK1/2). Next, we conducted an in vivo study and found that VitC induced intestinal HO-1 protein expression (mainly observed in the intestinal epithelial cells) and HO-1 activity in normal SD rats, and that these HO-1 levels were further enhanced by VitC in a rat model of HS. The HS related intestinal injuries, including histological damage, pro-inflammatory cytokine levels (tumor necrosis factor and interleukin-6), neutrophil infiltration and apoptosis decreased after VitC pretreatment, and this alleviating of organ injuries was abrogated after the inhibition of HO-1 activity by zinc protoporphyrin-IX. It was of note that VitC did little histological damage to the intestine of the sham rats. These data suggested that VitC might be applied as a safe inducer of intestinal HO-1 and that VitC pretreatment attenuated HS related intestinal injuries via the induction of HO-1.


Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Heme Oxygenase-1/metabolism , Intestinal Diseases/prevention & control , Intestines/injuries , Shock, Hemorrhagic/complications , Animals , Antioxidants/administration & dosage , Apoptosis/drug effects , Ascorbic Acid/administration & dosage , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation/drug effects , Intestinal Diseases/enzymology , Intestines/drug effects , Intestines/enzymology , MAP Kinase Signaling System/drug effects , Rats , Rats, Sprague-Dawley , Shock, Hemorrhagic/enzymology , Shock, Hemorrhagic/pathology
18.
Scand J Clin Lab Invest ; 74(5): 447-53, 2014 Aug.
Article En | MEDLINE | ID: mdl-24734782

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.


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
19.
J Pediatr Gastroenterol Nutr ; 59(2): 264-9, 2014 Aug.
Article En | MEDLINE | ID: mdl-24625969

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.


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
20.
J Pharmacol Exp Ther ; 348(1): 86-95, 2014 Jan.
Article En | MEDLINE | ID: mdl-24135073

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.


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
...