Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Tissue Barriers ; : 2327776, 2024 Mar 17.
Article in English | MEDLINE | ID: mdl-38494646

ABSTRACT

Gut-vascular barrier (GVB) is the second barrier in mucosa to control systemic dissemination of gut bacteria. Severe burns induce enteroglial cells to produce S100B and endothelial cells to generate ADAM10 and cause vitamin D3 insufficiency/deficiency and GVB disruption. It is not clear whether vitamin D3 supplementation attenuates GVB damage via regulation of S100B/ADAM10 pathway. Here, GVB disruption was induced by 30% of total body surface area scalds. Rats were treated with 1,25(OH)2D3 (0.05, 0.5 or 5 µg/kg) or S100B monoclonal antibody (S100BmAb, 10 µg/kg) or GI254023X (ADAM10 inhibitor, 100 mg/kg). Rat enteric glial cell-line CRL2690 and rat intestinal microvascular endothelial cells (RIMECs) were treated with S100B (5 µM) or plus 1,25(OH)2D3 (0.05, 0.5 or 5 µM) or GI254023X (5 µM). S100B, TNF-α, 25(OH)D3 and 1,25(OH)2D3 in serum and gut mucosa were determined by enzyme-linked immunosorbent assay. The endothelial permeability was measured using FITC-dextran 70 kDa. ADAM10 and ß-catenin expression was assayed by Western blot. The results showed that 1,25(OH)2D3 and 25(OH)D3 concentration in serum reduced whereas TNF-α and S100B in serum and gut mucosa increased in burned rats. S100BmAb, GI254023X and 1,25(OH)2D3 treatment lowered burns-increased GVB permeability. 1,25(OH)2D3 also decreased S100B concentration in serum and gut mucosa. 1,25(OH)2D3 inhibited S100B release from TNF-α-treated CRL2690 and raised ß-catenin while decreasing ADAM10 protein in S100B-treated RIMECs. 1,25(OH)2D3 and GI254023X also decreased the endothelial permeability of S100B-treated RIMECs. Collectively, these findings provide evidence that severe burns lower serum 25(OH)D3 and 1,25(OH)2D3 concentration. 1,25(OH)2D3 supplementation alleviates burns-elicited GVB disruption via inhibition of S100B/ADAM10 signaling.

2.
Pharm Biol ; 62(1): 53-61, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38108311

ABSTRACT

Context: Berberine (BBR) can regulate enteric glial cells (EGCs) and the gut vascular barrier (GVB).Objective: To explore whether BBR regulates GVB permeability via the S100B pathway.Materials and methods: GVB hyperpermeability in C57BL/6J mice was induced by burns or S100B enema. BBR (25 or 50 mg/kg/d, 3 d) was gavaged preburn. S100B monoclonal antibody (S100BmAb) was i.v. injected postburn. Mouse intestinal microvascular endothelial cells (MIMECs) were treated with S100B, S100B plus BBR, or Z-IETD-FMK. GVB permeability was assayed by FITC-dextran, S100B by ELISA, caspase-8, ß-catenin, occludin and PV-1 by immunoblot.Results: Burns elevated S100B in serum and in colonic mucosa to a peak (147.00 ± 4.95 ng/mL and 160.30 ± 8.50 ng/mg, respectively) at 36 h postburn, but BBR decreased burns-induced S100B in serum (126.20 ± 6.30 or 90.60 ± 3.78 ng/mL) and in mucosa (125.80 ± 12.40 or 91.20 ± 8.54 ng/mg). Burns raised GVB permeability (serum FITC-dextran 111.40 ± 8.56 pg/mL) at 48 h postburn, but BBR reduced GVB permeability (serum FITC-dextran 89.20 ± 6.98 or 68.60 ± 5.50 ng/mL). S100B enema (1 µM) aggravated burns-raised GVB permeability (142.80 ± 8.07 pg/mL) and PV-1, but the effect of S100B was antagonized by BBR. Z-IETD-FMK (5 µM) increased S100B-induced permeability to FITC-dextran (205.80 ± 9.70 to 263.80 ± 11.04 AUs) while reducing ß-catenin in MIMECs. BBR (5 µM) reduced S100B-induced permeability (104.20 ± 9.65 AUs) and increased caspase-8, ß-catenin and occludin.Discussion and conclusion: BBR decreases burns-induced GVB hyperpermeability via modulating S100B/caspase-8/ß-catenin pathway and may involve EGCs.


Subject(s)
Berberine , Burns , Animals , Mice , Mice, Inbred C57BL , Berberine/pharmacology , Caspase 8 , Endothelial Cells , Occludin , beta Catenin , Burns/drug therapy
3.
Life Sci ; 261: 118460, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32961234

ABSTRACT

AIMS: The hyperpermeability of gut-vascular barrier (GVB) plays a role in gut-derived sepsis. The goal of this study was to evaluate if berberine might improve hepatic apolipoprotein M (ApoM) generation and raise plasma ApoM level to protect the compromised GVB. MATERIALS AND METHODS: The compromised GVB was induced by sepsis. Hepatic ApoM mRNA and phosphoenolpyruvate carboxykinase (PEPCK) mRNA and plasma ApoM level were assayed by qRT-PCR and ELISA, respectively. The permeability of intestinal capillary in vivo and of rat intestinal microvascular endothelial cells (RIMECs) in vitro was assayed by FITC-dextran. The blood glucose was detected by a glucometer. Plasma insulin, TNF-α and IL-1ß were assayed by ELISA. The plasmalemma vesicle-associated protein-1 (PV1), ß-catenin and occludin in RIMECs were assayed by Western blot. KEY FINDINGS: Sepsis decreased hepatic ApoM mRNA and plasma ApoM level, but raised hepatic PEPCK mRNA and plasma glucose, insulin, TNF-α, and IL-1ß levels. The increased vascular endothelial permeability was abrogated by recombinant rat ApoM in vivo or ApoM-bound S1P in vitro. ApoM-bound S1P decreased PV1 but increased occludin and ß-catenin expression in LPS-treated RIMECs. Berberine in a dose-dependent manner raised hepatic ApoM mRNA and plasma ApoM level, but decreased septic hyperglycemia, insulin resistance and plasma TNF-α and IL-1ß levels. Berberine reduced sepsis-induced PEPCK and TLR4 mRNA overexpression in the liver. SIGNIFICANCE: This study demonstrated berberine inhibited TLR4-mediated hyperglycemia, insulin resistance and proinflammatory molecule production, thereby increasing ApoM gene expression and plasma ApoM. Berberine protected the damaged GVB via modulation of ApoM/S1P pathway.


Subject(s)
Apolipoproteins M/metabolism , Berberine/therapeutic use , Capillary Permeability/drug effects , Lysophospholipids/metabolism , Sepsis/drug therapy , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Animals , Berberine/pharmacology , Disease Models, Animal , Gastrointestinal Tract/blood supply , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/physiopathology , Hep G2 Cells , Humans , Male , Rats, Wistar , Sepsis/metabolism , Sepsis/physiopathology , Sphingosine/metabolism
4.
Life Sci ; 233: 116697, 2019 Sep 15.
Article in English | MEDLINE | ID: mdl-31351968

ABSTRACT

AIMS: The present study investigated if berberine might induce Zrt-Irt-like protein 14 (ZIP14) and affect zinc redistribution to protect intestinal barrier in sepsis. MAIN METHODS: Rodent model of sepsis was induced by cecal ligation and puncture (CLP). Plasma endotoxin was assayed by LAL test and plasma zinc was measured by flame atomic spectrophotometer. Gut mucosal permeability was determined by plasma FITC-dextran. Zinc content and ZIP14 mRNA in gut mucosa were assayed by spectrophotometer and qRT-PCR, respectively. Tight junction integrity of Caco-2 was evaluated by transepithelial electrical resistance (TEER). Tight junction (TJ) protein expression was detected by Western blotting. KEY FINDINGS: Berberine and zinc gluconate pretreatment to CLP rats improved survival rate, reduced plasma endotoxin level, alleviated hypozincemia, increased zinc accumulation and ZIP14 mRNA expression in the intestinal mucosa. Berberine and zinc gluconate pretreatment decreased CLP-elicited intestinal hyperpermeability to FITC-dextran. These effects of berberine in vivo were abolished by AG1024. In vitro, lipopolysaccharide (LPS) repressed zinc transfer into Caco-2 cells exposed to zinc gluconate. Berberine and IGF-I treatment increased ZIP14 protein expression and promoted zinc transfer into Caco-2 cells exposed to zinc gluconate plus LPS. Berberine treatment induced TJ protein (claudin-1 and occludin) and raised TEER in LPS-treated Caco-2 cells. These effects of berberine in vitro were partially inhibited by ZIP14 siRNA. SIGNIFICANCE: The present study reveals that berberine induces ZIP14 expression and affects zinc re- distribution to protect intestinal barrier in sepsis, which is partially linked with the activation of IGF-I signaling.


Subject(s)
Berberine/pharmacology , Cation Transport Proteins/metabolism , Coinfection/prevention & control , Gluconates/pharmacology , Intestinal Mucosa/drug effects , Sepsis/prevention & control , Tyrphostins/pharmacology , Zinc/metabolism , Animals , Caco-2 Cells , Cell Membrane Permeability/drug effects , Coinfection/metabolism , Coinfection/microbiology , Humans , Male , Protective Agents/pharmacology , Rats , Rats, Wistar , Sepsis/metabolism , Sepsis/microbiology , Signal Transduction/drug effects
5.
Cell Physiol Biochem ; 49(4): 1342-1351, 2018.
Article in English | MEDLINE | ID: mdl-30205381

ABSTRACT

BACKGROUND/AIMS: The gut-vascular barrier (GVB) has recently been depicted to dampen the bacterial invasion of the bloodstream. The intestinal mucosa is a tissue rich in small vessels including capillaries. In this study, the protective effect of berberine on GVB in small bowel mucosa was investigated. METHODS: The rat cecal ligation and puncture (CLP) sepsis model was employed to evaluate the effect of berberine on serum endotoxin level and intestinal vascular permeability to Evans blue in vivo. The rat intestinal microvascular endothelial cells (RIMECs) treated by lipopolysaccharide (LPS) were used to assess the effect of berberine on endothelial permeability to FITC-labeled dextran, transendothelial electrical resistance (TEER), and tight junction (TJ) and adherens junction (AJ) expression in vitro. RESULTS: After 24-hr CLP operation the serum endotoxin concentration and gut vascular permeability were significantly increased, while berberine markedly reduced endotoxin level and vascular leakage. In vitro, LPS not only dramatically increased endothelial permeability of RIMECs to FITC-dextran, but also decreased TEER and inhibited claudin-12, beta-catenin and VE-cadherin expression. These effects of LPS were antagonized by berberine. In addition, our in vivo and vitro studies also confirmed that the effect of berberine on GVB could be partially abolished by ICG001. CONCLUSION: Berberine exerted a protective effect on GVB function in sepsis, which was strictly related to the modulation of the Wnt/beta-catenin signaling pathway.


Subject(s)
Berberine/pharmacology , Capillary Permeability/drug effects , Intestinal Mucosa/metabolism , Protective Agents/pharmacology , Wnt Signaling Pathway/drug effects , Animals , Antigens, CD/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cadherins/metabolism , Claudins/metabolism , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endotoxins/blood , Intestinal Mucosa/drug effects , Lipopolysaccharides/pharmacology , Male , Pyrimidinones/pharmacology , Rats , Rats, Long-Evans , Sepsis/drug therapy , Sepsis/mortality , Sepsis/pathology , Survival Rate , Tight Junctions/metabolism , beta Catenin/metabolism
6.
J Photochem Photobiol B ; 183: 147-153, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29705507

ABSTRACT

Inhibiting component of therapy with (-)-epigallocatechin-3-gallate (EGCG) is low bioavailability of fresh tea polyphenols that outcome from insecurity under stomach related circumstances, insufficient transcellular transport. As needs are, fucose- carboxymethyl chitosan (FU-CMC) graft EGCG with gold nanoparticles (GNPs) (FU-CMC-EGCG-GNPs) nanocomposites were prepared and managed peritumorally to assess their anticancer action. The physicochemical properties of as-prepared nanocomposite were evaluated by FTIR spectroscopy, UV-visible absorption spectra, XRD, FESEM-EDX and HRTEM-SAD. Additionally, the viability and cell uptake assays revealed that the as-prepared nanocomposite successfully repressed the propagation of gastric tumor cells. In vivo anticancer treatment of FU-CMC-EGCG-GNPs nanocomposites showed more anticancer action contrasted with pure EGCG. Immuno-histological investigations established a superior numeral of apoptotic tissues in the as-prepared FU-CMC-EGCG-GNPs nanocomposites contrasted with pure EGCG. Overall, the as-prepared FU-CMC-EGCG-GNPs nanocomposite affords a proficient medicine delivery stage for chemotherapy.


Subject(s)
Catechin/analogs & derivatives , Drug Carriers/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Nanocomposites/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Catechin/chemistry , Catechin/therapeutic use , Catechin/toxicity , Cell Line, Tumor , Chitosan/analogs & derivatives , Chitosan/chemistry , Fucose/chemistry , Helicobacter pylori/drug effects , Humans , Kidney/pathology , Liver/pathology , Mice , Microscopy, Electron, Transmission , Nanocomposites/therapeutic use , Nanocomposites/toxicity , Particle Size , Spectroscopy, Fourier Transform Infrared , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transplantation, Heterologous , X-Ray Diffraction
7.
Fitoterapia ; 124: 200-205, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29154866

ABSTRACT

Insulin-like growth factor I (IGF-I) and binding protein 3 (IGFBP-3) play a role in the maintenance of gut mucosal barrier function. Nevertheless, IGF-I/IGFBP-3 and tight junction protein (TJP) expression in small intestinal mucosa are often impaired during endotoxemia. In this model of acute endotoxemia, the regulatory effect of berberine on IGF-I/IGFBP-3 and TJP expression in ileal mucosa was evaluated. The findings revealed systemic injection of lipopolysaccharide (LPS) suppressed mRNA and protein expression of IGF-I and IGFBP-3, but berberine ameliorated their production. LPS injection inhibited occludin and claudin-1 protein generation, and this inhibitory effect of LPS was abolished by berberine. Inhibition of IGF-I/IGFBP-3 signaling by AG1024 or siRNAs reduced berberine-induced occludin and claudin-1 production. Additionally, GW9662 was found to repress berberine-induced IGF-I/IGFBP-3 expression, indicating of a cross-link between PPARγ and IGF-I/IGFBP-3 axis.


Subject(s)
Berberine/pharmacology , Endotoxemia/drug therapy , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Intestinal Mucosa/drug effects , Signal Transduction/drug effects , Anilides , Animals , Ileum/drug effects , Male , Rats , Rats, Wistar , Tyrphostins
8.
Oncol Rep ; 38(5): 2752-2760, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29048674

ABSTRACT

MicroRNAs (miRNAs) are a group of critical players in gastric cancer (GC). Among numerous cancer-related miRNAs, the expression level and functional role of miR-345 in GC has not been investigated. This study showed that miR-345 expression was decreased in GC. Decreased expression level of miR-345 was associated with occurrence of lymph metastasis and advanced TNM stage of GC patients. Patients with low expression level of miR-345 had reduced overall survival (OS) and disease-free survival (DFS). In vitro experiments showed that miR-345 could inhibit the migration and invasion of GC cells. In vivo experiments showed that miR-345 knockdown could promote lung metastasis of GC cells in nude mice. miR-345 was found to prevent the metastasis by inhibiting epithelial-mesenchymal transition (EMT) of GC cells. Furthermore, FOXQ1 was confirmed to be the downstream target of miR-345 in GC cells. Forced expression of FOXQ1 could reverse the inhibitory effects of miR-345 on GC metastasis, while knockdown of FOXQ1 prevented the promoting effects of miR-345 knockdown on GC metastasis. In summary, this study demonstrates miR-345 is a promising biomarker and therapeutic target in GC.


Subject(s)
Epithelial-Mesenchymal Transition , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , MicroRNAs/genetics , Stomach Neoplasms/pathology , Adult , Aged , Animals , Cell Line, Tumor , Cell Movement , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Neoplasm Transplantation , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Survival Analysis
9.
World J Surg Oncol ; 14(1): 252, 2016 Sep 26.
Article in English | MEDLINE | ID: mdl-27669818

ABSTRACT

BACKGROUND: Epithelial dysplasia and adenomatous polyps of colorectum are precancerous lesions. Surgical removal is still one of the important treatment approaches for colorectal polyps. CASE PRESENTATION: A male patient over 78 years was admitted due to bloody stool and abdominal pain. Colonoscopic biopsy showed a high-grade epithelial dysplasia in an adenomatous polyp of sigmoid colon. Anemia, COPD, ischemic heart disease (IHD), arrhythmias, and hypoproteinemia were comorbidities. The preoperative preparation was carefully made consisting of oral nutritional supplements (ONS), blood transfusion, cardiorespiratory management, and hemostatic therapy. However, his illness did not improve but deteriorate mainly due to polyp rebleeding during preparative period. The open polypectomy was performed within 60 min under epidural anesthesia. Postoperative treatments included oxygen inhalation, bronchodilation, parenteral and enteral nutrition, human serum albumin, antibiotics, and blood transfusion. Unluckily, these did not significantly facilitate to surgical recovery on account of severe comorbidities and complications. The most serious complications were colonic leakage and secondary abdominal severe infection. The patient finally gave up treatment due to multiple organ dysfunction syndromes. CONCLUSIONS: The polypectomy for colonic polyp is a seemingly minor but potentially deadly surgery for patients with severe comorbidities, and prophylactic ostomy should be considered for the safety.

10.
PLoS One ; 8(12): e80997, 2013.
Article in English | MEDLINE | ID: mdl-24312512

ABSTRACT

The mechanism by which baicalin modulated the expression of inducible nitric oxide synthase (iNOS) and nitric oxide (NO) in the mucosa of distal ileum was investigated in a rat model of acute endo-toxemia induced by intraperitoneal injection of bacterial lipopolysaccharide (LPS). The experiment demonstrated that LPS upregulated iNOS mRNA and protein expression as well as NO produc-tion (measured as the stable degradation production, nitrites). LPS not only increased toll-like receptor 4 (TLR4) and peroxisome proliferator-activated receptor gamma (PPARγ) content, but also activated p38 and activating transcription factor 2 (ATF2) and inactivated PPARγ via phosphorylation. Inhibition of p38 signalling pathway by chemical inhibitor SB202190 and small interfering RNA (siRNA) ameliorated LPS-induced iNOS generation, while suppression of PPARγ pathway by SR-202 boosted LPS-elicited iNOS expression. Baicalin treatment (I) attenuated LPS-induced iNOS mRNA and protein as well as nitrites generation, and (II) ameliorated LPS-elicited TLR4 and PPARγ production, and (III) inhibited p38/ATF2 phosphorylation leading to suppression of p38 signalling, and (IV) prevented PPARγ from phosphorylation contributing to maintainence of PPARγ bioactivity. However, SR-202 co-treatment (I) partially abrogated the inhibitory effect of baicalin on iNOS mRNA expression, and (II) partially reversed baicalin-inhibited p38 phosphorylation. In summary, baicalin could ameliorate LPS-induced iNOS and NO overproduction in mucosa of rat terminal ileum via inhibition of p38 signalling cascade and activation of PPARγ pathway. There existed a interplay between the two signalling pathways.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Endotoxemia/enzymology , Flavonoids/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Intestinal Mucosa/enzymology , Nitric Oxide Synthase Type II/biosynthesis , Acute Disease , Animals , Endotoxemia/chemically induced , Endotoxemia/drug therapy , Endotoxemia/pathology , Intestinal Mucosa/pathology , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Male , Nitric Oxide/biosynthesis , PPAR gamma/metabolism , Rats , Rats, Sprague-Dawley , Toll-Like Receptor 4/metabolism
11.
Int Immunopharmacol ; 12(1): 182-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22155099

ABSTRACT

Berberine hydrochloride (BBR), a plant alkaloid, has been used to treat intestinal inflammation or infection for years. Cyclooxygenase-2 (COX-2) is pro-inflammatory mediator and involved in the induction of gut inflammation. The expression of COX-2 in small bowel mucosa was determined and the mechanism by which BBR modulated COX-2 expression was explored in a rat model of endotoxemia induced by lipopolysaccharide (LPS). The results showed that without LPS stimulation COX-2 was constitutively expressed at low levels in control rats. LPS challenge rapidly induced COX-2 gene transcription resulting in high levels of inducible COX-2 expression in endotoxemic rats. BBR pre- and post-treatment had no marked effect on constitutive COX-2 expression but inhibited inducible COX-2 overexpression. LPS challenge increased the expression and phosphorylation of peroxisome proliferator-activated receptor gamma (PPARγ), p38 and activating transcription factor 2 and 3 (ATF2, ATF3), but the effects of LPS were inhibited by BBR treatment. GW9662 did not influence constitutive COX-2 expression but enhanced inducible COX-2 overproduction. Besides, GW9662 abolished the inhibitory effect of BBR on inducible COX-2, p38, ATF2, 3 expression and phosphorylation. Collectively, these results indicated that BBR gavage could attenuate the overexpression of inducible COX-2, not constitutive COX-2, in ileal mucosa during acute endotoxemia in part via activation of PPARγ pathway, which negatively interfered with p38/ATFs cascade.


Subject(s)
Berberine/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Endotoxemia/metabolism , Intestinal Mucosa/drug effects , PPAR gamma/agonists , Anilides/pharmacology , Animals , Cyclooxygenase 2/genetics , Disease Models, Animal , Endotoxemia/chemically induced , Escherichia coli , Ileum/drug effects , Ileum/metabolism , Intestinal Mucosa/metabolism , Lipopolysaccharides , Male , PPAR gamma/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
12.
Respir Physiol Neurobiol ; 178(2): 235-41, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21726671

ABSTRACT

The recent pandemic influenza A (H1N1 2009) virus infection has caused acute lung injury in susceptible population resulting in high mortality in ICU patients. In this report, we observed the effect of pre-B cell colony-enhancing factor (PBEF) on the inflammation and apoptosis in H1N1-infected human pulmonary microvascular endothelial cells (HPMECs). We constructed an in vitro HPMEC monolayer model. The results showed that H1N1 2009 induced the increased expression of inflammatory cytokines (IL-6/IL-8/TNF-α/IP-10) and apoptosis factors (FasL/TRAIL) in infected HPMECs. However, PBEF silencing with siRNA inhibited the expression of some inflammatory cytokines and decreased the apoptosis mediated by FasL. We conclude that PBEF might be partially responsible for the localized inflammatory response to H1N1 2009 in the lung microvascular endothelium and the H1N1-induced endothelial cell apoptosis probably through the FasL-mediated pathway.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Apoptosis/physiology , Inflammation Mediators/antagonists & inhibitors , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/prevention & control , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/physiology , Pandemics , Respiratory Mucosa/enzymology , Acute Lung Injury/enzymology , Acute Lung Injury/pathology , Acute Lung Injury/virology , Animals , Apoptosis Regulatory Proteins/physiology , Cells, Cultured , Child , Cytokines/biosynthesis , Cytokines/physiology , Dogs , Gene Knockdown Techniques/methods , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Influenza A Virus, H1N1 Subtype/genetics , Influenza, Human/enzymology , Influenza, Human/pathology , Microcirculation/genetics , Microcirculation/physiology , Respiratory Mucosa/blood supply , Respiratory Mucosa/pathology
13.
Fitoterapia ; 82(7): 976-82, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21641970

ABSTRACT

The effect of berberine hydrochloride (BBR) on inducible cyclooxygenase-2 (COX-2) in small intestinal mucosa and related mechanisms was investigated in a rat model of acute endotoxemia. The results showed that lipopolysaccharide (LPS) increased COX-2 expression, whereas SB202190 and BBR curtailed it. LPS increased phosphorylation of mucosal p38 MAPK and ATF2 as well as production of ATF2, whereas BBR attenuated these effects. LPS upregulated mucosal peroxisome proliferator-activated receptor gamma (PPARγ), but BBR reduced this receptor. GW9662 aggravated LPS-induced and reversed BBR-attenuated COX-2 expression. The findings showed that BBR ameliorated COX-2 overexpression partially via modulation of p38 and PPARγ pathways during acute endotoxemia.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Berberine/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , Cyclooxygenase 2/metabolism , Endotoxemia/drug therapy , Intestinal Mucosa/drug effects , Phytotherapy , Acute Disease , Anilides , Animals , Coptis/chemistry , Cyclooxygenase 2 Inhibitors/pharmacology , Disease Models, Animal , Endotoxemia/metabolism , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Intestinal Mucosa/metabolism , Intestine, Small/drug effects , Intestine, Small/metabolism , Lipopolysaccharides , Male , PPAR gamma/metabolism , Phosphorylation , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Rhizome , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Fitoterapia ; 82(4): 709-15, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21376787

ABSTRACT

Growing evidence suggests that TLRs/NF-κB signaling pathway plays a critical role in the pathogenesis of Crohn's disease (CD). We have reported that triptolide, an active component from Tripterygium wilfordii Hook, showed therapeutic activity in IL-10-deficeint (IL-10-/- mice, a murine CD model. However the full mechanisms of action of this agent in CD remain largely unknown. We hypothesized that triptolide would ameliorate the experimental colitis by inhibiting TLRs/NF-κB signaling pathway. We found TLR2 and TLR4 were upregulated in IL-10-)/- mice, triptolide inhibited the TLRs/NF-κB signaling pathway in vivo. In addition, triptolide in vitro was able to downregulate the TLRs/NF-κB pathway in cultured colonic explants from CD patients. Our results confirm the therapeutic effect of triptolide in experimental colitis, and suggest it as a promising compound for CD treatment. These findings also support the possibility that targeted inhibition of TLR signaling pathway is an approach deserving further investigation as a therapeutic strategy for CD.


Subject(s)
Crohn Disease/drug therapy , Diterpenes/therapeutic use , Phenanthrenes/therapeutic use , Phytotherapy , Toll-Like Receptors/antagonists & inhibitors , Transcription Factor RelA/antagonists & inhibitors , Animals , Diterpenes/pharmacology , Drug Evaluation, Preclinical , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Humans , Interleukin-10/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/metabolism , Phenanthrenes/pharmacology , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Signal Transduction/drug effects , Tissue Culture Techniques , Toll-Like Receptors/metabolism , Transcription Factor RelA/metabolism , Tripterygium
SELECTION OF CITATIONS
SEARCH DETAIL
...