Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
Can J Physiol Pharmacol ; 95(2): 122-128, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27992235

ABSTRACT

Baicalin has been reported to attenuate lung edema in the process of lung injury. However, the effect of baicalin on alveolar fluid clearance (AFC) and epithelial sodium channel (ENaC) expression has not been tested. Sprague-Dawley rats were anesthetized and intratracheally injected with either 1 mg/kg lipopolysaccharide (LPS) or saline vehicle. Baicalin with various concentrations (10, 50, and 100 mg/kg) was injected intraperitoneally 30 min before administration of LPS. Then lungs were isolated for measurement of AFC, cyclic adenosine monophosphate (cAMP) level, and cellular localization of α-ENaC. Moreover, mouse alveolar type II (ATII) epithelial cell line was incubated with baicalin (30 µmol/L), adenylate cyclase inhibitor SQ22536 (10 µmol/L), or cAMP-dependent protein kinase inhibitor (PKA) KT5720 (0.3 µmol/L) 15 min before LPS (1 µg/mL) incubation. Protein expression of α-ENaC was detected by Western blot. Baicalin increased cAMP concentration and AFC in a dose-dependent manner in rats with LPS-induced acute lung injury. The increase of AFC induced by baicalin was associated with an increase in the abundance of α-ENaC protein. SQ22536 and KT5720 prevented the increase of α-ENaC expression caused by baicalin in vitro. These findings suggest that baicalin prevents LPS-induced reduction of AFC by upregulating α-ENaC protein expression, which is activated by stimulating cAMP/PKA signaling pathway.


Subject(s)
Acute Lung Injury/metabolism , Epithelial Sodium Channels/metabolism , Flavonoids/pharmacology , Lung/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/physiopathology , Adenylyl Cyclase Inhibitors/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carbazoles/pharmacology , Cells, Cultured , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Edema/complications , Edema/drug therapy , Edema/physiopathology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Flavonoids/antagonists & inhibitors , Flavonoids/therapeutic use , Ionomycin/pharmacology , Lipopolysaccharides , Lung/metabolism , Lung/physiopathology , Male , Mice , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Rats , Water/metabolism
2.
Acta Pharmacol Sin ; 37(8): 1091-100, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27345626

ABSTRACT

AIM: Postoperative ileus (POI) is a postoperative dysmotility disorder of gastrointestinal tract, which remains one of the most perplexing problems in medicine. In the present study we investigated the effects of hesperidin, a major flavonoid in sweet oranges and lemons, on POI in rats. METHODS: SD rats were administered hesperidin (5, 20, and 80 mg·kg(-1)·d(-1), ig) for 3 consecutive days. POI operation (gently manipulating the cecum for 1 min) was performed on d 2. The gastrointestinal motility and isolated intestinal contraction were examined 1 d after the operation. Then the myosin phosphorylation and inflammatory responses in cecum tissue were assessed. Smooth muscle cells were isolated from rat small intestine for in vitro experiments. RESULTS: The gastric emptying and intestinal transit were significantly decreased in POI rats, which were reversed by administration of hesperidin. In ileum and cecum preparations of POI rats in vitro, hesperidin (2.5-160 µmol/L) dose-dependently increased the spontaneous contraction amplitudes without affecting the contractile frequency, which was blocked by the myosin light chain kinase (MLCK) inhibitor ML-7 or verapamil, but not by TTX. Furthermore, administration of hesperidin increased the phosphorylation of MLC20 in the cecum tissue of POI rats. Moreover, administration of hesperidin reversed the increased levels of inflammatory cytokines, iNOS and COX-2 in cecum tissue of POI rats. In freshly isolated intestinal smooth muscle cells, hesperidin (5-80 µmol/L) dose-dependently increased the intracellular Ca(2+) concentration as well as the phosphorylation of MLC20, which was abrogated by ML-7 or siRNA that knocked down MLCK. CONCLUSION: Oral administration of hesperidin effectively alleviates rat POI through inhibition of inflammatory responses and stimulation of Ca(2+)-dependent MLC phosphorylation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Hesperidin/pharmacology , Ileus/drug therapy , Inflammation/prevention & control , Myosins/metabolism , Phosphorylation/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Azepines/pharmacology , Calcium/metabolism , Cecum/metabolism , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Dose-Response Relationship, Drug , Gastric Emptying/drug effects , Gastrointestinal Motility/drug effects , Hesperidin/antagonists & inhibitors , Hesperidin/therapeutic use , Intestine, Small/physiology , Male , Muscle Contraction/drug effects , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/antagonists & inhibitors , Naphthalenes/pharmacology , Nitric Oxide Synthase Type II/metabolism , Postoperative Complications/drug therapy , RNA, Small Interfering/pharmacology , Rats , Verapamil/pharmacology
4.
Shock ; 44(4): 371-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26196844

ABSTRACT

Acute lung injury (ALI) is characterized by lung inflammation and diffuse infiltration of neutrophils. Neutrophil apoptosis is recognized as an important control point in the resolution of inflammation. Maresin 1 (MaR1) is a new docosahexaenoic acid-derived proresolving agent that promotes the resolution of inflammation. However, its function in neutrophil apoptosis is unknown. In this study, isolated human neutrophils were incubated with MaR1, the pan-caspase inhibitor z-VAD-fmk, and lipopolysaccharide (LPS) to determine the mechanism of neutrophil apoptosis. Acute lung injury was induced by intratracheal instillation of LPS. In addition, mice were treated with MaR1 intravenously at the peak of inflammation and administered z-VAD-fmk intraperitoneally. We found that culture of isolated human neutrophils with LPS dramatically delayed neutrophil apoptosis through the phosphorylation of AKT, ERK, and p38 to upregulate the expression of the antiapoptotic proteins Mcl-1 and Bcl-2, which was blocked by pretreatment with MaR1 in vitro. In mice, MaR1 accelerated the resolution of inflammation in LPS-induced ALI through attenuation of neutrophil accumulation, pathohistological changes, and pulmonary edema. Maresin 1 promoted resolution of inflammation by accelerating caspase-dependent neutrophil apoptosis. Moreover, MaR1 also reduced the LPS-induced production of proinflammatory cytokines and upregulated the production of the anti-inflammatory cytokine interleukin-10. In contrast, treatment with z-VAD-fmk inhibited the proapoptotic action of MaR1 and attenuated the protective effects of MaR1 in LPS-induced ALI. Taken together, MaR1 promotes the resolution of LPS-induced ALI by overcoming LPS-mediated suppression of neutrophil apoptosis.


Subject(s)
Acute Lung Injury/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Docosahexaenoic Acids/therapeutic use , Neutrophils/drug effects , Acute Lung Injury/pathology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Bronchoalveolar Lavage Fluid/cytology , Caspase Inhibitors/pharmacology , Cell Survival/drug effects , Cells, Cultured , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/antagonists & inhibitors , Docosahexaenoic Acids/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Humans , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/antagonists & inhibitors , Male , Mice, Inbred BALB C , Neutrophils/pathology , Signal Transduction/drug effects
5.
Acta Neuropsychiatr ; 27(1): 48-52, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25467825

ABSTRACT

BACKGROUND: Dipyrone is one of the most commonly used non-opioid analgesic and antipyretic drug. Its anti-nociceptive and hypothermic effects have long been suspected to be centrally mediated. The involvement of the most recently discovered opioid peptide, nociceptin/orphanin FQ (N/OFQ), and its receptor (NOP) in pain transmission is controversial. It appears to be pro-nociceptive when administered supra-spinally, but exerts anti-nociceptive effects when injected spinally or systemically. OBJECTIVE: Investigation of the role of the N/OFQ system in paracetamol-induced anti-nociception and hypothermia led us to determine its role in the anti-nociceptive and hypothermic effects of dipyrone. Material and Methods Hot-plate and tail-flick tests were used to assess nociception, and a rectal thermometer was used to measure rectal temperature in mice. RESULTS: Mice injected with dipyrone (150, 300, 600 mg/kg, i.p.) displayed dose-related anti-nociception and hypothermia. The NOP receptor antagonist JTC-801 (3 mg/kg, i.p.), at a dose that exerted no effect when used alone, alleviated dipyrone-induced anti-nociception but did not reverse dipyrone-induced hypothermia. CONCLUSION: We conclude that NOP receptors participate in the anti-nociceptive, but not in the hypothermic, effects of dipyrone.


Subject(s)
Dipyrone/pharmacology , Hypothermia/chemically induced , Nociception/drug effects , Nociception/physiology , Receptors, Opioid/physiology , Aminoquinolines/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzamides/pharmacology , Dipyrone/antagonists & inhibitors , Dose-Response Relationship, Drug , Hypothermia/physiopathology , Male , Mice , Pain Measurement/drug effects , Receptors, Opioid/drug effects , Nociceptin Receptor
6.
Toxicol Mech Methods ; 24(8): 560-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25137224

ABSTRACT

Escin, a natural mixture of triterpenoid saponin isolated from the seed of the horse chestnut, is reported to have a potent antiulcer activity against ethanol-induced gastric mucosal lesions. This study investigated the possible mechanisms underlying the gastroprotective effect of escin against indomethacin-induced gastric ulcer in mice. Gastric ulceration was induced by a single intragastric administration of indomethacin (18 mg/kg). The mice underwent intragastric treatment with escin at doses of 0.45, 0.9 or 1.8 mg/kg. Gastric lesion was estimated morphometrically and histopathologically 6 h after the indomethacin administration. The antioxidative parameters in gastric mucosa were measured. Moreover, the activity of myeloperoxidase and the contents of TNF-α, P-selectin and VCAM-1 in gastric tissues were determined. The results showed that escin protected gastric tissues against indomethacin-induced gastropathy as demonstrated from a reduction in the ulcer index and an attenuation of histopathologic changes. Escin caused significant reductions of the contents of malondialdehyde, TNF-α, P-selectin, VCAM-1 and myeloperoxidase activity. The altered activities of superoxide dismutase, catalase and glutathione peroxidase in the stomach tissues were also ameliorated by escin treatment. The present study demonstrated that escin had a protective effect against indomethacin-induced gastric ulcer in mice, not only by virtue of its antioxidant potential, but also due to its anti-inflammatory effect.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Ulcer Agents/therapeutic use , Escin/therapeutic use , Gastric Mucosa/drug effects , Indomethacin/antagonists & inhibitors , Protective Agents/therapeutic use , Stomach Ulcer/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anti-Ulcer Agents/administration & dosage , Antioxidants/administration & dosage , Antioxidants/therapeutic use , Dose-Response Relationship, Drug , Escin/administration & dosage , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , H(+)-K(+)-Exchanging ATPase/chemistry , H(+)-K(+)-Exchanging ATPase/metabolism , Indomethacin/adverse effects , Lipid Peroxidation/drug effects , Mice , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/metabolism , P-Selectin/antagonists & inhibitors , P-Selectin/chemistry , P-Selectin/metabolism , Protective Agents/administration & dosage , Random Allocation , Severity of Illness Index , Stomach/drug effects , Stomach/immunology , Stomach/pathology , Stomach Ulcer/chemically induced , Stomach Ulcer/metabolism , Stomach Ulcer/pathology , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/chemistry , Vascular Cell Adhesion Molecule-1/metabolism
7.
J Neuroimmune Pharmacol ; 9(4): 582-90, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25035139

ABSTRACT

Recent evidence has suggested that microglial activation plays an important role in the pathogenesis of depression. Activated microglia can secrete various pro-inflammatory cytokines, which may contribute to the development and maintenance of depression. Thus, inhibition of microglial activation may have a therapeutic benefit in the treatment of depression. In the present study, we found that fluoxetine significantly inhibited lipopolysaccharide (LPS)-induced production of tumor necrosis factor-alpha (TNF-α), interleukin- 6 (IL-6) and nitric oxide (NO) and reduced the phosphorylation of transforming growth factor-beta-activated kinase 1 (TAK1) and nuclear factor-kappa B (NF-κB) p65 nuclear translocation in microglia. We further found that fluoxetine increased the expression of ß-arrestin 2 and enhanced the association of ß-arrestin 2 with TAK1-binding protein 1 (TAB1) and disrupted TAK1-TAB1 interaction. Moreover, ß-arrestin 2 knock-down abolished the anti-inflammatory effects of fluoxetine in lipopolysaccharide-stimulated microglial cells. Collectively, our findings suggest that ß-arrestin 2 is necessary for the anti-inflammatory effects of fluoxetine and offers novel drug targets in the convergent fluoxetine/ß-arrestin 2 and inflammatory pathways for treating microglial inflammatory neuropathologies like depression.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arrestins/metabolism , Fluoxetine/pharmacology , Lipopolysaccharides/pharmacology , Microglia/drug effects , Microglia/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Arrestins/genetics , Cells, Cultured , Fluoxetine/antagonists & inhibitors , Gene Expression/drug effects , Gene Knockdown Techniques , Interleukin-6/metabolism , Lipopolysaccharides/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Male , Mice , NF-kappa B/metabolism , Nitric Oxide/metabolism , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism , beta-Arrestin 2 , beta-Arrestins
8.
J Nutr ; 144(7): 1002-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24790027

ABSTRACT

Dietary fiber intake is associated with lower incidence and mortality from disease, but the underlying mechanisms of these protective effects are unclear. We hypothesized that ß2→1-fructan dietary fibers confer protection on intestinal epithelial cell barrier function via Toll-like receptor 2 (TLR2), and we studied whether ß2→1-fructan chain-length differences affect this process. T84 human intestinal epithelial cell monolayers were incubated with 4 ß2→1-fructan formulations of different chain-length compositions and were stimulated with the proinflammatory phorbol 12-myristate 13-acetate (PMA). Transepithelial electrical resistance (TEER) was analyzed by electric cell substrate impedance sensing (ECIS) as a measure for tight junction-mediated barrier function. To confirm TLR2 involvement in barrier modulation by ß2→1-fructans, ECIS experiments were repeated using TLR2 blocking antibody. After preincubation of T84 cells with short-chain ß2→1-fructans, the decrease in TEER as induced by PMA (62.3 ± 5.2%, P < 0.001) was strongly attenuated (15.2 ± 8.8%, P < 0.01). However, when PMA was applied first, no effect on recovery was observed during addition of the fructans. By blocking TLR2 on the T84 cells, the protective effect of short-chain ß2→1-fructans was substantially inhibited. Stimulation of human embryonic kidney human TLR2 reporter cells with ß2→1-fructans induced activation of nuclear factor kappa-light-chain-enhancer of activated B cells, confirming that ß2→1-fructans are specific ligands for TLR2. To conclude, ß2→1-fructans exert time-dependent and chain length-dependent protective effects on the T84 intestinal epithelial cell barrier mediated via TLR2. These results suggest that TLR2 located on intestinal epithelial cells could be a target of ß2→1-fructan-mediated health effects.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Colon/metabolism , Fructans/metabolism , Intestinal Mucosa/metabolism , Protective Agents/metabolism , Tight Junctions/metabolism , Toll-Like Receptor 2/agonists , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Antibodies, Blocking/pharmacology , Cell Line , Colon/drug effects , Colon/immunology , Diglycerides/pharmacology , Fructans/antagonists & inhibitors , Fructans/chemistry , Gastrointestinal Agents/antagonists & inhibitors , Gastrointestinal Agents/chemistry , Gastrointestinal Agents/metabolism , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Kidney/drug effects , Kidney/immunology , Kidney/metabolism , Ligands , Membrane Transport Modulators/antagonists & inhibitors , Membrane Transport Modulators/toxicity , Molecular Structure , NF-kappa B/agonists , NF-kappa B/metabolism , Oligopeptides/pharmacology , Prebiotics/analysis , Protective Agents/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/antagonists & inhibitors , Tetradecanoylphorbol Acetate/toxicity , Tight Junctions/drug effects , Tight Junctions/immunology , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Transcription Factor AP-1/agonists , Transcription Factor AP-1/metabolism
9.
Br J Nutr ; 111(7): 1221-30, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24252501

ABSTRACT

Several reports have indicated that dietary intake of DHA is associated with lower prevalence of periodontitis. In the present study, we investigated the effect of DHA on the production of proinflammatory mediators in murine macrophage-like RAW264.7 cells stimulated with lipopolysaccharide (LPS) isolated from Prevotella intermedia, a pathogen implicated in inflammatory periodontal disease, and its mechanisms of action. LPS was isolated from lyophilised P. intermedia ATCC 25,611 cells using the standard hot-phenol-water protocol. Culture supernatants were collected and assayed for NO, IL-1ß and IL-6. Real-time PCR analysis was carried out to detect the expression of inducible NO synthase (iNOS), IL-1ß, IL-6 and haeme oxygenase-1 (HO-1) mRNA. Immunoblot analysis was carried out to quantify the expression of iNOS and HO-1 protein and concentrations of signalling proteins. DNA-binding activities of NF-κB subunits were determined using an ELISA-based assay kit. DHA significantly attenuated the production of NO, IL-1ß and IL-6 at both gene transcription and translation levels in P. intermedia LPS-activated RAW264.7 cells. DHA induced the expression of HO-1 in cells treated with P. intermedia LPS. Selective inhibition of HO-1 activity by tin protoporphyrin IX significantly mitigated the inhibitory effects of DHA on LPS-induced NO production. DHA significantly attenuated the phosphorylation of c-Jun N-terminal kinase induced by LPS. In addition, DHA suppressed the transcriptional activity of NF-κB by regulating the nuclear translocation and DNA-binding activity of NF-κB p50 subunit and inhibited the phosphorylation of signal transducer and activator of transcription 1. Further in vivo studies are needed to better evaluate the potential of DHA in humans as a therapeutic agent to treat periodontal disease.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Dietary Fats/metabolism , Docosahexaenoic Acids/metabolism , Inflammation Mediators/antagonists & inhibitors , Lipopolysaccharides/antagonists & inhibitors , Macrophages/drug effects , Prevotella intermedia/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Cell Line, Transformed , Dietary Fats/antagonists & inhibitors , Docosahexaenoic Acids/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Inflammation Mediators/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/isolation & purification , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Macrophage Activation/drug effects , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/agonists , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Metalloporphyrins/pharmacology , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Periodontitis/immunology , Periodontitis/metabolism , Periodontitis/microbiology , Periodontitis/prevention & control , Protoporphyrins/pharmacology
10.
Eur J Nutr ; 53(3): 853-64, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24077694

ABSTRACT

PURPOSE: Urolithins, gut microbiota metabolites derived from ellagic acid and ellagitannins, reach micromolar concentrations in the colon lumen where can have anti-inflammatory and anticancer effects. The antiproliferative activity of urolithins (Uro-A, Uro-B, Uro-C and Uro-D) and their most relevant in vivo glucuronides were evaluated in three human colon cancer cell lines (Caco-2, SW480 and HT-29). METHODS: Cell proliferation was evaluated by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide and Trypan blue exclusion assays. Cell cycle was evaluated by flow cytometry and urolithins metabolism by HPLC­MS/MS. RESULTS: Urolithins inhibited cell proliferation and cell cycle progression in a time- and dose-dependent manner and arrested the cells at S and G2/M phases, depending on the urolithin. Uro-A exerted the highest antiproliferative activity, followed by Uro-C, Uro-D and Uro-B. Unlike Caco-2 and SW480 cells, HT-29 cells partially overcame the effects after 48 h, which was related to the complete glucuronidation of urolithins. Uro-A or Uro-B glucuronides did not affect cell cycle and showed lower antiproliferative activity than their aglycone counterparts. Uro-A or Uro-B plus inhibitors of drug efflux ABC transporters partially prevented the glucuronidation of urolithins in HT-29 cells which became more sensitive. CONCLUSIONS: Uro-A, Uro-B, Uro-C and Uro-D exerted different antiproliferative effects depending on the colon cancer cell line. We also report here, for the first time, the role of ABC transporters and Phase-II metabolism in HT-29 cells as a mechanism of cancer resistance against urolithins due to their conversion to glucuronide conjugates that exerted lower antiproliferative activity.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Colonic Neoplasms/drug therapy , Coumarins/pharmacology , Hydrolyzable Tannins/pharmacology , Metabolic Detoxication, Phase II , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colon/drug effects , Colon/metabolism , Colonic Neoplasms/metabolism , Coumarins/antagonists & inhibitors , Coumarins/chemistry , Coumarins/metabolism , Drug Resistance, Neoplasm/drug effects , G2 Phase/drug effects , Glucuronides/chemistry , Glucuronides/metabolism , Humans , Hydrolyzable Tannins/antagonists & inhibitors , Hydrolyzable Tannins/chemistry , Hydrolyzable Tannins/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Kinetics , Membrane Transport Modulators/pharmacology , S Phase/drug effects
11.
Br J Pharmacol ; 171(3): 735-45, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24172143

ABSTRACT

BACKGROUND AND PURPOSE: The COX-2/PGE2 pathway in hypoxic cancer cells has important implications for stimulation of inflammation and tumourigenesis. However, the mechanism by which glucocorticoid receptors (GRs) inhibit COX-2 during hypoxia has not been elucidated. Hence, we explored the mechanisms underlying glucocorticoid-mediated inhibition of hypoxia-induced COX-2 in human distal lung epithelial A549 cells. EXPERIMENTAL APPROACH: The expressions of COX-2 and glucocorticoid-induced leucine zipper (GILZ) in A549 cells were determined by Western blot and/or quantitative real time-PCR respectively. The anti-invasive effect of GILZ on A549 cells was evaluated using the matrigel invasion assay. KEY RESULTS: The hypoxia-induced increase in COX-2 protein and mRNA levels and promoter activity were suppressed by dexamethasone, and this effect of dexamethasone was antagonized by the GR antagonist RU486. Overexpression of GILZ in A549 cells also inhibited hypoxia-induced COX-2 expression levels and knockdown of GILZ reduced the glucocorticoid-mediated inhibition of hypoxia-induced COX-2 expression, indicating that the inhibitory effects of dexamethasone on hypoxia-induced COX-2 are mediated by GILZ. GILZ suppressed the expression of hypoxia inducible factor (HIF)-1α at the protein level and affected its signalling pathway. Hypoxia-induced cell invasion was also dramatically reduced by GILZ expression. CONCLUSION AND IMPLICATIONS: Dexamethasone-induced upregulation of GILZ not only inhibits the hypoxic-evoked induction of COX-2 expression and cell invasion but further blocks the HIF-1 pathway by destabilizing HIF-1α expression. Taken together, these findings suggest that the suppression of hypoxia-induced COX-2 by glucocorticoids is mediated by GILZ. Hence, GILZ is a potential key therapeutic target for suppression of inflammation under hypoxia.


Subject(s)
Alveolar Epithelial Cells/drug effects , Cyclooxygenase 2/metabolism , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Receptors, Glucocorticoid/agonists , Transcription Factors/agonists , Alveolar Epithelial Cells/immunology , Alveolar Epithelial Cells/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Hypoxia , Cell Line , Cell Movement/drug effects , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/genetics , Dexamethasone/antagonists & inhibitors , Dexamethasone/pharmacology , Genes, Reporter/drug effects , Glucocorticoids/antagonists & inhibitors , Hormone Antagonists/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , RNA Interference , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/metabolism , Signal Transduction/drug effects , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism
12.
J Physiol Pharmacol ; 64(3): 269-80, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23959723

ABSTRACT

Diclofenac (DIC), a nonsteroidal anti-inflammatory drug, is known to exert anti-nociceptive and anti-convulsant actions; however, its effects on ion currents, in neurons remain debatable. We aimed to investigate (1) potential effects of diclofenac on membrane potential and potassium currents in differentiated NSC-34 neuronal cells and dorsal root ganglion (DRG) neurons with whole-cell patch-clamp technology, and (2) firing of action potentials (APs), using a simulation model from hippocampal CA1 pyramidal neurons based on diclofenac's effects on potassium currents. In the NSC-34 cells, diclofenac exerted an inhibitory effect on delayed-rectifier K⁺ current (I(KDR)) with an IC50 value of 73 µM. Diclofenac not merely inhibited the I(KDR) amplitude in response to membrane depolarization, but also accelerated the process of current inactivation. The inhibition by diclofenac of IK(DR) was not reversed by subsequent application of either naloxone. Importantly, diclofenac (300 µM) increased the amplitude of M-type K⁺ current (I)(KM)), while flupirtine (10 µM) or meclofenamic acid (10 µM) enhanced it effectively. Consistently, diclofenac (100 µM) increased the amplitude of I(KM) and diminished the I(KDR) amplitude, with a shortening of inactivation time constant in DRG neurons. Furthermore, by using the simulation modeling, we demonstrated the potential electrophysiological mechanisms underlying changes in AP firing caused by diclofenac. During the exposure to diclofenac, the actions on both I(KM) and I(KDR) could be potential mechanism through which it influences the excitability of fast-spiking neurons. Caution needs to be made in attributing the effects of diclofenac primarily to those produced by the activation of I(KM).


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Diclofenac/pharmacology , Ganglia, Spinal/drug effects , KCNQ Potassium Channels/antagonists & inhibitors , Motor Neurons/drug effects , Potassium Channel Blockers/pharmacology , Shaw Potassium Channels/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anticonvulsants/antagonists & inhibitors , Anticonvulsants/pharmacology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , Cell Differentiation , Cell Line , Cells, Cultured , Diclofenac/antagonists & inhibitors , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , KCNQ Potassium Channels/agonists , KCNQ Potassium Channels/genetics , KCNQ Potassium Channels/metabolism , Kinetics , Membrane Potentials/drug effects , Membrane Transport Modulators/pharmacology , Mice , Models, Biological , Motor Neurons/cytology , Motor Neurons/metabolism , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Potassium Channel Blockers/antagonists & inhibitors , Protein Isoforms/agonists , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Rats , Shaw Potassium Channels/agonists , Shaw Potassium Channels/genetics , Shaw Potassium Channels/metabolism
13.
Biochem J ; 454(2): 283-93, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23782265

ABSTRACT

The atherogenic cytokine IL-6 (interleukin-6) induces pro-inflammatory gene expression in VECs (vascular endothelial cells) by activating the JAK (Janus kinase)/STAT3 (signal transducer and activator of transcription 3) signalling pathway, which is normally down-regulated by the STAT3-dependent induction of the E3 ubiquitin ligase component SOCS3 (suppressor of cytokine signalling 3). Novel treatments based on the regulation of SOCS3 protein levels could therefore have value in the treatment of diseases with an inflammatory component, such as atherosclerosis. To this end we carried out a screen of 1031 existing medicinal compounds to identify inducers of SOCS3 gene expression and identified the flavanoids naringenin and flavone as effective inducers of SOCS3 protein, mRNA and promoter activity. This was in contrast with the action of traditional JAK/STAT3 inhibitors and the polyphenol resveratrol, which effectively suppress SOCS3 gene expression. Both naringenin and flavone also effectively suppressed IL-6-stimulated phosphorylation of STAT3 (Tyr7°5) which led to suppression of IL-6-induction of the atherogenic STAT3 target gene MCP1 (monocyte chemotactic protein-1), suggesting that their ability to induce SOCS3 gene expression is STAT3-independent. Supporting this idea was the observation that the general kinase inhibitor compound C inhibits flavone- and cAMP-dependent, but not JAK-dependent, SOCS3 induction in VECs. Indeed, the ability of flavanoids to induce SOCS3 expression requires activation of the ERK (extracellular-signal-regulated kinase)-dependent transcription factor SP3, and not STAT3. In the present paper we therefore describe novel molecular actions of flavanoids, which control SOCS3 gene induction and suppression of STAT3 signalling in VECs. These mechanisms could potentially be exploited to develop novel anti-atherogenic therapies.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cytokine Receptor gp130/antagonists & inhibitors , Endothelium, Vascular/metabolism , Flavonoids/metabolism , Interleukin-6/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Suppressor of Cytokine Signaling Proteins/agonists , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Cell Line , Cells, Cultured , Chlorocebus aethiops , Cytokine Receptor gp130/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Flavonoids/antagonists & inhibitors , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-6/metabolism , Interleukin-6 Receptor alpha Subunit/metabolism , Mice , Mutant Proteins/agonists , Mutant Proteins/metabolism , Promoter Regions, Genetic/drug effects , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/agonists , Recombinant Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
14.
Diabetes Obes Metab ; 15(11): 1029-39, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23731386

ABSTRACT

AIM: Visceral adiposity and impaired glucose metabolism are common patho-physiological features in patients co-morbid with obesity and type-2 diabetes. We investigated the effects of the heme-oxygenase (HO) inducer hemin and the HO blocker stannous-mesoporphyrin (SnMP) on glucose metabolism, adipocyte hypertrophy and pro-inflammatory cytokines/mediators in Zucker diabetic fatty (ZDF) rats, a model characterized by obesity and type-2 diabetes. METHODS: Histological, morphological/morphometrical, Western immunoblotting, enzyme immunoassay, ELISA and spectrophotometric analysis were used. RESULTS: Treatment with hemin enhanced HO-1, HO activity and cGMP, but suppressed retroperitoneal adiposity and abated the elevated levels of macrophage-chemoattractant protein-1 (MCP-1), ICAM-1, tumour necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), IL-1ß, NF-κB, c-Jun-NH2-terminal-kinase (JNK) and activating-protein (AP-1), with parallel reduction of adipocyte hypertrophy. Correspondingly, important proteins of lipid metabolism and insulin-signalling such as lipoprotein lipase (LPL), insulin-receptor substrate-1 (IRS-1), GLUT4, PKB/Akt, adiponectin, the insulin-sensitizing and anti-inflammatory protein and adenosine-monophosphate-activated protein kinase (AMPK) were significantly enhanced in hemin-treated ZDF rats. CONCLUSION: Elevated retroperitoneal adiposity and the high levels of MCP-1, ICAM-1, TNF-α, IL-6, IL-1ß, NF-κB, JNK and AP-1 in untreated ZDF are patho-physiological factors that exacerbate inflammatory insults, aggravate adipocyte hypertrophy, with corresponding reduction of adiponectin and deregulation of insulin-signalling and lipid metabolism. Therefore, the suppression of MCP-1, ICAM-1, TNF-α, IL-6, IL-1ß, NF-κB, JNK, AP-1 and adipocyte hypertrophy, with the associated enhancement of LPL, adiponectin, AMPK, IRS-1, GLUT4, PKB/Akt and cGMP in hemin-treated ZDF are among the multifaceted mechanisms by which the HO system combats inflammation to potentiate insulin signalling and improve glucose and lipid metabolism. Thus, HO inducers may be explored in the search of novel remedies against the co-morbidities of obesity, dysfunctional lipid metabolism and impaired glucose metabolism.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Hemin/therapeutic use , Hypoglycemic Agents/therapeutic use , Insulin Resistance , Intra-Abdominal Fat/drug effects , Obesity/complications , Adiponectin/metabolism , Adiposity/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Enzyme Induction/drug effects , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/metabolism , Hemin/antagonists & inhibitors , Hyperglycemia/prevention & control , Hypertrophy , Hypoglycemic Agents/antagonists & inhibitors , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Lipid Metabolism/drug effects , Male , Metalloporphyrins/adverse effects , Random Allocation , Rats , Rats, Zucker , Signal Transduction/drug effects
15.
J Agric Food Chem ; 61(22): 5291-7, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23668856

ABSTRACT

The aim of this study was to determine the protective effect of quercetin, epigallocatechingallate, resveratrol, and rutin against the disruption of epithelial integrity induced by indomethacin in Caco-2 cell monolayers. Indomethacin decreased the transepithelial electrical resistance and increased the permeability of the monolayers to fluorescein-dextran. These alterations were abolished by all the tested polyphenols but rutin, with quercetin being the most efficient. The protective effect of quercetin was associated with its capacity to inhibit the redistribution of ZO-1 protein induced in the tight junction by indomethacin or rotenone, a mitochondrial complex-I inhibitor, and to prevent the decrease of ZO-1 and occludin expression induced by indomethacin. The fact that the antioxidant polyphenols assayed in this study differ in their protective capacity against the epithelial damage induced by indomethacin suggests that this damage is due to the ability of this agent to induce not only oxidative stress but also mitochondrial dysfunction.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Antioxidants/metabolism , Enterocytes/metabolism , Functional Food/analysis , Indomethacin/antagonists & inhibitors , Occludin/metabolism , Zonula Occludens-1 Protein/metabolism , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Antioxidants/analysis , Caco-2 Cells , Catechin/analogs & derivatives , Catechin/analysis , Catechin/metabolism , Enterocytes/drug effects , Gene Expression Regulation/drug effects , Humans , Indomethacin/adverse effects , Occludin/agonists , Occludin/antagonists & inhibitors , Occludin/genetics , Permeability/drug effects , Polyphenols/analysis , Polyphenols/metabolism , Protein Transport/drug effects , Quercetin/analysis , Quercetin/metabolism , Resveratrol , Stilbenes/analysis , Stilbenes/metabolism , Tight Junctions/drug effects , Zonula Occludens-1 Protein/agonists , Zonula Occludens-1 Protein/antagonists & inhibitors , Zonula Occludens-1 Protein/genetics
16.
J Immunol ; 191(1): 337-44, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23720811

ABSTRACT

The bacterial molecule N-3-oxo-dodecanoyl-l-homoserine lactone (C12) has critical roles in both interbacterial communication and interkingdom signaling. The ability of C12 to downregulate production of the key proinflammatory cytokine TNF-α in stimulated macrophages was suggested to contribute to the establishment of chronic infections by opportunistic Gram-negative bacteria, such as Pseudomonas aeruginosa. We show that, in contrast to TNF-α suppression, C12 amplifies production of the major anti-inflammatory cytokine IL-10 in LPS-stimulated murine RAW264.7 macrophages, as well as peritoneal macrophages. Furthermore, C12 increased IL-10 mRNA levels and IL-10 promoter reporter activity in LPS-stimulated RAW264.7 macrophages, indicating that C12 modulates IL-10 expression at the transcriptional level. Finally, C12 substantially potentiated LPS-stimulated NF-κB DNA-binding levels and prolonged p38 MAPK phosphorylation in RAW264.7 macrophages, suggesting that increased transcriptional activity of NF-κB and/or p38-activated transcription factors serves to upregulate IL-10 production in macrophages exposed to both LPS and C12. These findings reveal another part of the complex array of host transitions through which opportunistic bacteria downregulate immune responses to flourish and establish a chronic infection.


Subject(s)
4-Butyrolactone/analogs & derivatives , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cytokines/metabolism , Inflammation Mediators/physiology , Macrophage Activation/immunology , Pseudomonas aeruginosa/immunology , Quorum Sensing/immunology , Signal Transduction/immunology , 4-Butyrolactone/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Cell Line , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Luminescent Proteins/antagonists & inhibitors , Luminescent Proteins/biosynthesis , Luminescent Proteins/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Pseudomonas aeruginosa/pathogenicity , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
17.
J Clin Endocrinol Metab ; 98(7): E1163-72, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23633194

ABSTRACT

CONTEXT: Inhibition of dipeptidyl peptidase-4 (DPP-4) is a potent strategy to increase glucose-dependent insulinotropic polypeptide and glucagon like peptide 1 (GLP-1) induced insulin secretion in diabetes. It is important to know whether new drugs approved for the treatment of type 2 diabetes have direct effects on the ß-cell. OBJECTIVE: Herein we investigated the effect of linagliptin, a novel DPP-4 inhibitor, on ß-cell function and survival. DESIGN: Human islets were exposed to a diabetic milieu (11.1-33.3 mM glucose, 0.5 mM palmitate, the mixture of 2 ng/mL IL-1ß+1000 U/mL interferon-γ, or 50 µM H2O2) with or without 500 ng/mL IL-1 receptor antagonist (IL-1Ra) or 30-50 nM linagliptin. RESULTS: Linagliptin restored ß-cell function and turnover, which was impaired when islets were exposed to elevated glucose, palmitate, cytokines, or H2O2. Pretreatment with IL-1Ra was similarly effective, except against H2O2 treatment. Nitrotyrosine concentrations in islet lysates, an indicator of oxidative stress, were highly elevated under diabetic conditions but not in islets treated with linagliptin or IL-1Ra. Linagliptin also reduced cytokine secretion and stabilized GLP-1 in islet supernatants. CONCLUSIONS: We show that the novel DPP-4 inhibitor linagliptin protected from gluco-, lipo-, and cytokine-toxicity and stabilized active GLP-1 secreted from human islets. This provides a direct GLP-1 mediated protective effect of linagliptin on ß-cell function and survival.


Subject(s)
Antioxidants/pharmacology , Cell Survival/drug effects , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Glucagon-Like Peptide 1/metabolism , Insulin-Secreting Cells/drug effects , Purines/pharmacology , Quinazolines/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/chemistry , Cytokines/metabolism , Dipeptidyl Peptidase 4/chemistry , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Drug Inverse Agonism , Glucagon-Like Peptide 1/agonists , Glucagon-Like Peptide 1/chemistry , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Linagliptin , Palmitic Acid/metabolism , Peptide Fragments/pharmacology , Protein Stability/drug effects , Purines/antagonists & inhibitors , Pyrazines/pharmacology , Quinazolines/antagonists & inhibitors , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Sitagliptin Phosphate , Tissue Culture Techniques , Triazoles/pharmacology
18.
Neuropharmacology ; 71: 1-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23541720

ABSTRACT

Nitric oxide modulates pain development. However, there is no evidence on the effect of nitroxyl (HNO/NO⁻) in nociception. Therefore, we addressed whether nitroxyl inhibits inflammatory hyperalgesia and its mechanism using the nitroxyl donor Angeli's salt (AS; Na2N2O3). Mechanical hyperalgesia was evaluated using a modified Randall and Selitto method in rats, cytokine production by ELISA and nitroxyl was determined by confocal microscopy in DAF (a cell permeable reagent that is converted into a fluorescent molecule by nitrogen oxides)-treated dorsal root ganglia neurons in culture. Local pre-treatment with AS (17-450 µg/paw, 30 min) inhibited the carrageenin-induced mechanical hyperalgesia in a dose- and time-dependent manner with maximum inhibition of 97%. AS also inhibited carrageenin-induced cytokine production. AS inhibited the hyperalgesia induced by other inflammatory stimuli including lipopolysaccharide, tumor necrosis factor-α, interleukin-1ß and prostaglandin E2. Furthermore, the analgesic effect of AS was prevented by treatment with ODQ (a soluble guanylate cyclase inhibitor), KT5823 (a protein kinase G [PKG] inhibitor) or glybenclamide (an ATP-sensitive K⁺ channel blocker), but not with naloxone (an opioid receptor antagonist). AS induced concentration-dependent increase in fluorescence intensity of DAF-treated neurons in a l-cysteine (nitroxyl scavenger) sensitive manner. l-cysteine did not affect the NO⁺ donor S-Nitroso-N-acetyl-DL- penicillamine (SNAP)-induced anti-hyperalgesia or fluorescence of DAF-treated neurons. This is the first study to demonstrate that nitroxyl inhibits inflammatory hyperalgesia by reducing cytokine production and activating the cGMP/PKG/ATP-sensitive K⁺ channel signaling pathway in vivo.


Subject(s)
Analgesics, Non-Narcotic/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Disease Models, Animal , Hyperalgesia/prevention & control , Neurons/drug effects , Nitrites/therapeutic use , Nitrogen Oxides/agonists , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/antagonists & inhibitors , Analgesics, Non-Narcotic/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cells, Cultured , Cytokines/metabolism , Dose-Response Relationship, Drug , Free Radical Scavengers/pharmacology , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/immunology , Ganglia, Spinal/metabolism , Hyperalgesia/immunology , Hyperalgesia/metabolism , Male , Neurons/cytology , Neurons/immunology , Neurons/metabolism , Nitric Oxide Donors/pharmacology , Nitrites/administration & dosage , Nitrites/antagonists & inhibitors , Nitrites/pharmacology , Nitrogen Oxides/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , Touch
19.
Eur J Clin Pharmacol ; 69(7): 1375-90, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23508666

ABSTRACT

PURPOSE: To review available evidence on the effectiveness of N-acetylcysteine (NAC) as a prophylactic agent in the prevention of non-contrast media agent-induced kidney injury. METHOD: Data were collected by searching Scopus, PubMed, Medline, Science direct and Cochrane database systematic reviews. A total of 26 relevant experimental studies up to the date of publication were included in the review. RESULTS: Available evidence shows that NAC has the potential to exert significant protective or ameliorative effects against drug-induced kidney injury in experimental models. The possible suggested renoprotective mechanisms of NAC in different experimental settings were acting as an antioxidant by restoring the pool of intracellular reduced glutathione, scavenging of free radicals, and/or interacting with reactive oxygen species. CONCLUSION: Whether the administration of NAC could be an effective protective clinical strategy to prevent drug-induced kidney injury or not is a question that remains to be answered in future clinical trials.


Subject(s)
Acetylcysteine/therapeutic use , Evidence-Based Medicine , Kidney/drug effects , Protective Agents/therapeutic use , Renal Insufficiency/prevention & control , Animals , Anti-Infective Agents/adverse effects , Anti-Infective Agents/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Antineoplastic Agents/adverse effects , Antineoplastic Agents/antagonists & inhibitors , Antioxidants/therapeutic use , Humans , Renal Insufficiency/chemically induced
20.
J Eur Acad Dermatol Venereol ; 27(8): 1048-52, 2013 Aug.
Article in English | MEDLINE | ID: mdl-22348297

ABSTRACT

BACKGROUND: Active chronic urticaria, identified as a mast cell- and basophil-dependent inflammatory disorder of the skin is able to elicit acute phase response (APR). However, systemic inflammatory response in different types of urticaria is poorly characterized. AIM: To determine APR pattern in a clearly defined group of patients with acute urticaria and/or angioedema - induced by NSAIDs. METHODS: Plasma IL-6 and serum C-reactive protein (CRP) concentrations were studied in 17 patients with NSAIDs-induced acute urticaria/angioedema (NSAIDsAU) and in 20 healthy controls. Eleven patients who used NSAIDs were presented at the emergency room with acute urticaria/angioedema while the remaining six manifested the symptoms during the aspirin challenge test. Patients were examined in a dynamic manner: during the acute phase, and next, after subsidence of the symptoms. RESULTS: CRP and IL-6 concentrations increased significantly in patients with NSAIDsAU as compared with their asymptomatic period and the healthy subjects. In addition, NSAIDsAU patients showed elevated concentration of the biomarkers following aspirin provocation with the baseline values recovered in the asymptomatic period. CONCLUSION: These results indicate that an acute systemic inflammatory response is activated in patients with NSAIDs-induced urticaria and/or angioedema. The study supports the evidence proving that up-regulation of CRP and IL-6 in urticaria/angioedema does not necessarily reflect any concomitant infection or other inflammatory processes, but may be due to the disease itself.


Subject(s)
Angioedema/chemically induced , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Aspirin/administration & dosage , Biomarkers/blood , Inflammation/diagnosis , Urticaria/chemically induced , Adolescent , Adult , Angioedema/blood , C-Reactive Protein/analysis , Female , Humans , Interleukin-6/blood , Male , Middle Aged , Urticaria/blood , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL