Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-25276218

ABSTRACT

In the previous study, the mixture of the rhizome of Anemarrhena asphodeloides (AA, family Liliaceae) and the rhizome of Coptidis chinensis (CC, family Ranunculaceae) (AC-mix) improved TNBS- or oxazolone-induced colitis in mice. Therefore, to investigate its anticolitic mechanism, we measured its effect in acute and chronic DSS-induced colitic mice and investigated its anti-inflammatory mechanism in peritoneal macrophages. AC-mix potently suppressed DSS-induced body weight loss, colon shortening, myeloperoxidase activity, and TNF-α, IL-1ß, and IL-6 expressions in acute or chronic DSS-stimulated colitic mice. Among AC-mix ingredients, AA, CC, and their main constituents mangiferin and berberine potently inhibited the expression of proinflammatory cytokines TNF-α and IL-1ß, as well as the activation of NF-κB in LPS-stimulated peritoneal macrophages. AA and mangiferin potently inhibited IRAK phosphorylation, but CC and berberine potently inhibited the binding of LPS to TLR4 on macrophages, as well as the phosphorylation of IRAK1. AC-mix potently inhibited IRAK phosphorylation and LPS binding to TLR4 on macrophages. Based on these findings, AC-mix may ameliorate colitis by the synergistic inhibition of IRAK phosphorylation and LPS binding to TLR4 on macrophages.

2.
J Agric Food Chem ; 62(40): 9711-21, 2014 Oct 08.
Article in English | MEDLINE | ID: mdl-25213465

ABSTRACT

Ursolic acid, which was isolated from an ethanol extract of Cornus officinalis seed, potently inhibited nuclear factor κ light-chain enhancer of activated B cells (NF-κB) activation in lipopolysaccharide (LPS)-stimulated peritoneal macrophages. Therefore, we investigated the anti-inflammatory mechanism of ursolic acid in LPS-stimulated macrophages and colitic mice. Ursolic acid inhibited phosphorylation of interleukin 1 receptor-associated kinase (IRAK)1, TAK1, inhibitor of nuclear factor κB kinase subunit ß (IKKß), and IκBα as well as activation of NF-κB and MAPKs in LPS-stimulated macrophages. Ursolic acid suppressed LPS-stimulated interleukin (IL)-1ß, IL-6, tumor necrosis factor (TNF)-α, cyclooxygenase (COX)-2, and inducible NO synthetase (iNOS) expression as well as PGE2 and NO levels. Ursolic acid not only inhibited the Alexa Fluor 488-conjugated LPS-mediated shift of macrophages but also reduced the intensity of fluorescent LPS bound to the macrophages transiently transfected with or without MyD88 siRNA. However, ursolic acid did not suppress NF-κB activation in peptidoglycan-stimulated macrophages. Oral administration of ursolic acid significantly inhibited 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colon shortening and myeloperoxidase (MPO) activity in mice. Ursolic acid also suppressed TNBS-induced COX-2 and iNOS expression as well as NF-κB activation in colon tissues. Ursolic acid (20 mg/kg) also inhibited TNBS-induced IL-1ß, IL-6, TNF-α by 93, 86, and 85%, respectively (p < 0.05). However, ursolic acid reversed TNBS-mediated downregulation of IL-10 expression to 79% of the normal control group (p < 0.05). On the basis of these findings, ursolic acid may ameliorate colitis by regulating NF-κB and MAPK signaling pathways via the inhibition of LPS binding to TLR4 on immune cells.


Subject(s)
Colitis/drug therapy , Cornus/chemistry , Lipopolysaccharides/metabolism , Toll-Like Receptor 4/metabolism , Triterpenes/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colitis/chemically induced , Colitis/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Peptidoglycan/pharmacology , Seeds/chemistry , Toll-Like Receptor 4/genetics , Trinitrobenzenesulfonic Acid/toxicity , Ursolic Acid
3.
Eur J Pharmacol ; 740: 652-61, 2014 Oct 05.
Article in English | MEDLINE | ID: mdl-24972244

ABSTRACT

Mangiferin, a main constituent of the root of Anemarrhena asphodeloides and the leaves of Mangifera indica, inhibits NF-κB activation in macrophages. Therefore, we investigated effect of mangiferin on 2,3,4-trinitrobenzene sulfonic acid (TNBS)-induced colitis in mice and its anti-inflammatory mechanism in lipolysaccharide (LPS)- or peptidoglycan-stimulated mouse peritoneal macrophages. Mangiferin inhibited phosphorylation of nuclear factor-kappaB (NF-κB), interleukin-1 receptor-associated kinase 1 (IRAK1), and mitogen-activated protein kinases (MAPK) in peptidoglycan- or LPS-stimulated peritoneal macrophages. Mangiferin in the presence of SN50 inhibited LPS-stimulated NF-κB activation more potently than mangiferin alone. Mangiferin inhibited interaction of fluorescent p-IRAK1 antibody to LPS-stimulated peritoneal macrophages, but increased binding of fluorescent IRAK1 antibody. Mangiferin did not influence interaction of fluorescent LPS to toll-like receptor-4 on the macrophages. Molecular peak of mangiferin bound to IRAK1 was detected in the macrophages by mass analysis. Mangiferin (10 µM) inhibited LPS-stimulated expression of TNF-α, IL-1ß and IL-6 by 81.0%, 89.5% and 88.3%, respectively, whereas it increased IL-10 expression by 131.8% compared to LPS-nontreated group. Mangiferin furthermore inhibited colon shortening, macroscopic score, and colonic myeloperoxidase activity in TNBS-induced colitic mice. Mangiferin inhibited TNBS-induced IRAK1 phosphorylation and NF-κB activation. Mangiferin suppressed TNBS-induced up-regulation of cyclooxygenase-2 and inducible NO synthase. Furthermore, mangiferin (20mg/kg) significantly inhibited TNF-α by 78%, IL-1ß by 82%, and IL-6 expressions by 88% (P<0.05), but induced IL-10 expression to 79% of the normal control group (P<0.05). Based on these findings, mangiferin may ameliorate inflammatory diseases such as colitis by regulating NF-κB and MAPK signaling pathways through the inhibition of IRAK1 phosphorylation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Colitis/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Xanthones/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Colitis/chemically induced , Colitis/drug therapy , Cytokines/metabolism , Lipopolysaccharides , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice, Inbred C57BL , Peptidoglycan , Phosphorylation/drug effects , Trinitrobenzenesulfonic Acid , Xanthones/therapeutic use
4.
Mol Nutr Food Res ; 58(5): 963-72, 2014 May.
Article in English | MEDLINE | ID: mdl-24402767

ABSTRACT

SCOPE: ß-Sitosterol, a common phytosterol, has been shown to exhibit anti-inflammatory effects. Here, we investigated the effect of ß-sitosterol on high-fat diet (HFD) induced colitis in mice and on LPS-stimulated mouse intestinal macrophages. METHODS AND RESULTS: C57BL/6J mice were maintained on an LFD (10 kcal% fat), an HFD (60 kcal% fat), or an HFD with ß-sitosterol (20 mg/kg) administration for 8 weeks. The increased levels of body weight and epididymal fat pad weight as well as the concentrations of circulating proinflammatory cytokines and LPS in HFD mice compared with LFD mice were decreased by oral administration of ß-sitosterol. The HFD-induced colonic inflammation evidenced by the increased expression of proinflammatory cytokines and the activation of nuclear factor kappa B (NF-κB) in the colon was also inhibited by ß-sitosterol. In LPS-stimulated intestinal macrophages, ß-sitosterol inhibited the production of proinflammatory cytokines and inflammatory enzymes as well as NF-κB activation. In addition, ß-sitosterol significantly prevented the binding of LPS to intestinal as well as peritoneal macrophages. Furthermore, ß-sitosterol potently inhibited the interaction between LPS and toll-like receptor 4 in intestinal macrophages transfected with control siRNA or MyD88 siRNA. CONCLUSION: These findings indicate that ß-sitosterol ameliorates HFD-induced colitis by inhibiting the binding of LPS to toll-like receptor 4 in the NF-κB pathway.


Subject(s)
Diet, High-Fat/adverse effects , Intestines/drug effects , Lipopolysaccharides/metabolism , NF-kappa B/metabolism , Sitosterols/pharmacology , Toll-Like Receptor 4/metabolism , Animals , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Inflammation/drug therapy , Inflammation/pathology , Intestines/pathology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Toll-Like Receptor 4/genetics
5.
Biomol Ther (Seoul) ; 21(5): 398-404, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-24244828

ABSTRACT

During a screening program to search the anticolitic herbal medicines, 80% ethanol extract of the rhizome of Anemarrhena asphodeloides (AA) was found to potently inhibit the expression of proinflammatory cytokines TNF-α and IL-1ß, as well as the activation of NF-κB in LPS-stimulated colonic macrophages, followed by that of the rhizome of C. chinensis (CC). AA also potently inhibited TNBS-induced colitic markers, shortening of the colon and increase of macroscopic score, myeloperoxidase activity, TNF-α, IL-1ß, and IL-6, in mice. The synergistic effect of CC against the anticolitic effect of AA was investigated. CC synergistically inhibited the anticolitic effect of AA. AC-mix (AA+CC, 1:1) potently inhibited them. AC-mix also inhibited the activation of NF-κB, as well as the expression of TNF-α, IL-1ß, IL-6, iNOS and COX-2. The effects of AC-mix against oxazolone-induced colitis were investigated in mice. AC-mix also potently inhibited oxazolone-induced inflammatory markers, colon shortening, macroscopic score, myeloperoxidase activity, NF-κB activation and proinflammatory cytokines. Overall, the anti-colitic effect of AC-mix was superior to that of mesalazine. Based on these findings, AC-mix may improve colitis by inhibiting NF-κB activation.

6.
Br J Pharmacol ; 170(5): 1078-91, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23941302

ABSTRACT

BACKGROUND AND PURPOSE: The gallnut of Rhus chinensis MILL and its main constituent penta-O-galloyl-ß-D-glucose (PGG) inhibited NF-κB activation in LPS-stimulated peritoneal and colonic macrophages. Here we have investigated PGG mechanisms underlying anti-inflammatory effects of PGG in vitro and in vivo. EXPERIMENTAL APPROACH: Male C57BL/6 mice (18-22 g, 6 weeks old) were used to prepare peritoneal and colonic macrophages and for the induction of colitis by intrarectal administration of 2,3,4-trinitrobenzene sulphonic acid (TNBS). A range of inflammatory markers and transcription factors were evaluated by elisa, immunoblotting, flow cytometry and confocal microscopy. KEY RESULTS: Expression of Toll-like receptor (TLR)-4 or Lipopolysaccharide (LPS) binding to TLR-4 in LPS-stimulated peritoneal macrophages was not affected by PGG. However PGG inhibited binding of an anti-MyD88 antibody to peritoneal macrophages, but did not reduce binding of anti-IL-1 receptor-associated kinase (IRAK1) and IRAK4 antibodies to the macrophages with or without transfection with MyD88 siRNA. PGG potently reduced the activation of IRAK1, NF-κB, and MAPKs in LPS- or pepetidoglycan-stimulated peritoneal and colonic macrophages. PGG suppressed IL-1ß, TNF-α and IL-6 in LPS-stimulated peritoneal macrophages, while increasing expression of the anti-inflammatorycytokine IL-10. Oral administration of PGG inhibited colon shortening and myeloperoxidase activity in mice with TNBS-induced colitis, along with reducing NF-κB activation and IL-1ß, TNF-α, and IL-6 levels, whereas it increased IL-10. CONCLUSIONS AND IMPLICATIONS: PGG reduced activation of NF-κB and MAPK signalling pathways by directly interacting with the MyD88 adaptor protein. PGG may ameliorate inflammatory diseases such as colitis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Colitis/prevention & control , Colon/drug effects , Gastrointestinal Agents/pharmacology , Hydrolyzable Tannins/pharmacology , MAP Kinase Signaling System/drug effects , Macrophages, Peritoneal/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Myeloid Differentiation Factor 88/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Administration, Oral , Animals , Anti-Inflammatory Agents/administration & dosage , Cells, Cultured , Colitis/chemically induced , Colitis/enzymology , Colitis/immunology , Colon/enzymology , Colon/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Gastrointestinal Agents/administration & dosage , Hydrolyzable Tannins/administration & dosage , Inflammation Mediators/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Phosphorylation , RNA Interference , Toll-Like Receptor 4/metabolism , Transfection , Trinitrobenzenesulfonic Acid
7.
Eur J Pharmacol ; 708(1-3): 21-9, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23375938

ABSTRACT

Seeds of Arctium lappa, containing arctigenin and its glycoside arctiin as main constituents, have been used as a diuretic, anti-inflammatory and detoxifying agent in Chinese traditional medicine. In our preliminary study, arctigenin inhibited IKKß and NF-κB activation in peptidoglycan (PGN)- or lipopolysaccharide (LPS)-induced peritoneal macrophages. To understand the anti-inflammatory effect of arctigenin, we investigated its anti-inflammatory effect in LPS-stimulated peritoneal macrophages and on LPS-induced systemic inflammation as well as 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis in mice. Arctigenin inhibited LPS-increased IL-1ß, IL-6 and TNF-α expression in LPS-stimulated peritoneal macrophages, but increased LPS-reduced IL-10 and CD204 expression. Arctigenin inhibited LPS-induced PI3K, AKT and IKKß phosphorylation, but did not suppress LPS-induced IRAK-1 phosphorylation. However, arctigenin did not inhibit NF-κB activation in LPS-stimulated PI3K siRNA-treated peritoneal macrophages. Arctigenin suppressed the binding of p-PI3K antibody and the nucleus translocation of NF-κB p65 in LPS-stimulated peritoneal macrophages. Arctigenin suppressed blood IL-1ß and TNF-α level in mice systemically inflamed by intraperitoneal injection of LPS. Arctigenin also inhibited colon shortening, macroscopic scores and myeloperoxidase activity in TNBS-induced colitic mice. Arctigenin inhibited TNBS-induced IL-1ß, TNF-α and IL-6 expression, as well as PI3K, AKT and IKKß phosphorylation and NF-κB activation in mice, but increased IL-10 and CD204 expression. However, it did not affect IRAK-1 phosphorylation. Based on these findings, arctigenin may ameliorate inflammatory diseases, such as colitis, by inhibiting PI3K and polarizing M1 macrophages to M2-like macrophages.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Furans/pharmacology , Lignans/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/therapeutic use , Colitis/chemically induced , Colitis/drug therapy , Colitis/immunology , Cytokines/genetics , Cytokines/immunology , Furans/therapeutic use , Lignans/therapeutic use , Lipopolysaccharides , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Male , Mice , Mice, Inbred C57BL , Peroxidase/immunology , Phosphatidylinositol 3-Kinases/immunology , Proto-Oncogene Proteins c-akt/immunology , Trinitrobenzenesulfonic Acid
8.
Int Immunopharmacol ; 15(2): 433-41, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23352442

ABSTRACT

The rhizome of Codonopsis lanceolata (CL, family Campanulaceae), of which the main constituent is lancemaside A, has been used for cough and bronchitis in traditional Chinese medicine. To evaluate anti-colitic effect of CL, we examined anti-inflammatory effect of CL extracts, lancemaside A and its metabolites in lipopolysaccharide (LPS)-stimulated peritoneal macrophages and 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitic mice. Among CL extracts, CL BuOH extract inhibited LPS-induced IL-1ß, IL-6 and TNF-α expression, as well as NF-κB activation most potently. CL BuOH extract also inhibited colon shortening and myeloperoxidase activity in TNBS-induced colitic mice. Among lancemaside A, a main constituent of CL BuOH extract, and its metabolites (lancemaside X, echinocystic acid-3-O-ß-d-glucopyranoside and echinocystic acid), echinocystic acid inhibited the expression of the pro-inflammatory cytokines, IL-1ß, IL-6, and TNF-α, as well as the phosphorylation of IKKß and p65 in LPS-stimulated peritoneal macrophages most potently. Echinocystic acid also potently inhibited the binding of LPS to TLR4 on peritoneal macrophages. Lancemaside A and its metabolite, echinocystic acid, inhibited TNBS-induced colonic inflammation, including colon shortening, increased myeloperoxidase activity and pro-inflammatory cytokine expression, and NF-κB activation in mice. The anti-colitic effect of echinocystic acid was superior to that of lancemaside A. Based on these findings, orally administered lancemaside A may be metabolized to echinocystic acid, which may express anti-colitic effect by inhibiting the binding of LPS to TLR4 on the macrophages.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Colitis/drug therapy , Colon/drug effects , Macrophages, Peritoneal/drug effects , Oleanolic Acid/analogs & derivatives , Saponins/administration & dosage , Animals , Cells, Cultured , Codonopsis/immunology , Colitis/chemically induced , Colitis/immunology , Colon/pathology , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Macrophages, Peritoneal/immunology , Male , Medicine, Chinese Traditional , Mice , Mice, Inbred ICR , NF-kappa B/metabolism , Oleanolic Acid/administration & dosage , Protein Binding/drug effects , Rhizome , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Trinitrobenzenesulfonic Acid/metabolism
9.
J Agric Food Chem ; 60(38): 9595-602, 2012 Sep 26.
Article in English | MEDLINE | ID: mdl-22849695

ABSTRACT

Ginseng (the root of Panax ginseng C.A. Meyer, family Araliaceae), which contains protopanaxadiol ginsenoside Rb1 and protopanaxatriol ginsenoside Re as main constituents, is frequently used to treat cancer, inflammation, and stress. In the preliminary study, protopanaxatriol ginsenoside Re inhibited NF-κB activation in lipopolysaccharide (LPS)-stimulated murine peritoneal macrophages. Therefore, we investigated its anti-inflammatory effect in peptidoglycan (PGN)-, LPS-, or tumor necrosis factor-α (TNF-α)-stimulated peritoneal macrophages and, in addition, in LPS-induced systemic inflammation and 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis in mice. Ginsenoside Re inhibited IKK-ß phosphorylation and NF-κB activation, as well as the expression of proinflammatory cytokines, TNF-α and IL-1ß, in LPS-stimulated peritoneal macrophages, but it did not inhibit them in TNF-α- or PG-stimulated peritoneal macrophages. Ginsenoside Re also inhibited IRAK-1 phosphorylation induced by LPS, as well as IRAK-1 and IRAK-4 degradations in LPS-stimulated peritoneal macrophages. Ginsenoside Re inhibited the binding of Alexa Fluor 488-conjugated LPS to TLR4 on peritoneal macrophages. Furthermore, ginsenoside Re inhibited the binding of LPS to TLR4 on peritoneal macrophages transiently transfected with MyD88 siRNAs. Orally administered ginsenoside Re significantly inhibited the expression of IL-1ß and TNF-α on LPS-induced systemic inflammation and TNBS-induced colitis in mice. Ginsenoside Re inhibited colon shortening and myeloperoxidase activity in TNBS-treated mice. Ginsenoside Re reversed the reduced expression of tight-junction-associated proteins ZO-1, claudin-1, and occludin. Ginsenoside Re (20 mg/kg) inhibited the activation of NF-κB in TNBS-treated mice. On the basis of these findings, ginsenoside Re may ameliorate inflammation by inhibiting the binding of LPS to TLR4 on macrophages.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Ginsenosides/pharmacology , Inflammation/drug therapy , Lipopolysaccharides/metabolism , Macrophages, Peritoneal/drug effects , Toll-Like Receptor 4/metabolism , Animals , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Cyclooxygenase 2/metabolism , Drug Evaluation, Preclinical , Inflammation/metabolism , Interleukin-1beta/blood , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred ICR , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Peptidoglycan/pharmacology , Phosphorylation/drug effects , RNA, Small Interfering , Trinitrobenzenesulfonic Acid/toxicity , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...