Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Clin Invest Med ; 46(3): E19-33, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37769275

ABSTRACT

BACKGROUND: Stroke is a major contributor to disability and death worldwide. Studies have demonstrated that inflammasome/pyroptosis and its mediated inflammatory response are important factors aggravating brain injury after stroke. We aimed to investigate and map the knowledge structure and global trends on inflam- masome/pyroptosis in stroke. METHODS: All relevant documents were obtained from the Web of Science on 5 June 2023. Bibliometric visualization diagrams were created using VOSviewer and CiteSpace. Excel was used for statistical analysis and drawing graphs. RESULTS: A total of 1106 publications were included, with more articles published each year, especially since 2014. China (740 papers), Zhejiang University (57 papers), Wang J (25 papers), and the Journal of Neuroinflammation (45 papers) were the most productive countries, institutions, authors, and journals, respectively. The United States was the country with highest centrality (0.56) and total link strength (171), and all of the top 10 institutions were in China. China and the U.S. cooperated closely. The centralities of the top 10 authors were all lower than 0.01; no leader has yet emerged in this field. "NLRP3 inflammasome" ranked first with 447 occurrences among 2136 keywords, of which 56 terms appeared more than 10 times when categorized into four clusters: cluster 1 (inflammation), cluster 2 (pyroptosis), cluster 3 (NLRP3 inflammasome), and cluster 4 (neuroinflammation). The studies focused on the mechanisms of inflammasome/pyroptosis in stroke were mainly limited to cell and animal experiments. CONCLUSION: Interest in inflammasome/pyroptosis in stroke is progressively increasing. The NLRP3 inflammasome is the most extensively studied and has been a research hotspot. The mechanisms of cell death in stroke are complex and future studies are needed to strengthen the clinical research on the relationship between pyroptosis-related processes and stroke, determine at which stage NLRP3 inflammasome activation, and clarify the detailed mechanism of NLRP3 in stroke.


Subject(s)
Pyroptosis , Stroke , Humans , Animals , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Neuroinflammatory Diseases , Bibliometrics
2.
Adv Healthc Mater ; 10(18): e2100590, 2021 09.
Article in English | MEDLINE | ID: mdl-34292673

ABSTRACT

As an important part of tumor microenvironment, tumor associated macrophages (TAMs) play a vital role in the occurrence, development, invasion, and metastasis of many malignant tumors and can significantly promote the formation of tumor blood vessels and lymphatic vessels, hence TAMs are greatly associated with poor prognosis. The research on nanomedicine has achieved huge progress, and nano-drugs have been widely utilized to treat various diseases through different mechanisms. Therefore, developing nano-drugs that are based on TAMs-associated anti-tumor mechanisms to effectively suppress tumor growth is expected to be a promising research filed. This paper introduces relevant information about TAMs in terms of their origin, and their roles in tumor genesis, development and metastasis. Furthermore, TAMs-related anti-tumor nano-drugs are summarized. Specifically, a wide range of nano-drugs targeting at TAMs are introduced, and categorized according to their therapeutic mechanisms toward tumors. Additionally, various nano delivery platforms using TAMs as cell carriers which aim at inhibiting tumor growth are reviewed. These two parts elucidate that the exploration of nanomedicine is essential to the study on TAMs-related anti-tumor strategies. This review is also intended to provide novel ideas for in-depth investigation on anti-tumor molecular mechanisms and nano-drug delivery systems based on TAMs.


Subject(s)
Nanomedicine , Neoplasms , Humans , Neoplasms/drug therapy , Tumor Microenvironment , Tumor-Associated Macrophages
3.
Sci Rep ; 9(1): 16636, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31719637

ABSTRACT

Alantolactone (ALA) is a sesquiterpene lactone with potent anti-inflammatory activity. However, the effect of ALA on intestinal inflammation remains largely unknown. The present study demonstrated that ALA significantly ameliorated the clinical symptoms of dextran sulfate sodium (DSS)-induced mice colitis as determined by body weight loss, diarrhea, colon shortening, inflammatory infiltration and histological injury. In mice exposed to DSS, ALA treatment significantly lowered pro-inflammatory mediators, including nuclear factor-kappa B (NF-κB) activation. In vitro, ALA inhibited NF-κB nuclear translocation and dose-dependently activated human/mouse pregnane X receptor (PXR), a key regulator gene in inflammatory bowel disease (IBD) pathogenesis. However, the pocket occluding mutants of the ligand-binding domain (LBD) of hPXR, abrogated ALA-mediated activation of the receptor. Overexpression of hPXR inhibited NF-κB-reporter activity and in this setting, ALA further enhanced the hPXR-mediated inhibition of NF-κB-reporter activity. Furthermore, silencing hPXR gene demonstrated the necessity for hPXR in downregulation of NF-κB activation by ALA. Finally, molecular docking studies confirmed the binding affinity between hPXR-LBD and ALA. Collectively, the current study indicates a beneficial effect of ALA on experimental IBD possibly via PXR-mediated suppression of the NF-κB inflammatory signaling.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colitis/drug therapy , Lactones/therapeutic use , NF-kappa B/metabolism , Pregnane X Receptor/metabolism , Sesquiterpenes, Eudesmane/therapeutic use , Signal Transduction/drug effects , Animals , Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate/pharmacology , Disease Models, Animal , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Pregnane X Receptor/drug effects
4.
Int J Nanomedicine ; 13: 5971-5990, 2018.
Article in English | MEDLINE | ID: mdl-30323586

ABSTRACT

BACKGROUND: Abnormal expression of stromal cells and extracellular matrix in tumor stroma creates a tight barrier, leading to insufficient extravasation and penetration of therapeutic agents. Cancer-associated fibroblasts (CAFs) take on pivotal roles encouraging tumor progression. METHOD: To surmount the refractoriness of stroma, we constructed a multi-targeting combined scenario of anti-CAFs agent tranilast and antitumor agent docetaxel micelles (DTX-Ms). Tranilast cut down crosstalk between tumor cells and stromal cells, ameliorated the tumor microenvironment, and enhanced the antiproliferation efficacy of DTX-Ms on cancer cells. RESULTS: Diverse experiments demonstrated that tranilast enhanced DTX-Ms' antitumor effect in a two-stage pattern by CAFs ablation, tumor cell migration blocking, and metastasis inhibition. Along with activated CAFs decreasing in vivo, the two-stage therapy succeeded in reducing interstitial fluid pressure, normalizing microvessels, improving micelles penetration and retention, and inhibiting tumor growth and metastasis. Interestingly, tranilast alone failed to inhibit tumor growth in vivo, and it could only be used as an adjuvant medicine together with an antitumor agent. CONCLUSION: Our proposed two-stage therapy offers a promising strategy to enhance antitumor effects by breaking down CAFs barrier and increasing micellar delivery efficiency.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Micelles , Neoplasms/drug therapy , Taxoids/therapeutic use , 3T3 Cells , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Body Weight , Cancer-Associated Fibroblasts/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Docetaxel , Extracellular Fluid/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Microvessels/drug effects , Microvessels/pathology , Neoplasm Metastasis , Neoplasms/blood supply , Neoplasms/pathology , Organ Specificity , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Tissue Distribution , Tumor Microenvironment/drug effects , ortho-Aminobenzoates/pharmacology , ortho-Aminobenzoates/therapeutic use
5.
Molecules ; 23(6)2018 Jun 07.
Article in English | MEDLINE | ID: mdl-29880739

ABSTRACT

Fargesin is a bioactive lignan from Flos Magnoliae, an herb widely used in the treatment of allergic rhinitis, sinusitis, and headache in Asia. We sought to investigate whether fargesin ameliorates experimental inflammatory bowel disease (IBD) in mice. Oral administration of fargesin significantly attenuated the symptoms of dextran sulfate sodium (DSS)-induced colitis in mice by decreasing the inflammatory infiltration and myeloperoxidase (MPO) activity, reducing tumor necrosis factor (TNF)-α secretion, and inhibiting nitric oxide (NO) production in colitis mice. The degradation of inhibitory κBα (IκBα), phosphorylation of p65, and mRNA expression of nuclear factor κB (NF-κB) target genes were inhibited by fargesin treatment in the colon of the colitis mice. In vitro, fargesin blocked the nuclear translocation of p-p65, downregulated the protein levels of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2), and dose-dependently inhibited the activity of NF-κB-luciferase in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. Taken together, for the first time, the current study demonstrated the anti-inflammatory effects of fargesin on chemically induced IBD might be associated with NF-κB signaling suppression. The findings may contribute to the development of therapies for human IBD by using fargesin or its derivatives.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Benzodioxoles/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Lignans/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Benzodioxoles/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Dextran Sulfate/administration & dosage , Gene Expression Regulation/drug effects , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/chemically induced , Lignans/pharmacology , Luciferases/antagonists & inhibitors , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/antagonists & inhibitors , Peroxidase/antagonists & inhibitors , Proteolysis , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/antagonists & inhibitors
6.
Sci Rep ; 7(1): 16374, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29180692

ABSTRACT

Baicalein (5,6,7-trihydroxyflavone), a predominant bioactive component isolated from the root of Scutellaria baicalensis Georgi, has established potent anti-inflammatory activity via multi-targeted mechanisms. However, little is known about the effect of baicalein on 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis, which shares pathology related to human Crohn's disease (CD). The present study demonstrated that baicalein alleviated the severity of TNBS-induced colitis in mice by decreasing the activity of myeloperoxidase (MPO) and the expression of pro-inflammatory mediators. The decline in the activation of nuclear factor-kappa B (NF-κB) and p38 mitogen-activated protein kinase (MAPK) correlated with a decrease in the expression of mucosal toll-like receptor 4 (TLR4) and its adaptor myeloid differentiation factor 88 (MyD88). In vitro, baicalein down-regulated the TLR4/MyD88 signaling cascades (NF-κB and MAPKs) in lipopolysaccharide (LPS)-stimulated macrophages. At the upstream level, baicalein bound to the hydrophobic region of the myeloid differentiation protein-2 (MD-2) pocket and inhibited the formation of the LPS-induced MD-2/TLR4 complex. Furthermore, baicalein reduced NOD-like receptor 3 (NLRP3) inflammasome activation and downstream interleukin-1ß expression in a dose-dependent manner. Our study provided evidence for the first time that baicalein attenuated TNBS-induced colitis, at least in part, via inhibition of TLR4/MyD88 signaling cascade as well as inactivation of NLRP3 inflammasome.


Subject(s)
Colitis/etiology , Colitis/metabolism , Flavanones/pharmacology , Inflammasomes/metabolism , Myeloid Differentiation Factor 88/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Biomarkers , Cell Line , Cell Survival/drug effects , Colitis/drug therapy , Colitis/pathology , Cytokines/genetics , Cytokines/metabolism , Flavanones/chemistry , Humans , Inflammation Mediators/metabolism , MAP Kinase Signaling System , Mice , Models, Molecular , Molecular Conformation , Myeloid Differentiation Factor 88/chemistry , NF-kappa B/metabolism , Structure-Activity Relationship , Toll-Like Receptor 4/chemistry , Trinitrobenzenesulfonic Acid/adverse effects
7.
Am J Physiol Gastrointest Liver Physiol ; 309(7): G517-27, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26251468

ABSTRACT

Cardamonin is a naturally occurring chalcone with strong anti-inflammatory activity. However, the direct effect of cardamonin on intestinal inflammation remains elusive. In the present study, we found that cardamonin markedly ameliorated dextran sulfate sodium-induced mouse body weight loss, diarrhea, colon shortening, spleen swelling, and histological damage, which correlated with a decline in the activity of myeloperoxidase and the production of nitric oxide, tumor necrosis factor-α and interleukin-6 in the colon. The upregulation of toll-like receptor 4 after dextran sulfate sodium treatment was associated with an increase in the activation of myeloid differentiation factor 88, interleukin-1 receptor-associated kinase-1, nuclear factor-κB (NF-κB) p65, inhibitor κBα, and inhibitor κB kinase-α/ß, as well as the mitogen-activated protein kinase molecules of extracellular signal-regulated kinase and c-Jun NH2-terminal kinase, and this upregulation was reversed by cardamonin administration. Moreover, cardamonin blocked the nuclear translocation of NF-κB p65, inhibited NF-κB-luciferase activity, and downregulated NF-κB target genes expression. The present study clearly demonstrates a beneficial effect of cardamonin on experimental inflammatory bowel disease via a mechanism associated with suppression of toll-like receptor 4 expression and inactivation of NF-κB and mitogen-activated protein kinase pathways. This study may give insight into the further evaluation of the therapeutic potential of cardamonin or its derivatives for human inflammatory bowel disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chalcones/pharmacology , Colitis/drug therapy , Animals , Cell Line , Colitis/chemically induced , Colitis/metabolism , Colon/metabolism , Colon/pathology , Cytokines/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Fluorescent Antibody Technique , Genes, Reporter , Humans , Immunoblotting , Interleukin-1 Receptor-Associated Kinases/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Polymerase Chain Reaction , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism
8.
J Pharmacol Exp Ther ; 352(2): 315-24, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25472953

ABSTRACT

Notoginsenoside R1 (R1) is the main bioactive component in Panax notoginseng, an old herb medicine widely used in Asian countries in the treatment of microcirculatory diseases. However, little is known about the effect of R1 on inflammatory bowel disease (IBD). The present study demonstrated that R1 alleviated the severity of dextran sulfate sodium-induced colitis in mice by decreasing the activity of myeloperoxidase, the production of cytokines, the expression of proinflammatory genes, and the phosphorylation of IκB kinase, IκBα, and p65 in the colon. Further studies indicated that R1 dose-dependently activated human/mouse pregnane X receptor (PXR), a known target for decreasing inflammation in IBD, and upregulated the expression of genes involved in xenobiotic metabolism in colorectal cells and the colon. Ligand pocket-filling mutant (S247W/C284W or S247W/C284W/S208W) of the human PXR abrogated the effect of R1 on PXR activation. Time-resolved fluorescence resonance energy transfer PXR competitive binding assay confirmed R1 (ligand) binding affinity. In addition, PXR overexpression inhibited nuclear factor-κB (NF-κB)-luciferase activity, which was potentiated by R1 treatment. PXR knockdown by small interfering RNA demonstrated the necessity of PXR in R1-induced upregulation of the expression of xenobiotic-metabolizing enzymes and downregulation of NF-κB activity. Finally, the anti-inflammatory effect of R1 was confirmed in trinitrobenzene sulfonic acid-induced colitis in mice. These findings suggest that R1 attenuates experimental IBD possibly via the activation of intestinal PXR signaling.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colon/drug effects , Ginsenosides/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Receptors, Steroid/metabolism , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Colon/immunology , Colon/metabolism , Colon/pathology , Disease Models, Animal , Female , Gene Expression/drug effects , Ginsenosides/administration & dosage , Ginsenosides/pharmacology , HT29 Cells , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Interleukin-6/metabolism , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Peroxidase/metabolism , Pregnane X Receptor , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
9.
Int Immunopharmacol ; 23(1): 170-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25194678

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic and relapsing inflammatory disorder of the gastrointestinal (GI) tract, and currently no curative treatment is available. Mangiferin, a natural glucosylxanthone mainly from the fruit, leaves and stem bark of a mango tree, has a strong anti-inflammatory activity. We sought to investigate whether mangiferin attenuates inflammation in a mouse model of chemically induced IBD. Pre-administration of mangiferin significantly attenuated dextran sulfate sodium (DSS)-induced body weight loss, diarrhea, colon shortening and histological injury, which correlated with the decline in the activity of myeloperoxidase (MPO) and the level of tumor necrosis factor-α (TNF-α) in the colon. DSS-induced degradation of inhibitory κBα (IκBα) and the phosphorylation of nuclear factor-kappa B (NF-κB) p65 as well as the mRNA expression of pro-inflammatory mediators (inducible NO synthase (iNOS), intercellular adhesion molecule-1 (ICAM-1), TNF-α, interleukin-1ß (IL-1ß) and IL-6) in the colon were also downregulated by mangiferin treatment. Additionally, the phosphorylation/activation of DSS-induced mitogen-activated protein kinase (MAPK) proteins was also inhibited by mangiferin treatment. In accordance with the in vivo results, mangiferin exposure blocked TNF-α-stimulated nuclear translocation of NF-κB in RAW264.7 mouse macrophage cells. Transient transfection gene reporter assay performed in TNF-α-stimulated HT-29 human colorectal adenocarcinoma cells indicated that mangiferin inhibits NF-κB transcriptional activity in a dose-dependent manner. The current study clearly demonstrates a protective role for mangiferin in experimental IBD through NF-κB and MAPK signaling inhibition. Since mangiferin is a natural compound with little toxicity, the results may contribute to the effective utilization of mangiferin in the treatment of human IBD.


Subject(s)
Cell Nucleus/metabolism , Colon/drug effects , Inflammatory Bowel Diseases/drug therapy , Phytotherapy/methods , Xanthones/administration & dosage , Animals , Cell Line , Colon/immunology , Cytokines/genetics , Cytokines/metabolism , Dextran Sulfate/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fruit , Humans , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/chemically induced , Mangifera/immunology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Protein Transport/drug effects , Signal Transduction/drug effects , Transcriptional Activation/drug effects
10.
Am J Physiol Gastrointest Liver Physiol ; 306(1): G27-36, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24232001

ABSTRACT

Paeonia lactiflora Pall is one of the most well-known herbs in China, Korea, and Japan for more than 1,200 years. Paeoniflorin, the major bioactive component of peony root, has recently been reported to have anticolitic activity. However, the underlying molecular mechanism is unclear. The present study was to explore the possible mechanism of paeoniflorin in attenuating dextran sulfate sodium (DSS)-induced colitis. Pre- and coadministration of paeoniflorin significantly reduced the severity of colitis and resulted in downregulation of several inflammatory parameters in the colon, including the activity of myeloperoxidase (MPO), the levels of TNF-α and IL-6, and the mRNA expression of proinflammatory mediators (MCP-1, Cox2, IFN-γ, TNF-α, IL-6, and IL-17). The decline in the activation of NF-κB p65, ERK, JNK, and p38 MAPK correlated with a decrease in mucosal Toll-like receptor 4 (TLR4) but not TLR2 or TLR5 expression. In accordance with the in vivo results, paeoniflorin downregulated TLR4 expression, blocked nuclear translocation of NF-κB p65, and reduced the production of IL-6 in LPS-stimulated mouse macrophage RAW264.7 cells. Transient transfection assay performed in LPS-stimulated human colon cancer HT-29 cells indicated that paeoniflorin inhibits NF-κB transcriptional activity in a dose-dependent manner. TLR4 knockdown and overexpression experiments demonstrated a requirement for TLR4 in paeoniflorin-mediated downregulation of inflammatory cytokines. Thus, for the first time, the present study indicates that paeoniflorin abrogates DSS-induced colitis via decreasing the expression of TLR4 and suppressing the activation of NF-κB and MAPK pathways.


Subject(s)
Benzoates/therapeutic use , Bridged-Ring Compounds/therapeutic use , Colitis , Glucosides/therapeutic use , Inflammation/metabolism , Paeonia , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biological Availability , Colitis/chemically induced , Colitis/drug therapy , Colitis/physiopathology , Dextran Sulfate/pharmacology , Drugs, Chinese Herbal , Gene Expression Profiling , HT29 Cells , Humans , Interleukin-6/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinases/metabolism , Models, Animal , Monoterpenes , NF-kappa B/metabolism , Peroxidase/metabolism , Protein Biosynthesis/drug effects , Toll-Like Receptor 4/metabolism , Transcriptional Activation/drug effects , Tumor Necrosis Factor-alpha/metabolism
11.
Br J Nutr ; 110(4): 599-608, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23506745

ABSTRACT

Naringenin, one of the most abundant flavonoids in citrus, grapefruits and tomatoes, has been used as a traditional anti-inflammatory agent for centuries. However, the molecular mechanism of naringenin in intestinal inflammation remains unknown so far. The present study investigated a molecular basis for the protective effect of naringenin in dextran sulphate sodium-induced murine colitis. Pre-administration of naringenin significantly reduced the severity of colitis and resulted in down-regulation of pro-inflammatory mediators (inducible NO synthase (iNOS), intercellular adhesion molecule-1 (ICAM-1), monocyte chemoattractant protein-1 (MCP-1), cyclo-oxygenase-2 (Cox2), TNF-α and IL-6 mRNA) in the colon mucosa. The decline in the production of pro-inflammatory cytokines, specifically TNF-α and IL-6, correlated with a decrease in mucosal Toll-like receptor 4 (TLR4) mRNA and protein. Phospho-NF-κB p65 protein was significantly decreased, which correlated with a similar decrease in phospho-IκBα protein. Consistent with the in vivo results, naringenin exposure blocked lipopolysaccharide-stimulated nuclear translocation of NF-κB p65 in mouse macrophage RAW264.7 cells. In addition, in vitro NF-κB reporter assays performed on human colonic HT-29 cells exposed to naringenin demonstrated a significant inhibition of TNF-α-induced NF-κB luciferase expression. Thus, for the first time, the present study indicates that targeted inhibition of the TLR4/NF-κB signalling pathway might be an important mechanism for naringenin in abrogating experimental colitis.


Subject(s)
Colitis/chemically induced , Colitis/prevention & control , Flavanones/pharmacology , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cell Line , Cytokines/metabolism , Dextran Sulfate/pharmacology , Dietary Supplements , Female , Flavonoids/pharmacology , Inflammation , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction
12.
J Pharmacol Exp Ther ; 345(3): 473-82, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23536316

ABSTRACT

Targeted activation of pregnane X receptor (PXR) in recent years has become a therapeutic strategy for inflammatory bowel disease. Chrysin is a naturally occurring flavonoid with anti-inflammation activity. The current study investigated the role of chrysin as a putative mouse PXR agonist in preventing experimental colitis. Pre-administration of chrysin ameliorated inflammatory symptoms in mouse models of colitis (dextran sodium sulfate- and 2,4,6-trinitrobenzene sulfonic acid-induced) and resulted in down-regulation of nuclear transcription factor κB (NF-κB) target genes (inducible NO synthase, intercellular adhesion molecule-1, monocyte chemotactic protein-1, cyclooxygenase 2, tumor necrosis factor-α, and interleukin 6) in the colon mucosa. Chrysin inhibited the phosphorylation/degradation of inhibitor κBα (IκBα), which correlated with the decrease in the activity of myeloperoxidase and the levels of tumor necrosis factor-α and interleukin 6 in the colon. Consistent with the in vivo results, chrysin blocked lipopolysaccharide -stimulated nuclear translocation of NF-κB p65 in mouse macrophage RAW264.7. Furthermore, chrysin dose-dependently activated human/mouse PXR in reporter gene assays and up-regulated xenobiotic detoxification genes in the colon mucosa, but not in the liver. Silencing of PXR by RNA interference demonstrated necessity of PXR in mediating chrysin's ability to induce xenobiotic detoxification genes and NF-κB inactivation. The repression of NF-κB transcription activity by chrysin was confirmed by in vitro PXR transduction. These findings suggest that the effect of chrysin in preventing chemically induced colitis is mediated in large part by a PXR/NF-κB pathway. The data also suggest that chrysin or chrysin-like flavonoids could be further developed as intestine-specific PXR activators.


Subject(s)
Colitis/drug therapy , Flavonoids/pharmacology , NF-kappa B/drug effects , Receptors, Steroid/drug effects , Signal Transduction/drug effects , Animals , Blotting, Western , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate , Female , Fluorescent Antibody Technique , Gene Silencing/drug effects , Gene Silencing/physiology , Genes, Reporter/drug effects , Humans , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/antagonists & inhibitors , Peroxidase/metabolism , Pregnane X Receptor , RNA/biosynthesis , RNA/isolation & purification , Receptors, Steroid/antagonists & inhibitors , Trinitrobenzenesulfonic Acid , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...