Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Bioengineered ; 12(2): 12461-12469, 2021 12.
Article in English | MEDLINE | ID: mdl-34931923

ABSTRACT

Severe mortality due to the COVID-19 pandemic resulted from the lack of effective treatment. Although COVID-19 vaccines are available, their side effects have become a challenge for clinical use in patients with chronic diseases, especially cancer patients. In the current report, we applied network pharmacology and systematic bioinformatics to explore the use of biochanin A in patients with colorectal cancer (CRC) and COVID-19 infection. Using the network pharmacology approach, we identified two clusters of genes involved in immune response (IL1A, IL2, and IL6R) and cell proliferation (CCND1, PPARG, and EGFR) mediated by biochanin A in CRC/COVID-19 condition. The functional analysis of these two gene clusters further illustrated the effects of biochanin A on interleukin-6 production and cytokine-cytokine receptor interaction in CRC/COVID-19 pathology. In addition, pathway analysis demonstrated the control of PI3K-Akt and JAK-STAT signaling pathways by biochanin A in the treatment of CRC/COVID-19. The findings of this study provide a therapeutic option for combination therapy against COVID-19 infection in CRC patients.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , Colorectal Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Genistein/therapeutic use , Phytoestrogens/therapeutic use , Atlases as Topic , COVID-19/immunology , COVID-19/pathology , COVID-19/virology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/virology , Cyclin D1/genetics , Cyclin D1/immunology , ErbB Receptors/genetics , ErbB Receptors/immunology , Humans , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Janus Kinases/genetics , Janus Kinases/immunology , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Molecular Targeted Therapy/methods , Multigene Family , Network Pharmacology/methods , PPAR gamma/genetics , PPAR gamma/immunology , Pharmacogenetics/methods , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/immunology , SARS-CoV-2/drug effects , SARS-CoV-2/growth & development , SARS-CoV-2/pathogenicity , STAT Transcription Factors/genetics , STAT Transcription Factors/immunology , Signal Transduction
2.
Front Immunol ; 12: 777522, 2021.
Article in English | MEDLINE | ID: mdl-35069549

ABSTRACT

Background: Si-Miao-San (SMS) is a well-known traditional Chinese medicine. This study aims to evaluate the anti-inflammatory effects of SMS on gouty arthritis and its potential mechanism of action. Methods: The effects and mechanism of SMS were evaluated in monosodium urate (MSU)-treated mice or macrophages. The expression of cytokines and PI3K/Akt was analyzed using real-time PCR and Western blotting analyses. Macrophage polarization was assessed with immunofluorescence assays, real-time PCR, and Western blotting. Mass spectrometry was used to screen the active ingredients of SMS. Results: Pretreatment with SMS ameliorated MSU-induced acute gouty arthritis in mice with increased PI3K/Akt activation and M2 macrophage polarization in the joint tissues. In vitro, SMS treatment significantly inhibited MSU-triggered inflammatory response, increased p-Akt and Arg-1 expression in macrophages, and promoted M2 macrophage polarization. These effects of SMS were inhibited when PI3K/Akt activation was blocked by LY294002 in the macrophages. Moreover, SMS significantly reduced serum uric acid levels in the hyperuricemia mice. Using mass spectrometry, the plant hormones ecdysone and estrone were detected as the potentially effective ingredients of SMS. Conclusion: SMS ameliorated MSU-induced gouty arthritis and inhibited hyperuricemia. The anti-inflammatory mechanism of SMS may exert anti-inflammatory effects by promoting M2 polarization via PI3K/Akt signaling. Ecdysone and estrone might be the potentially effective ingredients of SMS. This research may provide evidence for the application of SMS in the treatment of gout.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Gout , Macrophages , Uric Acid , Animals , Chromones/pharmacology , Gout/drug therapy , Gout/immunology , Gout/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , THP-1 Cells , Uric Acid/immunology , Uric Acid/metabolism
3.
Int Immunopharmacol ; 84: 106567, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32413737

ABSTRACT

Costunolide (CTL) is the major sesquiterpene lactone from Radix Aucklandiae, which is widely used on the treatment of gastrointestinal diseases. However, the therapeutic effect of costunolide in ulcerative colitis (UC) is still unknown. Herein, we sought to evaluate the therapeutic effects and underlying mechanisms of costunolide on UC. ICR mice were intraperitoneally administered with costunolide (10 mg/kg) for 10 days. Beginning on the 4th day of drug administration, acute colitis was induced by feeding 4% dextran sulfate sodium (DSS) for additional 7 days. Costunolide markedly attenuated DSS-induced body weight loss, colonic shortening, elevation in disease activity index, and pathological damage of colon, and decreased the number of CD4+ T cells in colon tissues. Furthermore, costunolide significantly inhibited myeloperoxidase (MPO) activity and nitric oxide (NO) level in colon tissues in DSS-exposed mice. Meanwhile, costunolide also suppressed DSS-induced expression of induced nitric oxide synthase (iNOS), interleukin-1ß (IL-1ß), IL-6, tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ) in both mRNA and protein levels. Mechanistically, costunolide repressed the phosphorylation of nuclear factor kappa-B (NF-κB) p65 and degradation of inhibitor of NF-κB (IκB), as well as the excessive activation of signal transducers and activators of transcription 1/3 (STAT1/3) and serine/threonine protein kinase Akt (Akt) in colon tissues in DSS-challenged mice. These findings successfully demonstrated that costunolide ameliorated DSS-induced murine acute colitis by suppressing inflammation through inactivation of NF-κB, STAT1/3, and Akt pathways. These results also suggested that costunolide may be a potential therapeutic agent for the treatment of acute UC.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colitis, Ulcerative/drug therapy , Sesquiterpenes/therapeutic use , Acute Disease , Animals , Anti-Inflammatory Agents/pharmacology , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Colon/drug effects , Colon/immunology , Colon/pathology , Cytokines/genetics , Cytokines/immunology , Dextran Sulfate , Male , Mice, Inbred ICR , NF-kappa B/immunology , Nitric Oxide/immunology , Peroxidase/immunology , Proto-Oncogene Proteins c-akt/immunology , STAT1 Transcription Factor/immunology , STAT3 Transcription Factor/immunology , Sesquiterpenes/pharmacology , Signal Transduction/drug effects
4.
Int J Mol Sci ; 19(8)2018 Aug 07.
Article in English | MEDLINE | ID: mdl-30087236

ABSTRACT

Osteoarthritis (OA) is a degenerative joint disease that is characterized by irreversible articular cartilage destruction by inflammatory reaction. Among inflammatory stimuli, interleukin-1ß (IL-1ß) is known to play a crucial role in OA pathogenesis by stimulating several mediators that contribute to cartilage degradation. Recently, the marine brown alga Sargassum serratifolium has been reported to exhibit antioxidant and anti-inflammatory effects in microglial and human umbilical vein endothelial cell models using lipopolysaccharide and tumor necrosis factor-α, but its beneficial effects on OA have not been investigated. This study aimed to evaluate the anti-osteoarthritic effects of ethanol extract of S. serratifolium (EESS) in SW1353 human chondrocytes and, in parallel, primary rat articular chondrocytes. Our results showed that EESS effectively blocked the generation of reactive oxygen species in IL-1ß-treated SW1353 and rat primary chondrocytes, indicating that EESS has a potent antioxidant activity. EESS also attenuated IL-1ß-induced production of nitric oxide (NO) and prostaglandin E2, major inflammatory mediators in these cells, which was associated with the inhibition of inducible NO synthase and cyclooxygenase-2 expression. Moreover, EESS downregulated the level of gene expression of matrix metalloproteinase (MMP)-1, -3 and -13 in SW1353 chondrocytes treated with IL-1ß, resulting in their extracellular secretion reduction. In addition, the IL-1ß-induced activation of nuclear factor-kappa B (NF-κB) was restored by EESS. Furthermore, EESS reduced the activation of p38 mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3-kinase (PI3K)/Akt signaling pathways upon IL-1ß stimulation. These results indicate that EESS has the potential to exhibit antioxidant and anti-inflammatory effects through inactivation of the NF-κB, p38 MAPK, and PI3K/Akt signaling pathways. Collectively, these findings demonstrate that EESS may have the potential for chondroprotection, and extracts of S. serratifolium could potentially be used in the prevention and treatment of OA.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Chondrocytes/drug effects , Interleukin-1beta/immunology , Plant Extracts/pharmacology , Sargassum , Animals , Anti-Inflammatory Agents/chemistry , Antioxidants/chemistry , Cell Line , Cells, Cultured , Chondrocytes/immunology , Humans , Inflammation/drug therapy , Inflammation/immunology , Male , NF-kappa B/immunology , Osteoarthritis/drug therapy , Osteoarthritis/immunology , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/immunology , Plant Extracts/chemistry , Proto-Oncogene Proteins c-akt/immunology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/immunology , Sargassum/chemistry , p38 Mitogen-Activated Protein Kinases/immunology
5.
J Med Food ; 20(12): 1152-1159, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29243967

ABSTRACT

Porphyra tenera, also known as nori, is a red algal species of seaweed. It is cultivated in Asia for culinary purposes. We report that P. tenera extract (PTE) enhances the immune response in mouse macrophages. We found that P. tenera extract regulates the NF-κB IκB kinase (IKK) signaling pathway, and we assessed the expression and translocation of p65, a subunit of NF-κB, in RAW264.7 mouse macrophage cells after treatment with PTE. We also investigated the effects of 10% ethanol PTE (PTE10) in RAW264.7 cells. The production of IL-10, IL-6, TNF-α, and IFN-γ was induced by PTE treatment of the macrophages, and PTE also enhanced p-IκB and p-AKT. PTE10 showed no cytotoxicity at 10-20 µg/mL in RAW264.7 cells. PTE10, in fact, increased cell viability at 24 h, stimulated macrophage cells, and induced the phosphorylation of Akt. Akt stimulates IKK activity through the phosphorylation of IKKα and enhances immune activity through the activation of NF-κB. In this study, NF-κB activation was induced by increasing p-NF-κB and p-IKK. A subunit of NF-κB, p65, was located in the nucleus and increased the expression of various cytokines. PTE thus enhanced the immune response through IκB-α immunostimulation signaling in RAW264.7 cells. PTE10 has potential therefore for development of future treatments requiring immune system stimulation.


Subject(s)
Macrophages/drug effects , Macrophages/immunology , NF-kappa B/immunology , Plant Extracts/pharmacology , Porphyra/chemistry , Seaweed/chemistry , Animals , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Mice , NF-kappa B/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction/drug effects
6.
Food Funct ; 8(9): 3165-3177, 2017 Sep 20.
Article in English | MEDLINE | ID: mdl-28782772

ABSTRACT

Alcohol intake is a major risk factor for the pathogenesis of alcoholic liver diseases. Accumulating evidence suggests that green tea protects against alcoholic liver injury; however, the underlying mechanisms remain unclear. The present study investigated the role of endothelial nitric oxide synthase (eNOS) in the protective effects of green tea against alcohol-induced liver injury and inflammation. Ethanol was intragastrically administered to male C57BL/6 mice once a day, and the mice were allowed free access to green tea infusion or water for two weeks. We assessed the plasma levels of alanine aminotransferase and aspartate aminotransferase, hepatic contents of thiobarbituric acid reactive substances, malondialdehyde and triglyceride and hepatic mRNA expression of pro-inflammatory cytokines (interleukin-1ß, tumor necrosis factor-α, and interleukin-6). Our results showed that compared with water alone, green tea infusion markedly reduced liver damage, hepatic oxidative stress, hepatic lipid accumulation and inflammatory response. Green tea infusion also significantly reduced hepatic nuclear factor-κB expression and its downstream inflammatory mediators (inducible nitric oxide synthase and cyclooxygenase-2) mRNA levels in ethanol-treated mice. Additionally, green tea infusion significantly activated hepatic phosphorylated phosphatidylinositol 3-kinase (PI3K) and phosphorylated protein kinase B (Akt), which are associated with the upregulation of phosphorylated eNOS expression and the increase of plasma nitric oxide levels in ethanol-treated mice. Furthermore, the protective effects of green tea infusion were considerably inhibited by the eNOS inhibitor NG-nitro-l-arginine methyl ester in ethanol-treated mice. In conclusion, our study demonstrated that the protective effects of green tea infusion on alcohol-induced liver injury and inflammation involve the modulation of the PI3K/AKT/eNOS pathway.


Subject(s)
Camellia sinensis/chemistry , Liver Diseases, Alcoholic/prevention & control , Nitric Oxide Synthase Type III/immunology , Phosphatidylinositol 3-Kinases/immunology , Plant Preparations/administration & dosage , Proto-Oncogene Proteins c-akt/immunology , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Ethanol/adverse effects , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Liver Diseases, Alcoholic/genetics , Liver Diseases, Alcoholic/immunology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Nitric Oxide Synthase Type III/genetics , Phosphatidylinositol 3-Kinases/genetics , Protective Agents/administration & dosage , Proto-Oncogene Proteins c-akt/genetics , Tea/chemistry
7.
Food Funct ; 8(3): 1299-1312, 2017 Mar 22.
Article in English | MEDLINE | ID: mdl-28251195

ABSTRACT

The aim of this study was to investigate the molecular mechanism underlying the immunomodulatory effect of the purified Artemisia sphaerocephala Krasch seed polysaccharide (ASKP-1) in RAW264.7 macrophages. Chemical characteristic analysis revealed that ASKP-1 consisted of 14.1% mannose, 56.9% glucose and 19.6% galactose with the average molecular weight of 9.08 × 105 Da and the mixed glycan backbone structure containing 1→4)-Glcp (39.8%), 1→6)-Galp (18.8%), 1→3,6)-Manp (19.6%), 1→)-Glcp (10.8%), 2→6)-Manp (4.0%) and 2→3,5)-Araf (7.0%). In vitro studies showed that ASKP-1 markedly induced the release of cytotoxic molecules (NO and ROS) and secretion of the cytokines (TNF-α, INF-ß, and IL-6) and significantly enhanced the phagocytosis of RAW264.7 macrophages. Furthermore, TLR4 was found to be a recognized target of ASKP-1 and its related mitogen-activated protein (MAPK) and phosphoinositide 3-kinase (PI3K)/Akt, including phosphorylated ERK, JNK, p38 and Akt, were rapidly activated by ASKP-1 in RAW264.7 macrophages. Moreover, ASKP-1 was found to cause the nuclear translocation of the nuclear factor NF-κB subunit p65 and the degradation of IκB-α in RAW264.7 macrophages. All these findings suggest that MAPK, PI3K/Akt and NF-κB pathways are involved in ASKP-1-induced macrophage activation, and ASKP-1 is a potential immunomodulating function food.


Subject(s)
Artemisia/chemistry , Macrophage Activation/drug effects , Mitogen-Activated Protein Kinase Kinases/immunology , NF-kappa B/immunology , Phosphatidylinositol 3-Kinases/immunology , Plant Extracts/pharmacology , Polysaccharides/pharmacology , Proto-Oncogene Proteins c-akt/immunology , Animals , Interleukin-6/genetics , Interleukin-6/immunology , Macrophages/drug effects , Macrophages/immunology , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , NF-kappa B/genetics , Phosphatidylinositol 3-Kinases/genetics , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Proto-Oncogene Proteins c-akt/genetics , RAW 264.7 Cells , Seeds/chemistry , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
8.
Biosci Biotechnol Biochem ; 80(5): 911-9, 2016 May.
Article in English | MEDLINE | ID: mdl-26943486

ABSTRACT

Pterocarpus indicus Willd has been widely used as a traditional medicine to treat edema, cancer, and hyperlipidemia, but its antiallergic properties and underlying mechanisms have not yet been studied. The purpose of this study was to evaluate the antiallergic activity of Pterocarpus indicus Willd water extract (PIW) using activated mast cells and an atopic dermatitis (AD)-like mouse model. PIW decreased IgE/Ag-induced mast cell degranulation and the phosphorylation of Syk and downstream signaling molecules such as PLC-γ, Akt, Erk 1/2, JNK compared to stimulated mast cells. In DNCB-induced AD-like mice, PIW reduced IgE level in serum, as well as AD-associated scratching behavior and skin severity score. These results indicate that PIW inhibits the allergic response by reducing mast cell activation and may have clinical potential as an antiallergic agent for disorders such as AD.


Subject(s)
Anti-Allergic Agents/pharmacology , Dermatitis, Atopic/drug therapy , Immunoglobulin E/blood , Mast Cells/drug effects , Pterocarpus/chemistry , Skin/drug effects , Animals , Cell Degranulation/drug effects , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Dinitrochlorobenzene , Disease Models, Animal , Gene Expression Regulation , Irritants , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/immunology , Male , Mast Cells/immunology , Mast Cells/pathology , Mice , Mice, Inbred ICR , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Phospholipase C gamma/genetics , Phospholipase C gamma/immunology , Phosphorylation/drug effects , Plant Extracts/chemistry , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction , Skin/immunology , Skin/pathology , Syk Kinase/genetics , Syk Kinase/immunology , Water
9.
Biochem Pharmacol ; 106: 56-69, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26883061

ABSTRACT

Class IA phosphoinositide 3-kinases (PI3Ks) are essential to function of normal and tumor cells, and to modulate immune responses. T lymphocytes express high levels of p110α and p110δ class IA PI3K. Whereas the functioning of PI3K p110δ in immune and autoimmune reactions is well established, the role of p110α is less well understood. Here, a novel dual p110α/δ inhibitor (ETP-46321) and highly specific p110α (A66) or p110δ (IC87114) inhibitors have been compared concerning T cell activation in vitro, as well as the effect on responses to protein antigen and collagen-induced arthritis in vivo. In vitro activation of naive CD4(+) T lymphocytes by anti-CD3 and anti-CD28 was inhibited more effectively by the p110δ inhibitor than by the p110α inhibitor as measured by cytokine secretion (IL-2, IL-10, and IFN-γ), T-bet expression and NFAT activation. In activated CD4(+) T cells re-stimulated through CD3 and ICOS, IC87114 inhibited Akt and Erk activation, and the secretion of IL-2, IL-4, IL-17A, and IFN-γ better than A66. The p110α/δ inhibitor ETP-46321, or p110α plus p110δ inhibitors also inhibited IL-21 secretion by differentiated CD4(+) T follicular (Tfh) or IL-17-producing (Th17) helper cells. In vivo, therapeutic administration of ETP-46321 significantly inhibited responses to protein antigen as well as collagen-induced arthritis, as measured by antigen-specific antibody responses, secretion of IL-10, IL-17A or IFN-γ, or clinical symptoms. Hence, p110α as well as p110δ Class IA PI3Ks are important to immune regulation; inhibition of both subunits may be an effective therapeutic approach in inflammatory autoimmune diseases like rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , CD4-Positive T-Lymphocytes/drug effects , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Subunits/antagonists & inhibitors , Pyrazines/pharmacology , Animals , Antibodies/pharmacology , Arthritis, Experimental/enzymology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , CD28 Antigens/genetics , CD28 Antigens/immunology , CD3 Complex/genetics , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/immunology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , Gene Expression , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Lymph Nodes/drug effects , Lymph Nodes/enzymology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , Protein Subunits/genetics , Protein Subunits/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology
10.
Immunobiology ; 221(2): 137-44, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26514297

ABSTRACT

Previous studies revealed the potent anti-inflammatory activity of tricin, the active component of Njavara rice bran. Here, we report the involvement of specific signaling pathways in the protective effect of tricin against LPS induced inflammation in hPBMCs and the role of tricin in modulating endothelial dysfunction in LPS induced HUVECs. Pretreatment with tricin (15µM) significantly inhibited the release of TNF-α and was comparable to the specific pathway blockers like ERK inhibitor (PD98059), JNK inhibitor (SP600125) and p38 inhibitor (SB203580), whereas an increased release of TNF-α was observed in PI3K/Akt inhibitor (LY294002) treated cells. Tricin alone and combination treatment of tricin and SB203580 showed more significant inhibition of activation of COX-2 and TNF-α than that of SB203580 alone treated group. Combination treatment of tricin and LY294002 showed increased activation of COX-2 and TNF-α, proved that PI3K activation is essential for the anti-inflammatory effect of tricin. Studies conducted on HUVECs revealed the protective effect of tricin against endothelial dysfunction associated with LPS induced inflammation by inhibiting the activation of proinflammatory mediators like TNF-α, IFN-γ, MCP 1 by modulating NF-κB and MAPK signaling pathways. ELISA and flow cytometric analysis again confirmed the protection of tricin against endothelial damage, especially from the decreased activation of cell adhesion molecules like ICAM-1, VCAM-1 and E-Selectin upon tricin treatment. This work establishes the mechanism behind the potent anti-inflammatory activity of the flavonoid tricin.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Flavonoids/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Oryza/chemistry , Proto-Oncogene Proteins c-akt/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , E-Selectin/genetics , E-Selectin/immunology , Flavonoids/isolation & purification , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/immunology , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lipopolysaccharides/pharmacology , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/immunology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Phosphoinositide-3 Kinase Inhibitors , Plant Extracts/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/immunology
11.
Mol Med Rep ; 11(6): 4087-92, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25634255

ABSTRACT

At least 50% of hypertrophic scarring (HS) is characterized by inflammation, for which there is currently no effective treatment available. Emodin is a major component of the widely used Chinese herb, rhubarb, which has been used to treat inflammation in several types of disease. However, few studies have investigated the efficacy of emodin in the treatment of HS. In the present study, a mouse model with mechanical stress­induced HS was used to investigate the effects of emodin (20, 40, 80, or 120 mg/ml) on HS, and to determine the potential underlying mechanisms. Treatment with emodin significantly attenuated HS inflammation, as determined by histopathological assessment of the scar elevation index, collagen structure and inflammation (P<0.05). Furthermore, treatment with emodin (40 mg/ml) markedly inhibited phosphoinositide 3­kinase (PI3K)/Akt activity (P<0.01) and this attenuation was associated with reduced expression levels of tumor necrosis factor­α, interleukin­6 and monocyte chemoattractant protein­1 (P<0.05) in the HS tissue. The results of the present study indicated that administration of emodin had therapeutic effects on the progression of HS and the underlying mechanism of this may be due to inhibition of the PI3K/Akt signaling pathway.


Subject(s)
Cicatrix, Hypertrophic/complications , Cicatrix, Hypertrophic/drug therapy , Drugs, Chinese Herbal/therapeutic use , Emodin/therapeutic use , Inflammation/complications , Inflammation/drug therapy , Animals , Cicatrix, Hypertrophic/etiology , Cicatrix, Hypertrophic/immunology , Female , Inflammation/etiology , Inflammation/immunology , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/immunology , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction/drug effects , Stress, Mechanical
12.
Zhongguo Zhong Yao Za Zhi ; 39(12): 2314-7, 2014 Jun.
Article in Chinese | MEDLINE | ID: mdl-25244766

ABSTRACT

To establish neonatal rat cardiac fibroblast inflammatory secretion model by using LPS 100 microg x L(-1) combined with ATP 5 mmol x L(-1), in order to study the inhibitory effect of quercetin on the secretion of inflammatory factors TNF-alpha, IL-1beta and IL-6 of cardiac fibroblasts, further investigate the effect of quercetin on the protein expression of p-NF-kappaB p65 (S276) and p-Akt (S473) by western blot, and discuss the inhibitory effect of quercetin on the inflammatory secretion of cardiac fibroblasts. According to the findings, quercetin with the concentrations between 51.74 micromol x L(-1) and 827.81 micromol x L(-1) had no significant effect on the activity of cardiac fibroblasts. Quercetin with the concentrations of 82.78, 41.39, 20.70 micromol x L(-1) could notably inhibit the increase of TNF-alpha and IL-1beta induced by LPS 100 microg x L(-1) for 3 h and then ATP 5 mmol x L(-1) for 36 h (P < 0.01 or P < 0.05). Quercetin with the concentrations of 82.78, 41.39 micromol x L(-1) could notably inhibit the increase of IL-6 induced LPS 100 microg x L(-1) for 3 h and then ATP 5 mmol x L(-1) for 36 h (P < 0.05), without any notable effect of quercetin with the concentration of 20.70 micromol x L(-1). Quercetin with the concentrations of 82.78, 41.39, 20. 70 micromol x L(-1) could notably inhibit the NF-kappaB p65 (S276) activation induced by LPS 100 microg x L(-1) for 3 h and then ATP 5 mmol x L(-1) for 15 min, with the most significant effect in 20.70 micromol x L(-1). Quercetin with the concentrations of 82.78, 41.39, 20.70 micromol x L(-1) could notably inhibit the increase of p-Akt(473) expression induced by LPS 100 microg x L(-1) for 3 h and then ATP 5 mmol x L(-1) for 240 min (P < 0.05). Therefore, this study believes that quercetin could attenuate the secretion of inflammatory factors TNF-alpha, IL-1beta and IL-6 of cardiac fibroblasts by inhibiting the activation of NF-kappaB p65 (S276) and Akt (473).


Subject(s)
Drugs, Chinese Herbal/administration & dosage , Endomyocardial Fibrosis/drug therapy , Fibroblasts/drug effects , Quercetin/administration & dosage , Animals , Cells, Cultured , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/immunology , Female , Fibroblasts/immunology , Heart/drug effects , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Male , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
13.
Cell Immunol ; 290(1): 21-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24859013

ABSTRACT

α-Viniferin is an oligostilbene of trimeric resveratrol and has anticancer activity; however, the molecular mechanism underlying the anti-inflammatory effects of α-viniferin has not been completely elucidated thus far. Therefore, we determined the mechanism by which α-viniferin regulates lipopolysaccharide (LPS)-induced expression of proinflammatory mediators in BV2 microglial cells. Treatment with α-viniferin isolated from Clematis mandshurica decreased LPS-induced production of nitric oxide (NO) and prostaglandin E2 (PGE2). α-Viniferin also downregulated the LPS-induced expression of proinflammatory genes such as iNOS and COX-2 by suppressing the activity of nuclear factor kappa B (NF-κB) via dephosphorylation of Akt/PI3K. Treatment with a specific NF-κB inhibitor, pyrrolidine dithiocarbamate (PDTC), indirectly showed that NF-κB is a crucial transcription factor for expression of these genes in the early stage of inflammation. Additionally, our results indicated that α-viniferin suppresses NO and PGE2 production in the late stage of inflammation through induction of heme oxygenase-1 (HO-1) regulated by nuclear factor erythroid 2-related factor (Nrf2). Taken together, our data indicate that α-viniferin suppresses the expression of proinflammatory genes iNOS and COX-2 in the early stage of inflammation by inhibiting the Akt/PI3K-dependent NF-κB activation and inhibits the production of proinflammatory mediators NO and PGE2 in the late stage by stimulating Nrf2-mediated HO-1 signaling pathway in LPS-stimulated BV2 microglial cells. These results suggest that α-viniferin may be a potential candidate to regulate LPS-induced inflammation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzofurans/pharmacology , Cyclooxygenase 2/biosynthesis , Microglia/immunology , NF-kappa B/antagonists & inhibitors , Nitric Oxide Synthase Type II/biosynthesis , Animals , Cell Line , Clematis , Dinoprostone/biosynthesis , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/immunology , Inflammation Mediators , Lipopolysaccharides , Membrane Proteins/biosynthesis , Membrane Proteins/immunology , Mice , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , NF-kappa B/genetics , Nitric Oxide/biosynthesis , Phosphatidylinositol 3-Kinases/immunology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Plant Extracts , Plant Roots , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/immunology , Pyrrolidines/pharmacology , RNA Interference , RNA, Small Interfering , Thiocarbamates/pharmacology
14.
Curr Med Chem ; 21(8): 1050-60, 2014.
Article in English | MEDLINE | ID: mdl-24180280

ABSTRACT

DEAD-box RNA helicase DDX3 is a well-known host factor that inhibits hepatitis B viral proliferation and boosts innate immune responses via TANK-binding kinase 1 (TBK1)/IKKε-mediated and/or interferon (IFN)-ß promoter stimulator-1 (IPS-1)-mediated IFN-ß induction. Previously, we demonstrated the anti-hepatitis B activity of Rg3 via stimulation of TRAF6/TAK1 degradation and inhibition of JNK/AP-1 signaling. To determine the effects of Rg3 on innate immunity, an IFN-ß promoter assay was performed. Rg3 ameliorated IFN-ß expression via upregulation of both the TBK1/IKKε pathway and DDX3 expression. In addition, Rg3 induced the phosphorylation of IRF3 and its translocation into nucleus, which is a key molecule to induction of IFN-ß expression. To evaluate the molecular mechanism of Rg3 on DDX3 expression, the DDX3 promoter (-1406/+105) was subjected to luciferase assay and ChIP analysis. p53 phosphorylation resulted in upregulation of DDX3 expression, which enhanced DDX3 promoter transactivation activity. Transient transfection with wild-type p53 increased DDX3 promoter activity in Hep3B cells which have null mutant of p53, whereas knockdown p53 by si-p53 reduced DDX3 promoter activity in HepG2.2.15 and HepG2 cells, respectively. Rg3- mediated phosphorylation of p53 resulted in inhibition of Akt phosphorylation, which in turn reduced MDM2-mediated p53 degradation. An Akt inhibitor augmented DDX3 promoter activity and reduced the secretion of hepatitis B surface antigen. Our data indicate that Rg3 enhances innate immunity by inducing IFN-ß expression through upregulation of DDX3 promoter activity via p53-mediated transactivation and activation of the TBK1/IKKε/IRF3 pathway.


Subject(s)
DEAD-box RNA Helicases/genetics , Ginsenosides/pharmacology , I-kappa B Kinase/immunology , Interferon Regulatory Factor-3/immunology , Protein Serine-Threonine Kinases/immunology , Proto-Oncogene Proteins c-akt/immunology , Tumor Suppressor Protein p53/immunology , Hep G2 Cells , Humans , Immunity, Innate/drug effects , Interferon-beta/genetics , Panax/chemistry , Signal Transduction/drug effects , Up-Regulation/drug effects
15.
Food Funct ; 5(2): 257-64, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24258363

ABSTRACT

Our previous data showed that the Momordica grosvenori Swingle extract (MSE) exhibited the anti-inflammatory effect through markedly suppressed LPS-induced up-regulation of inducible NO synthase (iNOS), cyclooxygenase-2 (COX-2) and ODC (ornithine decarboxylase) gene expression in RAW 264.7 cells. Regarding the link between inflammation and carcinogenesis, we further investigated the bio-molecular mechanisms of both anti-inflammatory and anti-tumor activities in vivo using a TPA (12-O-tetradecanoyl phorbol 13-acetate)-stimulated mouse skin model. Pretreatment with MSE in mouse skin has led to the reduction of TPA-induced nuclear translocation of the nuclear factor-κB (NFκB) subunits as well as phosphorylation of IκBα and p65 subsequent reduction of IκBα degradation. In addition, the MSE inhibitory effect on upstream of NFκB was found to involve the transcriptional effects of MAPK signaling as indicated by strong suppression on TPA-induced activation of extracellular signal regulate kinase (ERK)1/2, p38 mitogen-activated protein kinase (MAPK), c-Jun N-terminal kinase (JNK)1/2, phosphatidylinositol 3-kinase (PI3K) and Akt. Moreover, MSE significantly inhibited 7,12-dimethylbenz[a]anthracene (DMBA)-TPA-induced skin tumor formation in mice measured by the tumor multiplicity of papillomas at 20 weeks. The results suggested that MSE contained promising functional ingredients capable of preventing inflammation-associated tumorigenesis.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Drugs, Chinese Herbal/administration & dosage , Edema/drug therapy , Momordica/chemistry , Skin Neoplasms/prevention & control , Skin/immunology , Animals , Edema/genetics , Edema/immunology , Female , Humans , In Vitro Techniques , Mice , NF-kappa B/genetics , NF-kappa B/immunology , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Skin/drug effects , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/physiopathology , Tetradecanoylphorbol Acetate/adverse effects
16.
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
17.
Exp Parasitol ; 115(3): 226-32, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17049518

ABSTRACT

Angiostrongylus cantonensis is the major cause of human eosinophilic meningoencephalitis. ICR mice were infected orally with 35 infective larvae and sacrificed at 4-14 days, 25 days or 32 days post infection (dpi) for pathological and immunocytochemical examinations. In the non-treated group, no apoptosis signal was found in the meninges or parenchyma of the brains (4-14 dpi). Only a few apoptotic cells were noticed at 25 dpi (3%) and 32 dpi (10%). In the groups, the animals were given a single dose of mebendazole (20 mg/kg, per os at various times) or injections of interleukin 12 (IL-12) (10 ng/daily, intraperitoneally), all the animals were sacrificed at 14 dpi; the number of apoptotic cells was increased (17-21%). In the group that received a single dose of mebendazole (4 dpi) in combination with IL-12 injections (4-13 dpi), mild meningitis was observed, and most of the infiltrated inflammatory cells were in the apoptotic program (55%). Taken together, apoptosis of the inflammatory cells (most were eosinophils) could be induced when the infected mice were treated with mebendazole or/and IL-12.


Subject(s)
Angiostrongylus cantonensis/drug effects , Antinematodal Agents/pharmacology , Brain/pathology , Interleukin-12/pharmacology , Mebendazole/pharmacology , Strongylida Infections/drug therapy , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/therapeutic use , Angiostrongylus cantonensis/physiology , Animals , Antinematodal Agents/therapeutic use , Apoptosis/drug effects , Biomphalaria , Brain/drug effects , Brain/parasitology , Caspase 3/immunology , Drug Therapy, Combination , Eosinophilia/drug therapy , Eosinophilia/parasitology , Eosinophilia/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Interleukin-12/therapeutic use , Male , Mebendazole/therapeutic use , Meningitis/drug therapy , Meningitis/parasitology , Meningitis/pathology , Meningoencephalitis/drug therapy , Meningoencephalitis/parasitology , Meningoencephalitis/pathology , Mice , Mice, Inbred ICR , Proto-Oncogene Proteins c-akt/immunology , Rats , Rats, Wistar , Strongylida Infections/pathology
SELECTION OF CITATIONS
SEARCH DETAIL