17 results on '"Han, Sang-Bae"'
Search Results
2. K284-6111 alleviates memory impairment and neuroinflammation in Tg2576 mice by inhibition of Chitinase-3-like 1 regulating ERK-dependent PTX3 pathway
- Author
-
Ham, Hyeon Joo, Lee, Yong Sun, Yun, Jaesuk, Son, Dong Ju, Lee, Hee Pom, Han, Sang-Bae, and Hong, Jin Tae
- Published
- 2020
- Full Text
- View/download PDF
3. Prevention of multiple system atrophy using human bone marrow-derived mesenchymal stem cells by reducing polyamine and cholesterol-induced neural damages
- Author
-
Park, Kyung-Ran, Hwang, Chul Ju, Yun, Hyung-Mun, Yeo, In Jun, Choi, Dong-Young, Park, Pil-Hoon, Kim, Hyung Sook, Lee, Jung Tae, Jung, Young Suk, Han, Sang-Bae, and Hong, Jin Tae
- Published
- 2020
- Full Text
- View/download PDF
4. Chitinase 3 like 1 suppresses the stability and activity of p53 to promote lung tumorigenesis
- Author
-
Park, Kyung-Ran, Yun, Hyung-Mun, Yoo, Kyeongwon, Ham, Young Wan, Han, Sang Bae, and Hong, Jin Tae
- Published
- 2020
- Full Text
- View/download PDF
5. Bee venom phospholipase A2 ameliorates amyloidogenesis and neuroinflammation through inhibition of signal transducer and activator of transcription-3 pathway in Tg2576 mice
- Author
-
Ham, Hyeon Joo, Han, Sang-Bae, Yun, Jaesuk, Yeo, In Jun, Ham, Young Wan, Kim, Se Hyun, Park, Pil-Hoon, Choi, Dong-Young, and Hong, Jin Tae
- Published
- 2019
- Full Text
- View/download PDF
6. Parkin deficiency prevents chronic ethanol-induced hepatic lipid accumulation through β-catenin accumulation
- Author
-
Lee, Dong Hun, Park, Mi Hee, Hwang, Chul Ju, Kim, Youngsoo, Hwang, Dae Yeon, Han, Sang Bae, and Hong, Jin Tae
- Published
- 2019
- Full Text
- View/download PDF
7. K284-6111 prevents the amyloid beta-induced neuroinflammation and impairment of recognition memory through inhibition of NF-κB-mediated CHI3L1 expression
- Author
-
Choi, Ji Yeon, Yeo, In Jun, Kim, Ki Cheon, Choi, Won Rack, Jung, Jae-Kyung, Han, Sang-Bae, and Hong, Jin Tae
- Published
- 2018
- Full Text
- View/download PDF
8. Inhibition of skin carcinogenesis by suppression of NF-κB dependent ITGAV and TIMP-1 expression in IL-32γ overexpressed condition.
- Author
-
Lee, Yong Sun, Lee, Chung Hee, Bae, Jun Tae, Nam, Kyung Tak, Moon, Dae Bong, Hwang, Ok Kyung, Choi, Jeong Soon, Kim, Tae Hoon, Jun, Hyoung Ok, Jung, Young Suk, Hwang, Dae Yeon, Han, Sang-Bae, Yoon, Do Young, and Hong, Jin Tae
- Subjects
HUMAN carcinogenesis ,INTERLEUKIN-32 ,TISSUE inhibitors of metalloproteinases ,IMMUNOHISTOCHEMISTRY ,WESTERN immunoblotting ,LABORATORY mice ,SKIN tumors ,INTEGRINS - Abstract
Background: Interleukin-32 (IL-32) has been associated with various diseases. Previous studies have shown that IL-32 inhibited the development of several tumors. However, the role of IL-32γ, an isotype of IL-32, in skin carcinogenesis remains unknown. Methods: We compared 7,12-Dimethylbenz[a]anthracene/12-O-Tetradecanoylphorbol-13-acetate (DMBA/TPA)-induced skin carcinogenesis in wild type (WT) and IL-32γ-overexpressing mice to evaluate the role of IL-32γ. We also analyzed cancer stemness and NF-κB signaling in skin cancer cell lines with or without IL-32γ expression by western blotting, quantitative real-time PCR and immunohistochemistry analysis. Results: Carcinogen-induced tumor incidence in IL-32γ mice was significantly reduced in comparison to that in WT mice. Infiltration of inflammatory cells and the expression levels of pro-inflammatory mediators were decreased in the skin tumor tissues of IL-32γ mice compared with WT mice. Using a genome-wide association study analysis, we found that IL-32 was associated with integrin αV (ITGAV) and tissue inhibitor of metalloproteinase-1 (TIMP-1), which are critical factor for skin carcinogenesis. Reduced expression of ITGAV and TIMP-1 were identified in DMBA/TPA-induced skin tissues of IL-32γ mice compared to that in WT mice. NF-κB activity was also reduced in DMBA/TPA-induced skin tissues of IL-32γ mice. IL-32γ decreased cancer cell sphere formation and expression of stem cell markers, and increased chemotherapy-induced cancer cell death. IL-32γ also downregulated expression of ITGAV and TIMP-1, accompanied with the inhibition of NF-κB activity. In addition, IL-32γ expression with NF-κB inhibitor treatment further reduced skin inflammation, epidermal hyperplasia, and cancer cell sphere formation and downregulated expression levels of ITGAV and TIMP-1. Conclusions: These findings indicated that IL-32γ suppressed skin carcinogenesis through the inhibition of both stemness and the inflammatory tumor microenvironment by the downregulation of TIMP-1 and ITGAV via inactivation of NF-κB signaling. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
9. Bee venom ameliorates lipopolysaccharide-induced memory loss by preventing NF-kappaB pathway.
- Author
-
Gu, Sun Mi, Park, Mi Hee, Hwang, Chul Ju, Song, Ho Sueb, Lee, Ung Soo, Han, Sang Bae, Oh, Ki Wan, Ham, Young Wan, Song, Min Jong, Son, Dong Ju, and Hong, Jin Tae
- Abstract
Background: Accumulation of beta-amyloid and neuroinflammation trigger Alzheimer's disease. We previously found that lipopolysaccharide (LPS) caused neuroinflammation with concomitant accumulation of beta-amyloid peptides leading to memory loss. A variety of anti-inflammatory compounds inhibiting nuclear factor kappaB (NF-κB) activation have showed efficacy to hinder neuroinflammation and amyloidogenesis. We also found that bee venom (BV) inhibits NF-κB.Methods: A mouse model of LPS-induced memory loss used administration of BV (0.8 and 1.6 μg/kg/day, i.p.) to ICR mice for 7 days before injection of LPS (2.5 mg/kg/day, i.p.). Memory loss was assessed using a Morris water maze test and passive avoidance test. For in vitro study, we treated BV (0.5, 1, and 2 μg/mL) to astrocytes and microglial BV-2 cells with LPS (1 μg/mL).Results: We found that BV inhibited LPS-induced memory loss determined by behavioral tests as well as cell death. BV also inhibited LPS-induced increases in the level of beta-amyloid (Aβ), β-and γ-secretases activities, NF-κB and its DNA-binding activity and expression of APP, and BACE1 and neuroinflammation proteins (COX-2, iNOS, GFAP and IBA-1) in the brain and cultured cells. In addition, pull-down assay and molecular modeling showed that BV binds to NF-κB.Conclusions: BV attenuates LPS-induced amyloidogenesis, neuroinflammation, and therefore memory loss via inhibiting NF-κB signaling pathway. Thus, BV could be useful for treatment of Alzheimer's disease. [ABSTRACT FROM AUTHOR]- Published
- 2015
- Full Text
- View/download PDF
10. Inhibitory effect of ent-Sauchinone on amyloidogenesis via inhibition of STAT3-mediated NF-κB activation in cultured astrocytes and microglial BV-2 cells.
- Author
-
Song, Suk-Young, Jung, Yu Yeon, Hwang, Chul Ju, Lee, Hee Peom, Sok, Chang Hyun, Kim, Joo Hwan, Lee, Sang Min, Seo, Hyun Ok, Hyun, Byung Kook, Choi, Dong Young, Han, Sang Bae, Ham, Young Wan, Hwang, Bang Yeon, and Hong, Jin Tae
- Abstract
Background: ent-Sauchinone is a polyphenolic compound found in plants belonging to the lignan family. ent-Sauchinone has been shown to modulate the expression of inflammatory factors through the nuclear factor-kappa B (NF-κB) signaling pathway. It is well known that neuroinflammation is associated with amyloidogenesis. Thus, in the present study, we investigated whether ent-Sauchinone could have anti-amyloidogenic effects through the inhibition of NF-κB pathways via its anti-inflammatory property.Methods: To investigate the potential effect of ent-Sauchinone on anti-neuroinflammation and anti-amyloidogenesis in in vitro studies, we used microglial BV-2 cells and cultured astrocytes treated with ent-Sauchinone (1, 5, and 10 μM) for 24 hours. For the detection of anti-neuro-inflammatory responses, reative oxygen species (ROS) and Nitric oxide (NO) generation and inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) expression were measured with assay kits and western blotting. β-secretase and β-secretase activities and β-amyloid levels were determined for measuring the anti-amyloidogenic effects of ent-Sauchinone by enzyme assay kits. NF-κB and STAT3 signals were detected with electromobility shift assay (EMSA) to study the related signaling pathways. The binding of ent-Sauchinone to STAT3 was evaluated by a pull-down assay and by a docking model using Autodock VINA software (Hoover's Inc., Texas, United states).Results: ent-Sauchinone (1, 5, and 10 μM) effectively decreased lipopolysaccharide (LPS)-(1 μg/ml) induced inflammatory responses through the reduction of ROS and NO generations and iNOS and COX-2 expressions in cultured astrocytes and microglial BV-2 cells. ent-Sauchinone also inhibited LPS-induced amyloidogenesis through the inhibition of β-secretase and β-secretase activity. NF- κB amyloid and STAT3, critical transcriptional factors regulating not only inflammation but also amyloidogenesis, were also inhibited in a concentration dependent manner by ent-Sauchinone by blocking the phosphorylation of I κB and STAT3 in cultured astrocytes and microglial BV-2 cells. The docking model approach showed that ent-Sauchinone binds to STAT3, and the employment of a STAT3 inhibitor and siRNA reversed ent-Sauchinone-induced inhibition NF-κB activation and Aβ generation.Conclusions: These results indicated that ent-Sauchinone inhibited neuroinflammation and amyloidogenesis through the inhibition of STAT3-mediated NF-κB activity, and thus could be applied in the treatment of neuro-inflammatory diseases, including Alzheimer's disease. [ABSTRACT FROM AUTHOR]- Published
- 2014
- Full Text
- View/download PDF
11. Snake venom toxin from Vipera lebetina turanica induces apoptosis of colon cancer cells via upregulation of ROS- and JNK-mediated death receptor expression.
- Author
-
Park, Mi Hee, Jo, Miran, Won, Dohee, Song, Ho Sueb, Han, Sang Bae, Song, Min Jong, and Hong, Jin Tae
- Abstract
Background: Abundant research suggested that the cancer cells avoid destruction by the immune system through down-regulation or mutation of death receptors. Therefore, it is very important that finding the agents that increase the death receptors of cancer cells. In this study, we demonstrated that the snake venom toxin from Vipera lebetina turanica induce the apoptosis of colon cancer cells through reactive oxygen species (ROS) and c-Jun N-terminal kinases (JNK) dependent death receptor (DR4 and DR5) expression.Methods: We used cell viability assays, DAPI/TUNEL assays, as well as western blot for detection of apoptosis related proteins and DRs to demonstrate that snake venom toxin-induced apoptosis is DR4 and DR5 dependent. We carried out transient siRNA knockdowns of DR4 and DR5 in colon cancer cells.Results: We showed that snake venom toxin inhibited growth of colon cancer cells through induction of apoptosis. We also showed that the expression of DR4 and DR5 was increased by treatment of snake venom toxin. Moreover, knockdown of DR4 or DR5 reversed the effect of snake venom toxin. Snake venom toxin also induced JNK phosphorylation and ROS generation, however, pretreatment of JNK inhibitor and ROS scavenger reversed the inhibitory effect of snake venom toxin on cancer cell proliferation, and reduced the snake venom toxin-induced upregulation of DR4 and DR5 expression.Conclusions: Our results indicated that snake venom toxin could inhibit human colon cancer cell growth, and these effects may be related to ROS and JNK mediated activation of death receptor (DR4 and DR5) signals. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
12. Inhibitory effect of a tyrosine-fructose Maillard reaction product, 2,4-bis(p-hydroxyphenyl)-2-butenal on amyloid-β generation and inflammatory reactions via inhibition of NF-κB and STAT3 activation in cultured astrocytes and microglial BV-2 cells.
- Author
-
Lee, Young-Jung, Choi, Dong-Young, Choi, Im Seup, Han, Jin-Yi, Jeong, Heon-Sang, Han, Sang Bae, Oh, Ki-Wan, and Hong, Jin Tae
- Abstract
Background: Amyloidogenesis is linked to neuroinflammation. The tyrosine-fructose Maillard reaction product, 2,4-bis(p-hydroxyphenyl)-2-butenal, possesses anti-inflammatory properties in cultured macrophages, and in an arthritis animal model. Because astrocytes and microglia are responsible for amyloidogenesis and inflammatory reactions in the brain, we investigated the anti-inflammatory and anti-amyloidogenic effects of 2,4-bis(p-hydroxyphenyl)-2-butenal in lipopolysaccharide (LPS)-stimulated astrocytes and microglial BV-2 cells.Methods: Cultured astrocytes and microglial BV-2 cells were treated with LPS (1 μg/ml) for 24 h, in the presence (1, 2, 5 μM) or absence of 2,4-bis(p-hydroxyphenyl)-2-butenal, and harvested. We performed molecular biological analyses to determine the levels of inflammatory and amyloid-related proteins and molecules, cytokines, Aβ, and secretases activity. Nuclear factor-kappa B (NF-κB) DNA binding activity was determined using gel mobility shift assays.Results: We found that 2,4-bis(p-hydroxyphenyl)-2-butenal (1, 2, 5 μM) suppresses the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) as well as the production of nitric oxide (NO), reactive oxygen species (ROS), tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β) in LPS (1 μg/ml)-stimulated astrocytes and microglial BV-2 cells. Further, 2,4-bis(p-hydroxyphenyl)-2-butenal inhibited the transcriptional and DNA binding activity of NF-κB--a transcription factor that regulates genes involved in neuroinflammation and amyloidogenesis via inhibition of IκB degradation as well as nuclear translocation of p50 and p65. Consistent with the inhibitory effect on inflammatory reactions, 2,4-bis(p-hydroxyphenyl)-2-butenal inhibited LPS-elevated Aβ42 levels through attenuation of β- and γ-secretase activities. Moreover, studies using signal transducer and activator of transcription 3 (STAT3) siRNA and a pharmacological inhibitor showed that 2,4-bis(p-hydroxyphenyl)-2-butenal inhibits LPS-induced activation of STAT3.Conclusions: These results indicate that 2,4-bis(p-hydroxyphenyl)-2-butenal inhibits neuroinflammatory reactions and amyloidogenesis through inhibition of NF-κB and STAT3 activation, and suggest that 2,4-bis(p-hydroxyphenyl)-2-butenal may be useful for the treatment of neuroinflammatory diseases like Alzheimer's disease. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
13. Antiviral activity of micafungin against enterovirus 71.
- Author
-
Kim C, Kang H, Kim DE, Song JH, Choi M, Kang M, Lee K, Kim HS, Shin JS, Jeong H, Jung S, Han SB, Kim JH, Ko HJ, Lee CK, Kim M, and Cho S
- Subjects
- Animals, Cell Line, Drug Repositioning, Humans, Micafungin, Antiviral Agents pharmacology, Echinocandins pharmacology, Enterovirus A, Human drug effects, Lipopeptides pharmacology
- Abstract
Background: Enterovirus 71 (EV71) is a major causative agent of hand-foot-mouth disease (HFMD) and also causes severe neurological complications, leading to fatality in young children. However, no effective therapy is currently available for the treatment of this infection., Methods: We identified small-molecule inhibitors of EV71 from a screen of 968 Food and Drug Administration (FDA)-approved drugs, with which clinical application for EV71-associated diseases would be more feasible, using EV71 subgenomic replicon system. Primary hits were extensively evaluated for their antiviral activities in EV71-infected cells., Results: We identified micafungin, an echinocandin antifungal drug, as a novel inhibitor of EV71. Micafungin potently inhibits the proliferation of EV71 as well as the replication of EV71 replicon in cells with a low micromolar IC50 (~5 μM). The strong antiviral effect of micafungin on EV71 replicon and the result from time-of-addition experiment demonstrated a targeting of micafungin on virion-independent intracellular process(es) during EV71 infection. Moreover, an extensive analysis excluded the involvement of 2C and 3A proteins, IRES-dependent translation, and also that of polyprotein processing in the antiviral effect of micafungin., Conclusions: Our research revealed a new indication of micafungin as an effective inhibitor of EV71, which is the first case reporting antiviral activity of micafungin, an antifungal drug.
- Published
- 2016
- Full Text
- View/download PDF
14. Anticancer effect of tectochrysin in colon cancer cell via suppression of NF-kappaB activity and enhancement of death receptor expression.
- Author
-
Park MH, Hong JE, Park ES, Yoon HS, Seo DW, Hyun BK, Han SB, Ham YW, Hwang BY, and Hong JT
- Subjects
- Animals, Antineoplastic Agents chemistry, Apoptosis drug effects, Apoptosis Regulatory Proteins metabolism, Caspase 3 metabolism, Cell Proliferation drug effects, Drug Resistance, Neoplasm drug effects, Flavonoids chemistry, HCT116 Cells, Humans, Male, Mice, Inbred BALB C, Mice, Nude, RNA, Small Interfering metabolism, TNF-Related Apoptosis-Inducing Ligand pharmacology, Xenograft Model Antitumor Assays, fas Receptor metabolism, Antineoplastic Agents pharmacology, Colonic Neoplasms metabolism, Colonic Neoplasms pathology, Flavonoids pharmacology, NF-kappa B metabolism, Receptors, Death Domain metabolism
- Abstract
Background: Flavonoids are a diverse family of natural phenolic compounds commonly found in fruits and vegetables. Epidemiologic studies showed that flavonoids also reduce the risk of colon cancer. Tectochrysin is one of the major flavonoids of Alpinia oxyphylla Miquel. However, the anti-cancer effects and the molecular mechanisms of tectochrysin in colon cancer cells have not yet been reported. We investigated whether tectochrysin could inhibit colon cancer cell growth at 1, 5, 10 μg/ml. In in vivo study, we injected a tectochrysin treatment dose of 5 mg/kg to each mouse., Results: Tectochrysin suppressed the growth of SW480 and HCT116 human colon cancer cells. The expression of DR3, DR4 and Fas were significantly increased, and pro-apoptotic proteins were also increased. Tectochrysin treatment also inhibited activity of NF-κB. A docking model indicated that tectochrysin binds directly to the p50 unit. In in vivo, tumor weights and volumes in mice were reduced when treated with tectochrysin. Tectochrysin leads to apoptotic cell death in colon cancer cells through activation of death receptors expression via the inhibition of NF-κB., Conclusions: Tectochrysin can be a useful agent for the treatment of colon cancer cell growth as well as an adjuvant agent for chemo-resistant cancer cells growth.
- Published
- 2015
- Full Text
- View/download PDF
15. Inhibitory effect of 4-O-methylhonokiol on lipopolysaccharide-induced neuroinflammation, amyloidogenesis and memory impairment via inhibition of nuclear factor-kappaB in vitro and in vivo models.
- Author
-
Lee YJ, Choi DY, Choi IS, Kim KH, Kim YH, Kim HM, Lee K, Cho WG, Jung JK, Han SB, Han JY, Nam SY, Yun YW, Jeong JH, Oh KW, and Hong JT
- Subjects
- Amyloid Precursor Protein Secretases metabolism, Analysis of Variance, Animals, Anti-Inflammatory Agents pharmacology, Aspartic Acid Endopeptidases metabolism, Astrocytes drug effects, Avoidance Learning drug effects, Biphenyl Compounds pharmacology, Brain drug effects, Brain metabolism, Cell Line, Transformed, Cyclooxygenase 2 metabolism, Cytokines metabolism, Dinoprostone metabolism, Disease Models, Animal, Electrophoretic Mobility Shift Assay, Glial Fibrillary Acidic Protein metabolism, In Situ Nick-End Labeling, Inflammation chemically induced, Lignans pharmacology, Lipopolysaccharides toxicity, Male, Maze Learning drug effects, Memory Disorders chemically induced, Memory Disorders pathology, Mice, Mice, Inbred ICR, Microglia drug effects, Nitric Oxide metabolism, Peptide Fragments metabolism, Amyloid beta-Peptides metabolism, Anti-Inflammatory Agents therapeutic use, Biphenyl Compounds therapeutic use, Inflammation drug therapy, Lignans therapeutic use, Memory Disorders drug therapy, NF-kappa B metabolism
- Abstract
Background: Neuroinflammation is important in the pathogenesis and progression of Alzheimer disease (AD). Previously, we demonstrated that lipopolysaccharide (LPS)-induced neuroinflammation caused memory impairments. In the present study, we investigated the possible preventive effects of 4-O-methylhonokiol, a constituent of Magnolia officinalis, on memory deficiency caused by LPS, along with the underlying mechanisms., Methods: We investigated whether 4-O-methylhonokiol (0.5 and 1 mg/kg in 0.05% ethanol) prevents memory dysfunction and amyloidogenesis on AD model mice by intraperitoneal LPS (250 μg/kg daily 7 times) injection. In addition, LPS-treated cultured astrocytes and microglial BV-2 cells were investigated for anti-neuroinflammatory and anti-amyloidogenic effect of 4-O-methylhonkiol (0.5, 1 and 2 μM)., Results: Oral administration of 4-O-methylhonokiol ameliorated LPS-induced memory impairment in a dose-dependent manner. In addition, 4-O-methylhonokiol prevented the LPS-induced expression of inflammatory proteins; inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) as well as activation of astrocytes (expression of glial fibrillary acidic protein; GFAP) in the brain. In in vitro study, we also found that 4-O-methylhonokiol suppressed the expression of iNOS and COX-2 as well as the production of reactive oxygen species, nitric oxide, prostaglandin E2, tumor necrosis factor-α, and interleukin-1β in the LPS-stimulated cultured astrocytes. 4-O-methylhonokiol also inhibited transcriptional and DNA binding activity of NF-κB via inhibition of IκB degradation as well as p50 and p65 translocation into nucleus of the brain and cultured astrocytes. Consistent with the inhibitory effect on neuroinflammation, 4-O-methylhonokiol inhibited LPS-induced Aβ1-42 generation, β- and γ-secretase activities, and expression of amyloid precursor protein (APP), BACE1 and C99 as well as activation of astrocytes and neuronal cell death in the brain, in cultured astrocytes and in microglial BV-2 cells., Conclusion: These results suggest that 4-O-methylhonokiol inhibits LPS-induced amyloidogenesis via anti-inflammatory mechanisms. Thus, 4-O-methylhonokiol can be a useful agent against neuroinflammation-associated development or the progression of AD.
- Published
- 2012
- Full Text
- View/download PDF
16. Anti-inflammatory and arthritic effects of thiacremonone, a novel sulfur compound isolated from garlic via inhibition of NF-kappaB.
- Author
-
Ban JO, Oh JH, Kim TM, Kim DJ, Jeong HS, Han SB, and Hong JT
- Subjects
- Animals, Blotting, Western, Electrophoretic Mobility Shift Assay, Garlic chemistry, Humans, Mice, NF-kappa B metabolism, Nitric Oxide metabolism, Nitric Oxide Synthase Type II drug effects, Nitric Oxide Synthase Type II metabolism, Plant Extracts pharmacology, Pyridines toxicity, Rats, Rats, Sprague-Dawley, Transfection, Anti-Inflammatory Agents pharmacology, Arthritis, Experimental drug therapy, Inflammation drug therapy, NF-kappa B drug effects, Phytotherapy, Thiophenes pharmacology
- Abstract
Introduction: Sulfur compounds isolated from garlic exert anti-inflammatory properties. We recently isolated thiacremonone, a novel sulfur compound from garlic. Here, we investigated the anti-inflammatory and arthritis properties of thiacremonone through inhibition of NF-kappaB since NF-kappaB is known to be a target molecule of sulfur compounds and an implicated transcription factor regulating inflammatory response genes., Methods: The anti-inflammatory and arthritis effects of thiacremone in in vivo were investigated in 12-O-tetradecanoylphorbol-13-acetate-induced ear edema, carrageenan and mycobacterium butyricum-induced inflammatory and arthritis models. Lipopolysaccharide-induced nitric oxide (NO) production was determined by Griess method. The DNA binding activity of NF-kappaB was investigated by electrophoretic mobility shift assay. NF-kappaB and inducible nitric oxide synthetase (iNOS) transcriptional activity was determined by luciferase assay. Expression of iNOS and cyclooxygenase-2 (COX-2) was determined by western blot., Results: The results showed that topical application of thiacremonone (1 or 2 microg/ear) suppressed the 12-O-tetradecanoylphorbol-13-acetate-induced (1 microg/ear) ear edema. Thiacremonone (1-10 mg/kg) administered directly into the plantar surface of hind paw also suppressed the carrageenan (1.5 mg/paw) and mycobacterium butyricum (2 mg/paw)-induced inflammatory and arthritic responses as well as expression of iNOS and COX-2, in addition to NF-kappaB DNA-binding activity. In further in vitro study, thiacremonone (2.5-10 microg/ml) inhibited lipopolysaccharide (LPS, 1 microg/ml)-induced nitric oxide (NO) production, and NF-kappaB transcriptional and DNA binding activity in a dose dependent manner. The inhibition of NO by thiacremonone was consistent with the inhibitory effect on LPS-induced inducible nitric oxide synthase (iNOS) and COX-2 expression, as well as iNOS transcriptional activity. Moreover, thiacremonone inhibited LPS-induced p50 and p65 nuclear translocation, resulting in an inhibition of the DNA binding activity of the NF-kappaB. These inhibitory effects on NF-kappaB activity and NO generation were suppressed by reducing agents dithiothreitol (DTT) and glutathione, and were abrogated in p50 (C62S)-mutant cells, suggesting that the sulfhydryl group of NF-kappaB molecules may be a target of thiacremonone., Conclusions: The present results suggested that thiacremonone exerted its anti-inflammatory and anti-arthritic properties through the inhibition of NF-kappaB activation via interaction with the sulfhydryl group of NF-kappaB molecules, and thus could be a useful agent for the treatment of inflammatory and arthritic diseases.
- Published
- 2009
- Full Text
- View/download PDF
17. JNK pathway is involved in the inhibition of inflammatory target gene expression and NF-kappaB activation by melittin.
- Author
-
Park HJ, Lee HJ, Choi MS, Son DJ, Song HS, Song MJ, Lee JM, Han SB, Kim Y, and Hong JT
- Abstract
Background: Bee venom therapy has been used to treat inflammatory diseases including rheumatoid arthritis in humans and in experimental animals. We previously found that bee venom and melittin (a major component of bee venom) have anti-inflammatory effect by reacting with the sulfhydryl group of p50 of nuclear factor-kappa B (NF-kappaB) and IkappaB kinases (IKKs). Since mitogen activated protein (MAP) kinase family is implicated in the NF-kappaB activation and inflammatory reaction, we further investigated whether activation of MAP kinase may be also involved in the anti-inflammatory effect of melittin and bee venom., Methods: The anti-inflammatory effects of melittin and bee venom were investigated in cultured Raw 264.7 cells, THP-1 human monocytic cells and Synoviocytes. The activation of NF-kappaB was investigated by electrophoretic mobility shift assay. Nitric oxide (NO) and prostaglandin E2 (PGE2) were determined either by Enzyme Linked Immuno Sorbent Assay or by biochemical assay. Expression of IkappaB, p50, p65, inducible nitric oxide synthetase (iNOS), cyclooxygenase-2 (COX-2) as well as phosphorylation of MAP kinase family was determined by Western blot., Results: Melittin (0.5-5 mug/ml) and bee venom (5 and 10 mug/ml) inhibited lipopolysaccharide (LPS, 1 mug/ml) and sodium nitroprusside (SNP, 200 muM)-induced activation of c-Jun NH2-terminal kinase (JNK) in RAW 264.7 cells in a dose dependent manner. However, JNK inhibitor, anthra [1,9-cd]pyrazole-6 (2H)-one (SP600215, 10-50 muM) dose dependently suppressed the inhibitory effects of melittin and bee venom on NF-kappaB dependent luciferase and DNA binding activity via suppression of the inhibitory effect of melittin and bee venom on the LPS and SNP-induced translocation of p65 and p50 into nucleus as well as cytosolic release of IkappaB. Moreover, JNK inhibitor suppressed the inhibitory effects of melittin and bee venom on iNOS and COX-2 expression, and on NO and PGE2 generation., Conclusion: These data show that melittin and bee venom prevent LPS and SNP-induced NO and PGE2 production via JNK pathway dependent inactivation of NF-kappaB, and suggest that inactivation of JNK pathways may also contribute to the anti-inflammatory and anti-arthritis effects of melittin and bee venom.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.