1
|
Duan Q, Yu B, Huang Z, Luo Y, Zheng P, Mao X, Yu J, Luo J, Yan H, He J. Sialyllactose Attenuates Inflammation and Injury of Intestinal Epithelial Cells upon Enterotoxigenic Escherichia coli Infection. Int J Mol Sci 2025; 26:3860. [PMID: 40332525 PMCID: PMC12027521 DOI: 10.3390/ijms26083860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Sialyllactose (SL), a bioactive trisaccharide abundant in porcine colostrum, demonstrates multifunctional properties including antimicrobial activity, immune regulation, and apoptosis inhibition. This research uncovers the mechanisms by which SL mitigates enterotoxigenic Escherichia coli (ETEC)-mediated damage to intestinal barrier integrity, employing IPEC-J2 porcine epithelial models. SL pre-treatment effectively blocked pathogen adhesion by competitively binding to cellular receptors, concurrently mitigating inflammation through significant suppression of TNF-α, IL-1β, and IL-6 expression (p < 0.05). Notably, SL exhibited functional parallels to the NF-κB inhibitor BAY11-7082, jointly enhancing tight junction integrity via ZO-1 protein stabilization and inhibiting pro-inflammatory signaling through coordinated suppression of IκB-α/NF-κB phosphorylation cascades. The dual-action mechanism combines molecular interception of microbial attachment with intracellular modulation of the TLR4/MyD88/NF-κB pathway, effectively resolving both pathogenic colonization and inflammatory amplification. These findings position SL as a potential therapeutic application nutraceutical for livestock, with the capacity to address post-weaning porcine enteritis through functional feed formulations that synergistically enhance intestinal barrier resilience while curbing ETEC-mediated inflammatory pathogenesis.
Collapse
Affiliation(s)
- Qiming Duan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Zhiqing Huang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| |
Collapse
|
2
|
Lunić T, Petković M, Rakić M, Lađarević J, Repac J, Nedeljković BB, Božić B. Anti-neuroinflammatory potential of hydroxybenzoic ester derivatives: In silico insight and in vitro validation. J Mol Struct 2025; 1321:139804. [DOI: 10.1016/j.molstruc.2024.139804] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
3
|
Kim JH, Lee JH, Nan Z, Choi JW, Song JW. Di(2-ethylhexyl) phthalate exposure aggravates hypoxia/reoxygenation injury in cerebral endothelial cells by downregulating epithelial cadherin expression. Toxicol Res (Camb) 2024; 13:tfae163. [PMID: 39371678 PMCID: PMC11447374 DOI: 10.1093/toxres/tfae163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/06/2024] [Accepted: 09/26/2024] [Indexed: 10/08/2024] Open
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is a widely used plasticizer that has adverse health effects. Most phthalates exhibit reproductive toxicity and are associated with diseases such as cardiovascular disorders. However, the effect of DEHP exposure on acute hypoxia/reperfusion injury remains unknown. Therefore, we assessed whether hypoxia/reperfusion injury is aggravated by exposure to DEHP and investigated plausible underlying mechanisms, including oxidative stress and expression of cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2) and endothelial junctional proteins. bEnd.3 cells were exposed to DEHP and subsequently subjected to oxygen-glucose deprivation (OGD). Cell viability was analyzed using 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) proliferation assay. The effect of DEHP/OGD/reoxygenation (R) was evaluated by assessing the levels of NO, reactive oxygen species (ROS), and PGE2. The expression of COX-2, cleaved caspase-3, cleaved PARP, inducible nitric oxide synthase (iNOS), and the endothelial tight junction proteins claudin-5 and ZO-1 was evaluated using quantitative polymerase chain reaction and western blotting. OGD/R decreased cell viability, and DEHP exposure before OGD/R further aggravated cell viability. DEHP/OGD/R significantly increased NO, PGE2, and ROS production following OGD/R. In the DEHP/OGD/R group, iNOS, COX-2, cleaved caspase-3, and cleaved PARP expression increased, and claudin-5 and ZO-1 levels decreased compared with those in the OGD/R group. E-Cadherin expression decreased significantly after DEHP/OGD/R exposure compared with that after OGD/R; this decrease in expression was recovered by treatment with the COX-2 inhibitor indomethacin and antioxidant N-acetylcysteine. Exposure to DEHP exacerbated hypoxia-reoxygenation injury. The enhanced damage upon DEHP exposure was associated with increased oxidative stress and COX-2 expression, leading to E-cadherin downregulation and increased apoptosis.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jae Hoon Lee
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Zhengyu Nan
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ja Woo Choi
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jong Wook Song
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
4
|
Shi T, Sun X, Yuan Q, Wang J, Shen X. Exploring the role of flavin-dependent monooxygenases in the biosynthesis of aromatic compounds. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2024; 17:46. [PMID: 38520003 PMCID: PMC10958861 DOI: 10.1186/s13068-024-02490-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 03/13/2024] [Indexed: 03/25/2024]
Abstract
Hydroxylated aromatic compounds exhibit exceptional biological activities. In the biosynthesis of these compounds, three types of hydroxylases are commonly employed: cytochrome P450 (CYP450), pterin-dependent monooxygenase (PDM), and flavin-dependent monooxygenase (FDM). Among these, FDM is a preferred choice due to its small molecular weight, stable expression in both prokaryotic and eukaryotic fermentation systems, and a relatively high concentration of necessary cofactors. However, the catalytic efficiency of many FDMs falls short of meeting the demands of large-scale production. Additionally, challenges arise from the limited availability of cofactors and compatibility issues among enzyme components. Recently, significant progress has been achieved in improving its catalytic efficiency, but have not yet detailed and informative viewed so far. Therefore, this review emphasizes the advancements in FDMs for the biosynthesis of hydroxylated aromatic compounds and presents a summary of three strategies aimed at enhancing their catalytic efficiency: (a) Developing efficient enzyme mutants through protein engineering; (b) enhancing the supply and rapid circulation of critical cofactors; (c) facilitating cofactors delivery for enhancing FDMs catalytic efficiency. Furthermore, the current challenges and further perspectives on improving catalytic efficiency of FDMs are also discussed.
Collapse
Affiliation(s)
- Tong Shi
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, 15 Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Xinxiao Sun
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, 15 Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Qipeng Yuan
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, 15 Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Jia Wang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, 15 Beisanhuan East Road, Chaoyang District, Beijing, 100029, China.
| | - Xiaolin Shen
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, 15 Beisanhuan East Road, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
5
|
Kim JH, Lee JH. Effect of miR-412-5p-loaded exosomes in H9c2 cardiomyocytes via the MAPK pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:755-760. [PMID: 38645496 PMCID: PMC11024402 DOI: 10.22038/ijbms.2024.75590.16365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/15/2024] [Indexed: 04/23/2024]
Abstract
Objectives MicroRNAs (miRNAs) are small non-coding RNAs that function in all biological processes. Recent findings suggest that exosomes, which are small vesicles abundantly secreted by various cell types, can transport miRNAs to target cells. Here, we elucidated the effect of miRNA-loaded exosomes on lipopolysaccharide (LPS)-induced inflammation in H9c2 cardiomyocytes. Materials and Methods Exosomes were isolated from mesenchymal stem cells (MSC) and loaded with miR-412-5p. Additionally, the effect of the miR-412-5p-loaded exosomes on LPS-induced inflammation in H9c2 cardiomyocytes was evaluated by assessing the levels of nitric oxide (NO), reactive oxygen species (ROS), and prostaglandin E2 (PGE2). The expression of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), inflammatory cytokines, and mitogen-activated protein kinase (MAPK) signaling factors was evaluated using reverse transcription-quantitative PCR and western blotting. Results miR-412-5p-loaded exosomes inhibited LPS-induced secretion of inflammatory mediators (NO, PGE2, and ROS), pro-inflammatory cytokines (IL-1β and IL-6), and COX-2 and iNOS expression. Additionally, miR-412-5p-loaded exosomes significantly decreased the expression of MAPK signaling molecules, including p-extracellular signal-regulated kinase (ERK), p-p38, and p-Jun kinase (JNK), in H9c2 cardiomyocytes. Conclusion These findings showed that miR-412-5p-loaded exosomes ameliorated LPS-induced inflammation in H9c2 cardiomyocytes by inhibiting COX-2 and iNOS expression, inflammatory mediators, and pro-inflammatory cytokines via the MAPK pathway. The findings indicate that miR-412-5p-loaded exosomes may be effective for the prevention of myocardial injury.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Anesthesiology and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - June Hwan Lee
- Department of Energy Information Technology, Fareast University, 76-32, Daehak-gil, Gamgok-myeon, Eumseong-gun, Chungcheongbuk-do 27601, Republic of Korea
| |
Collapse
|
6
|
Park JY, Yasir M, Lee HJ, Han ET, Han JH, Park WS, Kwon YS, Chun W. Caffeic acid methyl ester inhibits LPS‑induced inflammatory response through Nrf2 activation and NF‑κB inhibition in human umbilical vein endothelial cells. Exp Ther Med 2023; 26:559. [PMID: 37941589 PMCID: PMC10628649 DOI: 10.3892/etm.2023.12257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/27/2023] [Indexed: 11/10/2023] Open
Abstract
Caffeic acid (CA) derivatives have been reported to exert anti-inflammatory activities in various inflammatory conditions. However, the impact of CA methyl ester (CAME) on the inflammatory response in vascular endothelial cells has not been thoroughly elucidated. In the present study, the aim was to understand how CAME can reduce inflammation in human umbilical vein endothelial cells (HUVECs), which were challenged with lipopolysaccharide (LPS), and elucidate its mechanisms. CAME significantly attenuated LPS-induced TNF-α and IL-1β release. Furthermore, CAME inhibited cyclooxygenase 2 expression and consequent secretion of prostaglandin E2. CAME also suppressed LPS-stimulated inducible nitric oxide synthase expression. In addition, CAME significantly enhanced the expression of heme oxygenase-1 (HO-1) and nuclear factor erythroid-derived 2-related factor 2 (Nrf2) phosphorylation in the absence or presence of LPS stimulation in HUVECs. CAME also significantly suppressed LPS-induced NF-κB phosphorylation and inhibitor of κB phosphorylation and degradation. In conclusion, the present results provide clear evidence that CAME exerts its anti-inflammatory activities by increasing HO-1/Nrf2-mediated cytoprotection and inhibiting NF-κB-mediated pro-inflammatory pathways in HUVECs.
Collapse
Affiliation(s)
- Jin-Young Park
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, Gangwon 24341, Republic of Korea
| | - Muhammad Yasir
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, Gangwon 24341, Republic of Korea
| | - Hee Jae Lee
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, Gangwon 24341, Republic of Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, Gangwon 24341, Republic of Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, Gangwon 24341, Republic of Korea
| | - Yong-Soo Kwon
- College of Pharmacy, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, Gangwon 24341, Republic of Korea
| |
Collapse
|
7
|
Niu W, Chen Y, Wang L, Li J, Cui Z, Lv J, Yang F, Huo J, Zhang Z, Ju J. The combination of sodium alginate and chlorogenic acid enhances the therapeutic effect on ulcerative colitis by the regulation of inflammation and the intestinal flora. Food Funct 2022; 13:10710-10723. [PMID: 36173280 DOI: 10.1039/d2fo01619b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Chlorogenic acid (CA) and sodium alginate (SA) each have good therapeutic effects on ulcerative colitis (UC) owing to their antioxidant and anti-inflammatory activity. This study aimed to investigate the effects of CA alone and in combination with SA on inflammatory cells and UC mice. In the Lipopolysaccharide (LPS)-induced RAW 264.7 inflammatory cell model, Nitric oxide (NO) and interleukin-6 (IL-6) levels were significantly lower after treatment with CA plus SA than with CA alone. In the DSS-induced UC mouse model, compared with CA alone, CA plus SA showed a better ability to alleviate weight loss, reduce the disease activity index (DAI), improve the colonic mucosa, reduce the expression of inflammatory factors in the serum and myeloperoxidase (MPO) in colonic tissue, increase superoxide dismutase (SOD) levels, protect the intestinal mucosa and regulate the abundance of Actinobacteriota, Lactobacillus, Bifidobacterium, Bacteroides, Subdoligranulum and Streptococcus. Thus, CA plus SA can improve the therapeutic efficacy of CA in UC by regulating inflammatory factors, oxidative stress, and the intestinal flora and by protecting ulcerative wounds. These findings broaden our understanding of the role of the combination of SA and CA in enhancing the effects of CA on UC and provide strategies for prevention and treatment.
Collapse
Affiliation(s)
- Wei Niu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Yuxuan Chen
- School of Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ligui Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Jia Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Zhao Cui
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Jiajie Lv
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Fuyan Yang
- Anhui University of Chinese Medicine, Hefei, China
| | - Jiege Huo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Zhenhai Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Jianming Ju
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| |
Collapse
|
8
|
Chen JS, Wang MX, Wang MM, Zhang YK, Guo X, Chen YY, Zhang MQ, Sun JY, Liu YF, Liu C. Synthesis and biological evaluation of geniposide derivatives as inhibitors of hyperuricemia, inflammatory and fibrosis. Eur J Med Chem 2022; 237:114379. [DOI: 10.1016/j.ejmech.2022.114379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/06/2022] [Accepted: 04/09/2022] [Indexed: 01/16/2023]
|
9
|
Chansiw N, Champakam S, Chusri P, Pangjit K, Srichairatanakool S. Quercetin-Rich Ethanolic Extract of Polygonum odoratum var Pakphai Leaves Decreased Gene Expression and Secretion of Pro-Inflammatory Mediators in Lipopolysaccharide-Induced Murine RAW264.7 Macrophages. Molecules 2022; 27:molecules27123657. [PMID: 35744785 PMCID: PMC9227601 DOI: 10.3390/molecules27123657] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/22/2022] [Accepted: 06/01/2022] [Indexed: 11/16/2022] Open
Abstract
Polygonum odoratum var. Pakphai has been used in traditional Thai medicine for the treatment of flatulence and constipation and to relieve the inflammation caused by insect bites. Quercetin (Q), which is abundant in plant-based foods, has been found to exert anti-inflammatory properties. This study evaluated the anti-inflammatory activity of P. odoratum ethanolic extract in RAW264.7 macrophage cells. Leaves were extracted with 50% ethanol, phenolics and flavonoids were then analyzed using UHPLC-QTOF-MS and HPLC-DAD. RAW264.7 cells were induced with lipopolysaccharides (LPSs). They were then treated with the extract and prostaglandin E2 (PGE2), and interleukin-6 (IL-6) and tumor necrotic factor-alpha (TNF-α) concentrations were determined. Levels of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), IL-6 and TNF-α mRNAs were analyzed using qRT-PCR. Chemical analysis demonstrated that the extract was abundant with Q while also containing catechin, gallic acid, epicatechin gallate and coumarin. The extract increased the viability of RAW264.7 cells and dose-dependently decreased nitric oxide production, PGE2, IL-6 and TNF-α levels in the medium from the LPS-induced RAW264.7 cell culture. Consistently, COX-2, iNOS, IL-6 and TNF-α mRNA levels were decreased in a concentration-dependent manner (p < 0.05). Thus, the quercetin-rich ethanolic extract derived from P. odoratum var Pakphai leaves can exert anti-inflammatory activity in LPS-induced RAW264.7 cells through a reduction of the pro-inflammatory mediator response.
Collapse
Affiliation(s)
- Nittaya Chansiw
- School of Medicine, Mae Fah Luang University, Chiang Rai 57100, Thailand; (N.C.); (P.C.)
| | - Sorraya Champakam
- School of Integrative Medicine, Mae Fah Luang University, Chiang Rai 57100, Thailand;
| | - Pattranuch Chusri
- School of Medicine, Mae Fah Luang University, Chiang Rai 57100, Thailand; (N.C.); (P.C.)
| | - Kanjana Pangjit
- College of Medicine and Public Health, Ubon Ratchathani University, Ubon Ratchathani 34190, Thailand;
| | - Somdet Srichairatanakool
- Oxidative Stress Cluster, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence: ; Tel.: +66-53935322
| |
Collapse
|
10
|
Fu Q, Lin Q, Chen D, Yu B, Luo Y, Zheng P, Mao X, Huang Z, Yu J, Luo J, Yan H, He J. β-defensin 118 attenuates inflammation and injury of intestinal epithelial cells upon enterotoxigenic Escherichia coli challenge. BMC Vet Res 2022; 18:142. [PMID: 35440001 PMCID: PMC9017018 DOI: 10.1186/s12917-022-03242-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 04/11/2022] [Indexed: 11/30/2022] Open
Abstract
Background Antimicrobial peptides including various defensins have been attracting considerable research interest worldwide, as they have potential to substitute for antibiotics. Moreover, AMPs also have immunomodulatory activity. In this study, we explored the role and its potential mechanisms of β-defensin 118 (DEFB118) in alleviating inflammation and injury of IPEC-J2 cells (porcine jejunum epithelial cell line) upon the enterotoxigenic Escherichia coli (ETEC) challenge. Results The porcine jejunum epithelial cell line (IPEC-J2) pretreated with or without DEFB118 (25 μg/mL) were challenged by ETEC (1×106 CFU) or culture medium. We showed that DEFB118 pretreatment significantly increased the cell viability (P<0.05) and decreased the expressions of inflammatory cytokines such as the interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) in IPEC-J2 cells exposure to ETEC (P<0.05). Interestingly, DEFB118 pretreatment significantly elevated the abundance of the major tight-junction protein zonula occludens-1 (ZO-1), but decreased the number of apoptotic cells upon ETEC challenge (P<0.05). The expression of caspase 3, caspase 8, and caspase 9 were downregulated by DEFB118 in the IPEC-J2 cells exposure to ETEC (P<0.05). Importantly, DEFB118 suppressed two critical inflammation-associated signaling proteins, nuclear factor-kappa-B inhibitor alpha (IκB-α) and nuclear factor-kappaB (NF-κB) in the ETEC-challenged IPEC-J2 cells. Conclusions DEFB118 can alleviate ETEC-induced inflammation in IPEC-J2 cells through inhibition of the NF-κB signaling pathway, resulting in reduced secretion of inflammatory cytokines and decreased cell apoptosis. Therefore, DEFB118 can act as a novel anti-inflammatory agent.
Collapse
Affiliation(s)
- Qingqing Fu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province, 611130, P. R. China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province, 611130, P. R. China
| | - Qian Lin
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province, 611130, P. R. China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province, 611130, P. R. China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province, 611130, P. R. China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province, 611130, P. R. China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province, 611130, P. R. China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province, 611130, P. R. China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province, 611130, P. R. China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province, 611130, P. R. China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province, 611130, P. R. China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province, 611130, P. R. China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province, 611130, P. R. China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province, 611130, P. R. China
| | - Zhiqing Huang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province, 611130, P. R. China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province, 611130, P. R. China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province, 611130, P. R. China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province, 611130, P. R. China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province, 611130, P. R. China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province, 611130, P. R. China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province, 611130, P. R. China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province, 611130, P. R. China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province, 611130, P. R. China. .,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province, 611130, P. R. China.
| |
Collapse
|
11
|
Zhou M, Tang Y, Liao L, Liu M, Deng Y, Zhao X, Li Y. Phillygenin inhibited LPS-induced RAW 264.7 cell inflammation by NF-κB pathway. Eur J Pharmacol 2021; 899:174043. [PMID: 33745957 DOI: 10.1016/j.ejphar.2021.174043] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 02/06/2023]
Abstract
Inflammation is a common pathological phenomenon when homeostasis is seriously disturbed. Phillygenin (PHI), a lignin component isolated from Forsythiae Fructus, has shown a good anti-inflammatory effect. However, the mechanisms of PHI on anti-inflammation have not yet been systematically elucidated. In this study, the lipopolysaccharide (LPS) - induced RAW264.7 cell inflammation model was established to investigate mechanisms of PHI on inflammation. The effect of PHI on the release of IL-1β and PGE2 inflammatory factors induced by LPS was detected by ELISA, and the mRNA expressions of IL-1β, IL-6 and TNF-α were detected by RT-qPCR. Proteomics studied the signaling pathways that might be affected by PHI and molecular docking technology was subsequently used to study the possible targets on proteomic screened pathways. Western blot was performed ultimately to detect progressive changes in protein expression on the related pathway. Our research showed that PHI significantly inhibited the robust increase of IL-1β and PGE2 and lowered the transcriptional level of inflammatory genes including IL-6, IL-1β and PGE2 in LPS-stimulated RAW264.7 cells. Proteomics results indicated that PHI was involved in the regulation of multiple signaling pathways. Molecular docking results indicated that PHI had an affinity for most proteins in NF-κB pathway. Western blot analysis proved that PHI inhibited LPS-induced NF-κB pathway activation. On the whole, PHI inhibited the activation of NF-κB pathway, thereby inhibiting the expression of related inflammatory genes and the release of cytokines, and showed a remarkable anti-inflammatory effect.
Collapse
Affiliation(s)
- Mengting Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yunqiu Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Li Liao
- State Key Laboratory of Southwestern Chinese Medicine Resources; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Meichen Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ying Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xingtao Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
12
|
Min JH, Kim MG, Kim SM, Park JW, Chun W, Lee HJ, Oh SR, Ahn KS, Lee JW. 3,4,5-Trihydroxycinnamic acid exerts a protective effect on pulmonary inflammation in an experimental animal model of COPD. Int Immunopharmacol 2020; 85:106656. [PMID: 32504994 DOI: 10.1016/j.intimp.2020.106656] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/12/2020] [Accepted: 05/29/2020] [Indexed: 01/23/2023]
Abstract
3,4,5-Trihydroxycinnamic acid (THCA), a derivative of hydroxycinnamic acid, has been reported to exert anti-inflammatory and antioxidant activities. However, its anti-inflammatory effects in chronic obstructive pulmonary disease (COPD) have not yet been elucidated. Therefore, we explored the protective effects of THCA on pulmonary inflammation in an experimental COPD model elicited by cigarette smoke (CS) and lipopolysaccharide (LPS). Oral administration of THCA significantly inhibited the activity of elastase, the release of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and monocyte chemoattractant protein-1 (MCP-1), myeloperoxidase (MPO) and the numbers of neutrophils and macrophages in the bronchoalveolar lavage fluid (BALF) of experimental COPD mice. THCA also exerted inhibitory effects on the recruitment of inflammatory cells, the levels of PAS positive cells and cAMP-response-element-binding protein (CREB) activation, and the expression of phosphodiesterase 4 (PDE4) in the lungs of experimental COPD mice. In addition, THCA exerted a regulatory effect on the activation of p38, ERK and nuclear factor-κB (NF-κB) in the lungs of experimental COPD mice. THCA also significantly upregulated the expression of NAD(P)H dehydrogenase (quinone 1) 1 (NQO1) and the activation of nuclear factor erythroid-derived 2-related factor 2 (Nrf2) in the lungs of mice. Furthermore, THC restored the reduction of NAD-dependent protein deacetylase sirtuin-1 (SIRT1) in the lungs of experimental COPD mice. In phorbol myristate acetate (PMA)-stimulated A549 or H292 airway epithelial cells, pretreatment of THCA dose-dependently inhibited the generation of IL-6. THCA also led to increased NQO1 expression in H292 cells. Collectively, these protective effects of antioxidant THCA were notably excellent and are thought to be associated with the downregulation of MAPK (partial)/NF-κB signaling and upregulation of NQO1 and SIRT1 expression.
Collapse
Affiliation(s)
- Jae-Hong Min
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; College of Pharmacy, Chungbuk National University, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Min-Gu Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; College of Pharmacy, Chungbuk National University, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Seong-Man Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Ji-Won Park
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Wanjoo Chun
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Kangwon 24341, Republic of Korea
| | - Hee Jae Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Kangwon 24341, Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea.
| | - Jae-Won Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea.
| |
Collapse
|
13
|
McGarrity S. How 3,4,5-Trihydroxycinnamic Acid Targets Vascular Leakage: Potential Targeted Sepsis Treatment. J Vasc Res 2020; 57:311-312. [PMID: 32599582 DOI: 10.1159/000507629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 03/26/2020] [Indexed: 11/19/2022] Open
Affiliation(s)
- Sarah McGarrity
- Department of Medicine, Centre for Systems Biology, University of Iceland, Reykjavik, Iceland, .,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA,
| |
Collapse
|
14
|
Park JY, Lee HJ, Kwon YS, Chun W. 3,4,5-Trihydroxycinnamic Acid Inhibits LPS-Induced Inflammatory Response by Increasing SIRT1 Expression in Human Umbilical Vein Endothelial Cells. J Vasc Res 2020; 57:302-310. [PMID: 32564014 DOI: 10.1159/000507628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/19/2020] [Indexed: 11/19/2022] Open
Abstract
3,4,5-Trihydroxycinnamic acid (THC) has been demonstrated to exert anti-inflammatory activities in LPS-induced RAW264.7 murine macrophage cells and in LPS-induced septic mice. However, the effect of THC on the inflammatory response in vascular endothelial cells has not been clearly examined. The goal of the present study was to elucidate the anti-inflammatory properties of THC and its underlying mechanism in LPS-challenged human umbilical vein endothelial cells (HUVECs). THC significantly suppressed LPS-induced interleukin-1β production and intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 expression and significantly decreased LPS-induced nuclear factor-κB activation by attenuating p65 phosphorylation and inhibitor of kappa B degradation. To understand the underlying mechanism of the anti-inflammatory effect of THC, the involvement of the sirtuin 1 (SIRT1) signaling pathway was examined. THC resulted in increased expression of SIRT1 in LPS-challenged HUVECs. Among the downstream molecular targets of SIRT1, the level of LPS-induced acetylated p53 was significantly decreased by THC treatment, whereas no noticeable change was observed in the levels of forkhead box O3 and peroxisome proliferator activated receptor gamma coactivator 1 alpha. In conclusion, the results clearly demonstrate that THC possesses anti-inflammatory properties by increasing SIRT1 expression and subsequent suppression of p53 activation in LPS-challenged HUVECs.
Collapse
Affiliation(s)
- Jin-Young Park
- Department of Pharmacology, College of Medicine, Chuncheon, Republic of Korea
| | - Hee Jae Lee
- Department of Pharmacology, College of Medicine, Chuncheon, Republic of Korea
| | - Yong-Soo Kwon
- College of Pharmacy, Kangwon National University, Chuncheon, Republic of Korea
| | - Wanjoo Chun
- Department of Pharmacology, College of Medicine, Chuncheon, Republic of Korea,
| |
Collapse
|
15
|
Maenpuen S, Pongsupasa V, Pensook W, Anuwan P, Kraivisitkul N, Pinthong C, Phonbuppha J, Luanloet T, Wijma HJ, Fraaije MW, Lawan N, Chaiyen P, Wongnate T. Creating Flavin Reductase Variants with Thermostable and Solvent-Tolerant Properties by Rational-Design Engineering. Chembiochem 2020; 21:1481-1491. [PMID: 31886941 DOI: 10.1002/cbic.201900737] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Indexed: 02/06/2023]
Abstract
We have employed computational approaches-FireProt and FRESCO-to predict thermostable variants of the reductase component (C1 ) of (4-hydroxyphenyl)acetate 3-hydroxylase. With the additional aid of experimental results, two C1 variants, A166L and A58P, were identified as thermotolerant enzymes, with thermostability improvements of 2.6-5.6 °C and increased catalytic efficiency of 2- to 3.5-fold. After heat treatment at 45 °C, both of the thermostable C1 variants remain active and generate reduced flavin mononucleotide (FMNH- ) for reactions catalyzed by bacterial luciferase and by the monooxygenase C2 more efficiently than the wild type (WT). In addition to thermotolerance, the A166L and A58P variants also exhibited solvent tolerance. Molecular dynamics (MD) simulations (6 ns) at 300-500 K indicated that mutation of A166 to L and of A58 to P resulted in structural changes with increased stabilization of hydrophobic interactions, and thus in improved thermostability. Our findings demonstrated that improvements in the thermostability of C1 enzyme can lead to broad-spectrum uses of C1 as a redox biocatalyst for future industrial applications.
Collapse
Affiliation(s)
- Somchart Maenpuen
- Department of Biochemistry, Faculty of Science, Burapha University, 169 Long-Hard Bangsaen Road, Chonburi, 20131, Thailand
| | - Vinutsada Pongsupasa
- School of Biomolecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), 555 Moo 1 Payupnai, Wangchan, Rayong, 21210, Thailand
| | - Wiranee Pensook
- Department of Biochemistry, Faculty of Science, Burapha University, 169 Long-Hard Bangsaen Road, Chonburi, 20131, Thailand
| | - Piyanuch Anuwan
- School of Biomolecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), 555 Moo 1 Payupnai, Wangchan, Rayong, 21210, Thailand
| | | | - Chatchadaporn Pinthong
- Department of Chemistry, Faculty of Science, Srinakharinwirot University, 114 Sukhumvit 23 Road, Bangkok, 10110, Thailand
| | - Jittima Phonbuppha
- School of Biomolecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), 555 Moo 1 Payupnai, Wangchan, Rayong, 21210, Thailand
| | - Thikumporn Luanloet
- Center for Excellence in Protein and Enzyme Technology, Faculty of Science, Mahidol University, 272 Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Hein J Wijma
- Molecular Enzymology Group, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 4, 9747 AG, Groningen, The Netherlands
| | - Marco W Fraaije
- Molecular Enzymology Group, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 4, 9747 AG, Groningen, The Netherlands
| | - Narin Lawan
- Department of Chemistry, Faculty of Science, Chiang Mai University, 239 Huaykaew Road, Suthep, Chiang Mai, 50200, Thailand
| | - Pimchai Chaiyen
- School of Biomolecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), 555 Moo 1 Payupnai, Wangchan, Rayong, 21210, Thailand.,Center for Excellence in Protein and Enzyme Technology, Faculty of Science, Mahidol University, 272 Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Thanyaporn Wongnate
- School of Biomolecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), 555 Moo 1 Payupnai, Wangchan, Rayong, 21210, Thailand
| |
Collapse
|
16
|
Zhang Y, Huang X, Chen H, Zhou D, Yang Z, Wang K, Liu W, Deng S, Yang R, Li J, He R. Discovery of anti-inflammatory terpenoids from Mallotus conspurcatus croizat. JOURNAL OF ETHNOPHARMACOLOGY 2019; 231:170-178. [PMID: 30445108 DOI: 10.1016/j.jep.2018.11.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/16/2018] [Accepted: 11/01/2018] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Mallotus conspurcatus croizat (Euphorbiaceae), a plant native to Jinxiu in Guangxi, is popularly used in folk medicine to treat pelvic inflammatory disease. The anti-inflammatory activities of the compounds obtained from M. conspurcatus root were evaluated in this study. AIM OF THE STUDY This study explored the major anti-inflammatory components of this plant. MATERIALS AND METHODS The ethyl acetate fraction of the ethanol extract from M. conspurcatus was separated using chromatographic techniques. The structures of the isolates were elucidated from NMR, MS and X-ray data as well as from ECD. The anti-inflammatory activities of the isolates from M. conspurcatus were evaluated using LPS-stimulated RAW 264.7 cell models. The production of NO, TNF-α and PGE-2 was determined by ELISA and Griess tests. The expression levels of COX-2, NF-κB/p65 and iNOS were measured by western blotting. RESULTS Two new diterpenoids, malloconspur A (1) and malloconspur B (2), and sixteen known terpenoids (3-18) were identified by comprehensive spectroscopic analyses and comparison with literature data. Malloconspur B (2) and 17-hydroxycleistantha-12,15-dien-3-one (3) substantially inhibited the release of NO with IC50 values of 10.47 μM and 9.32 μM, respectively. Compounds 1, 2 and 3 markedly decreased the secretion of PGE2 and TNF-α (P < 0.01) by LPS-induced RAW264.7 cells. Compounds 2 and 3 markedly decreased iNOS, NF-κB/p65 and COX-2 protein expression. CONCLUSIONS Our identification of these diterpenoids provides strong evidence for the use of M. conspurcatus among the Yao people as a medicinal plant for the treatment of inflammation. The dramatic differences in the chemical structures of the active diterpenoids of this plant from those on the market suggest these compounds have potential as anti-inflammatory lead compounds for follow-up research.
Collapse
Affiliation(s)
- Yanjun Zhang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemical & Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China; School of Petroleum and Chemical Engineering, Qinzhou University, Qinzhou 535000, China
| | - Xishan Huang
- School of Chemistry, SunYat-sen University, Guangzhou 510275, China
| | - Huangcan Chen
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemical & Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Dexiong Zhou
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemical & Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Zhengmin Yang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemical & Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Ke Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemical & Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Wei Liu
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemical & Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Shengping Deng
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemical & Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Ruiyun Yang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemical & Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Jun Li
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemical & Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Ruijie He
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemical & Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China; Guangxi Key Laboratory of Functional Phytochemicals Research and Utilization, Guangxi Institute of Botany, Guilin 541006, China.
| |
Collapse
|
17
|
Zou YH, Zhao L, Xu YK, Bao JM, Liu X, Zhang JS, Li W, Ahmed A, Yin S, Tang GH. Anti-inflammatory sesquiterpenoids from the Traditional Chinese Medicine Salvia plebeia: Regulates pro-inflammatory mediators through inhibition of NF-κB and Erk1/2 signaling pathways in LPS-induced Raw264.7 cells. JOURNAL OF ETHNOPHARMACOLOGY 2018; 210:95-106. [PMID: 28847754 DOI: 10.1016/j.jep.2017.08.034] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Salvia plebeia R. Brown, a traditional Chinese medicinal herb, has been used to treat inflammatory diseases such as cough, hepatitis, and diarrhea for a long history. AIM OF THE STUDY The aim of the present study was to isolate and identify potential anti-inflammatory agents from the herb of S. plebeia, which may have contributed to its folk pharmacological use in the treatment of inflammatory diseases. MATERIAL AND METHODS The aerial parts of S. plebeia were extracted with 95% ethanol and separated by silica gel, RP-C18, Sephadex LH-20, and HPLC. The structures of the isolated compounds were elucidated by extensive spectroscopic analysis (MS, NMR, and X-ray). Anti-inflammatory activities of all compounds were evaluated by the model of LPS-induced up-regulated of NO in Raw264.7 macrophages. The expression levels of cytokine (TNF-α) and proteins (iNOS and COX-2) were assessed by ELISA kit and Western blotting analysis, respectively. Furthermore, the influences of salviplenoid A (1) on NF-κB and MAPK signaling pathways were determined by Western blotting analysis and immunofluorescence assay. RESULTS Six new (1-6, salviplenoids A-F) and ten known (7-16) sesquiterpenoids were isolated from the herb of S. plebeia. The absolute configurations of compounds 1, 2, and 7 were determined by X-ray diffraction. The new eudesmane-type sesquiterpenoid, salviplenoid A (1), significantly decreased the release of NO and TNF-α and the expression of proteins iNOS and COX-2. In addition, the biochemical mechanistic study indicated that 1 regulated the NF-κB dependent transcriptional activity through inhibiting the nuclear translocation of p50/p65 dimer and decreasing the phosphorylation of IκB and Erk1/2. CONCLUSIONS Among all sesquiterpenoids isolated from S. plebeian, the new salviplenoid A (1) exhibited the most potent anti-inflammatory activity in LPS-induced Raw264.7 cells via inhibition of NF-κB and Erk1/2 signaling pathways.
Collapse
Affiliation(s)
- Yi-Hong Zou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Liang Zhao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - You-Kai Xu
- Key Laboratory of Tropical Plant Resource and Sustainable Use, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Menglun, Yunnan 666303, China
| | - Jing-Mei Bao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Xin Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Jun-Sheng Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Wei Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Abrar Ahmed
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Sheng Yin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Gui-Hua Tang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
18
|
|
19
|
Park JY, Kim SI, Lee HJ, Kim SS, Kwon YS, Chun W. Isorhamnetin-3-O-Glucuronide Suppresses JNK and p38 Activation and Increases Heme-Oxygenase-1 in Lipopolysaccharide-Challenged RAW264.7 Cells. Drug Dev Res 2017; 77:143-51. [PMID: 27113811 DOI: 10.1002/ddr.21301] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 03/08/2016] [Indexed: 12/24/2022]
Abstract
Preclinical Research Isorhanmetin (ISH) exhibits a wide range of biological properties including anticancer, anti-oxidant and anti-inflammatory activities. However, the pharmacological properties of isorhamnetin-3-O-glucuronide (IG), a glycoside derivative of ISH, have not been extensively examined. The objective of this study was to examine the anti-inflammatory properties of IG and its underlying mechanism in lipopolysaccharide (LPS)-challenged RAW264.7 macrophage cells in comparison with its aglycone, ISH. IG suppressed LPS-induced extracellular secretion of the proinflammatory mediators, nitric oxide (NO) and PGE2 , and proinflammatory protein expressions of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2. IG also increased expression of heme oxygenase-1 (HO-1). IG attenuated LPS-induced activation of c-Jun N-terminal kinase (JNK) and p38 in a concentration-dependent manner with negligible suppression of extracellular signal-regulated kinases (ERK) phosphorylation. In conclusion, this study demonstrates that IG exerts anti-inflammatory activity by increasing HO-1 expression and by suppressing JNK and p38 signaling pathways in LPS-challenged RAW264.7 macrophage cells. Drug Dev Res 77 : 143-151, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jin-Young Park
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, 200-701, Korea
| | - Song-In Kim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, 200-701, Korea
| | - Hee Jae Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, 200-701, Korea
| | - Sung-Soo Kim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, 200-701, Korea
| | - Yong-Soo Kwon
- College of Pharmacy, Kangwon National University, Chuncheon, 200-701, Korea
| | - Wanjoo Chun
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, 200-701, Korea
| |
Collapse
|
20
|
Bhatt NP, Park JY, Lee HJ, Kim SS, Kwon YS, Chun W. Apocynin protects mesangial cells from lipopolysaccharide-induced inflammation by exerting heme oxygenase 1-mediated monocyte chemoattractant protein-1 suppression. Int J Mol Med 2017; 40:1294-1301. [DOI: 10.3892/ijmm.2017.3090] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/01/2017] [Indexed: 11/06/2022] Open
|
21
|
Zhai XT, Chen JQ, Jiang CH, Song J, Li DY, Zhang H, Jia XB, Tan W, Wang SX, Yang Y, Zhu FX. Corydalis bungeana Turcz. attenuates LPS-induced inflammatory responses via the suppression of NF-κB signaling pathway in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2016; 194:153-161. [PMID: 27616027 DOI: 10.1016/j.jep.2016.09.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 08/26/2016] [Accepted: 09/07/2016] [Indexed: 05/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Corydalis bungeana Turcz. (C. bungeana) is one of traditionally used medicines in China and possesses various biological effects, such as anti-inflammatory, antibacterial activity and inhibition of the immune function of the host. AIM OF THE STUDY we studied the anti-inflammatory effect and molecular mechanism involved of C. bungeana both in vitro and in vivo model system in which the inflammatory response was induced by LPS treatment. MATERIALS AND METHODS Anti-inflammatory activity of C. bungeana was investigated by LPS-induced RAW 264.7 macrophages and BALB/c mice. The production and expression of pro-inflammatory cytokines were evaluated by Griess reagent, ELISA kits and RT-qPCR, respectively. Phosphorylation status of IκBα and p65 was illustrated by western blot assay. RESULTS C. bungeana reduced the secretion of NO, TNF-α, IL-6 and IL-1β through inhibiting the protein expression of iNOS, TNF-α, IL-6 and IL-1β in vitro and in vivo. Western blot analysis suggested that C. bungeana supressed NF-κB activation via regulating the phosphorylation of IκBα and p65. Immunohistochemical assay also demostrated the histological inflammatory change in liver tissue. CONCLUSIONS The results indicate that C. bungeana supresses the activation of NF-κB signaling pathway through inhibiting phosphorylation of IκBα and p65, which results in good anti-inflammatory effect. In addition, C. bungeana attenuates inflammatory reaction by supressing the expression of various inflammatory cytokines both in vivo and in vitro.
Collapse
Affiliation(s)
- Xiao-Ting Zhai
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Key Laboratory of New Drug Delivery System of Chinese Meteria Medica, Jiangsu Province Academy of Chinese Medicine, Nanjing 210028, PR China; Department of Analytical Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jia-Quan Chen
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Cui-Hua Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, PR China
| | - Jie Song
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Key Laboratory of New Drug Delivery System of Chinese Meteria Medica, Jiangsu Province Academy of Chinese Medicine, Nanjing 210028, PR China
| | - Dong-Yu Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Key Laboratory of New Drug Delivery System of Chinese Meteria Medica, Jiangsu Province Academy of Chinese Medicine, Nanjing 210028, PR China
| | - Hao Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Key Laboratory of New Drug Delivery System of Chinese Meteria Medica, Jiangsu Province Academy of Chinese Medicine, Nanjing 210028, PR China
| | - Xiao-Bin Jia
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Key Laboratory of New Drug Delivery System of Chinese Meteria Medica, Jiangsu Province Academy of Chinese Medicine, Nanjing 210028, PR China
| | - Wei Tan
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Key Laboratory of New Drug Delivery System of Chinese Meteria Medica, Jiangsu Province Academy of Chinese Medicine, Nanjing 210028, PR China
| | - Shu-Xia Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Department of Clinical Laboratory, Jiangsu Province Hospital on Integration Medicine, Nanjing 210028, PR China
| | - Yi Yang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Key Laboratory of New Drug Delivery System of Chinese Meteria Medica, Jiangsu Province Academy of Chinese Medicine, Nanjing 210028, PR China
| | - Fen-Xia Zhu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Key Laboratory of New Drug Delivery System of Chinese Meteria Medica, Jiangsu Province Academy of Chinese Medicine, Nanjing 210028, PR China.
| |
Collapse
|
22
|
Liu Q, Chen Y, Shen C, Xiao Y, Wang Y, Liu Z, Liu X. Chicoric acid supplementation prevents systemic inflammation‐induced memory impairment and amyloidogenesis via inhibition of NF‐κB. FASEB J 2016; 31:1494-1507. [DOI: 10.1096/fj.201601071r] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/12/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Qian Liu
- Laboratory of Functional Chemistry and Nutrition of FoodCollege of Food Science and Engineering, Northwest A&F University Yangling China
| | - Yuwei Chen
- Laboratory of Functional Chemistry and Nutrition of FoodCollege of Food Science and Engineering, Northwest A&F University Yangling China
| | - Chun Shen
- Laboratory of Functional Chemistry and Nutrition of FoodCollege of Food Science and Engineering, Northwest A&F University Yangling China
| | - Yating Xiao
- Laboratory of Functional Chemistry and Nutrition of FoodCollege of Food Science and Engineering, Northwest A&F University Yangling China
| | - Yutang Wang
- Laboratory of Functional Chemistry and Nutrition of FoodCollege of Food Science and Engineering, Northwest A&F University Yangling China
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of FoodCollege of Food Science and Engineering, Northwest A&F University Yangling China
| | - Xuebo Liu
- Laboratory of Functional Chemistry and Nutrition of FoodCollege of Food Science and Engineering, Northwest A&F University Yangling China
| |
Collapse
|
23
|
Park JY, Lim MS, Kim SI, Lee HJ, Kim SS, Kwon YS, Chun W. Quercetin-3- O- β-D-Glucuronide Suppresses Lipopolysaccharide-Induced JNK and ERK Phosphorylation in LPS-Challenged RAW264.7 Cells. Biomol Ther (Seoul) 2016; 24:610-615. [PMID: 27257013 PMCID: PMC5098540 DOI: 10.4062/biomolther.2016.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/04/2016] [Accepted: 03/29/2016] [Indexed: 01/21/2023] Open
Abstract
Quercetin, a flavonol, has been reported to exhibit a wide range of biological properties including anti-oxidant and anti-inflammatory activities. However, pharmacological properties of quercetin-3-O-β-D-glucuronide (QG), a glycoside derivative of quercetin, have not been extensively examined. The objective of this study is to elucidate the anti-inflammatory property and underlying mechanism of QG in lipopolysaccharide (LPS)-challenged RAW264.7 macrophage cells in comparison with quercetin. QG significantly suppressed LPS-induced extracellular secretion of pro-inflammatory mediators such as nitric oxide (NO) and PGE2, and pro-inflammatory protein expressions of iNOS and COX-2. To elucidate the underlying mechanism of the anti-inflammatory property of QG, involvement of MAPK signaling pathways was examined. QG significantly attenuated LPS-induced activation of JNK and ERK in concentration-dependent manners with a negligible effect on p38. In conclusion, the present study demonstrates QG exerts anti-inflammatory activity through the suppression of JNK and ERK signaling pathways in LPS-challenged RAW264.7 macrophage cells.
Collapse
Affiliation(s)
- Jin-Young Park
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Man-Sup Lim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Song-In Kim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hee Jae Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Sung-Soo Kim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Yong-Soo Kwon
- College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Wanjoo Chun
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
24
|
Hwang YJ, Lee SJ, Park JY, Chun W, Nam SJ, Park JM, Park SC, Choi DH, Kang CD. Apocynin Suppresses Lipopolysaccharide-Induced Inflammatory Responses Through the Inhibition of MAP Kinase Signaling Pathway in RAW264.7 Cells. Drug Dev Res 2016; 77:271-7. [DOI: 10.1002/ddr.21321] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 07/10/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Young-Jae Hwang
- Department of Internal Medicine; Kangwon National University School of Medicine; Chuncheon Korea
| | - Sung Joon Lee
- Department of Internal Medicine; Kangwon National University School of Medicine; Chuncheon Korea
| | - Jin-Young Park
- Department of Pharmacology, College of Medicine; Kangwon National University; Chuncheon Korea
| | - Wanjoo Chun
- Department of Pharmacology, College of Medicine; Kangwon National University; Chuncheon Korea
| | - Seung-Joo Nam
- Department of Internal Medicine; Kangwon National University School of Medicine; Chuncheon Korea
| | - Jin Myung Park
- Department of Internal Medicine; Kangwon National University School of Medicine; Chuncheon Korea
| | - Sung Chul Park
- Department of Internal Medicine; Kangwon National University School of Medicine; Chuncheon Korea
| | - Dae Hee Choi
- Department of Internal Medicine; Kangwon National University School of Medicine; Chuncheon Korea
| | - Chang Don Kang
- Department of Internal Medicine; Kangwon National University School of Medicine; Chuncheon Korea
| |
Collapse
|
25
|
Jin SE, Kim OS, Yoo SR, Seo CS, Kim Y, Shin HK, Jeong SJ. Anti-inflammatory effect and action mechanisms of traditional herbal formula Gamisoyo-san in RAW 264.7 macrophages. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:219. [PMID: 27422559 PMCID: PMC4946171 DOI: 10.1186/s12906-016-1197-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 07/07/2016] [Indexed: 11/10/2022]
Abstract
BACKGROUND Gamisoyo-san (GMSYS) is a traditional herbal formula used to treat insomnia, dysmenorrhea, and infertility in Korea. The purpose of this study was to investigate the anti-inflammatory effect and action mechanisms of GMSYS in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. METHODS The anti-inflammatory effects of GMSYS were investigated using nitric oxide (NO) assay and ELISAs for prostaglandin E2 (PGE2), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6). The anti-inflammatory action mechanisms of GMSYS were evaluated using Western blotting for inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and activation of nuclear transcription factor kappa B (NF-κB) and mitogen-activated protein kinases (MAPKs). RESULTS GMSYS significantly inhibited the LPS-induced production of NO, PGE2, TNF-α, and IL-6 compared with the vehicle-treated cells. GMSYS consistently downregulated the expression of iNOS and COX-2 mRNA induced by LPS. In addition, pretreatment with GMSYS suppressed the LPS-induced activation of NF-κB and MAPKs such as p38, extracellular signal-regulated kinase (ERK), and c-Jun N-terminal kinase (JNK). CONCLUSIONS Our results indicate that the anti-inflammatory effects of GMSYS in RAW 264.7 macrophages are associated with inhibition of the release of inflammatory mediators and cytokines through the suppression of MAPK and NF-κB activation. These findings suggest that GMSYS may be a useful therapeutic candidate for the prevention or treatment of inflammatory diseases.
Collapse
|
26
|
Zhai XT, Zhang ZY, Jiang CH, Chen JQ, Ye JQ, Jia XB, Yang Y, Ni Q, Wang SX, Song J, Zhu FX. Nauclea officinalis inhibits inflammation in LPS-mediated RAW 264.7 macrophages by suppressing the NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2016; 183:159-165. [PMID: 26806575 DOI: 10.1016/j.jep.2016.01.018] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 01/14/2016] [Accepted: 01/18/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Nauclea officinalis has been traditionally used in China for the treatment of fever, pneumonia and enteritidis etc. This study aims to investigate effects of N. officinalis on the inflammatory response as well as the possible molecular mechanism in LPS-stimulated RAW 264.7 murine macrophage cells. MATERIALS AND METHODS Anti-inflammatory activity of N. officinalis (10, 20, 50, and 100µg/mL) was investigated by using LPS-induced RAW 264.7 macrophages. The NO production was determined by assaying nitrite in culture supernatants with the Griess reagent. The levels of TNF-α, IL-6 and IL-1β in culture media were measured with ELISA kits. Real time fluorescence quantitative PCR was detected for mRNA expression of iNOS, TNF-α, IL-6 and IL-1β. Western blot assay was performed to illustrate the inhibitory effects of N. officinalis on phosphorylation of IκB-α and NF-κB p65. RESULTS Treatment with N. officinalis (10-100µg/mL) dose-dependently inhibited the production as well as mRNA expression of NO, TNF-α, IL-6 and IL-1β in RAW 264.7 macrophages. Western blot assay suggested that the mechanism of the anti-inflammatory effect was associated with the inhibition of phosphorylation of IκB-α and NF-κB p65. CONCLUSIONS The results indicated that N. officinalis potentially inhibited the activation of upstream mediator NF-κB signaling pathway via suppressing phosphorylation of IκB-α and NF-κB p65 to inhibit LPS-stimulated inflammation.
Collapse
Affiliation(s)
- Xiao-Ting Zhai
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China; Department of Analytical Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Zhi-Yuan Zhang
- College of Pharmacy, Hainan Medical University, Haikou 570102, PR China
| | - Cui-Hua Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Jia-Quan Chen
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Ji-Qing Ye
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xiao-Bin Jia
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Yi Yang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Qian Ni
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Shu-Xia Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Jie Song
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Fen-Xia Zhu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China.
| |
Collapse
|
27
|
Jnawali HN, Jeon D, Park YG, Lee E, Heo YS, Kim Y. Rhamnetin Is a Potent Inhibitor of Extracellular Signal-regulated Kinase 1 and c-Jun N-Terminal Kinase 1. B KOREAN CHEM SOC 2015. [DOI: 10.1002/bkcs.10413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Hum Nath Jnawali
- Department of Bioscience and Biotechnology, Bio-Molecular Informatics Center; Konkuk University; Seoul 143-701 Korea
| | - Dasom Jeon
- Department of Bioscience and Biotechnology, Bio-Molecular Informatics Center; Konkuk University; Seoul 143-701 Korea
| | - Young-Gun Park
- Department of Bioscience and Biotechnology, Bio-Molecular Informatics Center; Konkuk University; Seoul 143-701 Korea
| | - Eunjung Lee
- Department of Bioscience and Biotechnology, Bio-Molecular Informatics Center; Konkuk University; Seoul 143-701 Korea
| | - Yong-Seok Heo
- Department of Chemistry; Konkuk University; Seoul 143-701 Korea
| | - Yangmee Kim
- Department of Bioscience and Biotechnology, Bio-Molecular Informatics Center; Konkuk University; Seoul 143-701 Korea
| |
Collapse
|
28
|
Dhammaraj T, Phintha A, Pinthong C, Medhanavyn D, Tinikul R, Chenprakhon P, Sucharitakul J, Vardhanabhuti N, Jiarpinitnun C, Chaiyen P. p-Hydroxyphenylacetate 3-Hydroxylase as a Biocatalyst for the Synthesis of Trihydroxyphenolic Acids. ACS Catal 2015. [DOI: 10.1021/acscatal.5b00439] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Taweesak Dhammaraj
- Department of Biochemistry and Center of
Excellence in Protein Structure and Function, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok 10400, Thailand
| | - Aisaraphon Phintha
- Department of Biochemistry and Center of
Excellence in Protein Structure and Function, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok 10400, Thailand
| | - Chatchadaporn Pinthong
- Department of Biochemistry and Center of
Excellence in Protein Structure and Function, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok 10400, Thailand
- Institute for Innovative Learning, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Dheeradhach Medhanavyn
- Department of Biochemistry and Center of
Excellence in Protein Structure and Function, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok 10400, Thailand
| | - Ruchanok Tinikul
- Mahidol University, Nakhonsawan Campus, Nakhonsawan 60130, Thailand
| | - Pirom Chenprakhon
- Institute for Innovative Learning, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Jeerus Sucharitakul
- Department
of Biochemistry, Faculty of Dentistry, Chulalongkorn University, Henri-Dunant
Road, Patumwan, Bangkok 10300, Thailand
| | - Nontima Vardhanabhuti
- Department of Pharmacy,
Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10300, Thailand
| | - Chutima Jiarpinitnun
- Department of Chemistry
and Center of Excellence for Innovation in Chemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Pimchai Chaiyen
- Department of Biochemistry and Center of
Excellence in Protein Structure and Function, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok 10400, Thailand
| |
Collapse
|
29
|
Cho BO, Yin HH, Fang CZ, Ha HO, Kim SJ, Jeong SI, Jang SI. Synergistic Anti-inflammatory Effect of Rosmarinic Acid and Luteolin in Lipopolysaccharide-Stimulated RAW264.7 Macrophage Cells. ACTA ACUST UNITED AC 2015. [DOI: 10.9721/kjfst.2015.47.1.119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
30
|
Ryu JH, Park HJ, Jeong YY, Han S, Shin JH, Lee SJ, Kang MJ, Sung NJ, Kang D. Aged red garlic extract suppresses nitric oxide production in lipopolysaccharide-treated RAW 264.7 macrophages through inhibition of NF-κB. J Med Food 2015; 18:439-45. [PMID: 25584924 DOI: 10.1089/jmf.2014.3214] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Lipopolysaccharides (LPS) activate nuclear factor kappa B (NF-κB), a transcription factor that is involved in inflammatory response. The pathways that activate NF-κB can be modulated by phytochemicals derived from garlic. We recently demonstrated that aged red garlic extract (ARGE), a new formulation of garlic, decreases nitric oxide (NO) generation by upregulating of heme oxygenase-1 (HO-1) in RAW 264.7 cells activated by LPS. However, the effects of ARGE on LPS-induced NF-κB activation are unknown. This study was performed to evaluate whether ARGE regulates LPS-induced NO production by modulation of NF-κB activation in macrophages. The inhibition of NF-κB by Bay 11-7085, an inhibitor of NF-κB, decreased LPS-induced NO production. ARGE treatment markedly reduced LPS-induced NO production and NF-κB nuclear translocation. ARGE downregulated expression of inducible nitric oxide synthase (iNOS) and upregulated expression of HO-1, a cytoprotective and anti-inflammatory protein. However, Bay 11-7085 only reduced iNOS expression. The NO production and iNOS expressions upregulated by suppression of HO-1 were suppressed by treatment with ARGE and Bay 11-7085. These results show that ARGE reduces LPS-induced NO production in macrophages through inhibition of NF-κB nuclear translocation and HO-1 activation. Compared to Bay 11-7085, ARGE may enhance anti-inflammatory effects by controlling other anti-inflammatory signals as well as regulation of NF-κB.
Collapse
Affiliation(s)
- Ji Hyeon Ryu
- 1 Department of Physiology, Institute of Health Sciences, Gyeongsang National University School of Medicine , Jinju, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Hur J, Lee P, Kim MJ, Cho YW. Regulatory Effect of 25-hydroxyvitamin D3 on Nitric Oxide Production in Activated Microglia. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2014; 18:397-402. [PMID: 25352759 PMCID: PMC4211123 DOI: 10.4196/kjpp.2014.18.5.397] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 08/07/2014] [Accepted: 08/20/2014] [Indexed: 11/15/2022]
Abstract
Microglia are activated by inflammatory and pathophysiological stimuli in neurodegenerative diseases, and activated microglia induce neuronal damage by releasing cytotoxic factors like nitric oxide (NO). Activated microglia synthesize a significant amount of vitamin D3 in the rat brain, and vitamin D3 has an inhibitory effect on activated microglia. To investigate the possible role of vitamin D3 as a negative regulator of activated microglia, we examined the effect of 25-hydroxyvitamin D3 on NO production of lipopolysaccharide (LPS)-stimulated microglia. Treatment with LPS increased the production of NO in primary cultured and BV2 microglial cells. Treatment with 25-hydroxyvitamin D3 inhibited the generation of NO in LPS-activated primary microglia and BV2 cells. In addition to NO production, expression of 1-α-hydroxylase and the vitamin D receptor (VDR) was also upregulated in LPS-stimulated primary and BV2 microglia. When BV2 cells were transfected with 1-α-hydroxylase siRNA or VDR siRNA, the inhibitory effect of 25-hydroxyvitamin D3 on activated BV2 cells was suppressed. 25-Hydroxyvitamin D3 also inhibited the increased phosphorylation of p38 seen in LPS-activated BV2 cells, and this inhibition was blocked by VDR siRNA. The present study shows that 25-hydroxyvitamin D3 inhibits NO production in LPS-activated microglia through the mediation of LPS-induced 1-α-hydroxylase. This study also shows that the inhibitory effect of 25-hydroxyvitamin D3 on NO production might be exerted by inhibiting LPS-induced phosphorylation of p38 through the mediation of VDR signaling. These results suggest that vitamin D3 might have an important role in the negative regulation of microglial activation.
Collapse
Affiliation(s)
- Jinyoung Hur
- Korea Food Research Institute, Seongnam 463-746, Korea
| | - Pyeongjae Lee
- Department of Natural Medicine Resources, Semyung University, Jecheon 390-711, Korea
| | - Mi Jung Kim
- Department of Physiology, Biomedical Science Institute and Medical Research Center for Reactive Oxygen Species, School of Medicine, Kyung Hee University, Seoul 130-701, Korea
| | - Young-Wuk Cho
- Department of Physiology, Biomedical Science Institute and Medical Research Center for Reactive Oxygen Species, School of Medicine, Kyung Hee University, Seoul 130-701, Korea
| |
Collapse
|
32
|
Yang EJ, Moon JY, Kim SS, Yang KW, Lee WJ, Lee NH, Hyun CG. Jeju seaweeds suppress lipopolysaccharide-stimulated proinflammatory response in RAW 264.7 murine macrophages. Asian Pac J Trop Biomed 2014; 4:529-37. [PMID: 25183272 DOI: 10.12980/apjtb.4.2014c1099] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 06/25/2014] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To investigate the anti-inflammatory effects of Jeju seaweeds on macrophage RAW 264.7 cells under lipopolysaccharide (LPS) stimulation. METHODS Ethyl acetate fractions were prepared from five different types of Jeju seaweeds, Dictyopteris divaricata (D. divaricata), Dictyopteris prolifera (D. prolifera), Prionitis cornea (P. cornea), Grateloupia lanceolata (G. lanceolata), and Grateloupia filicina (G. filicina). They were screened for inhibitory effects on proinflammatory mediators and cytokines such as nitric oxide (NO), prostaglandin E2, tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6). RESULTS Our results revealed that D. divaricata, D. prolifera, P. cornea, G. lanceolata, and G. filicina potently inhibited LPS-stimulated NO production (IC50 values were 18.0, 38.36, 38.43, 32.81 and 37.14 µg/mL, respectively). Consistent with these findings, D. divaricata, D. prolifera, P. cornea, and G. filicina also reduced the LPS-induced and prostaglandin E2 production in a concentration-dependent manner. Expectedly, they suppressed the expression of inducible NO synthase and cyclooxygenase-2 at the protein level in a dose-dependent manner in the RAW 264.7 cells, as determined by western blotting. In addition, the levels of TNF-α and IL-6, released into the medium, were also reduced by D. divaricata, D. prolifera, P. cornea, G. lanceolata, and G. filicina in a dose-dependent manner (IC50 values for TNF-α were 16.11, 28.21, 84.27, 45.52 and 74.75 µg/mL, respectively; IC50 values for IL-6 were 37.35, 80.08, 103.28, 62.53 and 84.28 µg/mL, respectively). The total phlorotannin content was measured by the Folin-Ciocalteu method and expressed as phloroglucinol equivalents. The content was 92.0 µg/mg for D. divaricata, 151.8 µg/mg for D. prolifera, 57.2 µg/mg for P. cornea, 53.0 µg/mg for G. lanceolata, and 40.2 µg/mg for G. filicina. CONCLUSIONS Thus, these findings suggest that Jeju seaweed extracts have potential therapeutic applications for inflammatory responses.
Collapse
Affiliation(s)
- Eun-Jin Yang
- Cosmetic Science Center, Department of Chemistry, Jeju National University, Jeju 690-756, Korea ; Jeju Technopark, Ara-1-dong, Jeju-si, Jeju 690-121, Korea
| | - Ji-Young Moon
- Cosmetic Science Center, Department of Chemistry, Jeju National University, Jeju 690-756, Korea ; Jeju Technopark, Ara-1-dong, Jeju-si, Jeju 690-121, Korea
| | - Sang Suk Kim
- Cosmetic Science Center, Department of Chemistry, Jeju National University, Jeju 690-756, Korea ; Citrus Research Station, National Institute of Horticultural & Herbal Science, RDA, Jeju 699-946, Korea
| | - Kyong-Wol Yang
- Jeju Love Co., Ltd., 542-5 Haengwon-ri, Gujwa-eup, Jeju 695-975, Korea
| | - Wook Jae Lee
- Jeju Technopark, Ara-1-dong, Jeju-si, Jeju 690-121, Korea
| | - Nam Ho Lee
- Cosmetic Science Center, Department of Chemistry, Jeju National University, Jeju 690-756, Korea
| | - Chang-Gu Hyun
- Cosmetic Science Center, Department of Chemistry, Jeju National University, Jeju 690-756, Korea ; LINC Agency, Jeju National University, Ara-1-dong, Jeju 690-756, Korea
| |
Collapse
|
33
|
Cho BO, Ryu HW, So Y, Lee CW, Jin CH, Yook HS, Jeong YW, Park JC, Jeong IY. Anti-Inflammatory Effect of Mangostenone F in Lipopolysaccharide-Stimulated RAW264.7 Macrophages by Suppressing NF-κB and MAPK Activation. Biomol Ther (Seoul) 2014; 22:288-94. [PMID: 25143806 PMCID: PMC4131528 DOI: 10.4062/biomolther.2014.052] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 06/05/2014] [Accepted: 06/10/2014] [Indexed: 11/06/2022] Open
Abstract
Mangostenone F (MF) is a natural xanthone isolated from Garcinia mangostana. However, little is known about the biological activities of MF. This study was designed to investigate the anti-inflammatory effect and underlying molecular mechanisms of MF in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. MF dose-dependently inhibited the production of NO, iNOS, and pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β) in LPS-stimulated RAW264.7 macrophages. Moreover, MF decreased the NF-κB luciferase activity and NF-κB DNA binding capacity in LPS-stimulated RAW264.7 macrophages. Furthermore, MF suppressed the NF-κB activation by inhibiting the degradation of IκBα and nuclear translocation of p65 subunit of NF-κB. In addition, MF attenuated the AP-1 luciferase activity and phosphorylation of ERK, JNK, and p38 MAP kinases. Taken together, these results suggest that the anti-inflammatory effect of MF is associated with the suppression of NO production and iNOS expression through the down-regulation of NF-κB activation and MAPK signaling pathway in LPS-stimulated RAW264.7 macrophages.
Collapse
Affiliation(s)
- Byoung Ok Cho
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 580-185
| | - Hyung Won Ryu
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon-gun 363-883
| | - Yangkang So
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 580-185
| | - Chang Wook Lee
- Department of Food Science and Human Nutrition, Chungnam National University, Daejeon 305-764
| | - Chang Hyun Jin
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 580-185
| | - Hong Sun Yook
- Department of Food Science and Human Nutrition, Chungnam National University, Daejeon 305-764
| | - Yong Wook Jeong
- Department of Microbiology, College of Medicine, Seonam University, Namwon 590-711, Republic of Korea
| | - Jong Chun Park
- Department of Microbiology, College of Medicine, Seonam University, Namwon 590-711, Republic of Korea
| | - Il Yun Jeong
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 580-185
| |
Collapse
|
34
|
IMM-H004, a novel coumarin derivative compound, attenuates the production of inflammatory mediatory mediators in lipopolysaccharide-activated BV2 microglia. Brain Res Bull 2014; 106:30-8. [PMID: 24878446 DOI: 10.1016/j.brainresbull.2014.05.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 05/16/2014] [Accepted: 05/19/2014] [Indexed: 01/11/2023]
Abstract
Therapeutic strategies designed to inhibit the activation of microglia may lead to significant advancement in the treatment of most neurodegenerative diseases. 7-hydroxy-5-methoxy-4-methyl-3-(4-methylpiperazin-1-yl)-coumarin (IMM-H004) is a novel compound and has been reported exerting potent neuroprotective effects which may be related to anti-inflammation. In the present study, the anti-inflammatory effects of IMM-H004 were investigated in lipopolysaccharide (LPS)-treated BV2 microglia. Our observations indicated that treatment with IMM-H004 significantly inhibited BV2 microglia activation, protected PC12 cells and primary neurons against indirect toxicity mediated by exposure to conditioned medium (CM) from LPS-treated BV2 cells. Additionally, IMM-H004 significantly suppressed the release of TNF-α, IL-1β and NO, and suppressed the expression of pro-inflammatory mediators and cytokines such as iNOS, COX-2, and IL-6 in LPS-stimulated BV2 microglia. The nuclear translocation of NF-κB and the phosphorylation level of JNK and p38 MAPK pathways were also inhibited by IMM-H004 in LPS-treated BV2 microglia. Moreover, IMM-H004 also was a strong selective OH scavenger whose effect was similar with vitamin C. Overall, our findings suggested that IMM-H004 might be a promising therapeutic agent for alleviating the progress of neurodegenerative diseases associated with microglia activation.
Collapse
|
35
|
Vo VA, Lee JW, Park JH, Kwon JH, Lee HJ, Kim SS, Kwon YS, Chun W. N-(p-Coumaryol)-Tryptamine Suppresses the Activation of JNK/c-Jun Signaling Pathway in LPS-Challenged RAW264.7 Cells. Biomol Ther (Seoul) 2014; 22:200-6. [PMID: 25009700 PMCID: PMC4060082 DOI: 10.4062/biomolther.2014.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 03/17/2014] [Accepted: 03/24/2014] [Indexed: 01/01/2023] Open
Abstract
N-(p-Coumaryol) tryptamine (CT), a phenolic amide, has been reported to exhibit anti-oxidant and anti-inflammatory activities. However, the underlying mechanism by which CT exerts its pharmacological properties has not been clearly demonstrated. The objective of this study is to elucidate the anti-inflammatory mechanism of CT in lipopolysaccharide (LPS)-challenged RAW264.7 macrophage cells. CT significantly inhibited LPS-induced extracellular secretion of pro-inflammatory mediators such as nitric oxide (NO) and PGE2, and protein expressions of iNOS and COX-2. In addition, CT significantly suppressed LPS-induced secretion of pro-inflammatory cytokines such as TNF-α and IL-1β. To elucidate the underlying anti-inflammatory mechanism of CT, involvement of MAPK and Akt signaling pathways was examined. CT significantly attenuated LPS-induced activation of JNK/c-Jun, but not ERK and p38, in a concentration-dependent manner. Interestingly, CT appeared to suppress LPS-induced Akt phosphorylation. However, JNK inhibition, but not Akt inhibition, resulted in the suppression of LPS-induced responses, suggesting that JNK/c-Jun signaling pathway significantly contributes to LPS-induced inflammatory responses and that LPS-induced Akt phosphorylation might be a compensatory response to a stress condition. Taken together, the present study clearly demonstrates CT exerts anti-inflammatory activity through the suppression of JNK/c-Jun signaling pathway in LPS-challenged RAW264.7 macrophage cells.
Collapse
Affiliation(s)
- Van Anh Vo
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701
| | - Jae-Won Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701
| | - Jun-Ho Park
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701
| | - Jae-Hyun Kwon
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701
- Department of Radiology, Dongguk University Ilsan Hospital, Ilsan 410-773, Republic of Korea
| | - Hee Jae Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701
| | - Sung-Soo Kim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701
| | - Yong-Soo Kwon
- College of Pharmacy, Kangwon National University, Chuncheon 200-701
| | - Wanjoo Chun
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701
| |
Collapse
|
36
|
A novel synthetic dibenzocyclooctadiene lignan analog XLYF-104-6 attenuates lipopolysaccharide-induced inflammatory response in RAW264.7 macrophage cells and protects BALB/c mice from sepsis. Eur J Pharmacol 2014; 729:22-9. [DOI: 10.1016/j.ejphar.2014.01.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 12/31/2013] [Accepted: 01/09/2014] [Indexed: 11/19/2022]
|
37
|
Vo VA, Lee JW, Shin SY, Kwon JH, Lee HJ, Kim SS, Kwon YS, Chun W. Methyl p-Hydroxycinnamate Suppresses Lipopolysaccharide-Induced Inflammatory Responses through Akt Phosphorylation in RAW264.7 Cells. Biomol Ther (Seoul) 2014; 22:10-6. [PMID: 24596616 PMCID: PMC3936424 DOI: 10.4062/biomolther.2013.095] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 12/17/2013] [Accepted: 12/20/2013] [Indexed: 12/02/2022] Open
Abstract
Derivatives of caffeic acid have been reported to possess diverse pharmacological properties such as anti-inflammatory, anti-tumor, and neuroprotective effects. However, the biological activity of methyl p-hydroxycinnamate, an ester derivative of caffeic acid, has not been clearly demonstrated. This study aimed to elucidate the anti-inflammatory mechanism of methyl p-hydroxycinnamate in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophage cells. Methyl p-hydroxycinnamate significantly inhibited LPS-induced excessive production of pro-inflammatory mediators such as nitric oxide (NO) and PGE2 and the protein expression of iNOS and COX-2. Methyl p-hydroxycinnamate also suppressed LPS-induced overproduction of pro-inflammatory cytokines such as IL-1β and TNF-α. In addition, methyl p-hydroxycinnamate significantly suppressed LPS-induced degradation of IκB, which retains NF-κB in the cytoplasm, consequently inhibiting the transcription of pro-inflammatory genes by NF-κB in the nucleus. Methyl p-hydroxycinnamate exhibited significantly increased Akt phosphorylation in a concentration-dependent manner. Furthermore, inhibition of Akt signaling pathway with wortmaninn abolished methyl p-hydroxycinnamate-induced Akt phosphorylation. Taken together, the present study clearly demonstrates that methyl p-hydroxycinnamate exhibits anti-inflammatory activity through the activation of Akt signaling pathway in LPS-stimulated RAW264.7 macrophage cells.
Collapse
Affiliation(s)
- Van Anh Vo
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701
| | - Jae-Won Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701
| | - Seung-Yeon Shin
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701
| | - Jae-Hyun Kwon
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701 ; Department of Radiology, Dongguk University Ilsan Hospital, Ilsan 410-773, Republic of Korea
| | - Hee Jae Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701
| | - Sung-Soo Kim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701
| | - Yong-Soo Kwon
- College of Pharmacy, Kangwon National University, Chuncheon 200-701
| | - Wanjoo Chun
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701
| |
Collapse
|
38
|
Jnawali HN, Lee E, Jeong KW, Shin A, Heo YS, Kim Y. Anti-inflammatory activity of rhamnetin and a model of its binding to c-Jun NH2-terminal kinase 1 and p38 MAPK. JOURNAL OF NATURAL PRODUCTS 2014; 77:258-263. [PMID: 24397781 DOI: 10.1021/np400803n] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Rhamnetin (1), a commonly occurring plant O-methylated flavonoid, possesses antioxidant properties. To address the potential therapeutic efficacy of 1, its anti-inflammatory activity and mode of action in mouse macrophage-derived RAW264.7 cells stimulated with lipopolysaccharide (LPS) or interferon (IFN)-γ were investigated. Rhamnetin (1) suppressed mouse tumor necrosis factor (mTNF)-α, mouse macrophage inflammatory protein (mMIP)-1, and mMIP-2 cytokine production in LPS-stimulated macrophages. A nontoxic dose of 1 suppressed nitric oxide production. It was found that the anti-inflammatory effects of 1 are mediated by actions on the p38 mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and cyclooxygenase (COX)-2 pathways in LPS- or IFN-γ-stimulated RAW264.7 cells. It was determined that 1 binds to human JNK1 (9.7 × 10(8) M(-1)) and p38 MAPK (2.31 × 10(7) M(-1)) with good affinity. The binding model showed interactions with the 3'- and 4'-hydroxy groups of the B-ring and the 5-hydroxy group of the A-ring of 1. Further, 1 exerted an anti-inflammatory effect, reducing the levels of pro-inflammatory cytokines and mediators.
Collapse
Affiliation(s)
- Hum Nath Jnawali
- Department of Bioscience and Biotechnology, Bio-Molecular Informatics Center, Konkuk University , Seoul 143-701, South Korea
| | | | | | | | | | | |
Collapse
|
39
|
Vo VA, Lee JW, Kim JY, Park JH, Lee HJ, Kim SS, Kwon YS, Chun W. Phosphorylation of Akt Mediates Anti-Inflammatory Activity of 1-p-Coumaroyl β-D-Glucoside Against Lipopolysaccharide-Induced Inflammation in RAW264.7 Cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2014; 18:79-86. [PMID: 24634601 PMCID: PMC3951828 DOI: 10.4196/kjpp.2014.18.1.79] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 12/17/2013] [Accepted: 12/31/2013] [Indexed: 11/15/2022]
Abstract
Hydroxycinnamic acids have been reported to possess numerous pharmacological activities such as antioxidant, anti-inflammatory, and anti-tumor properties. However, the biological activity of 1-p-coumaroyl β-D-glucoside (CG), a glucose ester derivative of p-coumaric acid, has not been clearly examined. The objective of this study is to elucidate the anti-inflammatory action of CG in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophage cells. In the present study, CG significantly suppressed LPS-induced excessive production of pro-inflammatory mediators such as nitric oxide (NO) and PGE2 and the protein expression of iNOS and COX-2. CG also inhibited LPS-induced secretion of pro-inflammatory cytokines, IL-1β and TNF-α. In addition, CG significantly suppressed LPS-induced degradation of IκB. To elucidate the underlying mechanism by which CG exerts its anti-inflammatory action, involvement of various signaling pathways were examined. CG exhibited significantly increased Akt phosphorylation in a concentration-dependent manner, although MAPKs such as Erk, JNK, and p38 appeared not to be involved. Furthermore, inhibition of Akt/PI3K signaling pathway with wortmannin significantly, albeit not completely, abolished CG-induced Akt phosphorylation and anti-inflammatory actions. Taken together, the present study demonstrates that Akt signaling pathway might play a major role in CG-mediated anti-inflammatory activity in LPS-stimulated RAW264.7 macrophage cells.
Collapse
Affiliation(s)
- Van Anh Vo
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701, Korea
| | - Jae-Won Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701, Korea
| | - Ji-Young Kim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701, Korea
| | - Jun-Ho Park
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701, Korea
| | - Hee Jae Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701, Korea
| | - Sung-Soo Kim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701, Korea
| | - Yong-Soo Kwon
- College of Pharmacy, Kangwon National University, Chuncheon 200-701, Korea
| | - Wanjoo Chun
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701, Korea
| |
Collapse
|
40
|
3,4,5-Trihydroxycinnamic acid inhibits lipopolysaccharide (LPS)-induced inflammation by Nrf2 activation in vitro and improves survival of mice in LPS-induced endotoxemia model in vivo. Mol Cell Biochem 2014; 390:143-53. [DOI: 10.1007/s11010-014-1965-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 01/14/2014] [Indexed: 10/25/2022]
|
41
|
Lee JW, Kim NH, Kim JY, Park JH, Shin SY, Kwon YS, Lee HJ, Kim SS, Chun W. Aromadendrin Inhibits Lipopolysaccharide-Induced Nuclear Translocation of NF-κB and Phosphorylation of JNK in RAW 264.7 Macrophage Cells. Biomol Ther (Seoul) 2013; 21:216-21. [PMID: 24265867 PMCID: PMC3830120 DOI: 10.4062/biomolther.2013.023] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 04/05/2013] [Accepted: 04/12/2013] [Indexed: 11/05/2022] Open
Abstract
Aromadendrin, a flavonol, has been reported to possess a variety of pharmacological activities such as anti-inflammatory, antioxidant, and anti-diabetic properties. However, the underlying mechanism by which aromadendrin exerts its biological activity has not been extensively demonstrated. The objective of this study is to elucidate the anti-inflammatory mechanism of aromadedrin in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophage cells. Aromadendrin significantly suppressed LPS-induced excessive production of pro-inflammatory mediators such as nitric oxide (NO) and PGE2. In accordance, aromadendrin attenuated LPSinduced overexpression iNOS and COX-2. In addition, aromadendrin significantly suppressed LPS-induced degradation of IκB, which sequesters NF-κB in cytoplasm, consequently inhibiting the nuclear translocation of pro-inflammatory transcription factor NF- κB. To elucidate the underlying signaling mechanism of anti-inflammatory activity of aromadendrin, MAPK signaling pathway was examined. Aromadendrin significantly attenuated LPS-induced activation of JNK, but not ERK and p38, in a concentration-dependent manner. Taken together, the present study clearly demonstrates that aromadendrin exhibits anti-inflammatory activity through the suppression of nuclear translocation of NF-κB and phosphorylation of JNK in LPS-stimulated RAW 264.7 macrophage cells.
Collapse
Affiliation(s)
- Jae-Won Lee
- Department of Pharmacology, College of Medicine
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Tsermentseli SK, Manesiotis P, Assimopoulou AN, Papageorgiou VP. Molecularly imprinted polymers for the isolation of bioactive naphthoquinones from plant extracts. J Chromatogr A 2013; 1315:15-20. [DOI: 10.1016/j.chroma.2013.09.044] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 08/07/2013] [Accepted: 09/11/2013] [Indexed: 10/26/2022]
|
43
|
Jnawali HN, Lee E, Jeong KW, Heo YS, Kim Y. Binding Model of Fisetin and Human c-Jun NH 2-Terminal Kinase 1 and Its Anti-inflammatory Activity. B KOREAN CHEM SOC 2013. [DOI: 10.5012/bkcs.2013.34.9.2629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
44
|
Lee JW, Choi YJ, Park JH, Sim JY, Kwon YS, Lee HJ, Kim SS, Chun W. 3,4,5-Trihydroxycinnamic Acid Inhibits Lipopolysaccharide-Induced Inflammatory Response through the Activation of Nrf2 Pathway in BV2 Microglial Cells. Biomol Ther (Seoul) 2013; 21:60-5. [PMID: 24009860 PMCID: PMC3762302 DOI: 10.4062/biomolther.2012.091] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 01/03/2013] [Accepted: 01/04/2013] [Indexed: 11/05/2022] Open
Abstract
3,4,5-Trihydroxycinnamic acid (THC) is a derivative of hydroxycinnamic acids, which have been reported to possess a variety of biological properties such as anti-inflammatory, anti-tumor, and neuroprotective activities. However, biological activity of THC has not been extensively examined. Recently, we reported that THC possesses anti-inflammatory activity in LPS-stimulated BV2 microglial cells. However, its precise mechanism by which THC exerts anti-inflammatory action has not been clearly identified. Therefore, the present study was carried out to understand the anti-inflammatory mechanism of THC in BV2 microglial cells. THC effectively suppressed the LPS-induced induction of pro-inflammatory mediators such as NO, TNF-α, and IL-1β. THC also suppressed expression of MCP-1, which plays a key role in the migration of activated microglia. To understand the underlying mechanism by which THC exerts these anti-inflammatory properties, involvement of Nrf2, which is a cytoprotective transcription factor, was examined. THC resulted in increased phosphorylation of Nrf2 with consequent expression of HO-1 in a concentration-dependent manner. THC-induced phosphorylation of Nrf2 was blocked with SB203580, a p38 MAPK inhibitor, indicating that p38 MAPK is the responsible kinase for the phosphorylation of Nrf2. Taken together, the present study for the first time demonstrates that THC exerts anti-inflammatory properties through the activation of Nrf2 in BV2 microglial cells, suggesting that THC might be a valuable therapeutic adjuvant for the treatment of inflammation-related disorders in the CNS.
Collapse
Affiliation(s)
- Jae-Won Lee
- Department of Pharmacology, College of Medicine, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Vo VA, Lee JW, Chang JE, Kim JY, Kim NH, Lee HJ, Kim SS, Chun W, Kwon YS. Avicularin Inhibits Lipopolysaccharide-Induced Inflammatory Response by Suppressing ERK Phosphorylation in RAW 264.7 Macrophages. Biomol Ther (Seoul) 2013; 20:532-7. [PMID: 24009846 PMCID: PMC3762284 DOI: 10.4062/biomolther.2012.20.6.532] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 11/16/2012] [Accepted: 11/20/2012] [Indexed: 11/26/2022] Open
Abstract
suppresAvicularin, quercetin-3-α-L-arabinofuranoside, has been reported to possess diverse pharmacological properties such as anti-inflammatory and anti-infectious effects. However, the underlying mechanism by which avicularin exerts its anti-inflammatory activity has not been clearly demonstrated. This study aimed to elucidate the anti-inflammatory mechanism of avicularin in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophage cells. Avicularin significantly inhibited LPS-induced excessive production of pro-inflammatory mediators such as nitric oxide (NO) and PGE2 and the protein levels of iNOS and COX-2, which are responsible for the production of NO and PGE2, respectively. Avicularin also suppressed LPS-induced overproduction of pro-inflammatory cytokine IL-1β. Furthermore, avicularin significantly suppressed LPS-induced degradation of IκB, which retains NF-κB in the cytoplasm, consequently inhibiting the transcription of pro-inflammatory genes by NF-κB in the nucleus. To understand the underlying signaling mechanism of anti-inflammatory activity of avicularin, involvement of multiple kinases was examined. Avicularin significantly attenuated LPS-induced activation of ERK signaling pathway in a concentration-dependent manner. Taken together, the present study clearly demonstrates that avicularin exhibits anti-inflammatory activity through the suppression of ERK signaling pathway in LPS-stimulated RAW 264.7 macrophage cells.
Collapse
Affiliation(s)
- Van Anh Vo
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 200-701, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Furuya T, Kino K. Catalytic activity of the two-component flavin-dependent monooxygenase from Pseudomonas aeruginosa toward cinnamic acid derivatives. Appl Microbiol Biotechnol 2013; 98:1145-54. [PMID: 23666444 DOI: 10.1007/s00253-013-4958-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 04/24/2013] [Accepted: 04/26/2013] [Indexed: 11/28/2022]
Abstract
4-Hydroxyphenylacetate 3-hydroxylases (HPAHs) of the two-component flavin-dependent monooxygenase family are attractive enzymes that possess the catalytic potential to synthesize valuable ortho-diphenol compounds from simple monophenol compounds. In this study, we investigated the catalytic activity of HPAH from Pseudomonas aeruginosa strain PAO1 toward cinnamic acid derivatives. We prepared Escherichia coli cells expressing the hpaB gene encoding the monooxygenase component and the hpaC gene encoding the oxidoreductase component. E. coli cells expressing HpaBC exhibited no or very low oxidation activity toward cinnamic acid, o-coumaric acid, and m-coumaric acid, whereas they rapidly oxidized p-coumaric acid to caffeic acid. Interestingly, after p-coumaric acid was almost completely consumed, the resulting caffeic acid was further oxidized to 3,4,5-trihydroxycinnamic acid. In addition, HpaBC exhibited oxidation activity toward 3-(4-hydroxyphenyl)propanoic acid, ferulic acid, and coniferaldehyde to produce the corresponding ortho-diphenols. We also investigated a flask-scale production of caffeic acid from p-coumaric acid as the model reaction for HpaBC-catalyzed syntheses of hydroxycinnamic acids. Since the initial concentrations of the substrate p-coumaric acid higher than 40 mM markedly inhibited its HpaBC-catalyzed oxidation, the reaction was carried out by repeatedly adding 20 mM of this substrate to the reaction mixture. Furthermore, by using the HpaBC whole-cell catalyst in the presence of glycerol, our experimental setup achieved the high-yield production of caffeic acid, i.e., 56.6 mM (10.2 g/L) within 24 h. These catalytic activities of HpaBC will provide an easy and environment-friendly synthetic approach to hydroxycinnamic acids.
Collapse
Affiliation(s)
- Toshiki Furuya
- Department of Applied Chemistry, Faculty of Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku-ku, Tokyo, 169-8555, Japan,
| | | |
Collapse
|
47
|
Shin JH, Ryu JH, Kang MJ, Hwang CR, Han J, Kang D. Short-term heating reduces the anti-inflammatory effects of fresh raw garlic extracts on the LPS-induced production of NO and pro-inflammatory cytokines by downregulating allicin activity in RAW 264.7 macrophages. Food Chem Toxicol 2013; 58:545-51. [PMID: 23583806 DOI: 10.1016/j.fct.2013.04.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/18/2013] [Accepted: 04/02/2013] [Indexed: 02/07/2023]
Abstract
Garlic has a variety of biologic activities, including anti-inflammatory properties. Although garlic has several biologic activities, some people dislike eating fresh raw garlic because of its strong taste and smell. Therefore, garlic formulations involving heating procedures have been developed. In this study, we investigated whether short-term heating affects the anti-inflammatory properties of garlic. Fresh and heated raw garlic extracts (FRGE and HRGE) were prepared with incubation at 25 °C and 95 °C, respectively, for 2 h. Treatment with FRGE and HRGE significantly reduced the LPS-induced increase in the pro-inflammatory cytokine concentration (TNF-α, IL-1β, and IL-6) and NO through HO-1 upregulation in RAW 264.7 macrophages. The anti-inflammatory effect was greater in FRGE than in HRGE. The allicin concentration was higher in FRGE than in HRGE. Allicin treatment showed reduced production of pro-inflammatory cytokines and NO and increased HO-1 activity. The results show that the decrease in LPS-induced NO and pro-inflammatory cytokines in RAW 264.7 macrophages through HO-1 induction was greater for FRGE compared with HRGE. Additionally, the results indicate that allicin is responsible for the anti-inflammatory effect of FRGE. Our results suggest a potential therapeutic use of allicin in the treatment of chronic inflammatory disease.
Collapse
Affiliation(s)
- Jung-Hye Shin
- Namhae Garlic Research Institute, Namhae 668-812, Republic of Korea
| | | | | | | | | | | |
Collapse
|