101
|
Methyl Jasmonate Reduces Inflammation and Oxidative Stress in the Brain of Arthritic Rats. Antioxidants (Basel) 2019; 8:antiox8100485. [PMID: 31618993 PMCID: PMC6826661 DOI: 10.3390/antiox8100485] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/02/2019] [Accepted: 10/04/2019] [Indexed: 12/29/2022] Open
Abstract
Methyl jasmonate (MeJA), common in the plant kingdom, is capable of reducing articular and hepatic inflammation and oxidative stress in adjuvant-induced arthritic rats. This study investigated the actions of orally administered MeJA (75–300 mg/kg) on inflammation, oxidative stress and selected enzyme activities in the brain of Holtzman rats with adjuvant-induced arthritis. MeJA prevented the arthritis-induced increased levels of nitrites, nitrates, lipid peroxides, protein carbonyls and reactive oxygen species (ROS). It also prevented the enhanced activities of myeloperoxidase and xanthine oxidase. Conversely, the diminished catalase and superoxide dismutase activities and glutathione (GSH) levels caused by arthritis were totally or partially prevented. Furthermore, MeJA increased the activity of the mitochondrial isocitrate dehydrogenase, which helps to supply NADPH for the mitochondrial glutathione cycle, possibly contributing to the partial recovery of the GSH/oxidized glutathione (GSSG) ratio. These positive actions on the antioxidant defenses may counterbalance the effects of MeJA as enhancer of ROS production in the mitochondrial respiratory chain. A negative effect of MeJA is the detachment of hexokinase from the mitochondria, which can potentially impair glucose phosphorylation and metabolism. In overall terms, however, it can be concluded that MeJA attenuates to a considerable extent the negative effects caused by arthritis in terms of inflammation and oxidative stress.
Collapse
|
102
|
Study on the Mechanism of Curcumin Regulating Lung Injury Induced by Outdoor Fine Particulate Matter (PM2.5). Mediators Inflamm 2019; 2019:8613523. [PMID: 31530996 PMCID: PMC6721509 DOI: 10.1155/2019/8613523] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 05/16/2019] [Accepted: 06/10/2019] [Indexed: 12/23/2022] Open
Abstract
Background Epidemiological studies have shown that exposure to PM induces oxidative stress, leading to a variety of health problems. In particular, PM2.5 contains a lot of substances harmful to the human body and penetrates into the lungs to induce lung injury. At the same time, there is increasing evidence that oxidative stress also affects the severity of lung injury. However, there is still no good way to reduce or eliminate these hazards. In the future, more experimental research is needed to further confirm the mechanisms of these hazards and formulate effective preventive measures and treatment plans for their hazard mechanisms. Curcumin has been reported to reduce oxidative stress and inflammatory damage and protect organs. Objective To investigate whether curcumin can play a protective role against PM2.5-induced oxidative stress and inflammatory damage by inducing expression of the HO-1/CO/P38 MAPK pathway. Methods In this experiment, PM2.5 was dropped into the trachea to establish a lung injury model in mice. 28 SPF-grade male Kunming mice were randomly divided into 4 groups: normal control group, saline control group, PM2.5 treatment group, and curcumin intervention group. Albumin (ALB), lactate dehydrogenase (LDH), and alkaline phosphatase (ALP) were measured in alveolar lavage fluid (BALF) to assess lung tissue damage. Colorimetric detection of oxidative stress indicators such as MDA, GSH-PX, T-AOC, and CAT in the lung tissue was performed. The levels of IL-6 and TNF-α in the lung tissue were determined by ELISA. Histopathological examination was used for the assessment of alveolar epithelial damage. The protein expression of the HO-1/P38 MAPK pathway in the lung tissue was determined by Western blot and immunohistochemistry. Endogenous CO was detected by spectrophotometry. The results showed that the expression of the HO-1/CO/P38 MAPK protein in the lung tissue was significantly increased in the curcumin intervention group compared with the PM2.5 treatment group, and it was statistically significant (P < 0.05). Compared with the PM2.5 treatment group, the curcumin intervention group can reduce the amount of ALB, LDH, and ALP in BALF; reduce the levels of MDA, IL-1, and TNF-α in the lung tissue; and improve GSH-PX, T-AOC, and CAT levels, but there is no statistical difference (P > 0.05). Conclusion We found that PM2.5 can cause lung damage through oxidative stress and inflammatory responses. Oxidative stress and inflammatory responses increase the expression of HO-1/CO/P38 MAPK. The intervention of curcumin can further increase the expression of HO-1/CO/P38 MAPK.
Collapse
|
103
|
Moon SW, Ahn CB, Oh Y, Je JY. Lotus (Nelumbo nucifera) seed protein isolate exerts anti-inflammatory and antioxidant effects in LPS-stimulated RAW264.7 macrophages via inhibiting NF-κB and MAPK pathways, and upregulating catalase activity. Int J Biol Macromol 2019; 134:791-797. [DOI: 10.1016/j.ijbiomac.2019.05.094] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/15/2019] [Accepted: 05/15/2019] [Indexed: 12/20/2022]
|
104
|
Meng XL, Chen ML, Chen CL, Gao CC, Li C, Wang D, Liu HS, Xu CB. Bisbenzylisoquinoline alkaloids of lotus (Nelumbo nucifera Gaertn.) seed embryo inhibit lipopolysaccharide-induced macrophage activation via suppression of Ca2+-CaM/CaMKII pathway. FOOD AGR IMMUNOL 2019. [DOI: 10.1080/09540105.2019.1638889] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Affiliation(s)
- Xue-Lian Meng
- School of Pharmaceutical Science, Liaoning University, Shenyang, People’s Republic of China
- Research Center for Natural Product Pharmacy of Liaoning Province, Shenyang, China
| | - Man-Ling Chen
- School of Pharmaceutical Science, Liaoning University, Shenyang, People’s Republic of China
| | - Chang-Lan Chen
- School of Pharmaceutical Science, Liaoning University, Shenyang, People’s Republic of China
- Research Center for Natural Product Pharmacy of Liaoning Province, Shenyang, China
| | - Cheng-Cheng Gao
- School of Pharmaceutical Science, Liaoning University, Shenyang, People’s Republic of China
| | - Chao Li
- School of Pharmaceutical Science, Liaoning University, Shenyang, People’s Republic of China
| | - Dan Wang
- School of Pharmaceutical Science, Liaoning University, Shenyang, People’s Republic of China
- Research Center for Natural Product Pharmacy of Liaoning Province, Shenyang, China
| | - Hong-Sheng Liu
- Research Center for Computer Simulating and Information Processing of Bio-macromolecules of Liaoning Province, Shenyang, People’s Republic of China
| | - Cheng-Bin Xu
- School of Environmental Science, Liaoning University, Shenyang, People’s Republic of China
| |
Collapse
|
105
|
Li Y, Zeng Y, Meng T, Gao X, Huang B, He D, Ran X, Du J, Zhang Y, Fu S, Hu G. Farrerol protects dopaminergic neurons in a rat model of lipopolysaccharide-induced Parkinson's disease by suppressing the activation of the AKT and NF-κB signaling pathways. Int Immunopharmacol 2019; 75:105739. [PMID: 31351366 DOI: 10.1016/j.intimp.2019.105739] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/30/2019] [Accepted: 07/02/2019] [Indexed: 12/26/2022]
Abstract
Neuroinflammation, characterized by the activation of microglia, is one of the major pathologic processes of Parkinson's disease (PD). Overactivated microglia can release many pro-inflammatory cytokines, which cause an excessive inflammatory response and eventually damage dopaminergic neurons. Therefore, the inhibition of neuroinflammation that results from the overactivation of microglia may be an method for the treatment of PD. Farrerol is a 2,3-dihydro-flavonoid obtained from Rhododendron, and it possesses various biological functions, including anti-inflammatory, antibacterial and antioxidant activities. However, the effect of farrerol on neuroinflammation has not been investigated. The present study uncovered a neuroprotective role for farrerol. In vitro, farrerol markedly decreased the production of inflammatory mediators, including interleukin-6 (IL-6), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), cyclooxygenase 2 (COX-2) and induced nitric oxide synthase (iNOS), induced by lipopolysaccharide (LPS) in BV-2 cells. This anti-inflammatory effect was regulated via inhibiting NF-κB p65 and AKT phosphorylation. Furthermore, we found that farrerol alleviated microglial activation and dopaminergic neuronal death in rats with LPS-induced PD. Pretreatment with farrerol markedly improved motor deficits in rats with LPS-induced PD. Taken together, our results indicate that the neuroprotective effect of the farrerol, which prevents microglial overactivation in rats with LPS-induced PD, may provide a potential therapy for patients suffering from PD.
Collapse
Affiliation(s)
- Yuhang Li
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Yalong Zeng
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Tianyu Meng
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Xiyu Gao
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Bingxu Huang
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Dewei He
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Xin Ran
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Jian Du
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Yufei Zhang
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Shoupeng Fu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Guiqiu Hu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
106
|
Hung YL, Wang SC, Suzuki K, Fang SH, Chen CS, Cheng WC, Su CC, Yeh HC, Tu HP, Liu PL, Huang MY, Li CY. Bavachin attenuates LPS-induced inflammatory response and inhibits the activation of NLRP3 inflammasome in macrophages. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 59:152785. [PMID: 31009850 DOI: 10.1016/j.phymed.2018.12.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 10/22/2018] [Accepted: 12/09/2018] [Indexed: 05/10/2023]
Abstract
BACKGROUND Bavachin is a natural product isolated from Psoralea corylifolia L. that has been applied as a traditional medicine in Asian countries. However, the anti-inflammatory effects of bavachin on LPS-induced inflammation and NLRP3 inflammasome activation by macrophages remain unclear. PURPOSE We investigated the anti-inflammatory effects of bavachin on LPS-activated murine macrophage cell line J774A.1 cells and murine peritoneal macrophages. METHODS J774A.1 cells and murine peritoneal macrophages were pre-treated with bavachin following LPS treatment. The concentrations of NO, PGE2, IL-6 and IL-12p40 in cell culture supernatant were analyzed. The expressions of iNOS, COX-2, mPGES-1 and MAPKs were analyzed using Western blotting, while NF-κB activity was detected using promoter reporter assay. To examine the activation of NLRP3 inflammasome, J774A.1 cells were incubated with LPS, and then treated with bavachin following treatment with ATP. The concentration of IL-1β in the cell culture supernatant was measured. The expressions of NLRP3, ASC, caspase-1 and IL-1β were analyzed using Western blotting. The formation of inflammasome complex was observed by immunofluorescence microscopy. RESULTS Bavachin suppressed LPS-induced NO and PGE2 production, and decreased iNOS and mPGES-1 expression. Bavachin also reduced LPS-induced IL-6 and IL-12p40 production and decreased the activation of MAPKs and NF-κB. Additionally, bavachin suppressed NLRP3 inflammasome-derived IL-1β secretion, decreased caspase-1 activation, repressed mature IL-1β expression, and inhibited inflammasome complex formation. Furthermore, bavachin also suppressed the production of NO, IL-6 and IL-12p40 by LPS-stimulated murine peritoneal macrophages. CONCLUSION Our experimental results indicated anti-inflammatory effects of bavachin exhibit attenuation of LPS-induced inflammation and inhibit activation of NLRP3 inflammasome in macrophages. These results suggest that bavachin might have potential in treating inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Yung-Li Hung
- Graduate School of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan
| | - Shu-Chi Wang
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan
| | - Shih-Hua Fang
- Institute of Athletics, National Taiwan University of Sport, Taichung 40404, Taiwan
| | - Chi-Shuo Chen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Wei-Chung Cheng
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Chia-Cheng Su
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Urology, Department of Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan; Department of Senior Citizen Service Management, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan
| | - Hsin-Chih Yeh
- Department of Urology, Kaohsiung Medical University Hospital and Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Urology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Hung-Pin Tu
- Department of Public Health and Environmental Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Po-Len Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ming-Yii Huang
- Department of Radiation Oncology, Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
107
|
Galangin Suppresses Renal Inflammation via the Inhibition of NF- κB, PI3K/AKT and NLRP3 in Uric Acid Treated NRK-52E Tubular Epithelial Cells. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3018357. [PMID: 31240210 PMCID: PMC6556363 DOI: 10.1155/2019/3018357] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/22/2019] [Accepted: 04/16/2019] [Indexed: 01/10/2023]
Abstract
Renal inflammation can result in renal injury. Uric acid (UA) is the final product of purine metabolism in humans and because of the lack of urate oxidase, UA may accumulate in tissues, including kidney, causing inflammation. Galangin was isolated from a traditional Chinese medicine plant and possesses several beneficial effects, working as an anti-oxidant, anti-mutagenic, anti-tumor, anti-inflammatory, anti-microbial, and anti-viral agent. Therefore, this study aimed at investigating the molecular mechanism of galangin in the attenuation of UA induced renal inflammation in normal rat kidney epithelial cells NRK-52E. Our findings suggested that galangin treatment efficiently protected NRK-52E cells against UA induced renal inflammation by decreasing tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-18, prostaglandin E2 (PGE2), and nitric oxide (NO) release, and it inhibited nitric oxide synthase (iNOS), prostaglandin endoperoxide synthase 2 (PTGS2), TNF-α, IL-1β, and IL-18 mRNA expression. In addition, galangin was not exerting any cytotoxicity at the concentrations that were effective against inflammation as assessed by CCK8 assay. Moreover, western blotting showed that galangin treatment effectively inhibited nuclear factor-kappa B (NF-κB), phosphatidylinositol 3 kinase (PI3K)/protein kinase B (AKT) and nucleotide-binding domain- (NOD-) like receptor protein 3 (NLRP3) signaling pathway activation. Taken together, these findings suggested that galangin plays a pivotal role in renal inflammation by suppressing inflammatory responses, which might be closely associated with the inhibition of NLRP3 inflammasome, NF-κB and PI3K/AKT signaling pathway activation.
Collapse
|
108
|
Shen T, Wang Y, Zhu Z, Wang X, Tian T. Lignans and Terpenoids From the Fruits of Vitex kwangsiensia and Their Inhibitory Activity on Nitric Oxide Production in Macrophages. Nat Prod Commun 2019. [DOI: 10.1177/1934578x19848178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Phytochemical investigation of the fruits from Vitex kwangsiensia led to the isolation of 18 constituents, including 6 lignans (1-6), 3 diterpenoids (7-9), 2 sesquiterpenoids (10, 11), 2 triterpenoids (16, 17), and 5 other compounds. Their structures were identified by spectroscopic methods (mass spectroscopy, infrared, UV, 1-dimensional and 2-dimensional nuclear magnetic resonance) and comparison with reported data in the literatures. Among these constituents, compound 1 was a new lignan and named as vitekwangin A. The spectroscopic data of 2 was reported herein for the first time, and given a trivial name vitekwangin B. The isolated lignans, diterpenoids, and sesquiterpenoids were evaluated for their inhibitory activity to prevent nitric oxide production in lipopolysaccharide-stimulated RAW264.7 macrophages.
Collapse
Affiliation(s)
- Tong Shen
- School of Chemical and Biological Engineering, Lanzhou Jiaotong University, Gansu, People's Republic of China
| | - Yongdong Wang
- School of Chemical and Biological Engineering, Lanzhou Jiaotong University, Gansu, People's Republic of China
| | - Zhiming Zhu
- School of Chemical and Biological Engineering, Lanzhou Jiaotong University, Gansu, People's Republic of China
| | - Xiujie Wang
- Department of Pharmacy, College of Marine Science, Shandong University, Weihai, People's Republic of China
| | - Tian Tian
- School of Chemical and Biological Engineering, Lanzhou Jiaotong University, Gansu, People's Republic of China
| |
Collapse
|
109
|
Craddock JC, Neale EP, Peoples GE, Probst YC. Vegetarian-Based Dietary Patterns and their Relation with Inflammatory and Immune Biomarkers: A Systematic Review and Meta-Analysis. Adv Nutr 2019; 10:433-451. [PMID: 30947338 PMCID: PMC6520040 DOI: 10.1093/advances/nmy103] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/25/2018] [Accepted: 11/01/2018] [Indexed: 12/27/2022] Open
Abstract
Dietary patterns with substantial proportions of energy from plant sources have been associated with favorable biomarkers of low-grade inflammation. Less is known of the relation between vegetarian-based dietary patterns and markers of inflammation and immune status. This systematic review and meta-analysis aimed to determine the relation between vegetarian-based dietary patterns and inflammatory and immune markers (C-reactive protein, tumour necrosis factor α, fibrinogen, natural killer cells, leukocytes, lymphocytes, thrombocytes, interleukins, and immunoglobulins). PubMed, Medline, and Cochrane scientific databases were searched to identify relevant studies. Random effects meta-analyses were conducted to assess the weighted mean differences (WMDs) for each outcome variable between vegetarian and non-vegetarian groups. Thirty observational and 10 intervention studies were included in the review. Pooled effects of vegetarian-based dietary patterns were associated with significantly lower concentrations of CRP (WMD: -0.61 mg/L; 95% CI: -0.91, -0.32 mg/L; P = 0.0001), fibrinogen (WMD: -0.22 g/L; 95% CI: -0.41, -0.04 mg/L; P = 0.02), and total leukocyte (WMD: -0.62 × 10(3)/μL; 95% CI -1.13 × 10(3), -0.10 × 10(3)/μL; P = 0.02) compared with those following non-vegetarian dietary patterns in observational studies. Insufficient data were identified for a meta-analysis of intervention studies. This study provides evidence that vegetarian-based dietary patterns are associated with lowered serum C-reactive protein, fibrinogen, and total leukocyte concentrations. Future research should focus on large-scale intervention trials, contrasting differences in inflammation and immune status and function between vegetarian and non-vegetarian-based populations.
Collapse
Affiliation(s)
- Joel C Craddock
- School of Medicine, Faculty of Science Medicine and Health, University of Wollongong, Wollongong, NSW, Australia,Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia,Address correspondence to JCC (e-mail: )
| | - Elizabeth P Neale
- School of Medicine, Faculty of Science Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Gregory E Peoples
- School of Medicine, Faculty of Science Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Yasmine C Probst
- School of Medicine, Faculty of Science Medicine and Health, University of Wollongong, Wollongong, NSW, Australia,Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
| |
Collapse
|
110
|
Kageyama H, Waditee-Sirisattha R. Antioxidative, Anti-Inflammatory, and Anti-Aging Properties of Mycosporine-Like Amino Acids: Molecular and Cellular Mechanisms in the Protection of Skin-Aging. Mar Drugs 2019; 17:E222. [PMID: 31013795 PMCID: PMC6521297 DOI: 10.3390/md17040222] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/02/2019] [Accepted: 04/10/2019] [Indexed: 12/17/2022] Open
Abstract
Prolonged exposure to ultraviolet (UV) radiation causes photoaging of the skin and induces a number of disorders, including sunburn, fine and coarse wrinkles, and skin cancer risk. Therefore, the application of sunscreen has gained much attention to reduce the harmful effects of UV irradiation on our skin. Recently, there has been a growing demand for the replacement of chemical sunscreens with natural UV-absorbing compounds. Mycosporine-like amino acids (MAAs), promising alternative natural UV-absorbing compounds, are a group of widely distributed, low molecular-weight, water-soluble molecules that can absorb UV radiation and disperse the absorbed energy as heat, without generating reactive oxygen species (ROS). More than 30 MAAs have been characterized, from a variety of organisms. In addition to their UV-absorbing properties, there is substantial evidence that MAAs have the potential to protect against skin aging, including antioxidative activity, anti-inflammatory activity, inhibition of protein-glycation, and inhibition of collagenase activity. This review will provide an overview of MAAs, as potential anti-aging ingredients, beginning with their structure, before moving on to discuss the most recent experimental observations, including the molecular and cellular mechanisms through which MAAs might protect the skin. In particular, we focus on the potential anti-aging activity of mycosporine-2-glycine (M2G).
Collapse
Affiliation(s)
- Hakuto Kageyama
- Department of Chemistry, Faculty of Science and Technology, Meijo University, 1-501 Shiogamaguchi, Tenpaku-ku, Nagoya, Aichi 468-8502, Japan.
| | - Rungaroon Waditee-Sirisattha
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Phayathai Road, Pathumwan, Bangkok 10330, Thailand.
| |
Collapse
|
111
|
Abstract
During sepsis, systemic inflammation is observed and is associated with multiple organ failure. Activation of NF-κB is crucial for inducing inflammation, which is controlled by degradation of inhibitor molecules (IκB). The ubiquitination proteasome pathway is responsible for the regulation of protein turnover. In this study, we hypothesized that administration of 4[4-(5-nitro-furan-2-ylmethylene)-3, -dioxo-pyrazolidin-1-yl]-benzoic acid ethyl ester (PYR-41), an inhibitor of ubiquitination, could reduce inflammation and organ injury in septic mice. PYR-41 prevented the reduction of IκB protein levels and inhibited release of tumor necrosis factor (TNF)-α in mouse macrophage RAW264.7 cells at 4 h after lipopolysaccharide stimulation dose-dependently. Male C57BL/6 mice were subjected to cecal ligation and puncture (CLP) to induce sepsis. PYR-41 (5 mg/kg) or dimethyl sulfoxide in saline (vehicle) was injected intravenously immediately after CLP. At 20 h after CLP, PYR-41 treatment significantly decreased serum levels of proinflammatory cytokines (TNF-α, interleukin [IL]-1β, and IL-6) and organ injury markers (aspartate aminotransferase, alanine aminotransferase, and lactate dehydrogenase). PYR-41 significantly improved microscopic structure, and reduced myeloperoxidase activity, number of apoptotic cells and caspase-3 degradation in the lungs of septic mice. The reduced protein levels of IκB in the lungs after CLP were restored by PYR-41 treatment. PYR-41 inhibited the expression of cytokines (IL-1β and IL-6), chemokines (keratinocyte-derived chemokine and macrophage inflammatory protein 2), and inflammatory mediators (cyclooxygenase-2 and inducible nitric oxide synthase) in the lungs of septic mice. Importantly, PYR-41 significantly increased 10-day survival in septic mice from 42% to 83%. Therefore, targeting ubiquitination by PYR-41 to inhibit NF-κB activation may represent a potential strategy of sepsis therapeutics.
Collapse
|
112
|
Neisi Z, Ansari-Asl Z, Jafarinejad-Farsangi S, Tarzi ME, Sedaghat T, Nobakht V. Synthesis, characterization and biocompatibility of polypyrrole/Cu(II) metal-organic framework nanocomposites. Colloids Surf B Biointerfaces 2019; 178:365-376. [PMID: 30903975 DOI: 10.1016/j.colsurfb.2019.03.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 02/24/2019] [Accepted: 03/14/2019] [Indexed: 11/27/2022]
Abstract
The main objective of composite science is to fabricate new materials with desired properties such as high chemical, mechanical, and/or biological performances. In this research, new conductive nanocomposites of copper metal-organic frameworks (Cu-MOF) and polypyrrole (PPy) were fabricated with the aim of exploiting the electrical conductivity of polypyrrole and the porosity of MOFs in the final products. The prepared compounds (PPy/x%Cu-MOF, x = 20, 50, and 80) were investigated by FTIR, PXRD, SEM, TEM, DLS, BET, EDS mapping, cyclic voltammetry (CV), and zeta potential (ξ) measurements. Spherical morphology was confirmed by SEM and TEM analysis. The PPy/80%Cu-MOF nanocomposite showed the highest ξ potential (-40 mV), demonstrating the stability of dispersed particles. The CV results revealed that the nanocomposites have higher capacitance in comparison to the pure materials. In vitro degradation of the as-prepared compounds in simulated body fluid (SBF) was studied by EIS (electrochemical impedance spectroscopy) and Tafel polarization tests. Furthermore, in vitro biocompatibility of the PPy/x%Cu-MOF composite was evaluated on a group of cells including 3T3 fibroblasts, MCF-7 breast cancer cells, J774.A1 macrophages and red blood cells (RBCs). Viability of 3T3 fibroblasts, MCF-7, and J774.A1 cells, by Methylthiazolyldiphenyl-tetrazolium bromide (MTT) method, was dependent on Cu-MOF percent and amount of composites. Hemolytic assay for RBCs exposed to different amounts of the PPy/x%Cu-MOF composites showed hematological toxicity less than 5% in most concentrations. In addition, to investigate pro-inflammatory activity, J774.A1 macrophages were exposed to non-toxic concentrations of the PPy/x%Cu-MOF and no significant change in the expression of two inflammatory genes COX-2 and iNOS was observed. Injection of the PPy/x%Cu-MOF (5 mg kg-1) into bloodstream of mice did not increase liver damage marker enzymes alanine transaminase (ALT) and aspartate transaminase (AST) level in serum 1 week post injection. Moreover, we observed slight but not significant increase in serum copper level in mice 1 week after injection. According to the results, the PPy/x%Cu-MOF nanocomposites exhibited a good in vitro and in vivo biocompatibility without inducing pro-inflammatory responses in macrophages and show promising potential for different biomedical applications such as biosensors and drug delivery. The release of curcumin from curcumin-loaded PPy/x%Cu-MOF nanocomposites was detectable in plasma of mice 4 days after administration.
Collapse
Affiliation(s)
- Zeinab Neisi
- Department of Chemistry, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Zeinab Ansari-Asl
- Department of Chemistry, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Saeideh Jafarinejad-Farsangi
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mojdeh Esmaeili Tarzi
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Tahereh Sedaghat
- Department of Chemistry, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Valiollah Nobakht
- Department of Chemistry, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
113
|
Abdel-Diam MM, Samak DH, El-Sayed YS, Aleya L, Alarifi S, Alkahtani S. Curcumin and quercetin synergistically attenuate subacute diazinon-induced inflammation and oxidative neurohepatic damage, and acetylcholinesterase inhibition in albino rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:3659-3665. [PMID: 30535736 DOI: 10.1007/s11356-018-3907-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 12/04/2018] [Indexed: 06/09/2023]
Abstract
The ubiquitous use of diazinon (DZN, an organophosphorus insecticide) has increased the probability of occupational, public, and the ecosystem exposure; these exposures are linked to negative health outcomes. The flavonoids curcumin (CUR) and quercetin (QUE) exert significant anti-inflammatory and antioxidant activities against toxicants, including insecticides. However, it is unclear whether their combination enhances these activities. Therefore, 40 albino rat were divided randomly into the CTR, DZN, CUR + DZN, QUE + DZN, and CUR + QUE + DZN groups, which are treated daily via gavage for 28 days. DZN induced neurohepatic inflammation and oxidative damage, which was confirmed by significant (P < 0.05) induction of aspartate and alanine aminotransferases, alkaline phosphatase, lactate dehydrogenase, γ-glutamyl transferase, and tumor necrosis factor-α and inhibition of acetylcholinesterase activity. Furthermore, the liver and brain of DZN-exposed rats exhibited a notable elevation in MDA level paralleled with reduction in antioxidant molecules, i.e., glutathione, superoxide dismutase, glutathione peroxidase, and catalase. The pretreatment of DZN-intoxicated rats with CUR or QUE substantially mitigated neurohepatic dysfunction and inflammation and improved liver and brain antioxidant status with reducing oxidative stress levels. Furthermore, pretreatment with CUR + QUE synergistically restored the neurohepatic dysfunction and oxidative levels to approximately normal levels. The overall results suggested that CUR or QUE inhibits DZN-mediated neurohepatic toxicity via their favorable anti-inflammatory, antioxidant, and free radical-scavenging activities. Moreover, both QUE and CUR may be mutual adjuvant agents against oxidative stress neurohepatic damages.
Collapse
Affiliation(s)
- Mohamed M Abdel-Diam
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt.
| | - Dalia H Samak
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt
| | - Yasser S El-Sayed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, 25030, Besançon Cedex, France
| | - Saud Alarifi
- Department of Zoology, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Saad Alkahtani
- Department of Zoology, Science College, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
114
|
Amini P, Saffar H, Nourani MR, Motevaseli E, Najafi M, Ali Taheri R, Qazvini A. Curcumin Mitigates Radiation-induced Lung Pneumonitis and Fibrosis in Rats. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2019; 7:212-219. [PMID: 31516880 PMCID: PMC6709933 DOI: 10.22088/ijmcm.bums.7.4.212] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/22/2019] [Indexed: 12/18/2022]
Abstract
Radiation-induced lung injury is one of the most prominent factors that interfere with chest cancer radiotherapy, and poses a great threat to patients exposed to total body irradiation. Upregulation of pro-oxidant enzymes is one of the main mechanisms through which the late effects of ionizing radiation on lung injury can be exerted. Interleukin (IL)-4 and IL-13 are two important cytokines that have been proposed to be involved in this process. Through stimulation of dual oxidase 1 and 2 (DUOX 1 & 2), they induce chronic oxidative stress in irradiated tissues. In this study, we evaluated the effects of curcumin treatment on the regulation of IL-4 and IL-13, DUOX1 & 2 genes as well as the pathological changes developed by this treatment. Twenty male Wistar rats were divided into four groups: radiation only; curcumin only; radiation +curcumin; and control group with neither pharmacotherapy nor radiation. Curcumin was administered for 4 and 6 consecutive days before and after irradiation, respectively. Also, the chest area was irradiated with 15 Gy using a cobalt-60 gamma rays source. All rats were sacrificed 67 days after irradiation, followed by the assessment of the levels of IL-4 and IL-13; the expression of IL- 4 receptor-a1 (IL4Ra1), IL13Ra2, DUOX1 and DUOX2, and finally the histopathological changes were evaluated. Radiation led to the increased level of IL-4, while the level of IL-13 showed no change. QPCR results showed the upregulation of IL4Ra1, DUOX1 and DUOX2 following lung irradiation. Histopathological evaluation also showed a remarkable increase in pneumonitis and fibrosis. Treatment with curcumin downregulated the expression of IL-4, IL4Ra1, DUOX1 & 2. Furthermore, it could mitigate pneumonitis and fibrosis following lung irradiation. The late effects of radiation- induced lung injury may be due to the upregulation of DUOX1 & 2 genes. Curcumin, through modulation of these genes, may contribute to the protection against ionizing radiation.
Collapse
Affiliation(s)
- Paiman Amini
- Nanobiotechnology Research Centre, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Hana Saffar
- Clinical and Anatomical Pathology, Tehran University of Medical Science, Imam Khomeini Hospital Complex, Tehran, Iran.
| | - Mohammad Reza Nourani
- Nanobiotechnology Research Centre, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Ramezan Ali Taheri
- Department of Pulmonology, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Ali Qazvini
- Department of Pulmonology, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
115
|
Gonzalez-Perilli L, Prolo C, Álvarez MN. Arachidonic Acid and Nitroarachidonic: Effects on NADPH Oxidase Activity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1127:85-95. [PMID: 31140173 DOI: 10.1007/978-3-030-11488-6_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Arachidonic acid (AA) is a polyunsaturated fatty acid that participates in the inflammatory response mainly through bioactive-lipids formation in macrophages and also in the phagocytic NADPH oxidase 2 (NOX2) activation. NOX2 is the enzyme responsible for a huge superoxide formation in macrophages, essential to eliminate pathogens inside the phagosome. The oxidase is an enzymatic complex comprised of a membrane-bound flavocytochrome b 558 (gp91phox/p22phox), three cytosolic subunits (p47phox, p40phox and p67phox) and a Rac-GTPase. The enzyme becomes active when macrophages are exposed to appropriate stimuli that trigger the phosphorylation of cytosolic subunits and its migration to plasmatic membrane to form the active complex. It is proposed that AA stimulates NOX2 activity through AA interaction with different components of the NADPH oxidase complex. In inflammatory conditions, there is an increase in reactive oxygen and nitrogen species that results in the production of nitrated derivatives of AA, such as nitroarachidonic acid (NO2-AA). NO2-AA is capable to inhibit NOX2 activity by interfering with p47phox migration to the membrane without affecting phosphorylation of cytosolic proteins. Also, NO2-AA is capable to interact with protein disulfide isomerase (PDI), which is involved on NOX2 active complex formation. It has been demonstrated that NO2-AA forms a covalent adduct with PDI that could prevent the interaction with NOX2 and it would explain the inhibitory effects of the fatty acid upon NOX2. Together, current data indicate that AA is an important activator of NOX2 formed in the early events of the inflammatory response, leading to a massive production of oxidants that may, in turn, promote NO2-AA formation and shutting down the oxidative burst. Hence, AA and its derivatives could have antagonistic roles on NOX2 activity regulation.
Collapse
Affiliation(s)
- Lucía Gonzalez-Perilli
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina-Universidad de la República, Montevideo, Uruguay
| | - Carolina Prolo
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina-Universidad de la República, Montevideo, Uruguay
| | - María Noel Álvarez
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina-Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
116
|
|
117
|
Luo Y, Tang H, Li H, Zhao R, Huang Q, Liu J. Recent advances in the development of neuroprotective agents and therapeutic targets in the treatment of cerebral ischemia. Eur J Med Chem 2019; 162:132-146. [DOI: 10.1016/j.ejmech.2018.11.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/30/2018] [Accepted: 11/06/2018] [Indexed: 11/25/2022]
|
118
|
Rezaei M, Ghafouri H, Aghamaali MR, Shourian M. Thiazolidinedione Derivative Suppresses LPS-induced COX-2 Expression and NO Production in RAW 264.7 Macrophages. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2019; 18:1371-1379. [PMID: 32641947 PMCID: PMC6934980 DOI: 10.22037/ijpr.2019.1100730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The present study was designed to investigate the inhibitory effect of 2,4 bis-[(4-ethoxyphenyl)azo] 5-(3-hydroxybenzylidene) thiazolidine-2,4-dione (TZD-OCH2CH3) on the cyclo-oxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) in RAW 264.7 cells. The effects of TZD-OCH2CH3 on COX-2 and iNOS mRNA expression in LPS-activated RAW 264.7 cells were detected by real time PCR. Also, to understand structure and substrate specificity, we have utilized molecular docking simulations (AutoDock Vina) and the active residues in the binding pocket were determined from COX-2 and iNOS. The treatment of RAW 264.7 cells with TZD-OCH2CH3 significantly inhibited LPS-induced COX-2 mRNA expression, corresponding to 46.1% and 61.06% at 30 and 60 μg/mL, respectively. The present study revealed that the TZD-OCH2CH3 had a little effect on iNOS mRNA expression. Meanwhile, the TZD-OCH2CH3 also could inhibit the production of NO compared to single LPS-stimulated cell. According to the results obtained, TZD-OCH2CH3 dramatically suppressed lipopolysaccharide (LPS) induced nitric oxide (NO) production after 24 h, in a concentration-dependent manner with an IC50 of 65 μg/mL. Our data suggest that TZD-OCH2CH3, as a functionally novel agent, inhibits the inflammatory pathway via suppression of COX-2 mRNA expression and also by the inhibition of the iNOS activity. Therefore, this compound could be suggested as a novel therapeutic strategy for inflammation-associated disorders.
Collapse
Affiliation(s)
- Mehrnaz Rezaei
- Department of Biology, Faculty of Science, University of Guilan, University Campus 2, Rasht, Iran.
| | - Hossein Ghafouri
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran.
| | | | - Mostafa Shourian
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran.
| |
Collapse
|
119
|
Shin WB, Dong X, Kim YS, Park JS, Kim SJ, Go EA, Kim EK, Park PJ. Anti-inflammatory Effects of Batillaria multiformis Water Extracts via NF-кB and MAPK Signaling Pathways in LPS-Induced RAW 264.7 Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1155:1001-1014. [PMID: 31468463 DOI: 10.1007/978-981-13-8023-5_83] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Batillaria multiformis (B. multiformis) belong to gastropods. They live generally in the sandpit of the lagoons and the estuaries of the intertidal zone. Most of them are distributed in Korea, Japan and China. In this study, we investigated the anti-inflammatory potential of B. multiformis water extracts (BMW). The results showed that the extracts significantly decreased the production of nitric oxide (NO) and pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in LPS-induced RAW 264.7 macrophages. In addition, the extracts suppressed the protein levels of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in a dose dependent manner. Further investigation indicated that BMW suppressed phosphorylated c-Jun N-terminal kinase (JNK), extracellular regulated protein kinase (ERK) and p38 through the MAPK signaling pathway and influenced the NF-κB signaling pathway by suppressing the IκBα degradation in LPS-induced RAW 264.7 macrophages.
Collapse
Affiliation(s)
- Woen-Bin Shin
- Department of Applied Life Science, Konkuk University, Chungju, Republic of Korea
| | - Xin Dong
- Department of Applied Life Science, Konkuk University, Chungju, Republic of Korea
| | - Yon-Suk Kim
- BK21plus Glocal Education Program of Nutraceuticals Development, Konkuk University, Chungju, Republic of Korea
| | - Jin-Su Park
- Department of Applied Life Science, Konkuk University, Chungju, Republic of Korea
| | - Su-Jin Kim
- Department of Applied Life Science, Konkuk University, Chungju, Republic of Korea
| | - Eun-Ae Go
- Department of Applied Life Science, Konkuk University, Chungju, Republic of Korea
| | - Eun-Kyung Kim
- Division of Food Bio Science, Konkuk University, Chungju, Republic of Korea
| | - Pyo-Jam Park
- Department of Applied Life Science, Konkuk University, Chungju, Republic of Korea. .,Department of Integrated Biosciences, Konkuk University, Chungju, Republic of Korea.
| |
Collapse
|
120
|
Wang C, Song Y, Wang X, Mao R, Song L. Baicalin Ameliorates Collagen-Induced Arthritis Through the Suppression of Janus Kinase 1 (JAK1)/Signal Transducer and Activator of Transcription 3 (STAT3) Signaling in Mice. Med Sci Monit 2018; 24:9213-9222. [PMID: 30562763 PMCID: PMC6322370 DOI: 10.12659/msm.910347] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Rheumatoid arthritis is an autoimmune disease that causes chronic joint inflammation and there is no cure. Baicalin, as an ingredient in the roots of Scutellaria baicalensis, is supposed to possess an anti-inflammatory effect. However, the protective effect of baicalin on collagen-induced arthritis requires further investigation. Material/Methods A model of rheumatoid arthritis was established in 20 mice (8- to 10-weeks old). The mice were randomly divided into 2 groups after modeling and then injected with saline or baicalin, respectively. The synovial fluids and tissues were collected, and the pressure pain threshold and clinical arthritis score were measured. The levels of tumor necrosis factor (TNF)-α, interlukin-1β (IL-1β), IL-6, matrix metalloproteinases (MMP)-2, MMP-9, nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2) and their downstream inflammatory mediators Janus kinase 1 (JAK1)/signal transducer and activator of transcription 3 (STAT3), extracellular signal-regulated kinases 1/2 (ERK1/2), p38, Jun N-terminal kinases (JNK) activation were detected using enzyme-linked immunosorbent assay (ELISA), and western blotting analyses. The mononuclear cells apoptosis ratio was calculated by flowcytometry analyses. Results Baicalin significantly reduced disease activities in a rheumatoid arthritis mouse model, which were reflected by pressure pain thresholds and clinical arthritis scores. Relevant proinflammatory cytokines such as TNF-α, IL-1β, IL-6, gelatinases (MMP-2, MMP-9) and inducible enzymes (iNOS, COX-2) were generally suppressed. Moreover, baicalin treatment induced cells apoptosis in synovial fluid monocytes and markedly down regulated JAK1/STAT3 but not mitogen-activated protein kinases (MAPKs) expressions in synovium of arthritis. Conclusions These observations confirm the relief of rheumatoid arthritis by baicalin. Our results indicate the effect is related with the modulation of decreased proinflammatory cytokines and inflammatory markers. And the apoptosis promotion of monocytes in synovial fluid were also inhibited. Moreover, the molecular mechanism implies suppressed JAK1/STAT3 signaling with baicalin treatment.
Collapse
Affiliation(s)
- Chunxiao Wang
- Department of Rheumatology, Qilu Hospital of Shandong University, Ji'nan, Shandong, China (mainland).,Department of Joint Surgery, Shandong General Team Hospital of Chinese People's Armed Police Force, Ji'nan, Shandong, China (mainland)
| | - Yong Song
- Department of Blood Purification, People's Hospital of Binhai Economic and Technological Development Zone, Weifang, Shandong, China (mainland)
| | - Xin Wang
- Department of Blood Purification, People's Hospital of Binhai Economic and Technological Development Zone, Weifang, Shandong, China (mainland)
| | - Ruijun Mao
- Department of Joint Surgery, Shandong General Team Hospital of Chinese People's Armed Police Force, Ji'nan, Shandong, China (mainland)
| | - Lijun Song
- Department of Rheumatology, Qilu Hospital of Shandong University, Ji'nan, Shandong, China (mainland)
| |
Collapse
|
121
|
Yang B, Guo J, Xiao C. Effect of PM2.5 environmental pollution on rat lung. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:36136-36146. [PMID: 30357727 DOI: 10.1007/s11356-018-3492-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/16/2018] [Indexed: 05/28/2023]
Abstract
Particulate matter smaller than 2.5 μm (PM2.5) is a continuing challenge to pulmonary health. Here, we investigated the mechanisms involved in PM2.5 exposure-induced acute lung injury in rats. We analyzed biochemical and morphological changes following a 2-week "real-world" exposure. And then we found that PM2.5 exposure increased the concentrations of total protein, malondialdehyde, hydrogen peroxide, nitric oxide, and soluble elastin in bronchoalveolar lavage fluid, levels of cytokines in blood, and expression of MMP-9 in airways. Further, alveolar macrophage and neutrophil counts increased following PM2.5 exposure, and edema and lung lesions were observed. Our results suggest that PM2.5 exposure can induce oxidative stress and acute inflammatory responses, which can damage the micro-environment and decrease the repair ability of the lung, resulting in tissue damage.
Collapse
Affiliation(s)
- Biao Yang
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang, People's Republic of China
| | - Jie Guo
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang, People's Republic of China
| | - Chunling Xiao
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang, People's Republic of China.
| |
Collapse
|
122
|
Tarasuntisuk S, Palaga T, Kageyama H, Waditee-Sirisattha R. Mycosporine-2-glycine exerts anti-inflammatory and antioxidant effects in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Arch Biochem Biophys 2018; 662:33-39. [PMID: 30502329 DOI: 10.1016/j.abb.2018.11.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/31/2018] [Accepted: 11/27/2018] [Indexed: 10/27/2022]
Abstract
Mycosporine-like amino acids (MAAs) are a group of water-soluble low-molecular-weight secondary metabolites, which are well-documented UV-screening molecules and antioxidants. We have recently demonstrated that a rare MAA, mycosporine-2-glycine (M2G), efficiently inhibited the formation of advanced glycation end-products (AGEs). Because AGEs contribute significantly to the aging process, including the pathogenesis and progression of age-related diseases, the present study further evaluated anti-inflammatory effects of M2G using an in vitro model of RAW 264.7 macrophages. We measured the inflammatory signaling molecule nitric oxide (NO) under inflammatory stimulation by lipopolysaccharide (LPS), revealing that M2G diminished LPS-induced NO production. M2G inhibited NO production approximately 2-3-fold more potently than other MAAs, including shinorine, porphyra-334, and palythine. Transcriptional analyses revealed that M2G significantly suppressed iNOS and COX-2 expression. Therefore, M2G inhibits the production of inflammatory mediators by suppressing the NF-κB pathway. Furthermore, under H2O2-induced oxidative stress, M2G down-regulated Sod1, Cat, and Nrf2 expression. Our findings clearly demonstrate anti-inflammatory and antioxidant effects of M2G in LPS-stimulated RAW 264.7 macrophages. Structure-activity relationships of biologically active MAAs are also discussed.
Collapse
Affiliation(s)
- Supamate Tarasuntisuk
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand; The Chemical Approaches for Food Applications Research Group, Faculty of Science, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand
| | - Tanapat Palaga
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand
| | - Hakuto Kageyama
- Department of Chemistry, Faculty of Science and Technology, Meijo University, Nagoya, Aichi, 468-8502, Japan; Graduate School of Environmental and Human Sciences, Meijo University, Nagoya, Aichi, 468-8502, Japan.
| | - Rungaroon Waditee-Sirisattha
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand; The Chemical Approaches for Food Applications Research Group, Faculty of Science, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand.
| |
Collapse
|
123
|
Lambertellin from Pycnoporus sanguineus MUCL 51321 and its anti-inflammatory effect via modulation of MAPK and NF-κB signaling pathways. Bioorg Chem 2018; 80:216-222. [DOI: 10.1016/j.bioorg.2018.06.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 02/02/2023]
|
124
|
Ju IG, Choi JG, Kim N, Kwak C, Lee JK, Oh MS. Peucedani Japonici Radix ameliorates lipopolysaccharide-induced neuroinflammation by regulating microglial responses. Neurosci Lett 2018; 686:161-167. [PMID: 30213621 DOI: 10.1016/j.neulet.2018.09.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/01/2018] [Accepted: 09/05/2018] [Indexed: 11/26/2022]
Abstract
Neuroinflammation is an inflammatory process within the central nervous system that is mediated by microglial activation, which releases pro-inflammatory mediators leading to neurodegeneration. In this study, we investigated the effects of Peucedani Japonici Radix (PJR), a medicinal herb traditionally used in East Asia to treat neuroinflammation both in vitro and in vivo. First, we examined the effects of PJR on pro-inflammatory mediators in lipopolysaccharide (LPS)-stimulated BV-2 microglial cells. The results showed that PJR suppressed the LPS-induced increase of several inflammatory factors, such as nitric oxide, inducible nitric oxide synthase, cyclooxygenase-2, prostaglandin E2, interleukin-1β, and tumor necrosis factor-α. We also revealed that PJR inhibited the nuclear factor kappa B (NF-κB) pathway, which is the upstream modulator of inflammatory processes. Furthermore, to confirm the regulatory effects of PJR on microglia in vivo, we measured the number of ionized calcium-binding adapter molecule 1-positive cells in mouse brains and found that PJR treatment reduced microglial activation. Taken together, these results suggest that PJR inhibits microglia-mediated neuroinflammation through the modulation of NF-κB signaling and has the therapeutic potential to prevent inflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- In Gyoung Ju
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Jin Gyu Choi
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Namkwon Kim
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Chaewon Kwak
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Jong Kil Lee
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dozgdaemun-gu, Seoul 02447, Republic of Korea
| | - Myung Sook Oh
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; Department of Oriental Pharmaceutical Science, College of Pharmacy and Kyung Hee East-West Pharmaceutical Research Institute, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| |
Collapse
|
125
|
Huang MY, Tu CE, Wang SC, Hung YL, Su CC, Fang SH, Chen CS, Liu PL, Cheng WC, Huang YW, Li CY. Corylin inhibits LPS-induced inflammatory response and attenuates the activation of NLRP3 inflammasome in microglia. Altern Ther Health Med 2018; 18:221. [PMID: 30107806 PMCID: PMC6092865 DOI: 10.1186/s12906-018-2287-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/12/2018] [Indexed: 02/08/2023]
Abstract
Background Inflammation has been found to be associated with many neurodegenerative diseases, including Parkinson’s and dementia. Attenuation of microglia-induced inflammation is a strategy that impedes the progression of neurodegenerative diseases. Methods We used lipopolysaccharide (LPS) to simulate murine microglia cells (BV2 cells) as an experimental model to mimic the inflammatory environment in the brain. In addition, we examined the anti-inflammatory ability of corylin, a main compound isolated from Psoralea corylifolia L. that is commonly used in Chinese herbal medicine. The production of nitric oxide (NO) by LPS-activated BV2 cells was measured using Griess reaction. The secretion of proinflammatory cytokines including tumor necrosis factor (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6) by LPS-activated BV2 cells was analyzed using enzyme-linked immunosorbent assay (ELISA). The expression of inducible NO synthase (iNOS), cyclooxygenase-2 (COX-2), nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3), apoptosis-associated speck-like protein containing a caspase-activation and recruitment domain (ASC), caspase-1, IL-1β and mitogen-activated protein kinases (MAPKs) in LPS-activated BV2 cells was examined by Western blot. Results Our experimental results demonstrated that corylin suppressed the production of NO and proinflammatory cytokines by LPS-activated BV2 cells. In addition, corylin inhibited the expression of iNOS and COX-2, attenuated the phosphorylation of ERK, JNK and p38, decreased the expression of NLRP3 and ASC, and repressed the activation of caspase-1 and IL-1β by LPS-activated BV2 cells. Conclusion Our results indicate the anti-inflammatory effects of corylin acted through attenuating LPS-induced inflammation and inhibiting the activation of NLRP3 inflammasome in LPS-activated BV2 cells. These results suggest that corylin might have potential in treating brain inflammation and attenuating the progression of neurodegeneration diseases.
Collapse
|
126
|
Lee S, Lee D, Lee JC, Kang KS, Ryoo R, Park HJ, Kim KH. Bioactivity-Guided Isolation of Anti-Inflammatory Constituents of the Rare Mushroom Calvatia nipponica
in LPS-Stimulated RAW264.7 Macrophages. Chem Biodivers 2018; 15:e1800203. [DOI: 10.1002/cbdv.201800203] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/21/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Seulah Lee
- School of Pharmacy; Sungkyunkwan University; Suwon 16419 Korea
| | - Dahae Lee
- School of Pharmacy; Sungkyunkwan University; Suwon 16419 Korea
| | - Joo Chan Lee
- School of Pharmacy; Sungkyunkwan University; Suwon 16419 Korea
| | - Ki Sung Kang
- College of Korean Medicine; Gachon University; Seongnam 13120 Korea
| | - Rhim Ryoo
- Special Forest Products Division; Forest Bioresources Department; National Institute of Forest Science; Suwon 16631 Korea
| | - Hyun-Ju Park
- School of Pharmacy; Sungkyunkwan University; Suwon 16419 Korea
| | - Ki Hyun Kim
- School of Pharmacy; Sungkyunkwan University; Suwon 16419 Korea
| |
Collapse
|
127
|
Jayesh K, Karishma R, Vysakh A, Gopika P, Latha MS. Terminalia bellirica (Gaertn.) Roxb fruit exerts anti-inflammatory effect via regulating arachidonic acid pathway and pro-inflammatory cytokines in lipopolysaccharide-induced RAW 264.7 macrophages. Inflammopharmacology 2018; 28:10.1007/s10787-018-0513-x. [PMID: 30003465 DOI: 10.1007/s10787-018-0513-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/03/2018] [Indexed: 12/31/2022]
Abstract
Terminalia bellirica (Gaertn.) Roxb. (Family: Combretaceae), known as Bhibhitaki in Sanskrit and locally known as Behera in India, has been used for centuries in Ayurveda, a universal system of medicine in India. The dried fruit of T. bellirica is used for the treatment of several disorders. The present study aims to explore the anti-inflammatory effects of aqueous acetone extracts isolated from T. bellirica (AATB) in RAW 264.7 cell lines. The AATB was prepared from the fruits of T. bellirica. Different concentrations of AATB (6.25-100 μg/ml) were used for MTT assay. The anti-inflammatory effect of AATB was evaluated by using different assays such as total cyclooxygenase (COX), 5-lipoxygenase (5-LOX) activity, nitrate and reactive oxygen species (ROS) production. The mRNA level expression of COX-2, tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6) were studied in LPS stimulated RAW 264.7 cells. AATB treatment significantly diminished the elevated levels of inflammatory markers. Moreover, AATB downregulated the mRNA level expression of TNF-α, IL-6 and COX-2 genes. The result of our study suggest the use of AATB and is able to reduce inflammatory conditions associated with various diseases.
Collapse
Affiliation(s)
- Kuriakose Jayesh
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India
| | - Raj Karishma
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India
| | - A Vysakh
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India
| | - Prasad Gopika
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India
| | - M S Latha
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India.
| |
Collapse
|
128
|
Extract of Fructus Schisandrae chinensis Inhibits Neuroinflammation Mediator Production from Microglia via NF-κ B and MAPK Pathways. Chin J Integr Med 2018; 25:131-138. [PMID: 29790065 DOI: 10.1007/s11655-018-3001-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2016] [Indexed: 10/16/2022]
Abstract
OBJECTIVE To investigate the anti-neuroinflammation effect of extract of Fructus Schisandrae chinensis (EFSC) on lipopolysaccharide (LPS)-induced BV-2 cells and the possible involved mechanisms. METHODS Primary cortical neurons were isolated from embryonic (E17-18) cortices of Institute of Cancer Research (ICR) mouse fetuses. Primary microglia and astroglia were isolated from the frontal cortices of newborn ICR mouse. Different cells were cultured in specific culture medium. Cells were divided into 5 groups: control group, LPS group (treated with 1 μg/mL LPS only) and EFSC groups (treated with 1 μg/mL LPS and 100, 200 or 400 mg/mL EFSC, respectively). The effect of EFSC on cells viability was tested by methylthiazolyldiphenyltetrazolium bromide (MTT) colorimetric assay. EFSC-mediated inhibition of LPS-induced production of pro-inflammatory mediators, such as nitrite oxide (NO) and interleukin-6 (IL-6) were quantified and neuron-protection effect against microglia-mediated inflammation injury was tested by hoechst 33258 apoptosis assay and crystal violet staining assay. The expression of pro-inflammatory marker proteins was evaluated by Western blot analysis or immunofluorescence. RESULTS EFSC (200 and 400 mg/mL) reduced NO, IL-6, inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2) expression in LPS-induced BV-2 cells (P<0.01 or P<0.05). EFSC (200 and 400 mg/mL) reduced the expression of NO in LPS-induced primary microglia and astroglia (P<0.01). In addition, EFSC alleviated cell apoptosis and inflammation injury in neurons exposed to microglia-conditioned medium (P<0.01). The mechanistic studies indicated EFSC could suppress nuclear factor (NF)-?B phosphorylation and its nuclear translocation (P<0.01). The anti-inflammatory effect of EFSC occurred through suppressed activation of mitogen-activated protein kinase (MAPK) pathway (P<0.01 or P<0.05). CONCLUSION EFSC acted as an anti-inflammatory agent in LPS-induced glia cells. These effects might be realized through blocking of NF-κB activity and inhibition of MAPK signaling pathways.
Collapse
|
129
|
Choi YK, Ye BR, Kim EA, Kim J, Kim MS, Lee WW, Ahn GN, Kang N, Jung WK, Heo SJ. Bis (3-bromo-4,5-dihydroxybenzyl) ether, a novel bromophenol from the marine red alga Polysiphonia morrowii that suppresses LPS-induced inflammatory response by inhibiting ROS-mediated ERK signaling pathway in RAW 264.7 macrophages. Biomed Pharmacother 2018; 103:1170-1177. [PMID: 29864895 DOI: 10.1016/j.biopha.2018.04.121] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 12/11/2022] Open
Abstract
Inflammation is a pathophysiological defense response against various factors for maintaining homeostasis in the body. However, when continued excessive inflammation becomes chronic, various chronic diseases can develop. Therefore, effective treatment before chronic inflammation development is essential. Bis (3-bromo-4,5-dihydroxybenzyl) ether (BBDE, C14H12Br2O5) is a novel bromophenol isolated from the red alga Polysiphonia morrowii. The beneficial physiological functions of various bromophenols are known, but whether BBDE has beneficial physiological functions is unknown. Therefore, we first investigated whether BBDE exerts any anti-inflammatory effect. We demonstrated that BBDE inhibits inflammation by reducing inflammatory mediators, such as nitric oxide, prostaglandin E2, iNOS, COX2, and pro-inflammatory cytokines (tumor necrosis factor-α, interleukin-1β, and interleukin-6), in LPS-induced macrophage cells. To examine the mechanism of action by which BBDE inhibits inflammation, we confirmed its effect on signal transduction and ROS generation. BBDE selectively inhibited ERK phosphorylation in the mitogen-activated protein kinase pathways. Moreover BBDE suppressed LPS-induced ROS generation in RAW 264.7 macrophage cells. Inhibition of LPS-induced ROS generation by BBDE also caused ERK inactivation and an inflammatory reaction. Therefore, BBDE inhibits LPS-induced inflammation by inhibiting the ROS-mediated ERK signaling pathway in RAW 264.7 macrophage cells and thus can be useful for treating inflammatory diseases.
Collapse
Affiliation(s)
- Youn Kyung Choi
- Jeju International Marine Science Center for Research & Education, Korea Institute of Ocean Science & Technology (KIOST), Jeju 63349, Korea; Department of Medicine, School of Medicine, Jeju National University, 102 Jejudaehakno, Jeju 63243, Korea
| | - Bo-Ram Ye
- Jeju International Marine Science Center for Research & Education, Korea Institute of Ocean Science & Technology (KIOST), Jeju 63349, Korea
| | - Eun-A Kim
- Jeju International Marine Science Center for Research & Education, Korea Institute of Ocean Science & Technology (KIOST), Jeju 63349, Korea
| | - Junseong Kim
- Jeju International Marine Science Center for Research & Education, Korea Institute of Ocean Science & Technology (KIOST), Jeju 63349, Korea
| | - Min-Sun Kim
- Jeju International Marine Science Center for Research & Education, Korea Institute of Ocean Science & Technology (KIOST), Jeju 63349, Korea
| | - Won Woo Lee
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Gin-Nae Ahn
- Department of Marine Bio-food Science, College of Fisheries and Ocean Sciences, Chonnam National University 59626, Republic of Korea
| | - Nalae Kang
- Department of Marine Bio-food Science, College of Fisheries and Ocean Sciences, Chonnam National University 59626, Republic of Korea
| | - Won-Kyo Jung
- Marine-Integrated Bionics Research Center, Pukyong National University, Busan 48513, Republic of Korea
| | - Soo-Jin Heo
- Jeju International Marine Science Center for Research & Education, Korea Institute of Ocean Science & Technology (KIOST), Jeju 63349, Korea; Department of Marine Biology, University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
130
|
Lipoxygenase inhibitors flavonoids from Cyperus rotundus aerial parts. REVISTA BRASILEIRA DE FARMACOGNOSIA-BRAZILIAN JOURNAL OF PHARMACOGNOSY 2018. [DOI: 10.1016/j.bjp.2018.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
131
|
Kim AY, Shim HJ, Shin HM, Lee YJ, Nam H, Kim SY, Youn HS. Andrographolide suppresses TRIF-dependent signaling of toll-like receptors by targeting TBK1. Int Immunopharmacol 2018. [DOI: 10.1016/j.intimp.2018.02.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
132
|
Sáenz J, Alba G, Reyes-Quiroz ME, Geniz I, Jiménez J, Sobrino F, Santa-María C. Curcumin enhances LXRα in an AMP-activated protein kinase-dependent manner in human macrophages. J Nutr Biochem 2018; 54:48-56. [DOI: 10.1016/j.jnutbio.2017.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 09/11/2017] [Accepted: 11/11/2017] [Indexed: 12/19/2022]
|
133
|
Desai SJ, Prickril B, Rasooly A. Mechanisms of Phytonutrient Modulation of Cyclooxygenase-2 (COX-2) and Inflammation Related to Cancer. Nutr Cancer 2018; 70:350-375. [PMID: 29578814 DOI: 10.1080/01635581.2018.1446091] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The link between chronic inflammation and cancer involves cytokines and mediators of inflammatory pathways. Cyclooxygenase-2 (COX-2), a key enzyme in fatty acid metabolism, is upregulated during both inflammation and cancer. COX-2 is induced by pro-inflammatory cytokines at the site of inflammation and enhanced COX-2-induced synthesis of prostaglandins stimulates cancer cell proliferation, promotes angiogenesis, inhibits apoptosis, and increases metastatic potential. As a result, COX-2 inhibitors are a subject of intense research interest toward potential clinical applications. Epidemiological studies highlight the potential benefits of diets rich in phytonutrients for cancer prevention. Plants contain numerous phytonutrient secondary metabolites shown to modulate COX-2. Studies have shown that these metabolites, some of which are used in traditional medicine, can reduce inflammation and carcinogenesis. This review describes the molecular mechanisms by which phytonutrients modulate inflammation, including studies of carotenoids, phenolic compounds, and fatty acids targeting various inflammation-related molecules and pathways associated with cancer. Examples of pathways include those of COX-2, mitogen-activated protein kinase kinase kinase, mitogen-activated protein kinase, pro-inflammatory cytokines, and transcription factors like nuclear factor kappa B. Such phytonutrient modulation of COX-2 and inflammation continue to be explored for applications in the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Shreena J Desai
- a Office of Cancer Complementary and Alternative Medicine , National Cancer Institute , Rockville , Maryland , USA
| | - Ben Prickril
- a Office of Cancer Complementary and Alternative Medicine , National Cancer Institute , Rockville , Maryland , USA
| | - Avraham Rasooly
- a Office of Cancer Complementary and Alternative Medicine , National Cancer Institute , Rockville , Maryland , USA
| |
Collapse
|
134
|
Gallic Acid-L-Leucine Conjugate Protects Mice against LPS-Induced Inflammation and Sepsis via Correcting Proinflammatory Lipid Mediator Profiles and Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1081287. [PMID: 29765489 PMCID: PMC5889890 DOI: 10.1155/2018/1081287] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 12/19/2017] [Accepted: 01/08/2018] [Indexed: 12/20/2022]
Abstract
The pathology of endotoxin LPS-induced sepsis is hallmarked by aberrant production of proinflammatory lipid mediators and nitric oxide (NO). The aim of the present study was to determine whether the new product gallic acid-L-leucine (GAL) conjugate could ameliorate the LPS-induced dysregulation of arachidonic acid metabolism and NO production. We first investigated the effects of GAL conjugate on the expression of proinflammatory enzymes and the production of proinflammatory NO and lipid mediators in mouse macrophage cell line RAW264.7, primary peritoneal macrophages, and mouse model. Western blot analyses revealed that GAL attenuated LPS-induced expression of iNOS, COX-2, and 5-LOX in a concentration-dependent manner. Consistently, probing NO-mediated fluorescence revealed that GAL antagonized the stimulatory effect of LPS on iNOS activity. By profiling of lipid mediators with ESI-MS-based lipidomics, we found that GAL suppressed LPS-induced overproduction of prostaglandin E2, prostaglandin F2, leukotriene B4, and thromboxane B2. We further discovered that GAL might exhibit anti-inflammatory activities by the following mechanisms: (1) suppressing LPS-induced activation of MAP kinases (i.e., ERK1/2, JNK, and p38); (2) reducing the production of reactive oxygen species (ROS); and (3) preventing LPS-induced nuclear translocation of transcription factors NF-κB and AP-1. Consequently, GAL significantly decreased the levels of COX-2 and iNOS expression and the plasma levels of proinflammatory lipid mediators in LPS-treated mice. GAL pretreatment enhanced the survival of mice against LPS-induced endotoxic shock. Taken together, our results suggest that GAL may be a potential anti-inflammatory drug for the treatment of endotoxemia and sepsis.
Collapse
|
135
|
Najafi M, Motevaseli E, Shirazi A, Geraily G, Rezaeyan A, Norouzi F, Rezapoor S, Abdollahi H. Mechanisms of inflammatory responses to radiation and normal tissues toxicity: clinical implications. Int J Radiat Biol 2018; 94:335-356. [DOI: 10.1080/09553002.2018.1440092] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Shirazi
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghazale Geraily
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolhasan Rezaeyan
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Norouzi
- Science and Research Branch, Azad University, Tehran, Iran
| | - Saeed Rezapoor
- Department of Radiology, Faculty of Paramedical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Abdollahi
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
136
|
Murakami A. Non-specific protein modifications may be novel mechanism underlying bioactive phytochemicals. J Clin Biochem Nutr 2018; 62:115-123. [PMID: 29610550 PMCID: PMC5874230 DOI: 10.3164/jcbn.17-113] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 11/27/2017] [Indexed: 12/27/2022] Open
Abstract
In a variety of experimental models, dietary phytochemicals have been demonstrated to exhibit pronounced and versatile bioactivities. Importantly, the possibility of such phytochemicals for human application has been supported in part by epidemiological surveys, which have demonstrated that frequent ingestion of vegetables and fruits containing abundant phytochemicals lowers the risk of onset of various diseases. However, the action mechanisms underlying those dietary phytochemical activities remain to be fully elucidated. For example, even though the anti-oxidant effects of natural polyphenols have long received widespread attention from food scientists, their roles in and contribution to those bioactivities remain controversial because of their poor bioavailability, resulting in extremely low concentrations in the bloodstream. Meanwhile, another important question is why phytochemicals have beneficial effects for animals, including humans, since they are biosynthesized by plants as compounds necessary for adaptation to environmental stress. In regard to that fundamental question, we recently reported novel and unique mechanisms of action of zerumbone, a sesquiterpene with anti-inflammatory and chemopreventive properties. This agent was found to partially exhibit bioactivity through its non-specific interactions with cellular proteins. More strikingly, a non-specific protein binding action of zerumbone was revealed to partially contribute to its anti-inflammatory functions via activation of heat shock factor 1. The present review article highlights and introduces our recent findings regarding the proteo-stress-mediated mechanisms of this phytochemical, along with the concept of hormesis.
Collapse
Affiliation(s)
- Akira Murakami
- Food Hormesis Laboratory, Department of Food Science & Nutrition, School of Human Science & Environment, Research Institute for Food and Nutritional Sciences, University of Hyogo, 1-1-12 Shinzaike-Honcho, Himeji, Hyogo 670-0092, Japan
| |
Collapse
|
137
|
Annamalai G, Suresh K. [6]-Shogaol attenuates inflammation, cell proliferation via modulate NF-κB and AP-1 oncogenic signaling in 7,12-dimethylbenz[a]anthracene induced oral carcinogenesis. Biomed Pharmacother 2018; 98:484-490. [PMID: 29287195 DOI: 10.1016/j.biopha.2017.12.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 11/22/2017] [Accepted: 12/04/2017] [Indexed: 12/17/2022] Open
Abstract
Nuclear factor-kappaB (NF-κB) and activator protein 1 (AP-1) is a major transcription factor which regulates many biological and pathological processes such as inflammation and cell proliferation, which are major implicates in cancer progression. [6]-Shogaol ([6]-SHO) is a major constituent of ginger, exhibits various biological properties such as anti-oxidants, anti-inflammation and anti-tumor. Recently, we proven that [6]-SHO prevents oral squamous cell carcinoma by activating proapoptotic factors in in vitro and in vivo experimental model. However, the preventive efficacy of [6]-SHO in 7,12-dimethylbenz[a]anthracene (DMBA) induced hamster buccal pouch carcinogenesis (HBP) has not been fully elucidated, so far. Hence, we aimed to investigate the effect of [6]-SHO on inflammation and cell proliferation by inhibiting the translocation of NF-κB and AP-1 in DMBA induced HBP carcinogenesis. In this study, we observed upregulation of inflammatory markers (COX-2, iNOS, TNF-α, interleukin-1 and -6), cell proliferative markers (Cyclin D1, PCNA and Ki-67) and aberrant activation of NF-κB, AP-1, IKKβ, c-jun, c-fos and decreased IκB-α in DMBA induced hamsters. Conversely, oral administration of [6]-SHO strongly inhibited constitutive phosphorylation and degradation of IκB and inhibit phosphorylation of c-jun, c-fos, resulting in inhibition of nuclear translocation of NF-κBp65 and AP-1. Thus, inhibition of NF-κB and AP-1 activation by [6]-SHO attenuates inflammation and cell proliferative response in DMBA induced hamsters. Our finding suggested that [6]-SHO is a novel functional agent capable of preventing DMBA induced inflammation and cell proliferation associated tumorigenesis by modulating multiple signalling molecules.
Collapse
Affiliation(s)
- Govindhan Annamalai
- Department of Biochemistry and Biotechnology, Annnamalai University, Annamalainagar, Tamil Nadu, 608 002, India
| | - Kathiresan Suresh
- Department of Biochemistry and Biotechnology, Annnamalai University, Annamalainagar, Tamil Nadu, 608 002, India.
| |
Collapse
|
138
|
Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, Li Y, Wang X, Zhao L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2018; 9:7204-7218. [PMID: 29467962 PMCID: PMC5805548 DOI: 10.18632/oncotarget.23208] [Citation(s) in RCA: 2399] [Impact Index Per Article: 399.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 11/03/2017] [Indexed: 02/07/2023] Open
Abstract
Inflammation is a biological response of the immune system that can be triggered by a variety of factors, including pathogens, damaged cells and toxic compounds. These factors may induce acute and/or chronic inflammatory responses in the heart, pancreas, liver, kidney, lung, brain, intestinal tract and reproductive system, potentially leading to tissue damage or disease. Both infectious and non-infectious agents and cell damage activate inflammatory cells and trigger inflammatory signaling pathways, most commonly the NF-κB, MAPK, and JAK-STAT pathways. Here, we review inflammatory responses within organs, focusing on the etiology of inflammation, inflammatory response mechanisms, resolution of inflammation, and organ-specific inflammatory responses.
Collapse
Affiliation(s)
- Linlin Chen
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
| | - Huidan Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Junliang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Yinglun Li
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Xun Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Ling Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| |
Collapse
|
139
|
Mishra V, Banga J, Silveyra P. Oxidative stress and cellular pathways of asthma and inflammation: Therapeutic strategies and pharmacological targets. Pharmacol Ther 2018; 181:169-182. [PMID: 28842273 PMCID: PMC5743757 DOI: 10.1016/j.pharmthera.2017.08.011] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Asthma is a complex inflammatory disease characterized by airway inflammation and hyperresponsiveness. The mechanisms associated with the development and progression of asthma have been widely studied in multiple populations and animal models, and these have revealed involvement of various cell types and activation of intracellular signaling pathways that result in activation of inflammatory genes. Significant contributions of Toll-like-receptors (TLRs) and transcription factors such as NF-кB, have been reported as major contributors to inflammatory pathways. These have also recently been associated with mechanisms of oxidative biology. This is of important clinical significance as the observed inefficacy of current available treatments for severe asthma is widely attributed to oxidative stress. Therefore, targeting oxidizing molecules in conjunction with inflammatory mediators and transcription factors may present a novel therapeutic strategy for asthma. In this review, we summarize TLRs and NF-кB pathways in the context of exacerbation of asthma pathogenesis and oxidative biology, and we discuss the potential use of polyphenolic flavonoid compounds, known to target these pathways and possess antioxidant activity, as potential therapeutic agents for asthma.
Collapse
Affiliation(s)
- Vikas Mishra
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Departments of Pediatrics, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Jaspreet Banga
- The Feinstein Institute for Medical Research, Center for Autoimmune and Musculoskeletal Diseases, Manhasset, NY, USA
| | - Patricia Silveyra
- Departments of Pediatrics, The Pennsylvania State University, College of Medicine, Hershey, PA, USA; Biochemistry and Molecular Biology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.
| |
Collapse
|
140
|
Kim YS, Shin WB, Dong X, Kim EK, Nawarathna WPAS, Kim H, Park PJ. Anti-inflammatory effect of the extract from fermented Asterina pectinifera with Cordyceps militaris mycelia in LPS-induced RAW264.7 macrophages. Food Sci Biotechnol 2017; 26:1633-1640. [PMID: 30263700 PMCID: PMC6049704 DOI: 10.1007/s10068-017-0233-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/27/2017] [Accepted: 08/10/2017] [Indexed: 12/29/2022] Open
Abstract
In our previous work, Asterina pectinifera was fermented with Cordyceps militaris mycelia to improve its bioactivities and was reported to have strong antioxidant activities. The aim of the current study was to investigate its anti-inflammatory effect and mechanisms of action. In this study, we observed the inhibitory effect of the extract from fermented A. pectinifera with C. militaris mycelia (FACM) on nitric oxide (NO) production and its molecular mechanism in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. FACM could decrease LPS-induced NO production. Western blot analysis showed that FACM could down-regulate LPS-induced expression of inducible NO synthase without affecting cyclooxygenase-2. Moreover, FACM exhibited anti-inflammatory activity in LPS-induced RAW264.7 mouse macrophage cells through proinflammatory mediators including TNF-α and IL-6 via nuclear factor kappa B pathway. FACM inhibited LPS-induced phosphorylation of extracellular-signal-regulated kinase expression. Our results suggest that FACM may be a potential candidate for inflammation therapy by attenuating the generation of cytokines, production of NO, and generation of ROS in RAW264.7 cells.
Collapse
Affiliation(s)
- Yon-Suk Kim
- Department of Biotechnology, Konkuk University, Chungju, Chungbuk 27478 Korea
- Nokyong Research Center, Konkuk University, Chungju, Chungbuk 27478 Korea
| | - Woen-Bin Shin
- Department of Biotechnology, Konkuk University, Chungju, Chungbuk 27478 Korea
| | - Xin Dong
- Department of Biotechnology, Konkuk University, Chungju, Chungbuk 27478 Korea
| | - Eun-Kyung Kim
- Division of Food and Bio Science, Konkuk University, Chungju, 27478 Korea
| | | | - Hakju Kim
- Seojin Biotech Co., Ltd., Yongin, Gyeonggi 17015 Korea
| | - Pyo-Jam Park
- Department of Biotechnology, Konkuk University, Chungju, Chungbuk 27478 Korea
- Nokyong Research Center, Konkuk University, Chungju, Chungbuk 27478 Korea
| |
Collapse
|
141
|
Lin CY, Wang WH, Chen SH, Chang YW, Hung LC, Chen CY, Chen YH. Lipopolysaccharide-Induced Nitric Oxide, Prostaglandin E2, and Cytokine Production of Mouse and Human Macrophages Are Suppressed by Pheophytin-b. Int J Mol Sci 2017; 18:ijms18122637. [PMID: 29211014 PMCID: PMC5751240 DOI: 10.3390/ijms18122637] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/29/2017] [Accepted: 12/04/2017] [Indexed: 02/06/2023] Open
Abstract
Sepsis is an overwhelming systemic response to infection that frequently results in tissue damage, organ failure, and even death. Nitric oxide (NO), prostaglandin E2 (PGE2), and cytokine overproduction are thought to be associated with the immunostimulatory cascade in sepsis. In the present study, we analyzed the anti-inflammatory efficacy of the pheophytin-b on both RAW 264.7 murine macrophage and purified human CD14+ monocytes stimulated with lipopolysaccharide (LPS) and elucidated the mechanisms by analyzing the cell signaling pathways known to be activated in sepsis. Pheophytin-b suppressed the overexpression of NO, PGE2, and cytokines in LPS-stimulated macrophages without inducing cytotoxicity. It also reduced NOS2 and COX-2 mRNA and protein levels. The inhibitory effects on NO, PGE2, and cytokine overproduction arose from the suppression of STAT-1 and PI3K/Akt pathways; no changes in NF-κB, MAPK, and AP-1 signaling were detected. Thus, pheophytin-b may represent a potential candidate to beneficially modulate the inflammatory response in sepsis.
Collapse
Affiliation(s)
- Chun-Yu Lin
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Sepsis Research Center, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Wen-Hung Wang
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Shin-Huei Chen
- Sepsis Research Center, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Yu-Wei Chang
- Sepsis Research Center, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Ling-Chien Hung
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chung-Yi Chen
- School of Medical and Health Sciences, Fooyin University, Kaohsiung 831, Taiwan.
| | - Yen-Hsu Chen
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Sepsis Research Center, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu 300, Taiwan.
| |
Collapse
|
142
|
Shin HM, Shim HJ, Kim AY, Lee YJ, Youn HS. Differential modulation of toll-like receptor agonists-induced iNOS expression by polyunsaturated and saturated fatty acids. FOOD AGR IMMUNOL 2017. [DOI: 10.1080/09540105.2017.1326468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Hyeon-Myeong Shin
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam 31538, Republic of Korea
| | - Hyun-Jin Shim
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam 31538, Republic of Korea
| | - Ah-Yeon Kim
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam 31538, Republic of Korea
| | - Yoo Jung Lee
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam 31538, Republic of Korea
| | - Hyung-Sun Youn
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam 31538, Republic of Korea
| |
Collapse
|
143
|
Kim YY, Lee S, Kim MJ, Kang BC, Dhakal H, Choi YA, Park PH, Choi H, Shin TY, Choi HG, Kwon TK, Khang D, Kim SH. Tyrosol attenuates lipopolysaccharide-induced acute lung injury by inhibiting the inflammatory response and maintaining the alveolar capillary barrier. Food Chem Toxicol 2017; 109:526-533. [DOI: 10.1016/j.fct.2017.09.053] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 09/19/2017] [Accepted: 09/29/2017] [Indexed: 01/07/2023]
|
144
|
Kwon DH, Cha HJ, Choi EO, Leem SH, Kim GY, Moon SK, Chang YC, Yun SJ, Hwang HJ, Kim BW, Kim WJ, Choi YH. Schisandrin A suppresses lipopolysaccharide-induced inflammation and oxidative stress in RAW 264.7 macrophages by suppressing the NF-κB, MAPKs and PI3K/Akt pathways and activating Nrf2/HO-1 signaling. Int J Mol Med 2017; 41:264-274. [PMID: 29115385 PMCID: PMC5746320 DOI: 10.3892/ijmm.2017.3209] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 10/19/2017] [Indexed: 12/15/2022] Open
Abstract
Schisandrin A is a bioactive lignan occurring in the fruits of plants of the Schisandra genus that have traditionally been used in Korea for treating various inflammatory diseases. Although the anti-inflammatory and antioxidant effects of lignan analogues similar to schisandrin A have been reported, the underlying molecular mechanisms have remained elusive. In the present study, schisandrin A significantly suppressed the lipopolysaccharide (LPS)-induced production of the key pro-inflammatory mediators nitric oxide (NO) and prostaglandin E2 by suppressing the expression of inducible NO synthase and cyclooxygenase-2 at the mRNA and protein levels in RAW 264.7 macrophages. Furthermore, schisandrin A was demonstrated to reduce the LPS-induced secretion of pro-inflammatory cytokines, including tumor necrosis factor-α and interleukin-1β; this was accompanied by a simultaneous decrease in the respective mRNA and protein levels in the macrophages. In addition, the LPS- induced translocation of nuclear factor-κB (NF-κB), as well as activation of mitogen-activated protein kinases (MAPKs) and phosphatidylinositol‑3 kinase (PI3K)/Akt pathways were inhibited by schisandrin A. Furthermore, schisandrin A significantly diminished the LPS-stimulated accumulation of intracellular reactive oxygen species, and effectively enhanced the expression of NF erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). These results suggested that schisandrin A has a protective effect against LPS-induced inflammatory and oxidative responses in RAW 264.7 cells by inhibiting the NF-κB, MAPK and PI3K/Akt pathways; these effects are mediated, at least in part, by the activation of the Nrf2/HO-1 pathway. Based on these results, it is concluded that schisandrin A may have therapeutic potential for treating inflammatory and oxidative disorders caused by over-activation of macrophages.
Collapse
Affiliation(s)
- Da Hye Kwon
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Republic of Korea
| | - Eun Ok Choi
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Sun-Hee Leem
- Department of Biological Science, College of Natural Sciences, Dong-A University, Busan 49315, Republic of Korea
| | - Gi-Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, College of Biotechnology and Natural Resource, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Young-Chae Chang
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu 42472, Republic of Korea
| | - Seok-Joong Yun
- Personalized Tumor Engineering Research Center, Department of Urology, Chungbuk National University College of Medicine, Cheongju 28644, Republic of Korea
| | - Hye Jin Hwang
- Department of Food and Nutrition, College of Nursing, Healthcare Sciences and Human Ecology, Dongeui University, Busan 47340, Republic of Korea
| | - Byung Woo Kim
- Department of Life Science and Biotechnology, College of Engineering, Dongeui University, Busan 47340, Republic of Korea
| | - Wun-Jae Kim
- Personalized Tumor Engineering Research Center, Department of Urology, Chungbuk National University College of Medicine, Cheongju 28644, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan 47227, Republic of Korea
| |
Collapse
|
145
|
Zhang X, Li W, Abudureheman A, Cheng T, Peng P. Imperatorin possesses notable anti‑inflammatory activity in vitro and in vivo through inhibition of the NF‑κB pathway. Mol Med Rep 2017; 16:8619-8626. [PMID: 28990061 PMCID: PMC5779915 DOI: 10.3892/mmr.2017.7706] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 05/18/2017] [Indexed: 11/24/2022] Open
Abstract
Imperatorin (IMT) is a furanocoumarin from the root of Phlomis younghusbandii (Lamiaceae) with various activities. In the present study, the anti-inflammatory effects of IMT were evaluated by examining dimethylbenzene-induced ear edema, acetic acid-induced vascular permeability and by performing cotton pellet granuloma assessments in mice. In addition, the expression of pro-inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β, were detected using enzyme-linked immunosorbent assay kits in mice and using reverse transcription polymerase chain reaction analysis in RAW 264.7 cells. The expression levels of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), nuclear p65, cytosolic p65 and inhibitor of nuclear factor (NF)-κB (IκB) in RAW 264.7 cells were determined using western blot analysis. The results showed that the oral administration of IMT significantly inhibited the inflammatory reactions and reduced the release of TNF-α, IL-6 and IL-1β reactions and reduced and suppressed the mRNA expression of TNF-A expressionact1o, and the protein expression of iNOS and COX-2 in the RAW 264.7 cells. The results also indicated that IMT suppressed the activity of NF-κB via upregulating p65 and IκB in the cytoplasm and downregulating p65 in the nucleus. In conclusion, IMT possessed notable anti-inflammatory activities in vitro and in vivo through inhibiting the NF-κB pathway.
Collapse
Affiliation(s)
- Xiaoxia Zhang
- Department of Emergency Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Wenchao Li
- Department of Emergency Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Aikebaier Abudureheman
- Department of Emergency Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Tao Cheng
- Department of Emergency Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Peng Peng
- Department of Emergency Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| |
Collapse
|
146
|
Vanamala JKP, Massey AR, Pinnamaneni SR, Reddivari L, Reardon KF. Grain and sweet sorghum (Sorghum bicolor L. Moench) serves as a novel source of bioactive compounds for human health. Crit Rev Food Sci Nutr 2017; 58:2867-2881. [PMID: 28662339 DOI: 10.1080/10408398.2017.1344186] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Grain sorghum is an important staple food crop grown globally while sweet sorghum is increasingly considered as a promising biofuel feedstock. Biofuels are the major economic products from the processing of large quantities of biomass, which is currently being utilized to make value-added products in the biorefinery approach. To date, these value-added products are typically commodity chemicals and waste materials used in agriculture. However, there are opportunities to generate high-value bioactive compounds from sorghum grain and biomass. Chronic diseases, such as cancers, are the top causes for morbidity and mortality in developed nations and are promoted by inflammation and oxidative stress. Globally, colorectal cancer results in approximately one-half million deaths annually. It is estimated that as much as 80% of colorectal cancer cases can be attributed to environmental and dietary factors. The sorghum grain and ligno-cellulosic biomass generated for biofuel production has been reported to be high in bioactive compounds, including phenolic acids and flavonoids, with antioxidant and anti-inflammatory properties. This review focuses on the bioactive compounds of grain and sweet sorghum (Sorghum bicolor L. Moench), for their anti-inflammatory, antioxidant, anti-colon cancer, and immune modulator functions. The review summarizes previous efforts to identify and quantify bioactive compounds in sorghum and documents their anti-cancer biological activities. Finally, this review discusses bioactive compound extraction methodologies and technologies as well as considerations for incorporating these technologies into current biorefining practices.
Collapse
Affiliation(s)
- Jairam K P Vanamala
- a Food Science , Pennsylvania State University , University Park , Pennsylvania , USA
| | - Aaron R Massey
- b Colorado State University , Fort Collins , Colorado , USA
| | | | - Lavanya Reddivari
- d Life Sciences , Pennsylvania State University , University Park , Pennsylvania , USA
| | - Kenneth F Reardon
- e Chemical and Biological Engineering , Colorado State University , Fort Collins , Colorado , USA
| |
Collapse
|
147
|
Zhang G, Zhao L, Zhu J, Feng Y, Wu X. Anti-inflammatory activities and glycerophospholipids metabolism in KLA-stimulated RAW 264.7 macrophage cells by diarylheptanoids from the rhizomes ofAlpinia officinarum. Biomed Chromatogr 2017; 32. [DOI: 10.1002/bmc.4094] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 08/31/2017] [Accepted: 09/07/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Guogai Zhang
- Central Laboratory; Guangdong Pharmaceutical University; Guangzhou Guangdong China
| | - Lifang Zhao
- Qingdao Yellow Sea Pharmaceutical Co. Ltd; Qingdao Shandong China
| | - Jiancheng Zhu
- Central Laboratory; Guangdong Pharmaceutical University; Guangzhou Guangdong China
| | - Yifan Feng
- Central Laboratory; Guangdong Pharmaceutical University; Guangzhou Guangdong China
| | - Xia Wu
- Central Laboratory; Guangdong Pharmaceutical University; Guangzhou Guangdong China
| |
Collapse
|
148
|
Meram C, Wu J. Anti-inflammatory effects of egg yolk livetins (α, β, and γ-livetin) fraction and its enzymatic hydrolysates in lipopolysaccharide-induced RAW 264.7 macrophages. Food Res Int 2017; 100:449-459. [DOI: 10.1016/j.foodres.2017.07.032] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/15/2017] [Accepted: 07/16/2017] [Indexed: 01/09/2023]
|
149
|
Inhibition of LPS induced pro-inflammatory responses in RAW 264.7 macrophage cells by PVP-coated naringenin nanoparticle via down regulation of NF-κB/P38MAPK mediated stress signaling. Pharmacol Rep 2017. [DOI: 10.1016/j.pharep.2017.04.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
150
|
Licochalcone A Prevents the Loss of Dopaminergic Neurons by Inhibiting Microglial Activation in Lipopolysaccharide (LPS)-Induced Parkinson's Disease Models. Int J Mol Sci 2017; 18:ijms18102043. [PMID: 28937602 PMCID: PMC5666725 DOI: 10.3390/ijms18102043] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/14/2017] [Accepted: 09/19/2017] [Indexed: 01/12/2023] Open
Abstract
The neuroprotective effects of Licochalcone A (Lico.A), a flavonoid isolated from the herb licorice, in Parkinson's disease (PD) have not been elucidated. The prominent pathological feature of PD is the loss of dopaminergic neurons. The crucial role of neuroinflammation induced by activated microglia in dopaminergic neurodegeneration has been validated. In this study, we explore the therapeutic effects of Lico.A in lipopolysaccharide (LPS)-induced PD models in vivo and in vitro. We find that Lico.A significantly inhibits LPS-stimulated production of pro-inflammatory mediators and microglial activation by blocking the phosphorylation of extracellular signal-regulated kinase (ERK1/2) and nuclear factor κB (NF-κB) p65 in BV-2 cells. In addition, through cultured primary mesencephalic neuron-glia cell experiments, we illustrate that Lico.A attenuates the decrease in [³H] dopamine (DA) uptake and the loss of tyrosine hydroxylase-immunoreactive (TH-ir) neurons in LPS-induced PD models in vitro. Furthermore, LPS intoxication in rats results in microglial activation, dopaminergic neurodegeneration and significant behavioral deficits in vivo. Lico.A treatment prevents microglial activation and reduction of dopaminergic neuron and ameliorates PD-like behavioral impairments. Thus, these results demonstrate for the first time that the neuroprotective effects of Lico.A are associated with microglia and anti-inflammatory effects in PD models.
Collapse
|