1
|
Balasubramaniam M, Sapuan S, Hashim IF, Ismail NI, Yaakop AS, Kamaruzaman NA, Ahmad Mokhtar AM. The properties and mechanism of action of plant immunomodulators in regulation of immune response - A narrative review focusing on Curcuma longa L. , Panax ginseng C. A. Meyer and Moringa oleifera Lam. Heliyon 2024; 10:e28261. [PMID: 38586374 PMCID: PMC10998053 DOI: 10.1016/j.heliyon.2024.e28261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 04/09/2024] Open
Abstract
Herbal treatments have been utilized for millennia to cure a variety of ailments. There are over 20, 000 herbal remedies available to treat cancer and other disease in humans. In Ayurveda, traditional plants having revitalizing and nourishing characteristics are known as "Rasayanas." They have anti-inflammatory, anticancer, anti-microbicidal, antiviral, and immunomodulatory effects on the immune system. Immunomodulation is a mechanism through which the body stimulates, suppresses, or boosts the immune system to maintain homeostasis. Plant-derived immunomodulators are typically phytocompounds, including carbohydrates, phenolics, lipids, alkaloids, terpenoids, organosulfur, and nitrogen-containing chemicals. Immunomodulation activity of phytocompounds from traditional plants is primarily mediated through macrophage activation, phagocytosis stimulation, peritoneal macrophage stimulation, lymphoid cell stimulation, and suppression or enhancement of specific and non-specific cellular immune systems via numerous signalling pathways. Despite extensive research, the precise mechanism of immunomodulation of most traditional plants has not yet been fully elucidated, justifying the need for further experimentation. Therefore, this review describes the immunomodulatory agents from traditional plants such as Curcuma longa L., Panax ginseng C.A. Meyer, and Moringa oleifera Lam, further highlighting the common molecular targets and immunomodulatory mechanism involved in eradicating diseases.
Collapse
Affiliation(s)
- Muggunna Balasubramaniam
- Small G protein Research Group, Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia
| | - Sarah Sapuan
- Department of Toxicology, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Kepala Batas, Penang, Malaysia
| | - Ilie Fadzilah Hashim
- Department of Clinical Medicine, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Kepala Batas, Penang, Malaysia
| | - Nurul Izza Ismail
- School of Biological Sciences, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia
| | - Amira Suriaty Yaakop
- School of Biological Sciences, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia
| | | | - Ana Masara Ahmad Mokhtar
- Small G protein Research Group, Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia
- Green Biopolymer Coating and Packaging Centre, School of Industrial Technology, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia
| |
Collapse
|
2
|
Lee C, Lee S, Jang YP, Park J. Anti-Inflammatory Activity of Vacuum Distillate from Panax ginseng Root on LPS-Induced RAW264.7 Cells. J Microbiol Biotechnol 2024; 34:262-269. [PMID: 38213284 PMCID: PMC10940780 DOI: 10.4014/jmb.2312.12001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 12/19/2023] [Accepted: 01/05/2024] [Indexed: 01/13/2024]
Abstract
Panax ginseng has been widely applied as an important herb in traditional medicine to treat numerous human disorders. However, the inflammatory regulation effect of P. ginseng distillate (GSD) has not yet been fully assessed. To determine whether GSD can ameliorate inflammatory processes, a GSD was prepared using the vacuum distillation process for the first time, and the regulation effect on lipopolysaccharide-induced macrophages was assessed. The results showed that GSD effectively inhibited nitric oxide (NO) formation and activation of inducible nitric oxide synthase (iNOS) mRNA in murine macrophage cell, but not cyclooxygenase-2 production. The mRNA expression pattern of tumor necrosis factor alpha and IL-6 were also reduced by GSD. Furthermore, we confirmed that GSD exerted its anti-inflammatory effects by downregulating c-Jun NH2-terminal kinase (JNK) phosphorylation, the extracellular signal-regulated kinase phosphorylation, and signaling pathway of nuclear factor kappa B (NF-κB). Our findings revealed that the inflammatory regulation activity of GSD could be induced by iNOS and NO formation inhibition mediated by regulation of nuclear factor kappa B and p38/JNK MAPK pathways.
Collapse
Affiliation(s)
- Chanwoo Lee
- College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seul Lee
- Department of Engineering Chemistry, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Young Pyo Jang
- College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Junseong Park
- Department of Engineering Chemistry, Chungbuk National University, Cheongju 28644, Republic of Korea
| |
Collapse
|
3
|
Kingkaew E, Konno H, Hosaka Y, Tanasupawat S. Probiogenomic Analysis of Lactiplantibacillus sp. LM14-2 from Fermented Mussel (Hoi-dong), and Evaluation of its Cholesterol-lowering and Immunomodulation Effects. Probiotics Antimicrob Proteins 2023; 15:1206-1220. [PMID: 35987935 DOI: 10.1007/s12602-022-09977-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2022] [Indexed: 02/01/2023]
Abstract
Lactiplantibacillus sp. LM14-2, isolated from Thai-fermented mussel (Hoi-dong), showed attractive probiotic properties. This strain was identified as Lactiplantibacillus plantarum based on its phenotypic, chemotaxonomic, and genetic characteristics including whole-genome sequencing (WGS). The draft genome sequence was analyzed and annotated for the molecular mechanisms involved in the safety assessment, the adaptation and adhesion of L. plantarum LM14-2 to the gastrointestinal tract (GIT), and the beneficial genes involved in bacteria-host interactions. The L. plantarum LM14-2 exhibited bile salt hydrolase (BSH) activity, assimilated cholesterol at 86.07 ± 5.03%, stimulated the secretion of interleukin-12, interferon-gamma, and human beta defensin-2, and induced nitric oxide production. In addition, L. plantarum LM14-2 showed excellent gastrointestinal tolerance and adhesion ability to Caco-2 cells. Furthermore, the in silico analysis showed that L. plantarum LM14-2 was a non-human pathogen and did not contain antibiotic resistance genes or plasmids. L. plantarum LM14-2 also contained potential genes associated with various probiotic characteristics and health-promoting effects. Consequently, this study suggested that L. plantarum LM14-2 could be considered safe, with potential probiotic properties and health-promoting impacts, which could facilitate its probiotic application.
Collapse
Affiliation(s)
- Engkarat Kingkaew
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Hiroshi Konno
- Akita Konno CO., LTD., 248 Aza Kariwano, Daisen-shi, Akita, 019-2112, Japan
| | - Yoshihito Hosaka
- Akita Konno CO., LTD., 248 Aza Kariwano, Daisen-shi, Akita, 019-2112, Japan
| | - Somboon Tanasupawat
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
4
|
Kingkaew E, Konno H, Hosaka Y, Phongsopitanun W, Tanasupawat S. Characterization of Lactic Acid Bacteria from Fermented Fish (pla-paeng-daeng) and Their Cholesterol-lowering and Immunomodulatory Effects. Microbes Environ 2023; 38. [PMID: 36754424 PMCID: PMC10037097 DOI: 10.1264/jsme2.me22044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
The cholesterol-lowering and immunomodulatory effects and probiotic properties of 25 lactic acid bacteria (LAB) isolated from fermented fish (pla-paeng-daeng) in Thailand were examined in the present study. Based on their phenotypic and genetic characteristics, LAB were identified as Lactiplantibacillus pentosus (Group I, 6 isolates), Lactiplantibacillus argentoratensis (Group II, 1 isolate), Limosilactobacillus fermentum (Group III, 2 isolates), Companilactobacillus pabuli (Group IV, 4 isolates), Companilactobacillus farciminis (Group V, 5 isolates), Companilactobacillus futsaii (Group VI, 6 isolates), and Enterococcus lactis (Group VII, 1 isolate). Lactiplantibacillus pentosus PD3-1 and PD9-2 and Enterococcus lactis PD3-2 exhibited bile salt hydrolase (BSH) activities. The percentage of cholesterol assimilated by all isolates ranged between 21.40 and 54.07%. Bile salt hydrolase-producing isolates tolerated acidic and bile conditions and possessed adhesion properties. They also exerted immunomodulatory effects that affected the production of interleukin-12 (IL-12), interferon-γ (IFN-γ), human β-defensin-2 (hBD-2), and nitric oxide (NO). These isolates meet standard probiotic requirements and exert beneficial effects.
Collapse
Affiliation(s)
- Engkarat Kingkaew
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University
| | | | | | - Wongsakorn Phongsopitanun
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University
| | - Somboon Tanasupawat
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University
| |
Collapse
|
5
|
Cao MY, Wu J, Xie CQ, Wu L, Gu Z, Hu JW, Xiong W. Antioxidant and anti-inflammatory activities of Gynura procumbens flowers extract through suppressing LPS-induced MAPK/NF-κB signalling pathways. FOOD AGR IMMUNOL 2022. [DOI: 10.1080/09540105.2022.2098935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Ming-Yuan Cao
- Institute of Applied Chemistry, Jiangxi Academy of Sciences, Nanchang, People’s Republic of China
- School of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, People’s Republic of China
| | - Jing Wu
- Institute of Applied Chemistry, Jiangxi Academy of Sciences, Nanchang, People’s Republic of China
| | - Chuan-Qi Xie
- Institute of Applied Chemistry, Jiangxi Academy of Sciences, Nanchang, People’s Republic of China
| | - Lei Wu
- Institute of Applied Chemistry, Jiangxi Academy of Sciences, Nanchang, People’s Republic of China
| | - Zhen Gu
- Institute of Applied Chemistry, Jiangxi Academy of Sciences, Nanchang, People’s Republic of China
| | - Ju-Wu Hu
- Institute of Applied Chemistry, Jiangxi Academy of Sciences, Nanchang, People’s Republic of China
- School of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, People’s Republic of China
| | - Wei Xiong
- Institute of Applied Chemistry, Jiangxi Academy of Sciences, Nanchang, People’s Republic of China
| |
Collapse
|
6
|
Kingkaew E, Konno H, Hosaka Y, Phongsopitanun W, Tanasupawat S. Distribution, cholesterol-lowering and immunomodulation effects of lactic acid bacteria from fermented mussel ( Hoi-dong). Heliyon 2022; 8:e12272. [PMID: 36590538 PMCID: PMC9800316 DOI: 10.1016/j.heliyon.2022.e12272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/22/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
Forty-eight lactic acid bacteria (LAB) isolated from fermented mussels in Thailand were evaluated for their probiotic properties, bile salt hydrolase (BSH), cholesterol assimilation and immunomodulatory effects. They were identified as Companilactobacillus formosensis (Group I, 10 isolates), Lentilactobacillus buchneri (Group II, 8 isolates), Lactiplantibacillus plantarum subsp. plantarum (Group III, 16 isolates), Lacticaseibacillus rhamnosus (Group IV, 1 isolate), Pediococcus pentosaceus (Group V, 5 isolates) and P. acidilactici (Group V, 1 isolate), Enterococcus thailandicus (Group VI, 2 isolates), En. hirae (Group VII, 1 isolate), En. durans (Group VI, 1 isolate), Lactococcus lactis subsp. lactis (Group VII, 1 isolate), Lc. lactis subsp. hordinae (Group VII, 1 isolate), and Leuconostoc lactis (Group VIII, 1 isolate), based on their phenotypic and genetic characteristics. Seven isolates, L. plantarum subsp. plantarum LM6-1, LM6-2, LM7-2-2B, LM12-1, LM14-1, LM15-1P and LM15-2 expressed bile salt hydrolase activity. All isolates assimilated cholesterol ranging from 20.73 to 79.40%. BSH-producing isolates were tolerant to acidic and bile conditions and showed the adhesion ability to Caco-2 cells. The BSH-producing and selected isolates showed the immunomodulatory effects to stimulate interleukin-12 (IL-12), interferon-gamma (IFN-γ), human beta defensin-2 (hBD-2) and nitric oxide (NO) production at various levels. Therefore, these results indicated that the isolates meet the standard probiotic criteria and beneficial effects.
Collapse
Affiliation(s)
- Engkarat Kingkaew
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Hiroshi Konno
- Akita Konno CO., LTD., 248 Aza Kariwano, Daisen-shi, Akita 019-2112, Japan
| | - Yoshihito Hosaka
- Akita Konno CO., LTD., 248 Aza Kariwano, Daisen-shi, Akita 019-2112, Japan
| | - Wongsakorn Phongsopitanun
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Somboon Tanasupawat
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand,Corresponding author.
| |
Collapse
|
7
|
Advances in the Bioactivities of Phytochemical Saponins in the Prevention and Treatment of Atherosclerosis. Nutrients 2022; 14:nu14234998. [PMID: 36501028 PMCID: PMC9735883 DOI: 10.3390/nu14234998] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease characterized by hardening and narrowing of arteries. AS leads to a number of arteriosclerotic vascular diseases including cardiovascular diseases, cerebrovascular disease and peripheral artery disease, which pose a big threat to human health. Phytochemicals are a variety of intermediate or terminal low molecular weight secondary metabolites produced during plant energy metabolism. Phytochemicals from plant foods (vegetables, fruits, whole grains) and traditional herb plants have been shown to exhibit multiple bioactivities which are beneficial for prevention and treatment against AS. Many types of phytochemicals including polyphenols, saponins, carotenoids, terpenoids, organic sulfur compounds, phytoestrogens, phytic acids and plant sterols have already been identified, among which saponins are a family of glycosidic compounds consisting of a hydrophobic aglycone (sapogenin) linked to hydrophilic sugar moieties. In recent years, studies have shown that saponins exhibit a number of biological activities such as anti-inflammation, anti-oxidation, cholesterol-lowering, immunomodulation, anti-platelet aggregation, etc., which are helpful in the prevention and treatment of AS. This review aims to summarize the recent advances in the anti-atherosclerotic bioactivities of saponins such as ginsenoside, soyasaponin, astra-galoside, glycyrrhizin, gypenoside, dioscin, saikosaponin, etc.
Collapse
|
8
|
Huang L, Ren C, Li HJ, Wu YC. Recent Progress on Processing Technologies, Chemical Components, and Bioactivities of Chinese Red Ginseng, American Red Ginseng, and Korean Red Ginseng. FOOD BIOPROCESS TECH 2021. [DOI: 10.1007/s11947-021-02697-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
9
|
Li H, Chen C, Li ZM, Yang Y, Xing CQ, Li Y, Jin YH. Specific Interaction With Human Serum Albumin Reduces Ginsenoside Cytotoxicity in Human Umbilical Vein Endothelial Cells. Front Pharmacol 2020; 11:498. [PMID: 32410989 PMCID: PMC7201041 DOI: 10.3389/fphar.2020.00498] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 03/30/2020] [Indexed: 11/16/2022] Open
Abstract
Human serum albumin (HSA) is an important component of plasma, which has the functions of maintaining colloid osmotic pressure and capillary membrane stability, promoting blood circulation, and anti-oxidation. Three-dimensional structure of HSA determines its ability to bind and transport hormones and other substances. In this study we examined the interactions between HSA and ginsenoside Rg3, Rg5, Rk1, Rh2, and Rh4, which are the main cytotoxic ginsenosides extracted from red ginseng. Heat transfer generated by the specific interaction between HSA and each ginsenoside was measured using isothermal titration calorimetry (ITC) assay, which demonstrated that all these 5 ginsenosides bound to HSA with binding constants of 3.25, 1.89, 6.04, 2.07, and 5.17 × 105 M−1, respectively. Molecular docking also displayed that these ginsenosides interact with HSA at different sites of the HSA surface. Importantly, cell viability assay showed that the cytotoxicity of these ginsenosides reduced significantly at the presence of HSA in human vascular endothelial cells (HUVEC). Taken together, this study reveals the mechanism by which these ginsenosides are transported in vivo by not causing damage in vascular endothelium, and also suggests HSA might be an ideal carrier help to transport and execute these ginsenoside functions in human body.
Collapse
Affiliation(s)
- He Li
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, College of Life Science, Jilin University, Changchun, China
| | - Chen Chen
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, College of Life Science, Jilin University, Changchun, China
| | - Zhong-Ming Li
- The First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Yang Yang
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, College of Life Science, Jilin University, Changchun, China
| | - Chao-Qun Xing
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, College of Life Science, Jilin University, Changchun, China
| | - Yang Li
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, College of Life Science, Jilin University, Changchun, China
| | - Ying-Hua Jin
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, College of Life Science, Jilin University, Changchun, China
| |
Collapse
|
10
|
Yang Z, Di YT, Zhang Y, Hu XJ. Four new compounds from Neoboletus magnificus. Nat Prod Res 2020; 34:1152-1157. [PMID: 30698021 DOI: 10.1080/14786419.2018.1553878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Four new compounds, compounds 1, 2, 4, 6, along with two known compounds 3, 5, were isolated from the methanol extract of the fruiting body of Neoboletus magnificus. The structures of compounds were elucidated by HRMS and NMR spectroscopic methods. The in vitro anti-inflammatory activity of the isolated compounds was evaluated.
Collapse
Affiliation(s)
- Zhi Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Ying-Tong Di
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Science, Kunming, China
| | - Yu Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Science, Kunming, China
| | - Xu-Jia Hu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
11
|
Lee JO, Kim JH, Kim S, Kim MY, Hong YH, Kim HG, Cho JY. Gastroprotective effects of the nonsaponin fraction of Korean Red Ginseng through cyclooxygenase-1 upregulation. J Ginseng Res 2019; 44:655-663. [PMID: 32617046 PMCID: PMC7322762 DOI: 10.1016/j.jgr.2019.11.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 10/07/2019] [Accepted: 11/01/2019] [Indexed: 01/16/2023] Open
Abstract
Background Korean Red Ginseng is known to exhibit immune-enhancing and anti-inflammatory properties. The immune-enhancing effects of the nonsaponin fraction (NSF) of Korean Red Ginseng have been studied in many reports. However, the gastroprotective effect of this fraction is not fully understood. In this study, we demonstrate the activities of NSF for gastrointestinal protection and its related critical factor. Methods The in vitro and in vivo regulatory functions of NSF on cyclooxygenase-1 (COX-1) messenger RNA and protein levels were examined by reverse transcription polymerase chain reaction and immunoblotting analyses. Gastroprotective effects of NSF were investigated by histological score, gastric juice pH, and myeloperoxidase activity on indomethacin-induced, cold stress-induced, and acetylsalicylic acid-induced gastritis and dextran sulfate sodium-induced colitis in in vivo mouse models. Results NSF did not show cytotoxicity, and it increased COX-1 messenger RNA expression and protein levels in RAW264.7 cells. This upregulation was also observed in colitis and gastritis in vivo models. In addition, NSF treatment in mice ameliorated the symptoms of gastrointestinal inflammation, including histological score, colon length, gastric juice pH, gastric wall thickness, and myeloperoxidase activity. Conclusion These results suggest that NSF has gastroprotective effects on gastritis and colitis in in vivo mouse models through COX-1 upregulation.
Collapse
Affiliation(s)
- Jeong-Oog Lee
- Department of Aerospace Information Engineering, Bio-Inspired Aerospace Information Laboratory, Konkuk University, Seoul, Republic of Korea
| | - Ji Hye Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sunggyu Kim
- Research and Business Foundation, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biocosmetics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul, Republic of Korea
| | - Yo Han Hong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
- Corresponding author: Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Suwon, 16419, Republic of Korea.
| | - Han Gyung Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
- Corresponding author: Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Suwon 16419, Republic of Korea.
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
- Research and Business Foundation, Sungkyunkwan University, Suwon, Republic of Korea
- Corresponding author: Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Suwon, 16419, Republic of Korea.
| |
Collapse
|
12
|
Li F, Cao Y, Luo Y, Liu T, Yan G, Chen L, Ji L, Wang L, Chen B, Yaseen A, Khan AA, Zhang G, Jiang Y, Liu J, Wang G, Wang MK, Hu W. Two new triterpenoid saponins derived from the leaves of Panax ginseng and their antiinflammatory activity. J Ginseng Res 2019; 43:600-605. [PMID: 31695566 PMCID: PMC6823746 DOI: 10.1016/j.jgr.2018.09.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/04/2018] [Accepted: 09/18/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The leaves and roots of Panax ginseng are rich in ginsenosides. However, the chemical compositions of the leaves and roots of P. ginseng differ, resulting in different medicinal functions. In recent years, the aerial parts of members of the Panax genus have received great attention from natural product chemists as producers of bioactive ginsenosides. The aim of this study was the isolation and structural elucidation of novel, minor ginsenosides in the leaves of P. ginseng and evaluation of their antiinflammatory activity in vitro. METHODS Various chromatographic techniques were applied to obtain pure individual compounds, and their structures were determined by nuclear magnetic resonance and high-resolution mass spectrometry, as well as chemical methods. The antiinflammatory effect of the new compounds was evaluated on lipopolysaccharide-stimulated RAW 264.7 cells. RESULTS AND CONCLUSIONS Two novel, minor triterpenoid saponins, ginsenoside LS1 (1) and 5,6-didehydroginsenoside Rg3 (2), were isolated from the leaves of P. ginseng. The isolated compounds 1 and 2 were assayed for their inhibitory effect on nitric oxide production in LPS-stimulated RAW 264.7 cells, and Compound 2 showed a significant inhibitory effect with IC50 of 37.38 μM compared with that of NG-monomethyl-L-arginine (IC50 = 90.76 μM). Moreover, Compound 2 significantly decreased secretion of cytokines such as prostaglandin E2 and tumor necrosis factor-α. In addition, Compound 2 significantly suppressed protein expression of inducible nitric oxide synthase and cyclooxygenase-2. These results suggested that Compound 2 could be used as a valuable candidate for medicinal use or functional food, and the mechanism is warranted for further exploration.
Collapse
Affiliation(s)
- Fu Li
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization and Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Yufeng Cao
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology Around Hongze Lake, Huaiyin Normal University, Huaian, China
| | - Yanyan Luo
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology Around Hongze Lake, Huaiyin Normal University, Huaian, China
| | - Tingwu Liu
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology Around Hongze Lake, Huaiyin Normal University, Huaian, China
| | - Guilong Yan
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology Around Hongze Lake, Huaiyin Normal University, Huaian, China
| | - Liang Chen
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology Around Hongze Lake, Huaiyin Normal University, Huaian, China
| | - Lilian Ji
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology Around Hongze Lake, Huaiyin Normal University, Huaian, China
| | - Lun Wang
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization and Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Bin Chen
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization and Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Aftab Yaseen
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization and Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Ashfaq A. Khan
- Department of Chemistry, Women University of Azad Jammu and Kashmir, Bagh, Pakistan
| | - Guolin Zhang
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization and Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Yunyao Jiang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianxun Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Gongcheng Wang
- Department of Urology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, China
| | - Ming-Kui Wang
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization and Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Weicheng Hu
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology Around Hongze Lake, Huaiyin Normal University, Huaian, China
| |
Collapse
|
13
|
Identification of 20(R, S)-protopanaxadiol and 20(R, S)-protopanaxatriol for potential selective modulation of glucocorticoid receptor. Food Chem Toxicol 2019; 131:110642. [DOI: 10.1016/j.fct.2019.110642] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 06/12/2019] [Accepted: 06/22/2019] [Indexed: 01/06/2023]
|
14
|
AKT1-targeted proapoptotic activity of compound K in human breast cancer cells. J Ginseng Res 2019; 43:692-698. [PMID: 31695573 PMCID: PMC6823769 DOI: 10.1016/j.jgr.2019.07.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/29/2019] [Accepted: 07/19/2019] [Indexed: 01/04/2023] Open
Abstract
Background Breast cancer is a severe disease and the second leading cause of cancer death in women worldwide. To surmount this, various diagnosis and treatment options for breast cancer have been developed. One of the most effective strategies for cancer treatment is to induce apoptosis using naturally occurring compounds. Compound K (CK) is a ginseng saponin metabolite generated by human intestinal bacteria. CK has been studied for its cardioprotective, antiinflammatory, and liver-protective effects; however, the role of CK in breast cancer is not fully understood. Methods To investigate the anticancer effects of CK in SKBR3 and MDA-MB-231 cells, cell viability assays and flow cytometry analysis were used. In addition, the direct targets of CK anticancer activity were identified using immunoblotting analysis and overexpression experiments. Invasion, migration, and clonogenic assays were carried out to determine the effects of CK on cancer metastasis. Results CK-induced cell apoptosis in SKBR3 cells as determined through 3-(4-5-dimethylthiazol-2-yl)-2-5-diphenyltetrazolium bromide assays, propidium iodide (PI) and annexin V staining, and morphological changes. CK increased the cleaved forms of caspase-7, caspase-8, and caspase-9, whereas the expression of Bcl-2 was reduced by CK. In assays probing the cell survival pathway, CK activated only AKT1 and not AKT2. Moreover, CK inhibited breast cancer cell invasion, migration, and colony formation. Through regulation of AKT1 activity, CK exerts anticancer effects by inducing apoptosis. Conclusion Our results suggest that CK could be used as a therapeutic compound for breast cancer.
Collapse
|
15
|
Song C, Hong YH, Park JG, Kim HG, Jeong D, Oh J, Sung GH, Hossain MA, Taamalli A, Kim JH, Kim JH, Cho JY. Suppression of Src and Syk in the NF-κB signaling pathway by Olea europaea methanol extract is leading to its anti-inflammatory effects. JOURNAL OF ETHNOPHARMACOLOGY 2019; 235:38-46. [PMID: 30710734 DOI: 10.1016/j.jep.2019.01.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 01/16/2019] [Accepted: 01/25/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Olea europaea L., (Oleaceae) has been used widely in folk medicine in the European Mediterranean islands, India, Asia, and other parts of the world. Although this plant has high ethnopharmacological value for treating inflammatory diseases, the molecular mechanisms of how it inhibits the inflammatory response are not fully understood. In this study, we sought to identify the anti-inflammatory mechanisms of this plant. MATERIALS AND METHODS Using macrophages, we investigated the effects of O. europaea L. methanol extract (Oe-ME) and ethanol extract (Oe-EE) on the production of inflammatory mediator nitric oxide (NO) and prostaglandin E2 (PGE2), the expression levels of pro-inflammatory genes and intracellular inflammatory signaling activities. RESULTS Oe-ME and Oe-EE suppressed the production of NO in lipopolysaccharide-(LPS-), Pam3CSK4-, and poly (I:C)-stimulated RAW264.7 cells; importantly, no cytotoxicity was observed. Oe-ME and Oe-EE reduced production of PGE2 without exhibiting cytotoxicity. The mRNA expression levels of cyclooxygenase-2 (COX-2), inducible NO synthase (iNOS), IL-6, IL-1β, and tumor necrosis factor (TNF)-α were down-regulated by Oe-ME and Oe-EE. Nuclear fraction and whole lysate immunoblotting analyses and overexpression experiments strongly suggested that Oe-ME decreased the translocation of p65 and p50 (nuclear factors of the NF-κB subunit) as well as Src and Syk. CONCLUSION These results suggest that Oe-ME exerts its anti-inflammatory effects by targeting Src and Syk in the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Chaoran Song
- Department of Integrative Biotechnology and Biomedical Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Yo Han Hong
- Department of Integrative Biotechnology and Biomedical Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Jae Gwang Park
- Department of Integrative Biotechnology and Biomedical Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Han Gyung Kim
- Department of Integrative Biotechnology and Biomedical Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Deok Jeong
- Department of Integrative Biotechnology and Biomedical Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Junsang Oh
- Institute for Bio-Medical Convergence, International St. Mary's Hospital and College of Medicine, Catholic Kwandong University, Incheon, Republic of Korea.
| | - Gi-Ho Sung
- Institute for Bio-Medical Convergence, International St. Mary's Hospital and College of Medicine, Catholic Kwandong University, Incheon, Republic of Korea.
| | - Mohammad Amjad Hossain
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea.
| | - Amani Taamalli
- Laboratory of Olive Biotechnology, Center of Biotechnology-Technopole of Borj-Cedria, BP 901, Hammam-Lif 2050, Tunisia.
| | - Ji Hye Kim
- Department of Integrative Biotechnology and Biomedical Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea.
| | - Jae Youl Cho
- Department of Integrative Biotechnology and Biomedical Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
16
|
Yang Y, Xing R, Liu S, Qin Y, Li K, Yu H, Li P. Immunostimulatory Effects of Chitooligosaccharides on RAW 264.7 Mouse Macrophages via Regulation of the MAPK and PI3K/Akt Signaling Pathways. Mar Drugs 2019; 17:md17010036. [PMID: 30626153 PMCID: PMC6357175 DOI: 10.3390/md17010036] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/28/2018] [Accepted: 01/07/2019] [Indexed: 01/18/2023] Open
Abstract
Chitooligosaccharides (COS), the hydrolyzed products of chitin and chitosan, can be obtained by various methods. In this study, water-soluble COS were prepared from α- and β-chitosan by microwave-assisted degradation and their immunostimulatory effects were investigated in RAW 264.7 macrophages. The results indicated that α-COS were more active than β-COS in promoting the production of nitric oxide (NO) and cytokines, such as tumor necrosis factor-α (TNF-α) and interleukin 6 (IL-6). Quantitative real-time reverse transcription polymerase chain reaction and Western blotting indicated that COS also enhanced the expression of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and TNF-α. Further analyses demonstrated that COS induced the phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), p38, p85 and Akt, and the nuclear translocation of p65, indicating that they are able to activate the mitogen-activated protein kinases (MAPKs) and phosphoinositide 3-kinases (PI3K)/Akt signaling pathways dependent on nuclear factor (NF)-κB activation. In conclusion, COS activate RAW 264.7 cells via the MAPK and PI3K/Akt signaling pathways and are potential novel immune potentiators.
Collapse
Affiliation(s)
- Yue Yang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China.
- College of earth and planetary sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China.
| | - Ronge Xing
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China.
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China.
| | - Song Liu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China.
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China.
| | - Yukun Qin
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China.
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China.
| | - Kecheng Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China.
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China.
| | - Huahua Yu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China.
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China.
| | - Pengcheng Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China.
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China.
| |
Collapse
|
17
|
Chen NH, Li W, Zhong YL, Niu QW, Li YY, Zhang YB, Li MM, Li YL, Wang GC. New Acetophenone Derivatives from Acronychia oligophlebia
and Their Anti-inflammatory and Antioxidant Activities. Chem Biodivers 2018; 15:e18000080. [DOI: 10.1002/cbdv.201800080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 03/29/2018] [Indexed: 01/22/2023]
Affiliation(s)
- Neng-Hua Chen
- Institute of Traditional Chinese Medicine and Natural Products; College of Pharmacy; Jinan University; Guangzhou 510632 P. R. China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 P. R. China
| | - Wen Li
- Guangdong Saskatchewan Oilseed Joint Laboratory; Guangdong Engineering Technology Research Center for Oils and Fats Biorefinery; Department of Food Science and Engineering; Jinan University; Guangzhou 510632 China
| | - Yuan-Lin Zhong
- Institute of Traditional Chinese Medicine and Natural Products; College of Pharmacy; Jinan University; Guangzhou 510632 P. R. China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 P. R. China
| | - Qian-Wen Niu
- Institute of Traditional Chinese Medicine and Natural Products; College of Pharmacy; Jinan University; Guangzhou 510632 P. R. China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 P. R. China
| | - Ying-Ying Li
- Institute of Traditional Chinese Medicine and Natural Products; College of Pharmacy; Jinan University; Guangzhou 510632 P. R. China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 P. R. China
| | - Yu-Bo Zhang
- Institute of Traditional Chinese Medicine and Natural Products; College of Pharmacy; Jinan University; Guangzhou 510632 P. R. China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 P. R. China
| | - Man-Mei Li
- Institute of Traditional Chinese Medicine and Natural Products; College of Pharmacy; Jinan University; Guangzhou 510632 P. R. China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 P. R. China
| | - Yao-Lan Li
- Institute of Traditional Chinese Medicine and Natural Products; College of Pharmacy; Jinan University; Guangzhou 510632 P. R. China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 P. R. China
| | - Guo-Cai Wang
- Institute of Traditional Chinese Medicine and Natural Products; College of Pharmacy; Jinan University; Guangzhou 510632 P. R. China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 P. R. China
| |
Collapse
|
18
|
Kim E, Kim D, Yoo S, Hong YH, Han SY, Jeong S, Jeong D, Kim JH, Cho JY, Park J. The skin protective effects of compound K, a metabolite of ginsenoside Rb1 from Panax ginseng. J Ginseng Res 2018; 42:218-224. [PMID: 29719469 PMCID: PMC5925615 DOI: 10.1016/j.jgr.2017.03.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 03/07/2017] [Accepted: 03/17/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Compound K (CK) is a ginsenoside, a metabolite of Panax ginseng. There is interest both in increasing skin health and antiaging using natural skin care products. In this study, we explored the possibility of using CK as a cosmetic ingredient. METHODS To assess the antiaging effect of CK, RT-PCR was performed, and expression levels of matrix metalloproteinase-1, cyclooxygenase-2, and type I collagen were measured under UVB irradiation conditions. The skin hydrating effect of CK was tested by RT-PCR, and its regulation was explored through immunoblotting. Melanin content, melanin secretion, and tyrosinase activity assays were performed. RESULTS CK treatment reduced the production of matrix metalloproteinase-1 and cyclooxygenase-2 in UVB irradiated NIH3T3 cells and recovered type I collagen expression level. Expression of skin hydrating factors-filaggrin, transglutaminase, and hyaluronic acid synthases-1 and -2-were augmented by CK and were modulated through the inhibitor of κBα, c-Jun N-terminal kinase, or extracellular signal-regulated kinases pathway. In the melanogenic response, CK did not regulate tyrosinase activity and melanin secretion, but increased melanin content in B16F10 cells was observed. CONCLUSION Our data showed that CK has antiaging and hydrating effects. We suggest that CK could be used in cosmetic products to protect the skin from UVB rays and increase skin moisture level.
Collapse
Affiliation(s)
- Eunji Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Donghyun Kim
- Material Lab, Basic Research & Innovation Division, R&D Center, AmorePacific Corporation, Republic of Korea
| | - Sulgi Yoo
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yo Han Hong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sang Yun Han
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seonggu Jeong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Deok Jeong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jong-Hoon Kim
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Junseong Park
- Department of Engineering Chemistry, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
19
|
Yang WS, Yang E, Kim MJ, Jeong D, Yoon DH, Sung GH, Lee S, Yoo BC, Yeo SG, Cho JY. Momordica charantia Inhibits Inflammatory Responses in Murine Macrophages via Suppression of TAK1. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:435-452. [PMID: 29463104 DOI: 10.1142/s0192415x18500222] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Momordica charantia known as bitter melon is a representative medicinal plant reported to exhibit numerous pharmacological activities such as antibacterial, antidiabetic, anti-inflammatory, anti-oxidant, antitumor, and hypoglycemic actions. Although this plant has high ethnopharmacological value for treating inflammatory diseases, the molecular mechanisms by which it inhibits the inflammatory response are not fully understood. In this study, we aim to identify the anti-inflammatory mechanism of this plant. To this end, we studied the effects of its methanol extract (Mc-ME) on lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. Specifically, we evaluated nitric oxide (NO) production, mRNA expression of inflammatory genes, luciferase reporter gene activity, and putative molecular targets. Mc-ME blocked NO production in a dose-dependent manner in RAW264.7 cells; importantly, no cytotoxicity was observed. Moreover, the mRNA expression levels of inducible NO synthase (iNOS) and cyclooxygenase (COX)-2 were decreased by Mc-ME treatment in a dose-dependent manner. Luciferase assays and nuclear lysate immunoblotting analyses strongly indicated that Mc-ME decreases the levels of p65 [a nuclear factor (NF)-[Formula: see text]B subunit] and c-Fos [an activator protein (AP)-1 subunit]. Whole lysate immunoblotting assays, luciferase assays, and overexpression experiments suggested that transforming growth factor [Formula: see text]-activated kinase 1 (TAK1) is targeted by Mc-ME, thereby suppressing NF-[Formula: see text]B and AP-1 activity via downregulation of extracellular signal-regulated kinases (ERKs) and AKT. These results strongly suggest that Mc-ME exerts its anti-inflammatory activity by reducing the action of TAK1, which also affects the activation of NF-[Formula: see text]B and AP-1.
Collapse
Affiliation(s)
- Woo Seok Yang
- * Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Eunju Yang
- † Gyeonggi Science High School for the Gifted, Suwon 16297, Republic of Korea
| | - Min-Jeong Kim
- ‡ Department of Radiology, Hallym Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Gyeonggi 14068, Republic of Korea
| | - Deok Jeong
- * Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Deok Hyo Yoon
- § Institute for Bio-Medical Convergence, International St. Mary's Hospital and College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea
| | - Gi-Ho Sung
- § Institute for Bio-Medical Convergence, International St. Mary's Hospital and College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea
| | - Seungihm Lee
- † Gyeonggi Science High School for the Gifted, Suwon 16297, Republic of Korea
| | - Byong Chul Yoo
- ¶ Colorectal Cancer Branch, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
| | - Seung-Gu Yeo
- ∥ Department of Radiation Oncology, Soonchunhyang University College of Medicine, Soonchunhyang University Hospital, Cheonan 31151, Republic of Korea
| | - Jae Youl Cho
- * Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
- † Gyeonggi Science High School for the Gifted, Suwon 16297, Republic of Korea
| |
Collapse
|
20
|
Syk and Src-targeted anti-inflammatory activity of aripiprazole, an atypical antipsychotic. Biochem Pharmacol 2018; 148:1-12. [DOI: 10.1016/j.bcp.2017.12.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 12/07/2017] [Indexed: 12/20/2022]
|
21
|
Hong YH, Kim D, Nam G, Yoo S, Han SY, Jeong SG, Kim E, Jeong D, Yoon K, Kim S, Park J, Cho JY. Photoaging protective effects of BIOGF1K, a compound-K-rich fraction prepared from Panax ginseng. J Ginseng Res 2018; 42:81-89. [PMID: 29348726 PMCID: PMC5766695 DOI: 10.1016/j.jgr.2017.01.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 12/25/2016] [Accepted: 01/02/2017] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND BIOGF1K, a compound-K-rich fraction, has been shown to display anti-inflammatory activity. Although Panax ginseng is widely used for the prevention of photoaging events induced by UVB irradiation, the effect of BIOGF1K on photoaging has not yet been examined. In this study, we investigated the effects of BIOGF1K on UVB-induced photoaging events. METHODS We analyzed the ability of BIOGF1K to prevent UVB-induced apoptosis, enhance matrix metalloproteinase (MMP) expression, upregulate anti-inflammatory activity, reduce sirtuin 1 expression, and melanin production using reverse transcription-polymerase chain reaction, melanin content assay, tyrosinase assay, and flow cytometry. We also evaluated the effects of BIOGF1K on the activator protein-1 signaling pathway, which plays an important role in photoaging, by immunoblot analysis and luciferase reporter gene assays. RESULTS Treatment of UVB-irradiated NIH3T3 fibroblasts with BIOGF1K prevented UVB-induced cell death, inhibited apoptosis, suppressed morphological changes, reduced melanin secretion, restored the levels of type I procollagen and sirtuin 1, and prevented mRNA upregulation of MMP-1, MMP-2, and cyclo-oxygenase-2; these effects all occurred in a dose-dependent manner. In addition, BIOGF1K markedly reduced activator-protein-1-mediated luciferase activity and decreased the activity of mitogen-activated protein kinases (extracellular response kinase, p38, and C-Jun N-terminal kinase). CONCLUSION Our results strongly suggest that BIOGF1K has anti-photoaging activity and that BIOGF1K could be used in anti-aging cosmeceutical preparations.
Collapse
Affiliation(s)
- Yo Han Hong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Donghyun Kim
- Amorepacific Research and Development Unit, Yongin, Republic of Korea
| | - Gibaeg Nam
- Amorepacific Research and Development Unit, Yongin, Republic of Korea
| | - Sulgi Yoo
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sang Yun Han
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seong-Gu Jeong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Eunji Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Deok Jeong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Keejung Yoon
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sunggyu Kim
- Research and Business Foundation, Sungkyunkwan University, Suwon, Republic of Korea
| | - Junseong Park
- Amorepacific Research and Development Unit, Yongin, Republic of Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
22
|
Kim MS, Yoo BC, Yang WS, Han SY, Jeong D, Song JM, Kim KH, Aravinthan A, Kim JH, Kim JH, Kim SC, Cho JY. Src is the primary target of aripiprazole, an atypical antipsychotic drug, in its anti-tumor action. Oncotarget 2017; 9:5979-5992. [PMID: 29464048 PMCID: PMC5814188 DOI: 10.18632/oncotarget.23192] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/01/2017] [Indexed: 12/22/2022] Open
Abstract
Aripiprazole (ARP) is an atypical anti-psychotic drug widely used to treat schizophrenia and bipolar disorder. The pharmacological effects of ARP on cancer cells are still poorly understood. In this study, anti-cancer effects of ARP on various malignant tumor cells and its molecular mechanism were further carefully examined by using cell proliferation assay, xenograft mouse model, immunoblotting analysis, migration assay, luciferase reporter gene assay, kinase assay, and overexpression strategy. Treatment with ARP induced cytotoxicity in U251 glioma cells, MKN-1 gastric adenosquamous carcinoma cells, and CT26 colon carcinoma cells. ARP suppressed cell proliferation of LN428, MDA-MB-231, and HEK293 cells. Pro-apoptotic factors active caspase-3, -8, and -9, as well as p53, were upregulated, whereas the protein and mRNA levels of anti-apoptotic factor B-cell lymphoma 2 (Bcl-2) decreased. In agreement with the in vitro results, ARP compound also significantly suppressed the growth of tumor masses formed by injecting CT26 colon cancer cells into mice. ARP treatment also effectively decreased the migratory ability of U251 glioma cells by downregulating metalloproteinase-9. Levels of phosphorylated Src, phosphorylated phosphatidylinositide 3-kinase (PI3K), and phosphorylated signal transducer and activator of transcription 3 (STAT3) were significantly decreased following ARP treatment. ARP compound reduced the kinase activity of Src. Our studies suggest that Src may be an important target molecule linked to the antitumor effects of ARP.
Collapse
Affiliation(s)
- Mi Seon Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Byong Chul Yoo
- Colorectal Cancer Branch, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
| | - Woo Seok Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sang Yun Han
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Deok Jeong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jun Min Song
- School of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| | - Kyung Hee Kim
- Colorectal Cancer Branch, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
| | - Adithan Aravinthan
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Ji Hye Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jong-Hoon Kim
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Seung Cheol Kim
- Department of Obstetrics and Gynecology, Ewha Womans University Mokdong Hospital, Ewha Womans University School of Medicine, Seoul 07985, Republic of Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
23
|
Park G, Kim E, Son YJ, Yoon DH, Sung GH, Aravinthan A, Park YC, Kim JH, Cho JY. Anti-inflammatory effect of torilidis fructus ethanol extract through inhibition of Src. PHARMACEUTICAL BIOLOGY 2017; 55:2074-2082. [PMID: 28832235 PMCID: PMC6130681 DOI: 10.1080/13880209.2017.1362011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
CONTEXT Torilidis fructus, fruits of Torilis japonica Decadolle (Umbelliferae), is a medicinal herb traditionally used as a pesticide, an astrictive, or a medicine for various inflammatory diseases. OBJECTIVES Due to the lack of pharmacological studies on this herbal medicine, we explored the inhibitory activity of torilidis fructus on the macrophage-mediated inflammatory response using its ethanol extract (Tf-EE). MATERIAL AND METHODS The Griess assay and prostaglandin (PGE2) ELISA assay were conducted with Tf-EE (0-75 µg/mL) and LPS (1 µg/mL) treated RAW264.7 cells in cultured media. Tf-EE pretreated RAW264.7 cells were incubated with LPS for 6 h and semi-quantitative PCR was performed. Reporter gene assays, overexpression of target enzymes and immunoblotting were performed on macrophages to determine the molecular targets of Tf-EE. RESULTS Tf-EE markedly suppressed the inflammatory response of macrophages, such as lipopolysaccharide (LPS)-induced nitric oxide (NO) and PGE2 production with IC50 values of 35.66 and 62.47 µg/mL, respectively. It was also found that Tf-EE reduced the expression of inducible NO synthase (iNOS) and cyclooxygenase (COX)-2 by 80%. Nuclear translocation and activation of nuclear factor (NF)-κB (p65 and p50) were declined by 60% and 30% respectively, and their regulatory events including the phosphorylation of AKT, IκBα, Src, and the formation of complexes between Src and p-p85 were also recognized to be diminished. CONCLUSIONS The signalling events managed by Src and p85 complex seemed to be critically involved in Tf-EE-mediated anti-inflammatory response. This might suggest that Tf-EE exhibited anti-inflammatory effects through Src-targeted inhibition of NF-κB.
Collapse
Affiliation(s)
- Gyubyung Park
- Gyeonggi Science High School for the Gifted, Suwon, Republic of Korea
| | - Eunji Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Young-Jin Son
- Department of Pharmacy, Sunchon National University, Suncheon, Korea
| | - Deok Hyo Yoon
- Institute for Bio-Medical Convergence, International St. Mary's Hospital and College of Medicine, Catholic Kwandong University Incheon, Incheon, Republic of Korea
| | - Gi-Ho Sung
- Institute for Bio-Medical Convergence, International St. Mary's Hospital and College of Medicine, Catholic Kwandong University Incheon, Incheon, Republic of Korea
| | - Adithan Aravinthan
- College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
| | - Yung Chul Park
- College of Forest and Environmental Sciences, Kangwon National University, Chuncheon, Republic of Korea
- Yung Chul ParkCollege of Forest and Environmental Sciences, Kangwon National University, Chuncheon24341, Republic of Korea
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
- Jong-Hoon KimCollege of Veterinary Medicine, Chonbuk National University, Iksan54596, Republic of Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
- CONTACT: Jae Youl ChoDepartment of Genetic Engineering, Sungkyunkwan University, Suwon16419, Republic of Korea
| |
Collapse
|
24
|
Yang Y, Xing R, Liu S, Qin Y, Li K, Yu H, Li P. Immunostimulatory effects of sulfated chitosans on RAW 264.7 mouse macrophages via the activation of PI3K/Akt signaling pathway. Int J Biol Macromol 2017; 108:1310-1321. [PMID: 29129634 DOI: 10.1016/j.ijbiomac.2017.11.042] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/08/2017] [Accepted: 11/08/2017] [Indexed: 01/15/2023]
Abstract
To investigate the immunostimulatory effects of chitosan sulfates, we prepared α- and β-chitosan sulfates with different molecular weights and compared their immunostimulatory activities in RAW 264.7 macrophages. Results suggest that β-chitosan sulfates were more active than α-chitosan in promoting nitric oxide (NO) production. Further study show that β-chitosan sulfate significantly promoted the production of NO, prostaglandin E2, tumor necrosis factor (TNF)-α, interleukin-6 and interleukin-1β at the levels of transcription and translation. Moreover, Western blots revealed that it induced the phosphorylation of p85 and Akt, and the nuclear translocation of p50/p65 and c-Fos/c-Jun. The luciferase activity of cells pretreated with β-chitosan sulfate further confirmed the nuclear translocation of p50/p65 and c-Fos/c-Jun. Determination of Toll-like receptor (TLR) 4 expression suggested that β-chitosan sulfate at least partly bound to TLR4. In conclusion, β-chitosan sulfates activate RAW 264.7 cells through the PI3K-Akt pathway, which is dependent on activator protein-1 and nuclear factor-κB activation.
Collapse
Affiliation(s)
- Yue Yang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1, Wenhai Road, Qingdao 266237, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ronge Xing
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1, Wenhai Road, Qingdao 266237, China.
| | - Song Liu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1, Wenhai Road, Qingdao 266237, China
| | - Yukun Qin
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1, Wenhai Road, Qingdao 266237, China
| | - Kecheng Li
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1, Wenhai Road, Qingdao 266237, China
| | - Huahua Yu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1, Wenhai Road, Qingdao 266237, China
| | - Pengcheng Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1, Wenhai Road, Qingdao 266237, China.
| |
Collapse
|
25
|
Kim Y, Kim HG, Han SY, Jeong D, Yang WS, Kim JI, Kim JH, Yi YS, Cho JY. Hydroquinone suppresses IFN-β expression by targeting AKT/IRF3 pathway. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:547-554. [PMID: 28883758 PMCID: PMC5587604 DOI: 10.4196/kjpp.2017.21.5.547] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 06/09/2017] [Indexed: 12/17/2022]
Abstract
Previous studies have demonstrated the role of hydroquinone (HQ), a hydroxylated benzene metabolite, in modulating various immune responses; however, its role in macrophage-mediated inflammatory responses is not fully understood. In this study, the role of HQ in inflammatory responses and the underlying molecular mechanism were explored in macrophages. HQ down-regulated the expression of interferon (IFN)-β mRNA in LPS-stimulated RAW264.7 cells without any cytotoxicity and suppressed interferon regulatory factor (IRF)-3-mediated luciferase activity induced by TIR-domain-containing adapter-inducing interferon-β (TRIF) and TANK-binding kinase 1 (TBK1). A mechanism study revealed that HQ inhibited IRF-3 phosphorylation induced by lipopolysaccharide (LPS), TRIF, and AKT by suppressing phosphorylation of AKT, an upstream kinase of the IRF-3 signaling pathway. IRF-3 phosphorylation is highly induced by wild-type AKT and poorly induced by an AKT mutant, AKT C310A, which is mutated at an inhibitory target site of HQ. We also showed that HQ inhibited IRF-3 phosphorylation by targeting all three AKT isoforms (AKT1, AKT2, and AKT3) in RAW264.7 cells and suppressed IRF-3-mediated luciferase activities induced by AKT in HEK293 cells. Taken together, these results strongly suggest that HQ inhibits the production of a type I IFN, IFN-β, by targeting AKTs in the IRF-3 signaling pathway during macrophage-mediated inflammation.
Collapse
Affiliation(s)
- Yong Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Han Gyung Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Sang Yun Han
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Deok Jeong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Woo Seok Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Jung-Il Kim
- Department of Information Statistics, Kangwon National University, Chucheon 24341, Korea
| | - Ji Hye Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Young-Su Yi
- Department of Pharmaceutical Engineering, Cheongju University, Cheongju 28503, Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
26
|
Anti-Inflammatory Effect of Piper attenuatum Methanol Extract in LPS-Stimulated Inflammatory Responses. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:4606459. [PMID: 28811826 PMCID: PMC5547706 DOI: 10.1155/2017/4606459] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 06/05/2017] [Accepted: 06/21/2017] [Indexed: 11/18/2022]
Abstract
Piper attenuatum is used as a traditional medicinal plant in India. One of the substances in P. attenuatum has been suggested to have anti-inflammatory effects. However, there is insufficient research about the anti-inflammatory mechanisms of action of P. attenuatum. The effects of P. attenuatum methanol extract (Pa-ME) on the production of inflammatory mediators nitric oxide (NO) and prostaglandin E2 (PGE2), the expression of proinflammatory genes, the translocation level of transcription factors, and intracellular signaling activities were investigated using macrophages. Pa-ME suppressed the production of NO and PGE2 in lipopolysaccharide- (LPS-), pam3CSK4-, and poly(I:C)-stimulated RAW264.7 cells without displaying cytotoxicity. The mRNA expression levels of inducible NO synthase (iNOS) and cyclooxygenase 2 (COX-2) were decreased by Pa-ME. P-ME reduced the translocation of p50/NF-κB and AP-1 (c-Jun and c-Fos), as well as the activity of their upstream enzymes Src, Syk, and TAK1. Immunoprecipitation analysis showed failure of binding between their substrates, phospho- (p-) p85 and p-MKK3/6. p-p85 and p-MKK3/6, which were induced by overexpression of Src, Syk, and TAK1, were also reduced by Pa-ME. Therefore, these results suggest that Pa-ME exerts its anti-inflammatory effects by targeting Src and Syk in the NF-κB signaling pathway and TAK1 in the AP-1 signaling pathway.
Collapse
|
27
|
Lee JO, Yang WS, Park JG, Jeong D, Kim HG, Yoon KD, Aravinthan A, Kim JH, Kim E, Cho JY. Src and Syk contribute to the anti-inflammatory activities of Achyranthes aspera ethanolic extract. JOURNAL OF ETHNOPHARMACOLOGY 2017; 206:1-7. [PMID: 28502904 DOI: 10.1016/j.jep.2017.05.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/23/2017] [Accepted: 05/10/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Nuclear factor-kappa B (NF-κB) plays pivotal roles in inflammation. Src and Syk are two tyrosine kinases that act upstream of NF-κB signaling. Although Achyranthes aspera L. (A. aspera) has been used as a traditional medicine to treat fevers and inflammatory ailments and heal wounds, the molecular mechanisms of its anti-inflammatory actions are not yet fully understood. MATERIALS AND METHODS In this study, we evaluated the anti-inflammatory effect of A. aspera ethanol extract (Aa-EE). To determine the mechanism by which Aa-EE dampens the inflammatory response, nitric oxide (NO) production and the mRNA expression levels of tumor necrosis factor (TNF)-α and inducible nitric oxide synthase (iNOS) were examined by Griess assay and RT-PCR. Luciferase assays and immunoblotting were also conducted to examine how Aa-EE regulates the NF-κB pathway. RESULTS Aa-EE reduced NO production up to 60% without any cytotoxicity. This extract was found to downregulate the mRNA expression levels of inflammatory genes. Aa-EE blocked NF-κB promoter activity induced by both TNF-α and adaptor molecule MyD88 (about 70% and 40%, respectively). Moreover, nuclear translocation of p65 and IκBα phosphorylation were also inhibited. Furthermore, Aa-EE inactivated two upstream signaling molecules, the Src and Syk kinases. In accordance with these data, the kinase activities of Src and Syk were decreased by 50% and 80%, respectively. The anti-inflammatory action of Aa-EE was also confirmed in a gastritis model. CONCLUSION Our data suggest that Aa-EE targets NF-κB to exert its anti-inflammatory properties by suppressing Src and Syk. Therefore, our study raises the possibility that this extract can be developed as a novel natural anti-inflammatory remedy.
Collapse
Affiliation(s)
- Jeong-Oog Lee
- Department of Aerospace Information Engineering, Bio-Inspired Aerospace Information Laboratory, Konkuk University, Seoul 05029, Republic of Korea
| | - Woo Seok Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Gwang Park
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Deok Jeong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Han Gyung Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Kee Dong Yoon
- College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Adithan Aravinthan
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea.
| | - Eunji Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
28
|
Anticancer Efficacy of Cordyceps militaris Ethanol Extract in a Xenografted Leukemia Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:8474703. [PMID: 28761499 PMCID: PMC5518515 DOI: 10.1155/2017/8474703] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/17/2017] [Accepted: 05/30/2017] [Indexed: 12/16/2022]
Abstract
Cordyceps militaris is used widely as a traditional medicine in East Asia. Although a few studies have attempted to elucidate the anticancer activities of C. militaris, the precise mechanism of C. militaris therapeutic effects is not fully understood. We examined the anticancer activities of C. militaris ethanolic extract (Cm-EE) and its cellular and molecular mechanisms. For this purpose, a xenograft mouse model bearing murine T cell lymphoma (RMA) cell-derived cancers was established to investigate in vivo anticancer mechanisms. MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay, immunoblotting analysis, and flow cytometric assay were employed to check in vitro cytotoxicity, molecular targets, and proapoptotic action of Cm-EE. Interestingly, cancer sizes and mass were reduced in a C. militaris-administered group. Levels of the phosphorylated forms of p85 and AKT were clearly decreased in the group administered with Cm-EE. This result indicated that levels of phosphoglycogen synthase kinase 3β (p-GSK3β) and cleaved caspase-3 were increased with orally administered Cm-EE. In addition, Cm-EE directly inhibited the viability of cultured RMA cells and C6 glioma cells. The number of proapoptotic cells was significantly increased in a Cm-EE treated group compared with a control group. Our results suggested that C. militaris might be able to inhibit cancer growth through regulation of p85/AKT-dependent or GSK3β-related caspase-3-dependent apoptosis.
Collapse
|
29
|
Yoo S, Kim MY, Cho JY. Beauvericin, a cyclic peptide, inhibits inflammatory responses in macrophages by inhibiting the NF-κB pathway. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:449-456. [PMID: 28706459 PMCID: PMC5507784 DOI: 10.4196/kjpp.2017.21.4.449] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/21/2017] [Accepted: 04/28/2017] [Indexed: 12/16/2022]
Abstract
Beauvericin (BEA), a cyclic hexadepsipeptide produced by the fungus Beauveria bassiana, is known to have anti-cancer, anti-inflammatory, and anti-microbial actions. However, how BEA suppresses macrophage-induced inflammatory responses has not been fully elucidated. In this study, we explored the anti-inflammatory properties of BEA and the underlying molecular mechanisms using lipopolysaccharide (LPS)-treated macrophage-like RAW264.7 cells. Levels of nitric oxide (NO), mRNA levels of transcription factors and the inflammatory genes inducible NO synthase (iNOS) and interleukin (IL)-1, and protein levels of activated intracellular signaling molecules were determined by Griess assay, semi-quantitative reverse transcriptase-polymerase chain reaction (RT-PCR), luciferase reporter gene assay, and immunoblotting analysis. BEA dose-dependently blocked the production of NO in LPS-treated RAW264.7 cells without inducing cell cytotoxicity. BEA also prevented LPS-triggered morphological changes. This compound significantly inhibited nuclear translocation of the NF-κB subunits p65 and p50. Luciferase reporter gene assays demonstrated that BEA suppresses MyD88-dependent NF-κB activation. By analyzing upstream signaling events for NF-κB activation and overexpressing Src and Syk, these two enzymes were revealed to be targets of BEA. Together, these results suggest that BEA suppresses NF-κB-dependent inflammatory responses by suppressing both Src and Syk.
Collapse
Affiliation(s)
- Sulgi Yoo
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
30
|
Syk Plays a Critical Role in the Expression and Activation of IRAK1 in LPS-Treated Macrophages. Mediators Inflamm 2017; 2017:1506248. [PMID: 28680194 PMCID: PMC5478860 DOI: 10.1155/2017/1506248] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 04/13/2017] [Indexed: 01/10/2023] Open
Abstract
To address how interleukin-1 receptor-associated kinase 1 (IRAK1) is controlled by other enzymes activated by toll-like receptor (TLR) 4, we investigated the possibility that spleen tyrosine kinase (Syk), a protein tyrosine kinase that is activated at an earlier stage during TLR4 activation, plays a central role in regulating the functional activation of IRAK1. Indeed, we found that overexpression of myeloid differentiation primary response gene 88 (MyD88), an adaptor molecule that drives TLR signaling, induced IRAK1 expression and that piceatannol, a Syk inhibitor, successfully suppressed the MyD88-dependent upregulation of IRAK1 under LPS treatment conditions. Interestingly, in Syk-knockout RAW264.7 cells, IRAK1 activity was almost completely blocked after LPS treatment, while providing a Syk-recovery gene to the knockout cells successfully restored IRAK1 expression. According to our measurements of IRAK1 mRNA levels, the transcriptional upregulation of IRAK1 was induced by LPS treatment between 4 and 60 min, and this can be suppressed in Syk knockout cells, providing an effect similar that that seen under piceatannol treatment. The overexpression of Syk reverses this effect and leads to a significantly higher IRAK1 mRNA level. Collectively, our results strongly suggest that Syk plays a critical role in regulating both the activity and transcriptional level of IRAK1.
Collapse
|
31
|
Anti-inflammatory activity of the sclerotia of edible fungus, Poria cocos Wolf and their active lanostane triterpenoids. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.02.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
32
|
Yi YS, Kim MY, Cho JY. JS-III-49, a hydroquinone derivative, exerts anti-inflammatory activity by targeting Akt and p38. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:345-352. [PMID: 28461777 PMCID: PMC5409119 DOI: 10.4196/kjpp.2017.21.3.345] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 01/29/2017] [Accepted: 02/03/2017] [Indexed: 12/25/2022]
Abstract
Since previous studies have reported that hydroquinone (HQ) exerted immunosuppressive and anti-inflammatory activity, various HQ derivatives have been synthesized and their biological activities investigated. In this study, we explored the anti-inflammatory activity of JS-III-49, a novel HQ derivative, in macrophage-mediated inflammatory responses. JS-III-49 suppressed the production of the inflammatory mediators nitric oxide (NO) and prostaglandin E2 (PGE2) and down-regulated the mRNA expression of the inflammatory enzymes cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) as well as the expression of the pro-inflammatory cytokines interleukin-6 (IL-6) and IL-1b without cytotoxicity in LPS-stimulated RAW264.7 cells. JS-III-49 inhibited nuclear translocation of the NF-kB transcription factors p65 and p50 by directly targeting Akt, an upstream kinase of the NF-kB pathway, in LPS-stimulated RAW264.7 cells. However, JS-III-49 did not directly inhibit the kinase activities of Src and Syk, which are upstream kinases of Akt, in LPS-stimulated RAW264.7 cells. Moreover, JS-III-49 suppressed the nuclear translocation of c-Fos, one of the components of AP-1, by specifically targeting p38, an upstream mitogen-activated protein kinase (MAPK) in the AP-1 pathway in LPS-stimulated RAW264.7 cells. These results suggest that JS-III-49 plays an anti-inflammatory role in LPS-stimulated macrophages by targeting Akt and p38 in the NF-kB and AP-1 pathways, respectively.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea.,Department of Pharmaceutical Engineering, Cheongju University, Cheongju 28503, Korea
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
33
|
Yang WS, Kim D, Yi YS, Kim JH, Jeong HY, Hwang K, Kim JH, Park J, Cho JY. AKT-targeted anti-inflammatory activity of the methanol extract of Chrysanthemum indicum var. albescens. JOURNAL OF ETHNOPHARMACOLOGY 2017; 201:82-90. [PMID: 28274893 DOI: 10.1016/j.jep.2017.03.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/24/2017] [Accepted: 03/01/2017] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wild chrysanthemum (Chrysanthemum indicum) is one of well-known medicinal plants traditionally used in Korea and China. As a variant of wild chrysanthemum, white wild chrysanthemum (Chrysanthemum indicum var. albescens) is also ethnopharmacologically applied to treat various symptoms such as inflammatory diseases. AIM OF STUDY Although the anti-inflammatory activity of Chrysanthemum indicum has been reported, the anti-inflammatory activity and underlying molecular mechanism of white wild chrysanthemum are poorly understood. MATERIALS AND METHODS The effects of Chrysanthemum indicum var. albescens methanol extract (Civ-ME) on the production of inflammatory mediators, expression of pro-inflammatory genes, cell viability, and the activities of intracellular signaling molecules and transcription factors were investigated in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. RESULTS Civ-ME suppressed the production of both nitric oxide (NO) and prostaglandin E2 (PGE2) without cytotoxicity in LPS-stimulated RAW264.7 cells. Civ-ME was found to reduce the mRNA levels of inflammatory genes such as inducible NO synthase (iNOS) and tumor necrosis factor (TNF)-α and reduced NF-κB-mediated transcriptional activation. Civ-ME inhibited the nuclear translocation of NF-κB (p65 and p50), and its upstream signaling composed of IκBα and IKKα/β. An NF-κB luciferase reporter gene assay and an in vitro kinase assay confirmed that AKT1 and AKT2 might be direct pharmacological targets of Civ-ME. In addition, luteolin was identified by HPLC analysis as the main active pharmacological components of Civ-ME. CONCLUSION Civ-ME exerts an anti-inflammatory effect by targeting AKT1 and AKT2 in the NF-κB signaling pathway in macrophage-mediated inflammatory responses.
Collapse
Affiliation(s)
- Woo Seok Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Donghyun Kim
- Heritage Material Research Team, Amorepacific R&D Unit, Yongin 446-729, Republic of Korea.
| | - Young-Su Yi
- Department of Pharmaceutical Engineering, Cheongju University, Cheongju 28503, Republic of Korea.
| | - Ji Hye Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Hye Yoon Jeong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Kyeonghwan Hwang
- Heritage Material Research Team, Amorepacific R&D Unit, Yongin 446-729, Republic of Korea.
| | - Jong-Hoon Kim
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea.
| | - Junseong Park
- Heritage Material Research Team, Amorepacific R&D Unit, Yongin 446-729, Republic of Korea.
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
34
|
Yu JS, Kim JH, Lee S, Jung K, Kim KH, Cho JY. Src/Syk-Targeted Anti-Inflammatory Actions of Triterpenoidal Saponins from Gac (Momordica cochinchinensis) Seeds. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:459-473. [PMID: 28367713 DOI: 10.1142/s0192415x17500288] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Momordica cochinchinensis Spreng (family Cucurbitaceae), also known as gac, or red melon, is an edible Southeast Asian fruit valued for its nutritional and medicinal properties. Specifically, Momordicae Semen, the seeds of the gac fruit, is used in traditional Chinese medicine to treat boils, rheumatic pain, muscle spasm, hemorrhoids, and hemangiomas. In this study, a chemical investigation into a gac seed ethanol (EtOH) extract resulted in the identification of three triterpenoidal saponins (1-3), which were investigated for their anti-inflammatory effects. Among the saponins, momordica saponin I (compound 3) reduced the production of nitric oxide (NO) in LPS-activated RAW264.7 cells without inducing cytotoxicity. The mRNA levels of inducible NO synthase (iNOS) and cyclooxygenase (COX)-2 were decreased by momordica saponin I. Additionally, the translocation of p65 and p50 (subunits of the transcription factor NF-[Formula: see text]B) into the nucleus was remarkably inhibited. Furthermore, the phosphorylation levels of inflammatory signaling proteins (I[Formula: see text]B[Formula: see text], Src, and Syk) known to be upstream regulatory molecules of p65 were decreased under momordica saponin I-treated conditions. The molecular targets of momordica saponin I were confirmed in overexpression experiments and through immunoblot analyses with Src and Syk. This study provides evidence that momordica saponin I could be beneficial in treating inflammatory diseases, and should be considered a bioactive immunomodulatory agent with anti-inflammatory properties.
Collapse
Affiliation(s)
- Jae Sik Yu
- * School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Jun Ho Kim
- † Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Seulah Lee
- * School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Kiwon Jung
- ‡ Institute of Pharmaceutical Sciences, College of Pharmacy, CHA University, Seongnam 13488, Republic of Korea
| | - Ki Hyun Kim
- * School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Jae Youl Cho
- † Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| |
Collapse
|
35
|
Hossen MJ, Yang WS, Kim D, Aravinthan A, Kim JH, Cho JY. Thymoquinone: An IRAK1 inhibitor with in vivo and in vitro anti-inflammatory activities. Sci Rep 2017; 7:42995. [PMID: 28216638 PMCID: PMC5316937 DOI: 10.1038/srep42995] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 01/18/2017] [Indexed: 12/14/2022] Open
Abstract
Thymoquinone (TQ) is a bioactive component of black seed (Nigella sativa) volatile oil and has been shown to have anti-oxidative, anti-inflammatory, and anti-cancer properties. In the present study, we explored the molecular mechanisms that underlie the anti-inflammatory effect of TQ and its target proteins using lipopolysaccharide (LPS)-stimulated murine macrophage-like RAW264.7 and human monocyte-like U937 cells, together with LPS/D-galactosamine (GalN)-induced acute hepatitis and HCl/EtOH-induced gastritis mouse models. TQ strongly inhibited the production of nitric oxide (NO) and repressed NO synthase (iNOS), tumor necrosis factor (TNF)-α, cyclooxygenase (COX)−2, interleukin (IL)−6, and IL-1β expression in LPS-activated RAW264.7 cells. Treatment of LPS/D-GalN–induced hepatitis and EtOH/HCl–induced gastritis mouse models with TQ significantly ameliorated disease symptoms. Using luciferase reporter gene assays, we also showed that the nuclear levels of transcription factors and phosphorylation patterns of signaling proteins, activator protein (AP)−1, and nuclear factor (NF)-κB pathways were all affected by TQ treatment. Finally, we used additional kinase and luciferase validation assays with interleukin-1 receptor-associated kinase 1 (IRAK1) to show that IRAK1 is directly suppressed by TQ treatment. Together, these findings strongly suggest that the anti-inflammatory actions of TQ are caused by suppression of IRAK-linked AP-1/NF-κB pathways.
Collapse
Affiliation(s)
- Muhammad Jahangir Hossen
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.,Department of Animal Science, Patuakhali Science and Technology University, Dumki, Patuakhali 8602, Bangladesh
| | - Woo Seok Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Daewon Kim
- Laboratory of Bio-informatics, Department of Multimedia Engineering, Dankook University, Cheonan 31116, Republic of Korea
| | - Adithan Aravinthan
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Jong-Hoon Kim
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
36
|
Dung TTM, Yi YS, Heo J, Yang WS, Kim JH, Kim HG, Park JG, Yoo BC, Cho JY, Hong S. Critical role of protein L-isoaspartyl methyltransferase in basic fibroblast growth factor-mediated neuronal cell differentiation. BMB Rep 2017; 49:437-42. [PMID: 26973341 PMCID: PMC5070731 DOI: 10.5483/bmbrep.2016.49.8.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Indexed: 11/29/2022] Open
Abstract
We aimed to study the role of protein L-isoaspartyl methyltransferase (PIMT) in neuronal differentiation using basic fibroblast growth factor (bFGF)-induced neuronal differentiation, characterized by cell-body shrinkage, long neurite outgrowth, and expression of neuronal differentiation markers light and medium neurofilaments (NF). The bFGF-mediated neuronal differentiation of PC12 cells was induced through activation of mitogen-activated protein kinase (MAPK) signaling molecules [MAPK kinase 1/2 (MEK1/2), extracellular signal-regulated kinase 1/2 (ERK1/2), and p90RSK], and phosphatidylinositide 3-kinase (PI3K)/Akt signaling molecules PI3Kp110β, PI3Kp110γ, Akt, and mTOR. Inhibitors (adenosine dialdehyde and S-adenosylhomocysteine) of protein methylation suppressed bFGF-mediated neuronal differentiation of PC12 cells. PIMT-eficiency caused by PIMT-specific siRNA inhibited neuronal differentiation of PC12 cells by suppressing phosphorylation of MEK1/2 and ERK1/2 in the MAPK signaling pathway and Akt and mTOR in the PI3K/Akt signaling pathway. Therefore, these results suggested that PIMT was critical for bFGF-mediated neuronal differentiation of PC12 cells and regulated the MAPK and Akt signaling pathways. [BMB Reports 2016; 49(8): 437-442]
Collapse
Affiliation(s)
- To Thi Mai Dung
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Young-Su Yi
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419; Department of Pharmaceutical Engineering, Cheongju University, Cheongju 28503, Korea
| | - Jieun Heo
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Woo Seok Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Ji Hye Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Han Gyung Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Jae Gwang Park
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Byong Chul Yoo
- Colorectal Cancer Branch, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Sungyoul Hong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
37
|
Yang WS, Nam GS, Kim MY, Cho JY. Syk-Mediated Suppression of Inflammatory Responses by Cordyceps bassiana. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:1217-1232. [DOI: 10.1142/s0192415x17500677] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
The fruit body of artificially cultivated Cordyceps bassiana has been reported to exhibit anti-inflammatory and anticancer activities. Although it has been suggested that the fruit body has neutraceutic and pharmaceutic biomaterial potential, the exact anti-inflammatory molecular mechanism has not been fully elucidated. In this study, we demonstrated the immunopharmacologic activity of Cordyceps bassiana under in vitro conditions and investigated its anti-inflammatory mechanism. Water extract (Cm-WE) of the fruit body of artificially cultivated Cordyceps bassiana without polysaccharide fractions reduced the expression of the proinflammatory genes cyclooxygenase (COX)-2, interleukin (IL)-12, and inducible nitric oxide synthase (iNOS) and promoted the expression of the anti-inflammatory gene IL-10 in lipopolysaccharide (LPS)-treated RAW264.7 cells. In addition, this fraction suppressed proliferation and interferon (IFN)-[Formula: see text] production in splenic T lymphocytes. Cm-WE blocked the activation of nuclear factor (NF)-[Formula: see text]B and activator protein (AP)-1 and their upstream inflammatory signaling cascades, including Syk, MEK, and JNK. Using kinase assays, Syk was identified as the target enzyme most strongly inhibited by Cm-WE. These results strongly suggest that Cm-WE suppresses inflammatory responses by inhibiting Syk kinase activity, with potential implications for novel neutraceutic and pharmaceutic biomaterials.
Collapse
Affiliation(s)
- Woo Seok Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Gyeong Sug Nam
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Republic of Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
38
|
Yi YS, Cho JY, Kim D. Cerbera manghas methanol extract exerts anti-inflammatory activity by targeting c-Jun N-terminal kinase in the AP-1 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2016; 193:387-396. [PMID: 27562319 DOI: 10.1016/j.jep.2016.08.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 07/26/2016] [Accepted: 08/20/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cerbera manghas L. (Apocynaceae) is a medicinal plant traditionally used to ameliorate the clinical signs of inflammatory diseases and hypertension. AIM OF STUDY Although C. manghas L. has long been used as a traditional remedy for various diseases, the underlying molecular and cellular mechanisms are poorly understood. A detailed investigation of these mechanisms is necessary to demonstrate the ethnopharmaceutical utility of this plant. MATERIALS AND METHODS The effects of C. manghas methanol extract (Cm-ME) on the production of inflammatory mediators and the expression of proinflammatory cytokines and identification of molecular targets were investigated using lipopolysaccharide (LPS)-treated macrophages in vitro. In addition, the inhibitory effects of Cm-ME orally administered were tested by LPS/D-galactosamine (D-GalN)-induced hepatitis and LPS-induced peritonitis mouse models in vivo. RESULTS Cm-ME downregulated the production of prostaglandin (PG)E2 and the mRNA expression of cyclooxygenase (COX)-2, tumor necrosis factor (TNF)-α, and interleukin (IL)-1β in LPS-stimulated RAW264.7 cells under non-toxic concentration of Cm-ME. This extract inhibited the nuclear translocation of c-Jun and p-ATF2, the phosphorylation of JNK and p38, and AP-1 activity. Western blot analysis and in vitro kinase assay confirmed that JNK is a direct pharmacological target of Cm-ME action. In addition, Cm-ME significantly ameliorated the clinical signs of LPS/D-GalN-induced hepatitis and lowered the production of nitric oxide (NO) and the phosphorylation of JNK in LPS-induced peritonitis conditions. CONCLUSION Cm-ME exerts anti-inflammatory actions on LPS-stimulated macrophages and in mouse models of acute inflammatory disease. These actions are predominantly mediated by targeting JNK in the AP-1 signaling pathway.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Pharmaceutical Engineering, Cheongju University, Cheongju 28503, Republic of Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Daewon Kim
- Laboratory of Bio-informatics, Department of Multimedia Engineering, Dankook University, Republic of Korea.
| |
Collapse
|
39
|
Lee SR, Lee S, Moon E, Park HJ, Park HB, Kim KH. Bioactivity-guided isolation of anti-inflammatory triterpenoids from the sclerotia of Poria cocos using LPS-stimulated Raw264.7 cells. Bioorg Chem 2016; 70:94-99. [PMID: 27912907 DOI: 10.1016/j.bioorg.2016.11.012] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 11/22/2016] [Indexed: 12/12/2022]
Abstract
Poria cocos Wolf (Polyporaceae) has been used as a medicinal fungus to treat various diseases since ancient times. This study aimed to investigate the anti-inflammatory chemical constituents of the sclerotia of P. cocos. Based on bioassay-guided fractionation using lipopolysaccharide (LPS)-stimulated Raw264.7 cells, chemical investigation of the EtOH extract of the sclerotia of P. cocos resulted in the isolation and identification of eight compounds including six triterpenoids, namely poricoic acid A (1), 3-O-acetyl-16α-hydroxydehydrotrametenolic acid (2), polyporenic acid C (3), 3β-hydroxylanosta-7,9(11),24-trien-21-oic acid (4), trametenolic acid (5), and dehydroeburicoic acid (6), as well as (-)-pinoresinol (7) and protocatechualdehyde (8). The structures of the isolated compounds were determined by spectroscopic analysis, including 1H and 13C NMR spectra, and LC/MS analysis. The anti-inflammatory activities of the isolates were evaluated by estimating their effect on the production of nitric oxide (NO) and prostaglandin E2 (PGE2) in LPS-stimulated Raw264.7 as well as on the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Compounds 1-5 inhibited NO production and iNOS expression in LPS-stimulated Raw264.7 cells. Among them, compound 1 exerted the highest anti-inhibitory activity and reduced PGE2 levels via downregulation of COX-2 protein expression. The findings of this study provide experimental evidence that the sclerotia of P. cocos are a potential source of natural anti-inflammatory agents for use in pharmaceuticals and functional foods. Furthermore, the most active compound 1, seco-lanostane triterpenoid, could be a promising lead compound for the development of novel anti-inflammatory agents.
Collapse
Affiliation(s)
- Seoung Rak Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Seulah Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Eunjung Moon
- Charmzone R&D Center, Charmzone Co. Ltd., Seoul 135-851, Republic of Korea
| | - Hye-Jin Park
- Department of Food Science and Biotechnology, College of BioNano Technology, Gachon University, Seongnam 461-701, Republic of Korea
| | - Hyun Bong Park
- Department of Chemistry, Yale University, New Haven, CT 06520, United States
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea.
| |
Collapse
|
40
|
Yang S, Kim Y, Jeong D, Kim JH, Kim S, Son YJ, Yoo BC, Jeong EJ, Kim TW, Lee ISH, Cho JY. Pyrrole-Derivative of Chalcone, ( E)-3-Phenyl-1-(2-Pyrrolyl)-2-Propenone, Inhibits Inflammatory Responses via Inhibition of Src, Syk, and TAK1 Kinase Activities. Biomol Ther (Seoul) 2016; 24:595-603. [PMID: 27469142 PMCID: PMC5098538 DOI: 10.4062/biomolther.2016.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/02/2016] [Accepted: 04/21/2016] [Indexed: 12/26/2022] Open
Abstract
(E)-3-Phenyl-1-(2-pyrrolyl)-2-propenone (PPP) is a pyrrole derivative of chalcone, in which the B-ring of chalcone linked to β-carbon is replaced by pyrrole group. While pyrrole has been studied for possible Src inhibition activity, chalcone, especially the substituents on the B-ring, has shown pharmaceutical, anti-inflammatory, and anti-oxidant properties via inhibition of NF-κB activity. Our study is aimed to investigate whether this novel synthetic compound retains or enhances the pharmaceutically beneficial activities from the both structures. For this purpose, inflammatory responses of lipopolysaccharide (LPS)-treated RAW264.7 cells were analyzed. Nitric oxide (NO) production, inducible NO synthase (iNOS) and tumor necrosis factor-α (TNF-α) mRNA expression, and the intracellular inflammatory signaling cascade were measured. Interestingly, PPP strongly inhibited NO release in a dose-dependent manner. To further investigate this anti-inflammatory activity, we identified molecular pathways by immunoblot analyses of nuclear fractions and whole cell lysates prepared from LPS-stimulated RAW264.7 cells with or without PPP pretreatment. The nuclear levels of p50, c-Jun, and c-Fos were significantly inhibited when cells were exposed to PPP. Moreover, according to the luciferase reporter gene assay after cotransfection with either TRIF or MyD88 in HEK293 cells, NF-κB-mediated luciferase activity dose-dependently diminished. Additionally, it was confirmed that PPP dampens the upstream signaling cascade of NF-κB and AP-1 activation. Thus, PPP inhibited Syk, Src, and TAK1 activities induced by LPS or induced by overexpression of these genes. Therefore, our results suggest that PPP displays anti-inflammatory activity via inhibition of Syk, Src, and TAK1 activity, which may be developed as a novel anti-inflammatory drug.
Collapse
Affiliation(s)
- Sungjae Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yong Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Deok Jeong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jun Ho Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sunggyu Kim
- Research and Business Foundation, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Young-Jin Son
- College of Pharmacy, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Byong Chul Yoo
- Colorectal Cancer Branch, Research Institute, National Cancer Center, Gyeonggi 10408, Republic of Korea
| | - Eun Jeong Jeong
- Department of Science Education, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Tae Woong Kim
- Department of Biochemistry, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - In-Sook Han Lee
- Department of Science Education, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
41
|
Eom HJ, Lee D, Lee S, Noh HJ, Hyun JW, Yi PH, Kang KS, Kim KH. Flavonoids and a Limonoid from the Fruits of Citrus unshiu and Their Biological Activity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:7171-7178. [PMID: 27608132 DOI: 10.1021/acs.jafc.6b03465] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The fruits of Citrus unshiu are one of the most popular and most enjoyed fruits in Korea. As we continue to seek for bioactive metabolites from Korean natural resources, our study on the chemical constituents of the fruits of C. unshiu resulted in the isolation of a new flavonoid glycoside, limocitrunshin 1, along with seven other flavonoids 2-8 and a limonoid 9. All structures were identified by spectroscopic methods, namely 1D and 2D NMR, including HSQC, HMBC, and (1)H-(1)H COSY experiments, HRMS, and other chemical methods. Compounds 3, 5, and 9 are reported to be isolated from this fruit for the first time. The isolated compounds were applied to activity tests to verify their inhibitory effects on inflammation and nephrotoxicity. Compounds 6 and 9 showed the most potent inhibitory activity on renal cell damage and nitric oxide production, respectively. Thus, the fruits of C. unshiu could serve as a valuable natural source of bioactive components with health benefits for potential application in functional foods.
Collapse
Affiliation(s)
- Hee Jeong Eom
- School of Pharmacy, Sungkyunkwan University , Suwon 440-746, Republic of Korea
| | - Dahae Lee
- School of Pharmacy, Sungkyunkwan University , Suwon 440-746, Republic of Korea
- College of Korean Medicine, Gachon University , Seongnam 461-701, Republic of Korea
| | - Seulah Lee
- School of Pharmacy, Sungkyunkwan University , Suwon 440-746, Republic of Korea
| | - Hyung Jun Noh
- Department of Herbal Crop Research, National Institute of Horticultural & Herbal Science, Rural Development Administration , Eumseoung 369-873, Republic of Korea
| | - Jae Wook Hyun
- Citrus Research Station, National Institute of Horticultural & Herbal Science, Rural Development Administration , Jeju 697-943, Republic of Korea
| | - Pyoung Ho Yi
- Citrus Research Station, National Institute of Horticultural & Herbal Science, Rural Development Administration , Jeju 697-943, Republic of Korea
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University , Seongnam 461-701, Republic of Korea
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University , Suwon 440-746, Republic of Korea
| |
Collapse
|
42
|
Boddawar GD, Dhawale SC, Shaikh SS. Assessment of anti-inflammatory potential of Sesbania bispinosa Linn. leaf extracts and fractions by acute and chronic models. ALEXANDRIA JOURNAL OF MEDICINE 2016. [DOI: 10.1016/j.ajme.2015.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
| | | | - Shafik S. Shaikh
- School of Physical Sciences, SRTM University, Nanded 431606, MS, India
| |
Collapse
|
43
|
Kim MY, Cho JY. Molecular association of CD98, CD29, and CD147 critically mediates monocytic U937 cell adhesion. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2016; 20:515-23. [PMID: 27610038 PMCID: PMC5014998 DOI: 10.4196/kjpp.2016.20.5.515] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/05/2016] [Accepted: 07/25/2016] [Indexed: 12/15/2022]
Abstract
Adhesion events of monocytes represent an important step in inflammatory responses induced by chemokines. The β1-integrin CD29 is a major adhesion molecule regulating leukocyte migration and extravasation. Although several adhesion molecules have been known as regulators of CD29, the molecular interactions between CD29 and its regulatory adhesion molecules (such as CD98 and CD147) have not been fully elucidated. Therefore, in this study, we examined whether these molecules are functionally, biochemically, and cell-biologically associated using monocytic U937 cells treated with aggregation-stimulating and blocking antibodies, as well as enzyme inhibitors. The surface levels of CD29, CD98, and CD147 (but not CD43, CD44, and CD82) were increased. The activation of CD29, CD98, and CD147 by ligation of them with aggregation-activating antibodies triggered the induction of cell-cell adhesion, and sensitivity to various enzyme inhibitors and aggregation-blocking antibodies was similar for CD29-, CD98-, and CD147-induced U937 cell aggregation. Molecular association between these molecules and the actin cytoskeleton was confirmed by confocal microscopy and immunoprecipitation. These results strongly suggest that CD29 might be modulated by its biochemical and cellular regulators, including CD98 and CD147, via the actin cytoskeleton.
Collapse
Affiliation(s)
- Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Korea
| | - Jae Youl Cho
- Depatment of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
44
|
Baek KS, Yi YS, Son YJ, Yoo S, Sung NY, Kim Y, Hong S, Aravinthan A, Kim JH, Cho JY. In vitro and in vivo anti-inflammatory activities of Korean Red Ginseng-derived components. J Ginseng Res 2016; 40:437-444. [PMID: 27746698 PMCID: PMC5052440 DOI: 10.1016/j.jgr.2016.08.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/07/2016] [Accepted: 08/09/2016] [Indexed: 11/05/2022] Open
Abstract
Background Although Korean Red Ginseng (KRG) has been traditionally used for a long time, its anti-inflammatory role and underlying molecular and cellular mechanisms have been poorly understood. In this study, the anti-inflammatory roles of KRG-derived components, namely, water extract (KRG-WE), saponin fraction (KRG-SF), and nonsaponin fraction (KRG-NSF), were investigated. Methods To check saponin levels in the test fractions, KRG-WE, KRG-NSF, and KRG-SF were analyzed using high-performance liquid chromatography. The anti-inflammatory roles and underlying cellular and molecular mechanisms of these components were investigated using a macrophage-like cell line (RAW264.7 cells) and an acute gastritis model in mice. Results Of the tested fractions, KGR-SF (but not KRG-NSF and KRG-WE) markedly inhibited the viability of RAW264.7 cells, and splenocytes at more than 500 μg/mL significantly suppressed NO production at 100 μg/mL, diminished mRNA expression of inflammatory genes such as inducible nitric oxide synthase, cyclooxygenase-2, tumor necrosis factor-α, and interferon-β at 200 μg/mL, and completely blocked phagocytic uptake by RAW264.7 cells. All three fractions suppressed luciferase activity triggered by interferon regulatory factor 3 (IRF3), but not that triggered by activator protein-1 and nuclear factor-kappa B. Phospho-IRF3 and phospho-TBK1 were simultaneously decreased in KRG-SF. Interestingly, all these fractions, when orally administered, clearly ameliorated the symptoms of gastric ulcer in HCl/ethanol-induced gastritis mice. Conclusion These results suggest that KRG-WE, KRG-NSF, and KRG-SF might have anti-inflammatory properties, mostly because of the suppression of the IRF3 pathway.
Collapse
Affiliation(s)
- Kwang-Soo Baek
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Korea
| | - Young-Su Yi
- Department of Pharmaceutical Engineering, Cheongju University, Cheongju, Korea
| | - Young-Jin Son
- Department of Pharmacy, Sunchon National University, Suncheon, Korea
| | - Sulgi Yoo
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Korea
| | - Nak Yoon Sung
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Korea
| | - Yong Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Korea
| | - Sungyoul Hong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Korea
| | - Adithan Aravinthan
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan, Korea
| | - Jong-Hoon Kim
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan, Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
45
|
Lin Y, Li Y, Song ZG, Zhu H, Jin YH. The interaction of serum albumin with ginsenoside Rh2 resulted in the downregulation of ginsenoside Rh2 cytotoxicity. J Ginseng Res 2016; 41:330-338. [PMID: 28701874 PMCID: PMC5489766 DOI: 10.1016/j.jgr.2016.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/21/2016] [Accepted: 06/23/2016] [Indexed: 12/22/2022] Open
Abstract
Background Ginsenoside Rh2 (G-Rh2) is a ginseng saponin that is widely investigated because of its remarkable antitumor activity. However, the molecular mechanism by which (20S) G-Rh2 triggers its functions and how target animals avoid its cytotoxic action remains largely unknown. Methods Phage display was used to screen the human targets of (20S) G-Rh2. Fluorescence spectroscopy and UV-visible absorption spectroscopy were used to confirm the interaction of candidate target proteins and (20S) G-Rh2. Molecular docking was utilized to calculate the estimated free energy of binding and to structurally visualize their interactions. MTT assay and immunoblotting were used to assess whether human serum albumin (HSA), bovine serum albumin (BSA), and bovine serum can reduce the cytotoxic activity of (20S) G-Rh2 in HepG2 cells. Results In phage display, (20S) G-Rh2-beads and (20R) G-Rh2-beads were combined with numerous kinds of phages, and a total of 111 different human complementary DNAs (cDNA) were identified, including HSA which had the highest rate. The binding constant and number of binding site in the interaction between (20S)-Rh2 and HSA were 3.5 × 105 M−1 and 1, and those in the interaction between (20S) G-Rh2 and BSA were 1.4 × 105 M−1 and 1. The quenching mechanism is static quenching. HSA, BSA and bovine serum significantly reduced the proapoptotic effect of (20S) G-Rh2. Conclusion HSA and BSA interact with (20S) G-Rh2. Serum inhibited the activity of (20S) G-Rh2 mainly due to the interaction between (20S) G-Rh2 and serum albumin (SA). This study proposes that HSA may enhance (20S) G-Rh2 water solubility, and thus might be used as nanoparticles in the (20S) G-Rh2 delivery process.
Collapse
Affiliation(s)
- Yingjia Lin
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University, Changchun, Jilin, China
| | - Yang Li
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University, Changchun, Jilin, China
| | - Zhi-Guang Song
- College of Chemistry, Jilin University, Changchun, China
| | - Hongyan Zhu
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University, Changchun, Jilin, China
| | - Ying-Hua Jin
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University, Changchun, Jilin, China
| |
Collapse
|
46
|
Syk and IRAK1 Contribute to Immunopharmacological Activities of Anthraquinone-2-carboxlic Acid. Molecules 2016; 21:molecules21060809. [PMID: 27338330 PMCID: PMC6272897 DOI: 10.3390/molecules21060809] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 06/16/2016] [Accepted: 06/18/2016] [Indexed: 12/12/2022] Open
Abstract
Anthraquinone-2-carboxlic acid (9,10-dihydro-9,10-dioxo-2-anthracenecarboxylic acid, AQCA) was identified as one of the major anthraquinones in Brazilian taheebo. Since there was no report explaining its immunopharmacological actions, in this study, we aimed to investigate the molecular mechanism of AQCA-mediated anti-inflammatory activity using reporter gene assays, kinase assays, immunoblot analyses, and overexpression strategies with lipopolysaccharide (LPS)-treated macrophages. AQCA was found to suppress the release of nitric oxide (NO) and prostaglandin (PG) E2 from LPS-treated peritoneal macrophages without displaying any toxic side effects. Molecular analysis revealed that AQCA was able to inhibit the activation of the nuclear factor (NF)-κB and activator protein (AP)-1 pathways by direct suppression of upstream signaling enzymes including interleukin-1 receptor-associated kinase 1 (IRAK1) and spleen tyrosine kinase (Syk). Therefore, our data strongly suggest that AQCA-mediated suppression of inflammatory responses could be managed by a direct interference of signaling cascades including IRAK and Syk, linked to the activation of NF-κB and AP-1.
Collapse
|
47
|
1-(2,3-Dibenzimidazol-2-ylpropyl)-2-methoxybenzene Is a Syk Inhibitor with Anti-Inflammatory Properties. Molecules 2016; 21:508. [PMID: 27096863 PMCID: PMC6274291 DOI: 10.3390/molecules21040508] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/12/2016] [Accepted: 04/14/2016] [Indexed: 01/15/2023] Open
Abstract
Inflammation is the protective action of our bodies against external pathogens by recognition of pathogen-associated molecular patterns (PAMPs) via pattern recognition receptors (PRRs). Proper regulation of inflammatory responses is required to maintain our body’s homeostasis, as well as there are demands to develop proper acute or chronic inflammation. In this study, we elucidated the regulatory mechanism of NF-κB-mediated inflammatory responses by a novel compound, 1-(2,3-dibenzimidazol-2-ylpropyl)-2-methoxybenzene (DBMB). We found that DBMB suppressed inflammatory mediators, nitric oxide (NO) and prostaglandin E2 (PGE2), reacted to exposure to a number of toll like receptor (TLR) ligands. Such observations occurred following to decreased mRNA expression of several pro-inflammatory mediators, and such diminished mRNA levels were caused by inhibited transcriptional factor nuclear factor (NF)-κB, as evaluated by luciferase reporter assay and molecular biological approaches. To find the potential targets of DBMB, we screened phosphorylated forms of NF-κB signal molecules: inhibitor of κBα (IκBα), IκB kinase (IKK)α/β, Akt, 3-phosphoinositide dependent protein kinase-1 (PDK1), p85, and spleen tyrosine kinase (Syk). We found that DBMB treatment could suppress signal transduction through these molecules. Additionally, we conducted in vitro kinase assays using immunoprecipitated Syk and its substrate, p85. Consequently, we could say that DBMB clearly suppressed the kinase activity of Syk kinase activity. Together, our results demonstrate that synthetic DBMB has an effect on the inflammatory NF-κB signaling pathway and suggest the potential for clinical use in the treatment of inflammatory diseases.
Collapse
|
48
|
Inhibition of hypoxia-induced cyclooxygenase-2 by Korean Red Ginseng is dependent on peroxisome proliferator-activated receptor gamma. J Ginseng Res 2016; 41:240-246. [PMID: 28701863 PMCID: PMC5489747 DOI: 10.1016/j.jgr.2016.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/19/2016] [Accepted: 04/02/2016] [Indexed: 12/22/2022] Open
Abstract
Background Korean Red Ginseng (KRG) is a traditional herbal medicine made by steaming and drying fresh ginseng. It strengthens the endocrine and immune systems to ameliorate various inflammatory responses. The cyclooxygenase-2 (COX-2)/prostaglandin E2 pathway has important implications for inflammation responses and tumorigenesis. Peroxisome proliferator-activated receptor gamma (PPARγ) is a transcription factor that regulates not only adipogenesis and lipid homeostasis, but also angiogenesis and inflammatory responses. Methods The effects of the KRG on inhibition of hypoxia-induced COX-2 via PPARγ in A549 cells were determined by luciferase assay, Western blot, and/or quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The antimigration and invasive effects of KRG were evaluated on A549 cells using migration and matrigel invasion assays. Results and conclusion We previously reported that hypoxia-induced COX-2 protein and mRNA levels were suppressed by KRG. This study examines the possibility of PPARγ as a cellular target of KRG for the suppression of hypoxia-induced COX-2. PPARγ protein levels and PPARγ-responsive element (PPRE)-driven reporter activities were increased by KRG. Reduction of hypoxia-induced COX-2 by KRG was abolished by the PPARγ inhibitor GW9662. In addition, the inhibition of PPARγ abolished the effect of KRG on hypoxia-induced cell migration and invasion. Discussion Our results show that KRG inhibition of hypoxia-induced COX-2 expression and cell invasion is dependent on PPARγ activation, supporting the therapeutic potential for suppression of inflammation under hypoxia. Further studies are required to demonstrate whether KRG activates directly PPARγ and to identify the constituents responsible for this activity.
Collapse
|
49
|
Cho YK, Kim JE, Woo JH. Genetic defects in the nef gene are associated with Korean Red Ginseng intake: monitoring of nef sequence polymorphisms over 20 years. J Ginseng Res 2016; 41:144-150. [PMID: 28413318 PMCID: PMC5386103 DOI: 10.1016/j.jgr.2016.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/19/2016] [Accepted: 02/19/2016] [Indexed: 01/11/2023] Open
Abstract
Background The presence of gross deletions in the human immunodeficiency virus nef gene (gΔnef) is associated with long-term nonprogression of infected patients. Here, we investigated how quickly genetic defects in the nef gene are associated with Korean Red Ginseng (KRG) intake in 10 long-term slow progressors. Methods This study was divided into three phases over a 20-yr period; baseline, KRG intake alone, and KRG plus highly active antiretroviral therapy (ART). nef gene amplicons were obtained using reverse transcription polymerase chain reaction (PCR) and nested PCR from 10 long-term slow progressors (n = 1,396), and nested PCR from 36 control patients (n = 198), and 28 ART patients (n = 157), and these were then sequenced. The proportion of gΔnef, premature stop codons, and not in-frame insertion or deletion of a nucleotide was compared between three phases, control, and ART patients. Results The proportion of defective nef genes was significantly higher in on-KRG patients (15.6%) than in baseline (5.7%), control (5.6%), on-KRG plus ART phase (7.8%), and on-ART patients (6.6%; p < 0.01). Small in-frame deletions or insertions were significantly more frequent among patients treated with KRG alone compared with controls (p < 0.01). Significantly fewer instances of genetic defects were detected in samples taken during the KRG plus ART phase (7.8%; p < 0.01). The earliest defects detected were gΔnef and small in-frame deletions after 7 mo and 67 mo of KRG intake, respectively. Conclusion KRG treatment might induce genetic defects in the nef gene. This report provides new insight into the importance of genetic defects in the pathogenesis of AIDS.
Collapse
Affiliation(s)
- Young-Keol Cho
- Department of Microbiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jung-Eun Kim
- Department of Microbiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jun-Hee Woo
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
50
|
Colzani M, Altomare A, Caliendo M, Aldini G, Righetti PG, Fasoli E. The secrets of Oriental panacea: Panax ginseng. J Proteomics 2016; 130:150-9. [DOI: 10.1016/j.jprot.2015.09.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/11/2015] [Accepted: 09/17/2015] [Indexed: 02/05/2023]
|