1
|
Zheng W, Li Y, Wang Y, He J, Zhao Q, Huang A. Identification of a novel peptide with anti-inflammatory activity from Binglangjiang buffalo fermented milk and its potential inhibitory mechanism in lipopolysaccharide-stimulated RAW264.7 cells. Food Chem 2024; 468:142451. [PMID: 39700804 DOI: 10.1016/j.foodchem.2024.142451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/08/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Binglangjiang buffalo fermented milk (BBFM) is rich in bioactive peptides and exhibits significant immunomodulatory activity, although the specific components and effects are not well understood. In this study, we investigated the anti-inflammatory activity peptides from BBFM using peptidomics and proteomics. A total of 769 peptides were identified using LC-MS/MS. Among these, a novel peptide (GPGAPADPGRPTG (GG13)), was screened out using LPS-stimulated RAW264.7 cells. The molecular weight of peptide GG13 was 1149.56 Da, and it exhibited high in-vitro safety and thermal stability. Furthermore, ELISA and Western blot analysis showed that peptide GG13 significantly inhibited the secretion of pro-inflammatory cytokines NO and TNF-α, as well as the expression of proteins iNOS and TNF-α in LPS-stimulated RAW264.7 cells, and proteomics analysis showed that peptide GG13 significantly down-regulated the protein expression of STAT1, NOS2, COX2, and CD40. The study provides a basis for further explore the development of health fermented milk products.
Collapse
Affiliation(s)
- Wentao Zheng
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Yufang Li
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Yuzhu Wang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Jinze He
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Qiong Zhao
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, Yunnan, China.
| | - Aixiang Huang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China.
| |
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
|
You X, Liu H, Chen Y, Zhao G. Multifunctional Liposomes Co-Modified with Ginsenoside Compound K and Hyaluronic Acid for Tumor-Targeted Therapy. Polymers (Basel) 2024; 16:405. [PMID: 38337294 DOI: 10.3390/polym16030405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Liposomes show promise for anti-cancer drug delivery and tumor-targeted therapy. However, complex tumor microenvironments and the performance limitations of traditional liposomes restrict clinical translation. Hyaluronic acid (HA)-modified nanoliposomes effectively target CD44-overexpressing tumor cells. Combination therapy enhances treatment efficacy and delays drug resistance. Here, we developed paclitaxel (PTX) liposomes co-modified with ginsenoside compound K (CK) and HA using film dispersion. Compared to cholesterol (Ch), CK substantially improved encapsulation efficiency and stability. In vitro release studies revealed pH-responsive behavior, with slower release at pH 7.4 versus faster release at pH 5. In vitro cytotoxicity assays demonstrated that replacing Ch with CK in modified liposomes considerably decreased HCT-116 cell viability. Furthermore, flow cytometry and fluorescence microscopy showed a higher cellular uptake of PTX-CK-Lip-HA in CD44-high cells, reflected in the lower half maximal inhibitory concentrations. Overall, CK/HA-modified liposomes represent an innovative, targeted delivery system for enhanced tumor therapy via pH-triggered drug release and CD44 binding.
Collapse
Affiliation(s)
- Xiaoyan You
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Hui Liu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China
- Haihe Laboratory of Synthetic Biology, Tianjin 300308, China
| | - Yue Chen
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Guoping Zhao
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
- Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
4
|
Kim H, Jeong EJ, Hwang B, Lee HD, Lee S, Jang M, Yeo K, Shin Y, Park S, Lim WT, Kim WJ, Moon SK. Pharmacological effects of biologically synthesized ginsenoside CK-rich preparation (AceCK40) on the colitis symptoms in DSS-induced Caco-2 cells and C57BL mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 124:155301. [PMID: 38181531 DOI: 10.1016/j.phymed.2023.155301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/13/2023] [Accepted: 12/17/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND Despite the notable pharmacological potential of natural ginsenosides, their industrial application is hindered by low oral bioavailability. Recent research centers on the production of less-glycosylated minor ginsenosides. PURPOSE This study aimed to explore the effect of a biologically synthesized ginsenoside CK-rich minor ginsenoside complex (AceCK40), on ameliorating colitis using DSS-induced colitis models in vitro and in vivo. METHODS The ginsenoside composition of AceCK40 was determined by HPLC-ELSD and UHPLC-MS/MS analyses. In vitro colitis model was established using dextran sodium sulfate (DSS)-induced Caco-2 intestinal epithelial model. For in vivo experiments, DSS-induced severe colitis mouse model was established. RESULTS In DSS-stimulated Caco-2 cells, AceCK40 downregulated mitogen-activated protein kinase (MAPK) activation (p < 0.05), inhibited monocyte chemoattractant protein-1 (MCP-1) production (p < 0.05), and enhanced MUC2 expression (p < 0.05), mediated via signaling pathway regulation. Daily AceCK40 administration at doses of 10 and 30 mg/kg/day was well tolerated by DSS-induced severe colitis mice. These doses led to significant alleviation of disease activity index score (> 36.0% decrease, p < 0.05), increased luminal immunoglobulin (Ig)G (> 37.6% increase, p < 0.001) and IgA (> 33.8% increase, p < 0.001), lowered interleukin (IL)-6 (> 65.7% decrease, p < 0.01) and MCP-1 (> 116.2% decrease, p < 0.05), as well as elevated serum IgA (> 51.4% increase, p < 0.001) and lowered serum IL-6 (112.3% decrease at 30 mg/kg, p < 0.001). Hematoxylin and eosin (H&E) and periodic acid-Schiff (PAS) staining revealed that DSS-mediated thickening of the muscular externa, extensive submucosal edema, crypt distortion, and decreased mucin droplets were significantly alleviated by AceCK40 administration. Additionally, daily administration of AceCK40 led to significant recovery of colonic tight junctions damaged by DSS through the elevation in the expression of adhesion molecules, including occludin, E-cadherin, and N-cadherin. CONCLUSION This study presents the initial evidence elucidating the anti-colitis effects of AceCK40 and its underlying mechanism of action through sequential in vitro and in vivo systems employing DSS stimulation. Our findings provide valuable fundamental data for the utilization of AceCK40 in the development of novel anti-colitis candidates.
Collapse
Affiliation(s)
- Hoon Kim
- Department of Food and Nutrition, Chung-Ang University, Anseong 17546, South Korea
| | - Eun-Jin Jeong
- Department of Integrated Biomedical and Life Sciences, Korea University, Seoul 02841, South Korea
| | - Byungdoo Hwang
- Department of Food and Nutrition, Chung-Ang University, Anseong 17546, South Korea
| | - Hak-Dong Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong 17546, South Korea
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong 17546, South Korea
| | - Mi Jang
- The Food Industry Promotional Agency of Korea, Iksan, South Korea
| | - Kwangeun Yeo
- The Food Industry Promotional Agency of Korea, Iksan, South Korea
| | - Yunjeong Shin
- The Food Industry Promotional Agency of Korea, Iksan, South Korea
| | - Sanghoon Park
- The Food Industry Promotional Agency of Korea, Iksan, South Korea
| | - Wan Taek Lim
- Research Institute, AceEMzyme, Anseong, South Korea
| | - Woo Jung Kim
- Biocenter, Gyeonggido Business and Science Accelerator, Suwon 16229, South Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung-Ang University, Anseong 17546, South Korea.
| |
Collapse
|
5
|
Kim WH, Choi WJ, Kim JE, Choi J, Hong YD, Nam J, Park WS, Shim SM. Kinetic conversion of BIOGF1K enriched in compound K from in vitro 3-D human tissue model. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2023; 5:100165. [PMID: 37869704 PMCID: PMC10589745 DOI: 10.1016/j.crphar.2023.100165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/24/2023] Open
Abstract
The purposes of current study were to investigate the effect of ginsenosides from BIOGF1K enriched in compound K (CK) and compound Y (CY) on the skin barrier function, the deposition in in vitro 3-D human tissue model (EpiDermFT™ Full Thickness 400), and to identify and quantify kinetic bioconversion of the ginsenosides in artificial skin by utilizing the Fourier transform infrared spectroscopy (FT-IR) and liquid chromatography mass spectrometry (LC-MS), respectively. Epidermal barrier integrity evaluated using transepithelial electrical resistance (TEER) was significantly higher in the BIOGF1K treatment than the CY or CK individual treatment throughout incubation (p < 0.05). Skin deposition (%) of CY and CK from BIOGF1K treatment was approximately 4 and 2 times higher than the CY and CK single component treatment, respectively. Total amount of CK found in human skin by deposition and bioconversion was approximately 1087.3, 528.82, and 867.76 μM after topical treatment of BIOGF1K, CK, and CY. Results from the current study reveal that topical treatment of BIOGF1K more effectively induced CK deposition as well as bioconversion of CY to CK than that of a single treatment of CY or CK, suggesting that BIOGF1K could be a useful cosmetic preparation for enhancing skin function.
Collapse
Affiliation(s)
- Woo-Hyun Kim
- Department of Food Science and Biotechnology, Sejong University, 98 Gunja-dong, Seoul, 05006, South Korea
| | - Won-Jo Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Jeong-Eun Kim
- Department of Food Science and Biotechnology, Sejong University, 98 Gunja-dong, Seoul, 05006, South Korea
| | - Joonho Choi
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si, 17074, Gyeonggi-do, South Korea
| | - Yong-Deok Hong
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si, 17074, Gyeonggi-do, South Korea
| | - Jin Nam
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si, 17074, Gyeonggi-do, South Korea
| | - Won-Seok Park
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si, 17074, Gyeonggi-do, South Korea
| | - Soon-Mi Shim
- Department of Food Science and Biotechnology, Sejong University, 98 Gunja-dong, Seoul, 05006, South Korea
| |
Collapse
|
6
|
Hu F, Liu C, Wang F, Zhou C, Zhu M, Sun-Waterhouse D, Wang Z. Phenolic compounds from Chaenomeles speciosa alleviate inflammation in lipopolysaccharide-treated RAW264.7 macrophages via the NF-κB and MAPK pathways. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2022.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
7
|
Choi S, Kim T. Compound K-An immunomodulator of macrophages in inflammation. Life Sci 2023; 323:121700. [PMID: 37068708 DOI: 10.1016/j.lfs.2023.121700] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/19/2023]
Abstract
Compound K (CK) is a secondary ginsenoside biotransformed from ginseng. This review discusses the function of CK as a potential ligand of the glucocorticoid receptor and a regulator of macrophage inflammatory responses. We provide findings on the ability of CK to inhibit the activation of M1 macrophages and promote the activation and differentiation of M2 macrophages. In addition, the effect of inhibiting the inflammasome response was collected. We summarized the evidences that CK is effective in the treatment of various inflammatory diseases such as rheumatoid arthritis, systemic lupus erythematosus, dermatitis, asthma, chronic obstructive pulmonary disease, sepsis associated encephalopathy, atherosclerosis, inflammatory bowel disease, and diabetes. These findings suggest the potential of CK as a therapeutic agent that can resolve inflammation and restore homeostasis.
Collapse
Affiliation(s)
- Susanna Choi
- KM Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero Yuseong-gu, Daejeon 34054, Republic of Korea.
| | - Taesoo Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero Yuseong-gu, Daejeon 34054, Republic of Korea
| |
Collapse
|
8
|
Kim WH, Kim JE, Kim S, Na Y, Hong YD, Choi J, Park WS, Shim SM. Bioconversion of BIOGF1K, a compound-K-rich fraction from ginseng root and its effect on epidermal barrier function. Heliyon 2023; 9:e14803. [PMID: 37025761 PMCID: PMC10070716 DOI: 10.1016/j.heliyon.2023.e14803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
BIOGF1K, the ginseng root-based and hydrolyzed ginsenoside-rich fraction, is known to improve skin damage, but there are rare studies on the kinetic of ginsenosides in the epidermis and their effects on epidermal barrier function. The current study investigated the effect of BIOGF1K on epidermal barrier function and its kinetics on epidermal transport. HPLC and LC/MS were used to verify the ginsenosides and the metabolites of BIOGF1K. Human immortalized keratinocytes (HaCaT) and epidermis-dermis artificial skin were treated with BIOGF1K and their metabolites were analyzed by HPLC and LC/MS. The epidermal barrier function was evaluated by transepithelial electrical resistance (TEER). In BIOGF1K, ginsenoside Rg1, Rd, F1, F2, compound Mc, compound Y (CY), and compound K (CK) were detected and CK and CY were the most and second abundant ginsenosides. TEER of HaCaT with 100 and 200 μg/mL BIOGF1K treatment was significantly higher than the control during 600 min of incubation. CK was permeated to the epidermis in a time-dependent manner and its maximum transported rate was observed at 600 min. In the case of artificial skin, CY and CK were permeated to the epidermis-dermis skin as time-dependent. Also, 24 h after treatment of CY, CK was detected as 19.59% of CY. It was proposed that CY was hydrolyzed into CK while permeating the epidermis. Results from the current study suggest that bioconversion of BIOGF1K rich in CK effectively enhances epidermal barrier function and it could be a useful cosmeceutical to exhibit its functionality to the skin.
Collapse
Affiliation(s)
- Woo-Hyun Kim
- Department of Food Science and Biotechnology, Sejong University, 98 Gunja-dong, Seoul 05006, South Korea
| | - Jeong-Eun Kim
- Department of Food Science and Biotechnology, Sejong University, 98 Gunja-dong, Seoul 05006, South Korea
| | - Sehyun Kim
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si 17074, Gyeonggi-do, South Korea
| | - Yongjoo Na
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si 17074, Gyeonggi-do, South Korea
| | - Yong-Deok Hong
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si 17074, Gyeonggi-do, South Korea
| | - Joonho Choi
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si 17074, Gyeonggi-do, South Korea
| | - Won-Seok Park
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si 17074, Gyeonggi-do, South Korea
- Corresponding author.
| | - Soon-Mi Shim
- Department of Food Science and Biotechnology, Sejong University, 98 Gunja-dong, Seoul 05006, South Korea
- Corresponding author.
| |
Collapse
|
9
|
Kim JH, Park JG, Hong YH, Shin KK, Kim JK, Kim YD, Yoon KD, Kim KH, Yoo BC, Sung GH, Cho JY. Sauropus brevipes ethanol extract negatively regulates inflammatory responses in vivo and in vitro by targeting Src, Syk and IRAK1. PHARMACEUTICAL BIOLOGY 2021; 59:74-86. [PMID: 33439064 PMCID: PMC7808742 DOI: 10.1080/13880209.2020.1866024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
CONTEXT Sauropus brevipes Müll. Arg. (Phyllanthaceae) has been used as an effective ingredient in a decoction for the treatment of diarrhoea. However, there was no report on its modulatory role in inflammation. OBJECTIVE This study investigates anti-inflammatory effect of S. brevipes in various inflammation models. MATERIALS AND METHODS The aerial part of S. brevipes was extracted with 95% ethanol to produce Sb-EE. RAW264.7 cells pre-treated with Sb-EE were stimulated by lipopolysaccharide (LPS), and Griess assay and PCR were performed. High-performance liquid chromatography (HPLC) analysis, luciferase assay, Western blotting and kinase assay were employed. C57BL/6 mice (10 mice/group) were orally administered with Sb-EE (200 mg/kg) once a day for five days, and peritonitis was induced by an intraperitoneal injection of LPS (10 mg/kg). ICR mice (four mice/group) were orally administered with Sb-EE (20 or 200 mg/kg) or ranitidine (positive control) twice a day for two days, and EtOH/HCl was orally injected to induce gastritis. RESULTS Sb-EE suppressed nitric oxide (NO) release (IC50=34 µg/mL) without cytotoxicity and contained flavonoids (quercetin, luteolin and kaempferol). Sb-EE (200 µg/mL) reduced the mRNA expression of inducible NO synthase (iNOS). Sb-EE blocked the activities of Syk and Src, while inhibiting interleukin-1 receptor associated kinases (IRAK1) by 68%. Similarly, orally administered Sb-EE (200 mg/kg) suppressed NO production by 78% and phosphorylation of Src and Syk in peritonitis mice. Sb-EE also decreased inflammatory lesions in gastritis mice. DISCUSSION AND CONCLUSIONS This study demonstrates the inhibitory effect of Sb-EE on the inflammatory response, suggesting that Sb-EE can be developed as a potential anti-inflammatory agent.
Collapse
Affiliation(s)
- Ji Hye Kim
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
| | - Jae Gwang Park
- Division of Translational Science, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Yo Han Hong
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
| | - Kon Kuk Shin
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
| | - Jin Kyeong Kim
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
| | - Young-Dong Kim
- Department of Life Science, Hallym University, Chuncheon, Republic of Korea
| | - Ki Dong Yoon
- College of Pharmacy, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Kyung-Hee Kim
- Proteomic Analysis Team, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Byong Chul Yoo
- Division of Translational Science, Research Institute, National Cancer Center, Goyang, Republic of Korea
- Byong Chul Yoo Division of Translational Science, Research Institute, National Cancer Center, Goyang, 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
- CONTACT Gi-Ho Sung Institute for Bio-Medical Convergence, International St. Mary’s Hospital and College of Medicine, Catholic Kwandong University, Incheon, Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
- Jae Youl Cho Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
10
|
Suwandi DW, Rostnawati T, Muchtaridi M, Subarnas A. In vitro evaluation of selligueain A effects on the proinflammatory mediators production in RAW264.7 murine macrophages. JOURNAL OF HERBMED PHARMACOLOGY 2021. [DOI: 10.34172/jhp.2021.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Introduction: Selligueain A derived from the roots of Polypodium feei was shown to have anti-inflammatory activity, which was tested in vivo on the rats’ paw edema induced by carrageenan. The aim of this study was to evaluate the anti-inflammatory mechanism of selligueain A in vitro against the production of pro-inflammatory mediators. Methods: In this study, RAW264.7 cells were used as an inflammatory cell model, and observations were made on the inflammatory mediators nitric oxide (NO), inducible nitric oxide synthase (iNOS), and tumour necrosis factor-α (TNF-α). The NO concentration was measured by the Griess reaction, and the iNOS enzyme and the TNF-α concentrations were determined by the ELISA method. Cell viability was assessed by the [3-(4,5-dimethylthiazol-2yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] (MTS) test. Results: Selligueain A at concentrations of 100 and 150 µM suppressed the production of NO, iNOS, and TNF-α in RAW264.7 cells stimulated by lipopolysaccharide (LPS). The concentration of 150 µM showed the highest inhibition of NO, iNOS, and TNF-α mediators with the percentage inhibition of 64.85, 55.01, and 48.54%, respectively. Conclusion: This study shows that selligueain A has anti-inflammatory activity through inhibition of NO, iNOS, and TNF-α production in RAW264.7 macrophage cells.
Collapse
Affiliation(s)
- Deden Winda Suwandi
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Sumedang, Indonesia
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Universitas Garut, Garut, Indonesia
| | - Tina Rostnawati
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Sumedang, Indonesia
| | - Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Sumedang, Indonesia
| | - Anas Subarnas
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Sumedang, Indonesia
| |
Collapse
|
11
|
Kim JK, Choi E, Hong YH, Kim H, Jang YJ, Lee JS, Choung ES, Woo BY, Hong YD, Lee S, Lee BH, Bach TT, Kim JH, Kim JH, Cho JY. Syk/NF-κB-targeted anti-inflammatory activity of Melicope accedens (Blume) T.G. Hartley methanol extract. JOURNAL OF ETHNOPHARMACOLOGY 2021; 271:113887. [PMID: 33539951 DOI: 10.1016/j.jep.2021.113887] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 05/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Melicope accedens (Blume) Thomas G. Hartley is a plant included in the family Rutaceae and genus Melicope. It is a native plant from Vietnam that has been used for ethnopharmacology. In Indonesia and Malaysia, the leaves of M. accedens are applied externally to decrease fever. AIM OF THE STUDY The molecular mechanisms of the anti-inflammatory properties of M. accedens are not yet understood. Therefore, we examined those mechanisms using a methanol extract of M. accedens (Ma-ME) and determined the target molecule in macrophages. MATERIALS AND METHODS We evaluated the anti-inflammatory effects of Ma-ME in lipopolysaccharide (LPS)-stimulated RAW264.7 cells and in an HCl/EtOH-triggered gastritis model in mice. To investigate the anti-inflammatory activity, we performed a nitric oxide (NO) production assay and ELISA assay for prostaglandin E2 (PGE2). RT-PCR, luciferase gene reporter assays, western blotting analyses, and a cellular thermal shift assay (CETSA) were conducted to identify the mechanism and target molecule of Ma-ME. The phytochemical composition of Ma-ME was analyzed by HPLC and LC-MS/MS. RESULTS Ma-ME suppressed the production of NO and PGE2 and the mRNA expression of proinflammatory genes (iNOS, IL-1β, and COX-2) in LPS-stimulated RAW264.7 cells without cytotoxicity. Ma-ME inhibited NF-κB activation by suppressing signaling molecules such as IκBα, Akt, Src, and Syk. Moreover, the CETSA assay revealed that Ma-ME binds to Syk, the most upstream molecule in the NF-κB signal pathway. Oral administration of Ma-ME not only alleviated inflammatory lesions, but also reduced the gene expression of IL-1β and p-Syk in mice with HCl/EtOH-induced gastritis. HPLC and LC-MS/MS analyses confirmed that Ma-ME contains various anti-inflammatory flavonoids, including quercetin, daidzein, and nevadensin. CONCLUSIONS Ma-ME exhibited anti-inflammatory activities in vitro and in vivo by targeting Syk in the NF-κB signaling pathway. Therefore, we propose that Ma-ME could be used to treat inflammatory diseases such as gastritis.
Collapse
Affiliation(s)
- Jin Kyeong Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Eunju Choi
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Yo Han Hong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Haeyeop Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Young-Jin Jang
- College of Veterinary Medicine, Chonbuk National University, Iksan, 54596, Republic of Korea.
| | - Jong Sub Lee
- DanjoungBio Co., Ltd., Wonju, 26303, Republic of Korea.
| | - Eui Su Choung
- DanjoungBio Co., Ltd., Wonju, 26303, Republic of Korea.
| | | | - Yong Deog Hong
- AMOREPACIFIC R&D Center, Yongin, 17074, Republic of Korea.
| | - Sarah Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon, 22689, Republic of Korea.
| | - Byoung-Hee Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon, 22689, Republic of Korea.
| | - Tran The Bach
- Institute of Ecology and Biological Resources, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Cau Giay, Ha Noi, Viet Nam.
| | - Ji Hye Kim
- Department of Integrative Biotechnology, 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, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
12
|
Lee CY, Park SH, Lim HY, Jang SG, Park KJ, Kim DS, Kim JH, Cho JY. In vivo anti-inflammatory effects of Prasiola japonica ethanol extract. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104440] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
13
|
Euodia pasteuriana Methanol Extract Exerts Anti-Inflammatory Effects by Targeting TAK1 in the AP-1 Signaling Pathway. Molecules 2020; 25:molecules25235760. [PMID: 33297427 PMCID: PMC7730574 DOI: 10.3390/molecules25235760] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/29/2020] [Accepted: 12/05/2020] [Indexed: 12/14/2022] Open
Abstract
Euodia pasteuriana A. Chev. ex Guillaumin, also known as Melicope accedens (Blume) T.G. Hartley, is a herbal medicinal plant native to Vietnam. Although Euodia pasteuriana is used as a traditional medicine to treat a variety of inflammatory diseases, the pharmacological mechanisms related to this plant are unclear. This study aimed to investigate the anti-inflammatory effects of a methanol extract of Euodia pasteuriana leaves (Ep-ME) on the production of inflammatory mediators, the mRNA expression of proinflammatory genes, and inflammatory signaling activities in macrophage cell lines. The results showed that Ep-ME strongly suppressed the release of nitric oxide (NO) in RAW264.7 cells induced with lipopolysaccharide (LPS), pam3CysSerLys4 (Pam3CSK), and polyinosinic-polycytidylic acid (poly I:C) without cytotoxicity. A reverse transcription-polymerase chain reaction further confirmed that Ep-ME suppressed the expression of interleukin 6 (IL-6), matrix metalloproteinase-1 (MMP1), matrix metalloproteinase-2 (MMP2), matrix metalloproteinase-3 (MMP3), tumor necrosis factor-α (TNF-α), and matrix metalloproteinase-9 (MMP9) at the transcriptional level and reduced the luciferase activities of activator protein 1 (AP-1) reporter promoters. In addition, immunoblotting analyses of the whole lysate and nuclear fraction, as well as overexpression assays demonstrated that Ep-ME decreased the translocation of c-Jun and suppressed the activation of transforming growth factor beta-activated kinase 1 (TAK1) in the AP-1 signaling pathways. These results imply that Ep-ME could be developed as an anti-inflammatory agent that targets TAK1 in the AP-1 signaling pathway.
Collapse
|
14
|
STAT3 Differentially Regulates TLR4-Mediated Inflammatory Responses in Early or Late Phases. Int J Mol Sci 2020; 21:ijms21207675. [PMID: 33081347 PMCID: PMC7589049 DOI: 10.3390/ijms21207675] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/02/2020] [Accepted: 10/14/2020] [Indexed: 12/17/2022] Open
Abstract
Toll-like receptor 4 (TLR4) signaling is an important therapeutic target to manage lipopolysaccharide (LPS)-induced inflammation. The transcription factor signal transducer and activator of transcription 3 (STAT3) has been identified as an important regulator of various immune-related diseases and has generated interest as a therapeutic target. Here, we investigated the time-dependent roles of STAT3 in LPS-stimulated RAW264.7 macrophages. STAT3 inhibition induced expression of the pro-inflammatory genes iNOS and COX-2 at early time points. STAT3 depletion resulted in regulation of nuclear translocation of nuclear factor (NF)-κB subunits p50 and p65 and IκBα/Akt/PI3K signaling. Moreover, we found that one Src family kinase, Lyn kinase, was phosphorylated in STAT3 knockout macrophages. In addition to using pharmacological inhibition of NF-κB, we found out that STAT3KO activation of NF-κB subunit p50 and p65 and expression of iNOS was significantly inhibited; furthermore, Akt tyrosine kinase inhibitors also inhibited iNOS and COX-2 gene expression during early time points of LPS stimulation, demonstrating an NF-κB- Akt-dependent mechanism. On the other hand, iNOS expression was downregulated after prolonged treatment with LPS. Activation of NF-κB signaling was also suppressed, and consequently, nitric oxide (NO) production and cell invasion were repressed. Overall, our data indicate that STAT3 differentially regulates early- and late-phase TLR4-mediated inflammatory responses.
Collapse
|
15
|
Yang WS, Kim JH, Jeong D, Hong YH, Park SH, Yang Y, Jang YJ, Kim JH, Cho JY. 3-Deazaadenosine, an S-adenosylhomocysteine hydrolase inhibitor, attenuates lipopolysaccharide-induced inflammatory responses via inhibition of AP-1 and NF-κB signaling. Biochem Pharmacol 2020; 182:114264. [PMID: 33035507 DOI: 10.1016/j.bcp.2020.114264] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/01/2020] [Indexed: 02/08/2023]
Abstract
3-Deazadenosine (3-DA) is a general methylation inhibitor that depletes S-adenosylmethionine, a methyl donor, by blocking S-adenosylhomocysteine hydrolase (SAHH). In this study, we investigated the inhibitory activity and molecular mechanisms of 3-DA in inflammatory responses. 3-DA suppressed the secretion of inflammatory mediators such as nitric oxide (NO) and prostaglandin E2 (PGE2) in lipopolysaccharide-treated RAW264.7 cells and phorbol 12-myristate 13-acetate (PMA)-differentiated U937 cells. It also reduced mRNA expression of inducible nitric oxide synthase, cyclooxygenase-2, tumor necrosis factor-α, interleukin-1β (IL-1 β), and IL-6, indicating that 3-DA has anti-inflammatory properties in murine and human macrophages. Moreover, 3-DA strongly blocked AP-1 and NF-κB luciferase activity under PMA-, MyD88-, and TRIF-stimulated conditions and decreased the translocation of c-Jun, c-Fos, p65, and p50 into the nucleus. In addition, the p-ERK level in AP-1 signaling and the p-IκBα level in NF-kB signaling were diminished by 3-DA treatment. Interestingly, 3-DA did not alter the phosphorylation of MEK1/2, an ERK modulator, or IKKα/β, an IκBα regulator. Instead, 3-DA prevented MEK1/2 and IKKα/β from combining with ERK and IκBα, respectively, and directly suppressed MEK1/2 and IKKα/β kinase activity. These results indicate that MEK1/2 and IKKα/β are direct targets of 3-DA. In addition, suppression of SAHH by siRNA or treatment with adenosine dialdehyde, another SAHH inhibitor, showed inhibitory patterns against p-ERK and IκBα similar to those of 3-DA. Taken together, this study demonstrates that 3-DA inhibits AP-1 and NF-κB signaling by directly blocking MEK1/2 and IKKα/β or indirectly mediating SAHH, resulting in anti-inflammatory activity.
Collapse
Affiliation(s)
- Woo Seok Yang
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ji Hye Kim
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Deok Jeong
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yo Han Hong
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Sang Hee Park
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yoonyong Yang
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon 22689, Republic of Korea
| | - Young-Jin Jang
- 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.
| | - Jae Youl Cho
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
16
|
Sharma A, Lee HJ. Ginsenoside Compound K: Insights into Recent Studies on Pharmacokinetics and Health-Promoting Activities. Biomolecules 2020; 10:E1028. [PMID: 32664389 PMCID: PMC7407392 DOI: 10.3390/biom10071028] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/14/2022] Open
Abstract
Ginseng (Panax ginseng) is an herb popular for its medicinal and health properties. Compound K (CK) is a secondary ginsenoside biotransformed from major ginsenosides. Compound K is more bioavailable and soluble than its parent ginsenosides and hence of immense importance. The review summarizes health-promoting in vitro and in vivo studies of CK between 2015 and 2020, including hepatoprotective, anti-inflammatory, anti-atherosclerosis, anti-diabetic, anti-cancer, neuroprotective, anti-aging/skin protective, and others. Clinical trial data are minimal and are primarily based on CK-rich fermented ginseng. Besides, numerous preclinical and clinical studies indicating the pharmacokinetic behavior of CK, its parent compound (Rb1), and processed ginseng extracts are also summarized. With the limited evidence available from animal and clinical studies, it can be stated that CK is safe and well-tolerated. However, lower water solubility, membrane permeability, and efflux significantly diminish the efficacy of CK and restrict its clinical application. We found that the use of nanocarriers and cyclodextrin for CK delivery could overcome these limitations as well as improve the health benefits associated with them. However, these derivatives have not been clinically evaluated, thus requiring a safety assessment for human therapy application. Future studies should be aimed at investigating clinical evidence of CK.
Collapse
Affiliation(s)
- Anshul Sharma
- Department of Food and Nutrition, College of Bionanotechnology, Gachon University, Gyeonggi-do 13120, Korea;
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of Bionanotechnology, Gachon University, Gyeonggi-do 13120, Korea;
- Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do 13120, Korea
| |
Collapse
|
17
|
LOMIX, a Mixture of Flaxseed Linusorbs, Exerts Anti-Inflammatory Effects through Src and Syk in the NF-κB Pathway. Biomolecules 2020; 10:biom10060859. [PMID: 32512905 PMCID: PMC7356372 DOI: 10.3390/biom10060859] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/30/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
Although flax (Linum usitatissimum L.) has long been used as Ayurvedic medicine, its anti-inflammatory role is still unclear. Therefore, we aimed to investigate the anti-inflammatory role of a linusorb mixture (LOMIX) recovered from flaxseed oil. Effects of LOMIX on inflammation and its mechanism of action were examined using several in vitro assays (i.e., NO production, real-time PCR analysis, luciferase-reporter assay, Western blot analysis, and kinase assay) and in vivo analysis with animal inflammation models as well as acute toxicity test. Results: LOMIX inhibited NO production, cell shape change, and inflammatory gene expression in stimulated RAW264.7 cells through direct targeting of Src and Syk in the NF-κB pathway. In vivo study further showed that LOMIX alleviated symptoms of gastritis, colitis, and hepatitis in murine model systems. In accordance with in vitro results, the in vivo anti-inflammatory effects were mediated by inhibition of Src and Syk. LOMIX was neither cytotoxic nor did it cause acute toxicity in mice. In addition, it was found that LOB3, LOB2, and LOA2 are active components included in LOMIX, as assessed by NO assay. These in vitro and in vivo results suggest that LOMIX exerts an anti-inflammatory effect by inhibiting the inflammatory responses of macrophages and ameliorating symptoms of inflammatory diseases without acute toxicity and is a promising anti-inflammatory medication for inflammatory diseases.
Collapse
|
18
|
Molecular Signatures of JMJD10/MINA53 in Gastric Cancer. Cancers (Basel) 2020; 12:cancers12051141. [PMID: 32370161 PMCID: PMC7281541 DOI: 10.3390/cancers12051141] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/17/2020] [Accepted: 04/28/2020] [Indexed: 12/18/2022] Open
Abstract
The JMJD10 gene and its encoded protein MYC-induced nuclear antigen (MINA53) are associated with multiple cancers. Besides having both an oncogenic and tumor suppressor function, the intricate role of JMJD10 in cancer is complex as it depends on the cancer type. In particular, the functional role of JMJD10/MINA53 in gastric cancer has been poorly understood. In this study, we have unraveled the molecular signatures and functional roles of JMJD10/MINA53 in gastric cancer by multiple approaches, i.e., multi-omics bioinformatics study, analysis of human gastric cancer tissues, and studies in vitro using knockdown or overexpression strategies in gastric cancer cell lines. The results indicated that the JMJD10 gene and MINA53 protein are commonly overexpressed in cancer patients. JMJD10/MINA53 is involved in the regulation of proliferation and survival of gastric cancer by controlling cell cycle gene expression. These processes are highly associated with MINA53 enzymatic activity in the regulation of H3K9me3 methylation status and controlling activation of AP-1 signaling pathways. This highlights the oncogenic role of JMJD10/MINA53 in gastric cancer and opens the opportunity to develop therapeutic targeting of JMJD10/MINA53 in gastric cancer.
Collapse
|
19
|
Kim E, Jang J, Park JG, Kim KH, Yoon K, Yoo BC, Cho JY. Protein Arginine Methyltransferase 1 (PRMT1) Selective Inhibitor, TC-E 5003, Has Anti-Inflammatory Properties in TLR4 Signaling. Int J Mol Sci 2020; 21:ijms21093058. [PMID: 32357521 PMCID: PMC7246892 DOI: 10.3390/ijms21093058] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/19/2020] [Accepted: 04/25/2020] [Indexed: 12/14/2022] Open
Abstract
Protein arginine methyltransferase 1 (PRMT1) is the most predominant PRMT and is type I, meaning it generates monomethylarginine and asymmetric dimethylarginine. PRMT1 has functions in oxidative stress, inflammation and cancers, and modulates diverse diseases; consequently, numerous trials to develop PRMT1 inhibitors have been attempted. One selective PRMT1 inhibitor is N,N′-(Sulfonyldi-4,1-phenylene)bis(2-chloroacetamide), also named TC-E 5003 (TC-E). In this study, we investigated whether TC-E regulated inflammatory responses. Nitric oxide (NO) production was evaluated by the Griess assay and the inflammatory gene expression was determined by conducting RT-PCR. Western blot analyzing was carried out for inflammatory signaling exploration. TC-E dramatically reduced lipopolysaccharide (LPS)-induced NO production and the expression of inflammatory genes (inducible NO synthase (iNOS), cyclooxygenase (COX)-2, tumor necrosis factor (TNF)-α and interleukin (IL)-6) as determined using RT-PCR. TC-E downregulated the nuclear translocation of the nuclear factor (NF)-κB subunits p65 and p50 and the activator protein (AP)-1 transcriptional factor c-Jun. Additionally, TC-E directly regulated c-Jun gene expression following LPS treatment. In NF-κB signaling, the activation of IκBα and Src was attenuated by TC-E. Taken together, these data show that TC-E modulates the lipopolysaccharide (LPS)-induced AP-1 and NF-κB signaling pathways and could possibly be further developed as an anti-inflammatory compound.
Collapse
Affiliation(s)
- Eunji Kim
- Department of Integrative Biotechnology, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (E.K.); (J.J.); (K.Y.)
| | - Jiwon Jang
- Department of Integrative Biotechnology, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (E.K.); (J.J.); (K.Y.)
| | - Jae Gwang Park
- Division of Translational Science, Research Institute, National Cancer Center, Goyang 10408, Korea;
| | - Kyung-Hee Kim
- Proteomic Analysis Team, Research Institute, National Cancer Center, Goyang 10408, Korea;
| | - Keejung Yoon
- Department of Integrative Biotechnology, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (E.K.); (J.J.); (K.Y.)
| | - Byong Chul Yoo
- Division of Translational Science, Research Institute, National Cancer Center, Goyang 10408, Korea;
- Correspondence: (B.C.Y.); (J.Y.C.); Tel.: +82-31-920-2342 (B.C.Y.); +82-31-290-7876 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (E.K.); (J.J.); (K.Y.)
- Correspondence: (B.C.Y.); (J.Y.C.); Tel.: +82-31-920-2342 (B.C.Y.); +82-31-290-7876 (J.Y.C.)
| |
Collapse
|
20
|
Anti-Inflammatory Functions of Alverine via Targeting Src in the NF-κB Pathway. Biomolecules 2020; 10:biom10040611. [PMID: 32326535 PMCID: PMC7225962 DOI: 10.3390/biom10040611] [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: 03/04/2020] [Revised: 04/03/2020] [Accepted: 04/11/2020] [Indexed: 12/25/2022] Open
Abstract
Alverine, a smooth muscle relaxant, is used to relieve cramps or spasms of the stomach and intestine. Although the effects of alverine on spontaneous and induced contractile activity are well known, its anti-inflammatory activity has not been fully evaluated. In this study, we investigated the anti-inflammatory effects of alverine in vitro and in vivo. The production of nitric oxide (NO) in RAW264.7 cells activated by lipopolysaccharide (LPS) or polyinosinic:polycytidylic acid (poly (I:C)) was reduced by alverine. The mRNA expression of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and tumor necrosis factor-α (TNF-α) was also dose-dependently inhibited by treatment with alverine. In reporter gene assays, alverine clearly decreased luciferase activity, mediated by the transcription factor nuclear factor κB (NF-κB) in TIR-domain-containing adapter-inducing interferon-β (TRIF)- or MyD88-overexpressing HEK293 cells. Additionally, phosphorylation of NF-κB subunits and upstream signaling molecules, including p65, p50, AKT, IκBα, and Src was downregulated by 200 μM of alverine in LPS-treated RAW264.7 cells. Using immunoblotting and cellular thermal shift assays (CETSAs), Src was identified as the target of alverine in its anti-inflammatory response. In addition, HCl/EtOH-stimulated gastric ulcers in mice were ameliorated by alverine at doses of 100 and 200 mg/kg. In conclusion, alverine reduced inflammatory responses by targeting Src in the NF-κB pathway, and these findings provide new insights into the development of anti-inflammatory drugs.
Collapse
|
21
|
Hong YH, Kim JH, Cho JY. Ranunculus bulumei Methanol Extract Exerts Anti-Inflammatory Activity by Targeting Src/Syk in NF-κB Signaling. Biomolecules 2020; 10:biom10040546. [PMID: 32260181 PMCID: PMC7226355 DOI: 10.3390/biom10040546] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 02/07/2023] Open
Abstract
(1) Background: Ranunculus bulumei is a flowering plant that belongs to the Ranunculus species. Several Ranunculus species, such as R. aquatilis and R. muricatus, have traditionally been used to treat fever and rheumatism throughout Asia, suggesting that plants belonging to the Ranunculus species may have anti-inflammatory effects. To our knowledge, the pharmacological activity of R. bulumei has not been reported. Therefore, in this study, we aim to assess the anti-inflammatory activity of a methanol extract that was derived from R. bulumei (Rb-ME) in macrophage-mediated inflammatory responses and to identify the molecular mechanism that underlies any anti-inflammatory action. (2) Methods: The anti-inflammatory efficacy of Rb-ME was evaluated while using in vitro and in vivo experiments. The RAW264.7 cells and peritoneal macrophages were stimulated by lipopolysaccharide (LPS). In addition, LPS-induced peritonitis and HCl/EtOH-triggered gastritis models were produced. A nitric oxide (NO) assay, real-time PCR, luciferase reporter gene assay, western blot analysis, plasmid overexpression strategy, and in vitro kinase assay were used to determine the molecular mechanisms and target molecules of Rb-ME. The phytochemical active ingredients of Rb-ME were also identified by high performance liquid chromatograph (HPLC). (3) Results: Rb-ME reduced the production of NO and mRNA expression of iNOS, COX-2, IL-1β, and IL-6 without cytotoxicity. The protein secretion of TNF-α and IL-6 was also decreased by Rb-ME. HPLC analysis indicates that quercetin, luteolin, and kaempferol are the main active ingredients in the anti-inflammatory efficacy of Rb-ME. Rb-ME also blocked MyD88-induced NF-κB promoter activity and nuclear translocation of NF-κB subunits (p65 and p50). Moreover, Rb-ME reduced the phosphorylation of IκBα, Akt, p85, Src, and Syk, which are NF-κB upstream signaling molecules in LPS-activated RAW264.7 cells. According to the in vitro kinase assay, Rb-ME directly inhibits Syk kinase activity. The oral administration of Rb-ME alleviated inflammatory responses and the levels of p-IκBα in mice with LPS-induced peritonitis and HCl/EtOH-induced gastritis. (4) Conclusions Rb-ME has anti-inflammatory capacity by suppressing NF-κB signaling and it has been found to target Src and Syk in the NF-κB pathway. Based on this efficacy, Rb-ME could be developed as an anti-inflammatory herbal medicine.
Collapse
|
22
|
Yu T, Wang Z, Jie W, Fu X, Li B, Xu H, Liu Y, Li M, Kim E, Yang Y, Cho JY. The kinase inhibitor BX795 suppresses the inflammatory response via multiple kinases. Biochem Pharmacol 2020; 174:113797. [DOI: 10.1016/j.bcp.2020.113797] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023]
|
23
|
Aziz N, Kang YG, Kim YJ, Park WS, Jeong D, Lee J, Kim D, Cho JY. Regulation of 8-Hydroxydaidzein in IRF3-Mediated Gene Expression in LPS-Stimulated Murine Macrophages. Biomolecules 2020; 10:biom10020238. [PMID: 32033247 PMCID: PMC7072285 DOI: 10.3390/biom10020238] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/17/2020] [Accepted: 01/26/2020] [Indexed: 12/20/2022] Open
Abstract
Cytokines and chemokines are transcriptionally regulated by inflammatory transcription factors such as nuclear factor-κB (NF-κB), activator protein-1 (AP-1), and interferon regulatory factor (IRF)-3. A daidzein derivative compound, 8-hydroxydaidzein (8-HD), isolated from soy products, has recently gained attention due to various pharmacological benefits, including anti-inflammatory activities. However, regulation of the inflammatory signaling mechanism for 8-HD is still poorly understood, particularly with respect to the IRF-3 signaling pathway. In this study, we explored the molecular mechanism of 8-HD in regulating inflammatory processes, with a focus on the IRF-3 signaling pathway using a lipopolysaccharide (LPS) and polyinosinic:polycytidylic acid [Poly (I:C)] stimulated murine macrophage cell line (RAW264.7). The 8-HD downregulated the mRNA expression level of IRF-3-dependent genes by inhibiting phosphorylation of the IRF-3 transcription factor. The inhibitory mechanism of 8-HD in the IRF-3 signaling pathway was shown to inhibit the kinase activity of IKKε to phosphorylate IRF-3. This compound can also interfere with the TRIF-mediated complex formation composed of TRAF3, TANK, and IKKε leading to downregulation of AKT phosphorylation and reduction of IRF-3 activation, resulted in inhibition of IRF-3-dependent expression of genes including IFN-β, C-X-C motif chemokine 10 (CXCL10), and interferon-induced protein with tetratricopeptide repeats 1 (IFIT1). Therefore, these results strongly suggest that 8-HD can act as a promising compound with the regulatory function of IRF-3-mediated inflammatory responses.
Collapse
Affiliation(s)
- Nur Aziz
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (N.A.); (D.J.)
| | - Young-Gyu Kang
- Basic Research & Innovation Division, R&D Center, AmorePacific Corporation, Yongin 17074, Korea; (Y.-G.K.); (Y.-J.K.); (W.-S.P.)
| | - Yong-Jin Kim
- Basic Research & Innovation Division, R&D Center, AmorePacific Corporation, Yongin 17074, Korea; (Y.-G.K.); (Y.-J.K.); (W.-S.P.)
| | - Won-Seok Park
- Basic Research & Innovation Division, R&D Center, AmorePacific Corporation, Yongin 17074, Korea; (Y.-G.K.); (Y.-J.K.); (W.-S.P.)
| | - Deok Jeong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (N.A.); (D.J.)
| | - Jongsung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (N.A.); (D.J.)
- Correspondence: (J.L.); (D.K.); (J.Y.C.); Tel.: +82-31-290-7861 (J.L.); +82-31-280-5869 (D.K.); +82-31-290-7868 (J.Y.C.)
| | - Donghyun Kim
- Basic Research & Innovation Division, R&D Center, AmorePacific Corporation, Yongin 17074, Korea; (Y.-G.K.); (Y.-J.K.); (W.-S.P.)
- Correspondence: (J.L.); (D.K.); (J.Y.C.); Tel.: +82-31-290-7861 (J.L.); +82-31-280-5869 (D.K.); +82-31-290-7868 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (N.A.); (D.J.)
- Correspondence: (J.L.); (D.K.); (J.Y.C.); Tel.: +82-31-290-7861 (J.L.); +82-31-280-5869 (D.K.); +82-31-290-7868 (J.Y.C.)
| |
Collapse
|
24
|
Trichosanthes tricuspidata Lour. Methanol Extract Exhibits Anti-Inflammatory Activity by Targeting Syk, Src, and IRAK1 Kinase Activity. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2019:6879346. [PMID: 31929819 PMCID: PMC6942823 DOI: 10.1155/2019/6879346] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 11/27/2019] [Indexed: 12/19/2022]
Abstract
Trichosanthes tricuspidata Lour., also known as T. palmata Roxb, T. bracteata Lam., T. puber Blume, and Modecca bracteata, is a vine belonging to the Cucurbitaceae family (English name: redball snake gourd). Distributed in China, South and East Asia, and tropical Australia, it has been traditionally used as a medicinal plant for its antifever, laxative, anthelmintic properties and for migraine treatment. In this paper, we examined the effects of Trichosanthes tricuspidata Lour. ethanol extract (Tt-ME) in vitro and in vivo. To confirm the effects of Tt-ME on inflammatory responses, we conducted experimental analyses including level of nitric oxide (NO) production, RT-PCR, and immunoblotting and using a HCl/EtOH-induced gastritis animal model. Tt-ME attenuated the release of NO and decreased mRNA levels of inducible NO synthase (iNOS), TNF-α, and IL-6 in lipopolysaccharide- (LPS-) induced macrophages in a concentration-dependent manner. Tt-ME time-dependently suppressed nuclear translocation of nuclear factor kappa B (NF-κB) subunits p50 and p65, activator protein (AP-1) subunits c-Fos and c-Jun, and STAT3 transcriptional activity by inhibiting nuclear translocation of p50, p65, c-Fos, c-Jun, and STAT3. Tt-ME significantly downregulated NF-κB, MAPK, and JAK2 signaling by targeting Syk, Src, and IRAK1 protein kinases. Furthermore, matrix metalloproteinase-9 (MMP-9) expression and cell migration were observed to be downregulated by Tt-ME in LPS-activated macrophages. In vivo studies on Tt-ME also produced similar trends in Hcl/EtOH-induced gastritis mouse models by inhibiting proinflammatory cytokines and the inflammatory signaling pathway. Our results strongly suggest that Tt-ME exerted anti-inflammatory activity in LPS-stimulated macrophages and mouse models of acute inflammatory disease.
Collapse
|
25
|
Choi E, Kang YG, Hwang SH, Kim JK, Hong YD, Park WS, Kim D, Kim E, Cho JY. In Vitro Effects of Dehydrotrametenolic Acid on Skin Barrier Function. Molecules 2019; 24:molecules24244583. [PMID: 31847353 PMCID: PMC6943483 DOI: 10.3390/molecules24244583] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/07/2019] [Accepted: 12/12/2019] [Indexed: 11/16/2022] Open
Abstract
Dehydrotrametenolic acid (DTA) is a lanostane-type triterpene acid isolated from Poria cocos Wolf (Polyporaceae). Several studies have reported the anti-inflammatory and antidiabetic effects of DTA; however, its effects on the skin are poorly understood. In this study, we investigated the effects of DTA on skin barrier function in vitro and its regulatory mechanism in human keratinocyte cell line HaCaT cells. DTA increased the microRNA (mRNA) expression of natural moisturizing factor-related genes, such as HAS-2, HAS-3, and AQP3 in HaCaT cells. DTA also upregulated the mRNA expression of various keratinocyte differentiation markers, including TGM-1, involucrin, and caspase-14. Moreover, the protein expression of HAS-2, HAS-3, and TGM-2 were significantly increased by DTA. To examine the regulatory mechanisms of DTA, Western blotting, luciferase-reporter assays, and RT-PCR were conducted. The phosphorylation of mitogen-activated protein kinases (MAPKs) and IκBα were increased in DTA-treated HaCaT cells. In addition, AP-1 and NF-κB transcriptional factors were dose-dependently activated by DTA. Taken together, our in vitro mechanism studies indicate that the regulatory effects of DTA on skin hydration and keratinocyte differentiation are mediated by the MAPK/AP-1 and IκBα/NF-κB pathways. In addition, DTA could be a promising ingredient in cosmetics for moisturizing and increased skin barrier function.
Collapse
Affiliation(s)
- Eunju Choi
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (E.C.); (S.-H.H.); (J.K.K.)
| | - Young-Gyu Kang
- Basic Research & Innovation Division, R&D Center, AmorePacific Corporation, Yongin 17074, Korea; (Y.-G.K.); (Y.D.H.); (W.-S.P.); (D.K.)
| | - So-Hyeon Hwang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (E.C.); (S.-H.H.); (J.K.K.)
| | - Jin Kyeong Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (E.C.); (S.-H.H.); (J.K.K.)
| | - Yong Deog Hong
- Basic Research & Innovation Division, R&D Center, AmorePacific Corporation, Yongin 17074, Korea; (Y.-G.K.); (Y.D.H.); (W.-S.P.); (D.K.)
| | - Won-Seok Park
- Basic Research & Innovation Division, R&D Center, AmorePacific Corporation, Yongin 17074, Korea; (Y.-G.K.); (Y.D.H.); (W.-S.P.); (D.K.)
| | - Donghyun Kim
- Basic Research & Innovation Division, R&D Center, AmorePacific Corporation, Yongin 17074, Korea; (Y.-G.K.); (Y.D.H.); (W.-S.P.); (D.K.)
| | - Eunji Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (E.C.); (S.-H.H.); (J.K.K.)
- Correspondence: (E.K.); or (J.Y.C.); Tel.: +82-31-290-7860 (E.K.); +82-31-290-7868 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (E.C.); (S.-H.H.); (J.K.K.)
- Correspondence: (E.K.); or (J.Y.C.); Tel.: +82-31-290-7860 (E.K.); +82-31-290-7868 (J.Y.C.)
| |
Collapse
|
26
|
Hwang SH, Lorz LR, Yi DK, Noh JK, Yi YS, Cho JY. Viburnum pichinchense methanol extract exerts anti-inflammatory effects via targeting the NF-κB and caspase-11 non-canonical inflammasome pathways in macrophages. JOURNAL OF ETHNOPHARMACOLOGY 2019; 245:112161. [PMID: 31419499 DOI: 10.1016/j.jep.2019.112161] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/11/2019] [Accepted: 08/12/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Viburnum pichinchense Benth. Mainly found in Ecuador and Colombia has been ethnopharmacologically utilized as a remedy for various female disorders with kidney inflammation and uterine relaxant. AIM OF THE STUDY The pharmacological activity of Viburnum pichinchense has never been studied, therefore, this study explored anti-inflammatory activity of Viburnum pichinchense methanol extract (Vp-ME). MATERIALS AND METHODS Anti-inflammatory activities of Vp-ME were evaluated in lipopolysaccharide (LPS)-stimulated RAW264.7 cells and HCl/EtOH-induced gastritis mice by MTT assay, nitric oxide (NO) production assay, semi-quantitative reverse-transcriptase-polymerase chain reaction (RT-PCR), luciferase reporter assay, Western blotting, and enzyme-linked immunosorbent assays (ELISA). Anti-inflammatory compounds in Vp-ME were identified by high performance liquid chromatography (HPLC). RESULTS Vp-ME inhibited NO production in RAW264.7 cells stimulated with pam3CSK4, poly I:C or LPS and in LPS-stimulated peritoneal macrophages without cytotoxicity and downregulated mRNA expression of inflammatory enzymes, inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2) and pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-6. The anti-inflammatory activity was accomplished by inhibiting nuclear factor-kappa B (NF-κB) transcriptional activation, upstream signaling molecules in the NF-κB pathway, and caspase-11 non-canonical inflammasome in RAW264.7 cells. Moreover, Vp-ME exhibited in vivo anti-inflammatory activity by ameliorating gastritis symptoms, inhibiting iNOS and IL-6 mRNA expression and IκBα activation in mice. HPLC analysis identified resveratrol, quercetin, luteolin, and kaempferol as the anti-inflammatory components in Vp-ME. CONCLUSION This study demonstrated Vp-ME has the anti-inflammatory activity via targeting NF-κB and caspase-11 non-canonical inflammasome pathways in macrophage-mediated inflammatory responses, suggesting Vp-ME could be developed as anti-inflammatory ethnopharmacological remedies to prevent and treat inflammatory diseases.
Collapse
Affiliation(s)
- So-Hyeon Hwang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, South Korea.
| | - Laura Rojas Lorz
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, South Korea.
| | - Dong-Keun Yi
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, South Korea.
| | - Jin Kyoung Noh
- Instituto de BioEconomia, El Batan, Quito, 170135, Ecuador.
| | - Young-Su Yi
- Department of Pharmaceutical and Biomedical Engineering, Cheongju University, Cheongju, 28503, South Korea.
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, South Korea.
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
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: 40] [Impact Index Per Article: 6.7] [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
|
30
|
Choi E, Kim MY, Cho JY. Anti-inflammatory activities of Canarium subulatum Guillaumin methanol extract operate by targeting Src and Syk in the NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2019; 238:111848. [PMID: 30951845 DOI: 10.1016/j.jep.2019.111848] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/24/2019] [Accepted: 03/31/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Canarium subulatum Guillaumin is an herbal medicinal plant native to Southeast Asia. Ethnopharmacological evidence suggests that plants of the genus Canarium cure a variety of inflammatory diseases. AIM OF THE STUDY The pharmacological mechanisms of C. subulatum Guillaumin remain poorly understood. In this study, we investigate inflammatory mechanisms and target molecules using C. subulatum Guillaumin methanol extract (Cs-ME) in inflammatory reactions managed by macrophages. MATERIALS AND METHODS To identify the anti-inflammatory activities of Cs-ME, lipopolysaccharide (LPS)-stimulated macrophages and a murine HCl/EtOH-induced gastritis model were chosen. The luciferase reporter gene assay, Western blot analysis, overexpression strategy, and the cellular thermal shift assay (CETSA) were employed to investigate the molecular mechanisms and target enzymes of Cs-ME. The active ingredients of this extract were also determined by HPLC. RESULTS Released levels of nitric oxide (NO) and mRNA expression levels of iNOS and IL-6 were downregulated by Cs-ME without exhibiting cytotoxicity. This extract inhibited MyD88-induced promoter activity and the nuclear translocation of nuclear factor (NF)-κB. Moreover, we found that Cs-ME reduced the phosphorylation of NF-κB upstream signaling molecules including IκBα, IKKα/β, Src, and Syk in LPS-stimulated macrophage-like RAW264.7 cells. The results of Western blot and CETSA confirmed that Src and Syk are anti-inflammatory targets of Cs-ME. In addition, orally injected Cs-ME alleviated HCl/EtOH-induced gastric ulcers in mice. HPLC analysis indicated that quercetin, luteolin, and kaempferol are major active components of this extract with anti-inflammatory activity. CONCLUSIONS Cs-ME exhibits anti-inflammatory effects in vitro and in vivo by targeting Src and Syk in the NF-κB signaling pathway. Consequently, Cs-ME could be developed as an anti-inflammatory herbal medicine.
Collapse
Affiliation(s)
- Eunju Choi
- Department of Integrative Biotechnology, 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 Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
31
|
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
|
32
|
Kim HG, Yang WS, Hong YH, Kweon DH, Lee J, Kim S, Cho JY. Anti-inflammatory functions of the CDC25 phosphatase inhibitor BN82002 via targeting AKT2. Biochem Pharmacol 2019; 164:216-227. [PMID: 30980807 DOI: 10.1016/j.bcp.2019.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 04/08/2019] [Indexed: 11/28/2022]
Abstract
This study presents BN82002 as an anti-inflammatory drug candidate. It was found that BN82002 inhibited the production of nitric oxide (NO) and prostaglandin E2 (PGE2) in RAW 264.7 cells and peritoneal macrophages that were activated by toll-like receptor (TLR) 4 ligand, lipopolysaccharide (LPS). BN82002 dose-dependently down-regulated mRNA levels of nitric oxide synthase, tumor necrosis factor-α, and cyclooxygenase-2. The nuclear translocation of nuclear factor (NF)-κB (p65 and p50) was also blocked by BN82002 in RAW265.7 cells stimulated by LPS. According to reporter gene assay performed with NF-κB construct, BN82002 clearly reduced increased level of luciferase activity mediated by transcription factor NF-κB in LPS-treated RAW264.7 cells and in MyD88- and AKT2-overexpressing HEK293 cells. However, BN82002 did not inhibit NF-κB activity in AKT1- or IKKβ-overexpressing HEK293 cells. NF-κB upstream signaling events specifically targeted AKT2 but had no effect on AKT1. The specific target of BN82002 was Tyr-178 in AKT2. BN82002 bound to Tyr-178 and interrupted the kinase activity of AKT2, according to a cellular thermal shift assay analysis of the interaction of BN82002 with AKT2 and an AKT2 mutant (Tyr-178 mutated to Ala; AKT2 Y178A). These results suggest that BN82002 could reduce inflammatory pathway by controlling NF-κB pathway and specifically targeting AKT2.
Collapse
Affiliation(s)
- Han Gyung Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Woo Seok Yang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yo Han Hong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dae-Hyuk Kweon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jongsung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Sunggyu Kim
- Research and Business Foundation, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
33
|
Dehydroabietic Acid Suppresses Inflammatory Response Via Suppression of Src-, Syk-, and TAK1-Mediated Pathways. Int J Mol Sci 2019; 20:ijms20071593. [PMID: 30934981 PMCID: PMC6480320 DOI: 10.3390/ijms20071593] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/27/2019] [Accepted: 03/27/2019] [Indexed: 12/20/2022] Open
Abstract
Dehydroabietic acid (DAA) is a naturally occurring diterpene resin acid derived from coniferous plants such as Pinus and Picea. Various bioactive effects of DAA have been studied including antibacterial, antifungal, and anticancer activities. However, the anti-inflammatory mechanism of DAA remains unclear. We evaluated the anti-inflammatory effect of DAA in macrophage cell lines. Dehydroabietic acid clearly reduced nitric oxide (NO) production and inflammatory gene expression decreased according to RT-PCR results. Dehydroabietic acid displayed anti-inflammatory activity at the transcriptional level in results from NF-κB- or AP-1-mediated luciferase assays. To identify the DAA target protein, we investigated NF-κB and AP-1 pathways by Western blotting analysis. Dehydroabietic acid suppressed the activity of proto-oncogene tyrosine protein kinase (Src) and spleen tyrosine kinase (Syk) in the NF-κB cascade and transforming growth factor beta-activated kinase 1 (TAK1) in the AP-1 cascade. Using overexpression strategies, we confirmed that DAA targeted these kinases. Our findings demonstrate the anti-inflammatory effects and molecular mechanism of DAA. This suggests that DAA has potential as a drug or supplement to ameliorate inflammation.
Collapse
|
34
|
A new triterpene diglycoside from the roots of Bupleurum chinense DC. and its inhibitory effect on adipogensis in 3T3-L1 cells. Med Chem Res 2018. [DOI: 10.1007/s00044-018-2279-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|