1
|
Hwang S, Park MS, Koo AJ, Yoo E, Song SH, Kim HK, Park MH, Kang JS. Compound K Promotes Megakaryocytic Differentiation by NLRP3 Inflammasome Activation. Biomolecules 2024; 14:1257. [PMID: 39456190 PMCID: PMC11506438 DOI: 10.3390/biom14101257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
Platelets are essential blood components that maintain hemostasis, prevent excessive bleeding, and facilitate wound healing. Reduced platelet counts are implicated in various diseases, including leukemia, hepatitis, cancer, and Alzheimer's disease. Enhancing megakaryocytic differentiation is a promising strategy to increase platelet production. Compound K (CK), a major bioactive metabolite of ginsenosides from Panax ginseng, has demonstrated anti-cancer and neuroprotective properties. In this study, we investigated the effects of CK on megakaryocytic differentiation and apoptosis in chronic myeloid leukemia (CML) cell lines K562 and Meg-01. CK treatment significantly upregulated the mRNA expression of key megakaryocytic differentiation markers, including CD61, CD41, and CD42a, and promoted the formation of large, multinucleated cells in K562 cells. Additionally, flow cytometry analysis revealed that CK at 5 µM induced apoptosis, a critical process in thrombocytopoiesis, in both K562 and Meg-01 cells. RT2 Profiler PCR array analysis further identified a marked increase in the expression of genes associated with the activation of the NLRP3 inflammasome in CK-treated K562 and Meg-01 cells. This study is the first to demonstrate that CK promotes megakaryocytic differentiation and apoptosis through the activation of the ERK/EGR1 and NLRP3 inflammasome pathways. These findings suggest that CK may enhance platelet production, indicating its potential as a therapeutic candidate for platelet-related disorders and other associated diseases.
Collapse
MESH Headings
- Humans
- Megakaryocytes/drug effects
- Megakaryocytes/metabolism
- Megakaryocytes/cytology
- Cell Differentiation/drug effects
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Inflammasomes/metabolism
- Inflammasomes/drug effects
- K562 Cells
- Apoptosis/drug effects
- Ginsenosides/pharmacology
- Platelet Membrane Glycoprotein IIb/metabolism
- Integrin beta3/metabolism
- Integrin beta3/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
Collapse
Affiliation(s)
- Seonhwa Hwang
- College of Pharmacy, Kyungsung University, 309 Suyeong-ro, Busan 48434, Republic of Korea; (S.H.); (M.-S.P.); (S.-H.S.); (H.-K.K.)
- Brain Busan 21 Plus Research Project Group, Kyungsung University, Busan 48434, Republic of Korea
| | - Min-Seo Park
- College of Pharmacy, Kyungsung University, 309 Suyeong-ro, Busan 48434, Republic of Korea; (S.H.); (M.-S.P.); (S.-H.S.); (H.-K.K.)
- Brain Busan 21 Plus Research Project Group, Kyungsung University, Busan 48434, Republic of Korea
| | - Anthony Junhoe Koo
- College of Engineering, North Carolina A&T State University, Greensboro, NC 27411, USA;
| | - Eunsoo Yoo
- Chemical, Biological, and Bioengineering Department, North Carolina A&T State University, Greensboro, NC 27411, USA;
| | - Seh-Hyon Song
- College of Pharmacy, Kyungsung University, 309 Suyeong-ro, Busan 48434, Republic of Korea; (S.H.); (M.-S.P.); (S.-H.S.); (H.-K.K.)
| | - Hye-Kyung Kim
- College of Pharmacy, Kyungsung University, 309 Suyeong-ro, Busan 48434, Republic of Korea; (S.H.); (M.-S.P.); (S.-H.S.); (H.-K.K.)
- Brain Busan 21 Plus Research Project Group, Kyungsung University, Busan 48434, Republic of Korea
| | - Min-Hi Park
- College of Pharmacy, Kyungsung University, 309 Suyeong-ro, Busan 48434, Republic of Korea; (S.H.); (M.-S.P.); (S.-H.S.); (H.-K.K.)
- Brain Busan 21 Plus Research Project Group, Kyungsung University, Busan 48434, Republic of Korea
| | - Jae-Seon Kang
- College of Pharmacy, Kyungsung University, 309 Suyeong-ro, Busan 48434, Republic of Korea; (S.H.); (M.-S.P.); (S.-H.S.); (H.-K.K.)
- Brain Busan 21 Plus Research Project Group, Kyungsung University, Busan 48434, Republic of Korea
| |
Collapse
|
2
|
Tan S, Zou Z, Luan X, Chen C, Li S, Zhang Z, Quan M, Li X, Zhu W, Yang G. Synthesis, Anti-Inflammatory Activities, and Molecular Docking Study of Novel Pyxinol Derivatives as Inhibitors of NF-κB Activation. Molecules 2024; 29:1711. [PMID: 38675532 PMCID: PMC11052049 DOI: 10.3390/molecules29081711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/06/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Pyxinol, an active metabolite of ginsenosides in human hepatocytes, exhibits various pharmacological activities. Here, a series of C-3 modified pyxinol derivatives was designed and virtually screened by molecular docking with the key inflammation-related proteins of the nuclear factor kappa B (NF-κB) pathway. Some of the novel derivatives were synthesized to assess their effects in inhibiting the production of nitric oxide (NO) and mitochondrial reactive oxygen species (MtROS) in lipopolysaccharide-triggered RAW264.7 cells. Derivative 2c exhibited the highest NO and MtROS inhibitory activities with low cytotoxicity. Furthermore, 2c decreased the protein levels of interleukin 1β, tumor necrosis factor α, inducible nitric oxide synthase, and cyclooxygenase 2 and suppressed the activation of NF-κB signaling. Cellular thermal shift assays indicated that 2c could directly bind with p65 and p50 in situ. Molecular docking revealed that 2c's binding to the p65-p50 heterodimer and p50 homodimer was close to their DNA binding sites. In summary, pyxinol derivatives possess potential for development as NF-κB inhibitors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Wei Zhu
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China; (S.T.); (Z.Z.); (X.L.); (C.C.); (S.L.); (Z.Z.); (M.Q.); (X.L.)
| | - Gangqiang Yang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China; (S.T.); (Z.Z.); (X.L.); (C.C.); (S.L.); (Z.Z.); (M.Q.); (X.L.)
| |
Collapse
|
3
|
Lv N, Wang L, Zeng M, Wang Y, Yu B, Zeng W, Jiang X, Suo Y. Saponins as therapeutic candidates for atherosclerosis. Phytother Res 2024; 38:1651-1680. [PMID: 38299680 DOI: 10.1002/ptr.8128] [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: 09/17/2023] [Revised: 12/25/2023] [Accepted: 01/06/2024] [Indexed: 02/02/2024]
Abstract
Drug development for atherosclerosis, the underlying pathological state of ischemic cardiovascular diseases, has posed a longstanding challenge. Saponins, classified as steroid or triterpenoid glycosides, have shown promising therapeutic potential in the treatment of atherosclerosis. Through an exhaustive examination of scientific literature spanning from May 2013 to May 2023, we identified 82 references evaluating 37 types of saponins in terms of their prospective impacts on atherosclerosis. These studies suggest that saponins have the potential to ameliorate atherosclerosis by regulating lipid metabolism, inhibiting inflammation, suppressing apoptosis, reducing oxidative stress, and modulating smooth muscle cell proliferation and migration, as well as regulating gut microbiota, autophagy, endothelial senescence, and angiogenesis. Notably, ginsenosides exhibit significant potential and manifest essential pharmacological attributes, including lipid-lowering, anti-inflammatory, anti-apoptotic, and anti-oxidative stress effects. This review provides a comprehensive examination of the pharmacological attributes of saponins in atherosclerosis, with particular emphasis on their role in the regulation of lipid metabolism regulation and anti-inflammatory effects. Thus, saponins may warrant further investigation as a potential therapy for atherosclerosis. However, due to various reasons such as low oral bioavailability, the clinical application of saponins in the treatment of atherosclerosis still needs further exploration.
Collapse
Affiliation(s)
- Nuan Lv
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Luming Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Miao Zeng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yijing Wang
- School of Nursing, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bin Yu
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenyun Zeng
- Oncology Department, Ganzhou people's hospital, Ganzhou, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanrong Suo
- Traditional Chinese Medicine Department, Ganzhou people's hospital, Ganzhou, China
| |
Collapse
|
4
|
Monmai C, Kim JS, Sim HB, Yun DW, Oh SD, Rha ES, Kim JJ, Baek SH. Protopanaxadiol-Enriched Rice Exerted Antiadipogenic Activity during 3T3-L1 Differentiation and Anti-Inflammatory Activity in 3T3-L1 Adipocytes. Pharmaceutics 2023; 15:2123. [PMID: 37631337 PMCID: PMC10458103 DOI: 10.3390/pharmaceutics15082123] [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: 07/18/2023] [Revised: 07/28/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Ginseng is a traditional medicine with health benefits for humans. Protopanaxadiol (PPD) is an important bioactive compound found in ginseng. Transgenic rice containing PPD has been generated previously. In the present study, extracts of this transgenic rice were evaluated to assess their antiadipogenic and anti-inflammatory activities. During adipogenesis, cells were treated with transgenic rice seed extracts. The results revealed that the concentrations of the rice seed extracts tested in this study did not affect cell viability at 3 days post-treatment. However, the rice seed extracts significantly reduced the accumulation of lipids in cells and suppressed the activation of CCAAT/enhancer-binding protein α (C/EBPα) and peroxisome proliferator-activated receptor γ (PPARγ), which in turn inhibited the expression of adipogenesis-related mRNAs, such as adiponectin, PPARγ, C/EBPα, sterol regulatory element-binding protein 1, glucose transport member 4, and fatty acid synthase. In adipocytes, the extracts significantly reduced the mRNA expression of inflammation-related factors following LPS treatment. The activation of NF-κB p65 and ERK 1/2 was inhibited in extract-treated adipocytes. Moreover, treatment with extract #8 markedly reduced the cell population of the G2/M phase. Collectively, these results indicate that transgenic rice containing PPD may act as an obesity-reducing and/or -preventing agent.
Collapse
Affiliation(s)
- Chaiwat Monmai
- Department of Agricultural Life Science, Sunchon National University, Sunchon 59722, Republic of Korea; (C.M.); (J.-S.K.); (E.-S.R.)
| | - Jin-Suk Kim
- Department of Agricultural Life Science, Sunchon National University, Sunchon 59722, Republic of Korea; (C.M.); (J.-S.K.); (E.-S.R.)
| | - Hyun Bo Sim
- Department of Biomedical Science, Sunchon National University, Sunchon 57922, Republic of Korea;
| | - Doh-Won Yun
- National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea; (D.-W.Y.); (S.-D.O.)
| | - Sung-Dug Oh
- National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea; (D.-W.Y.); (S.-D.O.)
| | - Eui-Shik Rha
- Department of Agricultural Life Science, Sunchon National University, Sunchon 59722, Republic of Korea; (C.M.); (J.-S.K.); (E.-S.R.)
| | - Jong-Jin Kim
- Department of Biomedical Science, Sunchon National University, Sunchon 57922, Republic of Korea;
| | - So-Hyeon Baek
- Department of Agricultural Life Science, Sunchon National University, Sunchon 59722, Republic of Korea; (C.M.); (J.-S.K.); (E.-S.R.)
| |
Collapse
|
5
|
Huangfu H, Du S, Zhang H, Wang H, Zhang Y, Yang Z, Zhang X, Ren S, Chen S, Wang C, Zhang Y, Zhou Y. Facile engineering of resveratrol nanoparticles loaded with 20(S)-protopanaxadiol for the treatment of periodontitis by regulating the macrophage phenotype. NANOSCALE 2023; 15:7894-7908. [PMID: 37060139 DOI: 10.1039/d2nr06452a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Periodontitis is an inflammatory disease, mainly caused by the formation of a subgingival plaque biofilm. In recent years, growing attention has been paid to immunotherapy in the treatment of periodontitis, and the importance of communal intervention associated with macrophage polarization was emphasized. Herein, resveratrol (RES) and 20(S)-protopanaxadiol (PPD) were successfully self-assembled into RES@PPD nanoparticles (NPs) by the phenolic resin reaction. RES@PPD NPs have good stability and biocompatibility. The combined application of PPD and RES enhances the anti-inflammatory and antioxidant properties of nanocomposites, remarkably reduces the level of reactive oxygen species, and finally realizes the coordinated regulation of host immunity in periodontitis. The detailed mechanism is as follows: RES@PPD NPs inhibit M1 polarization of macrophages, promote M2 polarization by scavenging ROS, and then inhibit the NF-κB signalling pathway to regulate host immunity. In the animal model of periodontitis, RES@PPD NPs can remarkably decrease the level of pro-inflammatory cytokines, up-regulate the anti-inflammatory cytokines, and exhibit a profound therapeutic effect on local inflammation. Therefore, RES@PPD NPs are effective in antioxidation and anti-inflammation, thus providing a promising candidate drug for the treatment of periodontitis.
Collapse
Affiliation(s)
- Huimin Huangfu
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Shulin Du
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China.
| | - Hao Zhang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Hanchi Wang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Yi Zhang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Zhen Yang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Xinwei Zhang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Sicong Ren
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Siyu Chen
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Cuizhu Wang
- School of Pharmaceutical Sciences Jilin University, Changchun, 130021, China.
| | - Yidi Zhang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China.
| | - Yanmin Zhou
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| |
Collapse
|
6
|
Dai Z, Wang H, Liu J, Zhang H, Li Q, Yu X, Zhang R, Yang C. Comparison of the Effects of Yucca saponin, Yucca schidigera, and Quillaja saponaria on Growth Performance, Immunity, Antioxidant Capability, and Intestinal Flora in Broilers. Animals (Basel) 2023; 13:ani13091447. [PMID: 37174484 PMCID: PMC10177514 DOI: 10.3390/ani13091447] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/10/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
The purpose of this study is to investigate the effects of Yucca saponin (YSa), Yucca schidigera (YS), and Quillaja Saponaria (QS) on growth performance, nitrogen metabolism, immune ability, antioxidant capability, and intestinal flora of yellow-feather broilers. This study randomly divided a total of 480 1-day yellow-feather broilers into 4 treatment groups. Factors in the 4 groups included CON group (basic diet), YSa group (basic diet mixed with 500 mg/kg YSa), YS group (basic diet mixed with 500 mg/kg YS), and QS group (basic diet mixed with 500 mg/kg QS). Throughout the 56-day study period, YSa, YS, and QS groups had higher average daily gain in broilers than the CON group (p < 0.01). The YS group had a lower feed gain ratio (F: G) in broilers than the CON group (p < 0.05). YSa, YS, and QS showed increased serum immunoglobin A (IgA), immunoglobin Y (IgY), immunoglobin M (IgM), and total antioxidant capacity (T-AOC) levels; enhanced acetic acid, butyric acid, and valeric acid levels of cecal content; and reduced contents of ammonia nitrogen, urea nitrogen, interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and malondialdehyde (MDA) in serum in broilers (p < 0.05). The relative abundance of Lachnoclostridium in the QS group was decreased compared with that in the CON group (p < 0.05). Higher IgA and IgY sera contents were observed in the YS group compared to the YSa and QS groups (p < 0.05). In contrast with the QS group, the serum IL-6 concentration of the YS group was reduced (p < 0.05). In conclusion, YSa, YS, and QS promoted growth performance, nitrogen metabolism, immunity, antioxidant capability, and intestinal flora in broilers. Through the comparison of YSa, YS, and QS, it was found that YS is more suitable as a feed additive to ameliorate the healthy growth of broilers.
Collapse
Affiliation(s)
- Zhenglie Dai
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| | - Huixian Wang
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| | - Jinsong Liu
- Key Agricultural Research Institute of Veagmax Green Animal Health Products of Zhejiang Province, Anji 313300, China
| | - Haoran Zhang
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| | - Qing Li
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| | - Xiaorong Yu
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| | - Ruiqiang Zhang
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| | - Caimei Yang
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| |
Collapse
|
7
|
Monmai C, Kim JS, Baek SH. Effect of Ginseng Sapogenin Protopanaxadiol-Enriched Rice (DJ-PPD) on Immunomodulation. PLANTS (BASEL, SWITZERLAND) 2023; 12:767. [PMID: 36840113 PMCID: PMC9959806 DOI: 10.3390/plants12040767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 06/18/2023]
Abstract
Protopanaxadiol (PPD), a gut microbiome-induced ginseng metabolite, has positive immune effects. We previously reported the immune-boosting and anti-inflammatory effects of PPD-enricshed rice seed extracts in normal and inflammatory cell environments, respectively. In the present study, the immunomodulatory activity of PPD-enriched transgenic rice seed extract (DJ-PPD), which exhibited the highest immune-related activity among all available extracts, was compared with that of commercially synthesized 20s-PPD (S-PPD) and natural ginseng root extract (GE), in RAW264.7 cells. Compared with S-PPD and GE treatment, DJ-PPD treatment (i) significantly promoted NF-κB p65 and c-Jun N-terminal protein kinase (JNK) phosphorylation; (ii) upregulated IL-1β, IL-6, COX-2, TLR-4, and TNF-α expression; (iii) and increased prostaglandin E2 (PGE2) production. However, there were no significant differences in the effects of the three treatments containing PPD-type sapogenin or saponins on nitric oxide (NO) production and phagocytic activity. In the inflammatory cell environment, DJ-PPD treatment markedly decreased the production of LPS-induced inflammatory factors, including NO and PGE2, as well as proinflammatory cytokine expression, by decreasing phosphorylated (p-)NF-κB p65, p-p38 MAPK, and p-JNK levels. Thus, DJ-PPD that does not require complex intestinal microbial processes to exert higher anti-inflammatory effects compared with S-PPD and GE. However, DJ-PPD exerted similar or higher immune-boosting effects (depending on inflammatory biomarkers) than S-PPD and GE. These findings indicate the potential of PPD-enriched transgenic rice as an alternative immunomodulatory agent.
Collapse
|
8
|
Wang Y, Mi X, Du Y, Li S, Yu L, Gao M, Yang X, Song Z, Yu H, Yang G. Design, Synthesis, and Anti-Inflammatory Activities of 12-Dehydropyxinol Derivatives. Molecules 2023; 28:molecules28031307. [PMID: 36770974 PMCID: PMC9921557 DOI: 10.3390/molecules28031307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 01/31/2023] Open
Abstract
Pyxinol skeleton is a promising framework of anti-inflammatory agents formed in the human liver from 20S-protopanaxadiol, the main active aglycone of ginsenosides. In the present study, a new series of amino acid-containing derivatives were produced from 12-dehydropyxinol, a pyxinol oxidation metabolite, and its anti-inflammatory activity was assessed using an NO inhibition assay. Interestingly, the dehydrogenation at C-12 of pyxinol derivatives improved their potency greatly. Furthermore, half of the derivatives exhibited better NO inhibitory activity than hydrocortisone sodium succinate, a glucocorticoid drug. The structure-activity relationship analysis indicated that the kinds of amino acid residues and their hydrophilicity influenced the activity to a great extent, as did R/S stereochemistry at C-24. Of the various derivatives, 5c with an N-Boc-protected phenylalanine residue showed the highest NO inhibitory activity and relatively low cytotoxicity. Moreover, derivative 5c could dose-dependently suppress iNOS, IL-1β, and TNF-α via the MAPK and NF-κB pathways, but not the GR pathway. Overall, pyxinol derivatives hold potential for application as anti-inflammatory agents.
Collapse
Affiliation(s)
- Yunxiao Wang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
| | - Xiaoliang Mi
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
| | - Yuan Du
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
| | - Shuang Li
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
| | - Liping Yu
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
| | - Meng Gao
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
| | - Xiaoyue Yang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
| | - Zhihua Song
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
| | - Hui Yu
- College of Food Engineering, Ludong University, Yantai 264025, China
- Correspondence: (H.Y.); (G.Y.)
| | - Gangqiang Yang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
- Correspondence: (H.Y.); (G.Y.)
| |
Collapse
|
9
|
Monmai C, Kim JS, Baek SH. Transgenic Rice Seed Extracts Exert Immunomodulatory Effects by Modulating Immune-Related Biomarkers in RAW264.7 Macrophage Cells. Nutrients 2022; 14:nu14194143. [PMID: 36235795 PMCID: PMC9573073 DOI: 10.3390/nu14194143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/17/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Protopanaxadiol (PPD), a native active triterpenoid present in Panax ginseng, has been reported to exert immune-related effects. We previously created PPD-producing transgenic rice by introducing the P. ginseng protopanaxadiol synthase and dammarenediol-II synthase genes into Dongjin rice. In the present study, the seeds of the T4 generation of this transgenic rice were tested for their immunomodulatory effects in RAW264.7 macrophage cells. Treatment with transgenic rice seed extract in RAW264.7 cells (i) significantly enhanced nitric oxide (NO) production in a dose-dependent manner without any cytotoxicity (up to 100 µg/mL), (ii) upregulated the expression of immune-related genes and increased production of the inflammation mediator prostaglandin E2 (PGE2), and (iii) activated nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) by promoting the phosphorylation of NF-κB p65, p38 MAPK, and c-Jun N-terminal protein kinase (JNK). In lipopolysaccharide (LPS)-treated RAW264.7 cells used to mimic the inflammation condition, treatment with transgenic rice seed extract significantly reduced NO production, proinflammatory cytokine expression, and PGE2 production, all of which are LPS-induced inflammation biomarkers, by inhibiting the phosphorylation of NF-κB p65, p38 MAPK, and JNK. Collectively, these results indicate that PPD-producing transgenic rice has immunomodulatory effects.
Collapse
|
10
|
Mitra A, Rahmawati L, Lee HP, Kim SA, Han CK, Hyun SH, Cho JY. Korean red ginseng water extract inhibits cadmium-induced lung injury via suppressing MAPK/ERK1/2/AP-1 pathway. J Ginseng Res 2022; 46:690-699. [PMID: 36090678 PMCID: PMC9459071 DOI: 10.1016/j.jgr.2022.04.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/05/2022] [Accepted: 04/18/2022] [Indexed: 11/06/2022] Open
Abstract
Background Few studies reported the therapeutic effect of Korean Red Ginseng (KRG) in lung inflammatory diseases. However, the anti-inflammatory role and underlying molecular in cadmium-induced lung injury have been poorly understood, directly linked to chronic lung diseases (CLDs): chronic obstructive pulmonary disease (COPD), cancer etc. Therefore, in this study we aim to investigate the therapeutic activities of water extract of KRG (KRG-WE) in mouse cadmium-induced lung injury model. Method The anti-inflammatory roles and underlying mechanisms of KRG-WE were evaluated in vitro under cadmium-stimulated lung epithelial cells (A549) and HEK293T cell line and in vivo in cadmium-induced lung injury mouse model using semi-quantitative polymerase chain reaction (RT-PCR), quantitative real-time PCR (qPCR), luciferase assay, immunoblotting, and FACS. Results KRG-WE strongly ameliorated the symptoms of CdSO4-induced lung injury in mice according to total cell number in bronchoalveolar lavage fluid (BALF) and severity scores as well as cytokine levels. KRG-WE significantly suppressed the upregulation of inflammatory signaling comprising mitogen-activated protein kinases (MAPK) and their upstream enzymes. In in vitro study, KRG-WE suppressed expression of interleukin (IL)-6, matrix metalloproteinase (MMP)-2, and IL-8 while promoting recovery in CdSO4-treated A549 cells. Similarly, KRG-WE reduced phosphorylation of MAPK and c-Jun/c-Fos in cadmium-exposed A549 cells. Conclusion KRG-WE was found to attenuate symptoms of cadmium-induced lung injury and reduce the expression of inflammatory genes by suppression of MAPK/AP-1-mediated pathway.
Collapse
|
11
|
Jia J, Wang Y, Huang R, Du F, Shen X, Yang Q, Li J. Protein disulfide-isomerase A3 knockdown attenuates oxidized low-density lipoprotein-induced oxidative stress, inflammation and endothelial dysfunction in human umbilical vein endothelial cells by downregulating activating transcription factor 2. Bioengineered 2022; 13:1436-1446. [PMID: 34983301 PMCID: PMC8805980 DOI: 10.1080/21655979.2021.2018980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/11/2021] [Indexed: 11/03/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease implicated in oxidative stress and endothelial dysfunction. Protein disulfide-isomerase A3 (PDIA3) has been reported to regulate oxidative stress and suppress inflammation. This study aimed to explore the function of PDIA3 in atherosclerosis and the underlying mechanisms. PDIA3 expression in oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cells (HUVECs) was detected using RT-qPCR and Western blotting. Following PDIA3 knockdown through transfection with small interfering RNA targeting PDIA3, cell viability, oxidative stress and inflammation in ox-LDL-induced HUVECs was examined using a Cell Counting Kit-8, corresponding kits and ELISA, respectively. The levels of CD31, α-smooth muscle, iNOS, p-eNOS, eNOS and NO were assessed using RT-qPCR, Western blotting and an NO kit to reflect endothelial dysfunction in ox-LDL-induced HUVECs. The relationship between PDIA3 and the activating transcription factor 2 (ATF2) was confirmed using co-immunoprecipitation. In addition, ATF2 expression was examined following PDIA3 silencing. The results indicated that PDIA3 was highly expressed in ox-LDL-induced HUVECs. PDIA3 silencing increased cell viability, and reduced oxidative stress and inflammation, as evidenced by the decreased levels of reactive oxygen species, malondialdehyde, TNF-α, IL-1β and IL-6, and increased superoxide dismutase and glutathione peroxidase activity. In addition, PDIA3 deletion improved endothelial dysfunction. PDIA3 interacted with ATF2, and PDIA3 deletion downregulated ATF2 expression. Furthermore, ATF2 overexpression reversed the effects of PDIA3 knockdown on ox-LDL-induced damage of HUVECs. Collectively, PDIA3 knockdown was found to attenuate ox-LDL-induced oxidative stress, inflammation and endothelial dysfunction in HUVECs by downregulating ATF2 expression, showing promise for the future treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jing Jia
- Department of Anesthetic Surgery, Baotou Steel Hospital, Baotou, China
| | - Yueping Wang
- Department of Cardiology, Baotou Steel Hospital, Baotou, China
| | - Ruijuan Huang
- Laser Treatment Center, Baotou Steel Hospital, Baotou, China
| | - Fengxia Du
- Department of Intensive Medicine, Baotou Steel Hospital, Baotou, China
| | - Xiaozhu Shen
- Department of Geriatrics, The Second People’s Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Qiurong Yang
- Nursing Department, The Second People’s Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Juan Li
- Nursing Department, The Second People’s Hospital of Lianyungang, Lianyungang, Jiangsu, China
| |
Collapse
|
12
|
Liang H, Deng P, Ma YF, Wu Y, Ma ZH, Zhang W, Wu JD, Qi YZ, Pan XY, Huang FS, Lv SY, Han JL, Dai WD, Chen Z. Advances in Experimental and Clinical Research of the Gouty Arthritis Treatment with Traditional Chinese Medicine. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:8698232. [PMID: 34721646 PMCID: PMC8550850 DOI: 10.1155/2021/8698232] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/12/2021] [Accepted: 09/20/2021] [Indexed: 12/27/2022]
Abstract
Gouty arthritis (GA) is a multifactorial disease whose pathogenesis is utterly complex, and the current clinical treatment methods cannot wholly prevent GA development. Western medicine is the primary treatment strategy for gouty arthritis, but it owns an unfavorable prognosis. Therefore, the prevention and treatment of GA are essential. In China, traditional Chinese medicine (TCM) has been adopted for GA prevention and treatment for thousands of years. Gout patients are usually treated with TCM according to their different conditions, and long-term results can be achieved by improving their physical condition. And TCM has been proved to be an effective method to treat gout in modern China. Nevertheless, the pharmacological mechanism of TCM for gout is still unclear, which limits its spread. The theory of prevention and treatment of gout with TCM is more well acknowledged in China than in abroad. In this article, Chinese herbs and ancient formula for gout were summarized first. A total of more than 570 studies published from 2004 to June 2021 in PubMed, Medline, CNKI, VIP, Web of Science databases and Chinese Pharmacopoeia and traditional Chinese books were searched; the current status of TCM in the treatment of GA was summarized from the following aspects: articular chondrocyte apoptosis inhibition, antioxidative stress response, inflammatory cytokine levels regulation, uric acid excretion promotion, immune function regulation, uric acid reduction, and intestinal flora improvement in subjects with gout. The literature review concluded that TCM has a specific curative effect on the prevention and treatment of GA, particularly when combined with modern medical approaches. However, lacking a uniform definition of GA syndrome differentiation and the support of evidence-based medicine in clinical practice have provoked considerable concern in previous studies, which needs to be addressed in future research.
Collapse
Affiliation(s)
- Huan Liang
- School of Graduates, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Pin Deng
- School of Graduates, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yu-Feng Ma
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, China
| | - Yan Wu
- School of Graduates, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhan-Hua Ma
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, China
| | - Wei Zhang
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, China
| | - Jun-De Wu
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, China
| | - Yin-Ze Qi
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, China
| | - Xu-Yue Pan
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, China
| | - Fa-Sen Huang
- School of Graduates, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Si-Yuan Lv
- School of Graduates, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jing-Lu Han
- School of Graduates, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wen-Da Dai
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, China
| | - Zhaojun Chen
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, China
| |
Collapse
|
13
|
Rahmawati L, Aziz N, Oh J, Hong YH, Woo BY, Hong YD, Manilack P, Souladeth P, Jung JH, Lee WS, Jeon MJ, Kim T, Hossain MA, Yum J, Kim JH, Cho JY. Cissus subtetragona Planch. Ameliorates Inflammatory Responses in LPS-induced Macrophages, HCl/EtOH-induced Gastritis, and LPS-induced Lung Injury via Attenuation of Src and TAK1. Molecules 2021; 26:molecules26196073. [PMID: 34641616 PMCID: PMC8512965 DOI: 10.3390/molecules26196073] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/24/2022] Open
Abstract
Several Cissus species have been used and reported to possess medicinal benefits. However, the anti-inflammatory mechanisms of Cissus subtetragona have not been described. In this study, we examined the potential anti-inflammatory effects of C. subtetragona ethanol extract (Cs-EE) in vitro and in vivo, and investigated its molecular mechanism as well as its flavonoid content. Lipopolysaccharide (LPS)-induced macrophage-like RAW264.7 cells and primary macrophages as well as LPS-induced acute lung injury (ALI) and HCl/EtOH-induced acute gastritis mouse models were utilized. Luciferase assays, immunoblotting analyses, overexpression strategies, and cellular thermal shift assay (CETSA) were performed to identify the molecular mechanisms and targets of Cs-EE. Cs-EE concentration-dependently reduced the secretion of NO and PGE2, inhibited the expression of inflammation-related cytokines in LPS-induced RAW264.7 cells, and decreased NF-κB- and AP-1-luciferase activity. Subsequently, we determined that Cs-EE decreased the phosphorylation events of NF-κB and AP-1 pathways. Cs-EE treatment also significantly ameliorated the inflammatory symptoms of HCl/EtOH-induced acute gastritis and LPS-induced ALI mouse models. Overexpression of HA-Src and HA-TAK1 along with CETSA experiments validated that inhibited inflammatory responses are the outcome of attenuation of Src and TAK1 activation. Taken together, these findings suggest that Cs-EE could be utilized as an anti-inflammatory remedy especially targeting against gastritis and acute lung injury by attenuating the activities of Src and TAK1.
Collapse
Affiliation(s)
- Laily Rahmawati
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (L.R.); (N.A.); (J.O.); (Y.H.H.)
| | - Nur Aziz
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (L.R.); (N.A.); (J.O.); (Y.H.H.)
| | - Jieun Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (L.R.); (N.A.); (J.O.); (Y.H.H.)
| | - Yo Han Hong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (L.R.); (N.A.); (J.O.); (Y.H.H.)
| | - Byoung Young Woo
- AmorePacific R&D Center, Yongin 17074, Korea; (B.Y.W.); (Y.D.H.)
| | - Yong Deog Hong
- AmorePacific R&D Center, Yongin 17074, Korea; (B.Y.W.); (Y.D.H.)
| | - Philaxay Manilack
- Department of Forestry, Ministry of Agriculture and Forestry, Vientiane P.O. Box 811, Laos;
| | - Phetlasy Souladeth
- Department of Forest Management, Faculty of Forest Science, National University of Laos, Vientiane P.O. Box 7322, Laos;
| | - Ji Hwa Jung
- Division of Zoology, Honam National Institute of Biological Resources, Mokpo 58762, Korea;
| | - Woo Shin Lee
- Department of Forest Sciences, College of Agriculture and Life Science, Seoul National University, Seoul 08826, Korea;
| | - Mi Jeong Jeon
- Animal Resources Division, National Institute of Biological Resources, Incheon 22689, Korea; (M.J.J.); (T.K.); (J.Y.)
| | - Taewoo Kim
- Animal Resources Division, National Institute of Biological Resources, Incheon 22689, Korea; (M.J.J.); (T.K.); (J.Y.)
| | - Mohammad Amjad Hossain
- Department of Veterinary Physiology, College of Medicine, Chonbuk National University, Iksan 54596, Korea;
| | - Jinwhoa Yum
- Animal Resources Division, National Institute of Biological Resources, Incheon 22689, Korea; (M.J.J.); (T.K.); (J.Y.)
| | - Jong-Hoon Kim
- Department of Veterinary Physiology, College of Medicine, Chonbuk National University, Iksan 54596, Korea;
- Correspondence: (J.-H.K.); (J.Y.C.); Tel.: +82-63-270-2563 (J.-H.K.); +82-31-290-7876 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (L.R.); (N.A.); (J.O.); (Y.H.H.)
- Correspondence: (J.-H.K.); (J.Y.C.); Tel.: +82-63-270-2563 (J.-H.K.); +82-31-290-7876 (J.Y.C.)
| |
Collapse
|
14
|
Tian C, Chang Y, Wang R, Kang Z, Wang Q, Tong Z, Zhou A, Cui C, Liu M. Optimization of ultrasound extraction of Tribulus terrestris L. leaves saponins and their HPLC-DAD-ESI-MS n profiling, anti-inflammatory activity and mechanism in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2021; 278:114225. [PMID: 34038799 DOI: 10.1016/j.jep.2021.114225] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tribulus terrestris L., as an annual herb plant from Zygophyllaceae, exhibits many biological activities, and its main chemical constituents are saponins. However, the extraction process, chemical compositions, anti-inflammatory effect and mechanism of total saponins from Tribulus terrestris L. leaves are still unclear. AIM OF THE STUDY The present study extensively evaluated the extraction process, major components, anti-inflammatory action and mechanism of Tribulus terrestris L. leaves saponins. MATERIALS AND METHODS The ultrasonic extraction and response surface methods were adopted for optimization of extraction technology of total saponins from Tribulus terrestris L. leaves, and its compositions were detected with LC-MSn method. The anti-inflammatory activity of total saponins was studied by lipopolysaccharide induced RAW 264.7 cells and acute lung injury mice models. RESULTS The ultrasonic extraction parameters of saponins fraction, including ethanol concentration 30%, extraction time 55 min, ratio of solvent to material 35:1 ml/g and extraction temperature 46 °C, were screened by response surface method with the extracting rate 5.49%, and thirty compositions were detected with LC-MSn method. Moreover, saponins fraction can play a stronger anti-inflammatory effect by reducing the phagocytic activity and pulmonary edema, and protection of morphology of RAW 264.7 cells and lung tissues, and decreasing the content of NO and TNF-α. Moreover, it was revealed that total saponins extract can exert the anti-inflammatory action by the inhibition of the activation of the TLR4-TRAF6-NF-κB signalling pathway. CONCLUSION These studies imply that Tribulus terrestris L. leaves saponins may be an important anti-inflammatory drug in clinic.
Collapse
Affiliation(s)
- Chunlian Tian
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No 120 Dongling Road Shenhe Dist., 110866, Shenyang Liaoning Prov., People's Republic of China; Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, 264005, Yantai Shangdong Prov., People's Republic of China
| | - Yu Chang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No 120 Dongling Road Shenhe Dist., 110866, Shenyang Liaoning Prov., People's Republic of China
| | - Ruxia Wang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No 120 Dongling Road Shenhe Dist., 110866, Shenyang Liaoning Prov., People's Republic of China
| | - Zhiruo Kang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No 120 Dongling Road Shenhe Dist., 110866, Shenyang Liaoning Prov., People's Republic of China
| | - Qi Wang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No 120 Dongling Road Shenhe Dist., 110866, Shenyang Liaoning Prov., People's Republic of China
| | - Zhifan Tong
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No 120 Dongling Road Shenhe Dist., 110866, Shenyang Liaoning Prov., People's Republic of China
| | - Aohua Zhou
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No 120 Dongling Road Shenhe Dist., 110866, Shenyang Liaoning Prov., People's Republic of China
| | - Cancan Cui
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No 120 Dongling Road Shenhe Dist., 110866, Shenyang Liaoning Prov., People's Republic of China
| | - Mingchun Liu
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No 120 Dongling Road Shenhe Dist., 110866, Shenyang Liaoning Prov., People's Republic of China.
| |
Collapse
|
15
|
Chen C, Lv Q, Li Y, Jin YH. The Anti-Tumor Effect and Underlying Apoptotic Mechanism of Ginsenoside Rk1 and Rg5 in Human Liver Cancer Cells. Molecules 2021; 26:molecules26133926. [PMID: 34199025 PMCID: PMC8271777 DOI: 10.3390/molecules26133926] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/20/2022] Open
Abstract
Ginsenoside Rk1 and Rg5 are minor ginseng saponins that have received more attention recently because of their high oral bioavailability. Each of them can effectively inhibit the survival and proliferation of human liver cancer cells, but the underlying mechanism remains largely unknown. Network pharmacology and bioinformatics analysis demonstrated that G-Rk1 and G-Rg5 yielded 142 potential targets, and shared 44 putative targets associated with hepatocellular carcinoma. Enrichment analysis of the overlapped genes showed that G-Rk1 and G-Rg5 may induce apoptosis of liver cancer cells through inhibition of mitogen-activated protein kinase (MAPK) and nuclear factor-kappa B (NF-κB) signal pathways. Methyl thiazolyl tetrazolium (MTT) assay was used to confirm the inhibition of cell viability with G-Rk1 or G-Rg5 in highly metastatic human cancer MHCC-97H cells. We evaluated the apoptosis of MHCC-97H cells by using flow cytometry and 4′,6-diamidino-2-phenylindole (DAPI) staining. The translocation of Bax/Bak led to the depolarization of mitochondrial membrane potential and release of cytochrome c and Smac. A sequential activation of caspase-9 and caspase-3 and the cleavage of poly(ADP-ribose) polymerase (PARP) were observed after that. The levels of anti-apoptotic proteins were decreased after treatment of G-Rk1 or G-Rg5 in MHCC-97H cells. Taken together, G-Rk1 and G-Rg5 promoted the endogenous apoptotic pathway in MHCC-97H cells by targeting and regulating some critical liver cancer related genes that are involved in the signal pathways associated with cell survival and proliferation.
Collapse
Affiliation(s)
| | | | - Yang Li
- Correspondence: (Y.L.); (Y.-H.J.)
| | | |
Collapse
|
16
|
Ma Y, Wang HY, Zhang XF, Zhao FL, Meng QG. Crystal structure of (3 S,8 R,10 R,12 R,14 R)-12-hydroxy-4,4,8,10,14-pentamethyl-17-(( R)-2,6,6-trimethyltetrahydro-2 H-pyran-2-yl) hexadecahydro-1 H-cyclopenta[ a]phenanthren-3-yl acetate, C 32H 54O 4. Z KRIST-NEW CRYST ST 2021. [DOI: 10.1515/ncrs-2020-0311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
C32H54O4, monoclinic, P21 (no. 4), a = 7.8727(1) Å, b = 19.9173(2) Å, c = 19.1801(2) Å, β = 94.539(1)°, V = 2998.06(6) Å3, Z = 4, R
gt
(F) = 0.0370, wR
ref
(F
2) = 0.0983, T = 293 K.
Collapse
Affiliation(s)
- Ying Ma
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education , Yantai University , Yantai , PR China
| | - Hui-Yun Wang
- College of Pharmacy , Jining Medical University , Rizhao , 276826, PR China
| | - Xiao-Fan Zhang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education , Yantai University , Yantai , PR China
| | - Feng-Lan Zhao
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education , Yantai University , Yantai , PR China
| | - Qing-Guo Meng
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education , Yantai University , Yantai , PR China
| |
Collapse
|
17
|
Li P, Xing J, Zhang J, Jiang J, Liu X, Zhao D, Zhang Y. Inhibition of long noncoding RNA HIF1A-AS2 confers protection against atherosclerosis via ATF2 downregulation. J Adv Res 2020; 26:123-135. [PMID: 33133688 PMCID: PMC7584671 DOI: 10.1016/j.jare.2020.07.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 01/17/2023] Open
Abstract
Introduction In atherosclerotic lesions, extensive inflammation of the vessel wall contributes to plaque instability. Long noncoding RNAs (lncRNAs) play important roles in diverse biological processes in atherosclerosis. Objectives Here, we aim to identify the functional role and regulatory mechanisms of lncRNA hypoxia-inducible factor 1 alpha-antisense RNA 2 (HIF1A-AS2) in atherosclerotic inflammation. Methods An atherosclerotic mouse model was induced in ApoE-/- mice by high fat diet (HFD). Endothelial cells (ECs), human aortic smooth muscle cells (SMCs) or human coronary artery endothelial cells (HCAECs) were exposed to ox-LDL to develop the in vitro model. The effects of lncRNA HIF1A-AS2 on inflammation were evaluated by determining levels of inflammatory factors tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6) and levels of adhesion molecules vascular cell adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), and macrophage cationic peptide 1 (MCP-1). Results It was established that lncRNA HIF1A-AS2 and ATF2 were highly expressed in atherosclerotic ApoE-/- mice. Downregulating lncRNA HIF1A-AS2 in ox-LDL-exposed ECs, SMCs and HCAECs inhibited inflammation by reducing levels of pro-inflammatory factors and adhesion molecules. LncRNA HIF1A-AS2 bound to the transcription factor USF1 to elevate ATF2 expression. USF1 overexpression counteracted the suppressive effect of lncRNA HIF1A-AS2 silencing on ox-LDL-induced inflammation. Knockdown of lncRNA HIF1A-AS2 or ATF2 could also attenuate inflammation in atherosclerotic mice. Collectively, the present study demonstrates that downregulation of lncRNA HIF1A-AS2 represses the binding of USF1 to the ATF2 promoter region and then inhibits ATF2 expression, thereby suppressing atherosclerotic inflammation. Conclusion This study suggests lncRNA HIF1A-AS2 as an promising therapeutic target for atherosclerosis.
Collapse
Key Words
- ATCC, American Type Culture Collection
- ATF2, activating transcription factor 2
- Activating transcription factor
- Atherosclerosis
- CAD, coronary artery disease
- CCK-8, cell counting kit-8
- ChIP, Chromatin immunoprecipitation
- DMEM, Dulbecco’s modified Eagle’s medium
- ECs, endothelial cells
- ELISA, enzyme linked immunosorbent assay
- GAPDH, Glyceraldehyde-3-phosphate dehydrogenase
- HCAECs, human coronary artery endothelial cells
- HE, Hematoxylin-eosin
- HFD, high fat diet
- HIF1A-AS2, hypoxia-inducible factor 1 alpha-antisense RNA 2
- Hypoxia-inducible factor 1 alpha-antisense RNA 2
- ICAM-1, intercellular adhesion molecule-1
- IL-1β, interleukin-1β
- IL-6, interleukin-6
- IgG, immunoglobulin G
- Inflammation
- LDL, low-density lipoprotein
- Long noncoding RNA
- MCP-1, monocyte chemoattractant protein-1
- ND, normal diet
- PBS, phosphate buffered saline
- RIP, RNA binding protein immunoprecipitation
- RT-qPCR, reverse transcription quantitative polymerase chain reaction
- SMCs, smooth muscle cells
- TNF-α, tumor necrosis factor-α
- Transcription factor
- USF1, upstream stimulatory factor 1
- Upstream transcription factor 1
- VCAM-1, vascular cell adhesion molecule 1
- lncRNAs, long noncoding RNAs
- ox-LDL, oxidized-low-density lipoprotein
- sh, short hairpin RNA
- si-NC, small interfering RNA-negative control
Collapse
Affiliation(s)
- Pengcheng Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Junhui Xing
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Jielei Zhang
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Jianwu Jiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Xuemeng Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Di Zhao
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
- Corresponding authors at: Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou 450052, Henan Province, PR China (D. Zhao). Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou 450052, Henan Province, PR China (Y. Zhang).
| | - Yanzhou Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
- Corresponding authors at: Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou 450052, Henan Province, PR China (D. Zhao). Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou 450052, Henan Province, PR China (Y. Zhang).
| |
Collapse
|
18
|
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
|
19
|
Sun Y, Fang X, Gao M, Wang C, Gao H, Bi W, Tang H, Cui Y, Zhang L, Fan H, Yu H, Yang G. Synthesis and Structure-Activity Relationship of Pyxinol Derivatives as Novel Anti-Inflammatory Agents. ACS Med Chem Lett 2020; 11:457-463. [PMID: 32292550 DOI: 10.1021/acsmedchemlett.9b00562] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/12/2020] [Indexed: 01/19/2023] Open
Abstract
Pyxinol, the main metabolite of 20S-protopanaxadiol in human liver, was chosen as a novel skeleton for the development of anti-inflammatory agents. Pyxinol derivatives modified at C-3, C-12, or C-25 and selected stereoisomers were designed, prepared, and investigated for in vitro anti-inflammatory activities. Structure-activity relationship (SAR), focused on skeleton, was analyzed based on their ability to inhibit lipopolysaccharide (LPS)-induced nitric oxide (NO) synthesis. The preliminary SAR results signified that the biological activity of the pyxinol derivatives is largely dependent on the R/S stereochemistry of pyxinol skeleton and the hydroxy at C-3 is a modifiable position. Among the tested compounds, the 3-oximinopyxinol (4a) exhibited the most potent NO-inhibitory activity and was even comparable to the steroid drug. Furthermore, compound 4a also significantly decreased LPS-induced TNF-α and IL-6 synthesis and iNOS and COX-2 expressions via the NF-κB pathway. This study proves that pyxinol is an interesting skeleton for anti-inflammatory drug discovery.
Collapse
Affiliation(s)
- Yixiao Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Xiaojuan Fang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Meng Gao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Conghui Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Hongyan Gao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Wenjing Bi
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Hanhan Tang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Yetong Cui
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Leiming Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Huaying Fan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Hui Yu
- College of Food Engineering, Ludong University, Yantai 264025, China
| | - Gangqiang Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| |
Collapse
|
20
|
Kim JW, Han SW, Cho JY, Chung IJ, Kim JG, Lee KH, Park KU, Baek SK, Oh SC, Lee MA, Oh D, Shim B, Ahn JB, Shin D, Lee JW, Kim YH. Korean red ginseng for cancer-related fatigue in colorectal cancer patients with chemotherapy: A randomised phase III trial. Eur J Cancer 2020; 130:51-62. [PMID: 32172198 DOI: 10.1016/j.ejca.2020.02.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/31/2020] [Accepted: 02/08/2020] [Indexed: 01/23/2023]
Abstract
BACKGROUND Cancer-related fatigue (CRF) is a common symptom and has a negative impact on prognosis in cancer patients. CRF could be improved by Korean red ginseng (KRG). PATIENTS AND METHODS For this randomised and double-blinded trial, colorectal cancer patients who received mFOLFOX-6 were randomly assigned to either KRG 2000 mg/day (n = 219) or placebo (n = 219) for 16 weeks. CRF was evaluated using the mean area under the curve (AUC) change from baseline of brief fatigue inventory (BFI) as the primary endpoint. Fatigue-related quality of life, stress, and adverse events were evaluated as secondary endpoints. RESULTS In the full analysis group, KRG up to 16 weeks improved CRF by the mean AUC change from baseline of BFI compared to placebo, particularly in "Mood" and "Walking ability" (P = 0.038, P = 0.023, respectively). In the per-protocol group, KRG led to improved CRF in the global BFI score compared with the placebo (P = 0.019). Specifically, there were improvements in "Fatigue right now," "Mood," "Relations with others," "Walking ability," and "Enjoyment of life" at 16 weeks (P = 0.045, P = 0.006, P = 0.028, P = 0.003, P = 0.036, respectively). In subgroups of female patients, ≥60 years old, with high compliance (≥80%) or more baseline fatigue, the beneficial effects of KRG were more enhanced than that of placebo. Although neutropenia was more frequent in KRG than placebo, the incidence of all adverse events was similar. CONCLUSIONS KRG could be safely combined with mFOLFOX-6 chemotherapy in colorectal cancer patients, and reduced CRF compared with placebo.
Collapse
Affiliation(s)
- Jin Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Sae Won Han
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jae Yong Cho
- Department of Internal Medicine, GangNam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ik-Joo Chung
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University College of Medicine, Hwasun, Republic of Korea
| | - Jong Gwang Kim
- Department of Oncology/Hematology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Kyung Hee Lee
- Department of Internal Medicine, Yeungnam University Medical Center, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Keon Uk Park
- Department of Internal Medicine, Keimyung University Dongsan Medical Center, College of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Sun Kyung Baek
- Department of Internal Medicine, Kyung Hee University Medical Center, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Sang Cheul Oh
- Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Myung Ah Lee
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Doyeun Oh
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Republic of Korea
| | - Byoungyong Shim
- Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Republic of Korea
| | - Joong Bae Ahn
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dongbok Shin
- Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Jae Won Lee
- Department of Statistics, Korea University, Seoul, Republic of Korea
| | - Yeul Hong Kim
- Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
21
|
Hong X, Mat Isa N, Fakurazi S, Safinar Ismail I. Phytochemical and anti-inflammatory properties of Scurrula ferruginea (Jack) Danser parasitising on three different host plants elucidated by NMR-based metabolomics. PHYTOCHEMICAL ANALYSIS : PCA 2020; 31:15-27. [PMID: 31243835 DOI: 10.1002/pca.2861] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 06/09/2023]
Abstract
INTRODUCTION Scurrula ferruginea (Jack) Danser is a hemi-parasitic shrub that is widely used as a traditional herbal medicine. OBJECTIVES To elucidate the anti-inflammatory activity of S. ferruginea parasitising on three different hosts of Vitex negundo L., Micromelum minutum (G. Forst.) Wight & Arn. and Tecoma stans (L.) Juss ex HBK., as well as, to determine the metabolite differences related to their anti-inflammatory properties. MATERIALS AND METHODS Two plant parts of S. ferruginea, stems and leaves, were extracted in water. The freeze-dried stem of S. ferruginea grown on T. stans was liquid-liquid partitioned into several solvents. Their potential anti-inflammatory activity was assessed via inhibition of nitric oxide (NO) production in lipopolysaccharide (LPS) and interferon-γ (IFN-γ)-induced RAW 264.7 macrophage cells. The metabolite variation was examined using proton nuclear magnetic resonance (1 H-NMR) combined with multivariate data analysis (MVDA). RESULTS Scurrula ferruginea stems parasitising on T. stans and V. negundo which were freeze dried exhibited higher anti-inflammatory activity with IC50 values of 114.47 ± 2.96 and 118.87 ± 2.31 μg/mL, respectively. The mid-polar ethyl acetate fraction of S. ferruginea hosted on T. stans displayed the highest NO inhibition with 84.80 ± 0.45% at 200 μg/mL. Principal component analysis (PCA) indicated notable and clear discriminations among the different plant parts and host plants based on the identified metabolites. Furthermore, partial least squares (PLS) regression model suggested the anti-inflammatory bioactivity might be associated with the presence of choline, isoleucine, catechin, leucine and chlorogenic acid. CONCLUSION This study suggests S. ferruginea could serve as a potential anti-inflammatory agent, highlighting the importance of T. stans as the host plant.
Collapse
Affiliation(s)
- Xia Hong
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Hebei Key Laboratory of Research and Development for Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei, P. R. China
| | - Nurulfiza Mat Isa
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Sharida Fakurazi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Intan Safinar Ismail
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
22
|
Zhao RL, Wang HY, Luan MZ, Zheng X, Zhao FL, Meng QG. Crystal structure of (3 R,5 R,8 R,9 R,10 R,12 R,13 R,14 R)-4,4,8,10,14-pentamethyl-17-(( R)-2,6,6-trimethyltetrahydro-2 H-pyran-2-yl)hexadecahydro-1 H-cyclopenta[ a]phenanthrene-3,12-diol, C 30H 52O 3. Z KRIST-NEW CRYST ST 2019. [DOI: 10.1515/ncrs-2019-0533] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
C30H52O3, orthorhombic, P212121 (no. 19), a = 6.71147(14) Å, b = 16.6565(4) Å, c = 25.0815(6) Å, V = 2803.85(11) Å3, Z = 4, R
gt (F) = 0.0405, wR
ref (F
2) = 0.1092, T = 293(2) K.
Collapse
Affiliation(s)
- Ruo-Lin Zhao
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University , Yantai , P.R. China
| | - Hui-Yun Wang
- College of Pharmacy, Jining Medical University , Rizhao 276826 , P.R. China
| | - Ming-Zhu Luan
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University , Yantai , P.R. China
| | - Xia Zheng
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University , Yantai , P.R. China
| | - Feng-Lan Zhao
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University , Yantai , P.R. China
| | - Qing-Guo Meng
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University , Yantai , P.R. China
| |
Collapse
|
23
|
Kim MH, Kim KT, Sohn SY, Lee JY, Lee CH, Yang H, Lee BK, Lee KW, Kim DD. Formulation And Evaluation Of Nanostructured Lipid Carriers (NLCs) Of 20(S)-Protopanaxadiol (PPD) By Box-Behnken Design. Int J Nanomedicine 2019; 14:8509-8520. [PMID: 31749618 PMCID: PMC6818673 DOI: 10.2147/ijn.s215835] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/24/2019] [Indexed: 12/15/2022] Open
Abstract
Background 20(S)-Protopanaxadiol (PPD) has a higher anti-wrinkle effect than the other glycone forms of ginsenosides. However, as PPD has low solubility in water and a high molecular weight, it cannot easily penetrate the stratum corneum layer, which is the rate-limiting step of topical skin delivery. Thus, the objective was to enhance the topical skin deposition of PPD using an optimized nanostructured lipid carriers (NLC) formulation. NLC formulations were optimized using a Box-Behnken design. Materials and methods NLC formulations were optimized using a Box-Behnken design, where the amount of PDD (X1), volume of the liquid lipid (X2), and amount of surfactant (X3) were set as the independent variables, while the particle size (Y1), polydispersity index (PDI) (Y2), and entrapment efficiency (EE) (Y3) were dependent factors. An in vitro deposition study was performed using Strat-M® and human cadaver skin, while in vivo skin irritation effect of the NLC formulation was evaluated in humans. Results An NLC was successfully prepared based on the optimized formulation determined using the Box-Behnken design. The particle size, PDI, and EE of the NLC showed less than 5% difference from the predicted values. The in vitro deposition of PPD after the application of the NLC formulation on a Strat-M® artificial membrane and human cadaver skin was significantly higher than that of the controls. Moreover, NLC formulations with and without PDD were not skin irritants in a human study. Conclusion An NLC formulation for the topical delivery of PPD was successfully optimized using the Box-Behnken design, and could be further developed to enhance the topical skin deposition of PPD.
Collapse
Affiliation(s)
- Min-Hwan Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ki-Taek Kim
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| | - Seo-Yeon Sohn
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jae-Young Lee
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Chang Hyung Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Hee Yang
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Bo Kyung Lee
- Research & Development Center, BOBSNU Co., Ltd., Suwon, Gyeonggi-Do 16229, Republic of Korea
| | - Ki Won Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea.,Research & Development Center, BOBSNU Co., Ltd., Suwon, Gyeonggi-Do 16229, Republic of Korea
| | - Dae-Duk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
24
|
Kim JC, Jeon JY, Yang WS, Kim CH, Eom DW. Combined Amelioration of Ginsenoside (Rg1, Rb1, and Rg3)-enriched Korean Red Ginseng and Probiotic Lactobacillus on Non-alcoholic Fatty Liver Disease. Curr Pharm Biotechnol 2019; 20:222-231. [PMID: 30854954 DOI: 10.2174/1389201020666190311143554] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 11/15/2018] [Accepted: 02/15/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Red ginseng is a traditional medicine that has been used to treat numerous metabolic and inflammatory diseases. Probiotic administration has been established to have beneficial effects in non-alcoholic fatty liver disease (NAFLD). The purpose of this study was to determine whether a combination of Korean red ginseng (KRG) and probiotics could synergistically reduce NAFLD and liver inflammation compared with the effects reported for each individual product. METHOD db/db and C57BL/6 mice were fed a normal chow diet and high-fat diet (HFD), respectively, and were treated with KRG, probiotics, or both. Samples were examined for lipid content, kinase protein phosphorylation, and gene expression patterns. RESULTS KRG- and probiotic-treated HFD-fed mice exhibited a reduction in body weight and a decrease in inflammatory cytokine secretion compared with the non-treated control mice. The same treatment was less successful in improving NAFLD parameters in the db/db mice while the combination of both products did not enhance their therapeutic potential. CONCLUSION The results of this study indicate that KRG and probiotics administration ameliorated NAFLD symptoms in a mouse model of dyslipidemia by reducing weight gain and liver inflammation. Coadministration of both products did not enhance their efficacy, and further research should be conducted to clarify their mechanisms of action.
Collapse
Affiliation(s)
- Jin-Chul Kim
- Natural Product Research Institute, Korea Institute of Science and Technology, Gangneung, Korea
| | - Joo-Yeong Jeon
- Natural Product Research Institute, Korea Institute of Science and Technology, Gangneung, Korea
| | | | - Cheorl-Ho Kim
- Department of Biological Sciences, SungKyunKwan University, Suwon, Kyungki-do, Korea
| | - Dae-Woon Eom
- Department of Pathology, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Korea
| |
Collapse
|
25
|
Thelenota ananas saponin extracts attenuate the atherosclerosis in apoE−/− mice by modulating lipid metabolism. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.04.065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
26
|
Comparative antitumor and anti-inflammatory effects of flavonoids, saponins, polysaccharides, essential oil, coumarin and alkaloids from Cirsium japonicum DC. Food Chem Toxicol 2019; 125:422-429. [DOI: 10.1016/j.fct.2019.01.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/13/2019] [Accepted: 01/20/2019] [Indexed: 11/18/2022]
|
27
|
Semi-synthesis and anti-tumor activity of novel 25-OCH3-PPD derivatives incorporating aromatic moiety. Bioorg Med Chem Lett 2019; 29:189-193. [DOI: 10.1016/j.bmcl.2018.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/19/2018] [Accepted: 12/03/2018] [Indexed: 12/16/2022]
|
28
|
Kim HG, Kim MY, Cho JY. Alisma canaliculatum ethanol extract suppresses inflammatory responses in LPS-stimulated macrophages, HCl/EtOH-induced gastritis, and DSS-triggered colitis by targeting Src/Syk and TAK1 activities. JOURNAL OF ETHNOPHARMACOLOGY 2018; 219:202-212. [PMID: 29574093 DOI: 10.1016/j.jep.2018.03.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 03/14/2018] [Accepted: 03/16/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alisma canaliculatum A.Braun & C.D.Bouché, distributed in Korea, Japan, China, and Taiwan, is a traditional medicine. In particular, the stem and root of Alisma canaliculatum A.Braun & C.D.Bouché are prescribed to relieve various inflammatory symptoms resulting from nephritis, cystitis, urethritis, and dropsy. AIM OF STUDY However, the curative mechanism of Alisma canaliculatum A.Braun & C.D.Bouché with respect to inflammatory symptoms is poorly understood. In this study, the curative roles of this plant in various inflammatory conditions as well as its inhibitory mechanism were aimed to examine using an ethanol extract (Ac-EE). MATERIALS AND METHODS Anti-inflammatory effects of Ac-EE were evaluated in lipopolysaccharide (LPS)-induced macrophages in vitro and HCl/EtOH-stimulated mouse model of gastritis and DSS-treated mouse model of colitis. To determine the potentially active anti-inflammatory components in this extracts, we employed HPLC. We also used kinase assays, reporter gene assay, immunoprecipitation analysis and target enzyme overexpressing cell analysis to analyze the molecular mechanisms and the target molecules. RESULTS This extract dose-dependently inhibited the production of nitric oxide (NO) and prostaglandin E2 (PGE2) from RAW264.7 cells and peritoneal macrophages activated by lipopolysaccharide (LPS). Additionally, Ac-EE ameliorated inflammatory symptoms resulting from gastritis and colitis. Ac-EE down-regulated the mRNA levels of inducible NO synthase (iNOS), tumor necrosis factor (TNF)-α, and cyclooxygenase-2 (COX-2). Ac-EE also blocked the nuclear translocation of nuclear factor (NF)-κB and activator protein (AP)- 1 in LPS-stimulated RAW264.7 cells. By analyzing the target signaling molecules activating these transcription factors, we found that Src and Syk, as well as molecular association between TAK1 and mitogen-activated protein kinase kinase 4/7 (MKK4/7), were targeted by Ac-EE. CONCLUSIONS Our data suggest that the Ac-EE NF-κB/AP-1-targeted anti-inflammatory potential is mediated by suppression of Src and Syk as well as the complex formation between TAK1 and its substrate proteins MKK4/7.
Collapse
Affiliation(s)
- Han Gyung Kim
- Department of Genetic Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongs il University, Seoul 06978, Republic of Korea.
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea.
| |
Collapse
|
29
|
Bai L, Gao J, Wei F, Zhao J, Wang D, Wei J. Therapeutic Potential of Ginsenosides as an Adjuvant Treatment for Diabetes. Front Pharmacol 2018; 9:423. [PMID: 29765322 PMCID: PMC5938666 DOI: 10.3389/fphar.2018.00423] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/11/2018] [Indexed: 12/14/2022] Open
Abstract
Ginseng, one of the oldest traditional Chinese medicinal herbs, has been used widely in China and Asia for thousands of years. Ginsenosides extracted from ginseng, which is derived from the roots and rhizomes of Panax ginseng C. A. Meyer, have been used in China as an adjuvant in the treatment of diabetes mellitus. Owing to the technical complexity of ginsenoside production, the total ginsenosides are generally extracted. Accumulating evidence has shown that ginsenosides exert antidiabetic effects. In vivo and in vitro tests revealed the potential of ginsenoside Rg1, Rg3, Rg5, Rb1, Rb2, Rb3, compound K, Rk1, Re, ginseng total saponins, malonyl ginsenosides, Rd, Rh2, F2, protopanaxadiol (PPD) and protopanaxatriol (PPT)-type saponins to treat diabetes and its complications, including type 1 diabetes mellitus, type 2 diabetes mellitus, diabetic nephropathy, diabetic cognitive dysfunction, type 2 diabetes mellitus with fatty liver disease, diabetic cerebral infarction, diabetic cardiomyopathy, and diabetic erectile dysfunction. Many effects are attributed to ginsenosides, including gluconeogenesis reduction, improvement of insulin resistance, glucose transport, insulinotropic action, islet cell protection, hepatoprotective activity, anti-inflammatory effect, myocardial protection, lipid regulation, improvement of glucose tolerance, antioxidation, improvement of erectile dysfunction, regulation of gut flora metabolism, neuroprotection, anti-angiopathy, anti-neurotoxic effects, immunosuppression, and renoprotection effect. The molecular targets of these effects mainly contains GLUTs, SGLT1, GLP-1, FoxO1, TNF-α, IL-6, caspase-3, bcl-2, MDA, SOD, STAT5-PPAR gamma pathway, PI3K/Akt pathway, AMPK-JNK pathway, NF-κB pathway, and endoplasmic reticulum stress. Rg1, Rg3, Rb1, and compound K demonstrated the most promising therapeutic prospects as potential adjuvant medicines for the treatment of diabetes. This paper highlights the underlying pharmacological mechanisms of the anti-diabetic effects of ginsenosides.
Collapse
Affiliation(s)
- Litao Bai
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jialiang Gao
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fan Wei
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Zhao
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Danwei Wang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junping Wei
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
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
|
33
|
Tabetri™ ( Tabebuia avellanedae Ethanol Extract) Ameliorates Osteoarthritis Symptoms Induced by Monoiodoacetate through Its Anti-Inflammatory and Chondroprotective Activities. Mediators Inflamm 2017; 2017:3619879. [PMID: 29317792 PMCID: PMC5727801 DOI: 10.1155/2017/3619879] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/10/2017] [Indexed: 02/02/2023] Open
Abstract
Although osteoarthritis (OA), a degenerative joint disease characterized by the degradation of joint articular cartilage and subchondral bones, is generally regarded as a degenerative rather than inflammatory disease, recent studies have indicated the involvement of inflammation in OA pathogenesis. Tabebuia avellanedae has long been used to treat various diseases; however, its role in inflammatory response and the underlying molecular mechanisms remain poorly understood. In this study, the pharmacological effects of Tabetri (Tabebuia avellanedae ethanol extract (Ta-EE)) on OA pathogenesis induced by monoiodoacetate (MIA) and the underlying mechanisms were investigated using experiments with a rat model and in vitro cellular models. In the animal model, Ta-EE significantly ameliorated OA symptoms and reduced the serum levels of inflammatory mediators and proinflammatory cytokines without any toxicity. The anti-inflammatory activity of Ta-EE was further confirmed in a macrophage-like cell line (RAW264.7). Ta-EE dramatically suppressed the production and mRNA expressions of inflammatory mediators and proinflammatory cytokines in lipopolysaccharide-stimulated RAW264.7 cells without any cytotoxicity. Finally, the chondroprotective effect of Ta-EE was examined in a chondrosarcoma cell line (SW1353). Ta-EE markedly suppressed the mRNA expression of matrix metalloproteinase genes. The anti-inflammatory and chondroprotective activities of Ta-EE were attributed to the targeting of the nuclear factor-kappa B (NF-κB) and activator protein-1 (AP-1) signaling pathways in macrophages and chondrocytes.
Collapse
|
34
|
Akanda MR, Park BY. Involvement of MAPK/NF-κB signal transduction pathways: Camellia japonica mitigates inflammation and gastric ulcer. Biomed Pharmacother 2017; 95:1139-1146. [PMID: 28926923 DOI: 10.1016/j.biopha.2017.09.031] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/07/2017] [Accepted: 09/08/2017] [Indexed: 12/25/2022] Open
|
35
|
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
|
36
|
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
|
37
|
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
|
38
|
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
|
39
|
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: 11] [Impact Index Per Article: 1.4] [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
|
40
|
Han SY, Kim J, Kim E, Kim SH, Seo DB, Kim JH, Shin SS, Cho JY. AKT-targeted anti-inflammatory activity of Panax ginseng calyx ethanolic extract. J Ginseng Res 2017; 42:496-503. [PMID: 30337810 PMCID: PMC6187086 DOI: 10.1016/j.jgr.2017.06.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/14/2017] [Accepted: 06/20/2017] [Indexed: 01/22/2023] Open
Abstract
Background Korean ginseng (Panax ginseng) plays an anti-inflammatory role in a variety of inflammatory diseases such as gastritis, hepatitis, and colitis. However, inflammation-regulatory activity of the calyx of the P. ginseng berry has not been thoroughly evaluated. To understand whether the calyx portion of the P. ginseng berry is able to ameliorate inflammatory processes, an ethanolic extract of P. ginseng berry calyx (Pg-C-EE) was prepared, and lipopolysaccharide-activated macrophages and HEK293 cells transfected with inflammation-regulatory proteins were used to test the anti-inflammatory action of Pg-C-EE. Methods The ginsenoside contents of Pg-C-EE were analyzed by HPLC. Suppressive activity of Pg-C-EE on NO production, inflammatory gene expression, transcriptional activation, and inflammation signaling events were examined using the Griess assay, reverse transcription-polymerization chain reaction, luciferase activity reporter gene assay, and immunoblotting analysis. Results Pg-C-EE reduced NO production and diminished mRNA expression of inflammatory genes such as cyclooxygenase-2, inducible NO synthase, and tumor necrosis factor-α in a dose-dependent manner. This extract suppressed luciferase activity induced only by nuclear factor-κB. Interestingly, immunoblotting analysis results demonstrated that Pg-C-EE reduced the activities of protein kinase B (AKT)1 and AKT2. Conclusion These results suggest that Pg-C-EE may have nuclear-factor-κB-targeted anti-inflammatory properties through suppression of AKT. The calyx of the P. ginseng berry is an underused part of the ginseng plant, and development of calyx-derived extracts may be useful for treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Sang Yun Han
- Department of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Juewon Kim
- Vital Beautie Research Division, Amorepacific Research and Development Center, Suwon, Republic of Korea
| | - Eunji Kim
- Department of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Su Hwan Kim
- Vital Beautie Research Division, Amorepacific Research and Development Center, Suwon, Republic of Korea
| | - Dae Bang Seo
- Vital Beautie Research Division, Amorepacific Research and Development Center, Suwon, Republic of Korea
| | - Jong-Hoon Kim
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
| | - Song Seok Shin
- Vital Beautie Research Division, Amorepacific Research and Development Center, Suwon, Republic of Korea
| | - Jae Youl Cho
- Department of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
41
|
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
|
42
|
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
|
43
|
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
|
44
|
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
|
45
|
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
|
46
|
Suh W, Nam G, Yang WS, Sung GH, Shim SH, Cho JY. Chemical Constituents Identified from Fruit Body of Cordyceps bassiana and Their Anti-Inflammatory Activity. Biomol Ther (Seoul) 2017; 25:165-170. [PMID: 27530115 PMCID: PMC5340541 DOI: 10.4062/biomolther.2016.063] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/05/2016] [Accepted: 06/20/2016] [Indexed: 11/05/2022] Open
Abstract
Cordyceps bassiana is one of Cordyceps species with anti-oxidative, anti-cancer, anti-inflammatory, anti-diabetic, anti-obesity, anti-angiogenic, and anti-nociceptive activities. This mushroom has recently demonstrated to have an ability to reduce 2,4-dinitrofluorobenzene-induced atopic dermatitis symptoms in NC/Nga mice. In this study, we further examined phytochemical properties of this mushroom by column chromatography and HPLC analysis. By chromatographic separation and spectroscopic analysis, 8 compounds, such as 1,9-dimethylguanine (1), adenosine (2), uridine (3), nicotinamide (4), 3-methyluracil (5), 1,7-dimethylxanthine (6), nudifloric acid (7), and mannitol (8) were identified from 6 different fractions and 4 more subfractions. Through evaluation of their anti-inflammatory activities using reporter gene assay and mRNA analysis, compound 1 was found to block luciferase activity induced by NF-κB and AP-1, suppress the mRNA levels of cyclooxygenase (COX)-2 and tumor necrosis factor (TNF)-α. Therefore, our data strongly suggests that compound 1 acts as one of major principles in Cordyceps bassiana with anti-inflammatory and anti-atopic dermatitis activities.
Collapse
Affiliation(s)
- Wonse Suh
- School of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Gyeongsug Nam
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Woo Seok Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, 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
| | - Sang Hee Shim
- College of Pharmacy, Duksung Women's University, Seoul 01369, Republic of Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
47
|
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
|
48
|
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
|
49
|
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
|
50
|
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
|