1
|
Wang F, Liu L, Zhu Z, Aisa HA, Xin X. Anti-inflammatory effect and mechanism of active parts of Artemisia mongolica in LPS-induced Raw264.7 cells based on network pharmacology analysis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117509. [PMID: 38030026 DOI: 10.1016/j.jep.2023.117509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/14/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Artemisia mongolica is well known for its use in folk medicine, it is commonly used to alleviate a variety of diseases associated with inflammation, such as laryngitis, tonsillitis, headaches and hepatitis in northwest China. However, its anti-inflammatory mechanism is still unknown. AIM OF THE STUDY The most potential anti-inflammatory part (AMPA) was identified by screening individual parts of A. Mongolica. After the network pharmacological analysis, the anti-inflammation effects and molecular mechanisms of AMPA were evaluated in RAW264.7 cells induced by LPS. MATERIALS AND METHODS AMPA was chosen as the most anti-inflammatory of the A. Mongolica, as measured by the effect of each part of the A. Mongolica on NO and COX-2. The chemical composition of AMPA was identified using HPLC-Q-TOF-MS/MS, and targets of bioactive chemicals and targets related to inflammation were found using open-source databases. The "Compound-targets" network and PPI network were established by combining compounds and overlapped targets, and targets in the PPI networks were analyzed by GO and KEGG enrichment. The RAW26.7 cells induced by LPS were used as a model of inflammation examination. MTT assay was performed to assess the cytotoxicity of AMPA on LPS-induced RAW264.7 cells. The level of NO was measured by the Griess method while the inflammatory factors were detected by ELISA. The protein expression levels of iNOS, COX-2, MAPK, NF-κB signaling pathway and AMPK/Nrf2-related proteins were determined by Western blot. The results of nuclear translocation of p65 and Nrf2 were analyzed by immunofluorescence assay. RESULTS A total of 18 compounds with potential bioactivity were identified, and after intersecting 640 compound-predicted targets and 1608 inflammation targets, the compounds and intersected targets were utilized to structure "compound-target" and PPI networks. Among AMPA, AM6, AM7, AM11, AM8 and AM1 compounds were essential in the "compound-targets" network, meanwhile, TNF, RELA, MAPK1, NOS2, PRKAG, and PTGS2 targets play important roles in the PPI network. The top 10 terms and pathways were obtained based on GO and KEGG. The cell experiments show that 50 μg/mL was the maximum concentration of AMPA without cytotoxicity in the LPS-induced RAW264.7 cell model. When compared with the LPS group, AMPA treatment not only effectively suppressed the generation of NO, TNF-α, IL-6, PGE2, IL-1β and MCP-1 in LPS-induced RAW264.7 cells, but also down-regulated the expression of COX-2, iNOS and the protein levels p-ERK, p-p38, p-IκB-α and p-p65, inhibited the nuclear translocation of p65. Furthermore, the expression levels of p-LKB1, p-AMPK, Nrf2 and HO-1 proteins were up-regulated and Nrf2 nuclear translocation was promoted. CONCLUSION AMPA should be considered an anti-inflammatory agent for the results of network pharmacology and in vitro, which could inhibit the MAPK pathway and NF-κB pathway and activate the AMPK/Nrf2 pathway in LPS-stimulated RAW264.7 cells.
Collapse
Affiliation(s)
- Fangsheng Wang
- The State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China; University of Chinese Academy of Sciences, Beijing 100039, China
| | - Liu Liu
- The State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| | - ZiWei Zhu
- The State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China; University of Chinese Academy of Sciences, Beijing 100039, China
| | - Haji Akber Aisa
- The State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China.
| | - Xuelei Xin
- The State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China.
| |
Collapse
|
2
|
Lee MS, Shim HJ, Cho YY, Lee JY, Kang HC, Song IS, Lee HS. Comparative metabolism of aschantin in human and animal hepatocytes. Arch Pharm Res 2024; 47:111-126. [PMID: 38182943 DOI: 10.1007/s12272-023-01483-w] [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: 11/01/2023] [Accepted: 12/25/2023] [Indexed: 01/07/2024]
Abstract
Aschantin, a tetrahydrofurofuran lignan with a 1,3-benzodioxole group derived from Flos Magnoliae, exhibits antioxidant, anti-inflammatory, cytotoxic, and antimicrobial activities. This study compared the metabolic profiles of aschantin in human, dog, mouse, and rat hepatocytes using liquid chromatography-high-resolution mass spectrometry. The hepatic extraction ratio of aschantin among the four species was 0.46-0.77, suggesting that it undergoes a moderate-to-extensive degree of hepatic metabolism. Hepatocyte incubation of aschantin produced 4 phase 1 metabolites, including aschantin catechol (M1), O-desmethylaschantin (M2 and M3), and hydroxyaschantin (M4), and 14 phase 2 metabolites, including O-methyl-M1 (M5 and M6) via catechol O-methyltransferase (COMT), six glucuronides of M1, M2, M3, M5, and M6, and six sulfates of M1, M2, M3, M5, and M6. Enzyme kinetic studies using aschantin revealed that the production of M1, a major metabolite, via O-demethylenation is catalyzed by cytochrome 2C8 (CYP2C8), CYP2C9, CYP2C19, CYP3A4, and CYP3A5 enzymes; the formation of M2 (O-desmethylaschantin) is catalyzed by CYP2C9 and CYP2C19; and the formation of M4 is catalyzed by CYP3A4 enzyme. Two glutathione (GSH) conjugates of M1 were identified after incubation of aschantin with human and animal liver microsomes in the presence of nicotinamide adenine dinucleotide phosphate and GSH, but they were not detected in the hepatocytes of all species. In conclusion, aschantin is extensively metabolized, producing 18 metabolites in human and animal hepatocytes catalyzed by CYP, COMT, UDP-glucuronosyltransferase, and sulfotransferase. These results can help in clarifying the involvement of metabolizing enzymes in the pharmacokinetics and drug interactions of aschantin and in elucidating GSH conjugation associated with the reactive intermediate formed from M1 (aschantin catechol).
Collapse
Affiliation(s)
- Min Seo Lee
- College of Pharmacy and BK21 Four-Sponsored Advanced Program for SmartPharma Leaders, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Hyun Joo Shim
- College of Pharmacy, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Yong-Yeon Cho
- College of Pharmacy and BK21 Four-Sponsored Advanced Program for SmartPharma Leaders, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Joo Young Lee
- College of Pharmacy and BK21 Four-Sponsored Advanced Program for SmartPharma Leaders, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Han Chang Kang
- College of Pharmacy and BK21 Four-Sponsored Advanced Program for SmartPharma Leaders, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Im-Sook Song
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Hye Suk Lee
- College of Pharmacy and BK21 Four-Sponsored Advanced Program for SmartPharma Leaders, The Catholic University of Korea, Bucheon, 14662, Republic of Korea.
| |
Collapse
|
3
|
Patel K, Patel DK. Biological Potential and Therapeutic Effectiveness of Phytoproduct 'Fargesin' in Medicine: Focus on the Potential of an Active Phytochemical of Magnolia fargesii. RECENT ADVANCES IN INFLAMMATION & ALLERGY DRUG DISCOVERY 2024; 18:79-89. [PMID: 38726781 DOI: 10.2174/0127722708286664240429093913] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/23/2024] [Accepted: 04/02/2024] [Indexed: 10/16/2024]
Abstract
Flos Magnoliae is one of the important medicinal plants in different traditional medicine, including Chinese herbal medicine. Lignans and neolignans, including tetrahydrofurofuran, tetrahydrofuran, and aryltetralin, are present in the Flos Magnoliae species. A wide range of pharmacological activity of Flos Magnoliae has been reported in medicine. Fargesin has been isolated from Magnolia fargesii and it is a lignan-class phytochemical. Fargesin has numerous pharmacological activities in medicine, including its effectiveness on lipid and glucose metabolism, oxidative stress, myocardial apoptosis, etc. In the present work, we have summarized the detailed scientific information of fargesin concerning its medicinal properties and pharmacological activities. Numerous biological and chemical aspects of fargesin are discussed here, including the detailed pharmacological activities and analytical aspects of fargesin. In this review, we have also compiled analytical data on fargesin based on available scientific literature. Ethnopharmacological information on fargesin was gathered by a literature survey on PubMed, Science Direct, Google, and Scopus using the terms fargesin, Flos Magnoliae, phytochemical, and herbal medicine. The present review paper compiled the scientific data on fargesin in medicine for its pharmacological activities and analytical aspects in a very concise manner with proper citations. The present work signified the biological importance of fargesin in medicine due to its significant impact on bone disorders, lung injury, colon cancer, atherosclerosis, neurological disorders, ischemia, sars-cov-2, allergy, lipid and glucose metabolism, melanin synthesis, and different classes of enzymes. Furthermore, fargesin also has anti-inflammatory, antihypertensive, antiprotozoal, antimycobacterial, and antifeedant activity. However, analytical methods used for the separation, identification and isolation of fargesin in different biological and non-biological samples were also covered in the present review. The present work revealed the pharmacological activities and analytical aspects of fargesin in medicine and other allied health sectors.
Collapse
Affiliation(s)
- Kanika Patel
- Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, 211007, Uttar Pradesh, India
| | - Dinesh Kumar Patel
- Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, 211007, Uttar Pradesh, India
| |
Collapse
|
4
|
Lee MS, Park EJ, Cho YY, Lee JY, Kang HC, Lee HS. Comparative metabolism of fargesin in human, dog, monkey, mouse, and rat hepatocytes. Toxicol Res 2024; 40:125-137. [PMID: 38223669 PMCID: PMC10786765 DOI: 10.1007/s43188-023-00211-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 01/16/2024] Open
Abstract
Fargesin, a bioactive lignan derived from Flos Magnoliae, possesses anti-inflammatory, anti-oxidative, anti-melanogenic, and anti-apoptotic effects. This study compared the metabolic profiles of fargesin in human, dog, monkey, mouse, and rat hepatocytes using liquid chromatography-high resolution mass spectrometry. In addition, we investigated the human cytochrome P450 (CYP), UDP-glucuronosyltransferase (UGT), and sulfotransferase (SULT) enzymes responsible for fargesin metabolism. The hepatic extraction ratio of fargesin among the five species ranged from 0.59 to 0.78, suggesting that it undergoes a moderate-to-extensive degree of hepatic metabolism. During metabolism, fargesin generates three phase 1 metabolites, including fargesin catechol (M1) and O-desmethylfargesin (M2 and M3), and 11 phase 2 metabolites, including O-methyl-M1 (M4 and M5) via catechol O-methyltransferase (COMT), glucuronides of M1, M2, M4, and M5, and sulfates of M1-M5. The production of M1 from fargesin via O-demethylenation is catalyzed by CYP2C9, CYP3A4, CYP2C19, and CYP2C8 enzymes, whereas the formation of M2 and M3 (O-desmethylfargesin) is catalyzed by CYP2C9, CYP2B6, CYP2C19, CYP3A4, CYP1A2, and CYP2D6 enzymes. M4 is metabolized to M4 glucuronide by UGT1A3, UGT1A8, UGT1A10, UGT2B15, and UGT2B17 enzymes, whereas M4 sulfate is generated by multiple SULT enzymes. Fargesin is extensively metabolized in human hepatocytes by CYP, COMT, UGT, and SULT enzymes. These findings help to elucidate the pharmacokinetics and drug interactions of fargesin.
Collapse
Affiliation(s)
- Min Seo Lee
- College of Pharmacy and BK21 Four-sponsored Advanced Program for SmartPharma Leaders, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Eun Jeong Park
- College of Pharmacy and BK21 Four-sponsored Advanced Program for SmartPharma Leaders, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Yong-Yeon Cho
- College of Pharmacy and BK21 Four-sponsored Advanced Program for SmartPharma Leaders, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Joo Young Lee
- College of Pharmacy and BK21 Four-sponsored Advanced Program for SmartPharma Leaders, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Han Chang Kang
- College of Pharmacy and BK21 Four-sponsored Advanced Program for SmartPharma Leaders, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Hye Suk Lee
- College of Pharmacy and BK21 Four-sponsored Advanced Program for SmartPharma Leaders, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| |
Collapse
|
5
|
Gil TY, Jin BR, Park YJ, Kim HM, An HJ. Effects of Magnoliae Flos on Atopic Dermatitis-Like Inflammation Evaluated via Extracellular Signal-regulated Kinase or Signal Transducers and Activators of Transcription 1/3 Signalling Pathways. Acta Derm Venereol 2023; 103:adv11593. [PMID: 37955529 PMCID: PMC10655128 DOI: 10.2340/actadv.v103.11593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 09/12/2023] [Indexed: 11/14/2023] Open
Abstract
Atopic dermatitis is a chronic inflammatory skin disease. Skin is the largest organ and plays a pivotal role in protecting the body. Not only does the skin act as a physical barrier against the external environment, but it also has its own immune system. Atopic dermatitis is caused by prolonged excessive inflammatory responses that worsen under imbalanced cutaneous immune system skin conditions. Although the prevalence and burden of atopic dermatitis is increasing, the standard therapeutic agents remain unclear due to the complicated pathophysiology of the condition. The objective of this study is to examine the use of Magnoliae flos, the dried flower bud of Magnolia biondii or related plants. The effects and underlying mechanism of action of aqueous extract of the buds of Magnoliae flos (MF) were evaluated. Immortalized human keratinocytes (HaCaT) stimulated with tumour necrosis factor-α and interferon-γ mixture and NC/Nga mice stimulated with 2,4-dinitrochlorobenzene were used as atopic dermatitis models, in vitro and in vivo, respectively. The effects of MF were determined by measuring the suppression of pro-inflammatory signalling pathways, such as extracellular signal-regulated kinase or signal transducers and activators of transcription 1/3 and restoring skin barrier molecules. In conclusion, MF is a potential therapeutic alternative for the treatment of atopic dermatitis through repressing inflammatory pathways.
Collapse
Affiliation(s)
- Tae-Young Gil
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Dongdaemun-gu, Seoul, Korea
| | - Bo-Ram Jin
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Dongdaemun-gu, Seoul, Korea
| | - Yea-Jin Park
- Department of Pharmacology, College of Korean Medicine, Sangji University, Wonju-si, Gangwon-do, Korea
| | - Hye-Min Kim
- Department of Pharmacology, College of Korean Medicine, Sangji University, Wonju-si, Gangwon-do, Korea
| | - Hyo-Jin An
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Dongdaemun-gu, Seoul, Korea.
| |
Collapse
|
6
|
Ding XY, Wen JR, Lin WY, Huang GY, Feng Q, Duan L, Zhang SJ, Liu Z, Zhang RR, Wang Y. Phloroglucinol derivatives, coumarins and an alkaloid from the roots of Evodia lepta Merr. PHYTOCHEMISTRY 2023:113774. [PMID: 37400011 DOI: 10.1016/j.phytochem.2023.113774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 07/05/2023]
Abstract
Two previously undescribed phloroglucinol derivatives [(±) evolephloroglucinols A and B], five unusual coumarins [evolecoumarins A and B and (±) evolecoumarins C-E], and one novel enantiomeric quinoline-type alkaloid [(±) evolealkaloid A], along with 20 known compounds, were isolated from the EtOH extract of the roots of Evodia lepta Merr. Their structures were elucidated by extensive spectroscopic analyses. The absolute configurations of the undescribed compounds were determined by X-ray diffraction or computational calculations. Their anti-neuroinflammatory effects were assayed. Among the identified compounds, compound 5a effectively reduced nitric oxide (NO) production with an EC50 value of 22.08 ± 0.46 μM. Hence, it could indeed inhibit the lipopolysaccharide (LPS)-induced Nod-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome.
Collapse
Affiliation(s)
- Xiao-Ying Ding
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jun-Ru Wen
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, People's Republic of China
| | - Wei-Yao Lin
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guo-Yong Huang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qian Feng
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lixin Duan
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shi-Jie Zhang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, People's Republic of China
| | - Zhongqiu Liu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Rong-Rong Zhang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Ying Wang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
7
|
Fargesin Inhibits EGF-Induced Cell Transformation and Colon Cancer Cell Growth by Suppression of CDK2/Cyclin E Signaling Pathway. Int J Mol Sci 2021; 22:ijms22042073. [PMID: 33669811 PMCID: PMC7922630 DOI: 10.3390/ijms22042073] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/05/2021] [Accepted: 02/16/2021] [Indexed: 01/15/2023] Open
Abstract
Although the lignan compound fargesin is a major ingredient in Shin-Yi, the roles of fargesin in carcinogenesis and cancer cell growth have not been elucidated. In this study, we observed that fargesin inhibited cell proliferation and transformation by suppression of epidermal growth factor (EGF)-stimulated G1/S-phase cell cycle transition in premalignant JB6 Cl41 and HaCaT cells. Unexpectedly, we found that signaling pathway analyses showed different regulation patterns in which fargesin inhibited phosphatidylinositol 3-kinase/AKT signaling without an alteration of or increase in mitogen activated protein kinase (MAPK) in JB6 Cl41 and HaCaT cells, while both signaling pathways were abrogated by fargesin treatment in colon cancer cells. We further found that fargesin-induced colony growth inhibition of colon cancer cells was mediated by suppression of the cyclin dependent kinase 2 (CDK2)/cyclin E signaling axis by upregulation of p21WAF1/Cip1, resulting in G1-phase cell cycle accumulation in a dose-dependent manner. Simultaneously, the suppression of CDK2/cyclin E and induction of p21WAF1/Cip1 were correlated with Rb phosphorylation and c-Myc suppression. Taken together, we conclude that fargesin-mediated c-Myc suppression inhibits EGF-induced cell transformation and colon cancer cell colony growth by the suppression of retinoblastoma (Rb)-E2F and CDK/cyclin signaling pathways, which are mainly regulated by MAPK and PKB signaling pathways.
Collapse
|
8
|
Tetrahydrofurofuranoid Lignans, Eudesmin, Fargesin, Epimagnolin A, Magnolin, and Yangambin Inhibit UDP-Glucuronosyltransferase 1A1 and 1A3 Activities in Human Liver Microsomes. Pharmaceutics 2021; 13:pharmaceutics13020187. [PMID: 33535454 PMCID: PMC7912740 DOI: 10.3390/pharmaceutics13020187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 11/17/2022] Open
Abstract
Eudesmin, fargesin, epimagnolin A, magnolin, and yangambin are tetrahydrofurofuranoid lignans with various pharmacological activities found in Magnoliae Flos. The inhibition potencies of eudesmin, fargesin, epimagnolin A, magnolin, and yangambin on six major human uridine 5'-diphospho-glucuronosyltransferase (UGT) activities in human liver microsomes were evaluated using liquid chromatography-tandem mass spectrometry and cocktail substrates. Eudesmin, fargesin, epimagnolin A, magnolin, and yangambin inhibited UGT1A1 and UGT1A3 activities, but showed negligible inhibition of UGT1A4, UGT16, UGT1A9, and UGT2B7 activities at 200 μM in pooled human liver microsomes. Moreover, eudesmin, fargesin, epimagnolin A, magnolin, and yangambin noncompetitively inhibited UGT1A1-catalyzed SN38 glucuronidation with Ki values of 25.7, 25.3, 3.6, 26.0, and 17.1 μM, respectively, based on kinetic analysis of UGT1A1 inhibition in pooled human liver microsomes. Conversely, the aforementioned tetrahydrofurofuranoid lignans competitively inhibited UGT1A3-catalyzed chenodeoxycholic acid 24-acyl-glucuronidation with 39.8, 24.3, 15.1, 37.6, and 66.8 μM, respectively in pooled human liver microsomes. These in vitro results suggest the necessity of evaluating whether the five tetrahydrofurofuranoid lignans can cause drug-drug interactions with UGT1A1 and UGT1A3 substrates in vivo.
Collapse
|
9
|
Hajra S, Garai S, Sen B. Chromatography-free “two-pots” asymmetric total synthesis of (+)-sesamin and (+)-aschantin. Tetrahedron 2020. [DOI: 10.1016/j.tet.2020.131483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
10
|
Neuroprotective effects of lignan 7-hydroxymatairesinol (HMR/lignan) in a rodent model of Parkinson's disease. Nutrition 2020; 69:110494. [DOI: 10.1016/j.nut.2019.04.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/24/2019] [Accepted: 04/01/2019] [Indexed: 01/01/2023]
|
11
|
Xin H, Cui Y, An Z, Yang Q, Zou X, Yu N. Attenuated glutamate induced ROS production by antioxidative compounds in neural cell lines. RSC Adv 2019; 9:34735-34743. [PMID: 35530670 PMCID: PMC9074000 DOI: 10.1039/c9ra03848e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/21/2019] [Indexed: 12/21/2022] Open
Abstract
Glutamate is an excitatory neurotransmitter involved in neural function. Excess accumulation of intercellular glutamate leads to increasing concentration of reactive oxygen species (ROS) and reactive nitrogen species (RNS) in neuronal cells. In this study, we investigated the antioxidant activity of several typical superior compounds among four neuronal cells, and determined the scavenging activity of free radicals. The in vivo assay was also carried out to compare the protective effect of glutamate-induced cell damage. Hierarchical clustering analysis was used to identify the common properties. Glutamate induced neurotoxicity and ROS production, suggesting glutamate cytotoxicity was related to oxidative stress and widely exists in different cell lines. Those screening compounds exhibited strong antioxidant ability, but low cytotoxicity to neuronal cells, acting as agents against neurodegenerative diseases. Finally, a hierarchical clustering analysis assay indicated that hyperoside and rutin hydrate are the most effective compounds for attenuating intercellular ROS levels. The results suggested the activity more or less relies on structure, rather than residues. These data generate new supporting ideas to remove intracellular ROS and the identified compounds serve as potential therapeutic agents in multiple neurological diseases. Glutamate is an excitatory neurotransmitter involved in neural function.![]()
Collapse
Affiliation(s)
- Haolin Xin
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases
- Department of Neurology
- Nankai University
- Huanhu Hospital
- Tianjin
| | - Ying Cui
- Tianjin University of Traditional Chinese Medicine
- Tianjin
- China
| | - Zhongping An
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases
- Department of Neurology
- Nankai University
- Huanhu Hospital
- Tianjin
| | - Qian Yang
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases
- Department of Neurology
- Nankai University
- Huanhu Hospital
- Tianjin
| | - Xuan Zou
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases
- Department of Neurology
- Nankai University
- Huanhu Hospital
- Tianjin
| | - Ning Yu
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases
- Department of Neurology
- Nankai University
- Huanhu Hospital
- Tianjin
| |
Collapse
|
12
|
García-Huertas P, Olmo F, Sánchez-Moreno M, Dominguez J, Chahboun R, Triana-Chávez O. Activity in vitro and in vivo against Trypanosoma cruzi of a furofuran lignan isolated from Piper jericoense. Exp Parasitol 2018; 189:34-42. [PMID: 29656102 DOI: 10.1016/j.exppara.2018.04.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 03/14/2018] [Accepted: 04/09/2018] [Indexed: 10/17/2022]
Abstract
Piperaceae species are abundant in the tropics and are important components of secondary vegetation. Many of these plants have received considerable attention due to their wide range of biological activities. Here, the trypanocidal activity of extracts and fractions with different polarities obtained from Colombian Piper jericoense plant was evaluated. A furofuran lignan, (1S,3aS,4S,6aS)-1-(3',4'-dimethoxyphenyl)-4-(3″,4″-methylendioxyphenyl)hexahydrofuro[3,4-c]furan, (1), was isolated from Colombian Piper jericoense leaves ethyl acetate extract. Its relative configuration at the stereogenic centers was established on the basis of various spectroscopic analyses, including 1D- (1H, 13C, and DEPT) and 2D-NMR (COSY, NOESY, HMQC and HMBC) and a 2D INADEQUATE NMR experiment as well as by comparison of their spectral data with those of related compounds such as (+)-Kobusin (2). The activity against Trypanosoma cruzi indicated that compound 1 was active against all parasite forms (epimastigote, amastigote and trypomastigote) and presented lower toxicity than the reference drug, benznidazole (Bz), evidenced by a selective index of 18.4 compared to that of Bz, which was 6.7. Moreover, this compound inhibited the infectious process, and it was active in infected mice in the acute phase. This compound significantly inhibited the T. cruzi Fe-SOD enzyme, whereas Cu/Zn-SOD from human cells was not affected. Ultrastructural analyses, together with metabolism-excretion studies in the parasite, were also performed to identify the possible mechanism of action of the tested compound. Interestingly, the lignan affected the parasite structure, but it did not alter the energetic metabolism.
Collapse
Affiliation(s)
- Paola García-Huertas
- Grupo Biología y Control de Enfermedades Infecciosas - BCEI, Universidad de Antioquia, Medellín, Colombia.
| | - Francisco Olmo
- Departamento de Parasitología, Instituto de Investigación Biosanitaria (IBS, Granada), Hospitales Universitarios de Granada, Universidad de Granada, Granada, Spain.
| | - Manuel Sánchez-Moreno
- Departamento de Parasitología, Instituto de Investigación Biosanitaria (IBS, Granada), Hospitales Universitarios de Granada, Universidad de Granada, Granada, Spain.
| | - Jorge Dominguez
- Grupo de Química Orgánica de Productos Naturales, Universidad de Antioquia, Medellín, Colombia.
| | - Rachid Chahboun
- Department of Organic Chemistry, Institute of Biotechnology, University of Granada, Avda. Fuentenueva, 18071, Granada, Spain.
| | - Omar Triana-Chávez
- Grupo Biología y Control de Enfermedades Infecciosas - BCEI, Universidad de Antioquia, Medellín, Colombia.
| |
Collapse
|
13
|
Kola PK, Akula A, NissankaraRao LS, Danduga RCSR. Protective effect of naringin on pentylenetetrazole (PTZ)-induced kindling; possible mechanisms of antikindling, memory improvement, and neuroprotection. Epilepsy Behav 2017; 75:114-126. [PMID: 28846920 DOI: 10.1016/j.yebeh.2017.07.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 07/07/2017] [Accepted: 07/09/2017] [Indexed: 12/14/2022]
Abstract
The present study investigated the effects of Naringin on seizure severity, progress of kindling, memory impairment, oxidative stress, neurochemicals, and neural damage in Pentylenetetrazole (PTZ)-induced kindling. Alternate intra-peritoneal injections of PTZ induced kindling at 22 injections of PTZ. In comparison with the PTZ group, pretreatment with Naringin 30 min prior to PTZ administration and on a PTZ-free day was found to lead to a decreased seizure score, a mitigated progress of kindling, decreased transfer latency, and increased total number of arm entries, % alternation behavior in Y maze, and % conditioned avoidance response in a pole climbing apparatus. Biochemical analysis of the frontal and temporal cortexes and the hippocampus of the brain showed that Naringin attenuated the level of lipid peroxidation (MDA) and augmented the reduced glutathione, superoxide dismutase, catalase, and total thiol results in decreased oxidative stress compared with the PTZ group and control group. Investigation of neurochemicals revealed a minute change in gamma amino butyric acid (GABA), glutamate and dopamine, and decreased AChE in the three regions. Increased CA1 neuronal density in the hippocampus and increased cell density in the frontal and temporal regions indicate the potential of naringin to act against PTZ-induced kindling, memory impairment, oxidative stress, neurochemical changes, and histological aberrations.
Collapse
Affiliation(s)
- Phani Kumar Kola
- University College of Pharmaceutical Sciences, Acharya Nagarjuna University, Guntur, Andhra Pradesh, India.
| | - Annapurna Akula
- University College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, Andhra Pradesh, India
| | | | - R Ch Sekhara Reddy Danduga
- University College of Pharmaceutical Sciences, Acharya Nagarjuna University, Guntur, Andhra Pradesh, India
| |
Collapse
|
14
|
Kim JH, Kwon SS, Jeong HU, Lee HS. Inhibitory Effects of Dimethyllirioresinol, Epimagnolin A, Eudesmin, Fargesin, and Magnolin on Cytochrome P450 Enzyme Activities in Human Liver Microsomes. Int J Mol Sci 2017; 18:ijms18050952. [PMID: 28468305 PMCID: PMC5454865 DOI: 10.3390/ijms18050952] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/25/2017] [Accepted: 04/27/2017] [Indexed: 12/13/2022] Open
Abstract
Magnolin, epimagnolin A, dimethyllirioresinol, eudesmin, and fargesin are pharmacologically active tetrahydrofurofuranoid lignans found in Flos Magnoliae. The inhibitory potentials of dimethyllirioresinol, epimagnolin A, eudesmin, fargesin, and magnolin on eight major human cytochrome P450 (CYP) enzyme activities in human liver microsomes were evaluated using liquid chromatography-tandem mass spectrometry to determine the inhibition mechanisms and inhibition potency. Fargesin inhibited CYP2C9-catalyzed diclofenac 4′-hydroxylation with a Ki value of 16.3 μM, and it exhibited mechanism-based inhibition of CYP2C19-catalyzed [S]-mephenytoin 4′-hydroxylation (Ki, 3.7 μM; kinact, 0.102 min−1), CYP2C8-catalyzed amodiaquine N-deethylation (Ki, 10.7 μM; kinact, 0.082 min−1), and CYP3A4-catalyzed midazolam 1′-hydroxylation (Ki, 23.0 μM; kinact, 0.050 min−1) in human liver microsomes. Fargesin negligibly inhibited CYP1A2-catalyzed phenacetin O-deethylation, CYP2A6-catalyzed coumarin 7-hydroxylation, CYP2B6-catalyzed bupropion hydroxylation, and CYP2D6-catalyzed bufuralol 1′-hydroxylation at 100 μM in human liver microsomes. Dimethyllirioresinol weakly inhibited CYP2C19 and CYP2C8 with IC50 values of 55.1 and 85.0 μM, respectively, without inhibition of CYP1A2, CYP2A6, CYP2B6, CYP2C9, CYP2D6, and CYP3A4 activities at 100 μM. Epimagnolin A, eudesmin, and magnolin showed no the reversible and time-dependent inhibition of eight major CYP activities at 100 μM in human liver microsomes. These in vitro results suggest that it is necessary to investigate the potentials of in vivo fargesin-drug interaction with CYP2C8, CYP2C9, CYP2C19, and CYP3A4 substrates.
Collapse
Affiliation(s)
- Ju-Hyun Kim
- Drug Metabolism and Bioanalysis Laboratory, College of Pharmacy, The Catholic University of Korea, Bucheon 420-743, Korea.
| | - Soon-Sang Kwon
- Drug Metabolism and Bioanalysis Laboratory, College of Pharmacy, The Catholic University of Korea, Bucheon 420-743, Korea.
| | - Hyeon-Uk Jeong
- Drug Metabolism and Bioanalysis Laboratory, College of Pharmacy, The Catholic University of Korea, Bucheon 420-743, Korea.
| | - Hye Suk Lee
- Drug Metabolism and Bioanalysis Laboratory, College of Pharmacy, The Catholic University of Korea, Bucheon 420-743, Korea.
| |
Collapse
|
15
|
Pham TH, Kim MS, Le MQ, Song YS, Bak Y, Ryu HW, Oh SR, Yoon DY. Fargesin exerts anti-inflammatory effects in THP-1 monocytes by suppressing PKC-dependent AP-1 and NF-ĸB signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 24:96-103. [PMID: 28160867 DOI: 10.1016/j.phymed.2016.11.014] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 10/24/2016] [Accepted: 11/19/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Fargesin is a lignan from Magnolia fargesii, an oriental medicine used in the treatment of nasal congestion and sinusitis. The anti-inflammatory properties of this compound have not been fully elucidated yet. PURPOSE This study focused on assessing the anti-inflammatory effects of fargesin on phorbal ester (PMA)-stimulated THP-1 human monocytes, and the molecular mechanisms underlying them. METHODS Cell viability was evaluated by MTS assay. Protein expression levels of inflammatory mediators were analyzed by Western blotting, ELISA, Immunofluorescence assay. mRNA levels were measured by Real-time PCR. Promoter activities were elucidated by Luciferase assay. RESULTS It was found that pre-treatment with fargesin attenuated significantly the expression of two major inflammatory mediators, cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS). Fargesin also inhibited the production of pro-inflammation cytokines (IL-1β, TNF-α) and chemokine (CCL-5). Besides, nuclear translocation of transcription factors nuclear factor-kappa B (NF-ĸB) and activator protein-1 (AP-1), which regulate multiple pro-inflammatory genes, was suppressed by fargesin in a PKC-dependent manner. Furthermore, among the mitogen-activated protein kinases (MAPKs), only c-Jun N-terminal kinase (JNK) was downregulated by fargesin in a PKC-dependent manner, and this reduction was involved in PMA-induced AP-1 and NF-ĸB nuclear translocation attenuation, demonstrated using a specific JNK inhibitor. CONCLUSION Taken together, our results found that fargesin exhibits anti-inflammation effects on THP-1 cells via suppression of PKC pathway including downstream JNK, nuclear factors AP-1 and NF-ĸB. These results suggest that fargesin has anti-inflammatory properties with potential applications in drug development against inflammatory disorders.
Collapse
Affiliation(s)
- Thu-Huyen Pham
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul 05029, Republic of Korea
| | - Man-Sub Kim
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul 05029, Republic of Korea
| | - Minh-Quan Le
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul 05029, Republic of Korea
| | - Yong-Seok Song
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul 05029, Republic of Korea
| | - Yesol Bak
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyung-Won Ryu
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gun, Chungbuk 28116, Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gun, Chungbuk 28116, Republic of Korea
| | - Do-Young Yoon
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
16
|
Huang Y, Zou X, Zhang X, Wang F, Zhu W, Zhang G, Xiao J, Chen M. Magnolin inhibits prostate cancer cell growth in vitro and in vivo. Biomed Pharmacother 2017; 87:714-720. [PMID: 28092770 DOI: 10.1016/j.biopha.2017.01.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/02/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Magnolin is the most active ingredient in the herb Magnolia fargesii, which has been traditionally used in oriental medicine to treat headaches and nasal congestion. Recent researches demonstrate that Magnolin inhibits cancer cell migration and invasion. MATERIALS AND METHODS This study used cell culture and the BALB/c nu/nu mouse xenograft model to investigate whether or not magnolin can inhibit the growth of PC3 and Du145 prostate cancer cells. MTT assay and flow cytometry were performed to estimate the proliferation, cycle, and apoptosis of the cells in vitro. Clone formation assay was also conducted. In the animal study, Ki-67 immunostaining and TUNEL assay were carried out to evaluate cell proliferation and apoptosis, respectively. To elucidate the possible mechanism by which magnolin attenuates prostate cancer cell growth, we estimated the expression levels of Akt/p-Akt, P53, P21, BCL-2, and cleaved Caspase3 by using Western blot 48h after magnolin-treatment of the cells. RESULTS Magnolin inhibited the proliferation and viability of the tumor cells by triggering cell cycle arrest via P53/P21 activation and inducing apoptosis in vitro and in vivo. Magnolin downregulated the phosphorylation of Akt protein kinase and upregulated cleaved Caspase3 during anti-proliferation and pro-apoptosis. CONCLUSION Magnolin may be a novel medicine for prostate cancer therapy.
Collapse
Affiliation(s)
- Yeqing Huang
- Department of Urology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiangyu Zou
- Department of Urology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaowen Zhang
- Department of Urology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Feng Wang
- Department of Nephrology and Rheumatology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Weidong Zhu
- Department of Urology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Guangyuan Zhang
- Department of Urology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| | - Jun Xiao
- Department of Urology, Anhui provincial hospital, Anhui Medical University, Hefei, China.
| | - Ming Chen
- Department of Urology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
17
|
Inhibitory Effects of Aschantin on Cytochrome P450 and Uridine 5'-diphospho-glucuronosyltransferase Enzyme Activities in Human Liver Microsomes. Molecules 2016; 21:molecules21050554. [PMID: 27128896 PMCID: PMC6273138 DOI: 10.3390/molecules21050554] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 04/22/2016] [Accepted: 04/23/2016] [Indexed: 01/13/2023] Open
Abstract
Aschantin is a bioactive neolignan found in Magnolia flos with antiplasmodial, Ca2+-antagonistic, platelet activating factor-antagonistic, and chemopreventive activities. We investigated its inhibitory effects on the activities of eight major human cytochrome P450 (CYP) and uridine 5′-diphospho-glucuronosyltransferase (UGT) enzymes of human liver microsomes to determine if mechanistic aschantin–enzyme interactions were evident. Aschantin potently inhibited CYP2C8-mediated amodiaquine N-de-ethylation, CYP2C9-mediated diclofenac 4′-hydroxylation, CYP2C19-mediated [S]-mephenytoin 4′-hydroxylation, and CYP3A4-mediated midazolam 1′-hydroxylation, with Ki values of 10.2, 3.7, 5.8, and 12.6 µM, respectively. Aschantin at 100 µM negligibly inhibited CYP1A2-mediated phenacetin O-de-ethylation, CYP2A6-mediated coumarin 7-hydroxylation, CYP2B6-mediated bupropion hydroxylation, and CYP2D6-mediated bufuralol 1′-hydroxylation. At 200 µM, it weakly inhibited UGT1A1-catalyzed SN-38 glucuronidation, UGT1A6-catalyzed N-acetylserotonin glucuronidation, and UGT1A9-catalyzed mycophenolic acid glucuronidation, with IC50 values of 131.7, 144.1, and 71.0 µM, respectively, but did not show inhibition against UGT1A3, UGT1A4, or UGT2B7 up to 200 µM. These in vitro results indicate that aschantin should be examined in terms of potential interactions with pharmacokinetic drugs in vivo. It exhibited potent mechanism-based inhibition of CYP2C8, CYP2C9, CYP2C19, and CYP3A4.
Collapse
|
18
|
Magnolin protects against contrast-induced nephropathy in rats via antioxidation and antiapoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:203458. [PMID: 25400863 PMCID: PMC4221873 DOI: 10.1155/2014/203458] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 10/01/2014] [Indexed: 12/17/2022]
Abstract
Background. Magnolin is the major active ingredient of the herb Magnolia fargesii which has anti-inflammatory and antioxidative effects. Oxidative stress and apoptosis are involved in the pathogenesis of contrast-induced nephropathy (CIN). We hypothesize that Magnolin could protect against CIN through antioxidative and antiapoptotic properties. Methods. To test whether Magnolin could attenuate CIN, oxidative stress and apoptosis, in vivo and in vitro, we utilized a rat model of ioversol-induced CIN and a cell model of oxidative stress in which HK2 cells were treated with H2O2. Rats were assigned to 4 groups (n = 6 per group): control group, ioversol group (ioversol-induced CIN), vehicle group (CIN rats pretreated with vehicle), and Magnolin group (CIN rats pretreated with 1 mg/kg Magnolin). Results. The results showed that magnolin ameliorated the renal tubular necrosis, apoptosis, and the deterioration of renal function (P < 0.05). Furthermore, Magnolin reduced the renal oxidative stress, suppressed caspase-3 activity, and increased Bcl-2 expression in vivo and in vitro. Conclusion. Magnolin might protect CIN in rats through antioxidation and antiapoptosis.
Collapse
|
19
|
Jung UJ, Kim SR. Effects of naringin, a flavanone glycoside in grapefruits and citrus fruits, on the nigrostriatal dopaminergic projection in the adult brain. Neural Regen Res 2014; 9:1514-7. [PMID: 25317167 PMCID: PMC4192967 DOI: 10.4103/1673-5374.139476] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2014] [Indexed: 11/30/2022] Open
Abstract
Recently, we have demonstrated the ability of naringin, a well-known flavanone glycoside of grapefruits and citrus fruits, to prevent neurodegeneration in a neurotoxin model of Parkinson's disease. Intraperitoneal injection of naringin protected the nigrostriatal dopaminergic projection by increasing glial cell line-derived neurotrophic factor expression and decreasing the level of tumor necrosis factor-alpha in dopaminergic neurons and microglia, respectively. These results suggest that naringin can impart to the adult dopaminergic neurons the ability to produce glial cell line-derived neurotrophic factor against Parkinson's disease with anti-inflammatory effects. Based on these results, we would like to describe an important perspective on its possibility as a therapeutic agent for Parkinson's disease.
Collapse
Affiliation(s)
- Un Ju Jung
- Center for Food and Nutritional Genomics Research, Kyungpook National University, Daegu, Korea
| | - Sang Ryong Kim
- School of Life Sciences, Kyungpook National University, Daegu, Korea ; BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea ; Institute of Life Science & Biotechnology, Kyungpook National University, Daegu, Korea ; Brain Science and Engineering Institute, Kyungpook National University, Daegu, Korea
| |
Collapse
|
20
|
Jun AY, Kim HJ, Park KK, Son KH, Lee DH, Woo MH, Chung WY. Tetrahydrofurofuran-type lignans inhibit breast cancer-mediated bone destruction by blocking the vicious cycle between cancer cells, osteoblasts and osteoclasts. Invest New Drugs 2013; 32:1-13. [PMID: 23673814 DOI: 10.1007/s10637-013-9969-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/29/2013] [Indexed: 10/26/2022]
Abstract
Breast cancer frequently spreads to bone. The interaction between bone metastases and microenvironment, referred as the "vicious cycle", increases both tumor burden and bone destruction. Therefore, inhibition at any point in this "vicious cycle" can reduce malignant osteolytic lesions in patients with advanced breast cancer. In this study, we evaluated whether tetrahydrofurofuran-type lignans derived from Magnoliae Flos, commonly used in traditional Asian medicine to treat inflammatory diseases, could block breast cancer-mediated bone loss. Aschatin, fargesin, lirioresinol B dimethyl ether, and magnolin at noncytotoxic concentrations suppressed mRNA expression and secretion of osteolytic factor PTHrP in MDA-MB-231 metastatic human breast cancer cells. Fargesin inhibited TGF-β-stimulated cell viability, migration, and invasion and decreased TGF-β-induced PTHrP production in MDA-MB-231 cells. In addition, these lignans reduced RANKL/OPG ratio in PTHrP-treated hFOB1.19 human osteoblastic cells and inhibited RANKL-mediated osteoclast differentiation in mouse bone marrow macrophages. Aschatin, fargesin, lirioresinol B dimethyl ether, and magnolin substantially reduced bone-resorbing activity of osteoclasts by inhibiting MMP-9 and cathepsin K activities. Furthermore, orally administered fargesin inhibited tumor growth and cancer-mediated bone destruction in mice with MDA-MB-231 cells injected into calvarial tissues. Aschatin, fargesin, lirioresinol B dimethyl ether, and magnolin blocked initiation and progression of the "vicious cycle" between breast cancer metastases and bone microenvironment by inhibiting PTHrP production in breast cancer cells and osteoclastic bone resorption. Therefore, these tetrahydrofurofuran-type lignans have the potential to serve as beneficial agents to prevent and treat cancer-induced bone destruction in breast cancer patients.
Collapse
Affiliation(s)
- Ah Young Jun
- Department of Applied Life Science, The Graduate School, Yonsei University, Seoul, 120-749, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
21
|
Fargesin, a component of Flos Magnoliae, stimulates glucose uptake in L6 myotubes. J Nat Med 2012; 67:320-6. [PMID: 22791412 DOI: 10.1007/s11418-012-0685-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 06/20/2012] [Indexed: 12/31/2022]
Abstract
Flos Magnoliae (FM) is a commonly used Chinese medicinal herb for symptomatic relief of allergic rhinitis, sinusitis and headache. Although several FM species have been used as substitutes or adulterants for clinical use, possible differences in their pharmacological actions have not been reported. To confirm the effects of FM on skeletal muscle glucose metabolism, we tested the effects of several compounds isolated from FM on glucose uptake by L6 myotubes. We found that fargesin, a component of FM, dose-dependently stimulated glucose consumption in L6 myotubes, which was accompanied by enhanced glucose transporter (GLUT)-4 translocation to the cell surface. Fargesin-stimulated glucose uptake was blocked by wortmannin, a phosphatidylinositol-3 kinase (PI3 K) inhibitor. Fargesin stimulated Akt phosphorylation, a key component in the insulin signaling pathway, which was completely inhibited by wortmannin. Here, we demonstrated that fargesin, a bioactive component of Flos Magnoliae, increases basal glucose uptake and GLUT4 translocation in L6 myotubes by activating the PI3 K-Akt pathway.
Collapse
|
22
|
Lee YS, Cha BY, Choi SS, Harada Y, Choi BK, Yonezawa T, Teruya T, Nagai K, Woo JT. Fargesin improves lipid and glucose metabolism in 3T3-L1 adipocytes and high-fat diet-induced obese mice. Biofactors 2012; 38:300-8. [PMID: 22674784 DOI: 10.1002/biof.1022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 04/02/2012] [Indexed: 12/20/2022]
Abstract
This study examined the effects of fargesin, a neolignan isolated from Magnolia plants, on obesity and insulin resistance and the possible mechanisms involved in these effects in 3T3-L1 adipocytes and high-fat diet (HFD)-induced obese mice. Fargesin promoted the glucose uptake in 3T3-L1 adipocytes. In HFD-induced obese mice, fargesin decreased the body weight gain, white adipose tissue (WAT), and plasma triglyceride, non-esterified fatty acid and glucose levels, and improved the glucose tolerance. Fargesin increased glucose transporter 4 (GLUT4) protein expression and phosphorylation of Akt, AMP-activated protein kinase (AMPK), and acetyl-CoA carboxylase (ACC) in both 3T3-L1 adipocytes and WAT of HFD-induced obese mice. Fargesin also decreased the mRNA expression levels of fatty acid oxidation-related genes, such as peroxisome proliferator-activated receptor α (PPARα), carnitine palmitoyltransferase-1 (CPT-1), uncoupling protein-2 (UCP-2) and leptin in WAT. Taken together, the present findings suggest that fargesin improves dyslipidemia and hyperglycemia by activating Akt and AMPK in WAT.
Collapse
Affiliation(s)
- Young-Sil Lee
- Research Institute for Biological Functions, Chubu University, Kasugai, Aichi, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kim KH, Moon E, Choi SU, Kim SY, Lee KR. Biological evaluation of phenolic constituents from the trunk of Berberis koreana. Bioorg Med Chem Lett 2011; 21:2270-3. [PMID: 21420296 DOI: 10.1016/j.bmcl.2011.02.104] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Revised: 02/23/2011] [Accepted: 02/24/2011] [Indexed: 01/24/2023]
Abstract
A bioassay-guided fractionation and chemical investigation of the trunk of Berberis koreana resulted in the isolation and identification of a new sesquilignan, named berbikonol (1), along with fourteen known lignan derivatives (2-15) and a new phenolic compound, named berfussinol (16), together with five known ones (17-21). The structures of these new compounds were elucidated on the basis of 1D and 2D NMR spectroscopic data analysis as well as circular dichroism (CD) spectroscopy studies. Compounds 1-5, 7-8, 11, and 14 showed significant cytotoxicity against the XF498 cell line with IC(50) values of 7.14-19.32 μM. In addition, compounds 3-8 and 15 strongly reduced nitric oxide (NO) production in lipopolysaccharide (LPS)-activated BV-2 cells, a microglial cell line.
Collapse
Affiliation(s)
- Ki Hyun Kim
- Natural Products Laboratory, School of Pharmacy, Sungkyunkwan University, 300 Chonchon-dong, Jangan-ku, Suwon 440-746, Republic of Korea
| | | | | | | | | |
Collapse
|
24
|
Kim DK, Liu KH, Jeong JH, Ji HY, Oh SR, Lee HK, Lee HS. In vitrometabolism of magnolin and characterization of cytochrome P450 enzymes responsible for its metabolism in human liver microsomes. Xenobiotica 2011; 41:358-71. [DOI: 10.3109/00498254.2010.549968] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
25
|
Jeong JH, Kim DK, Ji HY, Oh SR, Lee HK, Lee HS. Liquid chromatography-atmospheric pressure chemical ionization tandem mass spectrometry for the simultaneous determination of dimethoxyaschantin, dimethylliroresinol, dimethylpinoresinol, epimagnolin A, fargesin and magnolin in rat plasma. Biomed Chromatogr 2010; 25:879-89. [PMID: 21058411 DOI: 10.1002/bmc.1538] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 09/04/2010] [Accepted: 09/06/2010] [Indexed: 11/06/2022]
Abstract
A rapid, selective, and sensitive liquid chromatography-atmospheric pressure chemical ionization (APCI) tandem mass spectrometry method was developed for the simultaneous determination of dimethoxyaschantin, dimethylliroresinol, dimethylpinoresinol, epimagnolin A, fargesin and magnolin, the pharmacologically active ingredients of Magnolia fargesii in rat plasma. These tetrahydrofurofuranoid lignans were extracted from rat plasma using tert-butyl methyl ether at pH 7.4. The analytes were separated on a Pinnacle DB biphenyl column with 65% methanol in 10 mm ammonium formate (pH 3.0) and detected by APCI tandem mass spectrometry in the selective reaction monitoring mode. The calibration curves were linear (r(2) ≥ 0.996) over the concentration range of 20.0-1000 ng/mL for six tetrahydrofurofuranoid lignans. The lower limit of quantification for these lignans was 20.0 ng/mL with 50 µL of plasma sample. The intra- and inter-assay coefficient of variation and relative error for the six tetrahydrofurofuranoid lignans at four quality control concentrations were 0.2-9.9% and -8.5-8.2%, respectively. There was no matrix effect for the six tetrahydrofurofuranoid lignans and tolterodine (internal standard). The pharmacokinetics of dimethylliroresinol, dimethylpinoresinol, epimagnolin A, fargesin and magnolin were evaluated after oral administration of a purified extract isolated from dried flower buds of Magnolia fargesii at doses of 5.5, 11.0 and 22.0 mg/kg in male rats.
Collapse
Affiliation(s)
- Ji Hyun Jeong
- National Research Laboratory for Drug Metabolism and Bioanalysis, College of Pharmacy, Wonkwang University, Iksan 570-749, Korea
| | | | | | | | | | | |
Collapse
|
26
|
Neuroprotective phenolics in medicinal plants. Arch Pharm Res 2010; 33:1611-32. [DOI: 10.1007/s12272-010-1011-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 09/02/2010] [Accepted: 09/02/2010] [Indexed: 12/26/2022]
|