Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Toxicol Res ; 40(1): 125-137, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38223669

RESUMO

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.

2.
Nat Prod Res ; 37(16): 2759-2764, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36200684

RESUMO

The present study carried out to investigate the bioactive chemical compounds, total polyphenol content and antioxidant potential of different extracts of the Zanthoxylum armatum leaves collected from the Nainital, Uttarakhand. The GC-MS analysis of Z. armtum leaves extract resulted in the isolation of sixty, twelve, twenty-three and nineteen phytochemical constituents in methanol, ethanol, chloroform and water extracts respectively. The leaves extracts were strongly characterised by Heneicosane, Tetratetracontane, Phytol, Fargesin, (+)- Seasmin and Paulowin. Methanol extract showed maximum DPPH (2,2-Di-phenyl-1-picryl-hydrazyl) Free radical scavenging activity (IC 50 15.63 ± 0.31), Ferric Reducing Antioxidant Activity (88.98 ± 3.34 AAE ± SD) and Metal Chelating Activity (IC 50 9.89 ± 0.83). The results showed that the methanolic extract exhibited the highest phenolic content for total phenol content (98.26 ± 0.8 mg of Gallic acid equivalent/g of dry weight), total flavonoid content (61.50 ± 1.62 mg of Quercetin equivalent/g of dry weight) and total tannin content (79.96 ± 0.81 mg of Tannic acid equivalent/g of dry weight).

3.
J Biochem Mol Toxicol ; 36(11): e23197, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35983679

RESUMO

Fragesin, a traditional Chinese medicine, has been shown to exert anti-inflammatory effect. The aim of this study was to figure out the possible effectiveness of the fargesin, and to invest the mechanisms by which it works in the cadmium-induced lung injury in mice. Fargesin was given 1 h before cadmium treatment for 7 days. Then, the bronchoalveolar lavage fluid (BALF) were harvested to test inflammatory cells and pro-inflammatory cytokine production. Lung histopathological changes, myeloperoxidase (MPO) activity, and aryl hydrocarbon receptor (AhR) and nuclear factor kappa B (NF-κB) activation were measured. Fargesin dose-dependently reduced inflammatory cells and pro-inflammatory cytokines in BALF, improved lung histopathological injury, and inhibited lung wet/dry ratio and MPO activity. Furthermore, fargesin inhibited cadmium-induced NF-κB activation. In addition, fargesin was found to increase AhR expression. In conclusion, fargesin attenuates cadmium-induced lung injury may be via activating AhR, which subsequently suppressing the inflammatory response.


Assuntos
Lesão Pulmonar , NF-kappa B , Camundongos , Animais , NF-kappa B/metabolismo , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/prevenção & controle , Cádmio/toxicidade , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais , Líquido da Lavagem Broncoalveolar , Pulmão/metabolismo , Lipopolissacarídeos/farmacologia
4.
Arthritis Res Ther ; 23(1): 142, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33990219

RESUMO

BACKGROUND: To investigate the role and regulatory mechanisms of fargesin, one of the main components of Magnolia fargesii, in macrophage reprogramming and crosstalk across cartilage and synovium during osteoarthritis (OA) development. METHODS: Ten-week-old male C57BL/6 mice were randomized and assigned to vehicle, collagenase-induced OA (CIOA), or CIOA with intra-articular fargesin treatment groups. Articular cartilage degeneration was evaluated using the Osteoarthritis Research Society International (OARSI) score. Immunostaining and western blot analyses were conducted to detect relative protein. Raw264.7 cells were treated with LPS or IL-4 to investigate the role of polarized macrophages. ADTC5 cells were treated with IL-1ß and conditioned medium was collected to investigate the crosstalk between chondrocytes and macrophages. RESULTS: Fargesin attenuated articular cartilage degeneration and synovitis, resulting in substantially lower Osteoarthritis Research Society International (OARSI) and synovitis scores. In particular, significantly increased M2 polarization and decreased M1 polarization in synovial macrophages were found in fargesin-treated CIOA mice compared to controls. This was accompanied by downregulation of IL-6 and IL-1ß and upregulation of IL-10 in serum. Conditioned medium (CM) from M1 macrophages treated with fargesin reduced the expression of matrix metalloproteinase-13, RUNX2, and type X collagen and increased Col2a1 and SOX9 in OA chondrocytes, but fargesin alone did not affect chondrocyte catabolic processes. Moreover, fargesin exerted protective effects by suppressing p38/ERK MAPK and p65/NF-κB signaling. CONCLUSIONS: This study showed that fargesin switched the polarized phenotypes of macrophages from M1 to M2 subtypes and prevented cartilage degeneration partially by downregulating p38/ERK MAPK and p65/NF-κB signaling. Targeting macrophage reprogramming or blocking the crosstalk between macrophages and chondrocytes in early OA may be an effective preventive strategy.


Assuntos
Cartilagem Articular , Osteoartrite , Animais , Benzodioxóis , Condrócitos , Interleucina-1beta , Lignanas , Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B , Osteoartrite/tratamento farmacológico
5.
Korean J Physiol Pharmacol ; 25(3): 251-258, 2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-33859065

RESUMO

Flos magnoliae (FM), the dry flower buds of Magnolia officinalis or its related species, is a traditional herbal medicine commonly used in Asia for symptomatic relief of and treating allergic rhinitis, headache, and sinusitis. Although several studies have reported the effects of FM on store-operated calcium entry (SOCE) via the ORAI1 channel, which is essential during intracellular calcium signaling cascade generation for T cell activation and mast cell degranulation, the effects of its isolated constituents on SOCE remain unidentified. Therefore, we investigated which of the five major constituents of 30% ethanoic FM (vanillic acid, tiliroside, eudesmin, magnolin, and fargesin) inhibit SOCE and their physiological effects on immune cells. The conventional whole-cell patch clamp results showed that fargesin, magnolin, and eudesmin significantly inhibited SOCE and thus human primary CD4+ T lymphocyte proliferation, as well as allergen-induced histamine release in mast cells. Among them, fargesin demonstrated the most potent inhibitory effects not only on ORAI1 (IC50 = 12.46 ± 1.300 µM) but also on T-cell proliferation (by 87.74% ± 1.835%) and mast cell degranulation (by 20.11% ± 5.366%) at 100 µM. Our findings suggest that fargesin can be a promising candidate for the development of therapeutic drugs to treat allergic diseases.

6.
Int J Mol Sci ; 22(4)2021 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-33669811

RESUMO

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.


Assuntos
Benzodioxóis/farmacologia , Transformação Celular Neoplásica/patologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Ciclina E/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Fator de Crescimento Epidérmico/efeitos adversos , Lignanas/farmacologia , Transdução de Sinais , Morte Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
7.
Pharmaceutics ; 13(2)2021 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-33535454

RESUMO

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.

8.
Artigo em Inglês | MEDLINE | ID: mdl-31988050

RESUMO

BACKGROUND AND AIMS: Fargesin mainly functions in the improvement of lipid metabolism and the inhibition of inflammation, but the role of fargesin in atherogenesis and the molecular mechanisms have not been defined. We aimed to explore if and how fargesin affects atherosclerosis by regulating lipid metabolism and inflammatory response. METHODS AND RESULTS: ApoE-/- mice were fed a high-fat diet to form atherosclerotic plaques and then administrated with fargesin or saline via gavage. Oil Red O, HE and Masson staining were performed to assess atherosclerostic plaques in apoE-/- mice. [3H] labeled cholesterol was used to detect cholesterol efflux and reverse cholesterol transport (RCT) efficiency. Enzymatic methods were performed to analyze plasma lipid profile in apoE-/- mice. Immunohistochemistry was used to analyze macrophage infiltration. THP-1-derived macrophages were incubated with fargesin or not. Both Western blot and qRT-PCR were applied to detect target gene expression. Oil Red O staining was applied to examine lipid accumulation in THP-1-derived macrophages. ELISA and qRT-PCR were used to examine the levels of inflammatory mediotors. We found that fargesin reduced atherosclerotic lesions by elevating efficiency of RCT and decreasing inflammatory response via upregulation of ABCA1 and ABCG1 expression in apoE-/- mice. Further, fargesin reduced lipid accumulation in THP-1-derived macrophages. Besides, fargesin increased phosphorylation of CEBPα in Ser21 and then upregulated LXRα, ABCA1 and ABCG1 expression in THP-1-derived macrophages. In addition, fargesin could reduce ox-LDL-induced inflammatory response by inactivation of the TLR4/NF-κB pathway. CONCLUSION: These results suggest that fargesin inhibits atherosclerosis by promoting RCT process and reducing inflammatory response via CEBPαS21/LXRα and TLR4/NF-κB pathways, respectively.


Assuntos
Aterosclerose/tratamento farmacológico , Benzodioxóis/administração & dosagem , Colesterol/metabolismo , Lignanas/administração & dosagem , Metabolismo dos Lipídeos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Administração Oral , Animais , Aterosclerose/imunologia , Aterosclerose/metabolismo , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Knockout para ApoE , NF-kappa B/metabolismo , Transdução de Sinais/imunologia , Células THP-1 , Receptor 4 Toll-Like/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
9.
J Dermatol Sci ; 94(1): 213-219, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30956031

RESUMO

BACKGROUND: Fargesin is commonly used in the treatment of allergic rhinitis, inflammation, sinusitis and headache. OBJECTIVE: The aim of the study is to investigate a new function of fargesin against melanin production and its underlying molecular mechanism. METHODS: B16F10 mouse melanoma cells, Melan-a and human epidermal melanocytes were treated with different concentrations of fargesin for the indicated time. The extracellular and cellular melanin content was detected by spectrometry at 490 nm and 405 nm, respectively. RT-qPCR and Western blot analysis were used to exam the expression of melanogenic enzymes and the activities of PKA/CREB and p38 MAPK pathway components. Zebrafish was used as an in vivo model for studying the function of fargesin in regulating melanogenesis. RESULTS: Fargesin effectively inhibited melanin production at moderate dose in mouse B16F10 melanoma cells, normal melanocyte cell lines and zebrafish. The expression of microphthalmia-associated transcription factor (MITF), its downstream melanogenic enzymes and tyrosinase activity were also strongly reduced by fargesin. Moreover, the increase of melanin production induced by UVB and forskolin could be fully reversed by fargesin treatment. Fargesin also effectively inhibited the activation of PKA/CREB and p38 MAPK as well as their interactions, which in turn is responsible for the expression of MITF and melanogenic enzymes. CONCLUSIONS: These results show that fargesin can function as an anti-melanogenic agent, at least in part, by inhibiting PKA/CREB and p38/MAPK signaling pathways. Therefore, fargesin and its derivatives may potentially be used for preventing hyperpigmentation disorders in the future.


Assuntos
Benzodioxóis/farmacologia , Hiperpigmentação/tratamento farmacológico , Lignanas/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melaninas/biossíntese , Melanócitos/efeitos dos fármacos , Animais , Benzodioxóis/uso terapêutico , Linhagem Celular Tumoral , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Avaliação Pré-Clínica de Medicamentos , Embrião não Mamífero , Humanos , Lignanas/uso terapêutico , Melanócitos/metabolismo , Camundongos , Modelos Animais , Peixe-Zebra
10.
Molecules ; 23(6)2018 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-29880739

RESUMO

Fargesin is a bioactive lignan from Flos Magnoliae, an herb widely used in the treatment of allergic rhinitis, sinusitis, and headache in Asia. We sought to investigate whether fargesin ameliorates experimental inflammatory bowel disease (IBD) in mice. Oral administration of fargesin significantly attenuated the symptoms of dextran sulfate sodium (DSS)-induced colitis in mice by decreasing the inflammatory infiltration and myeloperoxidase (MPO) activity, reducing tumor necrosis factor (TNF)-α secretion, and inhibiting nitric oxide (NO) production in colitis mice. The degradation of inhibitory κBα (IκBα), phosphorylation of p65, and mRNA expression of nuclear factor κB (NF-κB) target genes were inhibited by fargesin treatment in the colon of the colitis mice. In vitro, fargesin blocked the nuclear translocation of p-p65, downregulated the protein levels of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2), and dose-dependently inhibited the activity of NF-κB-luciferase in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. Taken together, for the first time, the current study demonstrated the anti-inflammatory effects of fargesin on chemically induced IBD might be associated with NF-κB signaling suppression. The findings may contribute to the development of therapies for human IBD by using fargesin or its derivatives.


Assuntos
Anti-Inflamatórios/uso terapêutico , Benzodioxóis/uso terapêutico , Doenças Inflamatórias Intestinais/tratamento farmacológico , Lignanas/uso terapêutico , Animais , Anti-Inflamatórios/farmacologia , Benzodioxóis/farmacologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Sulfato de Dextrana/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Mediadores da Inflamação/metabolismo , Doenças Inflamatórias Intestinais/induzido quimicamente , Lignanas/farmacologia , Luciferases/antagonistas & inibidores , Camundongos , NF-kappa B/genética , NF-kappa B/metabolismo , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Peroxidase/antagonistas & inibidores , Proteólise , Células RAW 264.7 , Fator de Necrose Tumoral alfa/antagonistas & inibidores
11.
J Ethnopharmacol ; 216: 104-119, 2018 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-29309862

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: About 2-5% of the world's population is suffering from liver toxicity including Pakistan with the second highest rate of hepatitis prevalence. Liver is a vital body organ which not only performs metabolic activities but also aids in detoxification, storage and digestion of food. Now a day's malnutrition, alcohol consumption and drug addiction are major causes of liver diseases throughout the world. In fact, there is no possible outcome to compensate liver malfunction for long term, and transplantation of liver is the only option left after the irretrievable injury of hepatic function. Subsequently, natural based therapeutic approaches are in the process of scrupulous testing as strong hepatoprotective mediator. In this regard plants are well thought hepatoprotective agents having multiple active components. In this review, based on species' pharmacology and safety we have compiled some plants which show strong hepatoprotective activity, main phytoconstituents with biological activities and few commercially used herbal formulations. MATERIALS AND METHODS: Ethnopharmacological information was gathered by an extensive literature survey like WHO monographs on selected herbal medicinal plants (Vol 1-Vol 4); Principles and Practice of Phytotherapy, Mills S and Bone K, Churchill Livingstone, London, UK; Herbal Drugs and Phytopharmaceuticals, Wichtl M Medpharm Press, Stuttgart 3rd edn; Pharmacology and Applications of Chinese Materia Medica Vols 1 and 2, Chang H-M and But P P-H World Scientific, Singapore; British Herbal Compendium Vol. 2, Bradley P British Herbal Medicine Association, Bournemouth, UK; ESCOP Monographs 2nd edn. Thieme, Stuttgart, Germany; as well as by using electronic databases such as Pubchem, Chemspider, http://www.herbal-ahp.org; http://www.ahpa.org; http://whqlibdoc.who.int/publications/2003/9241546271.pdf; http://www.escop.com, Pubmed, HubMed and Scopus. RESULTS: Data for more about 29 plants have been accomplished for their bioactive constituent(s), biological activities and medicinal uses. Some of the plants have been identified as strong hepato-modulator. Such knowledge about traditional medicinal plants can be globally applied for safe and evidence based use in pharmacological applications. CONCLUSION: With the rise in liver risks a meek struggle has been made to draw attention toward herbal therapy. Hepatoprotective constituents of said plants are expressed with chemical structures. However, for certain plants active constituents are not still isolated/purified but overall plant extract was found effective in providing protection against hepatic injury. As a future perspective, there is need to purify plant active constituents for ethnomedical rationale.


Assuntos
Hepatopatias/prevenção & controle , Fígado/efeitos dos fármacos , Extratos Vegetais/uso terapêutico , Substâncias Protetoras/uso terapêutico , Animais , Etnofarmacologia , Humanos , Fígado/metabolismo , Fígado/patologia , Fígado/fisiopatologia , Hepatopatias/diagnóstico , Hepatopatias/epidemiologia , Hepatopatias/fisiopatologia , Fitoterapia , Extratos Vegetais/efeitos adversos , Plantas Medicinais/efeitos adversos , Plantas Medicinais/química , Plantas Medicinais/classificação , Substâncias Protetoras/efeitos adversos , Resultado do Tratamento
12.
Int J Mol Sci ; 18(5)2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-28468305

RESUMO

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.


Assuntos
Benzodioxóis/farmacologia , Inibidores das Enzimas do Citocromo P-450/farmacocinética , Furanos/farmacologia , Lignanas/farmacocinética , Microssomos Hepáticos/efeitos dos fármacos , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Lignanas/química , Lignanas/farmacologia , Microssomos Hepáticos/metabolismo
13.
Phytomedicine ; 24: 96-103, 2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-28160867

RESUMO

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-1ß, 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.


Assuntos
Anti-Inflamatórios/uso terapêutico , Inflamação/tratamento farmacológico , Lignanas/uso terapêutico , Magnolia/química , Monócitos/efeitos dos fármacos , Extratos Vegetais/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Linhagem Celular , Humanos , Inflamação/metabolismo , Camundongos , Fitoterapia
14.
Can J Physiol Pharmacol ; 94(8): 900-6, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27409158

RESUMO

Fargesin, a bioactive neolignan isolated from magnolia plants, is widely used in the treatment of managing rhinitis, inflammation, histamine, sinusitis, and headache. To provide more biological information about fargesin, we investigated the effects of fargesin on rat aortic rings and 2-kidney, 1-clip (2K1C) hypertensive rats. In vitro, fargesin caused concentration-dependent vasorelaxation in rat isolated aortic rings induced by KCl and norepinephrine. The effect was weakened by endothelium denudation and nitric oxide (NO) synthesis inhibition. In vivo, the evolution of systolic blood pressure (SBP) was followed by weekly measurements. Angiotensin II (Ang II) and endothelin (ET) levels, NO and nitric oxide synthase (NOS), and plasma and liver oxidative stress markers were determined at the end of the experimental period. After 5 weeks of fargesin treatment, we found that fargesin treatment reduced SBP, cardiac hypertrophy, and Ang II and ET levels of hypertensive rats. Increased NOS activity and NO level were observed in fargesin-treated rats. Normalisation of plasma MDA concentrations and improvement of the antioxidant defence system in plasma and liver accompanied the antihypertensive effect of fargesin. Taken together, these results provided substantial evidences that fargesin has antihypertensive effect in 2K1C hypertensive rats via inhibiting oxidative stress and promoting NO release.


Assuntos
Anti-Hipertensivos/uso terapêutico , Benzodioxóis/uso terapêutico , Hipertensão/tratamento farmacológico , Lignanas/uso terapêutico , Óxido Nítrico/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Anti-Hipertensivos/farmacologia , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/metabolismo , Benzodioxóis/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Hipertensão/metabolismo , Lignanas/farmacologia , Masculino , Técnicas de Cultura de Órgãos , Estresse Oxidativo/fisiologia , Ratos , Ratos Wistar , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia
15.
Fitoterapia ; 105: 16-25, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26025856

RESUMO

Fargesin displayed similar chromatographic retention peak to metoprolol in the cardiac muscle/cell membrane chromatography (CM/CMC) and ß1 adrenergic receptor/cell membrane chromatography (ß1AR/CMC) models. To provide more biological information about fargesin, we investigated the effects of fargesin on isoproterenol-(ISO-) induced cells injury in the high expression ß1 adrenergic receptor/Chinese hamster ovary-S (ß1AR/CHO-S) cells and occluding the left coronary artery- (LAD-) induced myocardial ischemia/reperfusion (MI/R) injury in rats. The results in vitro showed that ISO-induced canonical cyclic adenosine monophosphate (cAMP) and protein kinase A (PKA) levels were decreased by fargesin in ß1AR/CHO-S cells. Fargesin attenuated the serum creatine kinase (CK), lactate dehydrogenase (LDH), and improved histopathological changes of ischemic myocardium compared with the I/R rats. Similar results were obtained with Evans Blue/TTC staining, in which fargesin notably reduced infarct size. Moreover, compared with the I/R group, fargesin increased COX release and the activities of some endogenous antioxidative enzymes including superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), but suppressed malondialdehyde (MDA), and intracellular ROS release. Additionally, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay demonstrated fargesin suppressed myocardial apoptosis, which may be related to inhibition of caspase-3 activity. Taken together, these results provided substantial evidences that fargesin as a potential ß1AR antagonist through cAMP/PKA pathway could protect against myocardial ischemia/reperfusion injury in rats. The underlining mechanism may be related to inhibiting oxidative stress and myocardial apoptosis.


Assuntos
Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Apoptose/efeitos dos fármacos , Benzodioxóis/farmacologia , Cardiotônicos/farmacologia , Coração/efeitos dos fármacos , Lignanas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Células CHO , Catalase/metabolismo , Creatina Quinase/sangue , Cricetulus , Modelos Animais de Doenças , Glutationa Peroxidase/metabolismo , L-Lactato Desidrogenase/sangue , Malondialdeído/metabolismo , Estrutura Molecular , Miocárdio/patologia , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Superóxido Dismutase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA