Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Nat Prod ; 86(1): 24-33, 2023 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-36634312

RESUMO

Hyperuricemia is the result of overproduction and/or underexcretion of uric acid, and it is a well-known risk factor for gout, hypertension, and diabetes. However, available drugs for hyperuricemia in the clinic are limited. Recently, a lot of research has been conducted in order to discover new uric acid-lowering agents from plants and foods. We found that the extracts from the pericarp of mangosteen reduced urate. Bioactivity-guided study showed that α-mangostin was the principal constituent. Herein, we reported for the first time the hypouricemic activities and underling mechanism of α-mangostin. The α-mangostin dose- and time-dependently decreased the levels of serum urate in hyperuricemic mice and markedly increased the clearance of urate in hyperuricemic rats, exhibiting a promotion of urate excretion in the kidney. Further evidence showed that α-mangostin significantly decreased the protein levels of GLUT9 in the kidneys. The change in the expression of URAT1 was not observed. Moreover, α-mangostin did not inhibit the activities of xanthine oxidoreductase and uricase in vitro or in vivo. Taken together, these findings suggest that α-mangostin has potential to be developed as a new anti-hyperuricemic agent with promoting uric acid excretion.


Assuntos
Garcinia mangostana , Hiperuricemia , Ratos , Camundongos , Animais , Hiperuricemia/tratamento farmacológico , Hiperuricemia/metabolismo , Ácido Úrico/metabolismo , Xantina Oxidase , Rim/metabolismo
2.
Arch Virol ; 167(3): 953-957, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35112203

RESUMO

In this report, the full genome sequence of a novel mycovirus, designated as "Rhizoctonia solani partitivirus SM03" (RsPV-SM03), was determined in Rhizoctonia solani AG-4 HG III isolate SM03. RsPV-SM03 genome consists of two dsRNAs (dsRNA-1 and dsRNA-2), each of them contains one single open reading frame (ORF). ORF1 of dsRNA-1 encodes a putative RNA-dependent RNA polymerase (RdRp), while ORF2 of dsRNA-2 encodes a putative viral coat protein (CP). Phylogenetic analysis indicated that the RdRp and CP of RsPV-SM03 are closely related to those of other members of the genus Alphapartitivirus, family Partitiviridae, suggesting that RsPV-SM03 represents a novel species in the genus Alphapartitivirus.


Assuntos
Micovírus , Vírus de RNA , Sequência de Bases , Genoma Viral , Fases de Leitura Aberta , Filogenia , RNA de Cadeia Dupla , RNA Viral/genética , Rhizoctonia
3.
Arch Virol ; 167(1): 255-259, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34761285

RESUMO

The full-length nucleotide sequence and genome organization of a novel mycovirus designated as "Rhizoctonia fumigata partitivirus 1" (RfPV1) from Rhizoctonia fumigata AG-Ba strain C-314 Baishi was determined. The genome of RfPV1 consists of two double-stranded RNAs (dsRNAs): dsRNA1 (2003 bp) and dsRNA2 (1802 bp). Each of the two dsRNAs contains one open reading frame, coding for a putative RNA-dependent RNA polymerase and a coat protein, respectively. The 5' untranslated regions (UTRs) of both dsRNAs were conserved, and the 3'-UTRs of the two dsRNAs had interrupted poly(A) tails, similar to other partitiviruses. Phylogenetic analysis indicated that RfPV1 is a new species in the genus Alphapartitivirus, family Partitiviridae.


Assuntos
Micovírus , Vírus de RNA , Rhizoctonia/virologia , Micovírus/genética , Genoma Viral , Fases de Leitura Aberta , Filogenia , Vírus de RNA/genética , RNA de Cadeia Dupla/genética , RNA Viral/genética
4.
Biol Pharm Bull ; 43(11): 1653-1659, 2020 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-32863294

RESUMO

Hyperuricemia is mainly the result of relative underexcretion of urate. Urate is mainly eliminated by kidney and several important transporters expressed on the membrane of renal tubular cells involved in urate excretion. Olsalazine sodium was screened from 3167 authorized small compounds/drugs, targeting xanthine oxidoreductase. In previous study, we reported that olsalazine sodium significantly reduced the serum urate levels, and the anti-hyperuricemic activity linked with inhibiting urate formation by reducing the activity of xanthine oxidoreductase. The current research aimed to assess olsalazine sodium renal urate excretion and likely molecular mechanism. The results showed that administration of olsalazine sodium 5.0 mg/kg decreased the levels of serum urate in hyperuricemic rats, and noticeably improved the fractional excretion of urate and urate clearance, exhibiting an uricosuric action. Moreover, olsalazine sodium (2.5, 5.0, 10.0 mg/kg) reduced the level of blood urea nitrogen in rats. Further study showed that olsalazine sodium reduced the mRNA expression of urate reabsorptive transporter glucose transporter 9 (GLUT9), increased the mRNA expression of urate secretory transporters, organic anion transporter 1 (OAT1), OAT3 and type 1 sodium-dependent phosphate transporter (NPT1) as well as the protein expression of OAT3 in the kidney in hyperuricemic mice. In conclusion, olsalazine sodium exhibited a promotion of urate excretion in kidney by increasing the expression of OAT3.


Assuntos
Ácidos Aminossalicílicos/farmacologia , Hiperuricemia/tratamento farmacológico , Transportadores de Ânions Orgânicos Sódio-Independentes/agonistas , Eliminação Renal/efeitos dos fármacos , Ácido Úrico/metabolismo , Ácidos Aminossalicílicos/uso terapêutico , Animais , Nitrogênio da Ureia Sanguínea , Creatinina/sangue , Creatinina/urina , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Humanos , Hiperuricemia/sangue , Hiperuricemia/fisiopatologia , Hiperuricemia/urina , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/fisiopatologia , Masculino , Camundongos , Proteínas de Transporte de Monossacarídeos/antagonistas & inibidores , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/agonistas , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Ratos , Ratos Sprague-Dawley , Eliminação Renal/fisiologia , Reabsorção Renal/efeitos dos fármacos , Reabsorção Renal/fisiologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo I/agonistas , Proteínas Cotransportadoras de Sódio-Fosfato Tipo I/metabolismo , Ácido Úrico/sangue , Ácido Úrico/urina
5.
J Pharmacol Sci ; 137(2): 154-161, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29934052

RESUMO

Mangiferin, a natural glucosyl xanthone, was confirmed to be an effective uric acid (UA)- lowering agent with dual action of inhibiting production and promoting excretion of UA. In this study, we aimed to evaluate the effect of mangiferin on alleviating hypertension induced by hyperuricemia. Mangiferin (30, 60, 120 mg/kg) was administered intragastrically to hyperuricemic rats induced by gavage with potassium oxonate (750 mg/kg). Systolic blood pressure (SBP), serum levels of UA, nitric oxide (NO), C-reactionprotein (CRP) and ONOO- were measured. The mRNA and protein levels of endothelial nitric oxide synthase (eNOS), intercellular adhesion molecule-1 (ICAM-1), CRP were also analyzed. Human umbilical vein endothelial cells (HUVECs) were used in vitro studies. Administration of mangiferin significantly decreased the serum urate level and SBP at 8 weeks and last to 12 weeks. Further more, mangiferin could increase the release of NO and decrease the level of CRP in blood. In addition, mangiferin reversed the protein expression of eNOS, CRP, ICAM-1 and ONOO- in aortic segments in hyperuricemic rats. The results in vitro were consistent with the observed results in vivo. Taken together, these data suggested that mangiferin has played an important part in alleviating hypertension induced by hyperuricemia via increasing NO secretion and improving endothelial function.


Assuntos
Hipertensão/tratamento farmacológico , Hipertensão/etiologia , Hiperuricemia/tratamento farmacológico , Óxido Nítrico/metabolismo , Fitoterapia , Xantonas/administração & dosagem , Xantonas/farmacologia , Administração Oral , Animais , Aorta/metabolismo , Proteína C-Reativa/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Hipertensão/metabolismo , Hiperuricemia/induzido quimicamente , Hiperuricemia/complicações , Hiperuricemia/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Mangifera/química , Óxido Nítrico Sintase Tipo III/metabolismo , Ácido Oxônico/efeitos adversos , Folhas de Planta/química , Ratos Sprague-Dawley , Sístole/efeitos dos fármacos , Ácido Úrico/sangue , Xantonas/isolamento & purificação
6.
Biol Pharm Bull ; 41(1): 99-105, 2018 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-29093325

RESUMO

The mechanism of 3,5,2',4'-tetrahydroxychalcone on lowing urate level is still unknown. Here we investigated the effects of 3,5,2',4'-tetrahydroxychalcone on urate levels, xanthine oxidase/xanthine dehydrogenase (XOD/XDH) activities in hypoxanthine-induced hyperuricemic mice, as well as the effects of 3,5,2',4'-tetrahydroxychalcone on the mRNA expression levels and content of phosphoribosyl pyrophosphate synthetase (PRPS), phosphoribosyl pyrophosphate amidotransferase (PRPPAT) and hypoxanthine-guanine phosphoribosyl transferase (HGPRT). Our results demonstrated that 3,5,2',4'-tetrahydroxychalcone (1.0, 2.0, and 4.0 mg/kg) reduced the uric acid levels in serum of the hyperuricemic mice in dose- and time-dependent manners. The activities of XOD/XDH in serum and liver were also significantly inhibited by 3,5,2',4'-tetrahydroxychalcone; In addition, 3,5,2',4'-tetrahydroxychalcone decreased the mRNA expression of HGPRT in brain and content of PRPS and PRPPAT in liver. These findings demonstrated that 3,5,2',4'-tetrahydroxychalcone suppresses uric acid production by affecting the critical enzymes, XOD/XDH, PRPS, PRPPAT and HGPRT in purine nucleotide metabolism.


Assuntos
Chalconas/uso terapêutico , Hiperuricemia/tratamento farmacológico , Ácido Úrico/sangue , Xantina Desidrogenase/metabolismo , Xantina Oxidase/metabolismo , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Hiperuricemia/induzido quimicamente , Hiperuricemia/metabolismo , Hipoxantina/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos Endogâmicos , Purinas/metabolismo , Xantina Desidrogenase/genética , Xantina Oxidase/genética
7.
J Pharmacol Sci ; 135(3): 114-120, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29132796

RESUMO

Hyperuricemia, a long-term purine metabolic disorder, is a well-known risk factor for gout, hypertension and diabetes. In maintaining normal whole-body purine levels, xanthine oxidase (XOD) is a key enzyme in the purine metabolic pathway, as it catalyzes the oxidation of hypoxanthine to xanthine and finally to uric acid. Here we used the protein-ligand docking software idock to virtually screen potential XOD inhibitors from 3167 approved small compounds/drugs. The inhibitory activities of the ten compounds with the highest scores were tested on XOD in vitro. Interestingly, all the ten compounds inhibited the activity of XOD at certain degrees. Particularly, the anti-ulcerative-colitis drug olsalazine sodium demonstrated a great inhibitory activity for XOD (IC50 = 3.4 mg/L). Enzymatic kinetic studies revealed that the drug was a hybrid-type inhibitor of xanthine oxidase. Furthermore, the drug strikingly decreased serum urate levels, serum/hepatic activities of XOD at a dose-dependent manner in vivo. Thus, we demonstrated a successful hunting process of compounds/drugs for hyperuricemia through virtual screening, supporting a potential usage of olsalazine sodium in the treatment of hyperuricemia.


Assuntos
Ácidos Aminossalicílicos/farmacologia , Antiulcerosos/farmacologia , Ácido Úrico/sangue , Xantina Desidrogenase/antagonistas & inibidores , Xantina Desidrogenase/metabolismo , Ácidos Aminossalicílicos/uso terapêutico , Animais , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Hiperuricemia/tratamento farmacológico , Técnicas In Vitro , Masculino , Camundongos , Relação Estrutura-Atividade
8.
J Nat Prod ; 80(8): 2319-2327, 2017 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-28742368

RESUMO

An insidious increase in the incidence of obesity, insulin resistance, and hyperlipidemia has led to an epidemic of type 2 diabetes worldwide. Tinospora crispa (T. crispa) is a familiar plant traditionally used in herbal medicine for diabetes; however, the major active ingredients of this plant are still unclear. In this study, we identified the therapeutic effects of borapetoside E, a small molecule extracted from T. crispa, in high-fat-diet (HFD)-induced obesity in mice. The therapeutic effects of borapetoside E in HFD-induced obese mice were assessed physiologically, histologically, and biochemically following intraperitoneal injection. Furthermore, we analyzed the expression of glucose and lipid metabolism-related genes and proteins in borapetoside E-treated obese mice. Compared with vehicle-treated mice, borapetoside E markedly improved hyperglycemia, insulin resistance, hepatic steatosis, hyperlipidemia, and oxygen consumption in obese mice, and the effects were comparable to or better than the drug metformin. In addition, borapetoside E suppressed the expression of sterol regulatory element binding proteins (SREBPs) and their downstream target genes related to lipid synthesis in the liver and adipose tissue. Borapetoside E showed beneficial effects in vivo, demonstrating that borapetoside E may be a potential therapy for the treatment of diet-induced type 2 diabetes and related metabolic syndromes.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Diterpenos Clerodânicos/isolamento & purificação , Diterpenos Clerodânicos/farmacologia , Hiperglicemia/metabolismo , Hiperlipidemias/metabolismo , Fígado/efeitos dos fármacos , Plantas Medicinais/química , Tinospora/química , Animais , Dieta Hiperlipídica , Diterpenos Clerodânicos/química , Resistência à Insulina , Metabolismo dos Lipídeos , Fígado/metabolismo , Camundongos , Estrutura Molecular , Fitoterapia
9.
Am J Physiol Cell Physiol ; 311(5): C831-C836, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27653985

RESUMO

Adipose tissue plays a critical role in metabolic diseases and the maintenance of energy homeostasis. RACK1 has been identified as an adaptor protein involved in multiple intracellular signal transduction pathways and diseases. However, whether it regulates adipogenesis remains unknown. Here, we reported that RACK1 is expressed in 3T3-L1 cells and murine white adipose tissue and that RACK1 knockdown by shRNA profoundly suppressed adipogenesis by reducing the expression of PPAR-γ and C/EBP-ß. Depletion of RACK1 increased ß-catenin protein levels and activated Wnt signaling. Furthermore, RACK1 knockdown also suppressed the PI3K-Akt-mTOR-S6K signaling pathway by reducing the PI3K p85α, pAkt T473, and S6K p70. Taken together, these results demonstrate that RACK1 is a novel factor required for adipocyte differentiation by emerging Wnt/ß-catenin signaling and PI3K-Akt-mTOR-S6K signaling pathway(s).


Assuntos
Adipogenia/fisiologia , Neuropeptídeos/metabolismo , Células 3T3-L1 , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Animais , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular , Camundongos , PPAR gama/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/metabolismo , Receptores de Quinase C Ativada , Serina-Treonina Quinases TOR/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo
10.
Yao Xue Xue Bao ; 51(4): 613-5, 2016 04.
Artigo em Zh | MEDLINE | ID: mdl-29860745

RESUMO

To study the constituents of Karelinia caspia(Pall.) Less, three phenylpropanoid derivatives and other compounds were isolated by silica gel, RP-18 chromatographic methods. Compound 1 was a new phenylpropanoid glycoside named as kareline A. Their structures were determined by spectroscopic analysis, including 1D- and 2D-NMR and mass spectrometry.


Assuntos
Asteraceae/química , Glicosídeos/isolamento & purificação , Extratos Vegetais/química , Propanóis/isolamento & purificação , Glicosídeos/química , Estrutura Molecular , Propanóis/química
11.
Pharm Biol ; 54(9): 1680-6, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26916555

RESUMO

Context Mangiferin has been reported to possess a potential hypouricaemic effect. However, the pharmacokinetic studies in rats showed that its oral bioavailability was only 1.2%, suggesting that mangiferin metabolites might exert the action. Objective The hypouricaemic effect and the xanthine oxidase inhibition of mangiferin and norathyriol, a mangiferin metabolite, were investigated. Inhibition of norathyriol analogues (compounds 3-9) toward xanthine oxidase was also evaluated. Materials and methods For a dose-dependent study, mangiferin (1.5-6.0 mg/kg) and norathyriol (0.92-3.7 mg/kg) were administered intragastrically to mice twice daily for five times. For a time-course study, mice received mangiferin and norathyriol both at a single dose of 7.1 µmol/kg. In vitro, inhibition of test compounds (2.4-2.4 mM) against xanthine oxidase activity was evaluated by the spectrophotometrical method. The inhibition type was identified from Lineweaver-Burk plots. Results Norathyriol (0.92, 1.85 and 3.7 mg/kg) dose dependently decreased the serum urate levels by 27.0, 33.6 and 37.4%, respectively. The action was more potent than that of mangiferin at the low dose, but was equivalent at the higher doses. Additionally, the hypouricaemic action of them exhibited a time dependence. In vitro, norathyriol markedly inhibited the xanthine oxidase activities, with the IC50 value of 44.6 µM, but mangiferin did not. The kinetic studies showed that norathyriol was an uncompetitive inhibitor by Lineweaver-Burk plots. The structure-activity relationships exhibited that three hydroxyl groups in norathyriol at the C-1, C-3 and C-6 positions were essential for maintaining xanthine oxidase inhibition. Discussion and conclusion Norathyriol was responsible for the hypouricaemic effect of mangiferin via inhibiting xanthine oxidase activity.


Assuntos
Inibidores Enzimáticos/farmacologia , Supressores da Gota/farmacologia , Hiperuricemia/tratamento farmacológico , Ácido Úrico/sangue , Xantenos/farmacologia , Xantina Oxidase/antagonistas & inibidores , Xantonas/farmacologia , Administração Oral , Animais , Biomarcadores/sangue , Biotransformação , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/metabolismo , Supressores da Gota/administração & dosagem , Supressores da Gota/metabolismo , Hiperuricemia/sangue , Hiperuricemia/induzido quimicamente , Hiperuricemia/enzimologia , Cinética , Camundongos , Estrutura Molecular , Ácido Oxônico , Relação Estrutura-Atividade , Xantenos/administração & dosagem , Xantenos/metabolismo , Xantina Oxidase/metabolismo , Xantonas/administração & dosagem , Xantonas/metabolismo
12.
Biol Pharm Bull ; 38(10): 1591-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26228630

RESUMO

Mangiferin, a natural glucosyl xanthone from the leaves of Mangifera indica L., was previously shown to exert potent hypouricemic effects associated with inhibition of the activity of xanthine dehydrogenase/oxidase. The present study aimed to evaluate its uricosuric effect and possible molecular mechanisms underlying the renal urate transporters responsible for urate reabsorption in vivo. Mangiferin (1.5-24.0 mg/kg) was administered intragastrically to hyperuricemic mice and rats induced by the intraperitoneal injection of uric acid and potassium oxonate, respectively. The uricosuric effect was evaluated by determining the serum and urinary urate levels as well as fractional excretion of uric acid (FEUA). The mRNA and protein levels of renal urate-anion transporter 1 (URAT1), organic anion transporter 10 (OAT10), glucose transporter 9 (GLUT9), and PDZ domain-containing protein (PDZK1) were analyzed. The administration of mangiferin significantly decreased the serum urate levels in hyperuricemic mice in a dose- and time-dependent manner. In hyperuricemic rats, mangiferin also reduced the serum urate levels and increased the urinary urate levels and FEUA. These results indicate that mangiferin has uricosuric effects. Further examination showed that mangiferin markedly inhibited the mRNA and protein expression of renal URAT1, OAT10, and GLUT9 in hyperuricemic rats, but did not interfere with PDZK1 expression. Taken together, these findings suggest that mangiferin promotes urate excretion by the kidney, which may be related to the inhibition of urate reabsorption via downregulation of renal urate transporters.


Assuntos
Hiperuricemia/metabolismo , Rim/efeitos dos fármacos , Ácido Úrico/metabolismo , Uricosúricos/farmacologia , Xantonas/farmacologia , Animais , Proteínas de Transporte de Ânions/genética , Proteínas de Transporte de Ânions/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas do Citoesqueleto , Hiperuricemia/sangue , Hiperuricemia/induzido quimicamente , Rim/metabolismo , Masculino , Camundongos , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Ácido Oxônico , Ratos Sprague-Dawley , Ácido Úrico/sangue , Ácido Úrico/urina
13.
Zhongguo Zhong Yao Za Zhi ; 40(23): 4634-8, 2015 Dec.
Artigo em Zh | MEDLINE | ID: mdl-27141675

RESUMO

This study was to investigate the chemical constituents of the aerial part of Zygophyllumfabago, by phytochemical methods. The compounds were isolated by silica gel and Sephadex LH-20 column chromatographies from the EtOAc extract. Their structures were characterized by various spectroscopic data (1H-NMR, 13C-NMR, MS) and comparison with the literature. As a result, thirteen compounds were isolated and their structures were identified as 1-hydroxyhinesol(1), hinesol(2), atractylenolactam(3), beta-eudesmol (4), 5alpha-hydroperoxy-beta-eudesmol(5), 12-hydroxy-valenc-1(10)-en-2-one(6), pubinernoid A(7), (6S,7E)-6-hydroxy-4,7-megastigmadien-3,9-dione(8), 3-hydroxy-5alpha, 6alpha-epoxy-beta-ionone (9), (3S,5R, 6S, 7E)-3, 5, 6-trihydroxy-7-megastigmen-9-one(10), (6R,7E,9R)-9-hydroxy-4,7-megastigmadien-3-one(11), (S)-3-hydroxy-beta-ionone(12), and blumenol A(13). Compounds 1-7 were sesquiterpenoids and 8-13 were megastigmane type norsesquiterpenoids. All the compounds were obtained from Z. fabago for the first time, and compound 1 was a new natural product.


Assuntos
Medicamentos de Ervas Chinesas/química , Terpenos/química , Zygophyllum/química , Medicamentos de Ervas Chinesas/isolamento & purificação , Estrutura Molecular , Espectrometria de Massas por Ionização por Electrospray , Terpenos/isolamento & purificação
14.
Pharm Biol ; 52(1): 68-73, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24033319

RESUMO

CONTEXT: Mangiferin has been reported to possess antidiabetic activities. Norathyriol, a xanthone aglycone, has the same structure as mangiferin, except for a C-glucosyl bond. To our best knowledge, no study has been conducted to determine and compare those two compounds on glucose consumption in vitro. OBJECTIVE: In this study, the effects of norathyriol and mangiferin on glucose consumption in normal and insulin resistance (IR) L6 myotubes were evaluated. Simultaneously, the potential mechanism of this effect was also investigated. MATERIALS AND METHODS: Normal or IR L6 myotubes were incubated with norathyriol (2.5 ∼ 10 µM, 0.625 ∼ 2.5 µM), mangiferin (10 ∼ 40 µM, 2.5 ∼ 10 µM) or rosiglitazone (20 µM) and/or 0.05 nM insulin for 24 h, respectively. The glucose consumption was assessed using the glucose oxidase method. Immunoblotting was performed to detect protein kinase B (PKB/Akt) and AMP-activated protein kinase (AMPK) phosphorylation in L6 myotubes cells. RESULTS: Norathyriol and mangiferin treatment alone increased the glucose consumption 61.9 and 56.3%, respectively, in L6 myotubes and made additional increasing with 0.05 nM insulin. In IR L6 myotubes, norathyriol treatment made increasing with or without insulin, mangiferin treatment also made increasing but only when co-treated with insulin. Immunoblotting results showed that norathyriol and mangiferin produced an increase of 1.9 - and 1.8-fold in the phosphorylation levels of the AMPK, but not in Akt. DISCUSSION AND CONCLUSION: Our findings suggest that norathyriol and mangiferin could improve the glucose utilization and insulin sensitivity by up-regulation of the phosphorylation of AMPK. Norathyriol may be considered as an active metabolite responsible for the antidiabetic activity of mangiferin.


Assuntos
Glucose/metabolismo , Hipoglicemiantes/farmacologia , Xantenos/farmacologia , Xantonas/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Células Cultivadas , Hipoglicemiantes/administração & dosagem , Immunoblotting , Insulina/farmacologia , Resistência à Insulina , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Rosiglitazona , Tiazolidinedionas/farmacologia , Regulação para Cima/efeitos dos fármacos , Xantenos/administração & dosagem , Xantonas/administração & dosagem
15.
Pharm Biol ; 50(9): 1177-82, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22881143

RESUMO

CONTEXT: Mangiferin, a natural bioactive xanthone C-glycoside, is widely present in medicinal plants like the leaf of Mangifera indica L. (Anacardiaceae). It has been reported that mangiferin possesses a variety of biological activities, including antidiabetic, hepatoprotective, anti-inflammatory, antioxidant, and anticarcinogenic. OBJECTIVE: The hypouricemic effect and xanthine oxidoreductase (XOR) inhibitory activity of mangiferin were investigated here for the first time. MATERIALS AND METHODS: The hypouricemic effect of mangiferin was investigated in normal and hyperuricemic mice induced by potassium oxonate. Mangiferin at a dose of 0.75-100.0 mg/kg was given intragastrically to mice. The serum urate levels were determined using the phosphotungstic acid method. The hepatic activities of xanthine dehydrogenase (XDH) and xanthine oxidase (XOD) in hyperuricemic mice were assayed using commercially available kits. RESULTS: The results showed that mangiferin at a dose of 1.5, 3.0, and 6.0 mg/kg significantly reduced the serum urate levels (148.7 ± 37.8, 142.2 ± 44.5, 121.7 ± 21.7 µmmol/L) in hyperuricemic mice, compared with untreated hyperuricemic mice (201.8 ± 71.2 µmmol/L). However, mangiferin did not decrease the serum urate levels in normal mice until mangiferin was up to 100 mg/kg. In addition, the hepatic activities of XDH in hyperuricemic mice were significantly decreased by mangiferin, while no changes of XOD were observed. Acute toxicity study in mice showed that mangiferin was very safe at a dose of up to 25 g/kg. DISCUSSION AND CONCLUSION: These findings demonstrate that mangiferin has the potential to be developed as a new therapeutic agent for the treatment of hyperuricemia and gout.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Supressores da Gota/uso terapêutico , Hiperuricemia/tratamento farmacológico , Fígado/efeitos dos fármacos , Ácido Úrico/sangue , Xantina Desidrogenase/antagonistas & inibidores , Xantonas/uso terapêutico , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/efeitos adversos , Feminino , Gota/tratamento farmacológico , Supressores da Gota/administração & dosagem , Supressores da Gota/efeitos adversos , Hiperuricemia/sangue , Hiperuricemia/enzimologia , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos , Ácido Oxônico , Fatores de Tempo , Testes de Toxicidade Aguda , Xantina Desidrogenase/metabolismo , Xantina Oxidase/metabolismo , Xantonas/administração & dosagem , Xantonas/efeitos adversos
16.
Tissue Cell ; 79: 101967, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36351341

RESUMO

The proliferation and migration of Schwann cells is pivotal to peripheral nerve injury (PNI) repair. Recent studies have revealed that Ginkgetin has neuroprotective effects. Hence, we focused on identifying whether Ginkgetin could regulate the proliferation and migration of Schwann cells, thereby contributing to the repair of PNI. Rat Schwann cells RSC96 were treated with different concentrations of Ginkgetin. Short hairpin RNA targeting phosphatidylinositol glycan anchor biosynthesis class F (shPIGF) was employed to investigate the effects of PIGF on Ginkgetin-induced RSC96 cells. Viability of RSC96 cells was estimated via cell counting kit-8 (CCK-8) assay and proliferation of the cells was assessed by 5-ethynyl-2'-deoxyuridine (EdU) assay. Migration was estimated via wound healing assay and invasion was evaluated through transwell assay. Western blot was employed to test the expressions of PIGF, protein-38 (p38), and phosphorylated p-38 (p-p38). Ginkgetin (50 or 100 µg/ml) increased the viability, proliferation, migration, and invasion of RSC96 cells, up-regulated PIGF expression and raised the ratio of p-p38/p38, which were all reversed by PIGF silencing. Ginkgetin promotes proliferation, migration, and invasion of Schwann cells via PIGF/p38 MAPK signaling pathway.


Assuntos
Células de Schwann , Transdução de Sinais , Feminino , Animais , Ratos , Fator de Crescimento Placentário , Proliferação de Células/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética
17.
Eur J Pharmacol ; 888: 173490, 2020 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-32827538

RESUMO

Increasing evidence shows that the intestinal tract plays an important role in maintaining urate homeostasis and might be a potential therapeutic target for hyperuricaemia. However, uric acid-lowering drugs available in the clinic do not target intestinal excretion as a therapeutic strategy. We previously reported that mangiferin had potent hypouricaemic effects in hyperuricaemic animals. However, the underlying mechanisms are not completely clear. Here, we investigated the effects of mangiferin on the intestinal excretion of urate and its underlying mechanisms. The data revealed that mangiferin concentration-dependently promoted the intestinal secretion of endogenous urate in in situ intestinal closed loops in normal and hyperuricaemic mice, as well as inhibited the absorption of exogenous uric acid perfused into the intestinal loops in rats. Administration of mangiferin not only decreased the serum urate levels in the hyperuricaemic mice but also increased the protein expression of ATP-binding cassette transporter, subfamily G, member 2 (ABCG2) and inhibited the protein expression of glucose transporter 9 (GLUT 9) in the intestine. These findings suggested that intestinal ABCG2 and GLUT9 might be pivotal and possible action sites for the observed hypouricaemic effects. Moreover, no significant changes in intestinal xanthine oxidoreductase activities were observed, suggesting that mangiferin did not affect intestinal uric acid generation in the hyperuricaemic mice. Overall, promoting intestinal elimination of urate by upregulating ABCG2 expression and downregulating GLUT9 expression might be an important mechanism underlying mangiferin lowering serum uric acid levels. Mangiferin supplementation might be beneficial for the prevention and treatment of hyperuricaemia.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/biossíntese , Eliminação Intestinal/efeitos dos fármacos , Proteínas de Transporte de Monossacarídeos/biossíntese , Ácido Úrico/metabolismo , Xantonas/farmacologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/agonistas , Animais , Hiperuricemia/tratamento farmacológico , Hiperuricemia/metabolismo , Eliminação Intestinal/fisiologia , Masculino , Camundongos , Proteínas de Transporte de Monossacarídeos/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Xantonas/uso terapêutico
18.
Nat Prod Bioprospect ; 10(3): 131-140, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32314168

RESUMO

Eriocalyxin B, an ent-Kaurene diterpenoid extracted from a traditional Chinese herb Isodon eriocalyx, has been shown to possess multifunctional activities such as anti-cancer and anti-inflammatory. However, the function and mechanism of the compound in adipocyte differentiation is still unknown. Here we reported that eriocalyxin B blunted adipogenesis remarkably by inhibiting the accumulation of lipid droplets, triglycerides and the expressions of adipogenesis-related factors, including C/EBPß, C/EBPα, PPARγ, and FABP4. Moreover, we showed that the inhibition might be the consequence of cell cycle being arrested at the G2/M phase during the mitotic clonal expansion of adipocyte differentiation, most likely by suppressing mRNAs and proteins of CDK1, CDK2, Cyclin A and Cyclin B1. Overall, we conclude that eriocalyxin B is capable of inhibiting adipocyte differentiation at the early stage through downregulating the proteins involved in cell cycle progression.

19.
Nat Prod Bioprospect ; 10(4): 269, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32548686

RESUMO

In the original publication of this article, we found an error under the section "Introduction". The first sentence of the fourth paragraph appears incorrectly. The corrected sentence is given below. Eriocalyxin B, isolated and identified in 1982 [1], is the major component in Chinese plant Isodon eriocalyx (Dunn.) Hara (family Lamiaceae) showing many pharmacological activities, such as inhibiting inflammatory response, regulating immune cell differentiation, inhibiting tumor cells proliferation, causing cell cycle arrest affecting angiogenesis and promoting cancer cells apoptosis.

20.
Eur J Pharmacol ; 853: 371-380, 2019 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-31009635

RESUMO

Hyperuricaemia, which results from the overproduction and underexcretion of uric acid, has been linked with chronic renal dysfunction, cardiovascular diseases, diabetes and metabolic syndrome. However, available clinical drugs cannot simultaneously target the production and excretion of uric acid. Norathyriol, a natural xanthone, was expected to have the dual actions. We previously reported that norathyriol possessed potent anti-hyperuricaemic activity related to the inhibition of uric acid production. Here, we investigated the uricosuric actions in hyperuricaemic animals and explored the possible molecular mechanisms associated with renal urate transporters and xanthine oxidase (XO). The results showed that norathyriol (0.5-4.0 mg/kg) dose- and time-dependently decreased serum urate levels in uric acid-induced hyperuricaemic mice and markedly increased the fractional excretion of urate in oxonate-induced hyperuricaemic rats, demonstrating a promotion of urate excretion in the kidney. Further evidence showed that norathyriol markedly increased renal mRNA and protein expression of the secretory organic anion transporter 1 (OAT1) in hyperuricaemic mice and strengthened its transport function in vitro. Moreover, norathyriol also upregulated the mRNA expression of the secretory transporters OAT3, ATP-binding cassette transporter G2 and multidrug resistance protein 4, but not their protein expression. Changes in the expression of the reabsorptive transporters were not observed. This is the first report that norathyriol has uricosuric effects by targeting OAT1. Moreover, norathyriol significantly inhibited XO activity in an uncompetitive manner. Taken together, these findings suggest that norathyriol has the potential to be developed as a new anti-hyperuricaemic agent with dual actions that activate OAT1 and inhibit XO activity.


Assuntos
Inibidores Enzimáticos/farmacologia , Hiperuricemia/tratamento farmacológico , Ácido Úrico/sangue , Ácido Úrico/urina , Xantenos/farmacologia , Xantina Oxidase/antagonistas & inibidores , Animais , Inibidores Enzimáticos/uso terapêutico , Células HEK293 , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Camundongos , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Xantenos/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA