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










Base de datos
Intervalo de año de publicación
1.
Acta Pharmacol Sin ; 44(1): 133-144, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35705686

RESUMEN

Cytochrome P450s are important phase I metabolic enzymes located on endoplasmic reticulum (ER) involved in the metabolism of endogenous and exogenous substances. Our previous study showed that a hepatoprotective agent silybin restored CYP3A expression in mouse nonalcoholic fatty liver disease (NAFLD). In this study we investigated how silybin regulated P450s activity during NAFLD. C57BL/6 mice were fed a high-fat-diet (HFD) for 8 weeks to induce NAFLD, and were administered silybin (50, 100 mg ·kg-1 ·d-1, i.g.) in the last 4 weeks. We showed that HFD intake induced hepatic steatosis and ER stress, leading to significant inhibition on the activity of five primary P450s including CYP1A2, CYP2B6, CYP2C19, CYP2D6, and CYP3A in liver microsomes. These changes were dose-dependently reversed by silybin administration. The beneficial effects of silybin were also observed in TG-stimulated HepG2 cells in vitro. To clarify the underlying mechanism, we examined the components involved in the P450 catalytic system, membrane phospholipids and ER membrane fluidity, and found that cytochrome b5 (cyt b5) was significantly downregulated during ER stress, and ER membrane fluidity was also reduced evidenced by DPH polarization and lower polyunsaturated phospholipids levels. The increased ratios of NADP+/NADPH and PC/PE implied Ca2+ release and disruption of cellular Ca2+ homeostasis resulted from mitochondria dysfunction and cytochrome c (cyt c) release. The interaction between cyt c and cyt b5 under ER stress was an important reason for P450s activity inhibition. The effect of silybin throughout the whole course suggested that it regulated P450s activity through its anti-ER stress effect in NAFLD. Our results suggest that ER stress may be crucial for the inhibition of P450s activity in mouse NAFLD and silybin regulates P450s activity by attenuating ER stress.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Ratones , Animales , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Silibina/farmacología , Silibina/metabolismo , Citocromo P-450 CYP3A/metabolismo , Ratones Endogámicos C57BL , Sistema Enzimático del Citocromo P-450/metabolismo , Dieta Alta en Grasa/efectos adversos , Estrés del Retículo Endoplásmico , Hígado/metabolismo
2.
Prog Brain Res ; 262: 399-430, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33931189

RESUMEN

It has been increasingly recognized that tinnitus is likely to be generated by complex network changes. Acoustic trauma that causes tinnitus induces significant changes in multiple metabolic pathways in the brain. However, it is not clear whether those metabolic changes in the brain could also be reflected in blood samples and whether metabolic changes could discriminate acoustic trauma, hyperacusis and tinnitus. We analyzed brain and serum metabolic changes in rats following acoustic trauma or a sham procedure using metabolomics. Hearing levels were recorded before and after acoustic trauma and behavioral measures to quantify tinnitus and hyperacusis were conducted at 4 weeks following acoustic trauma. Tissues from 11 different brain regions and serum samples were collected at about 3 months following acoustic trauma. Among the acoustic trauma animals, eight exhibited hyperacusis-like behavior and three exhibited tinnitus-like behavior. Using Gas chromatography-mass spectrometry and multivariate statistical analysis, significant metabolic changes were found in acoustic trauma animals in both the brain and serum samples with a number of metabolic pathways significantly perturbated. Furthermore, metabolic changes in the serum were able to differentiate sham from acoustic trauma animals, as well as sham from hyperacusis animals, with high accuracy. Our results suggest that serum metabolic profiling in combination with machine learning analysis may be a promising approach for identifying biomarkers for acoustic trauma, hyperacusis and potentially, tinnitus.


Asunto(s)
Pérdida Auditiva Provocada por Ruido , Acúfeno , Estimulación Acústica , Animales , Encéfalo , Pérdida Auditiva Provocada por Ruido/complicaciones , Hiperacusia/etiología , Ruido , Ratas , Acúfeno/etiología
3.
Acta Pharmacol Sin ; 42(12): 2132-2143, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33658706

RESUMEN

Continuous docetaxel (DTX) treatment of non-small cell lung cancer induces development of drug resistance, but the mechanism is poorly understood. In this study we performed metabolomics analysis to characterize the metabolic patterns of sensitive and resistant A549 non-small cell lung cancer cells (A549/DTX cells). We showed that the sensitive and resistant A549 cells exhibited distinct metabolic phenotypes: the resistant cells were characterized by an altered microenvironment of redox homeostasis with reduced glutathione and elevated reactive oxygen species (ROS). DTX induction reprogrammed the metabolic phenotype of the sensitive cells, which acquired a phenotype similar to that of the resistant cells: it reduced cystine influx, inhibited glutathione biosynthesis, increased ROS and decreased glutathione/glutathione disulfide (GSH/GSSG); the genes involved in glutathione biosynthesis were dramatically depressed. Addition of the ROS-inducing agent Rosup (25, 50 µg/mL) significantly increased P-glycoprotein expression and reduced intracellular DTX in the sensitive A549 cells, which ultimately acquired a phenotype similar to that of the resistant cells. Supplementation of cystine (1.0 mM) significantly increased GSH synthesis, rebalanced the redox homeostasis of A549/DTX cells, and reversed DTX-induced upregulation of P-glycoprotein, and it markedly improved the effects of DTX and inhibited the growth of A549/DTX in vitro and in vivo. These results suggest that microenvironmental redox homeostasis plays a key role in the acquired resistance of A549 cancer cells to DTX. The enhancement of GSH synthesis by supplementary cystine is a promising strategy to reverse the resistance of tumor cells and has potential for translation in the clinic.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Cistina/uso terapéutico , Docetaxel/uso terapéutico , Homeostasis/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Células A549 , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Antineoplásicos/farmacología , Cistina/farmacología , Docetaxel/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Glutatión/metabolismo , Humanos , Masculino , Ratones Desnudos , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Microambiente Tumoral/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
4.
Acta Pharmacol Sin ; 41(1): 73-81, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31427695

RESUMEN

Kaempferol is a natural flavonol that possesses various pharmacological activities, including anti-arthritis effects, yet the underlying mechanisms remain controversial. To evaluate the anti-arthritis efficacy and the underlying mechanisms of kaempferol, collagen-induced arthritis (CIA) mice were treated with kaempferol intragastrically (200 mg · kg-1 · d-1) and intraperitoneally (20 mg · kg-1 · d-1). Pharmacodynamic and pharmacokinetic studies showed that the oral administration of kaempferol produced distinct anti-arthritis effects in model mice with arthritis in terms of the spleen index, arthritis index, paw thickness, and inflammatory factors; the bioavailability (1.5%, relative to that of the intraperitoneal injection) and circulatory exposure of kaempferol (Cmax = 0.23 ± 0.06 ng/mL) and its primary metabolite kaempferol-3-O-glucuronide (Cmax = 233.29 ± 89.64 ng/mL) were rather low. In contrast, the intraperitoneal injection of kaempferol caused marginal anti-arthritis effects, although it achieved a much higher in vivo exposure. The much higher kaempferol content in the gut implicated a potential mechanism involved in the gut. Analysis of 16S ribosomal RNA revealed that CIA caused imbalance of 14 types of bacteria at the family level, whereas kaempferol largely rebalanced the intestinal microbiota in CIA mice. A metabolomics study showed that kaempferol treatment significantly reversed the perturbation of metabolites involved in energy production and the tryptophan, fatty acid and secondary bile acid metabolisms in the gut contents of the CIA mice. In conclusion, we demonstrate for the first time that the high level of kaempferol in the gut regulates the intestinal flora and microbiotic metabolism, which are potentially responsible for the anti-arthritis activities of kaempferol.


Asunto(s)
Antiinflamatorios/farmacología , Artritis Experimental/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Quempferoles/farmacología , Quempferoles/uso terapéutico , Administración Oral , Animales , Antiinflamatorios/administración & dosificación , Artritis Experimental/inducido químicamente , Artritis Experimental/patología , Autoanticuerpos/análisis , Bovinos , Colágeno Tipo II , Citocinas/análisis , Modelos Animales de Enfermedad , Quempferoles/administración & dosificación , Masculino , Ratones , Ratones Endogámicos DBA
5.
Acta Pharmacol Sin ; 41(1): 129-137, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31341258

RESUMEN

Rheumatoid arthritis patients can be prescribed a combination of immunosuppressive drug leflunomide (LEF) and the antiviral drug acyclovir to reduce the high risk of infection. Acyclovir is a substrate of organic anion transporter (OAT) 1/3 and multidrug resistance-associated protein (MRP) 2. Considering the extraordinarily long half-life of LEF's active metabolite teriflunomide (TER) and the kidney injury risk of acyclovir, it is necessary to elucidate the potential impact of LEF on the disposition of acyclovir. Here we used a specific MRP inhibitor MK571 and probenecid (OAT1/3 and MRP2 inhibitor) to assess the effects of MRP2 and OAT1/3 on the pharmacokinetics and tissue distribution of acyclovir in rats. We showed that LEF and probenecid, but not MK571 significantly increased the plasma concentration of acyclovir. However, kidney and liver exposures of acyclovir were increased when coadministered with LEF, probenecid or MK571. The kidney/plasma ratio of acyclovir was increased to approximately 2-fold by LEF or probenecid, whereas it was increased to as much as 14.5-fold by MK571. Consistently, these drugs markedly decreased the urinary excretion of acyclovir. TER (0.5-100 µmol/L) dose-dependently increased the accumulation of acyclovir in MRP2-MDCK cells with an IC50 value of 4.91 µmol/L. TER (5 µmol/L) significantly inhibited the uptake of acyclovir in hOAT1/3-HEK293 cells. These results suggest that LEF/TER increased the kidney accumulation of acyclovir by inhibiting the efflux transporter MRP2, which increased its kidney/plasma ratio and renal injury risk. However, the inhibitory effects of LEF/TER on OAT1/3 reduced the tubular cells' uptake of acyclovir and increased the plasma concentration.


Asunto(s)
Aciclovir/farmacocinética , Riñón/metabolismo , Leflunamida/farmacología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Proteína 1 de Transporte de Anión Orgánico/antagonistas & inhibidores , Transportadores de Anión Orgánico Sodio-Independiente/antagonistas & inhibidores , Aciclovir/administración & dosificación , Aciclovir/metabolismo , Administración Intravenosa , Animales , Células Cultivadas , Crotonatos/administración & dosificación , Crotonatos/metabolismo , Crotonatos/farmacología , Perros , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Hidroxibutiratos , Leflunamida/administración & dosificación , Leflunamida/metabolismo , Células de Riñón Canino Madin Darby/efectos de los fármacos , Células de Riñón Canino Madin Darby/metabolismo , Masculino , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Nitrilos , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Probenecid/administración & dosificación , Probenecid/metabolismo , Probenecid/farmacología , Propionatos/administración & dosificación , Propionatos/metabolismo , Propionatos/farmacología , Quinolinas/administración & dosificación , Quinolinas/metabolismo , Quinolinas/farmacología , Ratas , Ratas Sprague-Dawley , Distribución Tisular , Toluidinas/administración & dosificación , Toluidinas/metabolismo , Toluidinas/farmacología
6.
Metabolomics ; 15(10): 128, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31541307

RESUMEN

INTRODUCTION: Clinical trials of Compound danshen dripping pills (CDDP) indicated distinct improvement in patients with chronic stable angina. Daily fluctuation of therapeutic effect agreed with a peak-valley PK profile during a 4-week CDDP regimen, but stabilized after 8-week treatment. OBJECTIVES: This article aims to explore the underlying mechanism for the time-dependent drug efficacy of the up-down fluctuation or stabilization in clinic trials. METHODS: A rat model of myocardial ischemia was established via isoproterenol induction. Metabolomics was employed to analyze the energy-related substances both in circulatory system and myocardium in the myocardial ischemia model. RESULTS: CDDP treatment ameliorated myocardial ischemia, reversed the reprogramming of the metabolism induced by ISO and normalized the level of most myocardial substrates and the genes/enzymes associated with those metabolic changes. After 1- or 2-week treatment, CDDP regulated plasma and myocardial metabolome in an analogous, time-dependent way, and modulated metabolic patterns of ischemic rats that perfectly matched with the fluctuated or stabilized effects observed in clinical trials with 4 or 8-week treatment, respectively. CONCLUSION: Metabolic modulation by CDDP contributes to the fluctuated or stabilized therapeutic outcome, and is a potential therapeutic approach for myocardial ischemia diseases.


Asunto(s)
Medicamentos Herbarios Chinos/uso terapéutico , Metabolómica , Isquemia Miocárdica/tratamiento farmacológico , Animales , Canfanos , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Isoproterenol , Masculino , Isquemia Miocárdica/inducido químicamente , Isquemia Miocárdica/metabolismo , Panax notoginseng , Ratas , Ratas Sprague-Dawley , Salvia miltiorrhiza , Factores de Tiempo
7.
Inflammopharmacology ; 27(6): 1193-1203, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31309485

RESUMEN

Paeoniflorin shows distinct anti-arthritis and immunoregulatory activities, but its rather low bioavailability via oral administration greatly challenges its known mechanism of in vivo activity. Our data showed that oral administration, instead of intraperitoneal injection, of paeoniflorin significantly reduced the polyarthritis index by 44.4%, reduced paw swelling by 18.4% and delayed the onset of arthritis in collagen-induced arthritis (CIA) mice. Oral paeoniflorin treatment also downregulated the systemic pro-inflammatory cytokines IL-6 (by 52.2%), TNF-α (by 57.7%) and IL-1ß (by 34.1%). A pharmacokinetic study revealed that the maximal plasma concentration of paeoniflorin after oral administration was 4.8 ± 1.9 µM in the CIA mice, much lower than the effective concentration in vitro (30 µM). In contrast, paeoniflorin was highly concentrated in the gut content, intestine and Peyer's patches. T cell analysis showed that paeoniflorin markedly reduced transcription factors of Th1 and Th17, inhibited Th1 by 22.2% and 23.1% and Th17 by 43.2% and 25.4% (p < 0.05) in the mesenteric lymph node and Peyer's patches, respectively. Paeoniflorin did not have a significant impact on Th1 and Th17 in the spleen. For the first time, these data suggest that paeoniflorin accumulates in the intestine and primarily modulates Th1 and Th17 responses in the mesenteric lymph nodes and Peyer's patches, rather than in the spleen, to exert anti-arthritis effects.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Glucósidos/farmacología , Mucosa Intestinal/efectos de los fármacos , Ganglios Linfáticos/efectos de los fármacos , Monoterpenos/farmacología , Ganglios Linfáticos Agregados/efectos de los fármacos , Células TH1/efectos de los fármacos , Células Th17/efectos de los fármacos , Animales , Citocinas/biosíntesis , Glucósidos/farmacocinética , Glucósidos/uso terapéutico , Mucosa Intestinal/inmunología , Ganglios Linfáticos/inmunología , Masculino , Ratones , Ratones Endogámicos DBA , Monoterpenos/farmacocinética , Monoterpenos/uso terapéutico , Ganglios Linfáticos Agregados/inmunología , Células TH1/inmunología , Células Th17/inmunología
8.
Acta Pharmacol Sin ; 40(1): 86-97, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29930278

RESUMEN

Epalrestat is an inhibitor of aldose reductase in the polyol pathway and is used for the management of diabetic neuropathy clinically. Our pilot experiments and accumulated evidences showed that epalrestat inhibited polyol pathway and reduced sorbitol production, and suggested the potential renal protection effects of epalrestat on diabetic nephropathy (DN). To evaluate the protective effect of epalrestat, the db/db mice were used and exposed to epalrestat for 8 weeks, both the physiopathological condition and function of kidney were examined. For the first time, we showed that epalrestat markedly reduced albuminuria and alleviated the podocyte foot process fusion and interstitial fibrosis of db/db mice. Metabolomics was employed, and metabolites in the plasma, renal cortex, and urine were profiled using a gas chromatography-mass spectrometry (GC/MS)-based metabolomic platform. We observed an elevation of sorbitol and fructose, and a decrease of myo-inositol in the renal cortex of db/db mice. Epalrestat reversed the renal accumulation of the polyol pathway metabolites of sorbitol and fructose, and increased myo-inositol level. Moreover, the upregulation of aldose reductase, fibronectin, collagen III, and TGF-ß1 in renal cortex of db/db mice was downregulated by epalrestat. The data suggested that epalrestat has protective effects on DN, and the inhibition of aldose reductase and the modulation of polyol pathway in nephritic cells be a potentially therapeutic strategy for DN.


Asunto(s)
Aldehído Reductasa/antagonistas & inhibidores , Nefropatías Diabéticas/prevención & control , Inhibidores Enzimáticos/uso terapéutico , Sustancias Protectoras/uso terapéutico , Rodanina/análogos & derivados , Tiazolidinas/uso terapéutico , Albuminuria/tratamiento farmacológico , Animales , Fructosa/sangre , Fructosa/metabolismo , Fructosa/orina , Inositol/sangre , Inositol/metabolismo , Inositol/orina , Riñón/metabolismo , Riñón/patología , Masculino , Metabolómica , Ratones , Rodanina/uso terapéutico , Sorbitol/sangre , Sorbitol/metabolismo , Sorbitol/orina
9.
Acta Pharmacol Sin ; 39(11): 1804-1815, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29921884

RESUMEN

Xuezhikang capsule (XZK) is a traditional Chinese medicine that contains lovastatin (Lv) for hyperlipidemia treatment, although it has fewer side effects than Lv. However, the pharmacokinetic mechanisms contributing to its distinct efficacy and low side effects are unclear. Mice were fed a high-fat diet (HFD) for 6 weeks to induce hyperlipidemia. We first conducted the pharmacokinetic studies in HFD mice following oral administration of Lv (10 mg/kg, i.g.) and found that HFD remarkably decreased the active form of Lv (the lovastatin acid, LvA) exposure in the circulation system, especially in the targeting organ liver, with a declined conversion from Lv to LvA, whereas the Lv (responsible for myotoxicity) exposure in muscle markedly increased. Then we compared the pharmacokinetic profiles of Lv in HFD mice after the oral administration of XZK (1200 mg/kg, i.g.) or an equivalent dose of Lv (10 mg/kg, i.g.). A higher exposure of LvA and lower exposure of Lv were observed after XZK administration, suggesting a pharmacokinetic interaction of some ingredients in XZK. Further studies revealed that HFD promoted the inflammation and inhibited carboxylesterase (CES) activities in the intestine and the liver, thus contributing to the lower transformation of Lv into LvA. In contrast, XZK inhibited the inflammation and upregulated CES in the intestine and the liver. Finally, we evaluated the effects of monacolins and phytosterols, the fractional extracts of isoflavones, on inflammatory LS174T or HepG2 cells, which showed that isoflavones inhibited inflammation, upregulated CES, and markedly enhanced the conversion of Lv into LvA. For the first time, we provide evidence that isoflavones and Lv in XZK act in concert to enhance the efficacy and reduce the side effects of Lv.


Asunto(s)
Medicamentos Herbarios Chinos/uso terapéutico , Hiperlipidemias/tratamiento farmacológico , Isoflavonas/farmacología , Lovastatina/análogos & derivados , Lovastatina/uso terapéutico , Administración Oral , Animales , Carboxilesterasa/genética , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/metabolismo , Medicamentos Herbarios Chinos/farmacocinética , Humanos , Inflamación/tratamiento farmacológico , Lovastatina/administración & dosificación , Lovastatina/metabolismo , Lovastatina/farmacocinética , Masculino , Ratones Endogámicos C57BL , Receptor X de Pregnano/genética , Regulación hacia Arriba/efectos de los fármacos
10.
Acta Pharmacol Sin ; 39(10): 1670-1680, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29770798

RESUMEN

Apatinib, a small-molecule inhibitor of VEGFR-2, has attracted much attention due to its encouraging anticancer activity in third-line clinical treatment for many malignancies, including non-small cell lung cancer (NSCLC). Its usage in second-line therapy with chemotherapeutic drugs is still under exploration. In this study we investigated the antitumor effect of apatinib combined with docetaxel against NSCLC and its cellular pharmacokinetic basis. A549 xenograft nude mice were treated with apatinib (100 mg/kg every day for 20 days) combined with docetaxel (8 mg/kg, ip, every four days for 5 times). Apatinib significantly enhanced the antitumor effect of docetaxel and alleviated docetaxel-induced liver damage as well as decreased serum transaminases (ALT and AST). LC-MS/MS analysis revealed that apatinib treatment significantly increased the docetaxel concentration in tumors (up to 1.77 times) without enhancing the docetaxel concentration in the serum, heart, liver, lung and kidney. Furthermore, apatinib decreased docetaxel-induced upregulation of P-glycoprotein in tumors. The effects of apatinib on the uptake, efflux and subcellular distribution of docetaxel were investigated in A549 and A549/DTX (docetaxel-resistant) cells in vitro. A cellular pharmacokinetic study revealed that apatinib significantly increased cellular/subcellular accumulation (especially in the cytosol) and decreased the efflux of docetaxel in A549/DTX cells through P-gp, while apatinib exerted no significant effect on the cellular pharmacokinetics of docetaxel in A549 cells. Consequently, the IC50 value of docetaxel in A549/DTX cells was more significantly decreased by apatinib than that in A549 cells. These results demonstrate that apatinib has potential for application in second-line therapy combined with docetaxel for NSCLC patients, especially for docetaxel-resistant or multidrug-resistant patients.


Asunto(s)
Docetaxel/uso terapéutico , Piridinas/uso terapéutico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Línea Celular Tumoral , Docetaxel/farmacocinética , Sinergismo Farmacológico , Humanos , Hígado/efectos de los fármacos , Masculino , Ratones Desnudos , Sustancias Protectoras/farmacocinética , Sustancias Protectoras/uso terapéutico , Piridinas/farmacocinética , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Chem Biol Interact ; 289: 15-22, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29689254

RESUMEN

The use of Chinese herbal medicines and natural products has become increasingly popular in both China and Western societies as an alternative medicine for the treatment of diseases or as a health supplement. Danshen, the dried root of Salvia miltiorrhiza (Fam.Labiatae), which is rich in phenolic acids and tanshinones, is a widely used herbal medicine for the treatment of cardio-cerebrovascular diseases. The goal of this study was to examine the inhibitory effects of fifteen components derived from Danshen on CYP2C8 and CYP2J2, which are expressed both in human liver and cardiovascular systems. Recombinant CYP2C8 and CYP2J2 were used, and the mechanism, kinetics, and type of inhibition were determined. Taxol 6-hydroxylation and astemizole O-desmethyastemizole were determined as probe activities for CYP2C8 and CYP2J2, respectively. Metabolites formations were analyzed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). The results demonstrated that salvianolic acid A was a competitive inhibitor of CYP2C8 (Ki = 2.5 µM) and mixed-type inhibitor of CYP2J2 (Ki = 7.44 µM). Salvianolic acid C had moderate noncompetitive and mixed-type inhibitions on CYP2C8 (Ki = 4.82 µM) and CYP2J2 (Ki = 5.75 µM), respectively. Tanshinone IIA was a moderate competitive inhibitor of CYP2C8 (Ki = 1.18 µM). Dihydrotanshinone I had moderate noncompetitive inhibition on CYP2J2 (Ki = 6.59 µM), but mechanism-based inhibition on CYP2C8 (KI = 0.43 µM, kinact = 0.097 min-1). Tanshinone I was a moderate competitive inhibitor of CYP2C8 (Ki = 4.20 µM). These findings suggested that Danshen preparations appear not likely to pose a significant risk of drug interactions mediated by CYP2C8 after oral administration; but their inhibitory effects on intestinal CYP2J2 mediated drug metabolism should not be neglected when they are given orally in combination with other drugs. Additionally, this study provided novel insights into the underling pharmacological mechanisms of Danshen components from the perspective of CYP2C8 and CYP2J2 inhibition.


Asunto(s)
Citocromo P-450 CYP2C8/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Medicamentos Herbarios Chinos/farmacología , Citocromo P-450 CYP2J2 , Inhibidores Enzimáticos del Citocromo P-450/química , Medicamentos Herbarios Chinos/química , Humanos , Concentración 50 Inhibidora , Cinética , Proteínas Recombinantes/metabolismo , Salvia miltiorrhiza , Taxoides/metabolismo , Factores de Tiempo
12.
Acta Pharmacol Sin ; 38(11): 1435-1444, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28836583

RESUMEN

Salvianolic acid A (SAA), a water-soluble phenolic acid isolated from the root of Dan Shen, displays distinct antioxidant activity and effectiveness in protection against cerebral ischemia/reperfusion (I/R) damage. However, whether SAA can enter the central nervous system and exert its protective effects by directly targeting brain tissue remains unclear. In this study, we evaluated the cerebral protection of SAA in rats subjected to transient middle cerebral artery occlusion (tMCAO) followed by reperfusion. The rats were treated with SAA (5, 10 mg/kg, iv) when the reperfusion was performed. SAA administration significantly decreased cerebral infarct area and the brain water content, attenuated the neurological deficit and pathology, and enhanced the anti-inflammatory and antioxidant capacity in tMCAO rats. The concentration of SAA in the plasma and brain was detected using LC-MS/MS. A pharmacokinetic study revealed that the circulatory system exposure to SAA was equivalent in the sham controls and I/R rats, but the brain exposure to SAA was significantly higher in the I/R rats than in the sham controls (fold change of 9.17), suggesting that the enhanced exposure to SAA contributed to its cerebral protective effect. Using a GC/MS-based metabolomic platform, metabolites in the serum and brain tissue were extracted and profiled. According to the metabolomic pattern of the tissue data, SAA administration significantly modulated the I/R-caused perturbation of metabolism in the brain to a greater extent than that in the serum, demonstrating that SAA worked at the brain tissue level rather than the whole circulation system. In conclusion, a larger amount of SAA enters the central nervous system in ischemia/reperfusion rats to facilitate its protective and regulatory effects on the perturbed metabolism.


Asunto(s)
Encéfalo/efectos de los fármacos , Ácidos Cafeicos/farmacocinética , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Lactatos/farmacocinética , Metabolómica/métodos , Fármacos Neuroprotectores/farmacocinética , Daño por Reperfusión/prevención & control , Animales , Disponibilidad Biológica , Encéfalo/metabolismo , Encéfalo/patología , Ácidos Cafeicos/administración & dosificación , Ácidos Cafeicos/sangre , Cromatografía Liquida , Citoprotección , Modelos Animales de Enfermedad , Cromatografía de Gases y Espectrometría de Masas , Infarto de la Arteria Cerebral Media/sangre , Infarto de la Arteria Cerebral Media/patología , Inyecciones Intravenosas , Lactatos/administración & dosificación , Lactatos/sangre , Masculino , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/sangre , Ratas Sprague-Dawley , Daño por Reperfusión/sangre , Daño por Reperfusión/patología , Espectrometría de Masas en Tándem
13.
J Pharm Biomed Anal ; 142: 190-200, 2017 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-28514718

RESUMEN

Cerebral ischemia-reperfusion (I/R) injury usually contributes to mortality and disability after ischemic stroke. Ginkgolides injection (GIn), a standard preparation composed of ginkgo diterpene lactones extract, is clinically used for neuroprotective treatment on reconvalescents of cerebral infarction. However, the understanding about its therapeutic mechanism is still lacking. In this study, a gas chromatography-mass spectrometry (GC-MS) based metabolomic approach coupled with multivariate data analysis (MVDA) was applied to explore the neuroprotective effects of GIn in a rodent model of focal ischemic stroke induced by transient middle cerebral artery occlusion (tMCAO). Metabolomic profiling revealed a series of metabolic perturbations that underlie the cerebral I/R pathological events. GIn can reverse the I/R induced brain metabolic deviations by modulating multiple metabolic pathways, such as glycolysis, Krebs cycle, pentose phosphate pathway (PPP), γ-aminobutyrate (GABA) shunt and lipid metabolism. Moreover, the main bioactive components of GIn were distributed to brain tissue much more easily in tMCAO rats than in normal rats after an intravenous administration, suggesting that the increased cerebral exposure to ginkgolides in I/R pathological condition potentially facilitated the neuroprotective effects of GIn by directly targeting at brain. The present study provided valuable information for our understanding about metabolic changes of cerebral I/R injury and clinical application of GIn.


Asunto(s)
Isquemia Encefálica , Animales , Cromatografía de Gases y Espectrometría de Masas , Ginkgólidos , Infarto de la Arteria Cerebral Media , Fármacos Neuroprotectores , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión
14.
Acta Pharmacol Sin ; 38(3): 351-361, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28042874

RESUMEN

Berberrubine (BRB) is the primary metabolite of berberine (BBR) that has shown a stronger glucose-lowering effect than BBR in vivo. On the other hand, BRB is quickly and extensively metabolized into berberrubine-9-O-ß-D-glucuronide (BRBG) in rats after oral administration. In this study we compared the pharmacokinetic properties of BRB and BRBG in rats, and explored the mechanisms underlying their glucose-lowering activities. C57BL/6 mice with HFD-induced hyperglycemia were administered BRB (50 mg·kg-1·d-1, ig) for 6 weeks, which caused greater reduction in the plasma glucose levels than those caused by BBR (120 mg·kg-1·d-1) or BRB (25 mg·kg-1·d-1). In addition, BRB dose-dependently decreased the activity of α-glucosidase in gut of the mice. After oral administration of BRB in rats, the exposures of BRBG in plasma at 3 different dosages (10, 40, 80 mg/kg) and in urine at different time intervals (0-4, 4-10, 10-24 h) were dramatically greater than those of BRB. In order to determine the effectiveness of BRBG in reducing glucose levels, we prepared BRBG from the urine pool of rats, and identified and confirmed it through LC-MS-IT-TOF and NMR spectra. In human normal liver cell line L-O2 in vitro, treatment with BRB or BRBG (5, 20, 50 µmol/L) increased glucose consumption, enhanced glycogenesis, stimulated the uptake of the glucose analog 2-NBDG, and modulated the mRNA levels of glucose-6-phosphatase and hexokinase. However, both BBR and BRB improved 2-NBDG uptake in insulin-resistant L-O2 cells, while BRBG has no effect. In conclusion, BRB exerts a stronger glucose-lowering effect than BBR in HFD-induced hyperglycemia mice. Although BRB significantly stimulated the insulin sensitivity and glycolysis in vitro, BRBG may have a greater contribution to the glucose-lowering effect because it has much greater system exposure than BRB after oral administration of BRB. The results suggest that BRBG is a potential agent for reducing glucose levels.


Asunto(s)
Berberina/análogos & derivados , Glucurónidos/uso terapéutico , Hiperglucemia/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Animales , Berberina/administración & dosificación , Berberina/sangre , Berberina/metabolismo , Berberina/farmacocinética , Berberina/uso terapéutico , Berberina/orina , Glucurónidos/sangre , Glucurónidos/orina , Humanos , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacocinética , Masculino , Ratones Endogámicos C57BL , Ratas Sprague-Dawley
15.
Sci Rep ; 6: 31364, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27527657

RESUMEN

Guizhi Fuling capsule (GZFL), a traditional Chinese medicine formulation, is widely used in China to relieve pain from dysmenorrhea and is now in a Phase II clinical trial in the USA. Due to the low exposure of the five main medicative ingredients (amygdalin, cinnamic acid, gallic acid, paeoniflorin and paeonol) of GZFL in human, a strategy was built to qualitatively and quantitatively identify the possible metabolites of GZFL and to describe the pharmacokinetic profiles of GZFL in human. In this strategy, LC-Q-TOF/MS was used to identify and structurally elucidate the possible metabolites of GZFL in vivo; and a time-based metabolite-confirming step (TBMCs) was used to confirm uncertain metabolites. The simultaneously quantitation results by LC-MS/MS showed low exposure of the five medicative ingredients. According to the strategy we built, a total of 36 metabolites were found and structurally elucidated. The simultaneously semi-quantitative analysis by LC-MS/MS showed that obvious time-concentration curves could be established for 12 of the metabolites, and most of them showed a relatively higher exposure. This study provides a better understanding of the metabolic processes of GZFL in human.


Asunto(s)
Medicamentos Herbarios Chinos/farmacocinética , Acetofenonas/administración & dosificación , Acetofenonas/química , Acetofenonas/farmacocinética , Amigdalina/administración & dosificación , Amigdalina/química , Amigdalina/farmacocinética , Cápsulas , Cromatografía Líquida de Alta Presión/métodos , Cinamatos/administración & dosificación , Cinamatos/química , Cinamatos/farmacocinética , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/química , Femenino , Ácido Gálico/administración & dosificación , Ácido Gálico/química , Ácido Gálico/farmacocinética , Glucósidos/administración & dosificación , Glucósidos/química , Glucósidos/farmacocinética , Voluntarios Sanos , Humanos , Masculino , Estructura Molecular , Monoterpenos/administración & dosificación , Monoterpenos/química , Monoterpenos/farmacocinética , Espectrometría de Masas en Tándem/métodos
16.
Artículo en Inglés | MEDLINE | ID: mdl-26657802

RESUMEN

Traditional Chinese herb medicines (TCHMs) have been used in the treatment of a variety of diseases for thousands of years in Asian countries. The active components of TCHMs usually exert combined synergistic therapeutic effects on multiple targets, but with less potential therapeutic effect based on routine indices than Western drugs. These complex effects make the assessment of the efficacy of TCHMs and the clarification of their underlying mechanisms very challenging, and therefore hinder their wider application and acceptance. Metabolomics is a crucial part of systems biology. It allows the quantitative measurement of large numbers of the low-molecular endogenous metabolites involved in metabolic pathways, and thus reflects the fundamental metabolism status of the body. Recently, dozens of metabolomic studies have been devoted to prove the efficacy/safety, explore the underlying mechanisms, and identify the potential biomarkers to access the action targets of TCHMs, with fruitful results. This article presents an overview of these studies, focusing on the progress made in exploring the pharmacology and toxicology of various herbal medicines.


Asunto(s)
Medicina Tradicional China , Metabolómica
17.
Acta Pharmacol Sin ; 36(12): 1462-72, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26592515

RESUMEN

AIM: Xuezhikang (XZK), an extract of red yeast rice, has been widely used in traditional Chinese medicine to treat cardiovascular disease. Three fractions F1, F2 and F3 (primarily containing isoflavones, monacolins or phytosterols, respectively) are extracted from Xuezhikang capsules. In this study we evaluated the lipid-lowering effects of these fractions and explored the potential mechanisms of actions. METHODS: Mice treated with a high-fat diet (HFD) were orally administered lovastatin (10 mg·kg(-1)·d(-1)), XZK (1200 mg·kg(-1)·d(-1)), F1 (27.5 mg·kg(-1)·d(-1)), F2 (11.3 mg·kg(-1)·d(-1)) or F3 (35 mg·kg(-1)·d(-1)) for 10 weeks. Lipids were measured using commercial enzymatic kits, and the mRNA and protein levels of genes involved in cholesterol and bile acid homeostasis were evaluated using qRT-PCR and Western blot analysis, respectively. RESULTS: XZK increased the fecal excretion of lipids and bile acids, reduced serum TC, TG and LDL-C levels by 40%, 55% and 46%, respectively, and increased serum HDL-C by 31%. Administration of F1 repressed serum TC and TG by 24% and 52%, respectively, and elevated hepatic synthesis of CYP7A1. It also increased hepatic elimination of bile acids in the fecal excretions by 79% through upregulating BSEP and downregulating NTCP. Administration of F3 decreased serum TC, TG and LDL-C levels by 33%, 29% and 39%, respectively, and increased serum HDL-C by 28%, significantly reduced intestinal absorption of cholesterol by inhibiting the transcription of NPC1L1, and elevated excretion of TC, FC and CE by 96%, 72% and 101%, respectively. Administration of F2 showed pharmacological effects similar to those of lovastatin. CONCLUSION: Isoflavones and phytosterols in XZK exert cholesterol-lowering effects in HFD mice through mechanisms that differ from those of lovastatin. Isoflavones and phytosterols act in a complimentary manner: through enhancing the elimination of bile acids and reducing intestinal cholesterol absorption, respectively.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Colesterol/metabolismo , Dieta Alta en Grasa , Medicamentos Herbarios Chinos/farmacología , Hipolipemiantes/farmacología , Isoflavonas/farmacología , Fitosteroles/farmacología , Animales , Ácidos y Sales Biliares/genética , Cápsulas , Colesterol/sangre , Colesterol/genética , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/química , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis/efectos de los fármacos , Hipolipemiantes/química , Isoflavonas/administración & dosificación , Isoflavonas/química , Metabolismo de los Lípidos/efectos de los fármacos , Lípidos/sangre , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Fitosteroles/administración & dosificación , Fitosteroles/química
18.
Acta Pharmacol Sin ; 36(2): 259-67, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25619394

RESUMEN

AIM: The pentose phosphate pathway (PPP) is involved in the activity of glucose-6-phosphate dehydrogenase (G6PD) and generation of NADPH, which plays a key role in drug metabolism. The aim of this study was to investigate the effects of modulation of the PPP on drug metabolism capacity in vitro. METHODS: A pair of hepatic cell lines, ie, the cancerous HepG2 cells and normal L02 cells, was used. The expression of CYP450 enzymes, p53 and G6PD in the cells were analyzed. The metabolism of testosterone (TEST, 10 µmol/L) and dextromethorphan (DEM, 1 µmol/L), the two typical substrates for CYP3A4 and CYP2D6, in the cells was examined in the presence of different agents. RESULTS: Both the expression and metabolic activities of CYP3A4 and CYP2D6 were considerably higher in HepG2 cells than in L02 cells. The metabolism of TEST and DEM in HepG2 cells was dose-dependently inhibited by the specific CYP3A4 inhibitor ketoconazole and CYP2D6 inhibitor quinidine. Addition of the p53 inhibitor cyclic PFT-α (5, 25 µmol/L) in HepG2 cells dose-dependently enhanced the metabolism of DEM and TEST, whereas addition of the p53 activator NSC 66811 (3, 10, 25 µmol/L) dose-dependently inhibited the metabolism. Furthermore, addition of the G6PD inhibitor 6-aminonicotinamide (5, 15 µmol/L) in HepG2 cells dose-dependently inhibited the metabolism of DEM and TEST, whereas addition of the PPP activity stimulator menadione (1, 5, 15 µmol/L) dose-dependently enhanced the metabolism. CONCLUSION: Modulation of p53 and the PPP alters the metabolism of DEM and TEST, suggesting that the metabolic flux pattern of PPP may be closely involved in drug metabolism and the individual variance.


Asunto(s)
Dextrometorfano/metabolismo , Fase I de la Desintoxicación Metabólica/fisiología , Vía de Pentosa Fosfato/fisiología , Testosterona/metabolismo , Línea Celular Tumoral , Citocromo P-450 CYP3A/metabolismo , Células Hep G2 , Humanos , Hígado/enzimología , Hígado/metabolismo
19.
Acta Pharmacol Sin ; 34(10): 1349-58, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23892274

RESUMEN

AIM: 20(S)-Ginsenoside Rh2 (Rh2) has shown potent inhibition on P-glycoprotein (P-gp), while most HIV protease inhibitors are both substrates and inhibitors of P-gp and CYP3A4. The aim of this study was to investigate the potential pharmacokinetic interactions between Rh2 and the HIV protease inhibitor ritonavir. METHODS: The effects of Rh2 on the cellular accumulation and transepithelial transport of ritonavir were studied in Caco-2 and MDCK-MDR1 cells. Male rats were administered Rh2 (25 or 60 mg/kg, po) or Rh2 (5 mg/kg, iv), followed by ritonavir (25 mg/kg, po). The P-gp inhibitors verapamil (20 mg/kg, po) or GF120918 (5 mg/kg, po) were used as positive controls. The concentrations of ritonavir in plasma, bile, urine, feces and tissue homogenates were analyzed using LC-MS. RESULTS: Rh2 (10 µmol/L) significantly increased the accumulation and inhibited the efflux of ritonavir in Caco-2 and MDCK-MDR1 cells, as verapamil did. But Rh2 did not significantly alter ritonavir accumulation or transport in MDCK-WT cells. Intravenous Rh2 significantly increased the plasma exposure of ritonavir while reducing its excretion in the bile, and oral verapamil or GF120918 also increased plasma exposure of ritonavir but without changing its excretion in the bile. Interestingly, oral Rh2 at both doses did not significantly change the plasma profile of ritonavir. Moreover, oral Rh2 (25 mg/kg) significantly elevated the ritonavir concentration in the hepatic portal vein, and markedly increased its urinary excretion and tissue distribution, which might counteract the elevated absorption of ritonavir. CONCLUSION: Rh2 inhibits the efflux of ritonavir through P-gp in vitro. The effects of Rh2 on ritonavir exposure in vivo depend on the administration route of Rh2: intravenous, but not oral, administration of Rh2 significantly increased the plasma exposure of ritonavir.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Ginsenósidos/farmacocinética , Inhibidores de la Proteasa del VIH/farmacocinética , Ritonavir/farmacocinética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Acridinas/farmacología , Administración Oral , Animales , Células CACO-2 , Cromatografía Liquida , Perros , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Ginsenósidos/administración & dosificación , Ginsenósidos/farmacología , Inhibidores de la Proteasa del VIH/administración & dosificación , Inhibidores de la Proteasa del VIH/farmacología , Humanos , Inyecciones Intravenosas , Células de Riñón Canino Madin Darby , Masculino , Espectrometría de Masas , Ratas , Ratas Wistar , Ritonavir/administración & dosificación , Ritonavir/farmacología , Tetrahidroisoquinolinas/farmacología , Distribución Tisular , Verapamilo/farmacología
20.
Acta Pharmacol Sin ; 31(8): 930-7, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20686518

RESUMEN

AIM: To investigate the regulatory effects of total ginsenosides and the conventional antihypertensive agents (captopril, amlodipine, terazosin and hydrochlorothiazide) on the blood pressure and perturbed metabolism in spontaneously hypertensive rats (SHRs) and to analyze the cause-effect relationships between high blood pressure and the metabolic disorders of hypertension. METHODS: SHRs were administrated with total ginsenosides or the antihypertensive agents for eight weeks. Systolic blood pressure (SP) was measured every week and low-molecular-weight compounds in blood plasma were quantitatively analyzed using a nontargeted high-throughput metabolomic tool: gas chromatography/time of flight mass spectrometry (GC/TOFMS) . The metabolic patterns were evaluated using principal components analysis and potential markers of hypertension were identified. RESULTS: Total ginsenosides and the antihypertensive agents differentially regulated SP and the metabolic pattern in SHRs. Total ginsenosides caused a progressive and prolonged reduction of SP and markedly normalized the perturbed metabolism with 14 of 27 (51.8%) markers of hypertension which were regulated toward normal. Total ginsenosides also reduced free fatty acids' level toward normal levels. In contrast, captopril, amlodipine and terazosin efficiently depressed SP, but had little effect on metabolic perturbation with only 8 (29.6%), 4 (14.8%), and 4 (14.8%) markers, respectively, which were regulated. CONCLUSION: The metabolic changes persisted when the blood pressure was lowered by the conventional antihypertensive agents, suggesting that hypertension may not be the cause of the metabolic perturbation in SHRs.


Asunto(s)
Antihipertensivos/farmacología , Presión Sanguínea/efectos de los fármacos , Ginsenósidos/farmacología , Hipertensión/tratamiento farmacológico , Animales , Ácidos Grasos no Esterificados/metabolismo , Cromatografía de Gases y Espectrometría de Masas/métodos , Hipertensión/fisiopatología , Masculino , Ratas , Ratas Endogámicas SHR
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...