Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 160
Filtrar
1.
Mol Pharm ; 21(5): 2284-2297, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38529622

RESUMEN

Organic anion-transporting polypeptides (OATP) 1B1 and OATP1B3, encoded by the SLCO gene family of the solute carrier superfamily, are involved in the disposition of many exogenous and endogenous compounds. Preclinical rodent models help assess risks of pharmacokinetic interactions, but interspecies differences in transporter orthologs and expression limit direct clinical translation. An OATP1B transgenic mouse model comprising a rodent Slco1a/1b gene cluster knockout and human SLCO1B1 and SLCO1B3 gene insertions provides a potential physiologically relevant preclinical tool to predict pharmacokinetic interactions. Pharmacokinetics of exogenous probe substrates, pitavastatin and pravastatin, and endogenous OATP1B biomarkers, coproporphyrin-I and coproporphyrin-III, were determined in the presence and absence of known OATP/Oatp inhibitors, rifampin or silymarin (an extract of milk thistle [Silybum marianum]), in wild-type FVB mice and humanized OATP1B mice. Rifampin increased exposure of pitavastatin (4.6- and 2.8-fold), pravastatin (3.6- and 2.2-fold), and coproporphyrin-III (1.6- and 2.1-fold) in FVB and OATP1B mice, respectively, but increased coproporphyrin-I AUC0-24h only (1.8-fold) in the OATP1B mice. Silymarin did not significantly affect substrate AUC, likely because the silymarin flavonolignan concentrations were at or below their reported IC50 values for the relevant OATPs/Oatps. Silymarin increased the Cmax of pitavastatin 2.7-fold and pravastatin 1.9-fold in the OATP1B mice. The data of the OATP1B mice were similar to those of the pitavastatin and pravastatin clinical data; however, the FVB mice data more closely recapitulated pitavastatin clinical data than the data of the OATP1B mice, suggesting that the OATP1B mice are a reasonable, though costly, preclinical strain for predicting pharmacokinetic interactions when doses are optimized to achieve clinically relevant plasma concentrations.


Asunto(s)
Interacciones Farmacológicas , Transportador 1 de Anión Orgánico Específico del Hígado , Ratones Transgénicos , Pravastatina , Rifampin , Silimarina , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos , Animales , Rifampin/farmacocinética , Ratones , Transportador 1 de Anión Orgánico Específico del Hígado/genética , Transportador 1 de Anión Orgánico Específico del Hígado/metabolismo , Humanos , Silimarina/farmacocinética , Pravastatina/farmacocinética , Pravastatina/administración & dosificación , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos/genética , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos/metabolismo , Quinolinas/farmacocinética , Coproporfirinas/metabolismo , Masculino , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo
2.
Int Immunopharmacol ; 97: 107715, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33933848

RESUMEN

Interferon beta (IFN-ß) has successfully been experimented with to treat multiple sclerosis (MS). However, patients sometimes do not respond effectively to treatment, and |adverse effects, including liver toxicity, accompany this therapy. |Accordingly, we decided to treat MS patients simultaneously with Silymarin (SM) as an immunomodulatory and hepatoprotective agent and IFN-ß in a clinical trial study. Complete blood count (CBC), liver enzyme levels, and the serum concentration of inflammatory and anti-inflammatory cytokines were measured. Also, the frequency of immune cells was determined by flow cytometry. Liver enzyme levels were significantly lower in the intervention group (p < 0.05). The percentage of Th17 cells in the intervention group was significantly reduced compared to the placebo group (P < 0.001). Also, the frequency of Treg cells after treatment with SM plus IFN-ß was significantly increased compared to the placebo group (p < 0.05). Furthermore, the IL-17 and IFNγ cytokine levels were significantly reduced in the intervention group (p < 0.05). Moreover, the levels of anti-inflammatory cytokines IL-10 and TGFß were significantly increased in the intervention group (P < 0.05).Overall, the results provide novel and supplementary information on SM's notable immunoregulatory effects on inflammatory response and liver function in MS patients. Clinical Trial Identifier Number: IRCTID: IRCT20171220037977N1.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Interferón beta/efectos adversos , Esclerosis Múltiple/tratamiento farmacológico , Silimarina/administración & dosificación , Adulto , Enfermedad Hepática Inducida por Sustancias y Drogas/diagnóstico , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Método Doble Ciego , Sinergismo Farmacológico , Quimioterapia Combinada/efectos adversos , Quimioterapia Combinada/métodos , Femenino , Humanos , Interferón beta/farmacocinética , Pruebas de Función Hepática , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/inmunología , Placebos/administración & dosificación , Silimarina/farmacocinética , Resultado del Tratamiento
3.
Molecules ; 25(12)2020 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-32599946

RESUMEN

Silybin is a flavonolignan extracted from Silybum marianum with chemopreventive activity against various cancers, including breast. This study was designed to develop an HPLC-MS/MS method for the determination of silybin in human plasma, urine and breast tissue in early breast cancer patients undergoing Siliphos® supplementation, an oral silybin-phosphatidylcholine complex. The determination of silybin was carried out by liquid-liquid extraction with methyl-tert-butyl ether (MTBE); total silybin concentration was determined by treating the samples with ß-glucuronidase, while for the determination of free silybin, the hydrolytic step was omitted. Naringenin and naproxen were selected as internal standards. The detection of the analyte was carried out by mass spectrometry and by chromatography. The HPLC-MS/MS method was evaluated in terms of selectivity, linearity, limit of quantification, precision and accuracy, and carryover. The method proved to be selective, linear, precise and accurate for the determination of silybin. To the best of our knowledge, this presents the first analytical method with the capacity to quantify the major bioactive components of milk thistle in three different biological matrices with a lower limit of quantification of 0.5 ng/mL for plasma. Silybin phosphatidylcholine, taken orally, can deliver high blood concentrations of silybin, which selectively accumulates in breast tumor tissue.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Silibina/análisis , Espectrometría de Masas en Tándem/métodos , Neoplasias de la Mama/química , Calibración , Femenino , Humanos , Límite de Detección , Extracción Líquido-Líquido , Fosfatidilcolinas/administración & dosificación , Fosfatidilcolinas/farmacocinética , Reproducibilidad de los Resultados , Silibina/sangre , Silibina/orina , Silimarina/administración & dosificación , Silimarina/farmacocinética , Solventes/química
4.
Adv Ther ; 37(4): 1279-1301, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32065376

RESUMEN

Silymarin, an extract from milk thistle seeds, has been used for centuries to treat hepatic conditions. Preclinical data indicate that silymarin can reduce oxidative stress and consequent cytotoxicity, thereby protecting intact liver cells or cells not yet irreversibly damaged. Eurosil 85® is a proprietary formulation developed to maximize the oral bioavailability of silymarin. Most of the clinical research on silymarin has used this formulation. Silymarin acts as a free radical scavenger and modulates enzymes associated with the development of cellular damage, fibrosis and cirrhosis. These hepatoprotective effects were observed in clinical studies in patients with alcoholic or non-alcoholic fatty liver disease, including patients with cirrhosis. In a pooled analysis of trials in patients with cirrhosis, silymarin treatment was associated with a significant reduction in liver-related deaths. Moreover, in patients with diabetes and alcoholic cirrhosis, silymarin was also able to improve glycemic parameters. Patients with drug-induced liver injuries were also successfully treated with silymarin. Silymarin is generally very well tolerated, with a low incidence of adverse events and no treatment-related serious adverse events or deaths reported in clinical trials. For maximum benefit, treatment with silymarin should be initiated as early as possible in patients with fatty liver disease and other distinct liver disease manifestations such as acute liver failure, when the regenerative potential of the liver is still high and when removal of oxidative stress, the cause of cytotoxicity, can achieve the best results.


Asunto(s)
Hepatopatías/tratamiento farmacológico , Sustancias Protectoras/farmacología , Sustancias Protectoras/uso terapéutico , Silimarina/farmacología , Silimarina/uso terapéutico , Glucemia/efectos de los fármacos , Diabetes Mellitus/fisiopatología , Hepatocitos , Humanos , Cirrosis Hepática/tratamiento farmacológico , Hepatopatías Alcohólicas/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Sustancias Protectoras/administración & dosificación , Sustancias Protectoras/farmacocinética , Silimarina/administración & dosificación , Silimarina/farmacocinética
5.
Int J Pharm ; 572: 118838, 2019 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-31715362

RESUMEN

Silymarin (SLM) is a mixture of flavonolignans extracted from the fruit of Silybum marianum L. Gaertn. which has been used for decades as a hepatoprotector. Silymarin has recently been proposed to be beneficial in type 2 diabetic patients. Constituents of SLM are poorly water-soluble and low permeable compounds, with consequently limited oral bioavailability. This study aimed to investigate the possibility of delivery of SLM via nanostructured lipid carriers (NLCs) to overcome these issues and for preparation of an oral dosage form. NLCs were prepared through an emulsion/evaporation/solidifying method. Cetyl palmitate:Lauroglycol 90 was selected as the lipid mixture and Brij S20 as surfactant. NLCs were chemically and physically characterized. Encapsulation efficiency was more than 92%. The storage stability of the NLC suspension was also investigated and the freeze-drying process was taken into consideration. After assessing the stability of the formulation in a simulated gastrointestinal environment, the release of SLM was monitored in different pH conditions. In vitro experiments with artificial membranes (PAMPA) and Caco-2 cells revealed that the NLCs enhanced the permeation of SLM. Active processes are involved in the internalization of NLCs, as evidenced by cellular uptake studies. After preliminary toxicological studies, the formulation was studied in vivo in a streptozotocin (STZ)-induced diabetic mouse model in the presence of metabolic syndrome. The formulation was also compared to an NLC containing stearic acid:Capryol 90, to evaluate the effect of the lipid matrix on the in vivo performance of nanocarriers. Finally, hepatic histopathological analyses were also conducted. Both SLM-loaded NLCs exhibited in vivo a significant down-regulation of blood glucose and triglyceride levels better than free SLM, with a liver-protective effect. Furthermore, both formulations showed a significant anti-hyperalgesic effect on STZ-induced neuropathy.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Síndrome Metabólico/tratamiento farmacológico , Silimarina/administración & dosificación , Administración Oral , Animales , Glucemia/efectos de los fármacos , Células CACO-2 , Química Farmacéutica , Diabetes Mellitus Experimental/tratamiento farmacológico , Portadores de Fármacos/química , Estabilidad de Medicamentos , Almacenaje de Medicamentos , Liofilización , Humanos , Lípidos/química , Masculino , Ratones , Silimarina/farmacocinética , Silimarina/farmacología , Solubilidad
6.
J Pharmacol Exp Ther ; 371(2): 385-393, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31420525

RESUMEN

Patients with nonalcoholic steatohepatitis (NASH) exhibit altered hepatic protein expression of metabolizing enzymes and transporters and altered xenobiotic pharmacokinetics. The botanical natural product silymarin, which has been investigated as a treatment of NASH, contains flavonolignans that inhibit organic anion-transporting polypeptide (OATP) transporter function. The purpose of this study was to assess the individual and combined effects of NASH and silymarin on the disposition of the model OATP substrate pitavastatin. Male Sprague Dawley rats were fed a control or a methionine- and choline-deficient diet (NASH model) for 8 weeks. Silymarin (10 mg/kg) or vehicle followed by pitavastatin (0.5 mg/kg) were administered intravenously, and the pharmacokinetics were determined. NASH increased mean total flavonolignan area under the plasma concentration-time curve (AUC0-120 min) 1.7-fold. Silymarin increased pitavastatin AUC0-120 min in both control and NASH animals approx. 2-fold. NASH increased pitavastatin plasma concentrations from 2 to 40 minutes, but AUC0-120 min was unchanged. The combination of silymarin and NASH had the greatest effect on pitavastatin AUC0-120 min, which increased 2.9-fold compared with control vehicle-treated animals. NASH increased the total amount of pitavastatin excreted into the bile 2.7-fold compared with control animals, whereas silymarin decreased pitavastatin biliary clearance approx. 3-fold in both control and NASH animals. This double hit of NASH and silymarin on hepatic uptake transporters is another example of a multifactorial pharmacokinetic interaction that may have a greater impact on drug disposition than each hit alone. SIGNIFICANCE STATEMENT: Multifactorial effects on xenobiotic pharmacokinetics are within the next frontier for precision medicine research and clinical application. The combination of silymarin and NASH is a probable clinical scenario that can affect drug uptake, liver concentrations, biliary elimination, and ultimately, efficacy and toxicity.


Asunto(s)
Antioxidantes/farmacocinética , Productos Biológicos/farmacocinética , Modelos Animales de Enfermedad , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Transportadores de Anión Orgánico/metabolismo , Silimarina/farmacocinética , Animales , Antioxidantes/uso terapéutico , Productos Biológicos/uso terapéutico , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas/fisiología , Células HEK293 , Humanos , Masculino , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley , Silimarina/uso terapéutico
7.
Pharmacol Rep ; 71(4): 703-712, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31207432

RESUMEN

BACKGROUND: Silymarin, a known hepatoprotectant, owing to its poor oral bioavailability, has limited pharmacological effects. The present study was designed to improve its in vitro and in vivo hepatoprotection and increase its oral bioavailability against alcohol intoxication by formulating it in four different liposomal formulations namely conventional, dicetyl phosphate, stearyl amine and PEGylated liposomes. METHOD: The liposomes were prepared using phosphatidylcholine, cholesterol, and silymarin in addition to dicetyl phosphate, stearyl amine and DSPE mPEG 2000 by film hydration method with 5% sucrose as a cryo-protectant. The optimized formulations were studied for their release profile at pH 1.2 and 6.8. Liposomes were studied for in vitro protection on Chang liver cells and efficacious liposomes were selected for in vivo hepatoprotection study. Further, conventional liposomes were studied for bioavailability in alcohol intoxicated Wistar rats. RESULTS: The conventional liposomes increased in vitro release profile at pH 1.2 and 6.8 and also showed better in vitro protection compared to silymarin alone. Conventional and PEGylated liposomes showed better improvement in liver function, better efficacy in combating inflammatory conditions, better improvement in antioxidant levels and reversal of histological changes compared to silymarin alone. Conventional also showed an almost fourfold increase in area under the curve compared to silymarin suspension. CONCLUSION: Conventional and PEGylated liposomes of silymarin were found to be more efficacious as hepatoprotective against alcohol-induced hepatotoxicity by its free radical scavenging and anti-inflammatory effects. Conventional liposomes showed enhanced bioavailability compared to silymarin alone.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Etanol/toxicidad , Hígado/efectos de los fármacos , Sustancias Protectoras/farmacología , Silimarina/farmacología , Animales , Disponibilidad Biológica , Supervivencia Celular/efectos de los fármacos , Composición de Medicamentos , Liberación de Fármacos , Células Hep G2 , Humanos , Liposomas , Hígado/metabolismo , Hígado/patología , Masculino , Sustancias Protectoras/administración & dosificación , Sustancias Protectoras/farmacocinética , Ratas Wistar , Silimarina/administración & dosificación , Silimarina/farmacocinética
8.
Molecules ; 24(11)2019 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-31181687

RESUMEN

Silymarin, a mixture of flavonolignan and flavonoid polyphenolic compounds extractable from milk thistle (Silybum marianum) seeds, has anti-oxidant, anti-inflammatory, anti-cancer and anti-viral activities potentially useful in the treatment of several liver disorders, such as chronic liver diseases, cirrhosis and hepatocellular carcinoma. Equally promising are the effects of silymarin in protecting the brain from the inflammatory and oxidative stress effects by which metabolic syndrome contributes to neurodegenerative diseases. However, although clinical trials have proved that silymarin is safe at high doses (>1500 mg/day) in humans, it suffers limiting factors such as low solubility in water (<50 µg/mL), low bioavailability and poor intestinal absorption. To improve its bioavailability and provide a prolonged silymarin release at the site of absorption, the use of nanotechnological strategies appears to be a promising method to potentiate the therapeutic action and promote sustained release of the active herbal extract. The purpose of this study is to review the different nanostructured systems available in literature as delivery strategies to improve the absorption and bioavailability of silymarin.


Asunto(s)
Síndrome Metabólico/tratamiento farmacológico , Silybum marianum/química , Silimarina/farmacocinética , Disponibilidad Biológica , Ensayos Clínicos como Asunto , Preparaciones de Acción Retardada , Humanos , Absorción Intestinal , Semillas/química , Silimarina/química , Silimarina/uso terapéutico
9.
Medicina (Kaunas) ; 55(5)2019 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-31117289

RESUMEN

Background and objectives: Ascorbic acid, alpha lipoic acid (ALA) and silymarin are well-known antioxidants that have hepatoprotective effects. This study aims to investigate the effects of these three compounds combined with attenuating drug-induced oxidative stress and cellular damage, taking acetaminophen (APAP)-induced toxicity in rats as a model both in vivo and in vitro. Materials and Methods: Freshly cultured primary rat hepatocytes were treated with ascorbic acid, ALA, silymarin and their combination, both with and without the addition of APAP to evaluate their in vitro impact on cell proliferation and mitochondrial activity. In vivo study was performed on rats supplemented with the test compounds or their combination for one week followed by two toxic doses of APAP. Results: Selected liver function tests and oxidative stress markers including superoxide dismutase (SOD), malondialdehyde (MDA) and oxidized glutathione (GSSG) were detected. The in vivo results showed that all three pretreatment compounds and their combination prevented elevation of SOD and GSSG serum levels indicating a diminished burden of oxidative stress. Moreover, ascorbic acid, ALA and silymarin in combination reduced serum levels of liver enzymes; however, silymarin markedly maintained levels of all parameters to normal ranges. Silymarin either alone or combined with ascorbic acid and ALA protected cultured rat hepatocytes and increased cellular metabolic activity. The subjected agents were capable of significantly inhibiting the presence of oxidative stress induced by APAP toxicity and the best result for protection was seen with the use of silymarin. Conclusions: The measured liver function tests may suggest an augmented hepatoprotection of the combination preparation than when compared individually.


Asunto(s)
Acetaminofén/efectos adversos , Ácido Ascórbico/farmacocinética , Factores Protectores , Silimarina/farmacocinética , Ácido Tióctico/farmacocinética , Acetaminofén/envenenamiento , Animales , Ácido Ascórbico/uso terapéutico , Enfermedad Hepática Inducida por Sustancias y Drogas/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/complicaciones , Modelos Animales de Enfermedad , Masculino , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley , Silimarina/uso terapéutico , Ácido Tióctico/uso terapéutico
10.
Molecules ; 24(9)2019 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-31052197

RESUMEN

Two novel nanomicellar formulations were developed to improve the poor aqueous solubility and the oral absorption of silymarin. Polymeric nanomicelles made of Soluplus and mixed nanomicelles combining Soluplus with d-α-tocopherol polyethylene glycol 1000 succinate (vitamin E TPGS) were prepared using the thin film method. Physicochemical parameters were investigated, in particular the average diameter, the homogeneity (expressed as polydispersity index), the zeta potential, the morphology, the encapsulation efficiency, the drug loading, the critical micellar concentration and the cloud point. The sizes of ~60 nm, the narrow size distribution (polydispersity index ≤0.1) and the encapsulation efficiency >92% indicated the high affinity between silymarin and the core of the nanomicelles. Solubility studies demonstrated that the solubility of silymarin increased by ~6-fold when loaded into nanomicelles. Furthermore, the physical and chemical parameters of SLM-loaded formulations stored at room temperature and in refrigerated conditions (4 °C) were monitored over three months. In vitro stability and release studies in media miming the physiological conditions were also performed. In addition, both formulations did not alter the antioxidant properties of silymarin as evidenced by the 1,1-Diphenyl-2-picrylhydrazyl radical (DPPH) assay. The potential of the nanomicelles to increase the intestinal absorption of silymarin was firstly investigated by the parallel artificial membrane permeability assay. Subsequently, transport studies employing Caco-2 cell line demonstrated that mixed nanomicelles statistically enhanced the permeability of silymarin compared to polymeric nanomicelles and unformulated extract. Finally, the uptake studies indicated that both nanomicellar formulations entered into Caco-2 cells via energy-dependent mechanisms.


Asunto(s)
Portadores de Fármacos/química , Composición de Medicamentos , Micelas , Nanopartículas/química , Silimarina/administración & dosificación , Silimarina/química , Administración Oral , Disponibilidad Biológica , Células CACO-2 , Permeabilidad de la Membrana Celular , Fenómenos Químicos , Liberación de Fármacos , Estabilidad de Medicamentos , Humanos , Microscopía Electrónica , Tamaño de la Partícula , Polímeros , Silimarina/farmacocinética , Solubilidad , Temperatura
11.
J Psychopharmacol ; 33(5): 615-631, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30896354

RESUMEN

BACKGROUND: The neuropathology of depression is quite complex. Thus, treatment failures are frequent with current antidepressants, raising the need for more effective ones. AIMS: This study aimed to investigate the influence of silymarin on depressive-like behavior induced by chronic unpredictable mild stress (CUMS) and explore the underlying mechanisms. METHODS: Silymarin was formulated as nanostructured lipid carriers (a lipid-based type of nanoparticle with the advantages of physical stability, good release profile, and targeted delivery). Mice were subjected to CUMS paradigm during 14 days. During this period, mice received silymarin (200 mg/kg, p.o.) per se or in its nanoparticle form or fluoxetine (10 mg/kg, p.o.). On the 15th day behavioral and biochemical parameters were analyzed. RESULTS: Oral administration of silymarin (200 mg/kg), particularly in its nanoparticulate form, exerted an antidepressant-like effect, comparable with fluoxetine in mice, as demonstrated in the behavioral despair tests. Silymarin also reversed prefrontal cortical and hippocampal CUMS-induced oxidative stress and neuroinflammation. Furthermore, silymarin augmented neurotransmitter levels, enhanced neurogenesis and inhibited nod-like receptor protein 3 inflammasome activation. Silymarin nanoparticles were superior to silymarin in certain parameters probably due to significantly higher brain silybinin (the major active component of silymarin) concentration by 12.46 fold in the group administered silymarin nanoparticles compared with the mice which were administered silymarin per se. CONCLUSIONS: The antidepressant-like effect of silymarin can be attributed to its antioxidant and anti-inflammatory effects as well as increased neurogenesis in the prefrontal cortex and hippocampus, which delineates silymarin, especially in nanoparticle form, as a promising strategy for treatment of depression.


Asunto(s)
Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Depresión/tratamiento farmacológico , Inflamasomas/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Nanopartículas/administración & dosificación , Neurogénesis/efectos de los fármacos , Silimarina/farmacocinética , Estrés Psicológico/complicaciones , Animales , Antidepresivos/administración & dosificación , Depresión/etiología , Fluoxetina/farmacología , Hipocampo/efectos de los fármacos , Inflamación/tratamiento farmacológico , Masculino , Ratones , Estrés Oxidativo/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Silimarina/administración & dosificación
12.
Eur Rev Med Pharmacol Sci ; 23(2): 885-891, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30720197

RESUMEN

OBJECTIVE: The liver is an important organ that is actively involved in metabolic functions and targeted by a number of toxicants. Galectin-8 (Gal-8) is downregulated in liver fibrosis. Reduced Gal-8 expression correlates with inflammation and metastasis. Therefore, this study aimed to further investigate the benefits of combined administration of silymarin and ginger for CCl4-induced liver injuries in mice. We also investigated the mechanisms underlying the hepatoprotective activity of these herbal drugs and evaluated the role of Gal-8 and apoptosis in liver fibrosis. MATERIALS AND METHODS: Eighty male albino mice were used in this study. Animals were divided into the following groups: control group, fibrotic group, silymarin and ginger group. The CCL4 model was used for the induction of liver fibrosis. RESULTS: Gal-8 expression was reduced in the fibrotic group, while Gal-8 expression was increased in the ginger group and silymarin and ginger group. Tissue levels of nitric oxide (NO) and malondialdehyde (MDA) were markedly increased in the fibrotic group but decreased in the silymarin and ginger group. Additionally, tissue caspase-3 activity and antioxidant markers were decreased in the fibrotic group. However, these markers were increased in the silymarin and ginger group. CONCLUSIONS: Gal-8 is a diagnostic and/or prognostic glycoprotein for liver fibrosis. The combination of silymarin and ginger has protective liver action and reduces the severity and incidence of liver fibrosis.


Asunto(s)
Galectinas/metabolismo , Cirrosis Hepática/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Sustancias Protectoras/administración & dosificación , Silimarina/administración & dosificación , Zingiber officinale/química , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Tetracloruro de Carbono/toxicidad , Sinergismo Farmacológico , Hígado/efectos de los fármacos , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/diagnóstico , Cirrosis Hepática/patología , Masculino , Ratones , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacocinética , Pronóstico , Sustancias Protectoras/farmacocinética , Especies Reactivas de Oxígeno/metabolismo , Índice de Severidad de la Enfermedad , Silimarina/farmacocinética
13.
Int J Pharm ; 555: 63-76, 2019 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-30448315

RESUMEN

The first objective of this study was to optimize a supersaturatable self-nanoemulsifying drug delivery system (S-SNEDDS) containing silymarin through the investigation of the single and synergistic effect of either SNEDDS or a precipitation inhibitor on dissolution efficiency (DE) of silymarin. The bioavailability and hepatoprotective activity of S-SNEDDS were then compared to those of a branded product (Legalon®, Meda). SNEDDS containing silymarin was developed by titration technique, and Poloxamer 407 was selected as the optimal precipitation inhibitor by using casting film and solvent-shift method. The interaction of silybin (the major active constituent of silymarin) and the polymer was then determined by differential scanning calorimetry, powder X-ray diffractometry (PXRD), Fourier transforms infrared spectroscopy and 1H NMR analysis. The combination of two techniques including SNEDDS and addition of 10% of Poloxamer 407 remarkably increased DE4h (88.28%) compared to the reference product (6.41%). The relative bioavailability of S-SNEDDS versus Legalon® was about 760%. The hepatoprotective activity of S-SNEDDS in CCl4-induced mice was also superior to the commercial product in declining both the levels of serum transaminases (ALT, AST) and lipid peroxidation as well as glutathione and superoxide dismutase (SOD) activities under tested doses calculated as silybin (10, 25 and 50 mg/kg). These biopharmaceutical and pharmacological advantages of S-SNEDDS indicated prospects in the development of a novel product that offers lower strength of silymarin while enhancing therapeutic outcomes.


Asunto(s)
Sistemas de Liberación de Medicamentos , Hepatopatías/prevención & control , Nanopartículas , Silimarina/administración & dosificación , Animales , Disponibilidad Biológica , Tetracloruro de Carbono/toxicidad , Química Farmacéutica/métodos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Portadores de Fármacos/química , Liberación de Fármacos , Emulsiones , Masculino , Ratones , Poloxámero/química , Sustancias Protectoras/administración & dosificación , Sustancias Protectoras/farmacocinética , Sustancias Protectoras/farmacología , Conejos , Silibina/administración & dosificación , Silibina/química , Silimarina/farmacocinética , Silimarina/farmacología , Solubilidad
14.
Xenobiotica ; 49(11): 1269-1278, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30489204

RESUMEN

1. Silymarin refers to a class of flavonoid lignans occurring in the fruits and seeds of the Silybum manalttlm (L). Gaertn, and is widely used in dietary supplements. 2. The main active ingredients of silymarin are silychristins A and B, silydianin, silybins A and B, and isosilybins A and B. However, the metabolism of silymarin has never been investigated. The major objectives of the present study were to investigate the metabolic pathways of silymarin isomers and to identify reactive metabolites. 3. Fourteen glutathione (GSH) conjugates were detected in rat/human liver microsomes incubations containing NADPH, GSH and seven individual isomers. Seven GSH conjugates (M1-M7) resulted from demethylated silymarin. M8-M14 originated from hydroxylated silymarin. Moreover, we found that GSH was probably conjugated on either ring A or ring E of silymarin based on the mass spectrometric fragments. In addition, recombinant enzyme incubation experiments demonstrated that CYP3A4 was the predominant P450 responsible for the metabolism of silymarin. 4. Several P450 enzymes were reportedly inactivated by some of bioactive constituents of silymarin to some extent. Our findings facilitate the understanding of mechanisms of the reported inactivation of P450 enzymes induced by silymarin.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Glutatión/metabolismo , Microsomas Hepáticos/metabolismo , Silimarina/metabolismo , Animales , Glutatión/química , Humanos , Hidroxilación , Isomerismo , Microsomas Hepáticos/efectos de los fármacos , Oxidación-Reducción , Ratas , Silibina/química , Silibina/metabolismo , Silibina/farmacocinética , Silimarina/análogos & derivados , Silimarina/química , Silimarina/aislamiento & purificación , Silimarina/farmacocinética , Espectrometría de Masas en Tándem
15.
Anticancer Agents Med Chem ; 18(14): 1970-1974, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30205806

RESUMEN

BACKGROUND: Silimarin is the dry mixture of a whole family of natural substances, extracted after the addition of ethanol, methanol, and acetone. Silimarin consists mainly of silibin A and silibin B, as well as other less important compounds. METHODS: Silimarin has been demonstrated to "inhibit cell proliferation and to induce apoptosis, while also having anti-angiogenic properties." The induction of apoptosis in cancer cells has been mediated by the involvement of ER stress. RESULTS: Silibinin has the potential to operate as a STAT3-targeted inhibitor as well as an inhibitor of the upregulation of the immune checkpoint regulator PD-L1 and also EMT regulators, thus being a promising adjuvant in NSCLC. It has also been documented to suppress cancer cells be means of down- regulating actin cytoskeleton and PI3K / Akt molecular pathways. Several studies have demonstrated that silibinin exerts its protective potential partly through interacting with the tumor suppressor gene p53. CONCLUSIONS: It is noteworthy that research has been carried out on the enhancement of silimarin's bioavailability, especially by the preparation of specific nanoformulas, and its probable additional use together with the chemotherapeutic regimens in the near future.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias/patología , Silimarina/farmacología , Inhibidores de la Angiogénesis/farmacocinética , Inhibidores de la Angiogénesis/farmacología , Antineoplásicos Fitogénicos/farmacocinética , Antígeno B7-H1/antagonistas & inhibidores , Disponibilidad Biológica , Humanos , Neoplasias/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Silimarina/farmacocinética
16.
J Nanobiotechnology ; 16(1): 64, 2018 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-30176941

RESUMEN

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is a chronic disease that causes excessive hepatic lipid accumulation. Reducing hepatic lipid deposition is a key issue in treatment and inhibition of NAFLD evolution. Silymarin is a potent hepatoprotective agent; however, it has low oral bioavailability due to its poor aqueous solubility and low membrane permeability. Unfortunately, few studies have addressed the development of convenient oral nanocarriers that can efficiently deliver silymarin to the liver and enhance its lipid-lowering effect. We designed silymarin-loaded lipid polymer hybrid nanoparticles containing chitosan (CS-LPNs) to improve silymarin bioavailability and evaluated their lipid-lowering effect in adiponutrin/patatin-like phospholipase-3 I148M transgenic mice, an NAFLD model. RESULTS: Compared to chitosan-free nanoparticles, CS-LPNs showed 1.92-fold higher uptake by fatty liver cells. Additionally, CS-LPNs significantly reduced TG levels in fatty liver cells in an in vitro lipid deposition assay, suggesting their potential lipid-lowering effects. The oral bioavailability of silymarin from CS-LPNs was 14.38-fold higher than that from suspensions in rats. Moreover, compared with chitosan-free nanoparticles, CS-LPNs effectively reduced blood lipid levels (TG), improved liver function (AST and ALT), and reduced lipid accumulation in the livers of mice in vivo. Reduced macrovesicular steatosis in pathological tissue after CS-LPN treatment indicated their protective effect against liver steatosis in NAFLD. CONCLUSIONS: CS-LPNs enhanced oral delivery of silymarin and exhibited a desirable lipid-lowering effect in a mouse model. These findings suggest that CS-LPNs may be a promising oral nanocarrier for NAFLD therapeutics.


Asunto(s)
Quitosano/química , Ácido Láctico/química , Lípidos/química , Nanopartículas/química , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Ácido Poliglicólico/química , Silimarina/farmacocinética , Administración Oral , Animales , Disponibilidad Biológica , Transporte Biológico , Células CACO-2 , Preparaciones de Acción Retardada/síntesis química , Portadores de Fármacos/síntesis química , Portadores de Fármacos/metabolismo , Portadores de Fármacos/uso terapéutico , Liberación de Fármacos , Femenino , Células Hep G2 , Humanos , Hígado , Masculino , Ratones , Ratones Transgénicos , Estructura Molecular , Nanopartículas/administración & dosificación , Nanopartículas/metabolismo , Nanopartículas/uso terapéutico , Tamaño de la Partícula , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas , Ratas Wistar , Silimarina/administración & dosificación , Silimarina/uso terapéutico , Propiedades de Superficie
17.
Biomed Pharmacother ; 102: 912-921, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29710546

RESUMEN

Diosmin and silibinin (SIL) are polyphenolic compounds which are the active components of several drugs and dietary supplements. After the oral administration of diosmin (flavonoid glycoside), only its aglycone diosmetin (DIO) reaches the systemic circulation. Both DIO and SIL form complexes with serum albumin and are able to inhibit several cytochrome P450 enzymes. Therefore, it is reasonable to hypothesize that these polyphenols may displace some drugs from serum albumin and inhibit their biotransformation, potentially leading to the disruption of drug therapy. In this study, the inhibitory action of DIO and SIL on CYP2C9-catalyzed metabolism of diclofenac to 4'-hydroxydiclofenac was examined, using warfarin as a positive control. Furthermore, interaction of DIO and SIL with human and bovine serum albumins as well as the displacement of warfarin from albumin by DIO and SIL were tested, employing steady-state fluorescence spectroscopy, fluorescence anisotropy, ultrafiltration, and molecular modeling. It is demonstrated that DIO and SIL are potent inhibitors of CYP2C9 enzyme and are able to displace the Site I ligand warfarin from human serum albumin. Because DIO and SIL may interfere with the pharmacokinetics of several drugs through both ways, we need to consider the potentially hazardous consequences of the consumption of diosmin or SIL together with other drugs.


Asunto(s)
Inhibidores del Citocromo P-450 CYP2C9/farmacología , Flavonoides/farmacocinética , Albúmina Sérica/metabolismo , Silimarina/farmacocinética , Anisotropía , Biotransformación/efectos de los fármacos , Citocromo P-450 CYP2C9 , Diclofenaco/farmacología , Interacciones Farmacológicas , Flavonoides/química , Humanos , Cinética , Simulación del Acoplamiento Molecular , Albúmina Sérica Bovina/metabolismo , Silibina , Silimarina/química , Espectrometría de Fluorescencia , Factores de Tiempo , Ultrafiltración , Warfarina
18.
Clin Ther ; 40(1): 103-113.e1, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29273470

RESUMEN

PURPOSE: The purpose of this study was to compare the bioavailability between 2 milk thistle-containing dietary supplements, Product B and IsaGenesis, in healthy volunteers. METHODS: Bioavailability between Product B, originally formulated as a powdered capsule, and IsaGenesis, reformulated as a soft gel, were compared by measuring silybin A and silybin B as surrogate pharmacokinetic markers for differences in absorption and bioavailability. For this randomized, open-label, crossover pharmacokinetic study, 12 healthy volunteers consumed a single-dose serving of each supplement separated by at least a 7-day washout period. Serial blood samples were obtained at 0, 0.5, 1, 1.5, 2, 3, 4, 6, and 8 hours and analyzed via LC-MS/MS. FINDINGS: Rapid absorption and elimination of silybin A and silybin B have been observed after oral administration of both Product B and IsaGenesis. However, the absorption rate and extent, as indicated by mean the Cmax and mean plasma AUC, were significantly higher for the IsaGenesis soft gel formulation. The dose-corrected mean Cmax was 365% and 450% greater for silybin A and B, respectively, relative to powdered Product B. The time to Tmax was reached, on average, at least 1 hour earlier with IsaGenesis relative to Product B for both silybin A and silybin B. IMPLICATIONS: The IsaGenesis soft gel formulation provided substantially greater absorption and bioavailability of silybin A and silybin B relative to the powdered Product B supplement. ClinicalTrials.gov Identifier: NCT02529605.


Asunto(s)
Antioxidantes/farmacocinética , Suplementos Dietéticos , Extractos Vegetales/farmacocinética , Silybum marianum , Silimarina/farmacocinética , Administración Oral , Adulto , Antioxidantes/administración & dosificación , Área Bajo la Curva , Disponibilidad Biológica , Cápsulas , Cromatografía Liquida , Estudios Cruzados , Composición de Medicamentos , Femenino , Geles , Humanos , Masculino , Extractos Vegetales/administración & dosificación , Polvos , Silibina , Silimarina/administración & dosificación , Silimarina/sangre , Espectrometría de Masas en Tándem , Equivalencia Terapéutica , Adulto Joven
19.
Int J Pharm ; 536(1): 231-240, 2018 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-29162374

RESUMEN

This study demonstrates the preparation of a silybin-loaded N-octyl-O, N-carboxymethyl chitosan micelle (OCC-SLB) to enhance the oral absorption efficiency of silybin (SLB) and investigate the related mechanisms of enhancement. Firstly, the physicochemical properties of OCC and OCC-SLB micelles, including critical micelle concentration (CMC), particle size, zeta potential, drug-loading, etc., were determined. Results of pharmacokinetic studies on rats then confirmed a desirable enhancement in the oral bioavailability of SLB by OCC-SLB micelles compared with a stock SLB suspension solution. Subsequently, uptake studies on the Caco-2 cell line demonstrated that OCC-SLB micelles effectively accumulated SLB or rhodamine-123 into cells through clathrin and caveolae-mediated endocytosis and the inhibition of P-glycoprotein (P-gp) efflux. In addition, results of the Caco-2 transport study further clarified that OCC-SLB micelles enhanced the permeability of SLB via tight junction opening and clathrin-mediated transcytosis across the endothelium. These findings indicated the OCC micelle platform as a potential delivery vehicle for oral administration of P-gp substrates such as SLB.


Asunto(s)
Quitosano/análogos & derivados , Absorción Intestinal/efectos de los fármacos , Silimarina/química , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Administración Oral , Animales , Disponibilidad Biológica , Células CACO-2 , Línea Celular Tumoral , Quitosano/química , Portadores de Fármacos/química , Excipientes/química , Humanos , Micelas , Tamaño de la Partícula , Ratas , Ratas Sprague-Dawley , Silibina , Silimarina/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...