Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Sci Rep ; 11(1): 15688, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34344946

RESUMEN

Hepatitis is one of earlier, but serious, signs of liver damage. High doses of statins for a long time can induce hepatitis. This study aimed to evaluate and compare the therapeutic potential of thymoquinone (TQ) and bee pollen (BP) on fluvastatin (F)-induced hepatitis in rats. Rats were randomly divided into: group 1 (G1, control), G2 (F, hepatitis), G3 (F + TQ), G4 (F + BP), and G5 (F + TQ + BP). Single treatment with TQ or BP relieved fluvastatin-induced hepatitis, with best effect for the combined therapy. TQ and/or BP treatment significantly (1) reduced serum levels of alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, gamma glutamyl transpeptidase, and total bilirubin, (2) decreased malondialdehyde levels and increased level of reduced glutathione, and activities of glutathione peroxidase and catalase in the liver, (3) improved liver histology with mild deposition of type I collagen, (4) increased mRNA levels of transforming growth factor beta 1, nuclear factor Kappa B, and cyclooxygenase 1 and 2, and (5) decreased tumor necrosis factor alpha and upregulated interleukin 10 protein in the liver. These data clearly highlight the ability of TQ and BP combined therapy to cause better ameliorative effects on fluvastatin-induced hepatitis than individual treatment by each alone.


Asunto(s)
Abejas , Benzoquinonas/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Fluvastatina/efectos adversos , Hepatitis Animal/tratamiento farmacológico , Polen , Animales , Antioxidantes/metabolismo , Biomarcadores , Enfermedad Hepática Inducida por Sustancias y Drogas/diagnóstico , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Expresión Génica , Hepatitis Animal/diagnóstico , Hepatitis Animal/etiología , Hepatitis Animal/metabolismo , Inmunohistoquímica , Pruebas de Función Hepática , Estrés Oxidativo/efectos de los fármacos , Ratas , Resultado del Tratamiento
2.
Mol Cell Biochem ; 476(2): 743-755, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33070276

RESUMEN

Statins are potent inhibitors of the mevalonate/cholesterol biosynthetic pathway and are widely prescribed for the prevention of cardiovascular diseases. Here, we carried out a comprehensive analysis of the effects of three statins, simvastatin, atorvastatin, and lovastatin, on six different cancer cell lines that include a P-glycoprotein-expressing, multidrug resistant variant of an ovarian cancer cell line. Incubation of all cancer cell lines with statins resulted in suppression of cell proliferation without inducing apoptotic cell death. The cell proliferation arrest could be reversed upon transfer of cells to statin-free growth media as well as by the supplementation of the growth media with mevalonate. Further analysis suggested that statins induced cell cycle arrest at G0/G1 phase in four cancer cell lines and the loss of c-Myc protein in three cancer cell lines. The c-Myc expression and the progression of cell division cycle were restored upon the addition of mevalonate to the culture media containing statins. Finally, cells incubated with statins contained an increased level of phosphorylated histone H2AX, an observation previously correlated to cellular senescence. Together, these data demonstrate that statins inhibit the mevalonate pathway which is tightly coupled to oxidative branch of the pentose phosphate pathway, c-Myc expression, cell division cycle progression, and cellular senescence. Implications of these observations in the application of statins as cancer therapeutics are discussed.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Atorvastatina/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Fluvastatina/farmacología , Humanos , Lovastatina/farmacología , Ácido Mevalónico/metabolismo , Neoplasias/patología , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Simvastatina/farmacología
3.
J Biochem Mol Toxicol ; 34(6): e22484, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32196851

RESUMEN

Statins are among the most commonly prescribed drugs for the treatment of high blood cholesterol. Myotoxicity of statins in certain individuals is often a severe side effect leading to withdrawal. Using C2C12 and H9c2 cells, both exhibiting characteristics of skeletal muscle cells, we addressed whether resveratrol (RSV) can prevent statin toxicity. Statins decreased cell viability in a dose and time-dependent manner. Among the five statins tested, atorvastatin, simvastatin, lovastatin, pravastatin, and fluvastatin, simvastatin is the most toxic one. Simvastatin at 10 µM caused about 65% loss of metabolic activity as measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays in C2C12 cells or H9c2 cells. Inhibition of metabolic activity correlates with an increase in caspase activity. RSV was found to protect H9c2 cells from simvastatin-induced activation of caspase-3/7. However, such protection was not found in C2C12 cells. This cell type-dependent effect of RSV adds to the complexity in muscle cell toxicity of statins.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Mioblastos/efectos de los fármacos , Sustancias Protectoras/farmacología , Resveratrol/farmacología , Animales , Atorvastatina/efectos adversos , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular , Fluvastatina/efectos adversos , Lovastatina/efectos adversos , Ratones , Mioblastos/metabolismo , Pravastatina/efectos adversos , Ratas , Transducción de Señal/efectos de los fármacos , Simvastatina/efectos adversos
4.
Biochem Biophys Res Commun ; 522(4): 862-868, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-31806372

RESUMEN

Ebola virus (EBOV), pathogen of Ebola hemorrhagic fever (EHF), is an enveloped filamental RNA virus. Recently, the EHF crisis occurred in the Democratic Republic of the Congo again highlights the urgency for its clinical treatments. However, no Food and Drug Administration (FDA)-approved therapeutics are currently available. Drug repurposing screening is a time- and cost-effective approach for identifying anti-EBOV therapeutics. Here, by combinatorial screening using pseudovirion and minigenome replicon systems we have identified several FDA-approved drugs with significant anti-EBOV activities. These potential candidates include azithromycin, clomiphene, chloroquine, digitoxin, epigallocatechin-gallate, fluvastatin, tetrandrine and tamoxifen. Mechanistic studies revealed that fluvastatin inhibited EBOV pseudovirion entry by blocking the pathway of mevalonate biosynthesis, while the inhibitory effect of azithromycin on EBOV maybe due to its intrinsic cationic amphiphilic structure altering the homeostasis of later endosomal vesicle similar as tamoxifen. Moreover, based on structure and pathway analyses, the anti-EBOV activity has been extended to other family members of statins, such as simvastatin, and multiple other cardiac glycoside drugs, some of which exhibited even stronger activities. More importantly, in searching for drug interaction, we found various synergy between several anti-EBOV drug combinations, showing substantial and powerful synergistic against EBOV infection. In conclusion, our work illustrates a successful and productive approach to identify new mechanisms and targets for treating EBOV infection by combinatorial screening of FDA-approved drugs.


Asunto(s)
Antivirales/análisis , Antivirales/farmacología , Técnicas Químicas Combinatorias , Aprobación de Drogas , Evaluación Preclínica de Medicamentos , Ebolavirus/efectos de los fármacos , Azitromicina/farmacología , Glicósidos Cardíacos/farmacología , Línea Celular , Colesterol/biosíntesis , Sinergismo Farmacológico , Ebolavirus/fisiología , Fluvastatina/farmacología , Humanos , Ácido Mevalónico/metabolismo , Modelos Biológicos , Tensoactivos/química , Virión/efectos de los fármacos , Virión/fisiología , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
5.
Sci Rep ; 9(1): 11461, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31391514

RESUMEN

Zika virus (ZIKV) is a mosquito-borne member of the Flaviviridae family. ZIKV infection has been associated with neurological complications such as microcephaly in newborns and Guillain-Barré syndrome in adults; thus, therapeutic agents are urgently needed. Statins are clinically approved for lowering cholesterol levels to prevent cardiovascular disease but have shown potential as antiviral drugs. In this study, we explored the possibility of utilizing statins as anti-ZIKV drugs. We found that, generally, lipophilic statins (atorvastatin, cerivastatin, fluvastatin, lovastatin, mevastatin, and simvastatin) could reduce ZIKV production in vitro and result in smaller foci of infection. Time-of-drug-addition assay revealed that early treatment with statins is more beneficial than late treatment; however, statins could not completely inhibit the entry stage of ZIKV infection. Furthermore, individual lipophilic statins differed in anti-ZIKV capacity, with fluvastatin being the most efficient at low concentrations. Taken together, this study shows that statins or their derivatives have the potential to be used as anti-ZIKV therapeutic agents.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Replicación Viral/efectos de los fármacos , Infección por el Virus Zika/tratamiento farmacológico , Virus Zika/efectos de los fármacos , Animales , Chlorocebus aethiops , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Fluvastatina/química , Fluvastatina/farmacología , Fluvastatina/uso terapéutico , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Inhibidores de Hidroximetilglutaril-CoA Reductasas/química , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Tiempo de Tratamiento , Células Vero , Virus Zika/fisiología , Infección por el Virus Zika/virología
6.
Curr Mol Med ; 19(7): 506-524, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31109273

RESUMEN

BACKGROUND: The incidence of fungal infections has increased significantly. Specifically the cases of candida albicans infection are increasing day by day and their resistance to clinically approved drugs is a major concern for humans. Various classes of antifungal drugs are available in the market for the treatment of these infections but unfortunately, none of them is able to treat the infection. OBJECTIVES: Thus, in the present investigation, we have repurposed the well-known drug (Fluvastatin) in the treatment of Candida albicans infections by using in silico, in vitro and ex vivo techniques. MATERIAL AND METHODS: Computational and in vitro techniques. RESULTS: Firstly, we developed and validated a simple model of CYP45014α-lanosterol demethylase of Candida albicans by using crystal structure of Mycobacterium tuberculosis (1EA1). Further, fluvastatin was docked with a validated model of CYP45014α-lanosterol demethylase and revealed good binding affinity as that of fluconazole. In vitro results (Percentage growth retardation, Fungal growth kinetics, Biofilm test and Post antifungal test) have shown good antifungal activity of fluvastatin. Finally, the results of MTT assay have shown non-cytotoxic effect of fluvastatin in murine splenocytes and thymocytes. CONCLUSION: However, further in vivo studies are required to confirm the complete role of fluvastatin as an antifungal agent.


Asunto(s)
Candida albicans/efectos de los fármacos , Candidiasis/tratamiento farmacológico , Fluvastatina/farmacología , Esterol 14-Desmetilasa/genética , Animales , Antifúngicos/farmacología , Candida albicans/patogenicidad , Candidiasis/genética , Candidiasis/microbiología , Biología Computacional , Reposicionamiento de Medicamentos , Fluvastatina/química , Humanos , Ratones , Pruebas de Sensibilidad Microbiana , Esterol 14-Desmetilasa/efectos de los fármacos
7.
J Pharm Sci ; 108(3): 1085-1089, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30339864

RESUMEN

Lilly Laboratories cell porcine kidney 1 (LLC-PK1) cells transfected with human P-glycoprotein (LLC-PK1-P-gp) are widely used in transport assays to identify drug candidates that function as substrates of this efflux transporter. Endogenous transporters expressed in LLC-PK1 cells may complicate the interpretation of findings from P-gp-mediated transport assays. We investigated the impact of porcine breast cancer resistance protein (Bcrp) in P-gp-mediated transport assays in LLC-PK1 cells. Porcine Bcrp mRNA was detected in both LLC-PK1 wildtype (WT) and LLC-PK1-P-gp cells by quantitative RT-PCR. To investigate the activity and impact of porcine Bcrp, we conducted transport assays using 6 typical BCRP substrates in LLC-PK1 cells. Efflux ratios (ER) of the 6 BCRP substrates in LLC-PK1 WT cells were >2, and were reduced in the presence of the BCRP inhibitor Ko143. The efflux activities of the 6 BCRP substrates were confirmed using MDCKII cells transfected with human BCRP. Net ERs of prazosin and fluvastatin, dual substrates of P-gp and BCRP, determined by dividing ERs in LLC-PK1-P-gp cells by those in LLC-PK1 WT cells, were <2, but increased to >2 in the presence of Ko143. These results indicated that endogenous Bcrp in LLC-PK1 cells was involved in the transport of BCRP substrates and may interfere with the identification of P-gp substrates.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/antagonistas & inhibidores , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Animales , Dicetopiperazinas/farmacología , Fluvastatina/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Células LLC-PK1 , Prazosina/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Porcinos , Transfección
8.
Eur J Clin Pharmacol ; 74(5): 601-609, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29368187

RESUMEN

PURPOSE: The objective of this study is to assess the effects of green tea and its major catechin component, (-)-epigallocatechin gallate (EGCG), on CYP2C9-mediated substrate metabolism in vitro, and the pharmacokinetics of fluvastatin in healthy volunteers. METHODS: The metabolism of diclofenac and fluvastatin in human recombinant CYP2C9 was investigated in the presence of EGCG. In a randomized three-phase crossover study, 11 healthy volunteers ingested a single 20-mg dose of fluvastatin with green tea extract (GTE), containing 150 mg of EGCG, along with water (300 mL), brewed green tea (300 mL), or water (300 mL) after overnight fasting. Plasma concentrations of fluvastatin and EGCG were measured by ultra-performance liquid chromatography with fluorescence detection and a single mass spectrometer. RESULTS: EGCG inhibited diclofenac 4'-hydroxylation and fluvastatin degradation with IC50 of 2.23 and 48.04 µM, respectively. Brewed green tea used in the clinical study also dose-dependently inhibited the metabolism of diclofenac and fluvastatin in vitro. However, no significant effects of GTE and brewed green tea were observed in plasma concentrations of fluvastatin. The geometric mean ratios with 90% CI for area under the plasma concentration-time curve (AUC0-∞) of fluvastatin were 0.993 (0.963-1.024, vs. brewed green tea) and 0.977 (0.935-1.020, vs. GTE). CONCLUSIONS: Although in vitro studies indicated that EGCG and brewed green tea produce significant inhibitory effects on CYP2C9 activity, the concomitant administration of green tea and fluvastatin in healthy volunteers did not influence the pharmacokinetics of fluvastatin.


Asunto(s)
Catequina/análogos & derivados , Citocromo P-450 CYP2C9/metabolismo , Ácidos Grasos Monoinsaturados/farmacocinética , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Indoles/farmacocinética , , Adulto , Antiinflamatorios no Esteroideos/farmacocinética , Catequina/análisis , Catequina/sangre , Catequina/farmacocinética , Catequina/farmacología , Estudios Cruzados , Diclofenaco/farmacocinética , Ácidos Grasos Monoinsaturados/sangre , Femenino , Fluvastatina , Interacciones Alimento-Droga , Voluntarios Sanos , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/sangre , Indoles/sangre , Masculino , Té/química , Adulto Joven
9.
Proc Natl Acad Sci U S A ; 114(21): 5503-5508, 2017 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-28500274

RESUMEN

Cerebral cavernous malformations (CCMs) are common vascular anomalies that develop in the central nervous system and, more rarely, the retina. The lesions can cause headache, seizures, focal neurological deficits, and hemorrhagic stroke. Symptomatic lesions are treated according to their presentation; however, targeted pharmacological therapies that improve the outcome of CCM disease are currently lacking. We performed a high-throughput screen to identify Food and Drug Administration-approved drugs or other bioactive compounds that could effectively suppress hyperproliferation of mouse brain primary astrocytes deficient for CCM3. We demonstrate that fluvastatin, an inhibitor of 3-hydroxy-3-methyl-glutaryl (HMG)-CoA reductase and the N-bisphosphonate zoledronic acid monohydrate, an inhibitor of protein prenylation, act synergistically to reverse outcomes of CCM3 loss in cultured mouse primary astrocytes and in Drosophila glial cells in vivo. Further, the two drugs effectively attenuate neural and vascular deficits in chronic and acute mouse models of CCM3 loss in vivo, significantly reducing lesion burden and extending longevity. Sustained inhibition of the mevalonate pathway represents a potential pharmacological treatment option and suggests advantages of combination therapy for CCM disease.


Asunto(s)
Difosfonatos/uso terapéutico , Ácidos Grasos Monoinsaturados/uso terapéutico , Hemangioma Cavernoso del Sistema Nervioso Central/tratamiento farmacológico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Imidazoles/uso terapéutico , Indoles/uso terapéutico , Animales , Astrocitos/efectos de los fármacos , Difosfonatos/farmacología , Drosophila , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Células Endoteliales/efectos de los fármacos , Femenino , Fluvastatina , Ensayos Analíticos de Alto Rendimiento , Imidazoles/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Embarazo , Prenilación de Proteína/efectos de los fármacos , Ácido Zoledrónico
10.
J Orthop Res ; 35(11): 2465-2475, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28303595

RESUMEN

Statins are cholesterol-lowering drugs that inhibit 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, a rate-limiting enzyme of the mevalonate pathway. The anti-inflammatory effect of statins has been reported in recent years. The present study investigated therapeutic effects of the local administration of statin in osteoarthritis (OA). We assessed clinically used statins and selected fluvastatin for further experimentation, as it showed potent anabolic and anti-catabolic effects on human OA chondrocytes. To achieve controlled intra-articular administration of statin, we developed an intra-articular injectable statin using poly(lactic-co-glycolic acid) (PLGA) as a drug delivery system (DDS). The release profile of the statin was evaluated in vitro. Finally, therapeutic effects of fluvastatin-loaded PLGA microspheres (FLU-PLGA) were tested in a rabbit OA model. Rabbit knees were divided into four subgroups: group 1-A, PLGA-treated group; group 1-B, PLGA contralateral saline control group; group 2-A, FLU-PLGA-treated group; and group 2-B, FLU-PLGA contralateral saline control group. Histological analysis 5 weeks after intra-articular injection revealed that OARSI scores were lower in group 2-A. No significant differences in OARSI scores were observed between groups 1-A, 1-B, and 2-B. This study indicates that a single intra-articular injection of fluvastatin-loaded PLGA microspheres could be a novel therapeutic approach for treating patients with OA. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2465-2475, 2017.


Asunto(s)
Anticolesterolemiantes/administración & dosificación , Artritis Experimental/tratamiento farmacológico , Cartílago Articular/efectos de los fármacos , Ácidos Grasos Monoinsaturados/administración & dosificación , Indoles/administración & dosificación , Osteoartritis/tratamiento farmacológico , Animales , Condrocitos/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Femenino , Fluvastatina , Humanos , Inyecciones Intraarticulares , Ácido Láctico , Microesferas , Ácido Poliglicólico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Cultivo Primario de Células , Conejos
11.
Bioorg Med Chem Lett ; 27(4): 1055-1061, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28089347

RESUMEN

Retinoid X receptor alpha (RXRα), an important ligand-dependent transcription factor, plays a critical role in the development of various cancers and metabolic and neurodegenerative diseases. Therefore, RXRα represents one of the most important targets in modern drug discovery. In this study, Drugbank 2.0 with 1280 old drugs were virtually screened by Glide according to the crystal structure of ligand-binding domain (LBP) of RXRα. 15 compounds selected were tested for their binding and transcriptional activity toward RXRα by Biacore and reporter gene assay, respectively. The identified new scafford ligand of RXRα, Pitavastatin (1), was chemically optimized. Our results demonstrated that statin compounds Pitavastatin (1) and Fluvastatin (4) could bind to the LBP of RXRα (KD=13.30µM and 11.04µM, respectively) and serve as transcriptional antagonists of RXRα. On the contrary, compound (12) (domperidone) and (13) (rosiglitazone maleate) could bind to the LBP of RXRα (KD=8.80µM and 15.01µM, respectively) but serve as transcriptional agonists of RXRα.


Asunto(s)
Bases de Datos Factuales , Receptor alfa X Retinoide/antagonistas & inhibidores , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos , Ácidos Grasos Monoinsaturados/química , Ácidos Grasos Monoinsaturados/farmacología , Fluvastatina , Indoles/química , Indoles/farmacología , Ligandos , Quinolinas/química , Quinolinas/farmacología , Receptor alfa X Retinoide/química
12.
Cochrane Database Syst Rev ; 11: CD009462, 2016 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-27888640

RESUMEN

BACKGROUND: Elevated levels of total cholesterol and low-density lipoprotein play an important role in the development of atheromas and, therefore, in cardiovascular diseases. Cholesterol biosynthesis follows a circadian rhythm and is principally produced at night (between 12:00 am and 6:00 am). The adjustment of hypolipaemic therapy to biologic rhythms is known as chronotherapy. Chronotherapy is based on the idea that medication can have different effects depending on the hour at which it is taken. Statins are one of the most widely used drugs for the prevention of cardiovascular events. In usual clinical practice, statins are administered once per day without specifying the time when they should be taken. It is unknown whether the timing of statin administration is important for clinical outcomes. OBJECTIVES: To critically evaluate and analyse the evidence available from randomised controlled trials regarding the effects of chronotherapy on the effectiveness and safety of treating hyperlipidaemia with statins. SEARCH METHODS: We searched the CENTRAL, MEDLINE, Embase, LILACS, ProQuest Health & Medical Complete, OpenSIGLE, Web of Science Conference Proceedings, and various other resources including clinical trials registers up to November 2015. We also searched the reference lists of relevant reviews for eligible studies. SELECTION CRITERIA: We included randomised controlled trials (RCTs), enrolling people with primary or secondary hyperlipidaemia. To be included, trials must have compared any chronotherapeutic lipid-lowering regimen with statins and any other statin lipid-lowering regimen not based on chronotherapy. We considered any type and dosage of statin as eligible, as long as the control and experimental arms differed only in the timing of the administration of the same statin. Quasi-randomised studies were excluded. DATA COLLECTION AND ANALYSIS: We used the standard methodological procedures expected by Cochrane. We extracted the key data from studies in relation to participants, interventions, and outcomes for safety and efficacy. We calculated odds ratios (OR) for dichotomous data and mean differences (MD) for continuous data with 95% confidence intervals (CI). Using the GRADE approach, we assessed the quality of the evidence and we used the GRADEpro Guideline Development Tool to import data from Review Manager to create 'Summary of findings' tables. MAIN RESULTS: This review includes eight RCTs (767 participants analysed in morning and evening arms). The trials used different lipid-lowering regimens with statins (lovastatin: two trials; simvastatin: three trials; fluvastatin: two trials; pravastatin: one trial). All trials compared the effects between morning and evening statin administration. Trial length ranged from four to 14 weeks. We found a high risk of bias in the domain of selective reporting in three trials and in the domain of incomplete outcome data in one trial of the eight trials included. None of the studies included were judged to be at low risk of bias.None of the included RCTs reported data on cardiovascular mortality, cardiovascular morbidity, incidence of cardiovascular events, or deaths from any cause. Pooled results showed no evidence of a difference in total cholesterol (MD 4.33, 95% CI -1.36 to 10.01), 514 participants, five trials, mean follow-up 9 weeks, low-quality evidence), low-density lipoprotein cholesterol (LDL-C) levels (MD 4.85 mg/dL, 95% CI -0.87 to 10.57, 473 participants, five trials, mean follow-up 9 weeks, low-quality evidence), high-density lipoprotein cholesterol (HDL-C) (MD 0.54, 95% CI -1.08 to 2.17, 514 participants, five trials, mean follow-up 9 weeks, low-quality evidence) or triglycerides (MD -8.91, 95% CI -22 to 4.17, 510 participants, five trials, mean follow-up 9 weeks, low-quality evidence) between morning and evening statin administration.With regard to safety outcomes, five trials (556 participants) reported adverse events. Pooled analysis found no differences in statins adverse events between morning and evening intake (OR 0.71, 95% CI 0.44 to 1.15, 556 participants, five trials, mean follow-up 9 weeks, low-quality evidence). AUTHORS' CONCLUSIONS: Limited and low-quality evidence suggested that there were no differences between chronomodulated treatment with statins in people with hyperlipidaemia as compared to conventional treatment with statins, in terms of clinically relevant outcomes. Studies were short term and therefore did not report on our primary outcomes, cardiovascular clinical events or death. The review did not find differences in adverse events associated with statins between both regimens. Taking statins in the evening does not have an effect on the improvement of lipid levels with respect to morning administration. Further high-quality trials with longer-term follow-up are needed to confirm the results of this review.


Asunto(s)
Anticolesterolemiantes/administración & dosificación , Cronoterapia de Medicamentos , Hiperlipidemias/tratamiento farmacológico , Anticolesterolemiantes/efectos adversos , Ácidos Grasos Monoinsaturados/administración & dosificación , Fluvastatina , Humanos , Indoles/administración & dosificación , Lovastatina/administración & dosificación , Pravastatina/administración & dosificación , Ensayos Clínicos Controlados Aleatorios como Asunto , Simvastatina/administración & dosificación
13.
Atherosclerosis ; 254: 215-227, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27764723

RESUMEN

BACKGROUND AND AIMS: Our study aims to evaluate the efficacy and safety of long-term treatment of statins for coronary heart disease (CHD). METHODS: Efficacy outcomes included changes in blood lipids, risk of CHD mortality and all-cause mortality. Safety outcomes were evaluated by the risk of adverse events (AE). Bayesian network meta-analysis was used to compare the direct and indirect effects between different statins. RESULTS: The systematic review showed that levels of blood lipids decreased during statin treatment. High dose of atorvastatin was the most obvious treatment for the reduction of blood lipids. Network meta-analysis showed that statins were significantly more effective than the control in reducing the risk of CHD mortality (Odds Ratio (OR) 0.69, 95% CI 0.61-0.77) and all-cause mortality (OR 0.84, 95% CI 0.80-0.87). In terms of reducing the risk of CHD morality, fluvastatin (77.3%), atorvastatin (72.3%) and lovastatin (68.4%) had higher cumulative probability than other statins, which were more effective treatments for the reduction of CHD morality. In terms of reducing all-cause mortality, atorvastatin (78.6%), fluvastatin (77.1%) and pitavastatin (74.1%) had higher cumulative probability than other statins, which were more effective treatment for reducing the all-cause mortality. Compared with placebo, statins increased the incidence risk of muscle disease (OR 1.05, 95% CI 1.00-1.10) and kidney disease (OR 1.11, 95% CI 1.05-1.72). CONCLUSIONS: Statins significantly reduced levels of blood lipids, with a high dose of atorvastatin being the most effective in blood-lipid level modification. Statins reduced the risk of CHD mortality and all-cause mortality, with atorvastatin and fluvastatin being the most effective in reducing the risk of CHD mortality and all-cause mortality. Statins increased the risk of muscle disease and kidney damage.


Asunto(s)
Enfermedad Coronaria/tratamiento farmacológico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Anticolesterolemiantes/farmacología , Atorvastatina/farmacología , Teorema de Bayes , Enfermedad Coronaria/patología , Ácidos Grasos Monoinsaturados/farmacología , Fluvastatina , Humanos , Indoles/farmacología , Enfermedades Renales/tratamiento farmacológico , Lípidos/sangre , Lovastatina/análogos & derivados , Lovastatina/farmacología , Músculos/efectos de los fármacos , Metaanálisis en Red , Seguridad del Paciente , Pravastatina/farmacología , Quinolinas/farmacología , Ensayos Clínicos Controlados Aleatorios como Asunto , Simvastatina/farmacología , Resultado del Tratamiento
14.
Basic Clin Pharmacol Toxicol ; 118(6): 499-502, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26617265

RESUMEN

The endogenous oxysterol 4ß-hydroxycholesterol may be used as a marker for the drug-metabolizing enzymes cytochrome P450 3A (CYP3A). The primary aim of this study was to investigate the effect of statin treatment on plasma 4ß-hydroxycholesterol concentrations. Plasma samples from a previously performed clinical study where gallstone patients had been treated with placebo (n = 6), 20 mg fluvastatin (n = 9) or 80 mg atorvastatin (n = 9) daily for 4 weeks were analysed. Hepatic CYP3A mRNA levels had previously been shown to be unchanged in all three treatment groups. Plasma 4ß-hydroxycholesterol did not change significantly (p = 0.92) in the placebo group, but treatment with low-dose fluvastatin or high-dose atorvastatin resulted in reductions in plasma concentration of 10.7% (p < 0.05) and 36.5% (p < 0.01), respectively. However, the 4ß-hydroxycholesterol/cholesterol ratio did not change significantly for the patients receiving placebo or patients receiving low-dose fluvastatin. The ratio for patients receiving high-dose atorvastatin increased by 12% (p < 0.05). In conclusion, the total plasma cholesterol level is an important determinant for the plasma 4ß-hydroxycholesterol level.


Asunto(s)
Atorvastatina/farmacología , Colesterol/sangre , Citocromo P-450 CYP3A/metabolismo , Ácidos Grasos Monoinsaturados/farmacología , Cálculos Biliares/tratamiento farmacológico , Hidroxicolesteroles/sangre , Indoles/farmacología , Atorvastatina/uso terapéutico , Biomarcadores/sangre , Interacciones Farmacológicas , Ácidos Grasos Monoinsaturados/uso terapéutico , Femenino , Fluvastatina , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Indoles/uso terapéutico , Masculino , Persona de Mediana Edad , Suecia
15.
J Am Chem Soc ; 137(26): 8490-8, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26070091

RESUMEN

H2S produced in small amounts by mammalian cells has been identified in mediating biological signaling functions. However, the in situ trapping of endogenous H2S generation is still handicapped by a lack of straightforward methods with high selectivity and fast response. Here, we encapsulate a semi-cyanine-BODIPY hybrid dye (BODInD-Cl) and its complementary energy donor (BODIPY1) into the hydrophobic interior of an amphiphilic copolymer (mPEG-DSPE), especially for building up a ratiometric fluorescent H2S nanoprobe with extraordinarily fast response. A remarkable red-shift in the absorption band with a gap of 200 nm in the H2S response can efficiently switch off the Förster resonance energy transfer (FRET) from BODIPY1 to BODInD-Cl, subsequently recovering the donor fluorescence. Impressively, both the interior hydrophobicity of supramolecular micelles and electron-withdrawing nature of indolium unit in BODInD-Cl can sharply increase aromatic nucleophilic substitution with H2S. The ratiometric strategy based on the unique self-assembled micellar aggregate NanoBODIPY achieves an extremely fast response, enabling in situ imaging of endogenous H2S production and mapping its physiological and pathological consequences. Moreover, the amphiphilic copolymer renders the micellar assembly biocompatible and soluble in aqueous solution. The established FRET-switchable macromolecular envelope around BODInD-Cl and BODIPY1 enables cellular uptake, and makes a breakthrough in the trapping of endogenous H2S generation within raw264.7 macrophages upon stimulation with fluvastatin. This study manifests that cystathione γ-lyase (CSE) upregulation contributes to endogenous H2S generation in fluvastatin-stimulated macrophages, along with a correlation between CSE/H2S and activating Akt signaling pathway.


Asunto(s)
Ácidos Grasos Monoinsaturados/química , Transferencia Resonante de Energía de Fluorescencia/métodos , Sulfuro de Hidrógeno/química , Indoles/química , Nanopartículas/química , Animales , Compuestos de Boro/química , Cistationina gamma-Liasa/química , Colorantes Fluorescentes/química , Fluvastatina , Macrófagos/metabolismo , Ratones , Micelas , Microscopía Confocal , Microscopía Fluorescente/métodos , Polímeros/química , Células RAW 264.7 , Regulación hacia Arriba
16.
J Muscle Res Cell Motil ; 36(3): 263-74, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25920381

RESUMEN

Myalgia and muscle weakness may appreciably contribute to the poor adherence to statin therapy. Although the pathomechanism of statin-induced myopathy is not completely understood, changes in calcium homeostasis and reduced coenzyme Q10 levels are hypothesized to play important roles. In our experiments, fluvastatin and/or coenzyme Q10 was administered chronically to normocholesterolaemic or hypercholaestherolaemic rats, and the modifications of the calcium homeostasis and the strength of their muscles were investigated. While hypercholesterolaemia did not change the frequency of sparks, fluvastatin increased it on muscles both from normocholesterolaemic and from hypercholesterolaemic rats. This effect, however, was not mediated by a chronic modification of the ryanodine receptor as shown by the unchanged ryanodine binding in the latter group. While coenzyme Q10 supplementation significantly reduced the frequency of the spontaneous calcium release events, it did not affect their amplitude and spatial spread in muscles from fluvastatin-treated rats. This indicates that coenzyme Q10 supplementation prevented the spark frequency increasing effect of fluvastatin without having a major effect on the amount of calcium released during individual sparks. In conclusion, we have found that fluvastatin, independently of the cholesterol level in the blood, consistently and specifically increased the frequency of calcium sparks in skeletal muscle cells, an effect which could be prevented by the addition of coenzyme Q10 to the diet. These results support theories favouring the role of calcium handling in the pathophysiology of statin-induced myopathy and provide a possible pathway for the protective effect of coenzyme Q10 in statin treated patients symptomatic of this condition.


Asunto(s)
Ácidos Grasos Monoinsaturados/farmacología , Hipercolesterolemia/tratamiento farmacológico , Hipercolesterolemia/metabolismo , Indoles/farmacología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Ubiquinona/análogos & derivados , Animales , Calcio/metabolismo , Colesterol/sangre , Femenino , Fluvastatina , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hipercolesterolemia/sangre , Enfermedades Musculares/sangre , Enfermedades Musculares/tratamiento farmacológico , Enfermedades Musculares/metabolismo , Ratas , Ratas Endogámicas F344 , Ubiquinona/metabolismo
17.
Eur J Intern Med ; 26(2): 82-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25640999

RESUMEN

The use of statins for cardiovascular disease prevention is clearly supported by clinical evidence. However, in January 2014 the U.S. Food and Drug Administration released an advice on statin risk reporting that "statin benefit is indisputable, but they need to be taken with care and knowledge of their side effects". Among them the by far most common complication is myopathy, ranging from common but clinically benign myalgia to rare but life-threatening rhabdomyolysis. This class side effect appears to be dose dependent, with more lipophilic statin (i.e., simvastatin) carrying a higher overall risk. Hence, to minimize statin-associated myopathy, clinicians should take into consideration a series of factors that potentially increase this risk (i.e., drug-drug interactions, female gender, advanced age, diabetes mellitus, hypothyroidism and vitamin D deficiency). Whenever it is appropriate to stop statin treatment, the recommendations are to stay off statin until resolution of symptoms or normalization of creatine kinase values. Afterwards, clinicians have several options to treat dyslipidemia, including the use of a lower dose of the same statin, intermittent non-daily dosing of statin, initiation of a different statin, alone or in combination with nonstatin lipid-lowering agents, and substitution with red yeast rice.


Asunto(s)
Anticolesterolemiantes/uso terapéutico , Dislipidemias/tratamiento farmacológico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Mialgia/inducido químicamente , Rabdomiólisis/inducido químicamente , Factores de Edad , Anciano , Anciano de 80 o más Años , Clorhidrato de Colesevelam/uso terapéutico , Creatina Quinasa/sangre , Interacciones Farmacológicas , Quimioterapia Combinada , Dislipidemias/epidemiología , Ezetimiba/uso terapéutico , Ácidos Grasos Monoinsaturados/efectos adversos , Femenino , Fluvastatina , Humanos , Indoles/efectos adversos , Masculino , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/metabolismo , Mialgia/sangre , Rabdomiólisis/sangre , Medición de Riesgo , Factores de Riesgo , Rosuvastatina Cálcica/efectos adversos , Factores Sexuales , Deficiencia de Vitamina D/epidemiología
18.
Biochem Biophys Res Commun ; 457(1): 23-30, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25529449

RESUMEN

The peroxisome proliferator-activated receptor-γ (PPARγ) is an important regulator of lipid and glucose metabolism, and its activation is reported to suppress the progression of atherosclerosis. We have reported that 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) activate PPARγ in macrophages. However, it is not yet known whether statins activate PPARγ in other vascular cells. In the present study, we investigated whether statins activate PPARγ in smooth muscle cells (SMCs) and endothelial cells (ECs) and thus mediate anti-atherosclerotic effects. Human aortic SMCs (HASMCs) and human umbilical vein ECs (HUVECs) were used in this study. Fluvastatin and pitavastatin activated PPARγ in HASMCs, but not in HUVECs. Statins induced cyclooxygenase-2 (COX-2) expression in HASMCs, but not in HUVECs. Moreover, treatment with COX-2-siRNA abrogated statin-mediated PPARγ activation in HASMCs. Statins suppressed migration and proliferation of HASMCs, and inhibited lipopolysaccharide-induced expression of monocyte chemoattractant protein-1 (MCP-1) and tumor necrosis factor-α (TNF-α) in HASMCs. These effects of statins were abrogated by treatment with PPARγ-siRNA. Treatment with statins suppressed atherosclerotic lesion formation in Apoe(-/-) mice. In addition, transcriptional activity of PPARγ and CD36 expression were increased, and the expression of MCP-1 and TNF-α was decreased, in the aorta of statin-treated Apoe(-/-) mice. In conclusion, statins mediate anti-atherogenic effects through PPARγ activation in SMCs. These effects of statins on SMCs may be beneficial for the prevention of atherosclerosis.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , PPAR gamma/metabolismo , Animales , Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/metabolismo , Aterosclerosis/enzimología , Aterosclerosis/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quimiocina CCL2/metabolismo , Ciclooxigenasa 2/metabolismo , Progresión de la Enfermedad , Ácidos Grasos Monoinsaturados/farmacología , Fluvastatina , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Indoles/farmacología , Lipopolisacáridos , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
19.
Sci Rep ; 4: 5285, 2014 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-24923838

RESUMEN

Mitochondria are semi-autonomous organelles regulated by a complex network of proteins that are vital for many cellular functions. Because mitochondrial modulators can impact many aspects of cellular homeostasis, their identification and validation has proven challenging. It requires the measurement of multiple parameters in parallel to understand the exact nature of the changes induced by such compounds. We developed a platform of assays scoring for mitochondrial function in two complementary models systems, mammalian cells and C. elegans. We first optimized cell culture conditions and established the mitochondrial signature of 1,200 FDA-approved drugs in liver cells. Using cell-based and C. elegans assays, we further defined the metabolic effects of two pharmacological classes that emerged from our hit list, i.e. imidazoles and statins. We found that these two drug classes affect respiration through different and cholesterol-independent mechanisms in both models. Our screening strategy enabled us to unequivocally identify compounds that have toxic or beneficial effects on mitochondrial activity. Furthermore, the cross-species approach provided novel mechanistic insight and allowed early validation of hits that act on mitochondrial function.


Asunto(s)
Caenorhabditis elegans/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Preparaciones Farmacéuticas/administración & dosificación , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular , Línea Celular Tumoral , Análisis por Conglomerados , Aprobación de Drogas , Evaluación Preclínica de Medicamentos/métodos , Ácidos Grasos Monoinsaturados/farmacología , Fluvastatina , Expresión Génica/efectos de los fármacos , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Imidazoles/farmacología , Indoles/farmacología , Lovastatina/farmacología , Células MCF-7 , Ratones , Mitocondrias/metabolismo , Preparaciones Farmacéuticas/clasificación , Reproducibilidad de los Resultados , Simvastatina/farmacología , Estados Unidos , United States Food and Drug Administration
20.
J Cardiopulm Rehabil Prev ; 34(3): 208-12, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24263076

RESUMEN

PURPOSE: We have previously shown that a "short-term, low-dose" treatment approach with statins, angiotensin receptor blockers, and especially their low-dose combination, is effective in improving arterial wall properties in apparently healthy middle-age men. This study was performed to expand investigation of its effects on inflammation and oxidative stress. METHODS: The study was performed supplementary to 3 previous studies, overall 65 treated participants (25 received fluvastatin 10 mg, 20 valsartan 20 mg, 20 their combination) and 65 participants placebo. The stored blood samples (collected at inclusion and after 30 days of treatment) were used to measure high-sensitivity CRP, interleukin-6, vascular cell adhesion molecule-1, total antioxidant status, glutathione peroxidase, and selenium concentration. RESULTS: A low-dose combination decreased inflammation parameters (high-sensitivity CRP: from 1.2 ± 0.1 to 0.7 ± 0.1 mg/L; P < .001; vascular cell adhesion molecule-1: from 523 ± 21 to 482 ± 12 pg/mL; P < .05; while interleukin-6 did not reach the level of significance). It also increased antioxidant defenses, as measured by total antioxidant status and glutathione peroxidase (from 1.4 ± 0.04 to 1.5 ± 0.04 mmol/L; P < .01, and from 1.2 ± 0.06 to 1.4 ± 0.06 µkat/g hemoglobin; P < .05, respectively), accompanied by decreased selenium levels. Low-dose valsartan was separately less effective than the combination. No changes were observed in the control groups. CONCLUSIONS: Low-dose combination of fluvastatin and valsartan and, to a lesser extent low-dose valsartan alone, produced important anti-inflammatory and anti-oxidative effects. These results confirm and extend the potential of the "short-term, low-dose" preventive strategy.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Ácidos Grasos Monoinsaturados/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Indoles/administración & dosificación , Inflamación/sangre , Estrés Oxidativo/efectos de los fármacos , Tetrazoles/administración & dosificación , Valina/análogos & derivados , Antioxidantes/análisis , Proteína C-Reactiva/análisis , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Fluvastatina , Glutatión Peroxidasa/sangre , Humanos , Interleucina-6/sangre , Masculino , Persona de Mediana Edad , Selenio/sangre , Valina/administración & dosificación , Valsartán , Molécula 1 de Adhesión Celular Vascular/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA