Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 408
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Neurosci ; 43(10): 1814-1829, 2023 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-36697260

RESUMEN

Juvenile neuronal ceroid lipofuscinosis (JNCL) is a fatal inherited neurodegenerative disease of children that occurs because of defective function of the lysosomal membrane glycoprotein CLN3. JNCL features glial activation and accumulation of autofluorescent storage material containing subunit c of mitochondrial ATP synthase (SCMAS), ultimately resulting into neuronal loss. Until now, no effective therapy is available for JNCL. This study underlines the possible therapeutic importance of gemfibrozil, an activator of peroxisome proliferator-activated receptor α (PPARα) and a lipid-lowering drug approved by the Food and Drug Administration in an animal model of JNCL. Oral gemfibrozil treatment reduced microglial and astroglial activation, attenuated neuroinflammation, restored the level of transcription factor EB (TFEB; the master regulator of lysosomal biogenesis), and decreased the accumulation of storage material SCMAS in somatosensory barrel field (SBF) cortex of Cln3Δex7/8 (Cln3ΔJNCL) mice of both sexes. Accordingly, gemfibrozil treatment also improved locomotor activities of Cln3ΔJNCL mice. While investigating the mechanism, we found marked loss of PPARα in the SBF cortex of Cln3ΔJNCL mice, which increased after gemfibrozil treatment. Oral gemfibrozil also stimulated the recruitment of PPARα to the Tfeb gene promoter in vivo in the SBF cortex of Cln3ΔJNCL mice, indicating increased transcription of Tfeb in the CNS by gemfibrozil treatment via PPARα. Moreover, disease pathologies aggravated in Cln3ΔJNCL mice lacking PPARα (Cln3ΔJNCLΔPPARα) and gemfibrozil remained unable to decrease SCMAS accumulation, reduce glial activation, and improve locomotor performance of Cln3ΔJNCLΔPPARα mice. These results suggest that activation of PPARα may be beneficial for JNCL and that gemfibrozil may be repurposed for the treatment of this incurable disease.SIGNIFICANCE STATEMENT Despite intense investigations, no effective therapy is available for JNCL, an incurable inherited lysosomal storage disorder. Here, we delineate that oral administration of gemfibrozil, a lipid-lowering drug, decreases glial inflammation, normalizes and/or upregulates TFEB, and reduces accumulation of autofluorescent storage material in SBF cortex to improve locomotor activities in Cln3Δex7/8 (Cln3ΔJNCL) mice. Aggravation of disease pathology in Cln3ΔJNCL mice lacking PPARα (Cln3ΔJNCLΔPPARα) and inability of gemfibrozil to decrease SCMAS accumulation, reduce glial activation, and improve locomotor performance of Cln3ΔJNCLΔPPARα mice delineates an important role of PPARα in this process. These studies highlight a new property of gemfibrozil and indicate its possible therapeutic use in JNCL patients.


Asunto(s)
Lipofuscinosis Ceroideas Neuronales , PPAR alfa , Ratones , Animales , Gemfibrozilo/farmacología , Lipofuscinosis Ceroideas Neuronales/tratamiento farmacológico , Lipofuscinosis Ceroideas Neuronales/patología , Neuroglía/patología , Microglía/patología , Modelos Animales de Enfermedad , Glicoproteínas de Membrana/genética , Chaperonas Moleculares/genética
2.
Int J Mol Sci ; 25(1)2023 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-38203431

RESUMEN

Recent studies observed, despite an anti-hyperlipidaemic effect, a positive impact of fibrates on septic conditions. This study evaluates the effects of gemfibrozil on microcirculatory variables, mitochondrial function, and lipid peroxidation levels with regard to its potential role as an indicator for oxidative stress in the colon and liver under control and septic conditions and dependencies on PPARα-mediated mechanisms of action. With the approval of the local ethics committee, 120 Wistar rats were randomly divided into 12 groups. Sham and septic animals were treated with a vehicle, gemfibrozil (30 and 100 mg/kg BW), GW 6471 (1 mg/kg BW, PPARα inhibitor), or a combination of both drugs. Sepsis was induced via the colon ascendens stent peritonitis (CASP) model. Then, 24 h post sham or CASP surgery, a re-laparotomy was performed. Measures of vital parameters (heart rate (HR), mean arterial pressure (MAP), and microcirculation (µHbO2)) were recorded for 90 min. Mitochondrial respirometry and assessment of lipid peroxidation via a malondialdehyde (MDA) assay were performed on colon and liver tissues. In the untreated sham animals, microcirculation remained stable, while pre-treatment with gemfibrozil showed significant decreases in the microcirculatory oxygenation of the colon. In the CASP animals, µHbO2 levels in the colon and the liver were significantly decreased 90 min after laparotomy. Pre-treatment with gemfibrozil prevented the microcirculatory aberrations in both organs. Gemfibrozil did not affect mitochondrial function and lipid peroxidation levels in the sham or CASP animals. Gemfibrozil treatment influences microcirculation depending on the underlying condition. Gemfibrozil prevents sepsis-induced microcirculatory aberrances in the colon and liver PPARα-independently. In non-septic animals, gemfibrozil impairs the microcirculatory variables in the colon without affecting those in the liver.


Asunto(s)
Enfermedades Transmisibles , Enfermedades Gastrointestinales , Peritonitis , Sepsis , Ratas , Animales , Gemfibrozilo/farmacología , Microcirculación , PPAR alfa , Ratas Wistar , Hígado , Peritonitis/tratamiento farmacológico , Sepsis/tratamiento farmacológico , Mitocondrias , Colon
3.
Int J Mol Sci ; 24(3)2023 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-36769295

RESUMEN

Gemfibrozil is a drug that has been used for over 40 years to lower triglycerides in blood. As a ligand for peroxisome proliferative-activated receptor-alpha (PPARα), which is expressed in many tissues, it induces the transcription of numerous genes for carbohydrate and lipid-metabolism. However, nothing is known about how intracellular lipid-homeostasis and, in particular, triglycerides are affected. As triglycerides are stored in lipid-droplets, which are known to be associated with many diseases, such as Alzheimer's disease, cancer, fatty liver disease and type-2 diabetes, treatment with gemfibrozil could adversely affect these diseases. To address the question whether gemfibrozil also affects intracellular lipid-levels, SH-SY5Y, HEK and Calu-3 cells, representing three different metabolically active organs (brain, lung and kidney), were incubated with gemfibrozil and subsequently analyzed semi-quantitatively by mass-spectrometry. Importantly, all cells showed a strong increase in intracellular triglycerides (SH-SY5Y: 170.3%; HEK: 272.1%; Calu-3: 448.1%), suggesting that the decreased triglyceride-levels might be due to an enhanced cellular uptake. Besides the common intracellular triglyceride increase, a cell-line specific alteration in acylcarnitines are found, suggesting that especially in neuronal cell lines gemfibrozil increases the transport of fatty acids to mitochondria and therefore increases the turnover of fatty acids for the benefit of additional energy supply, which could be important in diseases, such as Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Neuroblastoma , Humanos , Gemfibrozilo/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Neuroblastoma/tratamiento farmacológico , Triglicéridos/metabolismo , Ácidos Grasos , Hipolipemiantes/farmacología , Hipolipemiantes/uso terapéutico
4.
J Neurosci ; 41(10): 2287-2300, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33514677

RESUMEN

Parkinson's disease (PD) is the most common neurodegenerative movement disorder in humans. Despite intense investigations, effective therapies are not yet available to halt the progression of PD. Gemfibrozil, a Food and Drug Administration-approved lipid-lowering drug, is known to decrease the risk of coronary heart disease by increasing the level of high-density lipoprotein cholesterol and decreasing the level of low-density lipoprotein cholesterol. This study underlines the importance of gemfibrozil in protecting dopaminergic neurons in an animal model of PD. Oral administration of the human equivalent dose of gemfibrozil protected tyrosine hydroxylase (TH)-positive dopaminergic neurons in the substantia nigra pars compacta and TH fibers in the striatum of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-insulted mice of both sexes. Accordingly, gemfibrozil also normalized striatal neurotransmitters and improved locomotor activities in MPTP-intoxicated mice. Gemfibrozil-mediated protection of the nigrostriatal and locomotor activities in WT but not PPARα-/- mice from MPTP intoxication suggests that gemfibrozil needs the involvement of peroxisome proliferator-activated receptor α (PPARα) in protecting dopaminergic neurons. While investigating further mechanisms, we found that gemfibrozil stimulated the transcription of glial-derived neurotrophic factor (GDNF) gene in astrocytes via PPARα and that gemfibrozil protected nigral neurons, normalized striatal fibers and neurotransmitters, and improved locomotor activities in MPTP-intoxicated Gfafcre mice, but not GdnfΔastro mice lacking GDNF in astrocytes. These findings highlight the importance of the PPARα-dependent astroglial GDNF pathway in gemfibrozil-mediated protection of dopaminergic neurons in an animal model of PD and suggest the possible therapeutic use of gemfibrozil in PD patients.SIGNIFICANCE STATEMENT Increasing the level of glial cell-derived neurotrophic factor (GDNF) in the brain is important for the protection of dopamine neurons in Parkinson's disease (PD). Although gene manipulation and GDNF protein infusion into the brain are available options, it seems from the therapeutic angle that the best option would be to stimulate/induce the production of GDNF in vivo in the brain of PD patients. Here, we delineate that gemfibrozil, a lipid-lowering drug, stimulates GDNF in astrocytes via peroxisome proliferator-activated receptor α (PPARα). Moreover, gemfibrozil protected nigral neurons, normalized striatal fibers and neurotransmitters, and improved locomotor activities from MPTP toxicity via the PPARα-dependent astroglial GDNF pathway. These studies highlight a new property of gemfibrozil and suggest its possible therapeutic use in PD patients.


Asunto(s)
Astrocitos/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Gemfibrozilo/farmacología , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , PPAR alfa/metabolismo , Trastornos Parkinsonianos/patología , Animales , Astrocitos/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Inhibidores del Citocromo P-450 CYP2C8/farmacología , Neuronas Dopaminérgicas/metabolismo , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Trastornos Parkinsonianos/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Chem Res Toxicol ; 35(7): 1257-1266, 2022 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-35763595

RESUMEN

Gemfibrozil (GEM), a lipid regulator, is a fibric acid derivative widely used in the treatment of hyperlipidemia. It has been reported that GEM can induce acute liver injury in the course of therapy in clinical practice, so it is necessary to elucidate the mechanisms of toxic action. The present study focused on metabolic activation of GEM, possibly participating in GEM-mediated liver injury. A benzylic alcohol metabolite (M1), along with a phenol metabolite (M2), was detected in microsomal incubations, rat primary hepatocyte culturing, and rats given GEM. A GSH conjugate (M3) was detected in cultured rat hepatocytes after exposure to GEM. Formation of M1 was found to be NADPH dependent, and generation of M3 required M1 and 3'-phosphoadenosine-5'-phosphosulfate. It is most likely that GEM was biotransformed to M1, which was further metabolized to a sulfate. The resulting sulfate was reactive to bio-thiols. Cytochrome P450 and sulfotransferases participated in the phase I and phase II reactions, respectively. M1 and M3 were chemically synthesized, and their structures were characterized by mass spectrometry and NMR. The present study has particular value for elucidating the mechanism of liver injury caused by GEM.


Asunto(s)
Gemfibrozilo , Microsomas Hepáticos , Activación Metabólica , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Gemfibrozilo/metabolismo , Gemfibrozilo/farmacología , Microsomas Hepáticos/metabolismo , Ratas , Sulfatos/metabolismo , Sulfotransferasas/metabolismo
6.
Exp Cell Res ; 407(2): 112753, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34499887

RESUMEN

Peroxisome proliferator-activated receptor α (PPARα) activation has been reported to exert protective effects on podocytes, whereas angiopoietin-like 3 (ANGPTL3) has been shown to exert significant pathogenic effects on these cells. This study aimed to investigate the link between the protective effects of PPARα activation and the pathogenic effects of ANGPTL3 in podocytes. Both PPARα and ANGPTL3 were expressed in cultured podocytes. PPARα mRNA and protein levels decreased whereas ANGPTL3 mRNA and protein levels increased in a time-dependent manner in podocytes treated with puromycin aminonucleoside (PAN). Gemfibrozil, a pharmacological agonist of PPARα, increased PPARα levels and activity in podocytes. The drug also decreased ANGPTL3 levels by potentially weakening ANGPTL3 promoter activity in both normal and PAN-treated podocytes. Furthermore, gemfibrozil significantly decreased PAN-induced apoptosis and F-actin rearrangement. Primary podocytes from Angptl3-knockout mice were cultured. There was no significant difference between Angptl3-/- podocytes treated with or without gemfibrozil in the lamellipodia numbers after PAN treatment. The results suggested that the protective effects of gemfibrozil on podocytes were not exerted following knockout of the Angptl3 gene. This study identified a novel mechanism of the PPARα agonist gemfibrozil that exerts its protective effects by inhibiting PAN-induced apoptosis and cytoskeleton rearrangements through inhibition of ANGPTL3 expression.


Asunto(s)
Citoesqueleto de Actina/efectos de los fármacos , Proteínas Similares a la Angiopoyetina/fisiología , Gemfibrozilo/farmacología , PPAR alfa/agonistas , Podocitos/efectos de los fármacos , Seudópodos/efectos de los fármacos , Puromicina Aminonucleósido/farmacología , Proteína 3 Similar a la Angiopoyetina , Animales , Apoptosis , Hipolipemiantes/farmacología , Ratones , Ratones Noqueados , Podocitos/metabolismo , Podocitos/patología , Factores Protectores , Seudópodos/metabolismo
7.
Gen Comp Endocrinol ; 296: 113537, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32540489

RESUMEN

High carbohydrate diet (HCD) can induce lipid metabolism disorder, characterized by excessive lipid in farmed fish. Peroxisome proliferator activated receptor-α (PPARα) plays an important role in lipid homeostasis. In this study, we hypothesize that PPARα can improve lipid metabolism in fish fed HCD. Fish (3.03 ± 0.11 g) were fed with three diets: control (30% carbohydrate), HCD (45% carbohydrate) and HCG (HCD supplemented with 200 mg/kg gemfibrozil, an agonist of PPARα) for eight weeks. The fish fed HCG had higher growth rate and protein effiency than those fed the HCD diet, whereas the opposite trend was observed in feed conversion ratio, hepatosomatic index and mesenteric fat index. Additionally, fish fed HCG significantly decreased lipid accumulation in the whole body, liver and adipose tissues compared to those fed the HCD diet. Furthermore, fish in the HCG group significantly increased the mRNA and protein expression and protein dephosphorylation of PPARα. The HCG group also significantly increased the mRNA level of the downstream target genes of PPARα, whereas the opposite trend occured in the mRNA level of lipolysis-related genes compared to the HCD group. Besides, fish in the HCG group remarkably decreased the contents of alanine aminotransferase, aspartate aminotransferase and malondialdehyde, whereas the opposite occured in the activities of antioxidative enzymes and anti-inflammatory cytokine genes compared to the HCD group. This study indicates that gemfibrozil can improve lipid metabolism and maintain high antioxidant and anti-inflammatory capacity through activating PPARα in Nile tilapia fed a high carbohydrate diet.


Asunto(s)
Cíclidos/metabolismo , Carbohidratos de la Dieta/farmacología , Conducta Alimentaria , Gemfibrozilo/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , PPAR alfa/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Antioxidantes/metabolismo , Composición Corporal/efectos de los fármacos , Cíclidos/sangre , Cíclidos/genética , Cíclidos/crecimiento & desarrollo , Dieta , Inflamación/genética , Inflamación/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estrés Fisiológico/genética
8.
Molecules ; 25(2)2020 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-31963528

RESUMEN

Metformin is the first-line drug for type 2 diabetes mellitus control. It is established that this drug traffics through OCT-2 and MATE-1 transporters in kidney tubular cells and is excreted in its unaltered form in the urine. Hereby, we provide evidence that points towards the metformin-dependent upregulation of OCT-2 and MATE-1 in the kidney via the transcription factor proliferator-activated receptor alpha (PPARα). Treatment of wild type mice with metformin led to the upregulation of the expression of OCT-2 and MATE-1 by 34% and 157%, respectively. An analysis in a kidney tubular cell line revealed that metformin upregulated PPARα and OCT-2 expression by 37% and 299% respectively. MK-886, a PPARα antagonist, abrogated the OCT-2 upregulation by metformin and reduced MATE-1 expression. Conversely, gemfibrozil, an agonist of PPARα, elicited the increase of PPARα, OCT-2, and MATE-1 expression by 115%, 144%, and 376%, respectively. PPARα knockout mice failed to upregulate both the expression of OCT-2 and MATE-1 in the kidney upon metformin treatment, supporting the PPARα-dependent metformin upregulation of the transporters in this organ. Taken together, our data sheds light on the metformin-induced mechanism of transporter modulation in the kidney, via PPARα, and this effect may have implications for drug safety and efficacy.


Asunto(s)
Riñón/química , Metformina/administración & dosificación , Proteínas de Transporte de Catión Orgánico/genética , Transportador 2 de Cátion Orgánico/genética , PPAR alfa/genética , Animales , Línea Celular , Gemfibrozilo/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Indoles/farmacología , Riñón/efectos de los fármacos , Masculino , Metformina/farmacología , Ratones , Regulación hacia Arriba/efectos de los fármacos
10.
Drug Metab Dispos ; 47(4): 377-385, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30630815

RESUMEN

A recent in vitro study suggested that CYP2C8 is essential in the metabolism of desloratadine, an H1 receptor antagonist. If the proposed biotransformation mechanism takes place in vivo in humans, desloratadine could serve as a selective CYP2C8 probe substrate in drug-drug interaction studies. Glucuronide metabolites of clopidogrel and gemfibrozil act as time-dependent inhibitors of CYP2C8, but they have not been compared clinically. We conducted a randomized crossover study in 11 healthy subjects to characterize the involvement of CYP2C8 in desloratadine metabolism and to compare the CYP2C8 inhibitory strength of clopidogrel (300 and 75 mg on two following days) with that of gemfibrozil (600 mg BID for 5 days). Compared with placebo (control), clopidogrel increased the area under the plasma concentration-time curve (AUC0-∞) and peak plasma concentration (C max) of desloratadine to 280% (P = 3 × 10-7) and 165% (P = 0.0006), respectively. The corresponding increases by gemfibrozil were to 462% (P = 4 × 10-7) and 174% (P = 0.0006). Compared with placebo, clopidogrel and gemfibrozil decreased 3-hydroxyloratadine AUC0-71h to 52% (P = 5 × 10-5) and 6% (P = 2 × 10-8), respectively. Moreover, the 3-hydroxydesloratadine:desloratadine AUC0-71 h ratios were 21% (P = 7 × 10-10) and 1.7% (P = 8 × 10-11) of control during the clopidogrel and gemfibrozil phases. Our results confirm that CYP2C8 plays a critical role in the formation of 3-hydroxydesloratadine in humans, making desloratadine a potential CYP2C8 probe substrate. Furthermore, the findings corroborate the previous estimates that clinically relevant doses of clopidogrel cause strong CYP2C8 inhibition, whereas those of gemfibrozil almost completely inactivate the enzyme in humans.


Asunto(s)
Clopidogrel/farmacología , Inhibidores del Citocromo P-450 CYP2C8/envenenamiento , Citocromo P-450 CYP2C8/metabolismo , Gemfibrozilo/farmacología , Loratadina/análogos & derivados , Adulto , Área Bajo la Curva , Hidrocarburo de Aril Hidroxilasas/metabolismo , Biotransformación/fisiología , Estudios Cruzados , Interacciones Farmacológicas/fisiología , Femenino , Genotipo , Glucurónidos/farmacología , Humanos , Hipolipemiantes/envenenamiento , Loratadina/farmacología , Masculino , Adulto Joven
11.
Zhonghua Gan Zang Bing Za Zhi ; 27(7): 533-540, 2019 Jul 20.
Artículo en Zh | MEDLINE | ID: mdl-31357780

RESUMEN

Objective: To analyze non-alcoholic steatohepatitis (NASH)-related differentially expressed genes (DEGs) by bioinformatics methods to find key pathways and potential therapeutic targets for NASH. Methods: GSE61260 chip was downloaded from the public microarray database and liver biopsy samples from 24 NASH cases and 38 healthy controls were included. The Limma software package in R language was used to screen DEGs under the condition of difference multiple > 1.5 and adj. P < 0.05. The clusterProfiler software package was used for GO analysis and KEGG analysis. The STRING online database was used for protein-protein interaction analysis, and the L1000 and DrugBank databases were used for drug prediction. Results: Compared with healthy control group, 857 DEGs were screened out in NASH group including 167 up-regulated genes and 690 down-regulated genes. GO analysis showed that DEGs were mainly involved in inflammation and cholesterol metabolism. KEGG analysis showed that DEGs were mainly enriched in PPAR, non-alcoholic fatty liver disease, oxidative phosphorylation and other signaling pathways. Among them, eight genes of ACSL4, CYP7A1, FABP4, FABP5, lipoprotein lipase, ME1, OLR1 and PLIN1 were enriched in PPAR signaling pathway, and 165 interaction nodes were formed with 47 DEGs-encoded proteins. Lipoprotein lipase interacted with 21 DEGs, and its up-regulated expression had improved lipid metabolism, insulin resistance and anti-inflammatory effects. Four drugs (gemfibrozil, bezafibrate, omega-3 carboxylic acid and glycyrrhizic acid) were screened by L1000 and DrugBank to activate lipoprotein lipase. Presently, these four drugs are clinically used to treat hypertriglyceridemia or to improve inflammation. In this regard, we speculated that the pharmacological effects of these four drugs had improved NASH by activating lipoprotein lipase to promote liver lipid metabolism and alleviate inflammation. Conclusion: PPAR signaling pathway is closely associated to the occurrence and development of NASH, and thereby lipoprotein lipase agonist is a new attempt to treat NASH.


Asunto(s)
Activadores de Enzimas/farmacología , Metabolismo de los Lípidos , Lipoproteína Lipasa/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/genética , Bezafibrato/farmacología , Biopsia , Ácidos Carboxílicos/farmacología , Estudios de Casos y Controles , Biología Computacional , Gemfibrozilo/farmacología , Ácido Glicirrínico/farmacología , Humanos , Enfermedad del Hígado Graso no Alcohólico/enzimología , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Depuradores de Clase E
12.
Int J Obes (Lond) ; 42(3): 391-397, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28990588

RESUMEN

OBJECTIVE: Resistance at the brain receptors for leptin and insulin has been associated with increased feeding, obesity and cognitive impairments. The causal agent for central resistance is unknown but could be derived from the blood. Here we postulate whether hypertriglyceridemia, the major dyslipidemia of the metabolic syndrome, could underlie central leptin and insulin resistance. DESIGN: We used radioactively labeled triglycerides to measure blood-brain barrier (BBB) penetration, western blots to measure receptor activation, and feeding and cognitive tests to assess behavioral endpoints. RESULTS: Human CSF was determined to contain triglycerides, a finding previously unclear. The radioactive triglyceride triolein readily crossed the BBB and centrally administered triolein and peripherally administered lipids induced in vivo leptin and/or insulin resistance at hypothalamic receptors. Central triolein blocked the satiety effect of centrally administered leptin. Decreasing serum triglycerides with gemfibrozil improved both learning and memory inversely proportionate to triglyceride levels. CONCLUSIONS: Triglycerides cross the blood-brain barrier rapidly, are found in human cerebrospinal fluid, and induce central leptin and insulin receptor resistance, decreasing satiety and cognition.


Asunto(s)
Antígenos CD/metabolismo , Barrera Hematoencefálica/metabolismo , Resistencia a la Insulina/fisiología , Leptina/metabolismo , Receptor de Insulina/metabolismo , Triglicéridos/metabolismo , Anciano , Animales , Cognición/efectos de los fármacos , Femenino , Gemfibrozilo/farmacología , Humanos , Leptina/farmacología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Respuesta de Saciedad/efectos de los fármacos , Triglicéridos/sangre , Triglicéridos/líquido cefalorraquídeo , Trioleína/metabolismo , Trioleína/farmacología
13.
Br J Clin Pharmacol ; 84(5): 972-986, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29381228

RESUMEN

AIMS: Statistically significant positive correlations are reported for the abundance of hepatic drug-metabolizing enzymes. We investigate, as an example, the impact of CYP3A4-CYP2C8 intercorrelation on the predicted interindividual variabilities of clearance and drug-drug interactions (DDIs) for repaglinide using physiologically based pharmacokinetic (PBPK) modelling. METHODS: PBPK modelling and simulation were employed using Simcyp Simulator (v15.1). Virtual populations were generated assuming intercorrelations between hepatic CYP3A4-CYP2C8 abundances derived from observed values in 24 human livers. A repaglinide PBPK model was used to predict PK parameters in the presence and absence of gemfibrozil in virtual populations, and the results were compared with a clinical DDI study. RESULTS: Coefficient of variation (CV) of oral clearance was 52.5% in the absence of intercorrelation between CYP3A4-CYP2C8 abundances, which increased to 54.2% when incorporating intercorrelation. In contrast, CV for predicted DDI (as measured by AUC ratio before and after inhibition) was reduced from 46.0% in the absence of intercorrelation between enzymes to 43.8% when incorporating intercorrelation: these CVs were associated with 5th/95th percentiles (2.48-11.29 vs. 2.49-9.69). The range of predicted DDI was larger in the absence of intercorrelation (1.55-77.06) than when incorporating intercorrelation (1.79-25.15), which was closer to clinical observations (2.6-12). CONCLUSIONS: The present study demonstrates via a systematic investigation that population-based PBPK modelling incorporating intercorrelation led to more consistent estimation of extreme values than those observed in interindividual variabilities of clearance and DDI. As the intercorrelations more realistically reflect enzyme abundances, virtual population studies involving PBPK and DDI should avoid using Monte Carlo assignment of enzyme abundance.


Asunto(s)
Carbamatos/farmacocinética , Citocromo P-450 CYP2C8/metabolismo , Citocromo P-450 CYP3A/metabolismo , Gemfibrozilo/farmacología , Piperidinas/farmacocinética , Adulto , Ensayos Clínicos como Asunto/estadística & datos numéricos , Simulación por Computador , Inhibidores del Citocromo P-450 CYP2C8/farmacología , Interacciones Farmacológicas , Femenino , Humanos , Hipoglucemiantes/farmacocinética , Masculino , Microsomas Hepáticos/metabolismo , Modelos Biológicos , Adulto Joven
14.
Biopharm Drug Dispos ; 39(9): 420-430, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30335192

RESUMEN

GDC-0810 was under development as an oral anti-cancer drug for the treatment of estrogen receptor-positive breast cancer as a single agent or in combination. In vitro data indicated that GDC-0810 is a potent inhibitor of OATP1B1/1B3. To assess clinical risk, a PBPK model was developed to predict the transporter drug-drug interaction (tDDI) between GDC-0810 and pravastatin in human. The PBPK model was constructed in Simcyp® by integrating in vitro and in vivo data for GDC-0810. The prediction of human pharmacokinetics (PK) was verified using GDC-0810 phase I clinical PK data. The Simcyp transporter DDI model was verified using known OATP1B1/1B3 inhibitors (rifampicin, cyclosporine and gemfibrozil) and substrate (pravastatin), prior to using the model to predict GDC-0810 tDDI. The effect of GDC-0810 on pravastatin PK was then predicted based on the proposed clinical scenarios. Sensitivity analysis was conducted on the parameters with uncertainty. The developed PBPK model described the PK profile of GDC-0810 reasonably well. In the tDDI verification, the model reasonably predicted pravastatin tDDI caused by rifampicin and gemfibrozil OATP1B1/3 inhibition but under-predicted tDDI caused by cyclosporine. The effect of GDC-0810 on pravastatin PK was predicted to be low to moderate (pravastatin Cmax ratios 1.01-2.05 and AUC ratio 1.04-2.23). The observed tDDI (Cmax ratio 1.20 and AUC ratio 1.41) was within the range of the predicted values. This work demonstrates an approach using a PBPK model to prospectively assess tDDI caused by a new chemical entity as an OATP1B1/3 uptake transporter inhibitor to assess clinical risk and to support development strategy.


Asunto(s)
Cinamatos/farmacología , Indazoles/farmacología , Transportador 1 de Anión Orgánico Específico del Hígado/antagonistas & inhibidores , Modelos Biológicos , Pravastatina/farmacocinética , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos/antagonistas & inhibidores , Área Bajo la Curva , Ciclosporina/farmacología , Interacciones Farmacológicas , Gemfibrozilo/farmacología , Humanos , Transportador 1 de Anión Orgánico Específico del Hígado/metabolismo , Rifampin/farmacología , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos/metabolismo
15.
Biopharm Drug Dispos ; 39(2): 88-98, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29240983

RESUMEN

Clopidogrel is predominantly hydrolyzed to clopidogrel carboxylic acid (CCA) by carboxylesterase 1, and subsequently CCA is glucuronidated to clopidogrel acyl glucuronide (CAG) by uridine diphosphate-glucuronosyltransferases (UGTs); however, the UGT isoenzymes glucuronidating CCA remain unidentified to date. In this study, the glucuronidation of CCA was screened with pooled human liver microsomes (HLMs) and 7 human recombinant UGT (rUGT) isoforms. Results indicated that rUGT2B7 exhibited the highest catalytical activity for the CCA glucuronidation as measured with a mean Vmax value of 120.9 pmol/min/mg protein, 3- to 12-fold higher than that of the other rUGT isoforms tested. According to relative activity factor approach, the relative contribution of rUGT2B7 to CCA glucuronidation was estimated to be 58.6%, with the minor contributions (3%) from rUGT1A9. Moreover, the glucuronidation of CCA followed Michaelis-Menten kinetics with a mean Km value of 372.9 µM and 296.4 µM for pooled HLMs and rUGT2B7, respectively, showing similar affinity for both. The formation of CAG was significantly inhibited by azidothymidine and gemfibrozil (well-characterized UGT2B7 substrates) in a concentration-dependent manner, or by fluconazole (a typical UGT2B7-selective inhibitor) in a time-dependent manner, for both HLMs and rUGT2B7, respectively. In addition, CCA inhibited azidothymidine glucuronidation (catalyzed almost exclusively by UGT2B7) by HLMs and rUGT2B7 in a concentration-dependent manner, indicating that CCA is a substrate of UGT2B7. These results reveal that UGT2B7 is the major enzyme catalyzing clopidogrel glucuronidation in the human liver, and that there is the potential for drug-drug interactions between clopidogrel and the other substrate drugs of UGT2B7.


Asunto(s)
Glucurónidos/metabolismo , Glucuronosiltransferasa/metabolismo , Ticlopidina/análogos & derivados , Clopidogrel , Interacciones Farmacológicas , Fluconazol/farmacología , Gemfibrozilo/farmacología , Glucuronosiltransferasa/antagonistas & inhibidores , Humanos , Isoenzimas/metabolismo , Cinética , Microsomas Hepáticos/metabolismo , Proteínas Recombinantes/metabolismo , Ticlopidina/metabolismo , Zidovudina/farmacología
16.
Biopharm Drug Dispos ; 39(2): 116-121, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29136681

RESUMEN

The fraction of substrate metabolized (fm ) can be used to estimate drug interactions and can be determined by comparison of the intrinsic clearances (CLint ) of victim drugs obtained from inhibited and uninhibited hepatic enzymes. Commercially available human liver microsomes were recently developed in which one cytochrome P450 (P450) isoform is selectively inactivated. These inactivated liver microsomes were used to evaluate the roles of P450 2C isoforms in the depletion and oxidation of probe substrates. Determination of CLint with sets of control and P450 2C9-inactivated liver microsomes yielded fm,P450 2C9 values of 0.69-1.0 for celecoxib, diclofenac and warfarin. Apparent minor contributions of P450 1A2/2C8/3A4 were seen in depletion assays, yielding ~1 for the sum of the fm values. Selectively inactivated liver microsomes were thereby shown to be potentially useful for determining the in vitro fm values for major P450 2C9 contributions to substrate oxidations. Metabolite formations from diclofenac and warfarin were suppressed by 62-84% by the replacement of control liver microsomes with P450 2C9-inactivated liver microsomes. R-, S- and racemic omeprazole and troglitazone oxidation activities by liver microsomes at multiple substrate concentrations were suppressed by 26-36% and 22-50%, respectively, when P450 2C19- and 2C8-inactivated liver microsomes were used in place of control liver microsomes. This study provides important information to help elucidate the different roles of P450 isoforms in metabolite formation at different substrate concentrations. The data obtained allow the fractions metabolized to be calculated for victim drugs.


Asunto(s)
Familia 2 del Citocromo P450/metabolismo , Microsomas Hepáticos/metabolismo , Hidrocarburos Aromáticos con Puentes/farmacología , Celecoxib/farmacocinética , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Diclofenaco/farmacocinética , Gemfibrozilo/farmacología , Humanos , NADPH-Ferrihemoproteína Reductasa/metabolismo , Isoformas de Proteínas/metabolismo , Teofilina/análogos & derivados , Teofilina/farmacología , Ticrinafeno/farmacología , Triazoles/farmacología , Warfarina/farmacocinética
17.
Ren Fail ; 40(1): 371-378, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29707997

RESUMEN

BACKGROUND/AIM: Cisplatin-induced nephrotoxicity in large proportion of patients. The aim of this work is to clarify the effect of combination of sildenafil and gemfibrozil on cisplatin-induced nephrotoxicity either before or after cisplatin treatment and determination of nephrotoxicity predictors among the measured tissue markers. METHODS: Thirty two adult male albino rats were divided into four equal groups (G) GI control, GII received cisplatin, GIII received sildenafil and gemfibrozil before cisplatin, GIV received sildenafil and gemfibrozil after cisplatin. Creatinine and urea were measured and animals were sacrificed and kidney was taken for histopathology. The following tissue markers were measured, heme oxygenase-1 (HO-1) activity, reduced glutathione, quantitative (real-time polymerase chain reaction) RT-PCR for gene expression of tumor necrosis factor alpha (TNF-α) and endothelial nitric oxide synthase (ENOS) level. RESULTS: GII developed AKI demonstrated by significantly high urea and creatinine and severe diffuse (80-90%) tubular necrosis. TNF-α was highly and significantly elevated while the rest of tissue markers were significantly reduced in GI1 compared to other groups. GIV showed better results compared to GIII. There was a significant positive correlation between creatinine and TNF-α when combining GI and GII while there were significant negative correlation between creatinine and other tissue markers in same groups. Linear regression analysis demonstrated that HO-1 was the independent predictor of AKI demonstrated by elevated creatinine among GI and GII. CONCLUSIONS: Combination of sildenafil and gemfibrozil can be used in treatment of cisplatin-induced nephrotoxicity. HO-1 is a promising target for prevention and/or treatment of cisplatin-induced nephrotoxicity.


Asunto(s)
Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Gemfibrozilo/uso terapéutico , Insuficiencia Renal/tratamiento farmacológico , Citrato de Sildenafil/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/análisis , Creatinina/sangre , Modelos Animales de Enfermedad , Quimioterapia Combinada/métodos , Gemfibrozilo/farmacología , Glutatión/análisis , Hemo-Oxigenasa 1/análisis , Hemo-Oxigenasa 1/metabolismo , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Neoplasias/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Ratas , Insuficiencia Renal/sangre , Insuficiencia Renal/inducido químicamente , Insuficiencia Renal/patología , Citrato de Sildenafil/farmacología , Resultado del Tratamiento , Urea/sangre
18.
J Neurochem ; 141(3): 423-435, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28199020

RESUMEN

Late Infantile Neuronal Ceroid Lipofuscinosis (LINCL) is a rare neurodegenerative disease caused by mutations in the Cln2 gene that leads to deficiency or loss of function of the tripeptidyl peptidase 1 (TPP1) enzyme. TPP1 deficiency is known to cause the accumulation of autofluoroscent lipid-protein pigments in brain. Similar to other neurodegenerative disorders, LINCL is also associated with neuroinflammation and neuronal damage. Despite investigations, no effective therapy is currently available for LINCL. Therefore, we administered gemfibrozil (gem), an food and drug administration (FDA)-approved lipid-lowering drug, which has been shown to stimulate lysosomal biogenesis and induce anti-inflammation, orally, at a dose of 7.5 mg/kg body wt/day to Cln2(-/-) mice. We observed that gem-fed Cln2(-/-) mice lived longer by more than 10 weeks and had better motor activity compared to vehicle (0.1% Methyl cellulose) treatment. Gem treatment lowered the burden of storage materials, increased anti-inflammatory factors like SOCS3 and IL-1Ra, up-regulated anti-apoptotic molecule like phospho-Bad, and reduced neuronal apoptosis in the brain of Cln2(-/-) mice. Collectively, this study reinforces a neuroprotective role of gem that may be of therapeutic interest in improving the quality of life in LINCL patients.


Asunto(s)
Gemfibrozilo/farmacología , Gemfibrozilo/uso terapéutico , Hipolipemiantes/farmacología , Hipolipemiantes/uso terapéutico , Longevidad/efectos de los fármacos , Lipofuscinosis Ceroideas Neuronales/tratamiento farmacológico , Aminopeptidasas/genética , Aminopeptidasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratones , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Lipofuscinosis Ceroideas Neuronales/patología , Serina Proteasas/genética , Serina Proteasas/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Tripeptidil Peptidasa 1 , Proteína Letal Asociada a bcl/metabolismo
19.
Can J Physiol Pharmacol ; 95(2): 199-205, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27918198

RESUMEN

While gemfibrozil and fenofibrate are prescribed for anti-dyslipidemia treatment, a rational basis for the use of these drugs for treatment of dyslipidemia with concurrent metabolic syndrome has not been established. In this study, wild-type and Pparα-null mice were fed gemfibrozil- or fenofibrate-containing diets for 14 days. Urine output (24 h) was monitored, and urine, serum, and liver and kidney tissues were subjected to toxicity assessment. A 2-month challenge followed by a 2-week wash-out was performed for gemfibrozil to determine urine output and the potential toxicity. A therapeutically equivalent dose of gemfibrozil was more effective than fenofibrate in increasing urine output. This regulatory effect was not observed in Pparα-null mice. In contrast, hepatomegaly induced by fenofibrate was more pronounced than that of gemfibrozil. No significant toxicity was observed in liver or kidney in the 2-month treatment with gemfibrozil. These data demonstrated PPARα mediates the increased urine output by fibrates. Considering the relative action on hepatomegaly and the regulatory effect on urine output, gemfibrozil may be the preferable drug to increase urine output. These results revealed a new pharmacodynamic effect of clinically prescribed PPARα agonists and suggested the potential value of gemfibrozil in modification of blood pressure.


Asunto(s)
Diuréticos/farmacología , Fenofibrato/farmacología , Gemfibrozilo/farmacología , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , PPAR alfa/metabolismo , Alanina Transaminasa/sangre , Aldosterona/sangre , Animales , Arginina Vasopresina/sangre , Aspartato Aminotransferasas/sangre , Nitrógeno de la Urea Sanguínea , Creatinina/sangre , Fenofibrato/efectos adversos , Gemfibrozilo/efectos adversos , Hepatomegalia/inducido químicamente , Homeostasis , Riñón/patología , Hígado/patología , Masculino , Ratones , Ratones Noqueados
20.
Biopharm Drug Dispos ; 38(9): 535-542, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28946176

RESUMEN

Gemfibrozil is a fibrate drug used widely for dyslipidemia associated with atherosclerosis. Clinically, both gemfibrozil and its phase II metabolite gemfibrozil 1-O-ß-glucuronide (gem-glu) are involved in drug-drug interaction (DDI). But the DDI risk caused by gem-glu between human and mice has not been compared. In this study, six volunteers were recruited and took a therapeutic dose of gemfibrozil for 3 days for examination of the gemfibrozil and gem-glu level in human. Male mice were fed a gemfibrozil diet (0.75%) for 7 days, following which a cocktail-based inhibitory DDI experiment was performed. Plasma samples and liver tissues from mice were collected for determination of gemfibrozil, gem-glu concentration and cytochrome p450 enzyme (P450) induction analysis. In human, the molar ratio of gem-glu/gemfibrozil was 15% and 10% at the trough concentration and the concentration at 1.5 h after the 6th dose. In contrast, this molar ratio at steady state in mice was 91%, demonstrating a 6- to 9-fold difference compared with that in human. Interestingly, a net induction of P450 activity and in vivo inductive DDI potential in mice was revealed. The P450 activity was not inhibited although the gem-glu concentration was high. These data suggested species difference of relative gem-glu exposure between human and mice, as well as a net inductive DDI potential of gemfibrozil in mouse model.


Asunto(s)
Inductores de las Enzimas del Citocromo P-450/farmacocinética , Sistema Enzimático del Citocromo P-450/efectos de los fármacos , Gemfibrozilo/análogos & derivados , Glucuronatos/farmacocinética , Hipolipemiantes/farmacocinética , Adulto , Animales , Inductores de las Enzimas del Citocromo P-450/administración & dosificación , Inductores de las Enzimas del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Gemfibrozilo/farmacocinética , Gemfibrozilo/farmacología , Glucuronatos/farmacología , Humanos , Hipolipemiantes/administración & dosificación , Hipolipemiantes/farmacología , Hígado/metabolismo , Masculino , Ratones , Especificidad de la Especie , Factores de Tiempo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA