Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Biol Sci ; 8(3): 310-27, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22355267

RESUMEN

Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a promising therapeutic target for treating coronary heart disease. We report a novel antibody 1B20 that binds to PCSK9 with sub-nanomolar affinity and antagonizes PCSK9 function in-vitro. In CETP/LDLR-hemi mice two successive doses of 1B20, administered 14 days apart at 3 or 10 mpk, induced dose dependent reductions in LDL-cholesterol (≥ 25% for 7-14 days) that correlated well with the extent of PCSK9 occupancy by the antibody. In addition, 1B20 induces increases in total plasma antibody-bound PCSK9 levels and decreases in liver mRNA levels of SREBP-regulated genes PCSK9 and LDLR, with a time course that parallels decreases in plasma LDL-cholesterol (LDL-C). Consistent with this observation in mice, in statin-responsive human primary hepatocytes, 1B20 lowers PCSK9 and LDLR mRNA levels and raises serum steady-state levels of antibody-bound PCSK9. In addition, mRNA levels of several SREBP regulated genes involved in cholesterol and fatty-acid synthesis including ACSS2, FDPS, IDI1, MVD, HMGCR, and CYP51A1 were decreased significantly with antibody treatment of primary human hepatocytes. In rhesus monkeys, subcutaneous (SC) dosing of 1B20 dose-dependently induces robust LDL-C lowering (maximal ~70%), which is correlated with increases in target engagement and total antibody-bound PCSK9 levels. Importantly, a combination of 1B20 and Simvastatin in dyslipidemic rhesus monkeys reduced LDL-C more than either agent alone, consistent with a mechanism of action that predicts additive effects of anti-PCSK9 agents with statins. Our results suggest that antibodies targeting PCSK9 could provide patients powerful LDL lowering efficacy on top of statins, and lower cardiovascular risk.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticolesterolemiantes/uso terapéutico , LDL-Colesterol/sangre , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Inmunización Pasiva , Síndrome Metabólico/terapia , Proproteína Convertasas/antagonistas & inhibidores , Proproteína Convertasas/inmunología , Serina Endopeptidasas/inmunología , Simvastatina/uso terapéutico , Proteínas de Unión a los Elementos Reguladores de Esteroles/fisiología , Animales , Anticuerpos Monoclonales/farmacología , Afinidad de Anticuerpos , Anticolesterolemiantes/administración & dosificación , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Proteínas de Transferencia de Ésteres de Colesterol/genética , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , Perfilación de la Expresión Génica , Células Hep G2/efectos de los fármacos , Células Hep G2/metabolismo , Hepatocitos/metabolismo , Humanos , Metabolismo de los Lípidos/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Macaca mulatta , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/genética , Ratones , Ratones Transgénicos , Proproteína Convertasa 9 , Proproteína Convertasas/biosíntesis , Proproteína Convertasas/genética , ARN Mensajero/metabolismo , Receptores de LDL/biosíntesis , Receptores de LDL/genética , Proteínas Recombinantes/metabolismo , Serina Endopeptidasas/biosíntesis , Serina Endopeptidasas/genética , Simvastatina/administración & dosificación
2.
J Cardiovasc Transl Res ; 5(1): 75-83, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22194019

RESUMEN

To assess cardiovascular risk in both clinical and basic research settings, it is imperative to be able to accurately measure plasma lipid levels. Here, methods commonly used to measure lipoproteins and lipids: ultracentrifugation (UC), fast protein liquid chromatography (FPLC), Roche auto-analyzer, and enzymatic assays were tested and compared. Plasma samples from 20 healthy humans and 22 cynomolgus monkeys were analyzed for their total cholesterol (TC), cholesterol in low density lipoproteins (LDL) and high density lipoproteins (HDL), and triglycerides (TG). Major lipid classes from UC and FPLC separated lipoprotein fractions from human plasma were further characterized by liquid chromatography-mass spectrometry analysis. All the tested methods showed acceptable performance with Roche analyzer among the best in approximate dilution linearity and recovery for most lipids as well as in repeatability between measurements of the same samples. TC, LDL, HDL, and TG values measured in human vs. monkey were-183.9 ± 35.5 (mean ± SD) vs. 105.6 ± 24.6 mg/dl, 106.0 ± 30.1 vs. 42.8 ± 13.0 mg/dl, 50.0 ± 11.4 vs. 53.4 ± 14.8 mg/dl, and 107.6 ± 50.7 vs. 58.0 ± 52.3 mg/dl. While no single method was uniformly the best, we recommend the Roche analyzer for routine measurements. UC or FPLC separation is needed for further functional characterization for specific lipid fraction. We have shown athero-protective profile in cynomolgus monkey compared with humans.


Asunto(s)
Técnicas de Química Analítica , Lípidos/sangre , Lipoproteínas/sangre , Adulto , Animales , Autoanálisis , Biomarcadores/sangre , Colesterol/sangre , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Cromatografía Liquida , Femenino , Humanos , Macaca fascicularis , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Reproducibilidad de los Resultados , Triglicéridos/sangre , Ultracentrifugación
3.
Metabolism ; 61(4): 470-81, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22001333

RESUMEN

The objective was to assess whether pharmacological activation of lecithin cholesterol acyltransferase (LCAT) could exert beneficial effects on lipoprotein metabolism. A putative small molecule activator (compound A) was used as a tool compound in in vitro and in vivo studies. Compound A increased LCAT activity in vitro in plasma from mouse, hamster, rhesus monkey, and human. To assess the acute pharmacodynamic effects of compound A, C57Bl/6 mice and hamsters received a single dose (20 mg/kg) of compound A. Both species displayed a significant increase in high-density lipoprotein cholesterol (HDLc) and a significant decrease in non-HDLc and triglycerides acutely after dosing; these changes tracked with ex vivo plasma LCAT activity. To examine compound A's chronic effect on lipoprotein metabolism, hamsters received a daily dosing of vehicle or of 20 or 60 mg/kg of compound A for 2 weeks. At study termination, compound treatment resulted in a significant increase in HDLc, HDL particle size, plasma apolipoprotein A-I level, and plasma cholesteryl ester (CE) to free cholesterol ratio, and a significant reduction in very low-density lipoprotein cholesterol. The increase in plasma CE mirrored the increase in HDL CE. Triglycerides trended toward a dose-dependent decrease in very low-density lipoprotein and HDL, with multiple triglyceride species reaching statistical significance. Gallbladder bile acids content displayed a significant and more than 2-fold increase with the 60 mg/kg treatment. We characterized pharmacological activation of LCAT by a small molecule extensively for the first time, and our findings support the potential of this approach in treating dyslipidemia and atherosclerosis; our analyses also provide mechanistic insight on LCAT's role in lipoprotein metabolism.


Asunto(s)
Activación Enzimática/fisiología , Lipoproteínas/metabolismo , Hígado/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Tiadiazoles/farmacología , Animales , Ácidos y Sales Biliares/sangre , Colesterol/sangre , Ésteres del Colesterol/sangre , Cricetinae , Activación Enzimática/efectos de los fármacos , Femenino , Lipoproteínas HDL/sangre , Hígado/enzimología , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Mesocricetus , Ratones , Ratones Endogámicos C57BL , Tiadiazoles/química , Triglicéridos/sangre
4.
J Lipid Res ; 53(1): 51-65, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22021650

RESUMEN

In an attempt to understand the applicability of various animal models to dyslipidemia in humans and to identify improved preclinical models for target discovery and validation for dyslipidemia, we measured comprehensive plasma lipid profiles in 24 models. These included five mouse strains, six other nonprimate species, and four nonhuman primate (NHP) species, and both healthy animals and animals with metabolic disorders. Dyslipidemic humans were assessed by the same measures. Plasma lipoprotein profiles, eight major plasma lipid fractions, and FA compositions within these lipid fractions were compared both qualitatively and quantitatively across the species. Given the importance of statins in decreasing plasma low-density lipoprotein cholesterol for treatment of dyslipidemia in humans, the responses of these measures to simvastatin treatment were also assessed for each species and compared with dyslipidemic humans. NHPs, followed by dog, were the models that demonstrated closest overall match to dyslipidemic humans. For the subset of the dyslipidemic population with high plasma triglyceride levels, the data also pointed to hamster and db/db mouse as representative models for practical use in target validation. Most traditional models, including rabbit, Zucker diabetic fatty rat, and the majority of mouse models, did not demonstrate overall similarity to dyslipidemic humans in this study.


Asunto(s)
Modelos Animales de Enfermedad , Dislipidemias/sangre , Lípidos/sangre , Animales , Cricetinae , Perros , Dislipidemias/tratamiento farmacológico , Ácidos Grasos/sangre , Humanos , Ratones , Primates , Simvastatina/uso terapéutico , Triglicéridos/sangre
5.
J Cardiovasc Transl Res ; 4(3): 373-83, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21394531

RESUMEN

Emerging evidence suggests apolipoprotein B (apoB) and apolipoprotein AI (apoAI) are strong risk predictors for atherosclerosis. Non-human primates (NHP), including rhesus monkeys, cynomolgus monkeys, and African green monkeys, are important preclinical species for studying dyslipidemia and atherosclerosis as they more closely resemble humans in lipid metabolism and disease physiology compared to lower species such as rodents. However, no commercial assays are currently available for measuring apoB and apoAI in NHP. We therefore evaluated analytical methods for routinely measuring apoB and apoAI in our NHP dyslipidemia and atherosclerosis research. Since NHP apoB and apoAI sequences are likely highly similar to human, we focused on the clinically validated and widely utilized human apoB and apoAI immunoturbidity assays. We carried out technical validation of these assays with NHP samples and leveraged orthogonal technical platforms including mass spectrometry, independent ELISA assay, and absolute quantitation via SDS-PAGE for further characterization. Analysis of purified lipoproteins demonstrated that the immunoturbidity assays detect NHP apoAI and apoB, with good dilution linearity and spike recovery from NHP plasma. Orthogonal studies showed apoAI correlated with protein concentration and apoB levels correlated with LC/MS and an independent ELISA. NHP samples from a drug treatment study were analyzed with the immunoturbidity assays and levels of apoB and apoAI fit our understanding of biology and expectations from literature. These studies serve as important technical and biological validation of the immunoturbidity assays for NHP samples, and demonstrate that these assays provide a high-throughput, fully automated analytical platform for NHP samples. Our studies pave the way for future translational research in NHP for developing therapies for treating dyslipidemia and atherosclerosis.


Asunto(s)
Apolipoproteína A-I/sangre , Apolipoproteínas B/sangre , Aterosclerosis/sangre , Dislipidemias/sangre , Inmunoensayo , Nefelometría y Turbidimetría , Animales , Aterosclerosis/diagnóstico , Aterosclerosis/tratamiento farmacológico , Biomarcadores/sangre , Calibración , Modelos Animales de Enfermedad , Dislipidemias/diagnóstico , Dislipidemias/tratamiento farmacológico , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Haplorrinos , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inmunoensayo/normas , Espectrometría de Masas , Nefelometría y Turbidimetría/normas , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Simvastatina/farmacología
6.
Eur J Pharmacol ; 650(1): 86-93, 2011 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-20955694

RESUMEN

Hydrogen sulfide (H(2)S) is a recently discovered gasotransmitter found in mammalian tissues and blood. Treatment with H(2)S donor molecules has shown promising results in preclinical models of inflammatory and cardiovascular diseases. Augmentation of H(2)S levels thus holds promise as a novel therapeutic approach for treatment of disease in man. Cystathionine ß-synthase (CBS) has been shown to catalyze H(2)S production in vitro. CBS enzyme activity is allosterically regulated by the endogenous activator S-adenosyl methionine. This mode of regulation suggests the possibility for designing a small molecule activator of CBS to enhance H(2)S production. This hypothesis, however, has not been directly tested in vivo. We show here that CBS contributes significantly to endogenous H(2)S production in mice: adenovirus mediated over expression of CBS in the liver significantly increased circulating levels of H(2)S, whereas CBS deficiency resulted in reduced levels. We demonstrate that CBS enzyme from endogenous sources can be activated by S-adenosyl methionine to a greater extent compared to recombinant enzyme, suggesting greater potential for activation than previously anticipated. Importantly, we show that circulating H(2)S levels are increased by pharmacological activation of CBS in vivo; i.e. in the presence of the endogenous activator. Together, our data demonstrate that CBS activity partially regulates endogenous H(2)S in mice, and suggest that pharmacological activation of CBS is a promising approach for enhancing endogenous production of H(2)S for the treatment of cardiovascular and other diseases.


Asunto(s)
Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Ingeniería Genética , Homocisteína/sangre , Sulfuro de Hidrógeno/sangre , Adenoviridae/genética , Animales , Activación Enzimática/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , S-Adenosilmetionina/farmacología
7.
Assay Drug Dev Technol ; 7(5): 495-506, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19715455

RESUMEN

Label-free mass spectrometric (MS) technologies are particularly useful for enzyme assay design for drug discovery screens. MS permits the selective detection of enzyme substrates or products in a wide range of biological matrices without need for derivatization, labeling, or capture technologies. As part of a cardiovascular drug discovery effort aimed at finding modulators of cystathionine beta-synthase (CBS), we used the RapidFire((R)) label-free high-throughput MS (HTMS) technology to develop a high-throughput screening (HTS) assay for CBS activity. The in vitro assay used HTMS to quantify the unlabeled product of the CBS reaction, cystathionine. Cystathionine HTMS analyses were carried out with a throughput of 7 s per sample and quantitation over a linear range of 80-10,000 nM. A compound library of 25,559 samples (or 80 384-well plates) was screened as singlets using the HTMS assay in a period of 8 days. With a hit rate of 0.32%, the actives showed a 90% confirmation rate. The in vitro assay was applied to secondary screens in more complex matrices with no additional analytical development. Our results show that the HTMS method was useful for screening samples containing serum, for cell-based assays, and for liver explants. The novel extension of the in vitro analytical method, without modification, to secondary assays resulted in a significant and advantageous economy of development time for the drug discovery project.


Asunto(s)
Cistationina/análisis , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Animales , Calibración , Línea Celular , Cromatografía Líquida de Alta Presión , Interpretación Estadística de Datos , Relación Dosis-Respuesta a Droga , Humanos , Indicadores y Reactivos , Cinética , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Espectrometría de Masa por Ionización de Electrospray
8.
J Alzheimers Dis ; 3(2): 221-229, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12214063

RESUMEN

Epidemiological studies have demonstrated that hypercholsterolemia is a significant risk factor for Alzheimer's disease (AD). The mechanism by which increased cholesterol may contribute to AD is unknown. However, as the generation and accumulation of the amyloid Abeta peptide in the brain appears to be significant for the initiation and progression of AD, it is possible that cholesterol levels can regulate Abeta formation and/or clearance. To test the effects of altering cholesterol on Abeta formation, we incubated cells in the presence of lipid depleted serum, with or without the active metabolite of the HMG-CoA reductase inhibitor lovastatin. After confirming that cholesterol was depleted in the cells, we then measured the fraction of Abeta formed from its precursor betaPP under each condition. We observed that cholesterol depletion led to a profound decrease in the levels of Abeta released from the cells. This effect of lovastatin acid was observed at concentrations of 0.05-5 &mgr;M, ranges where this compound is effective at inhibiting HMG-CoA reductase, thereby inhibiting cholesterol synthesis. In contrast, the release of an additional AbetaPP fragment, AbetaPPs, was only modestly reduced by cholesterol treatment. In further studies, we determined that the decreased release of Abeta was not due to its accumulation in the cell, but rather due to decreased formation of Abeta. Finally, we were able to exclude decreased maturation (glycosylation and sulfation) of newly synthesized AbetaPP as a cause for the effects of lovastatin acid on betaPP processing and Abeta formation. Our results demonstrate that reducing cellular cholesterol by the use of an HMG-CoA reductase inhibitor regulates Abeta formation. This effect may involve alterations in the trafficking of AbetaPP and/or alterations in the activity of the proteases that cleave AbetaPP. The results suggest a mechanism by which hypercholesterolemia may increase risk for AD and indicate that reduction in cholesterol may delay the onset and/or slow the progression of AD.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...