Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-29524693

RESUMEN

Acetyl coenzyme A is involved in several key metabolic pathways. Its concentration can vary considerably in response to physiological or pathological conditions making it a potentially valuable biomarker. However, little information about the measurement and concentration of acetyl CoA in human whole blood is found in the literature. The aim of this study was the development of an accurate method for the determination of acetyl CoA in human whole blood by LC-MS/MS. The method, involving extraction from whole blood by a rapid protein precipitation procedure was thoroughly validated: limit of quantitation was 3.91 ng mL-1. Accuracy and precision were calculated at five concentrations and were within ±15%. The average endogenous level of acetyl CoA in human whole blood was determined in 17 healthy individuals to be 220.9 ng mL-1 (ranging from 124.0 to 308.0 ng mL-1). This represents, to our knowledge, the first report of acetyl CoA levels in human whole blood, and the first practical and reliable method for its determination.


Asunto(s)
Acetilcoenzima A/sangre , Cromatografía Líquida de Alta Presión/métodos , Espectrometría de Masas en Tándem/métodos , Adulto , Femenino , Humanos , Límite de Detección , Modelos Lineales , Masculino , Reproducibilidad de los Resultados
2.
PLoS One ; 13(3): e0192028, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29522513

RESUMEN

In cells, phosphorylation of pantothenic acid to generate phosphopantothenic acid by the pantothenate kinase enzymes is the first step in coenzyme A synthesis. Pantothenate kinase 2, the isoform localized in neuronal cell mitochondria, is dysfunctional in patients with pantothenate kinase-associated neurodegeneration. Fosmetpantotenate is a phosphopantothenic acid prodrug in clinical development for treatment of pantothenate kinase-associated neurodegeneration, which aims to replenish phosphopantothenic acid in patients. Fosmetpantotenate restored coenzyme A in short-hairpin RNA pantothenate kinase 2 gene-silenced neuroblastoma cells and was permeable in a blood-brain barrier model. The rate of fosmetpantotenate metabolism in blood is species-dependent. Following up to 700 mg/kg orally, blood exposure to fosmetpantotenate was negligible in rat and mouse, but measurable in monkey. Consistent with the difference in whole blood half-life, fosmetpantotenate dosed orally was found in the brains of the monkey (striatal dialysate) but was absent in mice. Following administration of isotopically labeled-fosmetpantotenate to mice, ~40% of liver coenzyme A (after 500 mg/kg orally) and ~50% of brain coenzyme A (after 125 µg intrastriatally) originated from isotopically labeled-fosmetpantotenate. Additionally, 10-day dosing of isotopically labeled-fosmetpantotenate, 12.5 µg, intracerebroventricularly in mice led to ~30% of brain coenzyme A containing the stable isotopic labels. This work supports the hypothesis that fosmetpantotenate acts to replace reduced phosphopantothenic acid in pantothenate kinase 2-deficient tissues.


Asunto(s)
Coenzima A/metabolismo , Modelos Animales de Enfermedad , Neurodegeneración Asociada a Pantotenato Quinasa/tratamiento farmacológico , Ácido Pantoténico/análogos & derivados , Profármacos/uso terapéutico , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Línea Celular Tumoral , Humanos , Macaca fascicularis , Masculino , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neurodegeneración Asociada a Pantotenato Quinasa/metabolismo , Ácido Pantoténico/farmacocinética , Ácido Pantoténico/farmacología , Ácido Pantoténico/uso terapéutico , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Profármacos/farmacocinética , Profármacos/farmacología , Interferencia de ARN , Ratas Sprague-Dawley , Especificidad de la Especie
3.
J Med Chem ; 58(3): 1159-83, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25590515

RESUMEN

We report on the development of a series of pyrimidine carboxylic acids that are potent and selective inhibitors of kynurenine monooxygenase and competitive for kynurenine. We describe the SAR for this novel series and report on their inhibition of KMO activity in biochemical and cellular assays and their selectivity against other kynurenine pathway enzymes. We describe the optimization process that led to the identification of a program lead compound with a suitable ADME/PK profile for therapeutic development. We demonstrate that systemic inhibition of KMO in vivo with this lead compound provides pharmacodynamic evidence for modulation of kynurenine pathway metabolites both in the periphery and in the central nervous system.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Enfermedad de Huntington/tratamiento farmacológico , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Pirimidinas/farmacología , Animales , Células CHO , Proliferación Celular/efectos de los fármacos , Cricetulus , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Enfermedad de Huntington/metabolismo , Quinurenina/metabolismo , Quinurenina 3-Monooxigenasa/metabolismo , Ratones , Modelos Moleculares , Estructura Molecular , Pirimidinas/síntesis química , Pirimidinas/química , Ratas , Relación Estructura-Actividad
4.
PLoS Curr ; 62014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24558637

RESUMEN

Huntington's disease is a neurodegenerative disorder caused by mutations in the CAG tract of huntingtin. Several studies in HD cellular and rodent systems have identified disturbances in cyclic nucleotide signaling, which might be relevant to pathogenesis and therapeutic intervention. To investigate whether selective phosphodiesterase (PDE) inhibitors can improve some aspects of disease pathogenesis in HD models, we have systematically evaluated the effects of a variety of cAMP and cGMP selective PDE inhibitors in various HD models. Here we present the lack of effect in a variety of endpoints of the PDE subtype selective inhibitor SCH-51866, a PDE1/5 inhibitor, in the R6/2 mouse model of HD, after chronic oral dosing.

5.
Drug Metab Dispos ; 40(12): 2297-306, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22942319

RESUMEN

Understanding whether regulation of tryptophan metabolites can ameliorate neurodegeneration is of high interest to investigators. A recent publication describes 3,4-dimethoxy-N-(4-(3-nitrophenyl)-5-(piperidin-1-ylmethyl)thiazol-2-yl)benzenesulfonamide (JM6) as a novel prodrug for the kynurenine 3-monooxygenase (KMO) inhibitor 3,4-dimethoxy-N-(4-(3-nitrophenyl)thiazol-2-yl)benzenesulfonamide (Ro-61-8048) that elicits therapeutic effects in mouse models of Huntington's and Alzheimer's diseases (Cell 145:863-874, 2011). Our evaluation of the metabolism and pharmacokinetics of JM6 and Ro-61-8048 indicate instead that Ro-61-8048 concentrations in mouse plasma after JM6 administration originate from a Ro-61-8048 impurity (<0.1%) in JM6. After a 0.05 mg/kg Ro-61-8048 oral dose alone or coadministered with 10 mg/kg JM6 to mice, the Ro-61-8048 areas under the concentration-time curves (AUCs) from 0 to infinity were similar (4300 and 4900 nM × h, respectively), indicating no detectable contributions of JM6 metabolism to the Ro-61-8048 AUCs. JM6 was stable in incubations under acidic conditions and Ro-61-8048 was not a product of JM6 metabolism in vitro (plasma, blood, or hepatic models). Species differences in the quantitative rate of oxidative metabolism indicate that major circulating JM6 metabolite(s) in mice are unlikely to be major in humans: JM6 is rapidly metabolized via the piperidyl moiety in mouse (forming an iminium ion reactive intermediate) but is slowly metabolized in human (in vitro), primarily via O-dealkylation at the phenyl ring. Our data indicate that JM6 is not a prodrug for Ro-61-8048 and is not a potent KMO inhibitor.


Asunto(s)
Profármacos/farmacocinética , Sulfonamidas/farmacocinética , Tiazoles/farmacocinética , Animales , Área Bajo la Curva , Línea Celular , Perros , Hepatocitos/enzimología , Hepatocitos/metabolismo , Humanos , Células de Riñón Canino Madin Darby , Masculino , Fase I de la Desintoxicación Metabólica , Ratones , Ratones Endogámicos C57BL , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/metabolismo , Ratas , Sulfonamidas/administración & dosificación , Tiazoles/administración & dosificación
6.
PLoS Curr ; 3: RRN1291, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22307216

RESUMEN

To evaluate the potential of memantine as a therapeutic agent for Huntington's disease (HD) we have undertaken a series of in vitro, ex vivo and whole animal studies to characterize its pharmacokinetics (PK) and pharmacodynamics (PD) in rats and mice. Results from these studies will enable determination of memantine exposures needed to engage the related functional PD marker and help predict the dose regimen for clinical trials to test its proposed mechanism of action; the selective blockade of extrasynaptic, but not synaptic, NMDA receptors. The studies reported here describe the PK of memantine in rats and mice at low (1 mg/kg) and high (10 mg/kg) doses. Our studies indicate that the clearance mechanisms of memantine in rats and mice are different from those in human, and that clearance needs to be taken into account when extrapolating to the human. In rats only, there is a significant metabolic contribution to memantine clearance at lower dose levels. While memantine is primarily cleared renally in all three species, the proportion of total systemic clearance above the glomerular filtration rate (GFR) is much higher in rats and mice (~13, 4.5, and 1.4 times higher than GFR in rats, mice, and humans, respectively), suggesting that the contribution of active transport to memantine elimination in rats and mice is more significant than in the human. In rats and mice, memantine had a short half-life (<4 h) and steep Cmax/Cmin ratios (>100). In the human, the half-life of memantine was reported to be very long (60-80 h) with a Cmax/Cmin ratio at steady state concentrations of ~1.5. A small change in the clearance of memantine - for example due to renal impairment or competition for the elimination pathway with a co-administered drug - will likely affect exposure and, therefore, the selectivity of memantine on NMDA receptors . The PK differences observed between these species demonstrate that the PK in mice and rats cannot be directly extrapolated to the human. Further, the relationship between the plasma concentration (and therefore dose) needed to elicit a mechanism-related in vivo functional effect (PD readout) while maintaining the selectivity of the extrasynaptic blockade of the NMDA receptors needs to be established before clinical trials can be appropriately planned.

7.
J Med Chem ; 53(2): 649-59, 2010 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-20000470

RESUMEN

The resurgence of tuberculosis (TB), the incidence of drug-resistant strains of Mycobacterium tuberculosis (MTB), and the coinfection between TB and HIV have led to serious infections, high mortality, and a global health threat, resulting in the urgent search for new classes of antimycobacterial agents. Herein, we report the identification of a novel class of tetrahydroindazole based compounds as potent and unique inhibitors of MTB. Compounds 6a, 6m, and 6q exhibited activity in the low micromolar range against replicating Mycobacterium tuberculosis (R-TB) phenotype, with minimum inhibitory concentrations (MICs) of 1.7, 1.9, and 1.9 muM, respectively, while showing no toxicity to Vero Ccells. Moreover, studies aimed to assess the in vitro metabolic stability of 6a and 6m in mouse liver microsomes and in vivo pharmacokinetic profiles in plasma levels gave satisfactory results. This research suggests that tetrahydroindazole based anti-TB compounds can serve as a promising lead scaffold in developing new drugs to combat tuberculosis infections.


Asunto(s)
Antituberculosos/síntesis química , Indazoles/síntesis química , Animales , Antituberculosos/farmacocinética , Antituberculosos/farmacología , Chlorocebus aethiops , Indazoles/farmacología , Indazoles/uso terapéutico , Ligandos , Ratones , Pruebas de Sensibilidad Microbiana , Microsomas Hepáticos , Mycobacterium tuberculosis/efectos de los fármacos , Relación Estructura-Actividad , Células Vero
9.
Drug Metab Dispos ; 35(4): 525-32, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17220241

RESUMEN

The pharmacokinetics, metabolism, and excretion of sitagliptin [MK-0431; (2R)-4-oxo-4-[3-(trifluoromethyl)-5,6-dihydro[1,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]-1-(2,4,5-trifluorophenyl)butan-2-amine], a potent dipeptidyl peptidase 4 inhibitor, were evaluated in male Sprague-Dawley rats and beagle dogs. The plasma clearance and volume of distribution of sitagliptin were higher in rats (40-48 ml/min/kg, 7-9 l/kg) than in dogs ( approximately 9 ml/min/kg, approximately 3 l/kg), and its half-life was shorter in rats, approximately 2 h compared with approximately 4 h in dogs. Sitagliptin was absorbed rapidly after oral administration of a solution of the phosphate salt. The absolute oral bioavailability was high, and the pharmacokinetics were fairly dose-proportional. After administration of [(14)C]sitagliptin, parent drug was the major radioactive component in rat and dog plasma, urine, bile, and feces. Sitagliptin was eliminated primarily by renal excretion of parent drug; biliary excretion was an important pathway in rats, whereas metabolism was minimal in both species in vitro and in vivo. Approximately 10 to 16% of the radiolabeled dose was recovered in the rat and dog excreta as phase I and II metabolites, which were formed by N-sulfation, N-carbamoyl glucuronidation, hydroxylation of the triazolopiperazine ring, and oxidative desaturation of the piperazine ring followed by cyclization via the primary amine. The renal clearance of unbound drug in rats, 32 to 39 ml/min/kg, far exceeded the glomerular filtration rate, indicative of active renal elimination of parent drug.


Asunto(s)
Inhibidores de la Dipeptidil-Peptidasa IV , Inhibidores Enzimáticos/farmacocinética , Hipoglucemiantes/farmacocinética , Pirazinas/farmacocinética , Triazoles/farmacocinética , Adenosina Desaminasa/metabolismo , Inhibidores de la Adenosina Desaminasa , Administración Oral , Animales , Bilis/metabolismo , Disponibilidad Biológica , Biotransformación , Ciclización , Dipeptidil Peptidasa 4/metabolismo , Perros , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/orina , Heces/química , Glucurónidos/metabolismo , Glicoproteínas/antagonistas & inhibidores , Glicoproteínas/metabolismo , Haplorrinos , Humanos , Hidroxilación , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/sangre , Hipoglucemiantes/orina , Técnicas In Vitro , Riñón/metabolismo , Masculino , Microsomas Hepáticos/metabolismo , Estructura Molecular , Oxidación-Reducción , Unión Proteica , Pirazinas/administración & dosificación , Pirazinas/sangre , Pirazinas/orina , Ratas , Ratas Sprague-Dawley , Fosfato de Sitagliptina , Especificidad de la Especie , Ésteres del Ácido Sulfúrico/metabolismo , Triazoles/administración & dosificación , Triazoles/sangre , Triazoles/orina
10.
Diabetes ; 54(10): 2988-94, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16186403

RESUMEN

Dipeptidyl peptidase (DPP)-IV inhibitors are a new approach to the treatment of type 2 diabetes. DPP-IV is a member of a family of serine peptidases that includes quiescent cell proline dipeptidase (QPP), DPP8, and DPP9; DPP-IV is a key regulator of incretin hormones, but the functions of other family members are unknown. To determine the importance of selective DPP-IV inhibition for the treatment of diabetes, we tested selective inhibitors of DPP-IV, DPP8/DPP9, or QPP in 2-week rat toxicity studies and in acute dog tolerability studies. In rats, the DPP8/9 inhibitor produced alopecia, thrombocytopenia, reticulocytopenia, enlarged spleen, multiorgan histopathological changes, and mortality. In dogs, the DPP8/9 inhibitor produced gastrointestinal toxicity. The QPP inhibitor produced reticulocytopenia in rats only, and no toxicities were noted in either species for the selective DPP-IV inhibitor. The DPP8/9 inhibitor was also shown to attenuate T-cell activation in human in vitro models; a selective DPP-IV inhibitor was inactive in these assays. Moreover, we found DPP-IV inhibitors that were previously reported to be active in models of immune function to be more potent inhibitors of DPP8/9. These results suggest that assessment of selectivity of potential clinical candidates may be important to an optimal safety profile for this new class of antihyperglycemic agents.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Dipeptidasas/antagonistas & inhibidores , Dipeptidil Peptidasa 4 , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/antagonistas & inhibidores , Hipoglucemiantes , Inhibidores de Proteasas/uso terapéutico , Animales , Dipeptidil Peptidasa 4/genética , Dipeptidil Peptidasa 4/fisiología , Perros , Femenino , Humanos , Hipoglucemiantes/uso terapéutico , Hipoglucemiantes/toxicidad , Isoleucina/análogos & derivados , Isoleucina/química , Isoleucina/uso terapéutico , Isoleucina/toxicidad , Isomerismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidores de Proteasas/toxicidad , Ratas , Proteínas Recombinantes/antagonistas & inhibidores , Tiazoles/química , Tiazoles/uso terapéutico , Tiazoles/toxicidad
11.
Drug Metab Dispos ; 33(1): 121-30, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15486074

RESUMEN

The current study evaluated the potential for two dipeptidyl peptidase-IV (DPP-IV) inhibitor analogs (1S)-1-(trans-4-([(4-trifluoromethoxyphenyl)sulfonyl]amino)cyclohexyl)-2-[(3S)-3-fluoropyrrolidin-1-yl]-2-oxoethanaminium chloride and (1S)-1-(trans-4-([(2,4-difluorophenyl)sulfonyl]amino)cyclohexyl)-2-[(3S)-3-fluoropyrrolidin-1-yl]-2-oxoethanaminium chloride (MRL-A and MRL-B), containing a fluoropyrrolidine moiety in the structure, to undergo metabolic activation. The irreversible binding of these tritium-labeled compounds to rat liver microsomal protein was time- and NADPH-dependent and was attenuated by the addition of reduced glutathione (GSH) or N-acetylcysteine (NAC) to the incubation, indicating that chemically reactive intermediates were formed and trapped by these nucleophiles. Mass spectrometric analyses and further trapping experiments with semicarbazide indicated that the fluoropyrrolidine ring had undergone sequential oxidation and defluorination events resulting in the formation of GSH or NAC conjugates of the pyrrolidine moiety. The bioactivation of MRL-A was catalyzed primarily by rat recombinant CYP3A1 and CYP3A2. Pretreatment of rats with prototypic CYP3A1 and 3A2 inducers (pregnenolone-16alpha-carbonitrile and dexamethasone) enhanced the extent of bioactivation which, in turn, led to a higher degree of in vitro irreversible binding to microsomal proteins (5- and 9-fold increase, respectively). Herein, we describe studies that demonstrate that the fluoropyrrolidine ring is prone to metabolic activation and that GSH or NAC can trap the reactive intermediates to form adducts that provide insight into the mechanisms of bioactivation.


Asunto(s)
Dipeptidil Peptidasa 4/metabolismo , Microsomas Hepáticos/enzimología , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacología , Pirrolidinas/metabolismo , Animales , Biotransformación , Flúor/química , Flúor/metabolismo , Flúor/farmacología , Masculino , Inhibidores de Proteasas/química , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Pirrolidinas/química , Pirrolidinas/farmacología , Ratas , Ratas Sprague-Dawley
12.
Drug Metab Dispos ; 31(10): 1269-77, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12975337

RESUMEN

The pharmacokinetics and metabolism of the l-threo isoleucine thiazolidide dipeptidyl peptidase IV inhibitor, di-[2S,3S]-2-amino-3-methyl-pentanoic-1,3-thiazolidine fumarate (ILT-threo) and its allo stereoisomer (ILT-allo) were evaluated in rats, dogs, and monkeys. Both compounds were well absorbed (>80%) in all species, and most of the dose (>60%) was recovered in urine. Metabolites identified in all species included a sulfoxide (M1), a sulfone (M2), and a carbamoyl glucuronide (M3). For both compounds, parent drug had moderate systemic clearance in rats and dogs ( approximately 20-35 ml/min/kg in both species) and lower clearance in monkeys ( approximately 6-9 ml/min/kg). In rats, M1 was present in systemic circulation in concentrations similar to that of parent drug, whereas in dogs and monkeys, exposures to M1 were higher than for parent drug. In dogs, exposures to the sulfoxide metabolite were approximately 2 to 3 times higher after administration of ILT-allo than after administration of ILT-threo. Carbamoyl glucuronidation was an important biotransformation pathway in dogs. Circulating levels of M3 were significant in the dog, and present only in trace levels in rats and monkeys. M3 could be produced in in vitro systems in a NaHCO3 buffer under a CO2-saturated atmosphere and in the presence of UDP-glucuronic acid and alamethicin.


Asunto(s)
Aminas/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Glucurónidos/metabolismo , Inhibidores de Serina Proteinasa/farmacocinética , Animales , Perros , Haplorrinos , Masculino , Ratas , Ratas Sprague-Dawley , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/metabolismo , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...