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.
Sci Transl Med ; 11(492)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31092695

RESUMEN

Sebum plays important physiological roles in human skin. Excess sebum production contributes to the pathogenesis of acne vulgaris, and suppression of sebum production reduces acne incidence and severity. We demonstrate that sebum production in humans depends on local flux through the de novo lipogenesis (DNL) pathway within the sebocyte. About 80 to 85% of sebum palmitate (16:0) and sapienate (16:1n10) were derived from DNL, based on stable isotope labeling, much higher than the contribution of DNL to triglyceride palmitate in circulation (~20%), indicating a minor contribution by nonskin sources to sebum lipids. This dependence on local sebocyte DNL was not recapitulated in two widely used animal models of sebum production, Syrian hamsters and Göttingen minipigs. Confirming the importance of DNL for human sebum production, an acetyl-CoA carboxylase inhibitor, ACCi-1, dose-dependently suppressed DNL and blocked synthesis of fatty acids, triglycerides, and wax esters but not free sterols in human sebocytes in vitro. ACCi-1 dose-dependently suppressed facial sebum excretion by ~50% (placebo adjusted) in human individuals dosed orally for 2 weeks. Sebum triglycerides, wax esters, and free fatty acids were suppressed by ~66%, whereas non-DNL-dependent lipid species, cholesterol, and squalene were not reduced, confirming selective modulation of DNL-dependent lipids. Last, individuals with acne vulgaris exhibited increased sebum production rates relative to individuals with normal skin, with >80% of palmitate and sapienate derived from DNL. These findings highlight the importance of local sebocyte DNL for human skin sebaceous gland biology and illuminate a potentially exploitable therapeutic target for the treatment of acne vulgaris.


Asunto(s)
Acetil-CoA Carboxilasa/antagonistas & inhibidores , Acné Vulgar/enzimología , Inhibidores Enzimáticos/farmacología , Lipogénesis , Sebo/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Adolescente , Adulto , Animales , Células Cultivadas , Cricetinae , Inhibidores Enzimáticos/química , Femenino , Humanos , Lipogénesis/efectos de los fármacos , Masculino , Malonil Coenzima A/metabolismo , Persona de Mediana Edad , Ratas Wistar , Glándulas Sebáceas/efectos de los fármacos , Glándulas Sebáceas/metabolismo , Glándulas Sebáceas/patología , Sebo/efectos de los fármacos , Porcinos , Porcinos Enanos , Triglicéridos/biosíntesis , Adulto Joven
2.
Metabolism ; 97: 68-80, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31132381

RESUMEN

PURPOSE: Exercise is recommended in addition to pharmacotherapies for the management of type 2 diabetes, but metformin and exercise training may have non-additive or even inhibitory effects on exercise-induced improvements in glycemic control and exercise capacity. The objectives of this report were to determine if co-treatment with a sodium-glucose cotransporter-2 inhibitor and exercise could (1) further improve glycemic control when compared to either monotherapy and (2) not worsen exercise capacity when compared to exercise alone. METHODS: A rodent model of type 2 diabetes (30 mg/kg streptozotocin and high-fat feeding in male Sprague-Dawley rats) was used to assess 12 weeks of co-treatment with a sodium-glucose cotransporter 2 inhibitor (SGLT2i) and exercise (EX; treadmill running) on glycemic control and exercise capacity. Animals were randomized to the following conditions (n = 7-10/group): vehicle (0.5% methyl cellulose) sedentary (VEH SED), VEH EX, canagliflozin (3 mg kg-1 d-1) SED (SGLT2i SED), or SGLT2i EX. RESULTS: Both EX and SGLT2i independently improved indices of glycemic control. The combination of SGLT2i and EX further improved glucose tolerance (glucose area under the curve 1109 ±â€¯51 vs 1427 ±â€¯82 mmol/ L 120 min-1 for SGLT2i EX vs. SGLT2i SED, respectively; p < 0.05) and insulin responses (insulin area under the curve 24,524 ±â€¯4126 vs. 41,208 ±â€¯2714 pmol L-1 120 min-1 for SGLT2i EX vs. VEH EX, respectively; p < 0.05) during an oral glucose tolerance test. Only the combination of SGLT2i EX lowered body weight compared to VEH SED (p < 0.01). SGLT2i caused several metabolic adaptations including increased ketone production and a greater reliance on fat as a source of energy during normal cage activity. Interestingly, animals that were given the SGLT2i and underwent exercise training (SGLT2i EX) had better submaximal exercise capacity than EX alone, as indicated by distance run prior to fatigue (882 ±â€¯183 vs.433 ±â€¯33 m for SGLT2i EX and VEH EX, respectively; p < 0.01), and this was accompanied by a greater reliance on fat as an energy source during exercise (p < 0.01). CONCLUSIONS: If these findings with the combination of SGLT2i and exercise translate to humans, they will have important clinical health implications.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Tolerancia al Ejercicio/efectos de los fármacos , Condicionamiento Físico Animal/fisiología , Roedores/metabolismo , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Transportador 2 de Sodio-Glucosa/metabolismo , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Tolerancia al Ejercicio/fisiología , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa/métodos , Insulina/metabolismo , Masculino , Metformina/farmacología , Ratas , Ratas Sprague-Dawley
3.
J Med Chem ; 60(18): 7835-7849, 2017 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28853885

RESUMEN

Increased fructose consumption and its subsequent metabolism have been implicated in hepatic steatosis, dyslipidemia, obesity, and insulin resistance in humans. Since ketohexokinase (KHK) is the principal enzyme responsible for fructose metabolism, identification of a selective KHK inhibitor may help to further elucidate the effect of KHK inhibition on these metabolic disorders. Until now, studies on KHK inhibition with small molecules have been limited due to the lack of viable in vivo pharmacological tools. Herein we report the discovery of 12, a selective KHK inhibitor with potency and properties suitable for evaluating KHK inhibition in rat models. Key structural features interacting with KHK were discovered through fragment-based screening and subsequent optimization using structure-based drug design, and parallel medicinal chemistry led to the identification of pyridine 12.


Asunto(s)
Diseño de Fármacos , Fructoquinasas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Cristalografía por Rayos X , Fructoquinasas/química , Fructoquinasas/metabolismo , Humanos , Masculino , Simulación del Acoplamiento Molecular , Piridinas/química , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley
4.
Drug Metab Dispos ; 44(10): 1633-42, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27417179

RESUMEN

Unbound partition coefficient (Kpuu) is important to an understanding of the asymmetric free drug distribution of a compound between cells and medium in vitro, as well as between tissue and plasma in vivo, especially for transporter-mediated processes. Kpuu was determined for a set of compounds from the SLC13A family that are inhibitors and substrates of transporters in hepatocytes and transporter-transfected cell lines. Enantioselectivity was observed, with (R)-enantiomers achieving much higher Kpuu (>4) than the (S)-enantiomers (<1) in human hepatocytes and SLC13A5-transfected human embryonic 293 cells. The intracellular free drug concentration correlated directly with in vitro pharmacological activity rather than the nominal concentration in the assay because of the high Kpuu mediated by SLC13A5 transporter uptake. Delivery of the diacid PF-06649298 directly or via hydrolysis of the ethyl ester prodrug PF-06757303 resulted in quite different Kpuu values in human hepatocytes (Kpuu of 3 for diacid versus 59 for prodrug), which was successfully modeled on the basis of passive diffusion, active uptake, and conversion rate from ester to diacid using a compartmental model. Kpuu values changed with drug concentrations; lower values were observed at higher concentrations possibly owing to a saturation of transporters. Michaelis-Menten constant (Km) of SLC13A5 was estimated to be 24 µM for PF-06649298 in human hepatocytes. In vitro Kpuu obtained from rat suspension hepatocytes supplemented with 4% fatty acid free bovine serum albumin showed good correlation with in vivo Kpuu of liver-to-plasma, illustrating the potential of this approach to predict in vivo Kpuu from in vitro systems.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Simportadores/metabolismo , Animales , Cromatografía Liquida , Medios de Cultivo/metabolismo , Células HEK293 , Hepatocitos/metabolismo , Humanos , Técnicas In Vitro , Ratas , Cotransportador de Sodio-Sulfato , Espectrometría de Masas en Tándem
5.
J Med Chem ; 59(3): 1165-75, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26734723

RESUMEN

Inhibition of the sodium-coupled citrate transporter (NaCT or SLC13A5) has been proposed as a new therapeutic approach for prevention and treatment of metabolic diseases. In a previous report, we discovered dicarboxylate 1a (PF-06649298) which inhibits the transport of citrate in in vitro and in vivo settings via a specific interaction with NaCT. Herein, we report the optimization of this series leading to 4a (PF-06761281), a more potent inhibitor with suitable in vivo pharmacokinetic profile for assessment of in vivo pharmacodynamics. Compound 4a was used to demonstrate dose-dependent inhibition of radioactive [(14)C]citrate uptake in liver and kidney in vivo, resulting in modest reductions in plasma glucose concentrations.


Asunto(s)
Citratos/metabolismo , Malatos/química , Malatos/farmacología , Fenilbutiratos/química , Fenilbutiratos/farmacología , Piridinas/química , Piridinas/farmacología , Simportadores/antagonistas & inhibidores , Animales , Transporte Biológico/efectos de los fármacos , Glucemia/metabolismo , Citratos/farmacocinética , Relación Dosis-Respuesta a Droga , Células HEK293 , Hepatocitos/efectos de los fármacos , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Malatos/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Estructura Molecular , Fenilbutiratos/administración & dosificación , Piridinas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Simportadores/metabolismo
6.
Sci Rep ; 5: 17391, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26620127

RESUMEN

Citrate is a key regulatory metabolic intermediate as it facilitates the integration of the glycolysis and lipid synthesis pathways. Inhibition of hepatic extracellular citrate uptake, by blocking the sodium-coupled citrate transporter (NaCT or SLC13A5), has been suggested as a potential therapeutic approach to treat metabolic disorders. NaCT transports citrate from the blood into the cell coupled to the transport of sodium ions. The studies herein report the identification and characterization of a novel small dicarboxylate molecule (compound 2) capable of selectively and potently inhibiting citrate transport through NaCT, both in vitro and in vivo. Binding and transport experiments indicate that 2 specifically binds NaCT in a competitive and stereosensitive manner, and is recognized as a substrate for transport by NaCT. The favorable pharmacokinetic properties of 2 permitted in vivo experiments to evaluate the effect of inhibiting hepatic citrate uptake on metabolic endpoints.


Asunto(s)
Ácido Cítrico/metabolismo , Simportadores/antagonistas & inhibidores , Células HEK293 , Humanos , Transporte Iónico/efectos de los fármacos , Simportadores/genética , Simportadores/metabolismo
7.
PLoS One ; 9(5): e97139, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24858947

RESUMEN

Hyperglycemia resulting from type 2 diabetes mellitus (T2DM) is the main cause of diabetic complications such as retinopathy and neuropathy. A reduction in hyperglycemia has been shown to prevent these associated complications supporting the importance of glucose control. Glucokinase converts glucose to glucose-6-phosphate and determines glucose flux into the ß-cells and hepatocytes. Since activation of glucokinase in ß-cells is associated with increased risk of hypoglycemia, we hypothesized that selectively activating hepatic glucokinase would reduce fasting and postprandial glucose with minimal risk of hypoglycemia. Previous studies have shown that hepatic glucokinase overexpression is able to restore glucose homeostasis in diabetic models; however, these overexpression experiments have also revealed that excessive increases in hepatic glucokinase activity may also cause hepatosteatosis. Herein we sought to evaluate whether liver specific pharmacological activation of hepatic glucokinase is an effective strategy to reduce hyperglycemia without causing adverse hepatic lipids changes. To test this hypothesis, we evaluated a hepatoselective glucokinase activator, PF-04991532, in Goto-Kakizaki rats. In these studies, PF-04991532 reduced plasma glucose concentrations independent of changes in insulin concentrations in a dose-dependent manner both acutely and after 28 days of sub-chronic treatment. During a hyperglycemic clamp in Goto-Kakizaki rats, the glucose infusion rate was increased approximately 5-fold with PF-04991532. This increase in glucose infusion can be partially attributed to the 60% reduction in endogenous glucose production. While PF-04991532 induced dose-dependent increases in plasma triglyceride concentrations it had no effect on hepatic triglyceride concentrations in Goto-Kakizaki rats. Interestingly, PF-04991532 decreased intracellular AMP concentrations and increased hepatic futile cycling. These data suggest that hepatoselective glucokinase activation may offer glycemic control without inducing hepatic steatosis supporting the evaluation of tissue specific activators in clinical trials.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Activadores de Enzimas/farmacología , Glucoquinasa/metabolismo , Hiperglucemia/complicaciones , Hiperglucemia/tratamiento farmacológico , Imidazoles/farmacología , Hígado/efectos de los fármacos , Hígado/enzimología , Ácidos Nicotínicos/farmacología , Animales , Activadores de Enzimas/efectos adversos , Activadores de Enzimas/uso terapéutico , Glucosa/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Imidazoles/efectos adversos , Imidazoles/uso terapéutico , Hígado/patología , Masculino , Ácidos Nicotínicos/efectos adversos , Ácidos Nicotínicos/uso terapéutico , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Especificidad de Órganos , Ratas
8.
Circulation ; 123(2): 186-94, 2011 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-21200001

RESUMEN

BACKGROUND: Adipose tissue expands in response to excess caloric intake, but individuals prone to deposit visceral instead of subcutaneous adipose tissue have higher risk of metabolic disease. The role of angiogenesis in the expandability of human adipose tissue depots is unknown. The objective of this study was to measure angiogenesis in visceral and subcutaneous adipose tissue and to establish whether there is a relationship between obesity, metabolic status, and the angiogenic properties of these depots. METHODS AND RESULTS: Angiogenic capacity was determined by quantifying capillary branch formation from human adipose tissue explants embedded in Matrigel, and capillary density was assessed by immunohistochemistry. Subcutaneous adipose tissue had a greater angiogenic capacity than visceral tissue, even after normalization to its higher initial capillary density. Gene array analyses revealed significant differences in expression of angiogenic genes between depots, including an increased subcutaneous expression of angiopoietin-like protein 4, which is proangiogenic in an adipose tissue context. Subcutaneous capillary density and angiogenic capacity decreased with morbid obesity, and subcutaneous, but not visceral, adipose tissue angiogenic capacity correlated negatively with insulin sensitivity. CONCLUSIONS: These data imply that subcutaneous adipose tissue has a higher capacity to expand its capillary network than visceral tissue, but this capacity decreases with morbid obesity. The decrease correlates with insulin resistance, suggesting that impairment of subcutaneous adipose tissue angiogenesis may contribute to metabolic disease pathogenesis.


Asunto(s)
Grasa Intraabdominal/irrigación sanguínea , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Obesidad/fisiopatología , Grasa Subcutánea/irrigación sanguínea , Adulto , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/metabolismo , Índice de Masa Corporal , Derivación Gástrica , Humanos , Resistencia a la Insulina/fisiología , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/fisiopatología , Persona de Mediana Edad , Obesidad/metabolismo , Obesidad/cirugía , Grasa Subcutánea/metabolismo , Grasa Subcutánea/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA