Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 9(1): 9046, 2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-31227742

RESUMEN

Obeticholic acid (OCA) and elafibranor (ELA) are selective and potent agonists for the farnesoid X receptor (FXR) and dual peroxisome proliferator-activated receptor α/δ (PPAR-α/δ), respectively. Both agents have demonstrated clinical efficacy in nonalcoholic steatohepatitis (NASH). The present study used OCA and ELA to compare the effects of mono- and combination therapies on metabolic and histological endpoints in Lepob/ob mice with established diet-induced and biopsy-confirmed NASH (ob/ob-NASH). ob/ob-NASH mice were fed the AMLN diet high in trans-fat, fructose and cholesterol for 15 weeks, whereafter they received vehicle, OCA (30 mg/kg, PO, QD), ELA (3, 10 mg/kg, PO, QD), or combinations (OCA + ELA) for eight weeks. Within-subject comparisons were performed on histomorphometric changes, including fractional area of liver fat, galectin-3 and Col1a1. OCA and ELA monotherapies improved all quantitative histopathological parameters and OCA + ELA combinations exerted additive effects on metabolic and histological endpoints. In agreement with their different molecular mechanisms of action, OCA and ELA monotherapies elicited distinct hepatic gene expression profiles and their combination led to profound transcriptome changes associated with further improvements in lipid handling and insulin signaling, suppression of immune responses and reduced extracellular matrix formation. In conclusion, these findings provide preclinical proof-of-concept for combined FXR and PPAR-α/δ agonist-based therapies in NASH.


Asunto(s)
Chalconas/uso terapéutico , Ácido Quenodesoxicólico/análogos & derivados , Cirrosis Hepática/tratamiento farmacológico , Propionatos/uso terapéutico , Animales , Biopsia , Ácido Quenodesoxicólico/uso terapéutico , Modelos Animales de Enfermedad , Cirrosis Hepática/patología , Ratones , PPAR alfa/metabolismo , Prueba de Estudio Conceptual , Transcripción Genética
2.
J Lipid Res ; 59(6): 982-993, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29559521

RESUMEN

Obeticholic acid (OCA) is a selective farnesoid X receptor (FXR) agonist that regulates bile acid and lipid metabolism. FXR activation induces distinct changes in circulating cholesterol among animal models and humans. The mechanistic basis of these effects has been elusive because of difficulties in studying lipoprotein homeostasis in mice, which predominantly package circulating cholesterol in HDLs. Here, we tested the effects of OCA in chimeric mice whose livers are mostly composed (≥80%) of human hepatocytes. Chimeric mice exhibited a human-like ratio of serum LDL cholesterol (LDL-C) to HDL cholesterol (HDL-C) at baseline. OCA treatment in chimeric mice increased circulating LDL-C and decreased circulating HDL-C levels, demonstrating that these mice closely model the cholesterol effects of FXR activation in humans. Mechanistically, OCA treatment increased hepatic cholesterol in chimeric mice but not in control mice. This increase correlated with decreased SREBP-2 activity and target gene expression, including a significant reduction in LDL receptor protein. Cotreatment with atorvastatin reduced total cholesterol, rescued LDL receptor protein levels, and normalized serum LDL-C. Treatment with two clinically relevant nonsteroidal FXR agonists elicited similar lipoprotein and hepatic changes in chimeric mice, suggesting that the increase in circulating LDL-C is a class effect of FXR activation.


Asunto(s)
Ácido Quenodesoxicólico/análogos & derivados , Quimera , Colesterol/metabolismo , Lipoproteínas/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Atorvastatina/farmacología , Ácido Quenodesoxicólico/farmacología , Colesterol/sangre , Humanos , Lipoproteínas/sangre , Hígado/citología , Masculino , Ratones
3.
World J Gastroenterol ; 24(2): 195-210, 2018 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-29375205

RESUMEN

AIM: To characterize the efficacy of the dual FXR/TGR5 receptor agonist INT-767 upon histological endpoints in a rodent model of diet-induced and biopsy-confirmed non-alcoholic steatohepatitis (NASH). METHODS: The effects of INT-767 on histological features of NASH were assessed in two studies using Lepob/ob (ob/ob) NASH mice fed the AMLN diet (high fat with trans-fat, cholesterol and fructose). In a proof-of-concept study, Lepob/ob (ob/ob) NASH mice were first dosed with INT-767 (3 or 10 mg/kg for 8 wk). A second ob/ob NASH study compared INT-767 (3 and 10 mg/kg) to obeticholic acid (OCA) (10 or 30 mg/kg; 16 wk). Primary histological endpoints included qualitative and quantitative assessments of NASH. Other metabolic and plasma endpoints were also assessed. A comparative assessment of INT-767 and OCA effects on drug distribution and hepatic gene expression was performed in C57Bl/6 mice on standard chow. C57Bl/6 mice were orally dosed with INT-767 or OCA (1-30 mg/kg) for 2 wk, and expression levels of candidate genes were assessed by RNA sequencing and tissue drug levels were measured by liquid chromatography tandem-mass spectrometry. RESULTS: INT-767 dose-dependently (3 and 10 mg/kg, PO, QD, 8 wk) improved qualitative morphometric scores on steatohepatitis severity, inflammatory infiltrates and fibrosis stage. Quantitative morphometric analyses revealed that INT-767 reduced parenchymal collagen area, collagen fiber density, inflammation (assessed by Galectin-3 immunohistochemistry) and hepatocyte lipid droplet area following INT-767 treatment. In a comparative study (16 wk), the FXR agonists OCA (10 and 30 mg/kg) and INT-767 (3 and 10 mg/kg) both improved NASH histopathology, with INT-767 exerting greater therapeutic potency and efficacy than OCA. Mechanistic studies suggest that both drugs accumulate similarly within the liver and ileum, however, the effects of INT-767 may be driven by enhanced hepatic, but not ileal, FXR function. CONCLUSION: These findings confirm the potential utility of FXR and dual FXR/TGR5 activation as disease intervention strategies in NASH.


Asunto(s)
Ácidos y Sales Biliares/farmacología , Dieta Alta en Grasa , Hígado/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Obesidad/tratamiento farmacológico , Animales , Ácidos y Sales Biliares/metabolismo , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/etiología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Cirrosis Hepática/prevención & control , Ratones Endogámicos C57BL , Ratones Obesos , Microscopía de Fluorescencia por Excitación Multifotónica , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Prueba de Estudio Conceptual , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Espectrometría de Masas en Tándem , Factores de Tiempo
4.
Pharmacol Rev ; 67(3): 564-600, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26071095

RESUMEN

Amylin is a pancreatic ß-cell hormone that produces effects in several different organ systems. Here, we review the literature in rodents and in humans on amylin research since its discovery as a hormone about 25 years ago. Amylin is a 37-amino-acid peptide that activates its specific receptors, which are multisubunit G protein-coupled receptors resulting from the coexpression of a core receptor protein with receptor activity-modifying proteins, resulting in multiple receptor subtypes. Amylin's major role is as a glucoregulatory hormone, and it is an important regulator of energy metabolism in health and disease. Other amylin actions have also been reported, such as on the cardiovascular system or on bone. Amylin acts principally in the circumventricular organs of the central nervous system and functionally interacts with other metabolically active hormones such as cholecystokinin, leptin, and estradiol. The amylin-based peptide, pramlintide, is used clinically to treat type 1 and type 2 diabetes. Clinical studies in obesity have shown that amylin agonists could also be useful for weight loss, especially in combination with other agents.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Animales , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético/fisiología , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Polipéptido Amiloide de los Islotes Pancreáticos/uso terapéutico , Pérdida de Peso/efectos de los fármacos
5.
Artículo en Inglés | MEDLINE | ID: mdl-24657407

RESUMEN

Peptide agonists of the glucagon-like peptide-1 receptor (GLP-1R) and the cholecystokinin-1 receptor (CCK1-R) have therapeutic potential because of their marked anorexigenic and weight lowering effects. Furthermore, recent studies in rodents have shown that co-administration of these agents may prove more effective than treatment either of the peptide classes alone. To correlate the pharmacodynamic effects to the pharmacokinetics of these peptide drugs in vivo, a sensitive and robust bioanalytical method is essential. Furthermore, the simultaneous determination of both analytes in plasma samples by a single method offers obvious advantages. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) is well suited to this goal through its ability to simultaneously monitor multiple analytes through selected reaction monitoring (SRM). However, it is a challenge to find appropriate conditions that allow two peptides with widely disparate physiochemical properties to be simultaneously analyzed while maintaining the necessary sensitivity for their accurate plasma concentrations. Herein, we report an on-line solid phase extraction (SPE) LC-MS/MS method for simultaneous quantification of the CCK1-R agonist AC170222 and the GLP-1R agonist AC3174 in rodent plasma. The assay has a linear range from 0.0975 to 100ng/mL, with lower limits of quantification of 0.0975ng/mL and 0.195ng/mL for AC3174 and AC170222, respectively. The intra- and inter-day precisions were below 15%. The developed LC-MS/MS method was used to simultaneously quantify AC3174 and AC170222, the results showed that the terminal plasma concentrations of AC3174 or AC170222 were comparable between groups of animals that were administered with the peptides alone (247±15pg/mL of AC3174 and 1306±48pg/mL of AC170222), or in combination (222±32pg/mL and 1136±47pg/mL of AC3174 and AC170222, respectively). These data provide information on the drug exposure to aid in assessing the combination effects of AC3174 and AC170222 on rodent metabolism.


Asunto(s)
Depresores del Apetito/análisis , Receptores de Colecistoquinina/agonistas , Receptores de Glucagón/agonistas , Extracción en Fase Sólida/métodos , Animales , Depresores del Apetito/aislamiento & purificación , Depresores del Apetito/farmacocinética , Depresores del Apetito/farmacología , Cromatografía Liquida/métodos , Receptor del Péptido 1 Similar al Glucagón , Límite de Detección , Masculino , Péptidos/sangre , Péptidos/aislamiento & purificación , Péptidos/farmacocinética , Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem/métodos
6.
Am J Physiol Gastrointest Liver Physiol ; 305(7): G483-95, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23886860

RESUMEN

Shortcomings of previously reported preclinical models of nonalcoholic steatohepatitis (NASH) include inadequate methods used to induce disease and assess liver pathology. We have developed a dietary model of NASH displaying features observed clinically and methods for objectively assessing disease progression. Mice fed a diet containing 40% fat (of which ∼18% was trans fat), 22% fructose, and 2% cholesterol developed three stages of nonalcoholic fatty liver disease (steatosis, steatohepatitis with fibrosis, and cirrhosis) as assessed by histological and biochemical methods. Using digital pathology to reconstruct the left lateral and right medial lobes of the liver, we made comparisons between and within lobes to determine the uniformity of collagen deposition, which in turn informed experimental sampling methods for histological, biochemical, and gene expression analyses. Gene expression analyses conducted with animals stratified by disease severity led to the identification of several genes for which expression highly correlated with the histological assessment of fibrosis. Importantly, we have established a biopsy method allowing assessment of disease progression. Mice subjected to liver biopsy recovered well from the procedure compared with sham-operated controls with no apparent effect on liver function. Tissue obtained by biopsy was sufficient for gene and protein expression analyses, providing the opportunity to establish an objective method of assessing liver pathology before subjecting animals to treatment. The improved assessment techniques and the observation that mice fed the high-fat diet exhibit many clinically relevant characteristics of NASH establish a preclinical model for identifying pharmacological interventions with greater likelihood of translating to the clinic.


Asunto(s)
Grasas de la Dieta/efectos adversos , Hígado Graso/etiología , Hígado Graso/patología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Hígado/metabolismo , Cirrosis Hepática/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico , Transcriptoma
7.
Curr Opin Endocrinol Diabetes Obes ; 20(1): 8-13, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23183359

RESUMEN

PURPOSE OF REVIEW: This review focuses on recent advances in receptor signaling, neurobiology, and pharmacological interactions of amylin with nutritive status, as well as other metabolism-related regulatory signals. RECENT FINDINGS: Manipulation of components of the amylin receptor complex revealed important roles for the accessory proteins of amylin receptors in energy balance. In-vitro findings point to potential novel sites of action and postreceptor signaling pathways activated by amylin. Neurobiological studies elucidated how amylin activation of hindbrain neural circuitry modulates hypothalamic signaling and responsiveness to leptin. The notion of 'amylin resistance' was addressed in several models (drug or diet-induced hyper-amylinemia). Finally, progress in the design and delivery of amylinomimetics is briefly discussed. SUMMARY: Collectively, these mechanistic studies deepen our understanding of the role of endogenous amylin in the regulation of appetite and adiposity, and hopefully will help guide research efforts towards the development of more effective amylin-based therapies for metabolic diseases.


Asunto(s)
Adiposidad/efectos de los fármacos , Depresores del Apetito/farmacología , Regulación del Apetito/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Obesidad/metabolismo , Transducción de Señal/efectos de los fármacos , Adiposidad/fisiología , Amiloide/metabolismo , Animales , Regulación del Apetito/fisiología , Metabolismo Energético , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/deficiencia , Leptina/metabolismo , Ratones , Ratones Endogámicos C57BL , Neurobiología/tendencias , Obesidad/tratamiento farmacológico , Farmacología/tendencias , Ratas , Ratas Endogámicas , Receptores de Polipéptido Amiloide de Islotes Pancreáticos/efectos de los fármacos , Receptores de Polipéptido Amiloide de Islotes Pancreáticos/metabolismo
8.
Eur J Pharmacol ; 698(1-3): 292-8, 2013 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-23178527

RESUMEN

Antagonism of opioid systems (e.g., with naltrexone) has been explored as an anti-obesity strategy, and is particularly effective when co-administered with dual inhibitors of dopamine and norepinephrine reuptake (e.g., bupropion). Previously, we demonstrated that amylin enhances the food intake lowering and weight loss effects of neurohormonal (e.g., leptin, cholecystokinin, melanocortins) and small molecule (e.g., phentermine, sibutramine) agents. Here, we sought to characterize the interaction of amylin with naltrexone/bupropion on energy balance. Wild-type and amylin knockout mice were similarly responsive to the food intake lowering effects of either naltrexone (1mg/kg, subcutaneous) or bupropion (50mg/kg, subcutaneous) suggesting that they act independently of amylinergic systems and could interact additively when given in combination with amylin. To test this, diet-induced obese rats were treated (for 11 days) with vehicle, rat amylin (50 µg/kg/d, infused subcutaneously), naltrexone/bupropion (1 and 20mg/kg, respectively by twice daily subcutaneous injection) or their combination. We found that amylin+naltrexone/bupropion combination therapy exerted additive effects to reduce cumulative food intake, body weight and fat mass. In a separate study, the effects of amylin and naltrexone/bupropion administered at the same doses (for 14 days) were compared to a pair-fed group. Although the combination and pair-fed groups lost a similar amount of body weight, rats treated with the combination lost 68% more fat and better maintained their lean mass. These findings support the strategy of combined amylin agonism with opioid and catecholaminergic signaling systems for the treatment of obesity.


Asunto(s)
Depresores del Apetito/farmacología , Peso Corporal/efectos de los fármacos , Bupropión/farmacología , Ingestión de Alimentos/efectos de los fármacos , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Naltrexona/farmacología , Animales , Composición Corporal/efectos de los fármacos , Dieta/efectos adversos , Interacciones Farmacológicas , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Polipéptido Amiloide de los Islotes Pancreáticos/deficiencia , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Masculino , Ratones , Obesidad/etiología , Obesidad/metabolismo , Obesidad/fisiopatología , Ratas , Factores de Tiempo
9.
Am J Physiol Gastrointest Liver Physiol ; 302(8): G762-72, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22268099

RESUMEN

These preclinical studies aimed to 1) increase our understanding the dietary induction of nonalcoholic steatohepatitis (NASH), and, 2) further explore the utility and mechanisms of glucagon-like peptide-1 receptor (GLP-1R) agonism in NASH. We compared the effects of a high trans-fat (HTF) or high lard fat (HLF) diet on key facets of nonalcoholic fatty liver disease (NAFLD)/NASH in Lep(ob)/Lep(ob) and C57BL6J (B6) mice. Although HLF-fed mice experienced overall greater gains in weight and adiposity, the addition of trans-fat better mirrored pathophysiological features of NASH (e.g., hepatomegaly, hepatic lipid, and fibrosis). Administration of AC3174, an exenatide analog, and GLP-1R agonist to Lep(ob)/Lep(ob) and B6 ameliorated hepatic endpoints in both dietary models. Next, we assessed whether AC3174-mediated improvements in diet-induced NASH were solely due to weight loss in HTF-fed mice. AC3174-treatment significantly reduced body weight (8.3%), liver mass (14.2%), liver lipid (12.9%), plasma alanine aminotransferase, and triglycerides, whereas a calorie-restricted, weight-matched group demonstrated only modest nonsignificant reductions in liver mass (9%) and liver lipid (5.1%) relative to controls. Treatment of GLP-1R-deficient (GLP-1RKO) mice with AC3174 had no effect on body weight, adiposity, liver or plasma indices pointing to the GLP-1R-dependence of AC3174's effects. Interestingly, the role of endogenous GLP-1Rs in NASH merits further exploration as the GLP-1RKO model was protected from the deleterious hepatic effects of HTF. Our pharmacological data further support the clinical evaluation of the utility of GLP-1R agonists for treatment of NASH.


Asunto(s)
Hígado Graso/tratamiento farmacológico , Péptidos/uso terapéutico , Receptores de Glucagón/agonistas , Animales , Composición Corporal/fisiología , Peso Corporal/efectos de los fármacos , Dieta , Dieta con Restricción de Grasas , Dieta Alta en Grasa , Determinación de Punto Final , Hígado Graso/inducido químicamente , Hígado Graso/metabolismo , Hígado Graso/patología , Expresión Génica/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón , Hormonas/sangre , Leptina/genética , Lípidos/química , Hígado/metabolismo , Hígado/patología , Pruebas de Función Hepática , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Enfermedad del Hígado Graso no Alcohólico , Receptores de Glucagón/genética , Ácidos Grasos trans/farmacología , Pérdida de Peso/efectos de los fármacos
10.
Br J Pharmacol ; 166(1): 121-36, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21671898

RESUMEN

The discoveries of the incretin hormone glucagon-like peptide-1 (GLP-1) and the ß-cell hormone amylin have translated into hormone-based therapies for diabetes. Both classes of molecules also exhibit weight-lowering effects and have been investigated for their anti-obesity potential. In the present review, we explore the mechanisms underlying the physiological and pharmacological actions of GLP-1 and amylin agonism. Despite their similarities (e.g. both molecular classes slow gastric emptying, decrease glucagon and inhibit food intake), there are important distinctions between the central and/or peripheral pathways that mediate their effects on glycaemia and energy balance. We suggest that understanding the similarities and differences between these molecules holds important implications for the development of novel, combination-based therapies, which are increasingly the norm for diabetes/metabolic disease. Finally, the future of GLP-1- and amylin agonist-based therapeutics is discussed.


Asunto(s)
Péptido 1 Similar al Glucagón/agonistas , Polipéptido Amiloide de los Islotes Pancreáticos/agonistas , Receptores de Glucagón/agonistas , Animales , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/fisiopatología , Ingestión de Alimentos/efectos de los fármacos , Vaciamiento Gástrico/efectos de los fármacos , Glucagón/efectos de los fármacos , Glucagón/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/fisiopatología , Obesidad/tratamiento farmacológico , Obesidad/fisiopatología , Receptores de Glucagón/metabolismo
11.
Endocrinology ; 151(12): 5657-68, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20962049

RESUMEN

In rodents, ovariectomy (OVX) elicits weight gain and diminished responsiveness to homeostatic signals. Here we characterized the response of obese OVX rats to peripheral amylin. Rats received sham surgery (SHAM), OVX, or OVX with hormonal replacement (17ß-estradiol, 2 µg per 4 d; OVX+E) and were infused with vehicle or amylin (50 µg/kg · d) for 28 d. Amylin reduced body weight (5.1 ± 1.1%) and food intake (10.9 ± 3.4%) in SHAM rats but was twice as efficacious in OVX rats in reducing weight (11.2 ± 1.9%) and food intake (23.0 ± 2.0%). There were no differences between amylin-treated SHAM and OVX+E rats. OVX decreased metabolic rate (∼24%) and increased respiratory exchange ratio relative to SHAM. Amylin partially normalized metabolic rate (13% increase) in OVX rats and decreased respiratory exchange ratio in OVX and SHAM rats. Regarding central mechanisms, amylin infusion corrected the OVX-induced decrease in hippocampal neurogenesis and increased immobility in the forced swim test. Additionally, amylin increased neurogenesis (∼2-fold) within the area postrema of OVX rats. To assess the contribution of endogenous leptin to amylin-mediated weight loss in OVX rats, amylin was administered to SHAM or OVX Zucker diabetic fatty rats. In SHAM rats, amylin infusion reduced food intake but not body weight, whereas in OVX Zucker diabetic fatty rats, food intake, body weight, and insulin were reduced. Overall, amylin induced greater body weight loss in the absence of estradiol via central and peripheral actions that did not require leptin. These findings support the clinical investigation of amylin in low estradiol (e.g. postmenopausal) states.


Asunto(s)
Dieta , Estradiol/deficiencia , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Obesidad/metabolismo , Pérdida de Peso/efectos de los fármacos , Animales , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Femenino , Leptina/metabolismo , Ovariectomía , Consumo de Oxígeno/efectos de los fármacos , Ratas , Ratas Zucker , Transducción de Señal , Natación
12.
Am J Physiol Regul Integr Comp Physiol ; 299(2): R623-31, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20554938

RESUMEN

Circulating amylin inhibits food intake via activation of the area postrema (AP). The aim of this study was to identify the neurochemical phenotype of the neurons mediating amylin's hypophagic action by immunohistochemical and feeding studies in rats. Expression of c-Fos protein was used as a marker for neuronal activation and dopamine-beta-hydroxylase (DBH), the enzyme-catalyzing noradrenaline synthesis, as a marker for noradrenergic neurons. We found that approximately 50% of amylin-activated AP neurons are noradrenergic. To clarify the functional role of these neurons in amylin's effect on eating, noradrenaline-containing neurons in the AP were lesioned using a saporin conjugated to an antibody against DBH. Amylin (5 or 20 microg/kg s.c.)-induced anorexia was observed in sham-lesioned rats with both amylin doses. Rats with a lesion of > 50% of the noradrenaline neurons were unresponsive to the low dose of amylin (5 microg/kg) and only displayed a reduction in food intake 60 min after injection of the high amylin dose (20 microg/kg). In a terminal experiment, the same rats received amylin (20 microg/kg) or saline. The AP and nucleus of the solitary tract (NTS) were stained for DBH to assess noradrenaline lesion success and for c-Fos expression to evaluate amylin-induced neuronal activation. In contrast to sham-lesioned animals, noradrenaline-lesioned rats did not show a significant increase in amylin-induced c-Fos expression in the AP and NTS. We conclude that the noradrenergic neurons in the AP mediate at least part of amylin's hypophagic effect.


Asunto(s)
Fibras Adrenérgicas/metabolismo , Amiloide/metabolismo , Regulación del Apetito , Área Postrema/metabolismo , Conducta Animal , Ingestión de Alimentos , Norepinefrina/metabolismo , Fibras Adrenérgicas/patología , Amiloide/administración & dosificación , Amiloide/toxicidad , Animales , Anorexia/inducido químicamente , Anorexia/metabolismo , Área Postrema/patología , Dopamina beta-Hidroxilasa/metabolismo , Inmunohistoquímica , Inyecciones Subcutáneas , Polipéptido Amiloide de los Islotes Pancreáticos , Masculino , Fenotipo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
13.
Trends Endocrinol Metab ; 21(8): 473-9, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20413324

RESUMEN

Although the adipokine leptin is regarded as the prototypical long-term signal of energy balance, obese individuals are largely nonresponsive to exogenous leptin administration. Restoration of leptin responsiveness in obesity has been elusive despite a detailed understanding of the molecular mechanisms of leptin signaling. Recent translational research findings point to a potential therapeutic approach that incorporates amylin (a beta-cell hormone) and leptin agonism, with amylin restoring or enhancing leptin sensitivity. Here we hypothesize various physiological, neurobiological and molecular mechanisms that could mediate the interaction of these two neurohormonal signals and discuss several methodological challenges. Understanding how amylin agonism improves leptin function could point to general therapeutic strategies for combating leptin resistance and associated obesity.


Asunto(s)
Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Polipéptido Amiloide de los Islotes Pancreáticos/fisiología , Leptina/farmacología , Leptina/fisiología , Obesidad/tratamiento farmacológico , Animales , Peso Corporal/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Hipotálamo/efectos de los fármacos , Polipéptido Amiloide de los Islotes Pancreáticos/uso terapéutico , Leptina/uso terapéutico , Obesidad/fisiopatología , Rombencéfalo/efectos de los fármacos
14.
Physiol Behav ; 100(2): 187-95, 2010 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-20206194

RESUMEN

Cholecystokinin (CCK) acutely synergizes with amylin to suppress food intake in lean mice. To extend on these findings, the present studies sought to identify neural correlates for the interaction of amylin and CCK, as well as further understand the therapeutic potential of CCK-based combinations in obesity. First, c-Fos activation was assessed in various brain nuclei after a single intraperitoneal injection of amylin (5microg/kg) and/or CCK (5microg/kg). Amylin and CCK additively increased c-Fos within the area postrema (AP), predominantly in noradrenergic (e.g., dopamine-beta-hydroxylase-containing) cells. Next, amylin (100 or 300microg/kg/d) and/or CCK (100 or 300microg/kg/d) were subcutaneously infused for 7days in diet-induced obese (DIO) rats. Amylin treatment of DIO rats for 7days induced significant body weight loss. CCK, while ineffective alone, significantly enhanced body weight loss when co-administered with the higher dose of amylin. Finally, the addition of CCK (300microg/kg/d) to leptin (125microg/kg/d), and to the combination of amylin (50microg/kg/d) and leptin (125microg/kg/d), was also explored in DIO rats via sustained subcutaneous infusion for 14days. Infusion of amylin/leptin/CCK for 14days exerted significantly greater body weight loss, inhibition of food intake, and reduction in adiposity compared to amylin/leptin treatment alone in DIO rats. However, co-infusion of CCK and leptin was an ineffective weight loss regimen in this model. Whereas CCK agonism alone is ineffective at eliciting or maintaining weight loss, it durably augmented the food intake and body weight-lowering effects of amylin and amylin/leptin in a relevant disease model, and when combined with amylin, cooperatively activated neurons within the caudal brainstem.


Asunto(s)
Colecistoquinina/uso terapéutico , Obesidad , Amiloide/farmacología , Amiloide/uso terapéutico , Análisis de Varianza , Animales , Depresores del Apetito/farmacología , Depresores del Apetito/uso terapéutico , Área Postrema/efectos de los fármacos , Área Postrema/metabolismo , Área Postrema/patología , Peso Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Dopamina beta-Hidroxilasa/metabolismo , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada/métodos , Ingestión de Alimentos/efectos de los fármacos , Polipéptido Amiloide de los Islotes Pancreáticos , Leptina/farmacología , Leptina/uso terapéutico , Masculino , Neuronas/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/etiología , Obesidad/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Pérdida de Peso/efectos de los fármacos
15.
Brain Res ; 1350: 86-94, 2010 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-20096672

RESUMEN

Active weight loss and the maintenance of a weight-reduced state elicit potent counter-regulatory responses in multiple neurochemical pathways rendering monotherapy-based anti-obesity agents relatively ineffective. Herein, we highlight potential strategies for overcoming counter-regulatory responses to states of negative energy balance using combinatorial approaches. We discuss methodological and practical considerations for preclinical modeling of additive/synergistic weight loss combinations that have emerged in our translational research program aimed at identifying naturally occurring neurohormonal synergies. As an example of synergy, pharmacological and mechanistic findings with the combined administration of the beta-cell hormone amylin and the adipokine leptin are reviewed. Finally, we briefly discuss what the future landscape of neurohormonal anti-obesity combinations may hold.


Asunto(s)
Polipéptido Amiloide de los Islotes Pancreáticos/uso terapéutico , Leptina/uso terapéutico , Obesidad/terapia , Pérdida de Peso/fisiología , Fármacos Antiobesidad/uso terapéutico , Terapia Combinada , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Leptina/metabolismo , Obesidad/metabolismo
16.
Obesity (Silver Spring) ; 18(1): 21-6, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19543217

RESUMEN

We have previously shown that combined amylin + leptin agonism elicits synergistic weight loss in diet-induced obese (DIO) rats. Here, we assessed the comparative efficacy of amylin, leptin, or amylin + leptin in the maintenance of amylin + leptin-mediated weight loss. DIO rats pretreated with the combination of rat amylin (50 microg/kg/day) and murine leptin (125 microg/kg/day) for 4 weeks were subsequently infused with either vehicle, amylin, leptin, or amylin + leptin for an additional 4 weeks. Food intake, body weight, body composition, plasma parameters, and the expression of key metabolic genes in liver and white adipose tissue (WAT) were assessed. Amylin + leptin treatment (weeks 0-4) reduced body weight to 87.5% of baseline. Rats subsequently maintained on vehicle or leptin regained all weight (to 104.2 and 101.2% of baseline, respectively), those maintained on amylin had partial weight regain (97.0%). By contrast, weight loss was largely maintained with continued amylin + leptin treatment (91.4%), associated with a 10% decrease in adiposity. Cumulative food intake (weeks 5-8) was reduced by amylin and amylin + leptin, but not by leptin alone. Amylin + leptin, but not amylin or leptin alone, reduced plasma triglycerides (by 55%), total cholesterol (by 19%), and insulin (by 57%) compared to vehicle. Amylin + leptin also reduced hepatic stearoyl-CoA desaturase-1 (Scd1) mRNA, and increased WAT mRNA levels of adiponectin, fatty acid synthase (Fasn), and lipoprotein lipase (Lpl). We conclude that, in DIO rats, maintenance of amylin + leptin-mediated weight loss requires continued treatment with both agonists, and is accompanied by sustained improvements in body composition, and indices of lipid metabolism and insulin sensitivity.


Asunto(s)
Amiloide/farmacología , Peso Corporal/efectos de los fármacos , Dieta , Leptina/farmacología , Obesidad/tratamiento farmacológico , Pérdida de Peso/efectos de los fármacos , Adiponectina/sangre , Tejido Adiposo Blanco/metabolismo , Amiloide/sangre , Análisis de Varianza , Animales , Depresores del Apetito/farmacología , Composición Corporal/efectos de los fármacos , Interacciones Farmacológicas , Ingestión de Alimentos/efectos de los fármacos , Perfilación de la Expresión Génica , Insulina/sangre , Polipéptido Amiloide de los Islotes Pancreáticos , Leptina/sangre , Hígado/metabolismo , Masculino , Obesidad/metabolismo , ARN Mensajero/metabolismo , Ratas
17.
Endocrinology ; 151(1): 143-52, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19875640

RESUMEN

The present studies aimed to identify mechanisms contributing to amylin/leptin synergy in reducing body weight and adiposity. We reasoned that if amylin/leptin harnessed complementary neuronal pathways, then in the leptin-sensitive state, amylin should augment leptin signaling/binding and that in the absence of endogenous amylin, leptin signaling should be diminished. Amylin (50 microg/kg, ip) amplified low-dose leptin-stimulated (15 microg/kg, ip) phosphorylated signal transducer and activator of transcription-3 signaling within the arcuate nucleus (ARC) in lean rats. Amylin (50 microg/kg x d) or leptin (125 microg/kg x d) infusion to lean rats decreased 28-d food intake (14 and 10%, respectively), body weight (amylin by 4.3%, leptin by 4.9%), and epididymal fat (amylin by 19%, leptin by 37%). Amylin/leptin co-infusion additively decreased food intake (by 26%) and reduced body weight (by 15%) and epididymal fat (by 78%; all P < 0.05 vs. all groups) in a greater than mathematically additive manner, consistent with synergy. Amylin increased leptin binding within the ventromedial hypothalamus (VMN) by 35% and dorsomedial hypothalamus by 47% (both P < 0.05 vs. vehicle). Amylin/leptin similarly increased leptin binding in the VMN by 40% and ARC by 70% (P < 0.05 vs. vehicle). In amylin-deficient mice, hypothalamic leptin receptor mRNA expression was reduced by 50%, leptin-stimulated phosphorylated signal transducer and activator of transcription-3 within ARC and VMN was reduced by 40%, and responsiveness to leptin's (1 mg/kg x d for 28 d) weight-reducing effects was attenuated (all P < 0.05 vs. wild-type controls). We suggest that amylin/leptin's marked weight- and fat-reducing effects are due to activation of intrinsic synergistic neuronal signaling pathways and further point to the integrated neurohormonal therapeutic potential of amylin/leptin agonism in obesity.


Asunto(s)
Amiloide/fisiología , Sinergismo Farmacológico , Leptina/fisiología , Modelos Animales , Roedores , Amiloide/genética , Amiloide/metabolismo , Amiloide/farmacología , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Área Postrema/efectos de los fármacos , Área Postrema/metabolismo , Femenino , Polipéptido Amiloide de los Islotes Pancreáticos , Leptina/genética , Leptina/metabolismo , Leptina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Roedores/genética , Roedores/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
18.
J Investig Med ; 57(7): 777-83, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20029268

RESUMEN

The discovery of leptin in 1994 was a seminal event in obesity research. It helped to establish that body weight is tightly regulated by a complex neurohormonal feedback system and that obesity should be viewed as a disorder with a strong biological basis rather than simply the result of poor lifestyle choices and lack of willpower.Leptin, secreted from adipocytes, acts as a prototypic long-term (tonic) adiposity signal. Although nonclinical and clinical studies have provided unequivocal evidence that leptin plays a unique, pivotal role in body weight regulation, efforts to develop recombinant leptin (metreleptin) as a monotherapy for obesity have proven unsuccessful. Amylin, secreted from pancreatic beta-cells, fulfills the criteria for a short-term (episodic) satiety signal. The amylin analog pramlintide elicits sustained reductions in food intake and body weight in obese rodents and humans.A translational research program aimed at elucidating the interaction between different islet-, gut-, and adipocyte-derived hormones led to the discovery that combined amylin/leptin agonism induces marked, synergistic, fat-specific weight loss in leptin-resistant diet-induced obese rodents. In obese humans, combination treatment with pramlintide/metreleptin led to an approximately 13% weight loss after 24 weeks, significantly more than after treatment with pramlintide or metreleptin alone.Collectively, these findings suggest that combined amylin/leptin agonism may have therapeutic utility as part of an integrated, neurohormonal approach to obesity pharmacotherapy.


Asunto(s)
Amiloide/agonistas , Fármacos Antiobesidad/uso terapéutico , Leptina/agonistas , Obesidad/tratamiento farmacológico , Amiloide/administración & dosificación , Animales , Fármacos Antiobesidad/administración & dosificación , Diseño de Fármacos , Quimioterapia Combinada , Retroalimentación Fisiológica , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos , Leptina/administración & dosificación , Leptina/análogos & derivados , Neurotransmisores/fisiología , Obesidad/fisiopatología , Respuesta de Saciedad/efectos de los fármacos , Respuesta de Saciedad/fisiología , Transducción de Señal , Pérdida de Peso/efectos de los fármacos , Pérdida de Peso/fisiología
19.
Arch Neurol ; 66(3): 306-10, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19273748

RESUMEN

Amylin receptor agonism is emerging as part of an integrated neurohormonal therapeutic approach for managing diabetes mellitus (DM) and body weight. Pramlintide acetate, an analogue of the pancreatic hormone amylin, has been studied in the United States as an antihyperglycemic agent in patients with type 1 or type 2 DM treated with mealtime insulin(1). Further clinical testing of pramlintide in subjects with obesity demonstrated that pramlintide monotherapy induced significant, sustained, and dose-dependent weight loss(2). Recent clinical observations point to its compatibility as a combination therapy with the hormone leptin, eliciting double-digit weight loss in patients with overweight and obesity(3). Herein, we link amylin activation of central neural circuits to these therapeutic effects, and we speculate on other potential therapeutic applications of amylin receptor agonism.


Asunto(s)
Amiloide/metabolismo , Diabetes Mellitus/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Receptores de Péptidos/agonistas , Amiloide/agonistas , Animales , Peso Corporal/efectos de los fármacos , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos , Receptores de Polipéptido Amiloide de Islotes Pancreáticos , Receptores de Péptidos/fisiología
20.
Expert Opin Biol Ther ; 8(11): 1733-47, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18847308

RESUMEN

BACKGROUND: The integrated central actions of hormones secreted from pancreatic islets, the gut and adipocytes regulate both energy homeostasis and body weight. Dysregulation in these neurohormonal pathways probably contributes to pathogenesis of obesity and type 2 diabetes. OBJECTIVE: To examine hormone-based therapies targeting these interrelated pathways as potential treatments for obesity and diabetes. METHODS: Preclinical and clinical data on therapies based on hormones secreted from the pancreas (glucagon, insulin, amylin and pancreatic polypeptide), gut (glucagon-like peptide-1, glucose dependent insulinotropic polypeptide, cholecystokinin and peptide YY) and adipose tissue (leptin and adiponectin) as potential treatments for diabetes and obesity are reviewed. RESULTS/CONCLUSIONS: In diabetes, hormone-based treatments have translated into new clinical platforms including insulin analogs, the GLP-1-like peptide receptor agonist exenatide and amylinomimetic pramlintide, which due to their complex interplay and the progressive nature of diabetes, can be utilized in different settings. Various peptide hormones and agonists/antagonists are currently under investigation as new approaches to treatment of obesity and diabetes.


Asunto(s)
Peso Corporal , Metabolismo Energético , Hormonas/metabolismo , Adipocitos/metabolismo , Adiponectina/metabolismo , Tejido Adiposo/metabolismo , Amiloide/farmacología , Animales , Diabetes Mellitus Tipo 2/metabolismo , Péptido 1 Similar al Glucagón/química , Homeostasis , Humanos , Mucosa Intestinal/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos , Leptina/metabolismo , Ratones , Obesidad/metabolismo , Páncreas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA