Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Nucl Med ; 63(5): 794-800, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34503957

RESUMEN

The glucagonlike peptide-1 receptor (GLP1R) is a gut hormone receptor, intricately linked to regulation of blood glucose homeostasis via several mechanisms. It is an established and emergent drug target in metabolic disease. The PET radioligand 68Ga-DO3A-VS-exendin4 (68Ga-exendin4) has the potential to enable longitudinal studies of GLP1R in the human pancreas. Methods:68Ga-exendin4 PET/CT examinations were performed on overweight-to-obese individuals with type 2 diabetes (n = 13) as part of a larger target engagement study (NCT03350191). A scanning protocol was developed to optimize reproducibility (target amount of 0.5 MBq/kg [corresponding to peptide amount of <0.2 µg/kg], blood sampling, and tracer stability assessment). The pancreas and abdominal organs were segmented, and binding was correlated with clinical parameters. Results: Uptake of 68Ga-exendin4 in the pancreas, but not in other abdominal tissues, was high but variable between individuals. There was no evidence of self-blocking of GLP1R by the tracer in this protocol, despite the high potency of exendin4. The results showed that a full dynamic scan can be simplified to a short static scan, potentially increasing throughput and reducing patient discomfort. The 68Ga-exendin4 concentration in the pancreas (i.e., GLP1R density) correlated inversely with the age of the individual and tended to correlate positively with body mass index. However, the total GLP1R content in the pancreas did not. Conclusion: In summary, we present an optimized and simplified 68Ga-exendin4 scanning protocol to enable reproducible imaging of GLP1R in the pancreas. 68Ga-exendin4 PET may enable quantification of longitudinal changes in pancreatic GLP1R during the development of type 2 diabetes, as well as target engagement studies of novel glucagonlike peptide-1 agonists.


Asunto(s)
Diabetes Mellitus Tipo 2 , Receptor del Péptido 1 Similar al Glucagón , Diabetes Mellitus Tipo 2/diagnóstico por imagen , Radioisótopos de Galio , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Humanos , Péptidos/química , Tomografía Computarizada por Tomografía de Emisión de Positrones , Tomografía de Emisión de Positrones/métodos , Reproducibilidad de los Resultados
2.
Diabetes ; 70(4): 842-853, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33547046

RESUMEN

Targeting of the glucose-dependent insulinotropic polypeptide receptor (GIPR) is an emerging strategy in antidiabetic drug development. The aim of this study was to develop a positron emission tomography (PET) radioligand for the GIPR to enable the assessment of target distribution and drug target engagement in vivo. The GIPR-selective peptide S02-GIP was radiolabeled with 68Ga. The resulting PET tracer [68Ga]S02-GIP-T4 was evaluated for affinity and specificity to human GIPR (huGIPR). The in vivo GIPR binding of [68Ga]S02-GIP-T4 as well as the occupancy of a drug candidate with GIPR activity were assessed in nonhuman primates (NHPs) by PET. [68Ga]S02-GIP-T4 bound with nanomolar affinity and high selectivity to huGIPR in overexpressing cells. In vivo, pancreatic binding in NHPs could be dose-dependently inhibited by coinjection of unlabeled S02-GIP-T4. Finally, subcutaneous pretreatment with a high dose of a drug candidate with GIPR activity led to a decreased pancreatic binding of [68Ga]S02-GIP-T4, corresponding to a GIPR drug occupancy of almost 90%. [68Ga]S02-GIP-T4 demonstrated a safe dosimetric profile, allowing for repeated studies in humans. In conclusion, [68Ga]S02-GIP-T4 is a novel PET biomarker for safe, noninvasive, and quantitative assessment of GIPR target distribution and drug occupancy.


Asunto(s)
Polipéptido Inhibidor Gástrico/metabolismo , Glucosa/metabolismo , Tomografía de Emisión de Positrones/métodos , Receptores de la Hormona Gastrointestinal/metabolismo , Animales , Femenino , Humanos , Hipoglucemiantes , Masculino , Radioquímica , Ratas , Transducción de Señal/fisiología
3.
J Nucl Med ; 62(6): 833-838, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33097629

RESUMEN

Despite the importance of the glucagon receptor (GCGR) in disease and in pharmaceutical drug development, there is a lack of specific and sensitive biomarkers of its activation in humans. The PET radioligand 68Ga-DO3A-VS-Tuna-2 (68Ga-Tuna-2) was developed to yield a noninvasive imaging marker for GCGR target distribution and drug target engagement in humans. Methods: The biodistribution and dosimetry of 68Ga-Tuna-2 was assessed by PET/CT in 13 individuals with type 2 diabetes as part of a clinical study assessing the occupancy of the dual GCGR/glucagon like peptide-1 receptor agonist SAR425899. Binding of 68Ga-Tuna-2 in liver and reference tissues was evaluated and correlated to biometrics (e.g., weight or body mass index) or other biomarkers (e.g., plasma glucagon levels). Results:68Ga-Tuna-2 binding was seen primarily in the liver, which is in line with the strong expression of GCGR on hepatocytes. The kidneys demonstrated high excretion-related retention, whereas all other tissue demonstrated rapid washout. The SUV55 min (SUV during the last 10-min time frame, 50-60 min after administration) uptake endpoint was sensitive to endogenous levels of glucagon. 68Ga-Tuna-2 exhibited a safe dosimetry profile and no adverse events after intravenous administration. Conclusion:68Ga-Tuna-2 can be used for safe and accurate assessment of the GCGR in human. It may serve as an important tool in understanding the in vivo pharmacology of novel drugs engaging the GCGR.


Asunto(s)
Diabetes Mellitus Tipo 2/diagnóstico por imagen , Diabetes Mellitus Tipo 2/metabolismo , Receptores de Glucagón/metabolismo , Adulto , Peso Corporal , Femenino , Radioisótopos de Galio , Humanos , Riñón/metabolismo , Masculino , Tomografía Computarizada por Tomografía de Emisión de Positrones , Radiometría , Distribución Tisular
4.
J Pharmacol Exp Ther ; 376(2): 190-203, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33203659

RESUMEN

As a gut-restricted, nonabsorbed therapy, polymeric bile acid sequestrants (BAS) play an important role in managing hyperlipidemia and hyperglycemia. Similarly, nonabsorbable sequestrants of dietary phosphate have been used for the management of hyperphosphatemia in end-stage renal disease. To evaluate the potential utility of such polymer sequestrants to treat type 2 diabetes (T2D) and its associated renal and cardiovascular complications, we synthesized a novel polymeric sequestrant, SAR442357, possessing optimized bile acid (BA) and phosphate sequestration characteristics. Long-term treatment of T2D obese cZucker fatty/Spontaneously hypertensive heart failure F1 hybrid (ZSF1) with SAR442357 resulted in enhanced sequestration of BAs and phosphate in the gut, improved glycemic control, lowering of serum cholesterol, and attenuation of diabetic kidney disease (DKD) progression. In comparison, colesevelam, a BAS with poor phosphate binding properties, did not prevent DKD progression, whereas losartan, an angiotensin II receptor blocker that is widely used to treat DKD, showed no effect on hyperglycemia. Analysis of hepatic gene expression levels of the animals treated with SAR442357 revealed upregulation of genes responsible for the biosynthesis of cholesterol and BAs, providing clear evidence of target engagement and mode of action of the new sequestrant. Additional hepatic gene expression pathway changes were indicative of an interruption of the enterohepatic BA cycle. Histopathological analysis of ZSF1 rat kidneys treated with SAR442357 further supported its nephroprotective properties. Collectively, these findings reveal the pharmacological benefit of simultaneous sequestration of BAs and phosphate in treating T2D and its associated comorbidities and cardiovascular complications. SIGNIFICANCE STATEMENT: A new nonabsorbed polymeric sequestrant with optimum phosphate and bile salt sequestration properties was developed as a treatment option for DKD. The new polymeric sequestrant offered combined pharmacological benefits including glucose regulation, lipid lowering, and attenuation of DKD progression in a single therapeutic agent.


Asunto(s)
Antihipertensivos/uso terapéutico , Ácidos y Sales Biliares/metabolismo , Nefropatías Diabéticas/tratamiento farmacológico , Hidrogeles/uso terapéutico , Hipertensión/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Animales , Antihipertensivos/síntesis química , Colesterol/metabolismo , Hidrogeles/síntesis química , Hipoglucemiantes/síntesis química , Hígado/metabolismo , Fosfatos/metabolismo , Poliaminas/química , Ratas , Ratas Zucker
5.
Obesity (Silver Spring) ; 28(11): 2163-2174, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33150746

RESUMEN

OBJECTIVE: Changes in the secretion of gut-derived peptide hormones have been associated with the metabolic benefits of Roux-en-Y gastric bypass (RYGB) surgery. In this study, the effects of RYGB on anthropometrics, postprandial plasma hormone responses, and mRNA expression in small intestinal mucosa biopsy specimens before and after RYGB were evaluated. METHODS: In a cross-sectional study, 20 individuals with obesity undergoing RYGB underwent mixed meal tests and upper enteroscopy with retrieval of small intestinal mucosa biopsy specimens 3 months before and after surgery. Concentrations of circulating gut and pancreatic hormones during mixed meal tests as well as full mRNA sequencing of biopsy specimens were evaluated. RESULTS: RYGB-induced improvements of body weight and composition, insulin resistance, and circulating cholesterols were accompanied by significant changes in postprandial plasma responses of pancreatic and gut hormones. Global gene expression analysis of biopsy specimens identified 2,437 differentially expressed genes after RYGB, including changes in genes that encode prohormones and G protein-coupled receptors. CONCLUSIONS: RYGB affects the transcription of a wide range of genes, indicating that the observed beneficial metabolic effects of RYGB may rely on a changed expression of several genes in the gut. RYGB-induced changes in the expression of genes encoding signaling peptides and G protein-coupled receptors may disclose new gut-derived treatment targets against obesity and diabetes.


Asunto(s)
Derivación Gástrica/métodos , Microbioma Gastrointestinal/genética , Expresión Génica/genética , Adulto , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad
6.
Sci Rep ; 9(1): 14960, 2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31628379

RESUMEN

The glucagon receptor (GCGR) is an emerging target in anti-diabetic therapy. Reliable biomarkers for in vivo activity on the GCGR, in the setting of dual glucagon-like peptide 1/glucagon (GLP-1/GCG) receptor agonism, are currently unavailable. Here, we investigated [68Ga]Ga-DO3A-S01-GCG as a biomarker for GCGR occupancy in liver, the tissue with highest GCGR expression, in non-human primates (NHP) by PET. [68Ga]Ga-DO3A-S01-GCG was evaluated by dynamic PET in NHPs by a dose escalation study design, where up to 67 µg/kg DO3A-S01-GCG peptide mass was co-injected. The test-retest reproducibility of [68Ga]Ga-DO3A-S01-GCG binding in liver was evaluated. Furthermore, we investigated the effect of pre-treatment with acylated glucagon agonist 1-GCG on [68Ga]Ga-DO3A-S01-GCG binding in liver. [68Ga]Ga-DO3A-S01-GCG bound to liver in vivo in a dose-dependent manner. Negligible peptide mass effect was observed for DO3A-S01-GCG doses <0.2 µg/kg. In vivo Kd for [68Ga]Ga-DO3A-S01-GCG corresponded to 0.7 µg/kg, which indicates high potency. The test-retest reproducibility for [68Ga]Ga-DO3A-S01-GCG binding in liver was 5.7 ± 7.9%. Pre-treatment with 1-GCG, an acylated glucagon agonist, resulted in a GCGR occupancy of 61.5 ± 9.1% in liver. Predicted human radiation dosimetry would allow for repeated annual [68Ga]Ga-DO3A-S01-GCG PET examinations. In summary, PET radioligand [68Ga]Ga-DO3A-S01-GCG is a quantitative biomarker of in vivo GCGR occupancy.


Asunto(s)
Biomarcadores/metabolismo , Receptores de Glucagón/metabolismo , Animales , Femenino , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Humanos , Ligandos , Hígado/diagnóstico por imagen , Hígado/metabolismo , Macaca fascicularis , Masculino , Péptidos/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones , Unión Proteica , Radiometría , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Bazo/diagnóstico por imagen
7.
J Clin Endocrinol Metab ; 104(12): 6403-6416, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31276156

RESUMEN

CONTEXT: After Roux-en-Y gastric bypass (RYGB) surgery, postprandial plasma glucagon concentrations have been reported to increase. This occurs despite concomitant improved glucose tolerance and increased circulating plasma concentrations of insulin and the glucagon-inhibiting hormone glucagon-like peptide 1 (GLP-1). OBJECTIVE: To investigate whether RYGB-induced hyperglucagonemia may be derived from the gut. DESIGN AND SETTING: Substudy of a prospective cross-sectional study at a university hospital in Copenhagen, Denmark. PARTICIPANTS: Morbidly obese individuals undergoing RYGB (n = 8) with or without type 2 diabetes. INTERVENTIONS: Three months before and after RYGB, participants underwent upper enteroscopy with retrieval of gastrointestinal mucosal biopsy specimens. Mixed-meal tests were performed 1 week and 3 months before and after RYGB. MAIN OUTCOME MEASURES: The 29-amino acid glucagon concentrations in plasma and in mucosal gastrointestinal biopsy specimens were assessed using mass spectrometry-validated immunoassays, and a new monoclonal antibody reacting with immunoreactive glucagon was used for immunohistochemistry. RESULTS: Postprandial plasma concentrations of glucagon after RYGB were increased. Expression of the glucagon gene in the small intestine increased after surgery. Glucagon was identified in the small-intestine biopsy specimens obtained after, but not before, RYGB. Immunohistochemically, mucosal biopsy specimens from the small intestine harbored cells costained for GLP-1 and immunoreactive glucagon. CONCLUSION: Increased concentrations of glucagon were observed in small-intestine biopsy specimens and postprandially in plasma after RYGB. The small intestine harbored cells immunohistochemically costaining for GLP-1 and glucagon-like immunoreactivity after RYGB. Glucagon derived from small-intestine enteroendocrine l cells may contribute to postprandial plasma concentrations of glucagon after RYGB.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Derivación Gástrica/métodos , Péptido 1 Similar al Glucagón/sangre , Glucagón/sangre , Insulina/sangre , Intestinos/fisiología , Obesidad Mórbida/sangre , Adolescente , Adulto , Enteroscopia de Balón , Biomarcadores/sangre , Glucemia/análisis , Estudios de Casos y Controles , Estudios Transversales , Femenino , Estudios de Seguimiento , Polipéptido Inhibidor Gástrico/sangre , Hemoglobina Glucada/análisis , Humanos , Masculino , Comidas , Persona de Mediana Edad , Obesidad Mórbida/complicaciones , Obesidad Mórbida/cirugía , Periodo Posprandial , Pronóstico , Estudios Prospectivos , Adulto Joven
8.
Diabetes Obes Metab ; 21(1): 120-128, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30091218

RESUMEN

AIMS: To evaluate the safety, pharmacokinetics and pharmacodynamics of SAR425899, a novel polypeptide, active as an agonist at both the glucagon-like peptide-1 receptor (GLP-1R) and the glucagon receptor (GCR), in healthy volunteers and in overweight/obese patients with type 2 diabetes (T2D). METHODS: Subcutaneous administrations of SAR425899 were tested in two randomized, placebo-controlled, double-blind clinical trials. In the first trial, healthy overweight volunteers (body mass index [BMI] 25-30 kg/m2 ; n = 32) received single-ascending doses (0.01-0.1 mg) of SAR425899 or placebo. In the second, a multiple-ascending-dose trial (NCT02411825), healthy normal- to overweight volunteers (BMI 20-30 kg/m2 ; n = 40) and overweight/obese patients with T2D (BMI 28-42 kg/m2 ; n = 36) received daily doses of SAR425899 or placebo over 21 or 28 days, respectively. RESULTS: The most frequently reported adverse events were gastrointestinal; gastrointestinal side effects were less pronounced in patients with T2D compared with healthy volunteers. SAR425899 significantly reduced levels of fasting plasma glucose (P < 0.05 vs. placebo) and glycated haemoglobin (P < 0.001 versus placebo) in patients with T2D. Additionally, SAR425899 led to reductions in body weight, with a maximal reduction of 5.32 kg in healthy volunteers and 5.46 kg in patients with T2D (P < 0.001 vs. placebo) at end of treatment. CONCLUSIONS: SAR425899 was well tolerated and led to favourable glycaemic effects in patients with T2D and weight reduction in both healthy volunteers and patients. Whether dual GLP-1R/GCR agonism represents a treatment method that is superior to pure GLP-1R agonists for obesity and diabetes treatment remains to be confirmed.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Receptor del Péptido 1 Similar al Glucagón/agonistas , Hipoglucemiantes , Receptores de Glucagón/agonistas , Adolescente , Adulto , Glucemia/análisis , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Diabetes Mellitus Tipo 2/complicaciones , Femenino , Humanos , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Lípidos/sangre , Masculino , Persona de Mediana Edad , Obesidad/complicaciones , Placebos , Adulto Joven
9.
Endocrinology ; 159(8): 3105-3119, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29992313

RESUMEN

We assessed the therapeutic contribution of the individual components of glucagon-like peptide-1 receptor (GLP-1R) and glucagon receptor (GCGR) agonists alone and in combination upon energy homeostasis and glycemic control in diet-induced obese, diabetic nonhuman primates. The pharmacological active dose ranges of selective agonists were established through a dose-finding study, followed by a 6-week chronic study. Repeated subcutaneous administration of a selective GCGR agonist (30 µg/kg once daily) did not affect food intake or body weight, whereas the selective GLP-1R agonist (3 µg/kg once daily) alone decreased energy intake by 18% and body weight by 3.8% ± 0.9%. Combination of both agonists reduced significantly cumulative food intake by 27% and body weight by 6.6% ± 0.9%. Fasting plasma glucose (FPG) was improved by GLP-1R agonist (baseline vs end of study, 176.7 ± 34.0 vs 115.9 ± 16.1 mg/dL). In contrast, groups exposed to GCGR agonist experienced nonsignificant elevations of FPG. More accurate assessment of therapeutic interventions on glucose homeostasis was tested by an IV glucose tolerance test. Glucose excursion was significantly elevated by chronic GCGR agonist administration, whereas it was significantly decreased in GLP-1R agonist-treated monkeys. In the combination group, a nonsignificant increase of glucose excursion was seen, concomitantly with significantly increased insulin secretion. We conclude that chronic glucagon agonism does not affect energy homeostasis in nonhuman primates. In combination with GLP-1R agonism, glucagon agonism synergistically enhances negative energy balance with resulting larger body weight loss. However, adding GCGR to GLP-1R agonism diminishes glycemic control in diabetic monkeys. Therefore, long-term therapeutic implications of using GLP-1R/GCGR coagonists for weight management in diabetes warrants further scrutiny.


Asunto(s)
Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/agonistas , Obesidad/metabolismo , Receptores de Glucagón/agonistas , Animales , Cirugía Bariátrica , Glucemia/metabolismo , Diabetes Mellitus/metabolismo , Diabetes Mellitus Tipo 2/cirugía , Quimioterapia Combinada , Metabolismo Energético/efectos de los fármacos , Macaca fascicularis , Ratones , Obesidad/cirugía
10.
Diabetes Obes Metab ; 20(8): 1836-1851, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29938884

RESUMEN

AIM: We performed acute and chronic studies in healthy and diet-induced obese animals using mouse-specific or monkey-specific dual GLP-1R/GCGR agonists to investigate their effects on food intake, body weight, blood glucose control and insulin secretion. The selective GLP-1R agonist liraglutide was used as comparator. METHODS: The mouse-specific dual agonist and liraglutide were tested in lean wild type, GLP-1R knockout and diet-induced obese mice at different doses. A chronic study was performed in DIO mice to investigate the effect on body weight, food consumption and total energy expenditure (TEE) in obese and diabetic monkeys with a focus on body weight and energy intake. RESULTS: The mouse-specific dual agonist and liraglutide similarly affected glycaemic control. A higher loss in body weight was measured in dual agonist-treated obese mice. The dual agonist significantly enhanced plasma glucose excursion in overnight fed GLP-1R-/- mice, probably reflecting a potent GCGR agonist activity. It increased TEE and enhanced fat and carbohydrate oxidation, while liraglutide produced no effect on TEE. In obese and diabetic monkeys, treatment with the monkey-specific dual agonist reduced total energy intake to 60%-70% of baseline TEI during chronic treatment. A decrease in body weight and significant improvement in glucose tolerance was observed. CONCLUSIONS: In DIO mice and non-human primates, dual agonists elicited robust glycaemic control, similar to the marketed GLP-1R agonist, while eliciting greater effects on body weight. Results from DIO mice suggest that the increase in TEE is caused not only by increased fat oxidation but also by an increase in carbohydrate oxidation.


Asunto(s)
Depresores del Apetito/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Receptor del Péptido 1 Similar al Glucagón/agonistas , Hiperglucemia/prevención & control , Hipoglucemiantes/uso terapéutico , Obesidad/tratamiento farmacológico , Receptores de Glucagón/agonistas , Animales , Animales no Consanguíneos , Depresores del Apetito/administración & dosificación , Depresores del Apetito/efectos adversos , Peso Corporal/efectos de los fármacos , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada/efectos adversos , Ingestión de Energía/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Femenino , Receptor del Péptido 1 Similar al Glucagón/genética , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/efectos adversos , Secreción de Insulina/efectos de los fármacos , Macaca fascicularis , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/sangre , Obesidad/etiología , Obesidad/metabolismo , Distribución Aleatoria , Receptores de Glucagón/metabolismo
11.
Mol Metab ; 3(3): 252-60, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24749054

RESUMEN

In most people with type 2 diabetes, progression from obesity to diabetes is accompanied by elevated tissue exposures to a variety of lipids. Among these lipid species, ceramides and more complex sphingolipids have gained recent attention as being pathophysiologically relevant for the development of insulin resistance and impaired glycemic control. Upon excess intake of saturated fat, ceramides accumulate in insulin sensitive tissues either as a consequence of de novo synthesis or through mobilization from complex sphingolipids. Clinical studies have confirmed positive correlation between plasma and tissue levels of several ceramide species and insulin resistance. At the cellular level, it has been demonstrated that ceramides impair insulin signaling and intracellular handling of glucose and lipids with resulting deleterious effects on cellular metabolism. Hence, we are reviewing whether therapeutic interventions aiming at reducing tissue exposure to ceramides or other sphingolipids represent viable therapeutic approaches to improve glucose metabolism in people with diabetes.

12.
Eur J Pharmacol ; 636(1-3): 88-95, 2010 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-20385125

RESUMEN

Tesofensine, a novel triple monoamine reuptake inhibitor, produces a significant weight loss in humans. The present study aimed at characterizing the weight-reducing effects of tesofensine in a rat model of diet-induced obesity. Sibutramine and rimonabant were used as reference comparators. Compared to baseline, long-term treatment with tesofensine (28 days, 1.0 or 2.5mg/kg, p.o.) resulted in a significant, dose-dependent and sustained weight loss of 5.7 and 9.9%, respectively. Sibutramine (7.5mg/kg, p.o.) treatment caused a sustained weight loss of 7.6%, whereas the employed dose of rimonabant (10mg/kg, p.o.) only produced a transient weight reduction. While all compounds exhibited a significant inhibitory effect on food intake which gradually wore off, the hypophagic effect of tesofensine was longer lasting than sibutramine and rimonabant. In contrast to tesofensine, the body weight of pair-fed rats returned to baseline at the end of the study, which may indicate that tesofensine stimulated energy expenditure. The differential efficacy on weight reduction was also reflected in lowered body fat depots, as tesofensine and sibutramine most efficiently reduced abdominal and subcutaneous fat mass which was paralleled by reduced plasma lipid levels. In an oral glucose tolerance test, only tesofensine significantly suppressed the plasma insulin response below the level that could be obtained by paired feeding, indicating that tesofensine further improved glycemic control. In conclusion, the robust weight loss with long-term tesofensine treatment is likely due to a combined synergistic effect of appetite suppression and increased energy expenditure.


Asunto(s)
Monoaminas Biogénicas/metabolismo , Glucemia/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Dieta/efectos adversos , Obesidad/metabolismo , Obesidad/fisiopatología , Pérdida de Peso/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Apetito/efectos de los fármacos , Ciclobutanos/farmacología , Ingestión de Alimentos/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Lípidos/sangre , Masculino , Inhibidores de la Captación de Neurotransmisores/farmacología , Obesidad/sangre , Obesidad/etiología , Piperidinas/farmacología , Pirazoles/farmacología , Ratas , Rimonabant , Factores de Tiempo
13.
Obesity (Silver Spring) ; 18(7): 1289-96, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19851307

RESUMEN

Using restriction fragment differential display (RFDD) technology, we have identified the imprinted gene neuronatin (Nnat) as a hypothalamic target under the influence of leptin. Nnat mRNA expression is decreased in several key appetite regulatory hypothalamic nuclei in rodents with impaired leptin signaling and during fasting conditions. Furthermore, peripheral administration of leptin to ob/ob mice normalizes hypothalamic Nnat expression. Comparative immunohistochemical analysis of human and rat hypothalami demonstrates that NNAT protein is present in anatomically equivalent nuclei, suggesting human physiological relevance of the gene product(s). A putative role of Nnat in human energy homeostasis is further emphasized by a consistent association between single nucleotide polymorphisms (SNPs) in the human Nnat gene and severe childhood and adult obesity.


Asunto(s)
Impresión Genómica/fisiología , Leptina/metabolismo , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Obesidad/genética , Obesidad/fisiopatología , Tejido Adiposo/fisiología , Animales , Metabolismo Energético/genética , Perfilación de la Expresión Génica , Genotipo , Homeostasis/genética , Hipotálamo/fisiología , Leptina/genética , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Proteínas del Tejido Nervioso/metabolismo , Células PC12 , Páncreas/fisiología , Hipófisis/fisiología , Polimorfismo de Nucleótido Simple , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/fisiología
14.
Obesity (Silver Spring) ; 18(2): 266-73, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19661956

RESUMEN

The selectively bred diet-induced obese (DIO) and diet-resistant (DR) rats represent a polygenetic animal model mimicking most clinical variables characterizing the human metabolic syndrome. When fed a high-energy (HE) diet DIO rats develop visceral obesity, dyslipidemia, hyperinsulinemia, and insulin resistance but never frank diabetes. To improve our understanding of the underlying cause for the deteriorating glucose and insulin parameters, we have investigated possible adaptive responses in DIO and DR rats at the level of the insulin-producing beta-cells. At the time of weaning, DR rats were found to have a higher body weight and beta-cell mass compared to DIO rats, and elevated insulin and glucose responses to an oral glucose load. However, at 2.5 months of age, and for the remaining study period, the effect of genotype became evident: the chow-fed DIO rats steadily increased their body weight and beta-cell mass, as well as insulin and glucose levels compared to the DR rats. HE feeding affected both DIO and DR rats leading to an increased body weight and an increased beta-cell mass. Interestingly, although the beta-cell mass in DR rats and chow-fed DIO rats appeared to constantly increase with age, the beta-cell mass in the HE-fed DIO rats did not continue to do so. This might constitute part of an explanation for their reduced glucose tolerance. Collectively, the data support the use of HE-fed DIO rats as a model of human obesity and insulin resistance, and accentuate its relevance for studies examining the benefit of pharmaceutical compounds targeting this disease complex.


Asunto(s)
Diabetes Mellitus/patología , Diabetes Mellitus/fisiopatología , Resistencia a la Insulina , Células Secretoras de Insulina/patología , Obesidad/patología , Obesidad/fisiopatología , Estado Prediabético/patología , Estado Prediabético/fisiopatología , Factores de Edad , Envejecimiento , Animales , Glucemia/metabolismo , Peso Corporal , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Dieta , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Prueba de Tolerancia a la Glucosa , Homeostasis , Insulina/sangre , Resistencia a la Insulina/genética , Células Secretoras de Insulina/metabolismo , Masculino , Obesidad/genética , Obesidad/metabolismo , Fenotipo , Estado Prediabético/genética , Estado Prediabético/metabolismo , Ratas
15.
Biochim Biophys Acta ; 1801(4): 421-9, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19962449

RESUMEN

Cold adaptation elicits a paradoxical simultaneous induction of fatty acid synthesis and beta-oxidation in brown adipose tissue. We show here that cold exposure coordinately induced liver X receptor alpha (LXRalpha), adipocyte determination and differentiation-dependent factor 1 (ADD1)/sterol regulatory element-binding protein-1c (SREBP1c) and peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC1alpha) in brown and inguinal white adipose tissues, but not in epididymal white adipose tissue. Using in vitro models of white and brown adipocytes we demonstrate that beta-adrenergic stimulation induced expression of LXRalpha, ADD1/SREBP1c and PGC1alpha in cells with a brown-like adipose phenotype. We demonstrate that ADD1/SREBP1c is a powerful inducer of PGC1alpha expression via a conserved E box in the proximal promoter and that beta-adrenergic stimulation led to recruitment of ADD1/SREBP1c to this E box. The ability of ADD1/SREBP1c to activate the PGC1alpha promoter exhibited a striking cell type dependency, suggesting that additional cell type-restricted factors contribute to ADD1/SREBP1c-mediated activation. In conclusion, our data demonstrate a novel role of ADD1/SREBP1c as a regulator of PGC1alpha expression in brown adipose tissue.


Asunto(s)
Adipocitos Marrones/metabolismo , Regiones Promotoras Genéticas/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Transactivadores/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Electroporación , Receptores X del Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Nucleares Huérfanos/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , ARN Mensajero/genética , ARN Mensajero/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Transactivadores/genética , Factores de Transcripción
16.
Neurosci Lett ; 449(1): 20-3, 2009 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-18926875

RESUMEN

The selective CB1 receptor antagonist rimonabant is a novel weight control agent. Although CB1 receptors and binding sites are present in both the rodent central and peripheral nervous systems, including the afferent vagus nerve, the role of gut afferents in mediating anorexia following CB1R blockade is still debated. In the present study we examined rimonabant-induced anorexia in male C57BL/6J mice with subdiaphragmatic vagotomy (VGX) as well as in male Sprague-Dawley rats subjected to either subdiaphragmatic vagal deafferentation (SDA) alone or in combination with a complete celiac-superior mesenteric ganglionectomy (CGX). Irrespective of the operational procedure, rimonabant (10mg/kg) effectively reduced standard chow as well as palatable diet (ensure) intake. In conclusion, the data clearly demonstrate that neither vagal gut afferents, nor gut afferents traveling via the sympathetic nervous system, are required for rimonabant to inhibit food intake leading to the hypothesis that centrally located CB1 receptors are the prime mediators of rimonabant-induced anorexia.


Asunto(s)
Anorexia/inducido químicamente , Anorexia/fisiopatología , Ganglios Simpáticos/fisiología , Piperidinas , Pirazoles , Nervio Vago/fisiología , Análisis de Varianza , Animales , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Ganglionectomía/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Rimonabant , Estilbamidinas/metabolismo , Vagotomía/métodos , Nervio Vago/cirugía
17.
Eur J Pharmacol ; 596(1-3): 173-9, 2008 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-18761337

RESUMEN

Balaglitazone is a novel thiazolidinedione in clinical development for the treatment of type 2 diabetes. Common side effects associated with PPARgamma receptor agonists are weight gain, oedema and adipogenesis. Balaglitazone is a selective partial PPARgamma agonist and it has been speculated that such compounds have a more favourable safety margin than full agonists. We have compared impact of equi-efficacious antihyperglycaemic doses of balaglitazone with full PPARgamma agonist rosiglitazone on body fluid accumulation, cardiac enlargement, and adipogenesis. Equi-efficacious antihyperglycaemic doses (ED(90)) of balaglitazone (3 mg/kg/day) and rosiglitazone (6 mg/kg/day) were determined in male diabetic db/db mice. In adult male rats treated for up to 42 days, feeding, drinking, anthropometry, and plasma volumes were measured. Total plasma volume was measured with dye dilution technique. Compared to vehicle, rosiglitazone consistently increased food intake throughout the 42 day treatment period. In contrast, balaglitazone increased food intake in the last week of the experiment. However, both rosiglitazone and balaglitazone increased water intake. After 42 days, rosiglitazone treated rats displayed significantly elevated adiposity. Rosiglitazone increased total blood and plasma volumes throughout the treatment. Twenty-one days of balaglitazone treatment had no significant impact on blood or plasma volumes, whilst 42 days of balaglitazone increased plasma volume but to a significantly lesser extent than seen for rosiglitazone (vehicle: 46.1+/-1.5; balaglitazone: 50.8+/-1.21; rosiglitazone: 54.6+/-1.6 ml/kg). Heart weight was significantly elevated only in rosiglitazone treated animals. At doses inducing comparable antihyperglycaemic control, the full PPARgamma agonist, rosiglitazone, induces more pronounced body fluid retention and heart enlargement than seen for the partial PPARgamma agonist, balaglitazone. Thus, partial agonists may pose safer alternative to current anti-diabetic therapy with full PPARgamma agonist.


Asunto(s)
Hipoglucemiantes/efectos adversos , Hipoglucemiantes/farmacología , PPAR gamma/agonistas , Quinazolinas/efectos adversos , Quinazolinas/farmacología , Tiazolidinedionas/efectos adversos , Tiazolidinedionas/farmacología , Adipogénesis/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Animales , Volumen Sanguíneo/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Corazón/anatomía & histología , Corazón/efectos de los fármacos , Humanos , Masculino , Ratones , Tamaño de los Órganos/efectos de los fármacos , PPAR gamma/genética , Ratas , Rosiglitazona
18.
Schizophr Res ; 103(1-3): 94-103, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18579346

RESUMEN

Therapeutic use of atypical antipsychotic agents is often associated with weight gain and impaired glucose tolerance. The once-daily human GLP-1 analog liraglutide improves glycemic control and reduces body weight. We have investigated the ability of liraglutide to improve olanzapine-induced metabolic effects in female rats. Female Sprague-Dawley rats were implanted with subcutaneous osmotic mini pumps for delivery of olanzapine (1.75 mg/24 h) or vehicle for 28 days (n=20). After 14 days, ten animals from each group were given liraglutide (0.2 mg/kg) or vehicle twice daily for the remainder of the study. Compared to vehicle treated animals, olanzapine infusion for 4 weeks significantly increased end point cumulated food intake (667.3+/-7.0 versus 593.2+/-13.2g, p<0.01), body weight (306.6+/-4.2 versus 276.4+/-3.6 g, p<0.001), subcutaneous inguinal fat (3.4+/-0.3 versus 1.9+/-0.1 g, p<0.001), mesenteric fat (3.1+/-0.2 versus 1.7+/-0.2g, p<0.001), retroperitoneal fat (6.2+/-0.6 versus 2.8+/-0.3 g, p<0.001), and impaired glucose tolerance, measured as total area under the glucose curve during an oral glucose tolerance test (1906+/-66 versus 1770+/-28 mMxmin, p<0.05). These olanzapine-induced elevations were significantly reduced by liraglutide (cumulated food intake: 601.8+/-20.4 g, p<0.01; body weight: 280.2+/-5.6 g, p<0.001; subcutaneous inguinal fat: 2.4+/-0.2 g, p<0.001; mesenteric fat: 1.8+/-0.1 g, p<0.001; retroperitoneal fat: 3.5+/-0.4 g, p<0.001; AUC: 1764+/-32 mMxmin, p<0.05). In conclusion, subcutaneous olanzapine infusion in female rats leads to weight gain and metabolic changes of which several are reversed following liraglutide treatment. It may therefore be relevant to study these effects of liraglutide in patients treated with atypically antipsychotics.


Asunto(s)
Antipsicóticos/efectos adversos , Benzodiazepinas/efectos adversos , Péptido 1 Similar al Glucagón/análogos & derivados , Intolerancia a la Glucosa/inducido químicamente , Aumento de Peso/efectos de los fármacos , Animales , Antipsicóticos/uso terapéutico , Benzodiazepinas/uso terapéutico , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Esquema de Medicación , Ingestión de Alimentos/efectos de los fármacos , Femenino , Péptido 1 Similar al Glucagón/efectos adversos , Péptido 1 Similar al Glucagón/uso terapéutico , Intolerancia a la Glucosa/prevención & control , Prueba de Tolerancia a la Glucosa , Grasa Intraabdominal/efectos de los fármacos , Liraglutida , Olanzapina , Ratas , Ratas Sprague-Dawley , Grasa Subcutánea Abdominal/efectos de los fármacos
19.
Peptides ; 27(8): 1981-6, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16762453

RESUMEN

Cocaine-amphetamine regulated transcript (CART) was first identified as a complete mRNA transcript 10 years ago. Since then it has been demonstrated that CART is a peptide neurotransmitter which has a role in a number of physiological processes, including body weight homeostasis and energy balance, in a number of vertebrate species. Research to date has demonstrated a role for CART in the control of food intake at a number of levels within the brain of both animals and man.


Asunto(s)
Peso Corporal/efectos de los fármacos , Homeostasis/efectos de los fármacos , Proteínas del Tejido Nervioso/farmacología , Animales , Peso Corporal/fisiología , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ingestión de Energía/efectos de los fármacos , Ingestión de Energía/fisiología , Metabolismo Energético/efectos de los fármacos , Humanos , Modelos Biológicos , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/fisiología , Fragmentos de Péptidos/farmacología
20.
Ugeskr Laeger ; 168(2): 152-5, 2006 Jan 09.
Artículo en Danés | MEDLINE | ID: mdl-16403339

RESUMEN

During the last few years, studies of the molecular pathogenesis of obesity both in mouse models and in the rare cases of monogenic obesity in humans have added significantly to our understanding of the key role of the hypothalamus in mediating hunger and satiety. These insights have brought us closer to the development of rational therapies of obesity, the epidemic of which is continuing in the post-industrial society, which is characterised by sedentary behaviour patterns.


Asunto(s)
Obesidad/etiología , Animales , Apetito/fisiología , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiología , Ratones , Obesidad/metabolismo , Obesidad/fisiopatología , Receptores de Superficie Celular/metabolismo , Receptores de Leptina , Saciedad/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA