Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Am J Physiol Regul Integr Comp Physiol ; 321(2): R250-R259, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34259025

RESUMEN

The peptide hormone amylin reduces food intake and body weight and is an attractive candidate target for novel pharmacotherapies to treat obesity. However, the short half-life of native amylin and amylin analogs like pramlintide limits these compounds' potential utility in promoting sustained negative energy balance. Here, we evaluate the ability of the novel long-acting amylin/calcitonin receptor agonist ZP5461 to reduce feeding and body weight in rats, and also test the role of calcitonin receptors (CTRs) in the dorsal vagal complex (DVC) of the hindbrain in the energy balance effects of chronic ZP5461 administration. Acute dose-response studies indicate that systemic ZP5461 (0.5-3 nmol/kg) robustly suppresses energy intake and body weight gain in chow- and high-fat diet (HFD)-fed rats. When HFD-fed rats received chronic systemic administration of ZP5461 (1-2 nmol/kg), the compound initially produced reductions in energy intake and weight gain but failed to produce sustained suppression of intake and body weight. Using virally mediated knockdown of DVC CTRs, the ability of chronic systemic ZP5461 to promote early reductions in intake and body weight gain was determined to be mediated in part by activation of DVC CTRs, implicating the DVC as a central site of action for ZP5461. Future studies should address other dosing regimens of ZP5461 to determine whether an alternative dose/frequency of administration would produce more sustained body weight suppression.


Asunto(s)
Agonistas de los Receptores de Amilina/farmacología , Depresores del Apetito/farmacología , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Receptores de Calcitonina/agonistas , Receptores de Polipéptido Amiloide de Islotes Pancreáticos/efectos de los fármacos , Rombencéfalo/efectos de los fármacos , Nervio Vago/efectos de los fármacos , Aumento de Peso/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Ingestión de Energía/efectos de los fármacos , Masculino , Ratas Sprague-Dawley , Receptores de Calcitonina/genética , Receptores de Calcitonina/metabolismo , Receptores de Polipéptido Amiloide de Islotes Pancreáticos/genética , Receptores de Polipéptido Amiloide de Islotes Pancreáticos/metabolismo , Rombencéfalo/metabolismo , Transducción de Señal , Factores de Tiempo , Nervio Vago/metabolismo
2.
Diabetes Obes Metab ; 20(5): 1223-1234, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29327400

RESUMEN

AIMS: While pharmacological glucagon-like peptide-1 receptor (GLP-1R) agonists are FDA-approved for treating type 2 diabetes mellitus (T2DM) and obesity, a major side effect is nausea/malaise. We recently developed a conjugate of vitamin B12 (B12) bound to the GLP-1R agonist exendin-4 (Ex4), which displays enhanced proteolytic stability and retention of GLP-1R agonism. Here, we evaluate whether the conjugate (B12-Ex4) can improve glucose tolerance without producing anorexia and malaise. MATERIALS AND METHODS: We evaluated the effects of systemic B12-Ex4 and unconjugated Ex4 on food intake and body weight change, oral glucose tolerance and nausea/malaise in male rats, and on intraperitoneal glucose tolerance in mice. To evaluate whether differences in the profile of effects of B12-Ex4 vs unconjugated Ex4 are the result of altered CNS penetrance, rats received systemic injections of fluorescein-Ex4 (Flex), Cy5-B12 or Cy5-B12-Ex4 and brain penetrance was evaluated using confocal microscopy. Uptake of systemically administered Cy5-B12-Ex4 in insulin-containing pancreatic beta cells was also examined. RESULTS: B12-Ex4 conjugate improves glucose tolerance, but does not elicit the malaise and anorexia produced by unconjugated Ex4. While Flex robustly penetrates into the brain (dorsal vagal complex, paraventricular hypothalamus), Cy5-B12 and Cy5-B12-Ex4 fluorescence were not observed centrally, supporting an absence of CNS penetrance, in line with observed reduction in CNS-associated Ex4 side effects. Cy5-B12-Ex4 colocalizes with insulin in the pancreas, suggesting direct pancreatic action as a potential mechanism underlying the hypoglycaemic effects of B12-Ex4. CONCLUSION: These novel findings highlight the potential clinical utility of B12-Ex4 conjugates as possible future T2DM therapeutics with reduced incidence of adverse effects.


Asunto(s)
Regulación del Apetito/efectos de los fármacos , Exenatida/análogos & derivados , Receptor del Péptido 1 Similar al Glucagón/agonistas , Intolerancia a la Glucosa/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Vitamina B 12/análogos & derivados , Animales , Conducta Animal/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Estabilidad de Medicamentos , Ingestión de Energía/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Exenatida/efectos adversos , Exenatida/farmacocinética , Exenatida/uso terapéutico , Femenino , Receptor del Péptido 1 Similar al Glucagón/genética , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Células HEK293 , Humanos , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/química , Hipoglucemiantes/farmacocinética , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Ratones Endogámicos C57BL , Náusea/inducido químicamente , Náusea/prevención & control , Ratas Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Distribución Tisular , Vitamina B 12/efectos adversos , Vitamina B 12/farmacocinética , Vitamina B 12/uso terapéutico
3.
J Neurosci ; 36(12): 3531-40, 2016 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-27013681

RESUMEN

Astrocytes are well established modulators of extracellular glutamate, but their direct influence on energy balance-relevant behaviors is largely understudied. As the anorectic effects of glucagon-like peptide-1 receptor (GLP-1R) agonists are partly mediated by central modulation of glutamatergic signaling, we tested the hypothesis that astrocytic GLP-1R signaling regulates energy balance in rats. Central or peripheral administration of a fluorophore-labeled GLP-1R agonist, exendin-4, localizes within astrocytes and neurons in the nucleus tractus solitarius (NTS), a hindbrain nucleus critical for energy balance control. This effect is mediated by GLP-1R, as the uptake of systemically administered fluorophore-tagged exendin-4 was blocked by central pretreatment with the competitive GLP-1R antagonist exendin-(9-39). Ex vivo analyses show prolonged exendin-4-induced activation (live cell calcium signaling) of NTS astrocytes and neurons; these effects are also attenuated by exendin-(9-39), indicating mediation by the GLP-1R. In vitro analyses show that the application of GLP-1R agonists increases cAMP levels in astrocytes. Immunohistochemical analyses reveal that endogenous GLP-1 axons form close synaptic apposition with NTS astrocytes. Finally, pharmacological inhibition of NTS astrocytes attenuates the anorectic and body weight-suppressive effects of intra-NTS GLP-1R activation. Collectively, data demonstrate a role for NTS astrocytic GLP-1R signaling in energy balance control. SIGNIFICANCE STATEMENT: Glucagon-like peptide-1 receptor (GLP-1R) agonists reduce food intake and are approved by the Food and Drug Administration for the treatment of obesity, but the cellular mechanisms underlying the anorectic effects of GLP-1 require further investigation. Astrocytes represent a major cellular population in the CNS that regulates neurotransmission, yet the role of astrocytes in mediating energy balance is largely unstudied. The current data provide novel evidence that astrocytes within the NTS are relevant for energy balance control by GLP-1 signaling. Here, we report that GLP-1R agonists activate and internalize within NTS astrocytes, while behavioral data suggest the pharmacological relevance of NTS astrocytic GLP-1R activation for food intake and body weight. These findings support a previously unknown role for CNS astrocytes in energy balance control by GLP-1 signaling.


Asunto(s)
Regulación del Apetito/fisiología , Astrocitos/fisiología , Conducta Alimentaria/fisiología , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Homeostasis/fisiología , Bulbo Raquídeo/metabolismo , Animales , Metabolismo Energético/fisiología , Retroalimentación Fisiológica/fisiología , Masculino , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley
4.
J Am Chem Soc ; 139(46): 16688-16695, 2017 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29130686

RESUMEN

Peptide hormones are attractive as injectable therapeutics and imaging agents, but they often require extensive modification by mutagenesis and/or chemical synthesis to prevent rapid in vivo degradation. Alternatively, the single-atom, O-to-S modification of peptide backbone thioamidation has the potential to selectively perturb interactions with proteases while preserving interactions with other proteins, such as target receptors. Here, we use the validated diabetes therapeutic, glucagon-like peptide-1 (GLP-1), and the target of clinical investigation, gastric inhibitory polypeptide (GIP), as proof-of-principle peptides to demonstrate the value of thioamide substitution. In GLP-1 and GIP, a single thioamide near the scissile bond renders these peptides up to 750-fold more stable than the corresponding oxopeptides toward cleavage by dipeptidyl peptidase 4, the principal regulator of their in vivo stability. These stabilized analogues are nearly equipotent with their parent peptide in cyclic AMP activation assays, but the GLP-1 thiopeptides have much lower ß-arrestin potency, making them novel agonists with altered signaling bias. Initial tests show that a thioamide GLP-1 analogue is biologically active in rats, with an in vivo potency for glycemic control surpassing that of native GLP-1. Taken together, these experiments demonstrate the potential for thioamides to modulate specific protein interactions to increase proteolytic stability or tune activation of different signaling pathways.


Asunto(s)
Polipéptido Inhibidor Gástrico/química , Péptido 1 Similar al Glucagón/química , Tioamidas/química , Polipéptido Inhibidor Gástrico/uso terapéutico , Péptido 1 Similar al Glucagón/uso terapéutico , Estabilidad Proteica , Proteolisis
5.
Am J Physiol Regul Integr Comp Physiol ; 306(3): R157-63, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24352410

RESUMEN

Glucagon-like peptide-1 (GLP-1) is an incretin hormone released from intestinal L-cells in response to food entering into the gastrointestinal tract. GLP-1-based pharmaceuticals improve blood glucose regulation and may hold promise for obesity treatment, as GLP-1 drugs reduce food intake and body weight in humans and animals. In an effort to improve GLP-1 pharmacotherapies, we focused our attention on macronutrients that, when present in the gastrointestinal tract, may enhance GLP-1 secretion and improve glycemic regulation and food intake suppression when combined with systemic administration of sitagliptin, a pharmacological inhibitor of DPP-IV (enzyme responsible for GLP-1 degradation). In particular, previous data suggest that specific macronutrient constituents found in dairy foods may act as potent secretagogues for GLP-1 and therefore may potentially serve as an adjunct dietary therapy in combination with sitagliptin. To directly test this hypothesis, rats received intraperitoneal injections of sitagliptin (6 mg/kg) or saline vehicle followed by intraduodenal infusions of either milk protein concentrate (MPC; 80/20% casein/whey; 4 kcal), soy protein (nondairy control infusate; 4 kcal), or 0.9% NaCl. Food intake was assessed 30 min postinfusion. In separate studies, regulation of blood glucose was examined via a 2-h oral glucose tolerance test (2 g/kg) following identical sitagliptin treatment and intraduodenal nutrient infusions. Collectively, results show that intraduodenal MPC, but not soy protein, significantly enhances both the food intake suppression and improved control of blood glucose produced by sitagliptin. These data support the hypothesis that dietary intake of dairy protein may be beneficial as an adjunct behavioral therapy to enhance the glycemic and food intake suppressive effects of GLP-1-based pharmacotherapies.


Asunto(s)
Glucemia/efectos de los fármacos , Dipeptidil Peptidasa 4/efectos de los fármacos , Ingestión de Alimentos , Péptido 1 Similar al Glucagón/efectos de los fármacos , Hipoglucemiantes/uso terapéutico , Proteínas de la Leche/metabolismo , Pirazinas/uso terapéutico , Triazoles/uso terapéutico , Animales , Glucemia/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Péptido 1 Similar al Glucagón/metabolismo , Masculino , Obesidad/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley , Fosfato de Sitagliptina
6.
Am J Physiol Endocrinol Metab ; 305(6): E751-9, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23900416

RESUMEN

Glucagon-like peptide-1 (GLP-1) receptors (GLP-1R) expressed in the nucleus tractus solitarius (NTS) are physiologically required for the control of feeding. Recently, NTS GLP-1R-mediated suppression of feeding was shown to occur via a rapid PKA-induced suppression of AMPK and activation of MAPK signaling. Unknown are the additional intracellular signaling pathways that account for the long-term hypophagic effects of GLP-1R activation. Because cAMP/PKA activity can promote PI3K/PIP3-dependent translocation of Akt to the plasma membrane, we hypothesize that hindbrain GLP-1R-mediated control of feeding involves a PI3K-Akt-dependent pathway. Importantly, the novel evidence presented here challenges the dogmatic view that PI3K phosphorylation results in an obligatory activation of Akt and instead supports a growing body of literature showing that activation of cAMP/PKA can inhibit Akt phosphorylation at the plasma membrane. Behavioral data show that inhibition of hindbrain PI3K activity by a fourth icv administration of LY-294002 (3.07 µg) attenuated the food intake- and body weight-suppressive effects of a fourth icv administration of the GLP-1R agonist exendin-4 (0.3 µg) in rats. Hindbrain administration of triciribine (10 µg), an inhibitor of PIP3-dependent translocation of Akt to the cell membrane, also attenuated the intake-suppressive effects of a fourth icv injection of exendin-4. Immunoblot analyses of ex vivo NTS tissue lysates and in vitro GLP-1R-expressing neurons (GT1-7) support the behavioral findings and show that GLP-1R activation decreases phosphorylation of Akt in a time-dependent fashion. Current data reveal the requirement of PI3K activation, PIP3-dependent translocation of Akt to the plasma membrane, and suppression in phosphorylation of membrane-bound Akt to mediate the food intake-suppressive effects of hindbrain GLP-1R activation.


Asunto(s)
Ingestión de Alimentos/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Glucagón/metabolismo , Rombencéfalo/metabolismo , Animales , Cromonas/farmacología , Ingestión de Alimentos/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Receptor del Péptido 1 Similar al Glucagón , Morfolinas/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Rombencéfalo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
7.
Sci Rep ; 9(1): 8447, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31186439

RESUMEN

A growing appreciation of the overlapping neuroendocrine mechanisms controlling energy balance has highlighted combination therapies as a promising strategy to enhance sustained weight loss. Here, we investigated whether amylin- and glucagon-like-peptide-1 (GLP-1)-based combination therapies produce greater food intake- and body weight-suppressive effects compared to monotherapies in both lean and diet-induced obese (DIO) rats. In chow-maintained rats, systemic amylin and GLP-1 combine to reduce meal size. Furthermore, the amylin and GLP-1 analogs salmon calcitonin (sCT) and liraglutide produce synergistic-like reductions in 24 hours energy intake and body weight. The administration of sCT with liraglutide also led to a significant enhancement in cFos-activation in the dorsal-vagal-complex (DVC) compared to mono-therapy, suggesting an activation of distinct, yet overlapping neural substrates in this critical energy balance hub. In DIO animals, long-term daily administration of this combination therapy, specifically in a stepwise manner, results in reduced energy intake and greater body weight loss over time when compared to chronic mono- and combined-treated groups, without affecting GLP-1 receptor, preproglucagon or amylin-receptor gene expression in the DVC.


Asunto(s)
Calcitonina/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Obesidad/tratamiento farmacológico , Pérdida de Peso/efectos de los fármacos , Animales , Fármacos Antiobesidad/metabolismo , Fármacos Antiobesidad/farmacología , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Calcitonina/genética , Calcitonina/farmacología , Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Energía/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Péptido 1 Similar al Glucagón/genética , Péptido 1 Similar al Glucagón/farmacología , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Masculino , Obesidad/metabolismo , Obesidad/patología , Ratas , Receptores de Glucagón/genética
8.
Neuropsychopharmacology ; 43(3): 627-637, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28920591

RESUMEN

The neurobiological substrates that mediate the anorectic effects of both endogenous glucagon-like peptide-1 (GLP-1) and exogenous GLP-1 receptor (GLP-1R) agonists are an active area of investigation. As the lateral dorsal tegmental nucleus (LDTg) expresses the GLP-1R and represents a potential neuroanatomical hub connecting the nucleus tractus solitarius (NTS), the major central source of GLP-1, with the other nuclei in the midbrain and forebrain, we tested the hypothesis that GLP-1R signaling in the LDTg controls food intake. Direct activation of LDTg GLP-1R suppresses food intake through a reduction in average meal size and independent of nausea/malaise. Immunohistochemical data show that GLP-1-producing neurons in the NTS project to the LDTg, providing anatomical evidence of endogenous central GLP-1 in the LDTg. Pharmacological blockade of LDTg GLP-1Rs with exendin-(9-39) dose-dependently increases food intake and attenuates the hypophagic effects of gastric distension. As GLP-1 mimetics are administered systemically in humans, we evaluated whether peripherally administered GLP-1R agonists access the LDTg to affect feeding. Immunohistochemical data show that a systemically administered fluorescent GLP-1R agonist accesses the LDTg and is juxtaposed with neurons. Additionally, blockade of LDTg GLP-1Rs attenuates the hypophagic effects of a systemic GLP-1R agonist. Together, these data indicate that LDTg GLP-1R signaling controls energy balance and underscores the role of the LDTg in integrating energy balance-relevant signals to modulate feeding.


Asunto(s)
Ingestión de Alimentos/fisiología , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Tegmento Mesencefálico/metabolismo , Animales , Fármacos del Sistema Nervioso Central/farmacología , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Exenatida , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Masculino , Vías Nerviosas/citología , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fragmentos de Péptidos/farmacología , Péptidos/farmacología , Ratas Sprague-Dawley , Núcleo Solitario/citología , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/metabolismo , Tegmento Mesencefálico/citología , Tegmento Mesencefálico/efectos de los fármacos , Ponzoñas/farmacología
9.
Physiol Behav ; 177: 122-128, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28433470

RESUMEN

Glucagon-like peptide-1 (GLP-1) is an incretin hormone released from intestinal L-cells in response to food entering into the gastrointestinal tract. GLP-1-based pharmaceuticals improve blood glucose regulation and reduce feeding. Specific macronutrients, when ingested, may trigger GLP-1 secretion and enhance the effects of systemic sitagliptin, a pharmacological inhibitor of DPP-IV (an enzyme that rapidly degrades GLP-1). In particular, macronutrient constituents found in dairy foods may act as potent secretagogues for GLP-1, and acute preclinical trials show that ingestion of dairy protein may represent a promising adjunct behavioral therapy in combination with sitagliptin. To test this hypothesis further, chow-maintained or high-fat diet (HFD)-induced obese rats received daily IP injections of sitagliptin (6mg/kg) or saline in combination with a twice-daily 8ml oral gavage of milk protein concentrate (MPC; 80/20% casein/whey; 0.5kcal/ml), soy protein (non-dairy control; 0.5kcal/ml) or 0.9% NaCl for two months. Food intake and body weight were recorded every 24-48h; blood glucose regulation was examined at baseline and at 3 and 6.5weeks via a 2h oral glucose tolerance test (OGTT; 25% glucose; 2g/kg). MPC and soy protein significantly suppressed cumulative caloric intake in HFD but not chow-maintained rats. AUC analyses for OGTT show suppression in glycemia by sitagliptin with MPC or soy in chow- and HFD-maintained rats, suggesting that chronic ingestion of dairy or soy proteins may augment endogenous GLP-1 signaling and the glycemic- and food intake-suppressive effects of DPP-IV inhibition.


Asunto(s)
Fármacos Antiobesidad/farmacología , Proteínas en la Dieta/administración & dosificación , Suplementos Dietéticos , Obesidad/dietoterapia , Obesidad/tratamiento farmacológico , Fosfato de Sitagliptina/farmacología , Alimentación Animal , Animales , Dieta Alta en Grasa , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Modelos Animales de Enfermedad , Ingestión de Energía/efectos de los fármacos , Ingestión de Energía/fisiología , Péptido 1 Similar al Glucagón/metabolismo , Prueba de Tolerancia a la Glucosa , Masculino , Obesidad/metabolismo , Ratas Sprague-Dawley
10.
Physiol Behav ; 173: 9-14, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28119159

RESUMEN

The beneficial glycemic and food intake-suppressive effects of glucagon-like peptide-1 (GLP-1) have made this neuroendocrine system a leading target for pharmacological approaches to the treatment of diabetes and obesity. One strategy to increase the activity of endogenous GLP-1 is to prevent the rapid degradation of the hormone by the enzyme dipeptidyl peptidase-IV (DPP-IV). However, despite the expression of both DPP-IV and GLP-1 in the brain, and the clear importance of central GLP-1 receptor (GLP-1R) signaling for glycemic and energy balance control, the metabolic effects of central inhibition of DPP-IV activity are unclear. To test whether hindbrain DPP-IV inhibition suppresses blood glucose, feeding, and body weight gain, the effects of 4th intracerebroventricular (ICV) administration of the FDA-approved DPP-IV inhibitor sitagliptin were evaluated. Results indicate that hindbrain delivery of sitagliptin improves glycemic control in a GLP-1R-dependent manner, suggesting that this effect is due at least in part to increased endogenous brainstem GLP-1 activity after sitagliptin administration. Furthermore, 4th ICV injection of sitagliptin reduced 24h body weight gain and energy intake, with a selective suppression of high-fat diet, but not chow, intake. These data reveal a novel role for hindbrain GLP-1R activation in glycemic control and also demonstrate that DPP-IV inhibition in the caudal brainstem promotes negative energy balance.


Asunto(s)
Glucemia/fisiología , Dipeptidil Peptidasa 4/metabolismo , Metabolismo Energético/fisiología , Rombencéfalo/metabolismo , Animales , Área Bajo la Curva , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Dieta Alta en Grasa/métodos , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Metabolismo Energético/efectos de los fármacos , Ayuno , Prueba de Tolerancia a la Glucosa , Inyecciones Intraventriculares , Masculino , Ratas , Ratas Sprague-Dawley , Rombencéfalo/efectos de los fármacos , Fosfato de Sitagliptina/farmacología
11.
Neuropharmacology ; 123: 67-79, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28552704

RESUMEN

Amylin is produced in the pancreas and the brain, and acts centrally to reduce feeding and body weight. Recent data show that amylin can act in the ventral tegmental area (VTA) to reduce palatable food intake and promote negative energy balance, but the behavioral mechanisms by which these effects occur are not fully understood. The ability of VTA amylin signaling to reduce intake of specific palatable macronutrients (fat or carbohydrate) was tested in rats in several paradigms, including one-bottle acceptance tests, two-bottle choice tests, and a free-choice diet. Data show that VTA amylin receptor activation with the amylin receptor agonist salmon calcitonin (sCT) preferentially and potently reduces intake of fat, with more variable suppression of sucrose intake. Intake of a non-nutritive sweetener is also decreased by intra-VTA administration of sCT. As several feeding-related signals that act in the mesolimbic system also impact motivated behaviors besides feeding, we tested the hypothesis that the suppressive effects of amylin signaling in the VTA extend to other motivationally relevant stimuli. Results show that intra-VTA sCT reduces water intake in response to central administration of the dipsogenic peptide angiotensin II, but has no effect on ad libitum water intake in the absence of food. Importantly, open field and social interaction studies show that VTA amylin signaling does not produce anxiety-like behaviors. Collectively, these findings reveal a novel ability of VTA amylin receptor activation to alter palatable macronutrient intake, and also demonstrate a broader role of VTA amylin signaling for the control of motivated ingestive behaviors beyond feeding.


Asunto(s)
Agonistas de los Receptores de Amilina/farmacología , Calcitonina/farmacología , Conducta Alimentaria/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos , Angiotensina II/farmacología , Animales , Ansiedad , Conducta de Elección/efectos de los fármacos , Carbohidratos de la Dieta , Grasas de la Dieta , Sacarosa en la Dieta , Agua Potable , Masculino , Ratas Sprague-Dawley , Receptores de Polipéptido Amiloide de Islotes Pancreáticos/metabolismo , Sacarina , Salmón , Área Tegmental Ventral/metabolismo
12.
Biol Psychiatry ; 82(11): 828-838, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28237459

RESUMEN

BACKGROUND: The pancreatic- and brain-derived hormone amylin promotes negative energy balance and is receiving increasing attention as a promising obesity therapeutic. However, the neurobiological substrates mediating amylin's effects are not fully characterized. We postulated that amylin acts in the lateral dorsal tegmental nucleus (LDTg), an understudied neural processing hub for reward and homeostatic feeding signals. METHODS: We used immunohistochemical and quantitative polymerase chain reaction analyses to examine expression of the amylin receptor complex in rat LDTg tissue. Behavioral experiments were performed to examine the mechanisms underlying the hypophagic effects of amylin receptor activation in the LDTg. RESULTS: Immunohistochemical and quantitative polymerase chain reaction analyses show expression of the amylin receptor complex in the LDTg. Activation of LDTg amylin receptors by the agonist salmon calcitonin dose-dependently reduces body weight, food intake, and motivated feeding behaviors. Acute pharmacological studies and longer-term adeno-associated viral knockdown experiments indicate that LDTg amylin receptor signaling is physiologically and potentially preclinically relevant for energy balance control. Finally, immunohistochemical data indicate that LDTg amylin receptors are expressed on gamma-aminobutyric acidergic neurons, and behavioral results suggest that local gamma-aminobutyric acid receptor signaling mediates the hypophagia after LDTg amylin receptor activation. CONCLUSIONS: These findings identify the LDTg as a novel nucleus with therapeutic potential in mediating amylin's effects on energy balance through gamma-aminobutyric acid receptor signaling.


Asunto(s)
Agonistas de los Receptores de Amilina/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Transducción de Señal/fisiología , Área Tegmental Ventral/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Calcitonina/farmacología , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Ingestión de Alimentos/efectos de los fármacos , Preferencias Alimentarias/efectos de los fármacos , GABAérgicos/farmacología , Masculino , Motivación/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Fosfopiruvato Hidratasa/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Modificadoras de la Actividad de Receptores/genética , Proteínas Modificadoras de la Actividad de Receptores/metabolismo , Receptores de Polipéptido Amiloide de Islotes Pancreáticos/antagonistas & inhibidores , Receptores de Polipéptido Amiloide de Islotes Pancreáticos/genética , Receptores de Polipéptido Amiloide de Islotes Pancreáticos/metabolismo , Transducción de Señal/efectos de los fármacos
13.
Obesity (Silver Spring) ; 24(10): 2118-25, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27558648

RESUMEN

OBJECTIVE: Binge eating is characterized by repeated intermittent bouts of compulsive overconsumption of food. Treatment is challenging given limited understanding of the mechanisms underlying this type of disordered eating. The hypothesis that dysregulation of mesocortical dopaminergic and GABAergic systems underlie binge eating was tested. METHODS: Analysis of gene expression within the ventral tegmental area and its terminal mesocortical regions was examined in bingeing rats before and after bingeing occurred. In addition, alterations in binge-type behavior induced by pharmacological inactivation of subnuclei of the prefrontal cortex (PFC) and by pharmacological activation and inhibition of cortical D1 and D2 receptors were examined. RESULTS: Correlative and functional evidence demonstrates dysregulated neurotransmitter processing by the PFC and ventral tegmental area, but not the amygdala or nucleus accumbens, in bingeing rats. Either GABAergic inactivation or D2-like receptor activation within the PFC increased consumption in bingeing rats, but not controls, suggesting that the PFC, and D2 receptors in particular, functions as a behavioral brake to limit bingeing. CONCLUSIONS: The act of bingeing resolved some gene expression differences that preceded binge onset, further suggesting that bingeing may partially serve to self-medicate a system driving this maladaptive behavior. However, the failure of bingeing to resolve other dopaminergic/GABAergic differences may render individuals vulnerable to future binge episodes.


Asunto(s)
Bulimia/metabolismo , Dopamina/metabolismo , Corteza Prefrontal/metabolismo , Transmisión Sináptica/fisiología , Área Tegmental Ventral/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Amígdala del Cerebelo/metabolismo , Animales , Ingestión de Alimentos/fisiología , Masculino , Núcleo Accumbens/metabolismo , Ratas
14.
Neuropsychopharmacology ; 40(2): 372-85, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25035079

RESUMEN

Amylin acts in the CNS to reduce feeding and body weight. Recently, the ventral tegmental area (VTA), a mesolimbic nucleus important for food intake and reward, was identified as a site-of-action mediating the anorectic effects of amylin. However, the long-term physiological relevance and mechanisms mediating the intake-suppressive effects of VTA amylin receptor (AmyR) activation are unknown. Data show that the core component of the AmyR, the calcitonin receptor (CTR), is expressed on VTA dopamine (DA) neurons and that activation of VTA AmyRs reduces phasic DA in the nucleus accumbens core (NAcC). Suppression in NAcC DA mediates VTA amylin-induced hypophagia, as combined NAcC D1/D2 receptor agonists block the intake-suppressive effects of VTA AmyR activation. Knockdown of VTA CTR via adeno-associated virus short hairpin RNA resulted in hyperphagia and exacerbated body weight gain in rats maintained on high-fat diet. Collectively, these findings show that VTA AmyR signaling controls energy balance by modulating mesolimbic DA signaling.


Asunto(s)
Agonistas de los Receptores de Amilina/farmacología , Depresores del Apetito/farmacología , Dopamina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Neuronas/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos , Animales , Dieta Alta en Grasa , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Masculino , Neuronas/metabolismo , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptores de Calcitonina/antagonistas & inhibidores , Receptores de Calcitonina/genética , Receptores de Calcitonina/metabolismo , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/metabolismo , Receptores de Polipéptido Amiloide de Islotes Pancreáticos/metabolismo , Área Tegmental Ventral/metabolismo , Aumento de Peso/efectos de los fármacos , Aumento de Peso/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA