Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Metab ; 66: 101641, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36396031

RESUMEN

OBJECTIVES: Glucagon-like peptide-1 receptor (GLP-1R) agonists reduce the rates of major cardiovascular events, including myocardial infarction in people with type 2 diabetes, and decrease infarct size while preserving ventricular function in preclinical studies. Nevertheless, the precise cellular sites of GLP-1R expression that mediate the cardioprotective actions of GLP-1 in the setting of ischemic cardiac injury are uncertain. METHODS: Publicly available single cell RNA sequencing (scRNA-seq) datasets on mouse and human heart cells were analyzed for Glp1r/GLP1R expression. Fluorescent activated cell sorting was used to localize Glp1r expression in cell populations from the mouse heart. The importance of endothelial and hematopoietic cells for the cardioprotective response to liraglutide in the setting of acute myocardial infarction (MI) was determined by inactivating the Glp1r in Tie2+ cell populations. Cardiac gene expression profiles regulated by liraglutide were examined using RNA-seq to interrogate mouse atria and both infarcted and non-infarcted ventricular tissue after acute coronary artery ligation. RESULTS: In mice, cardiac Glp1r mRNA transcripts were exclusively detected in endocardial cells by scRNA-seq. In contrast, analysis of human heart by scRNA-seq localized GLP1R mRNA transcripts to populations of atrial and ventricular cardiomyocytes. Moreover, very low levels of GIPR, GCGR and GLP2R mRNA transcripts were detected in the human heart. Cell sorting and RNA analyses detected cardiac Glp1r expression in endothelial cells (ECs) within the atria and ventricle in the ischemic and non-ischemic mouse heart. Transcriptional responses to liraglutide administration were not evident in wild type mouse ventricles following acute MI, however liraglutide differentially regulated genes important for inflammation, cardiac repair, cell proliferation, and angiogenesis in the left atrium, while reducing circulating levels of IL-6 and KC/GRO within hours of acute MI. Inactivation of the Glp1r within the Tie2+ cell expression domain encompassing ECs revealed normal cardiac structure and function, glucose homeostasis and body weight in Glp1rTie2-/- mice. Nevertheless, the cardioprotective actions of liraglutide to reduce infarct size, augment ejection fraction, and improve survival after experimental myocardial infarction (MI), were attenuated in Glp1rTie2-/- mice. CONCLUSIONS: These findings identify the importance of the murine Tie2+ endothelial cell GLP-1R as a target for the cardioprotective actions of GLP-1R agonists and support the importance of the atrial and ventricular endocardial GLP-1R as key sites of GLP-1 action in the ischemic mouse heart. Hitherto unexplored species-specific differences in cardiac GLP-1R expression challenge the exclusive use of mouse models for understanding the mechanisms of GLP-1 action in the normal and ischemic human heart.


Asunto(s)
Fibrilación Atrial , Receptor del Péptido 1 Similar al Glucagón , Liraglutida , Infarto del Miocardio , Animales , Humanos , Ratones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Células Endoteliales/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Liraglutida/farmacología , Infarto del Miocardio/tratamiento farmacológico , ARN Mensajero , Modelos Animales de Enfermedad , Receptor TIE-2/metabolismo
2.
Peptides ; 153: 170811, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35594964

RESUMEN

A group of long-acting, peptide-based, and selective GLP-1R/CCK-2R dual agonizts were identified by rational design. Guided by sequence analysis, structural elements of the CCK-2R agonist moiety were engineered into the GLP-1R agonist Xenopus GLP-1, resulting in hybrid peptides with potent GLP-1R/CCK-2R dual activity. Further modifications with fatty acids resulted in novel metabolically stable peptides, among which 3d and 3 h showed potent GLP-1R and CCK-2R activation potencies and comparable stability to semaglutide. In food intake tests, 3d and 3 h also showed a potent reduction in food intake, superior to that of semaglutide. Moreover, the acute hypoglycemic and insulinotropic activities of 3d and 3 h were better than that of semaglutide and ZP3022. Importantly, the limited pica response following 3d and 3 h administration in SD rats preliminarily indicated that the food intake reduction effects of 3d and 3 h are independent of nausea/vomiting. In a 35-day study in db/db mice, every two days administration of 3d and 3 h increased islet areas and numbers, insulin contents, ß-cell area, ß-cell proliferation, as well as improved glucose tolerance, and decreased HbA1c, to a greater extent than ZP3022 and semaglutide. In a 21-day study in DIO mice, once-weekly administration of 3d and 3 h significantly induced body weight loss, improved glucose tolerance, and normalized lipid metabolism, to a greater extent than semaglutide. The current study showed the antidiabetic and antiobesity potentials of GLP-1R/CCK-2R dual agonizts that warrant further investigation.


Asunto(s)
Diabetes Mellitus Tipo 2 , Receptor del Péptido 1 Similar al Glucagón , Hipoglucemiantes , Péptidos , Receptor de Colecistoquinina B , Animales , Fármacos Antiobesidad/farmacología , Colecistoquinina/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Glucosa , Hipoglucemiantes/farmacología , Ratones , Péptidos/química , Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Colecistoquinina B/agonistas , Receptor de Colecistoquinina B/efectos de los fármacos , Receptor de Colecistoquinina B/metabolismo
3.
Cell Rep ; 36(8): 109609, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34433018

RESUMEN

Agonists for glucagon-like-peptide-1 receptor (GLP-1R) are currently used for the treatment of type 2 diabetes and obesity. Their benefits have been centered on pancreas and hypothalamus, but their roles in other organ systems are not well understood. We studied the action of GLP-1R on secretions of adrenal medulla. Exendin-4, a synthetic analog of GLP-1, increases the synthesis and the release of catecholamines (CAs) by increasing cyclic AMP (cAMP) production, without apparent participation of cAMP-regulated guanine nucleotide exchange factor (Epac). Exendin-4, when incubated for 24 h, increases CA synthesis by promoting the activation of tyrosine hydroxylase. Short incubation (20 min) increases the quantum size of exocytotic events by switching exocytosis from partial to full fusion. Our results give a strong support to the role of GLP-1 in the fine control of exocytosis.


Asunto(s)
Células Cromafines/metabolismo , Exenatida/farmacología , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Animales , Células Cromafines/efectos de los fármacos , AMP Cíclico/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Exenatida/metabolismo , Exocitosis/efectos de los fármacos , Exocitosis/fisiología , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Factores de Intercambio de Guanina Nucleótido/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratas , Receptores de Glucagón/efectos de los fármacos , Receptores de Glucagón/metabolismo
4.
Neurotoxicol Teratol ; 87: 106984, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33864929

RESUMEN

Evidence supporting the use of glucagon-like peptide-1 (GLP-1) analogues to pharmacologically treat disorders beyond type 2 diabetes and obesity is increasing. However, little is known about how activation of the GLP-1 receptor (GLP-1R) during pregnancy affects maternal and offspring outcomes. We treated female C57Bl/6 J mice prior to conception and throughout gestation with a long-lasting GLP-1R agonist, Exendin-4. While GLP-1R activation has significant effects on food and drug reward, depression, locomotor activity, and cognition in adults, we found few changes in these domains in exendin-4-exposed offspring. Repeated injections of Exendin-4 had minimal effects on the dams and may have enhanced maternal care. Offspring exposed to the drug weighed significantly more than their control counterparts during the preweaning period and demonstrated alterations in anxiety-like outcomes, which indicate a developmental role for GLP-1R modulation in the stress response that may be sex-specific.


Asunto(s)
Exenatida/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Tiempo , Animales , Encéfalo/metabolismo , Cognición/efectos de los fármacos , Exenatida/efectos de los fármacos , Femenino , Péptido 1 Similar al Glucagón/metabolismo , Ratones Endogámicos C57BL
6.
JCI Insight ; 6(4)2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33434183

RESUMEN

Somatostatin (SS) inhibits glucagon-like peptide-1 (GLP-1) secretion in a paracrine manner. We hypothesized that blocking somatostatin subtype receptor 2 (SSTR2) and 5 (SSTR5) would improve glycemia by enhancing GLP-1 secretion. In the perfused mouse small intestine, the selective SSTR5 antagonist (SSTR5a) stimulated glucose-induced GLP-1 secretion to a larger degree than the SSTR2 antagonist (SSTR2a). In parallel, mice lacking the SSTR5R showed increased glucose-induced GLP-1 secretion. Both antagonists improved glycemia in vivo in a GLP-1 receptor-dependent (GLP-1R-dependent) manner, as the glycemic improvements were absent in mice with impaired GLP-1R signaling and in mice treated with a GLP-1R-specific antagonist. SSTR5a had no direct effect on insulin secretion in the perfused pancreas, whereas SSTR2a increased insulin secretion in a GLP-1R-independent manner. Adding a dipeptidyl peptidase 4 inhibitor (DPP-4i) in vivo resulted in additive effects on glycemia. However, when glucose was administered intraperitoneally, the antagonist was incapable of lowering blood glucose. Oral administration of SSTR5a, but not SSTR2a, lowered blood glucose in diet-induced obese mice. In summary, we demonstrate that selective SSTR antagonists can improve glucose control primarily through the intestinal GLP-1 system in mice.


Asunto(s)
Glucemia/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Hipoglucemiantes/farmacología , Receptores de Somatostatina/antagonistas & inhibidores , Animales , Glucemia/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Modelos Animales de Enfermedad , Femenino , Péptido 1 Similar al Glucagón/metabolismo , Insulina , Secreción de Insulina/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Receptores de Somatostatina/genética
7.
Mol Metab ; 43: 101118, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33221554

RESUMEN

OBJECTIVES: Combinatorial therapies are under intense investigation to develop more efficient anti-obesity drugs; however, little is known about how they act in the brain to produce enhanced anorexia and weight loss. The goal of this study was to identify the brain sites and neuronal populations engaged during the co-administration of GLP-1R and CCK1R agonists, an efficient combination therapy in obese rodents. METHODS: We measured acute and long-term feeding and body weight responses and neuronal activation patterns throughout the neuraxis and in specific neuronal subsets in response to GLP-1R and CCK1R agonists administered alone or in combination in lean and high-fat diet fed mice. We used PhosphoTRAP to obtain unbiased molecular markers for neuronal populations selectively activated by the combination of the two agonists. RESULTS: The initial anorectic response to GLP-1R and CCK1R co-agonism was mediated by a reduction in meal size, but over a few hours, a reduction in meal number accounted for the sustained feeding suppressive effects. The nucleus of the solitary tract (NTS) is one of the few brain sites where GLP-1R and CCK1R signalling interact to produce enhanced neuronal activation. None of the previously categorised NTS neuronal subpopulations relevant to feeding behaviour were implicated in this increased activation. However, we identified NTS/AP Calcrl+ neurons as treatment targets. CONCLUSIONS: Collectively, these studies indicated that circuit-level integration of GLP-1R and CCK1R co-agonism in discrete brain nuclei including the NTS produces enhanced rapid and sustained appetite suppression and weight loss.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón/metabolismo , Obesidad/tratamiento farmacológico , Receptores de Colecistoquinina/metabolismo , Animales , Fármacos Antiobesidad/farmacología , Regulación del Apetito , Encéfalo/metabolismo , Dieta Alta en Grasa , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Obesidad/metabolismo , Núcleo Solitario/metabolismo , Pérdida de Peso/efectos de los fármacos
8.
Diabetologia ; 64(1): 142-151, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33043402

RESUMEN

AIMS/HYPOTHESIS: The endocrine pancreas comprises the islets of Langerhans, primarily consisting of beta cells, alpha cells and delta cells responsible for secretion of insulin, glucagon and somatostatin, respectively. A certain level of intra-islet communication is thought to exist, where the individual hormones may reach the other islet cells and regulate their secretion. Glucagon has been demonstrated to importantly regulate insulin secretion, while somatostatin powerfully inhibits both insulin and glucagon secretion. In this study we investigated how secretion of somatostatin is regulated by paracrine signalling from glucagon and insulin. METHODS: Somatostatin secretion was measured from perfused mouse pancreases isolated from wild-type as well as diphtheria toxin-induced alpha cell knockdown, and global glucagon receptor knockout (Gcgr-/-) mice. We studied the effects of varying glucose concentrations together with infusions of arginine, glucagon, insulin and somatostatin, as well as infusions of antagonists of insulin, somatostatin and glucagon-like peptide 1 (GLP-1) receptors. RESULTS: A tonic inhibitory role of somatostatin was demonstrated with infusion of somatostatin receptor antagonists, which significantly increased glucagon secretion at low and high glucose, whereas insulin secretion was only increased at high glucose levels. Infusion of glucagon dose-dependently increased somatostatin secretion approximately twofold in control mice. Exogenous glucagon had no effect on somatostatin secretion in Gcgr-/- mice, and a reduced effect when combined with the GLP-1 receptor antagonist exendin 9-39. Diphtheria toxin-induced knockdown of glucagon producing cells led to reduced somatostatin secretion in response to 12 mmol/l glucose and arginine infusions. In Gcgr-/- mice (where glucagon levels are dramatically increased) overall somatostatin secretion was increased. However, infusion of exendin 9-39 in Gcgr-/- mice completely abolished somatostatin secretion in response to glucose and arginine. Neither insulin nor an insulin receptor antagonist (S961) had any effect on somatostatin secretion. CONCLUSIONS/INTERPRETATION: Our findings demonstrate that somatostatin and glucagon secretion are linked in a reciprocal feedback cycle with somatostatin inhibiting glucagon secretion at low and high glucose levels, and glucagon stimulating somatostatin secretion via the glucagon and GLP-1 receptors. Graphical abstract.


Asunto(s)
Glucagón/fisiología , Insulina/fisiología , Somatostatina/metabolismo , Animales , Arginina/administración & dosificación , Comunicación Celular , Toxina Diftérica/farmacología , Técnicas de Silenciamiento del Gen , Glucagón/administración & dosificación , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/fisiología , Células Secretoras de Glucagón/efectos de los fármacos , Células Secretoras de Glucagón/fisiología , Glucosa/administración & dosificación , Insulina/administración & dosificación , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Glucagón/deficiencia , Receptores de Glucagón/genética , Receptores de Glucagón/fisiología , Receptores de Somatostatina/antagonistas & inhibidores , Transducción de Señal/fisiología , Somatostatina/administración & dosificación
9.
Aging (Albany NY) ; 13(1): 525-536, 2020 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-33298623

RESUMEN

Background and aims Advanced glycation end products (AGEs) and receptor of advanced glycation end products (RAGE), are associated with cognition decline. We aim to investigate the effect of liraglutide on cognitive function in diabetic mice. Results Diabetic mice showed decreased cognitive function. Moreover, lower glucagon like peptide-1 (GLP-1) levels in plasma were detected in db/db mice. Additionally, up-regulated RAGE and down-regulated glucagon like peptide-1 (GLP-1R) levels were observed in db/db mice. However, decreased GLP-1R and increased RAGE were reversed by liraglutide. We also found decreased cellular activity in cells with AGEs. Moreover, AGEs up-regulated RAGE in PC12 and HT22 cells. However, liraglutide improved the cell activity damaged by AGEs. Although we did not discover the direct-interaction between RAGE and GLP-1R, elevated RAGE levels induced by AGEs were restored by liraglutide. Conclusion We demonstrated that the cognitive function of diabetic mice was improved by liraglutide via the down-regulation of RAGE. Methods db/db mice and db/m mice were used in this study. Liraglutide was used to remedy diabetic mice. Neurons and RAGE in hippocampus were shown by immunofluorescence. And then, PC12 cells or HT22 cells with AGEs were treated with liraglutide. GLP-1R and RAGE were measured by western blotting.


Asunto(s)
Cognición/efectos de los fármacos , Disfunción Cognitiva/metabolismo , Diabetes Mellitus/metabolismo , Péptido 1 Similar al Glucagón/efectos de los fármacos , Productos Finales de Glicación Avanzada/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipoglucemiantes/farmacología , Liraglutida/farmacología , Neuronas/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Disfunción Cognitiva/fisiopatología , Diabetes Mellitus/fisiopatología , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Productos Finales de Glicación Avanzada/farmacología , Hipocampo/citología , Hipocampo/metabolismo , Ratones , Ratones Obesos , Células PC12 , Ratas , Receptor para Productos Finales de Glicación Avanzada/efectos de los fármacos , Receptor para Productos Finales de Glicación Avanzada/metabolismo
10.
Alcohol Clin Exp Res ; 44(10): 2118-2129, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33043520

RESUMEN

BACKGROUND: The misuse and abuse of alcohol is a major public health issue. However, available treatments are limited with variable efficacy. Recently, preclinical studies show that glucagon-like-peptide-1 (GLP-1) and its analogue Exendin-4 (Ex4) potently reduce a range of alcohol intake behaviors, thus highlighting its potential as a treatment for alcohol use disorders. However, the neural mechanisms and sites of action mediating the effects of Ex4 on alcohol intake behaviors remain to be characterized. This study examined the ventral tegmental area (VTA) as a site of action for the effects of GLP-1 on alcohol intake. METHODS: Male Long-Evans rats were given intermittent access to 20% alcohol and trained to nose poke for 20% alcohol. Rats received intra-VTA injections of Ex4 (vehicle, 0.01, 0.05 µg), and the effects of VTA Ex4 on alcohol self-administration, motivation, and relapse were assessed. RESULTS: When compared to vehicle treatment, intra-VTA Ex4 (0.01, 0.05 µg) delivery significantly reduced alcohol self-administration, an effect that was particularly prominent in high alcohol drinkers. However, VTA Ex4 did not reduce reacquisition of alcohol self-administration after extinction nor the motivation to obtain alcohol. Importantly, the lower dose of Ex4 (0.01 µg) used had no effect on food intake or locomotor activity, suggesting that the reduction in alcohol self-administration observed was not secondary to caloric intake or motor deficits. CONCLUSIONS: Together, these findings provide support for the VTA as a key site of action for GLP-1 on alcohol self-administration but not the reacquisition of alcohol self-administration or motivation to work for alcohol.


Asunto(s)
Disuasivos de Alcohol/farmacología , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Exenatida/farmacología , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos , Disuasivos de Alcohol/metabolismo , Disuasivos de Alcohol/uso terapéutico , Consumo de Bebidas Alcohólicas/metabolismo , Consumo de Bebidas Alcohólicas/fisiopatología , Animales , Exenatida/metabolismo , Exenatida/uso terapéutico , Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/uso terapéutico , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/fisiología , Masculino , Actividad Motora/efectos de los fármacos , Ratas , Ratas Long-Evans , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/fisiopatología
11.
Life Sci ; 257: 118025, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32598933

RESUMEN

BACKGROUND: Glucagon-like peptide-1 receptor (GLP-1R) and glucose-dependent insulinotropic polypeptide receptor (GIPR) co-agonists have emerged as treatment options for reversing diabetes and obesity. Here, we screened the high potency receptor-biased GLP-1R agonists via a newly designed high-throughput GLP-1R extracellular domain (ECD)-based system and demonstrated its in vitro and in vivo therapeutic characters. METHODS: Twelve 9-mer peptides (named XEL1-XEL12) which were screened from a large phage-displayed peptide library were fused to the N-terminus of GIP (3-30) to generate another twelve fusion peptides, termed XEL13-24. Using the six lysine-altered XEL17 as leading sequences, eighteen fatty chain modified fusion peptides were further assessed via in vitro GLP-1R/GIPR-based cell assay. Moreover, the acute and long-acting in vivo effects of selected candidate on diabetic db/db mice and diet-induced obesity (DIO) rats were both carefully evaluated. RESULTS: XEL17 exhibited balanced activation potency on GLP-1R/GIPR in stable cell lines, and further assessment was performed to evaluate the XEL32, a fatty chain modified XEL17 derivative. Preclinical pharmacodynamic results in diabetic db/db mice demonstrated that XEL32 held outstanding insulinotropic and glucose-lowering activities. In addition, protracted antidiabetic effects of XEL32 were also proved by the hypoglycemic test and multiple oral glucose tolerance test. Furthermore, chronic treatment of XEL32 in DIO rats exhibited outstanding beneficial effects on body weight control, fat loss, food intake control, hemoglobin A1C (HbA1C) reduction as well as the glucose tolerance. CONCLUSIONS: XEL32, as a novel GLP-1/GIP dual receptor agonist, may supply efficient glycemic control and weight loss.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón/metabolismo , Péptidos/farmacología , Receptores de la Hormona Gastrointestinal/metabolismo , Pérdida de Peso/efectos de los fármacos , Animales , Glucemia/metabolismo , China , Diabetes Mellitus/tratamiento farmacológico , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Células HEK293 , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Hipoglucemiantes/farmacología , Masculino , Ratones , Obesidad/metabolismo , Ratas , Receptores de la Hormona Gastrointestinal/agonistas , Receptores de la Hormona Gastrointestinal/efectos de los fármacos
12.
JCI Insight ; 5(6)2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32213703

RESUMEN

Semaglutide, a glucagon-like peptide 1 (GLP-1) analog, induces weight loss, lowers glucose levels, and reduces cardiovascular risk in patients with diabetes. Mechanistic preclinical studies suggest weight loss is mediated through GLP-1 receptors (GLP-1Rs) in the brain. The findings presented here show that semaglutide modulated food preference, reduced food intake, and caused weight loss without decreasing energy expenditure. Semaglutide directly accessed the brainstem, septal nucleus, and hypothalamus but did not cross the blood-brain barrier; it interacted with the brain through the circumventricular organs and several select sites adjacent to the ventricles. Semaglutide induced central c-Fos activation in 10 brain areas, including hindbrain areas directly targeted by semaglutide, and secondary areas without direct GLP-1R interaction, such as the lateral parabrachial nucleus. Automated analysis of semaglutide access, c-Fos activity, GLP-1R distribution, and brain connectivity revealed that activation may involve meal termination controlled by neurons in the lateral parabrachial nucleus. Transcriptomic analysis of microdissected brain areas from semaglutide-treated rats showed upregulation of prolactin-releasing hormone and tyrosine hydroxylase in the area postrema. We suggest semaglutide lowers body weight by direct interaction with diverse GLP-1R populations and by directly and indirectly affecting the activity of neural pathways involved in food intake, reward, and energy expenditure.


Asunto(s)
Peso Corporal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Péptidos Similares al Glucagón/farmacología , Vías Nerviosas/efectos de los fármacos , Animales , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Ratones , Ratas
13.
Bioorg Chem ; 95: 103538, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31901754

RESUMEN

Oxyntomodulin (OXM) is an endogenous gastrointestinal hormone, which activates both the Glucagon-like peptide-1 receptor (GLP-1R) and the glucagon receptor (GCGR). However, OXM has shortcomings including poor GLP-1R agonism to control glycemia, short half-life and others. Inspired from the sequence relationship between OXM and glucagon, in this study, we introduced different C-terminus residues of GLP-1, exenatide and OXM to glucagon to get a series of hybrid peptides with enhanced GLP-1R activation. The formed glucagon-exenatide hybrid peptide shows higher GLP-1R activation properties than OXM. Then the peptides based on the glucagon-exenatide hybrid peptide were coupled with fatty acid side chains to prolong their half-lives. As a result, the most potent compound 16a could stimulate insulin secretion and maintain blood glucose in normal level for ~42.6 h in diabetic mice. 16a exhibited reduced HbA1c level in diabetic mice, lowered body weight significantly in obesity mice on chronic treatment assay. 16a, combined efficient GCGR/GLP-1R activity, is potential as novel treatment for obesity and diabetes. This finding provides new insights into balancing GLP-1/GCGR potency of glucagon-exenatide hybrid peptide and is helpful for discovery of novel anti-diabetic and bodyweight-reducing drugs.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Glucagón/química , Hipoglucemiantes/farmacología , Oxintomodulina/química , Péptidos/farmacología , Pérdida de Peso/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Glucemia/metabolismo , Ingestión de Energía , Prueba de Tolerancia a la Glucosa , Hipoglucemiantes/uso terapéutico , Ratones , Obesidad/tratamiento farmacológico , Obesidad/etiología , Péptidos/química , Péptidos/uso terapéutico , Homología de Secuencia de Aminoácido , Estreptozocina , Relación Estructura-Actividad
14.
FASEB J ; 33(11): 11836-11844, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31370679

RESUMEN

Currently, quantifying ß-cell mass (BCM) requires harvesting the pancreas. In this study, we investigated a potential noninvasive method to quantify BCM changes longitudinally using [Lys12(111In-BnDTPA-Ahx)]exendin-4 ([111In]-Ex4) and single-photon emission computed tomography (SPECT). We used autoradiography and transgenic mice expressing green fluorescent protein under the control of mouse insulin 1 gene promotor to evaluate the specificity of [111In]-Ex4 toward ß cells. Using nonobese diabetic (NOD) mice, we injected [111In]-Ex4 (3.0 MBq) intravenously and performed SPECT 30 min later, repeating this at a 2-wk interval. After the second scan, we harvested the pancreas and calculated BCM from immunohistochemically stained pancreatic sections. Specific accumulation of [111In]-Ex4 in ß cells was confirmed by autoradiography, with a significant correlation (r = 0.94) between the fluorescent and radioactive signal intensities. The radioactive signal from the pancreas in the second SPECT scan significantly correlated (r = 0.89) with BCM calculated from the immunostained pancreatic sections. We developed a regression formula to estimate BCM from the radioactive signals from the pancreas in SPECT scans. BCM can be quantified longitudinally and noninvasively by SPECT imaging with [111In]-Ex4. This technique successfully demonstrated longitudinal changes in BCM in NOD mice before and after onset of hyperglycemia.-Fujita, N., Fujimoto, H., Hamamatsu, K., Murakami, T., Kimura, H., Toyoda, K., Saji, H., Inagaki, N. Noninvasive longitudinal quantification of ß-cell mass with [111In]-labeled exendin-4.


Asunto(s)
Exenatida/farmacología , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Animales , Proteínas Fluorescentes Verdes/metabolismo , Ratones Transgénicos , Péptidos/metabolismo , Tomografía Computarizada de Emisión de Fotón Único/métodos
15.
Mol Biol Rep ; 46(2): 2273-2283, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30747384

RESUMEN

HFD animals were exposed to a low rate of different fractionated whole body gamma irradiation doses (0.5, 1 and 2 Gy, three fractions per week for two consecutive months) and the expression of certain genes involved in type 2 diabetes mellitus (T2DM) in livers and brains of HFD Wistar rats was investigated. Additionally, levels of diabetes-related proteins encoded by the studied genes were analyzed. Results indicated that mRNA level of incretin glucagon like peptite-1 receptor (GLP-1R) was augmented in livers and brains exposed to 1 and 2 Gy doses. Moreover, the mitochondrial uncoupling proteins 2 and 3 (UCP2/3) expressions in animals fed on HFD compared to those fed on normal chow diet were significantly increased at all applied doses. GLP-1R and UCP3 protein levels were up regulated in livers. Total protein content increased at 0.5 and 1 Gy gamma irradiation exposure and returned to its normal level at 2 Gy dose. Results could be an indicator of type 2 diabetes delayed development during irradiation exposure and support the importance of GLP-1R as a target gene in radiotherapy against T2DM and its chronic complications. A new hypothesis of brain-liver and intestine interface is speculated by which an increase in the hepatic GLP-1R is influenced by the effect of fractionated whole body gamma irradiation.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Rayos gamma/uso terapéutico , Animales , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Hígado/metabolismo , Masculino , ARN Mensajero/genética , Ratas , Ratas Wistar , Receptores de Glucagón/genética , Receptores de Glucagón/metabolismo , Proteína Desacopladora 2/efectos de los fármacos , Proteína Desacopladora 3/efectos de los fármacos
16.
Biomed Pharmacother ; 97: 1061-1065, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29136785

RESUMEN

BACKGROUD: In the early stage of diabetic retinopathy, the damage of retinal ganglion cells already exists, promoting the development of the disease. The aim of this study was to investigate the protective role and the mechanisms of obestatin against H2O2-induced damage in RGC-5 cells. METHODS: RGC-5 cells were incubated with various concentrations of obestatin for 24h before H2O2 added. The survival rates of RGC-5 were measured by MTT assay. The expression of apoptosis-related proteins and TrkB pathway-related proteins were detected by Western blot analysis. RESULTS: Our data showed that H2O2 evidently decreased the survival rate of RGC-5 cells. However, obestatin pretreatment reversed the decreased activity. Moreover, obestatin effectively increased the expression of Bcl-2 and decreased the expression of Bax. In addition, obestatin potentially plays a role in protecting RGC-5 by activating of TrkB. Obestatin notablely increased the phosphorylation of TrkB, AKT and ERK1/2. All these effects of obestatin can be inhibited by GLP-1R antagonist exendin (9-39). CONCLUSIONS: Obestatin prevents H2O2-induced damage in RGC-5 cells by activating TrkB pathway. Moreover, GLP-1R is closely related to the function of obestatin in RGC-5 cells.


Asunto(s)
Retinopatía Diabética/prevención & control , Ghrelina/farmacología , Receptor trkB/efectos de los fármacos , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Peróxido de Hidrógeno/toxicidad , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Receptor trkB/metabolismo , Células Ganglionares de la Retina/patología , Proteína X Asociada a bcl-2/genética
17.
Diabetes ; 66(4): 1062-1073, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28057699

RESUMEN

Glucagon-like peptide 1 (GLP-1) and serotonin play critical roles in energy balance regulation. Both systems are exploited clinically as antiobesity strategies. Surprisingly, whether they interact in order to regulate energy balance is poorly understood. Here we investigated mechanisms by which GLP-1 and serotonin interact at the level of the central nervous system. Serotonin depletion impaired the ability of exendin-4, a clinically used GLP-1 analog, to reduce body weight in rats, suggesting that serotonin is a critical mediator of the energy balance impact of GLP-1 receptor (GLP-1R) activation. Serotonin turnover and expression of 5-hydroxytryptamine (5-HT) 2A (5-HT2A) and 5-HT2C serotonin receptors in the hypothalamus were altered by GLP-1R activation. We demonstrate that the 5-HT2A, but surprisingly not the 5-HT2C, receptor is critical for weight loss, anorexia, and fat mass reduction induced by central GLP-1R activation. Importantly, central 5-HT2A receptors are also required for peripherally injected liraglutide to reduce feeding and weight. Dorsal raphe (DR) harbors cell bodies of serotonin-producing neurons that supply serotonin to the hypothalamic nuclei. We show that GLP-1R stimulation in DR is sufficient to induce hypophagia and increase the electrical activity of the DR serotonin neurons. Finally, our results disassociate brain metabolic and emotionality pathways impacted by GLP-1R activation. This study identifies serotonin as a new critical neural substrate for GLP-1 impact on energy homeostasis and expands the current map of brain areas impacted by GLP-1R activation.


Asunto(s)
Apetito/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Núcleo Dorsal del Rafe/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Hipoglucemiantes/farmacología , Receptor de Serotonina 5-HT2A/efectos de los fármacos , Receptor de Serotonina 5-HT2C/efectos de los fármacos , Serotonina/metabolismo , Aminopiridinas/farmacología , Animales , Anorexia , Exenatida , Conducta Alimentaria/efectos de los fármacos , Fenclonina/farmacología , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Indoles/farmacología , Liraglutida/farmacología , Masculino , Péptidos/farmacología , Pirrolidinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT2A/metabolismo , Receptor de Serotonina 5-HT2C/metabolismo , Antagonistas de la Serotonina/farmacología , Ponzoñas/farmacología , Pérdida de Peso/efectos de los fármacos
18.
Nutrients ; 8(10)2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27775601

RESUMEN

Food intake depends on homeostatic and non-homeostatic factors. In order to use grape seed proanthocyanidins (GSPE) as food intake limiting agents, it is important to define the key characteristics of their bioactivity within this complex function. We treated rats with acute and chronic treatments of GSPE at different doses to identify the importance of eating patterns and GSPE dose and the mechanistic aspects of GSPE. GSPE-induced food intake inhibition must be reproduced under non-stressful conditions and with a stable and synchronized feeding pattern. A minimum dose of around 350 mg GSPE/kg body weight (BW) is needed. GSPE components act by activating the Glucagon-like peptide-1 (GLP-1) receptor because their effect is blocked by Exendin 9-39. GSPE in turn acts on the hypothalamic center of food intake control probably because of increased GLP-1 production in the intestine. To conclude, GSPE inhibits food intake through GLP-1 signaling, but it needs to be dosed under optimal conditions to exert this effect.


Asunto(s)
Regulación del Apetito/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Extracto de Semillas de Uva/administración & dosificación , Proantocianidinas/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Femenino , Péptido 1 Similar al Glucagón/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Extracto de Semillas de Uva/farmacología , Mucosa Intestinal/metabolismo , Proantocianidinas/farmacología , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
19.
Cardiovasc Res ; 112(3): 669-676, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27702763

RESUMEN

AIMS: Although the nature of the humoral factor which mediates cardioprotection established by remote ischaemic conditioning (RIc) remains unknown, parasympathetic (vagal) mechanisms appear to play a critical role. As the production and release of many gut hormones is modulated by the vagus nerve, here we tested the hypothesis that RIc cardioprotection is mediated by the actions of glucagon-like peptide-1 (GLP-1). METHODS AND RESULTS: A rat model of myocardial infarction (coronary artery occlusion followed by reperfusion) was used. Remote ischaemic pre- (RIPre) or perconditioning (RIPer) was induced by 15 min occlusion of femoral arteries applied prior to or during the myocardial ischaemia. The degree of RIPre and RIPer cardioprotection was determined in conditions of cervical or subdiaphragmatic vagotomy, or following blockade of GLP-1 receptors (GLP-1R) using specific antagonist Exendin(9-39). Phosphorylation of PI3K/AKT and STAT3 was assessed. RIPre and RIPer reduced infarct size by ∼50%. In conditions of bilateral cervical or subdiaphragmatic vagotomy RIPer failed to establish cardioprotection. GLP-1R blockade abolished cardioprotection induced by either RIPre or RIPer. Exendin(9-39) also prevented RIPre-induced AKT phosphorylation. Cardioprotection induced by GLP-1R agonist Exendin-4 was preserved following cervical vagotomy, but was abolished in conditions of M3 muscarinic receptor blockade. CONCLUSIONS: These data strongly suggest that GLP-1 functions as a humoral factor of remote ischaemic conditioning cardioprotection. This phenomenon requires intact vagal innervation of the visceral organs and recruitment of GLP-1R-mediated signalling. Cardioprotection induced by GLP-1R activation is mediated by a mechanism involving M3 muscarinic receptors.


Asunto(s)
Arteria Femoral/cirugía , Péptido 1 Similar al Glucagón/metabolismo , Precondicionamiento Isquémico/métodos , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Nervio Vago/fisiopatología , Animales , Modelos Animales de Enfermedad , Exenatida , Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Antagonistas de Hormonas/farmacología , Ligadura , Masculino , Antagonistas Muscarínicos/farmacología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/patología , Fragmentos de Péptidos/farmacología , Péptidos/farmacología , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Receptor Muscarínico M3/efectos de los fármacos , Receptor Muscarínico M3/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Vagotomía , Nervio Vago/efectos de los fármacos , Nervio Vago/cirugía , Ponzoñas/farmacología
20.
Cardiovasc Diabetol ; 15: 65, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-27079193

RESUMEN

BACKGROUND: Glucagon-like peptide-1 (GLP-1) therapies are routinely used for glycaemic control in diabetes and their emerging cardiovascular actions have been a major recent research focus. In addition to GLP-1 receptor activation, the metabolically-inactive breakdown product, GLP-1(9-36)amide, also appears to exert notable cardiovascular effects, including protection against acute cardiac ischaemia. Here, we specifically studied the influence of GLP-1(9-36)amide on chronic post-myocardial infarction (MI) remodelling, which is a major driver of heart failure progression. METHODS: Adult female C57BL/6 J mice were subjected to permanent coronary artery ligation or sham surgery prior to continuous infusion with GLP-1(9-36)amide or vehicle control for 4 weeks. RESULTS: Infarct size was similar between groups with no effect of GLP-1(9-36)amide on MI-induced cardiac hypertrophy, although modest reduction of in vitro phenylephrine-induced H9c2 cardiomyoblast hypertrophy was observed. Whilst echocardiographic systolic dysfunction post-MI remained unchanged, diastolic dysfunction (decreased mitral valve E/A ratio, increased E wave deceleration rate) was improved by GLP-1(9-36)amide treatment. This was associated with modulation of genes related to extracellular matrix turnover (MMP-2, MMP-9, TIMP-2), although interstitial fibrosis and pro-fibrotic gene expression were unaltered by GLP-1(9-36)amide. Cardiac macrophage infiltration was also reduced by GLP-1(9-36)amide together with pro-inflammatory cytokine expression (IL-1ß, IL-6, MCP-1), whilst in vitro studies using RAW264.7 macrophages revealed global potentiation of basal pro-inflammatory and tissue protective cytokines (e.g. IL-1ß, TNF-α, IL-10, Fizz1) in the presence of GLP-1(9-36)amide versus exendin-4. CONCLUSIONS: These data suggest that GLP-1(9-36)amide confers selective protection against post-MI remodelling via preferential preservation of diastolic function, most likely due to modulation of infiltrating macrophages, indicating that this often overlooked GLP-1 breakdown product may exert significant actions in this setting which should be considered in the context of GLP-1 therapy in patients with cardiovascular disease.


Asunto(s)
Cardiotónicos/farmacología , Péptido 1 Similar al Glucagón/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Péptidos/uso terapéutico , Ponzoñas/uso terapéutico , Remodelación Ventricular/efectos de los fármacos , Animales , Exenatida , Femenino , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Función Ventricular Izquierda/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...