Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 277
Filtrar
1.
Neuropeptides ; 105: 102427, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38579490

RESUMEN

Obesity is a critical health condition worldwide that increases the risks of comorbid chronic diseases, but it can be managed with weight loss. However, conventional interventions relying on diet and exercise are inadequate for achieving and maintaining weight loss, thus there is significant market interest for pharmaceutical anti-obesity agents. For decades, receptor agonists for the gut peptide glucagon-like peptide 1 (GLP-1) featured prominently in anti-obesity medications by suppressing appetite and food reward to elicit rapid weight loss. As the neurocircuitry underlying food motivation overlaps with that for drugs of abuse, GLP-1 receptor agonism has also been shown to decrease substance use and relapse, thus its therapeutic potential may extend beyond weight management to treat addictions. However, as prolonged use of anti-obesity drugs may increase the risk of mood-related disorders like anxiety and depression, and individuals taking GLP-1-based medication commonly report feeling demotivated, the long-term safety of such drugs is an ongoing concern. Interestingly, current research now focuses on dual agonist approaches that include GLP-1 receptor agonism to enable synergistic effects on weight loss or associated functions. GLP-1 is secreted from the same intestinal cells as the anorectic gut peptide, Peptide YY3-36 (PYY3-36), thus this review assessed the therapeutic potential and underlying neural circuits targeted by PYY3-36 when administered independently or in combination with GLP-1 to curb the appetite for food or drugs of abuse like opiates, alcohol, and nicotine. Additionally, we also reviewed animal and human studies to assess the impact, if any, for GLP-1 and/or PYY3-36 on mood-related behaviors in relation to anxiety and depression. As dual agonists targeting GLP-1 and PYY3-36 may produce synergistic effects, they can be effective at lower doses and offer an alternative approach for therapeutic benefits while mitigating undesirable side effects.


Asunto(s)
Péptido 1 Similar al Glucagón , Péptido YY , Humanos , Animales , Péptido YY/metabolismo , Péptido YY/farmacología , Péptido 1 Similar al Glucagón/metabolismo , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Fragmentos de Péptidos/farmacología , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo
2.
J Physiol ; 601(16): 3461-3480, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37269207

RESUMEN

An understanding of the metabolic determinants of postexercise appetite regulation would facilitate development of adjunctive therapeutics to suppress compensatory eating behaviours and improve the efficacy of exercise as a weight-loss treatment. Metabolic responses to acute exercise are, however, dependent on pre-exercise nutritional practices, including carbohydrate intake. We therefore aimed to determine the interactive effects of dietary carbohydrate and exercise on plasma hormonal and metabolite responses and explore mediators of exercise-induced changes in appetite regulation across nutritional states. In this randomized crossover study, participants completed four 120 min visits: (i) control (water) followed by rest; (ii) control followed by exercise (30 min at ∼75% of maximal oxygen uptake); (iii) carbohydrate (75 g maltodextrin) followed by rest; and (iv) carbohydrate followed by exercise. An ad libitum meal was provided at the end of each 120 min visit, with blood sample collection and appetite assessment performed at predefined intervals. We found that dietary carbohydrate and exercise exerted independent effects on the hormones glucagon-like peptide 1 (carbohydrate, 16.8 pmol/L; exercise, 7.4 pmol/L), ghrelin (carbohydrate, -48.8 pmol/L; exercise: -22.7 pmol/L) and glucagon (carbohydrate, 9.8 ng/L; exercise, 8.2 ng/L) that were linked to the generation of distinct plasma 1 H nuclear magnetic resonance metabolic phenotypes. These metabolic responses were associated with changes in appetite and energy intake, and plasma acetate and succinate were subsequently identified as potential novel mediators of exercise-induced appetite and energy intake responses. In summary, dietary carbohydrate and exercise independently influence gastrointestinal hormones associated with appetite regulation. Future work is warranted to probe the mechanistic importance of plasma acetate and succinate in postexercise appetite regulation. KEY POINTS: Carbohydrate and exercise independently influence key appetite-regulating hormones. Temporal changes in postexercise appetite are linked to acetate, lactate and peptide YY. Postexercise energy intake is associated with glucagon-like peptide 1 and succinate levels.


Asunto(s)
Regulación del Apetito , Carbohidratos de la Dieta , Masculino , Apetito/fisiología , Regulación del Apetito/fisiología , Estudios Cruzados , Ingestión de Energía/fisiología , Ejercicio Físico/fisiología , Ghrelina/metabolismo , Ghrelina/farmacología , Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Insulina/farmacología , Péptido YY/metabolismo , Péptido YY/farmacología , Succinatos/farmacología , Humanos
3.
Pharm Res ; 38(8): 1369-1385, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34272643

RESUMEN

PURPOSE: The anorectic effect of PYY3-36 makes it a potential pharmacological weight loss treatment. Modifications of the endogenous peptide to obtain commercially attractive pharmacological and biophysical stability properties are examined. METHODS: Half-life extended PYY3-36 analogues were prepared and examined regarding Y2-receptor potency as well as biophysical and stability properties. RESULTS: Deamidation of asparagine in position 18 and 29 was observed upon incubation at 37°C. Asparagine in position 18 - but not position 29 - could be substituted to glutamine without detrimental effects on Y2-receptor potency. Covalent dimers were formed via the phenol impurity benzoquinone reacting with two N-terminal residues (Isoleucine-Lysine). Both residues had to be modified to suppress dimerization, which could be done without negatively affecting Y2-receptor potency or other stability/biophysical properties. Introduction of half-life extending modifications in position 30 and 35 eliminated aggregation at 37°C without negatively affecting other stability properties. Placement of a protracting moiety (fatty acid) in the receptor-binding C-terminal region reduced Y2-receptor potency substantially, whereas only minor effects of protractor position were observed on structural, biophysical or stability properties. Lipidated PYY3-36 analogues formed oligomers of various sizes depending on primary structure and solution conditions. CONCLUSIONS: By rational design, a chemically and physically stable Y2-receptor selective, half-life extended PYY3-36 peptide has been developed.


Asunto(s)
Fragmentos de Péptidos/química , Péptido YY/química , Receptores de Neuropéptido Y/agonistas , Asparagina/química , Desarrollo de Medicamentos , Células HEK293 , Humanos , Fragmentos de Péptidos/farmacología , Péptido YY/farmacología
4.
Angew Chem Int Ed Engl ; 60(15): 8268-8275, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33448563

RESUMEN

The two gut hormones GLP-1 and PYY3-36 , which are both secreted from the L-cells upon food stimuli, have a stronger inhibitory effect on food intake when they are combined, compared to their individual effects as single agonists. Although they are not homologous and share no sequence similarity, we show that a GLP-1 analogue can be designed to exhibit potent activity on both the Y2 and GLP-1 receptors. Dual acting hybrid analogues were realized by designing truncated and potent Y2 receptor PYY analogues, followed by integrating the critical residues into GLP-1. In this study, we show that one of these dual acting agonists acutely reduces food intake significantly more than the respective mono-agonist counterparts.


Asunto(s)
Diseño de Fármacos , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón/agonistas , Fragmentos de Péptidos/farmacología , Péptido YY/farmacología , Receptores de la Hormona Gastrointestinal/agonistas , Cristalografía por Rayos X , Péptido 1 Similar al Glucagón/síntesis química , Péptido 1 Similar al Glucagón/química , Humanos , Modelos Moleculares , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/química , Péptido YY/síntesis química , Péptido YY/química
5.
Nutrients ; 14(1)2021 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-35010991

RESUMEN

BACKGROUND: The hypothalamus is an important brain region for the regulation of energy balance. Roux-en-Y gastric bypass (RYGB) surgery and gut hormone-based treatments are known to reduce body weight, but their effects on hypothalamic gene expression and signaling pathways are poorly studied. METHODS: Diet-induced obese male Wistar rats were randomized into the following groups: RYGB, sham operation, sham + body weight-matched (BWM) to the RYGB group, osmotic minipump delivering PYY3-36 (0.1 mg/kg/day), liraglutide s.c. (0.4 mg/kg/day), PYY3-36 + liraglutide, and saline. All groups (except BWM) were kept on a free choice of high- and low-fat diets. Four weeks after interventions, hypothalami were collected for RNA sequencing. RESULTS: While rats in the RYGB, BWM, and PYY3-36 + liraglutide groups had comparable reductions in body weight, only RYGB and BWM treatment had a major impact on hypothalamic gene expression. In these groups, hypothalamic leptin receptor expression as well as the JAK-STAT, PI3K-Akt, and AMPK signaling pathways were upregulated. No significant changes could be detected in PYY3-36 + liraglutide-, liraglutide-, and PYY-treated groups. CONCLUSIONS: Despite causing similar body weight changes compared to RYGB and BWM, PYY3-36 + liraglutide treatment does not impact hypothalamic gene expression. Whether this striking difference is favorable or unfavorable to metabolic health in the long term requires further investigation.


Asunto(s)
Hormonas Gastrointestinales/farmacología , Hipotálamo/metabolismo , Liraglutida/farmacología , Fragmentos de Péptidos/farmacología , Péptido YY/farmacología , Transcriptoma/efectos de los fármacos , Animales , Peso Corporal , Restricción Calórica , Modelos Animales de Enfermedad , Metabolismo Energético , Derivación Gástrica , Expresión Génica/efectos de los fármacos , Masculino , Obesidad , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
6.
J Med Chem ; 63(17): 9660-9671, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32844654

RESUMEN

Bariatric surgery results in increased intestinal secretion of hormones GLP-1 and anorexigenic PYY, which is believed to contribute to the clinical efficacy associated with the procedure. This observation raises the question whether combination treatment with gut hormone analogs might recapitulate the efficacy and mitigate the significant risks associated with surgery. Despite PYY demonstrating excellent efficacy and safety profiles with regard to food intake reduction, weight loss, and glucose control in preclinical animal models, PYY-based therapeutic development remains challenging given a low serum stability and half-life for the native peptide. Here, combined peptide stapling and PEG-fatty acid conjugation affords potent PYY analogs with >14 h rat half-lives, which are expected to translate into a human half-life suitable for once-weekly dosing. Excellent efficacy in glucose control, food intake reduction, and weight loss for lead candidate 22 in combination with our previously reported long-acting GLP-1 analog is demonstrated in a diet-induced obesity mouse model.


Asunto(s)
Ingeniería , Receptor del Péptido 1 Similar al Glucagón/agonistas , Obesidad/tratamiento farmacológico , Péptido YY/química , Péptido YY/farmacología , Receptores de Neuropéptido Y/agonistas , Secuencia de Aminoácidos , Animales , Peso Corporal/efectos de los fármacos , Interacciones Farmacológicas , Ingestión de Alimentos/efectos de los fármacos , Semivida , Modelos Moleculares , Péptido YY/farmacocinética , Polietilenglicoles/química , Conformación Proteica , Ratas
7.
Hormones (Athens) ; 19(4): 549-558, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32572709

RESUMEN

PURPOSE: The daily circadian cycle is known to modulate both feeding behavior and metabolism. As such, the timing of food consumption can play a role in regulating overall health. The purpose of this study is to determine whether fasting at different times of the day alters subsequent food consumption and levels of PYY3-36, a hormone secreted after a meal which inhibits appetite. METHODS: Separate groups of mice were fasted at different times of the day: (1) start of the day, (2) middle of the day, (3) start of the night, and (4) middle of the night, and either injected with vehicle or PYY3-36 to assess their subsequent food consumption patterns, PYY3-36 levels, and glucose and insulin levels. We also investigated whether light exposure during the night would alter food consumption and PYY3-36 levels after fasting. RESULTS: Mice fasted during the start of the daytime exhibited increased food consumption post-fast compared to mice fasted during the night. Injections of PYY3-36 during the night were more effective in reducing food consumption compared to PYY3-36 administration during the day. Constant light exposure suppressed food consumption after fasting and increased fasting PYY3-36 levels. CONCLUSIONS: These results indicate that mice exhibit distinct food consumption patterns after being presented with a fast at different times of the day. Light exposure also modulates both food consumption after a fast and levels of PYY3-36.


Asunto(s)
Ritmo Circadiano/fisiología , Ayuno/fisiología , Conducta Alimentaria/fisiología , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Péptido YY/metabolismo , Péptido YY/farmacología , Fotoperiodo , Animales , Ayuno/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/administración & dosificación , Péptido YY/administración & dosificación
8.
Front Endocrinol (Lausanne) ; 11: 598843, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33551994

RESUMEN

Background: Combination therapies of anorectic gut hormones partially mimic the beneficial effects of bariatric surgery. Thus far, the effects of a combined chronic systemic administration of Glucagon-like peptide-1 (GLP-1) and peptide tyrosine tyrosine 3-36 (PYY3-36) have not been directly compared to Roux-en-Y gastric bypass (RYGB) in a standardized experimental setting. Methods: High-fat diet (HFD)-induced obese male Wistar rats were randomized into six treatment groups: (1) RYGB, (2) sham-operation (shams), (3) liraglutide, (4) PYY3-36, (5) PYY3-36+liraglutide (6), saline. Animals were kept on a free choice high- and low-fat diet. Food intake, preference, and body weight were measured daily for 4 weeks. Open field (OP) and elevated plus maze (EPM) tests were performed. Results: RYGB reduced food intake and achieved sustained weight loss. Combined PYY3-36+liraglutide treatment led to similar and plateaued weight loss compared to RYGB. Combined PYY3-36+liraglutide treatment was superior to PYY3-36 (p ≤ 0.0001) and liraglutide (p ≤ 0.05 or p ≤ 0.01) mono-therapy. PYY3-36+liraglutide treatment and RYGB also reduced overall food intake and (less pronounced) high-fat preference compared to controls. The animals showed no signs of abnormal behavior in OF or EPM. Conclusions: Liraglutide and PYY3-36 combination therapy vastly mimics reduced food intake, food choice and weight reducing benefits of RYGB.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos , Derivación Gástrica/métodos , Liraglutida/farmacología , Obesidad/terapia , Péptido YY/farmacología , Animales , Peso Corporal , Terapia Combinada , Preferencias Alimentarias , Hipoglucemiantes/farmacología , Masculino , Obesidad/etiología , Obesidad/patología , Ratas , Ratas Wistar
9.
J Pharmacol Exp Ther ; 372(1): 73-82, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31771994

RESUMEN

Itch stimuli are detected by specialized primary afferents that convey the signal to the spinal cord, but how itch transmission is regulated is still not completely known. Here, we investigated the roles of the neuropeptide Y (NPY)/Y2 receptor system on scratch behavior. The inhibitory Y2 receptor is expressed on mouse primary afferents, and intrathecal administration of the Y2 agonist peptide YY (PYY)3-36 reduced scratch episode frequency and duration induced by compound 48/80, an effect that could be reversed by intrathecal preadministration of the Y2 antagonist BIIE0246. Also, scratch episode duration induced by histamine could be reduced by PYY3-36 In contrast, scratch behavior induced by α-methyl-5HT, protease-activated receptor-2-activating peptide SLIGRL, chloroquine, topical dust mite extract, or mechanical itch induced by von Frey filaments was unaffected by stimulation of Y2 Primary afferent neurons expressing the Npy2r gene were found to coexpress itch-associated markers such as natriuretic peptide precursor b, oncostatin M receptor, and interleukin (IL) 31 receptor A. Accordingly, intrathecal PYY3-36 reduced the scratch behavior induced by IL-31. Our findings imply that the NPY/Y2 system reduces histaminergic and IL-31-associated itch through presynaptic inhibition of a subpopulation of itch-associated primary afferents. SIGNIFICANCE STATEMENT: The spinal neuropeptide Y system dampens scratching behavior induced by histaminergic compounds and interleukin 31, a cytokine involved in atopic dermatitis, through interactions with the Y2 receptor. The Y2 receptor is expressed by primary afferent neurons that are rich in itch-associated neurotransmitters and receptors such as somatostatin, natriuretic peptide precursor b, and interleukin 31 receptors.


Asunto(s)
Antipruriginosos/farmacología , Dermatitis Atópica/metabolismo , Neuronas Aferentes/metabolismo , Fragmentos de Péptidos/farmacología , Péptido YY/farmacología , Prurito/metabolismo , Receptores de Neuropéptido Y/metabolismo , Animales , Antipruriginosos/administración & dosificación , Antipruriginosos/uso terapéutico , Arginina/análogos & derivados , Arginina/toxicidad , Benzazepinas/toxicidad , Células Cultivadas , Cloroquina/farmacología , Dermatitis Atópica/tratamiento farmacológico , Ganglios Espinales/citología , Histamina/farmacología , Histamina/toxicidad , Interleucinas/farmacología , Interleucinas/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Péptido Natriurético Encefálico/genética , Péptido Natriurético Encefálico/metabolismo , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/fisiología , Oligopéptidos/farmacología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/uso terapéutico , Péptido YY/administración & dosificación , Péptido YY/uso terapéutico , Prurito/tratamiento farmacológico , Prurito/etiología , Receptores de Neuropéptido Y/genética , Receptores de Oncostatina M/genética , Receptores de Oncostatina M/metabolismo , Serotonina/farmacología
10.
Obes Surg ; 30(2): 697-706, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31701411

RESUMEN

AIMS: Roux-en-Y gastric bypass (RYGB) is one of the most effective surgical therapies for the rapid resolution of type 2 diabetes. However, the mechanisms underlying the entero-hormonal response after surgery and the role of peptide tyrosine tyrosine (PYY) in the restoration of normoglycemia are still not clear. METHODS: We reproduced the RYGB technique in Wistar and Goto-Kakizaki rats and performed serum hormonal, histological, and hormonal-infusion test. RESULTS: Using the diabetic Goto-Kakizaki (GK) rat model, we demonstrated that PYY plasma levels showed a remarkable peak approximately 30 min earlier than GLP-1 or GIP after mixed-meal administration in RYGB-operated rats with PYY. The GLP-1 and GIP areas under the curve (AUCs) increased after RYGB in GK rats. Additionally, the findings suggested that PYY (3-36) infusion led to increased GLP-1 and GIP plasma levels close to those obtained after a meal. Finally, the number of GLP-1-positive cells appeared to increase in the three segments of the small intestine in GK-RYGB-operated rats beyond the early presence of nutrient stimulation in the ileum. Nevertheless, PYY-positive cell numbers appeared to increase only in the ileum. CONCLUSION: At least in rats, these data demonstrate an earlier essential role for PYY in gut hormone regulation after RYGB. We understand that PYY contributes to GLP-1 and GIP release and there must be the existence of enteroendocrine communication routes between the distal and proximal small intestine.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/cirugía , Derivación Gástrica , Péptido YY/fisiología , Animales , Glucemia/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Hormonas Gastrointestinales/sangre , Hormonas Gastrointestinales/metabolismo , Prueba de Tolerancia a la Glucosa , Control Glucémico , Secreción de Insulina/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Intestinos/efectos de los fármacos , Intestinos/patología , Masculino , Péptido YY/sangre , Péptido YY/farmacología , Ratas , Ratas Wistar , Aumento de Peso/efectos de los fármacos
11.
J Clin Endocrinol Metab ; 105(3)2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31628465

RESUMEN

CONTEXT: Central and peripheral administration of peptide YY (PYY) has potent anorectic effects, and PYY analogs are under development as antiobesity treatments. Recent animal data suggest PYY may also influence the reproductive axis; however the effects of PYY on the human reproductive system are unknown. OBJECTIVE: To investigate the effects of PYY administration on the reproductive axis in healthy young men. DESIGN: Single-blind, randomized, placebo-controlled crossover study. SETTING: Clinical Research Facility, Imperial College Healthcare NHS Trust. PARTICIPANTS: Eighteen healthy eugonadal men (mean age 24.1 ± 0.9 years, mean body mass index 22.2 ± 0.4 kg/m2). INTERVENTION: Eight-hour intravenous infusion of 0.4 pmol/kg/min PYY3-36 or rate-matched vehicle infusion. MAIN OUTCOME MEASURES: Number of luteinizing hormone (LH) pulses, LH, follicle stimulating hormone (FSH), and testosterone levels. RESULTS: The number of LH pulses (mean number of LH pulses/8 hours: PYY 4.4 ± 0.3 vs vehicle 4.4 ± 0.4, P > .99), LH area under the curve (AUC) (PYY 1503 ± 79 IU.min/L vs vehicle 1574 ± 86 IU.min/L, P = .36), FSH AUC (PYY 1158 ± 513 IU.min/L vs vehicle 1199 ± 476 IU.min/L, P = .49) and testosterone AUC (PYY 10 485 ± 684 IU.min/L vs vehicle 11 133 ± 803 IU.min/L, P = .24) were similar during PYY and vehicle infusions. CONCLUSIONS: Acute intravenous infusion of 0.4 pmol/kg/min PYY does not affect the reproductive axis in healthy men.


Asunto(s)
Biomarcadores/sangre , Hormona Folículo Estimulante/sangre , Sistema Hipotálamo-Hipofisario/metabolismo , Hormona Luteinizante/sangre , Péptido YY/farmacología , Testosterona/sangre , Adolescente , Adulto , Estudios Cruzados , Estudios de Seguimiento , Voluntarios Sanos , Humanos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Masculino , Pronóstico , Método Simple Ciego , Adulto Joven
12.
PLoS Biol ; 17(12): e3000482, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31805040

RESUMEN

Better understanding of feeding behaviour will be vital in reducing obesity and metabolic syndrome, but we lack a standard model that captures the complexity of feeding behaviour. We construct an accurate stochastic model of rodent feeding at the bout level in order to perform quantitative behavioural analysis. Analysing the different effects on feeding behaviour of peptide YY3-36 (PYY3-36), lithium chloride, glucagon-like peptide 1 (GLP-1), and leptin shows the precise behavioural changes caused by each anorectic agent. Our analysis demonstrates that the changes in feeding behaviour evoked by the anorectic agents investigated do not mimic the behaviour of well-fed animals and that the intermeal interval is influenced by fullness. We show how robust homeostatic control of feeding thwarts attempts to reduce food intake and how this might be overcome. In silico experiments suggest that introducing a minimum intermeal interval or modulating upper gut emptying can be as effective as anorectic drug administration.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología , Animales , Depresores del Apetito/farmacología , Ingestión de Alimentos/fisiología , Péptido 1 Similar al Glucagón/farmacología , Homeostasis/efectos de los fármacos , Leptina/farmacología , Masculino , Ratones , Obesidad , Fragmentos de Péptidos/farmacología , Péptido YY/farmacología , Ratas
13.
Endocr Regul ; 53(1): 26-33, 2019 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-31517617

RESUMEN

OBJECTIVE: The aim of the present study was to assess the effect of the PYY3-36, as a potential therapy for the type 2 diabetes mellitus (T2DM), induced by high fat diet (HFD) and an intraperitoneal (i.p.) administration of streptozotocin (STZ) in albino rats. METHODS: Forty adult male albino Wistar rats were divided into: 1) control group (C, in which the rats were fed with a standard diet and received vehicle; 2) diabetic group (D, in which T2DM was induced by feeding the rats with HFD for four weeks followed by a single i.p. injection of 35 mg/kg STZ, this group was also allowed to have HFD till the end of the study; and 3) D+PYY3-36 group (in which the diabetic rats were treated with 50 µg/kg i.p. PYY3-36 twice a day for one week). Food intake, water intake, body weight (b.w.), visceral fat weight (VFW), liver glycogen content, serum levels of glucose, insulin, and interleukin-6 (IL-6), were measured. Homeostatic-model assessment of insulin resistance (HOMA-IR) was estimated. The gene expression of the hypothalamic neuropeptide Y (NPY) and visceral nuclear factor kappa B (NF-κB) were assessed by a reverse transcription polymerase chain reaction (RT-PCR). RESULTS: The PYY3-36 administration to the diabetic group of rats significantly increased the serum insulin levels and liver glycogen content, decreased the body weight, VFW, food intake, water intake, serum levels of the glucose, IL-6, and HOMA-IR. It also decreased the expression of both the hypothalamic NPY and the visceral fat NF-κB. CONCLUSION: With respect to the fact of improved insulin release and enhanced insulin sensitivity (an effect that may be mediated via suppressing accumulation of visceral fat and inflammatory markers), in the rats treated with PYY3-36, the PYY3-36 might be considered for the future as a promising therapeutic tool in T2DM.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Resistencia a la Insulina , Fragmentos de Péptidos/farmacología , Péptido YY/farmacología , Adiposidad/efectos de los fármacos , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/patología , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Resistencia a la Insulina/fisiología , Grasa Intraabdominal/efectos de los fármacos , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/patología , Masculino , Tamaño de los Órganos/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de Neuropéptido Y/agonistas , Estreptozocina
14.
PLoS Negl Trop Dis ; 13(8): e0007573, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31408466

RESUMEN

The metacercariae of the Clonorchis sinensis liver fluke excyst in the duodenum of mammalian hosts, and the newly excysted juveniles (CsNEJs) migrate along the bile duct via bile chemotaxis. Cholic acid is a major component of bile that induces this migration. We investigated the neuronal control of chemotactic behavior of CsNEJs toward cholic acid. The migration of CsNEJs was strongly inhibited at sub-micromolar concentration by dopamine D1 (LE-300 and SKF-83566), D2 (spiramide, nemonapride, and sulpiride), and D3 (GR-103691 and NGB-2904) receptor antagonists, as well as a dopamine reuptake inhibitor (BTCP). Neuropeptides, FMRFamide, peptide YY, and neuropeptide Y were also potent inhibitors of chemotaxis. Meanwhile, serotonergic, glutamatergic, and cholinergic inhibitors did not affect chemotaxis, with the exception of fluoxetine and CNQX. Confocal immunofluorescence analysis indicated that dopaminergic and cholinergic neurons were colocalized in the somatic muscle tissues of adult C. sinensis. Our findings suggest that dopaminergic neurons and neuropeptides play a major role in the chemotactic migration of CsNEJs to bile, and their inhibitors or modulators could be utilized to prevent their migration from the bile duct.


Asunto(s)
Quimiotaxis/efectos de los fármacos , Quimiotaxis/fisiología , Clonorchis sinensis/efectos de los fármacos , Clonorchis sinensis/fisiología , Fasciola hepatica/efectos de los fármacos , Neurotransmisores/farmacología , Animales , Benzamidas/farmacología , Compuestos de Bifenilo/farmacología , Ácido Cólico , Dopamina/farmacología , Inhibidores de Captación de Dopamina/farmacología , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , FMRFamida/farmacología , Fluorenos/farmacología , Neuropéptido Y/farmacología , Péptido YY/farmacología , Piperazinas/farmacología , Serotoninérgicos/farmacología , Compuestos de Espiro/farmacología , Sulpirida/farmacología
15.
Mol Pharm ; 16(8): 3665-3677, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31310716

RESUMEN

Peptide YY3-36 (PYY3-36) is an endogenous ligand of the neuropeptide Y2 receptor (Y2R), on which it acts to reduce food intake. Chemically modified PYY3-36 analogues with extended half-lives are potential therapeutics for the treatment of obesity. Here we show that the common half-life extending strategies PEGylation and lipidation not only control PYY3-36's pharmacokinetics but also affect central aspects of its pharmacodynamics. PEGylation of PYY3-36 inhibited endocytosis by increasing receptor dissociation rates (koff), which reduced arrestin-3 (Arr3) activity. This is the first link between Arr3 recruitment and Y2R residence time. C16-lipidation of PYY3-36 had a negligible impact on Y2R signaling, binding, and endocytosis. In contrast, C18acid-lipidation minimized endocytosis, which indicated a decreased internalization through non-arrestin-related mechanisms. We propose a temporal model that connects the properties and position of the half-life extender with receptor Gi versus Arr3 signaling bias. We believe that this will be important for future design of peptide therapeutics.


Asunto(s)
Fármacos Antiobesidad/farmacología , Diseño de Fármacos , Fragmentos de Péptidos/farmacología , Péptido YY/farmacología , Receptores de Neuropéptido Y/metabolismo , Fármacos Antiobesidad/química , Fármacos Antiobesidad/uso terapéutico , Arrestinas/metabolismo , Células HEK293 , Semivida , Humanos , Microscopía Intravital , Lípidos/química , Liposomas , Modelos Biológicos , Modelos Químicos , Estructura Molecular , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/uso terapéutico , Péptido YY/química , Péptido YY/uso terapéutico , Polietilenglicoles/química , Relación Estructura-Actividad
16.
Drug Dev Ind Pharm ; 45(7): 1101-1110, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31039626

RESUMEN

Peptide YY(3-36) (PYY(3-36)) is an endogenous appetite suppressing peptide. The present research was to perform pharmacokinetic/pharmacodynamic (PK/PD) analysis for predicting the concentration- and response-time profiles of PYY(3-36) after systemic and pulmonary delivery in mice, with the goal of suggesting a potential pulmonary dosing regimen in humans. A PK/PD model was developed to describe PYY(3-36) plasma concentration - and relative food intake rate ratio (as % of control) - time profiles after intraperitoneal and subcutaneous administration, and inhalation in mice. The absorption of inhaled PYY(3-36) from the lungs of mice could only be described with a combined slow (absorption rate of 0.147 L/h) and fast (absorption rate of 104.4 L/h) absorption process, presumably related to absorption from the central and peripheral regions of the lungs. The estimates for IC50 and Imax were 6.8 ng/mL and 63.5%, respectively, based on inhibitory Emax model. The PK parameters, such as clearance (CL), volume of distribution at steady state (Vdss), and the absorption rates (ka), were then scaled to human's. The scaled human CL and Vdss for obese subjects were 24.8 L/h and 9.0 L, respectively. The model predicted human plasma PYY(3-36) concentrations agreed reasonably well with placebo-normalized plasma PYY(3-36) concentrations after short-term infusion and SC injection in literature. An inhalation dose of PYY(3-36) of about 100 µg was proposed for obese subjects based on simulations. This PK/PD analysis satisfactorily described PYY(3-36) concentration-time and relative food intake rate ratio- time profiles at all doses and routes. The developed model might facilitate the inhalation dose selection of PYY(3-36).


Asunto(s)
Mucosa Gástrica/metabolismo , Pulmón/metabolismo , Péptido YY/farmacología , Péptido YY/farmacocinética , Administración por Inhalación , Animales , Apetito/efectos de los fármacos , Ingestión de Alimentos , Humanos , Ratones , Ratones Endogámicos C57BL , Obesidad/tratamiento farmacológico , Estómago
17.
Endocrinology ; 160(11): 2737-2747, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31074796

RESUMEN

To study the effects of an analog of the gut-produced hormone peptide YY (PYY3-36), which has increased selectivity for the Y2 receptor; specifically, to record its effects on food intake and on hypothalamic neuropeptide Y/agouti-related peptide (NPY/AgRP) neuron activity. NNC0165-1273, a modified form of the peptide hormone PYY3-36 with potent selectivity at Y2 receptor (>5000-fold over Y1, 1250-fold over Y4, and 650-fold over Y5 receptor), was tested in vivo and in vitro in mouse models. NNC0165-1273 has fivefold lower relative affinity for Y2 compared with PYY3-36, but >250-, 192-, and 400-fold higher selectivity, respectively, for the Y1, Y4, and Y5 receptors. NNC0165-1273 produced a reduction in nighttime feeding at a dose at which PYY3-36 loses efficacy. The normal behavioral satiety sequence observed suggests that NNC0165-1273 is not nauseating and, instead, reduces food intake by producing early satiety. Additionally, NNC0165-1273 blocked ghrelin-induced cFos expression in NPY/AgRP neurons. In vitro electrophysiological recordings showed that, opposite to ghrelin, NNC0165-1273 hyperpolarized NPY/AgRP neurons and reduced action potential frequency. Administration of NNC0165-1273 via subcutaneous osmotic minipump caused a dose-dependent decrease in body weight and fat mass in an obese mouse model. Finally, NNC0165-1273 attenuated the feeding response when NPY/AgRP neurons were activated using ghrelin or more selectively with designer receptors. NNC0165-1273 is nonnauseating and stimulates a satiety response through, at least in part, a direct action on hypothalamic NPY/AgRP neurons. Modification of PYY3-36 to produce compounds with increased affinity to Y2 receptors may be useful as antiobesity therapies in humans.


Asunto(s)
Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Fragmentos de Péptidos/química , Péptido YY/química , Receptores de Neuropéptido Y/agonistas , Respuesta de Saciedad/efectos de los fármacos , Animales , Núcleo Arqueado del Hipotálamo/citología , Ghrelina , Masculino , Ratones , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Péptido YY/farmacología
18.
Am J Physiol Regul Integr Comp Physiol ; 316(4): R406-R416, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30726118

RESUMEN

Neuropeptide Y (NPY), peptide YY (PYY), and their cognate receptors (YR) are expressed by subpopulations of central and peripheral nervous system neurons. Intracerebroventricular injections of NPY or PYY increase food intake, and intrahypothalamic NPY1 or NPY5 receptor agonist injections also increase food intake. In contrast, injection of PYY in the periphery reduces food intake, apparently by activating peripheral Y2R. The dorsal vagal complex (DVC) of the hindbrain is the site where vagal afferents relay gut satiation signals to the brain. While contributions of the DVC are increasingly investigated, a role for DVC YR in control of food intake has not been examined systematically. We used in situ hybridization to confirm expression of Y1R and Y2R, but not Y5R, in the DVC and vagal afferent neurons. We found that nanoinjections of a Y2R agonist, PYY-(3-36), into the DVC significantly increased food intake over a 4-h period in satiated male rats. PYY-(3-36)-evoked food intake was prevented by injection of a selective Y2R antagonist. Injection of a Y1R/Y5R-preferring agonist into the DVC failed to increase food intake at doses reported to increase food intake following hypothalamic injection. Finally, injection of PYY-(3-36) into the DVC prevented reduction of 30-min food intake following intraperitoneal injection of cholecystokinin (CCK). Our results indicate that activation of DVC Y2R, unlike hypothalamic or peripheral Y2R, increases food intake. Furthermore, in the context of available electrophysiological observations, our results are consistent with the hypothesis that DVC Y2R control food intake by dampening vagally mediated satiation signals in the DVC.


Asunto(s)
Colecistoquinina/farmacología , Ingestión de Alimentos/efectos de los fármacos , Receptores de Neuropéptido Y/agonistas , Saciedad/efectos de los fármacos , Nervio Vago/efectos de los fármacos , Animales , Inyecciones , Masculino , Péptido YY/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Neuropéptido Y/antagonistas & inhibidores , Receptores de Neuropéptido Y/efectos de los fármacos
19.
Cell Metab ; 29(4): 837-843.e5, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30773465

RESUMEN

The gut hormone PYY3-36 reduces food intake in humans and exhibits at least additive efficacy in combination with GLP-1. However, the utility of PYY analogs as anti-obesity agents has been severely limited by emesis and rapid proteolysis, a profile similarly observed with native PYY3-36 in obese rhesus macaques. Here, we found that antibody conjugation of a cyclized PYY3-36 analog achieved high NPY2R selectivity, unprecedented in vivo stability, and gradual infusion-like exposure. These properties permitted profound reduction of food intake when administered to macaques for 23 days without a single emetic event in any animal. Co-administration with the GLP-1 receptor agonist liraglutide for an additional 5 days further reduced food intake with only one animal experiencing a single bout of emesis. This antibody-conjugated PYY analog therefore may enable the long-sought potential of GLP-1/PYY-based combination treatment to achieve robust, well-tolerated weight reduction in obese patients.


Asunto(s)
Anorexia/inducido químicamente , Péptido YY/química , Péptido YY/farmacología , Vómitos , Animales , Células CHO , Cricetulus , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Células HEK293 , Humanos , Liraglutida/farmacología , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Péptido YY/administración & dosificación , Vómitos/inducido químicamente
20.
Neuropeptides ; 73: 89-95, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30471778

RESUMEN

Peptide YY(3-36) ((PYY(3-36)) and glucagon like peptide 1 (GLP-1) in combination reduce food intake and body weight in an additive or synergistic manner in animal models and in humans. Nevertheless, the mechanisms behind are not completely understood. The present study aims to investigate the effect of combining PYY(3-36) and the GLP-1 receptor agonist exendin-4 (Ex4) by examining acute food intake and global neuronal activation as measured by c-fos in C57BL/6 J mice. An additive reduction in food intake was found 1.5 h after s.c dosing with the combination of PYY(3-36) (200 µg/kg) and Ex4 (2.5 µg/kg). This was associated with a synergistic enhancement of c-fos reactivity in central amygdalar nucleus (CeA), rostral part of the mediobasal arcuate nucleus (ARH), supratrigeminal nucleus (SUT), lateral parabrachial nucleus (PB), area postrema (AP) and nucleus tractus solitarius (NTS) compared to vehicle, PYY(3-36) and Ex4 individually dosed mice. The regions activated by Ex4 individually and PYY(3-36) and Ex4 in combination resembled each other, but the combination group had a significantly stronger c-fos response. Twenty-five brain areas were activated by PYY(3-36) and Ex4 in combination compared to vehicle versus nine brain areas by Ex4 individually. No significant increase in c-fos reactivity was found by PYY(3-36) compared to vehicle dosed mice. The neuronal activation of ARH and the AP/NTS to PB to CeA pathway is important for appetite regulation while SUT has not previously been reported in the regulation of energy balance. As PYY(3-36) and Ex4 act on different neurons leading to recruitment of different signalling pathways within and to the brain, an interaction of these pathways may contribute to their additive/synergistic action. Thus, PYY(3-36) boosts the effect of Ex4 possibly by inducing less inhibition of neuronal activity leading to an enhanced neuronal activity induced by Ex4.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Exenatida/farmacología , Hipotálamo/efectos de los fármacos , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Péptido YY/farmacología , Amígdala del Cerebelo/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Hipotálamo/metabolismo , Ratones , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...