Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Physiol Behav ; 283: 114601, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38838800

RESUMEN

AIM: The hypothesis of this study is to determine the effects of intracerebroventricular (icv) prokineticin 2 infusion on food consumption and body weight and to elucidate whether it has effects on energy expenditure via the hypothalamus-pituitary-thyroid (HPT) axis in adipose tissue. MATERIAL AND METHODS: A total of 40 rats were used in the study and 4 groups were established: Control, Sham, Prokineticin 1.5 and Prokineticin 4.5 (n=10). Except for the Control group, rats were treated intracerebroventricularly via osmotic minipumps, the Sham group was infused with aCSF (vehicle), and the Prokineticin 1.5 and Prokineticin 4.5 groups were infused with 1.5 nMol and 4.5 nMol prokineticin 2, respectively. Food and water consumption and body weight were monitored during 7-day infusion in all groups. At the end of the infusion, the rats were decapitated and serum TSH, fT4 and fT3 levels were determined by ELISA. In addition, PGC-1α and UCP1 gene expression levels in white adipose tissue (WAT) and brown adipose tissue (BAT), TRH from rat hypothalamic tissue were determined by real-time PCR. RESULTS: Icv prokineticin 2 (4.5 nMol) infusion had no effect on water consumption but reduced daily food consumption and body weight (p<0.05). Icv prokineticin 2 (4.5 nMol) infusion significantly increased serum TSH, fT4 and fT3 levels when compared to Control and Sham groups (p<0.05). Also, icv prokineticin 2 (4.5 nMol) infusion increased the expression of TRH in the hypothalamus tissue and expression of PGC-1α UCP1 in the WAT and BAT (p<0.05). CONCLUSION: Icv prokineticin 2 (4.5 nMol) infusion may suppress food consumption via its receptors in the hypothalamus and reduce body weight by stimulating energy expenditure and thermogenesis in adipose tissue through the HPT axis.


Asunto(s)
Peso Corporal , Ingestión de Alimentos , Metabolismo Energético , Hormonas Gastrointestinales , Infusiones Intraventriculares , Glándula Tiroides , Animales , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Masculino , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/metabolismo , Ratas , Hormonas Gastrointestinales/metabolismo , Hormonas Gastrointestinales/administración & dosificación , Proteína Desacopladora 1/metabolismo , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Neuropéptidos/metabolismo , Neuropéptidos/administración & dosificación , Tirotropina/sangre , Tirotropina/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tiroxina/sangre , Tiroxina/administración & dosificación , Ingestión de Líquidos/efectos de los fármacos , Triyodotironina/administración & dosificación , Triyodotironina/sangre , Triyodotironina/farmacología , Ratas Wistar , Hipotálamo/metabolismo , Hipotálamo/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos
2.
J Clin Endocrinol Metab ; 107(2): e767-e782, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34460933

RESUMEN

CONTEXT: The gut-derived peptide hormones glucagon-like peptide-1 (GLP-1), oxyntomodulin (OXM), and peptide YY (PYY) are regulators of energy intake and glucose homeostasis and are thought to contribute to the glucose-lowering effects of bariatric surgery. OBJECTIVE: To establish the metabolomic effects of a combined infusion of GLP-1, OXM, and PYY (tripeptide GOP) in comparison to a placebo infusion, Roux-en-Y gastric bypass (RYGB) surgery, and a very low-calorie diet (VLCD). DESIGN AND SETTING: Subanalysis of a single-blind, randomized, placebo-controlled study of GOP infusion (ClinicalTrials.gov NCT01945840), including VLCD and RYGB comparator groups. PATIENTS AND INTERVENTIONS: Twenty-five obese patients with type 2 diabetes or prediabetes were randomly allocated to receive a 4-week subcutaneous infusion of GOP (n = 14) or 0.9% saline control (n = 11). An additional 22 patients followed a VLCD, and 21 underwent RYGB surgery. MAIN OUTCOME MEASURES: Plasma and urine samples collected at baseline and 4 weeks into each intervention were subjected to cross-platform metabolomic analysis, followed by unsupervised and supervised modeling approaches to identify similarities and differences between the effects of each intervention. RESULTS: Aside from glucose, very few metabolites were affected by GOP, contrasting with major metabolomic changes seen with VLCD and RYGB. CONCLUSIONS: Treatment with GOP provides a powerful glucose-lowering effect but does not replicate the broader metabolomic changes seen with VLCD and RYGB. The contribution of these metabolomic changes to the clinical benefits of RYGB remains to be elucidated.


Asunto(s)
Restricción Calórica/estadística & datos numéricos , Diabetes Mellitus Tipo 2/terapia , Derivación Gástrica/estadística & datos numéricos , Hormonas Gastrointestinales/administración & dosificación , Obesidad Mórbida/terapia , Adulto , Anciano , Glucemia/análisis , Restricción Calórica/métodos , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/orina , Quimioterapia Combinada/métodos , Femenino , Derivación Gástrica/métodos , Péptido 1 Similar al Glucagón/administración & dosificación , Humanos , Infusiones Subcutáneas , Masculino , Metabolómica/estadística & datos numéricos , Persona de Mediana Edad , Obesidad Mórbida/sangre , Obesidad Mórbida/metabolismo , Obesidad Mórbida/orina , Oxintomodulina/administración & dosificación , Péptido YY/administración & dosificación , Método Simple Ciego , Resultado del Tratamiento , Pérdida de Peso , Adulto Joven
3.
Acta Biochim Pol ; 67(3): 333-340, 2020 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-32894825

RESUMEN

Ulcerative colitis (UC) is a major type of inflammatory bowel disease (IBD) and significantly impacts patient quality of life. Previous research revealed that the guanylate cyclase-C (GC-C) signaling pathway is associated with the severity of UC. We aimed to investigate the effect of the GC-C agonist, guanylin (Gn), on inflammatory injury in mice with colitis. An experimental UC model was established in Balb/c mice. Mesalamine served as a positive control. The Gn overexpression vector was administered once per day for 1 week. Intestinal permeability of the mice was measured using fluorescein isothiocyanate-dextran after the treatment. Histopathologic grading was estimated to assess the inflammatory injury of the colon. The expression level of crucial mediators of the GC-C signaling pathway (Gn, Ugn and GC-C) and tight junction proteins (occludin, claudin-1 and ZO-1) was measured in the colon. Additionally, the level of pro-inflammatory cytokines (IL-8 and TNF-α) in serum was measured. After injecting the UC mice with the Gn overexpression vector, the body weight increased, and the frequency of loose stools and bloody stools was decreased. Intestinal permeability and histopathologic score were significantly reduced (P<0.05). The expression level of GC-C, Gn, Ugn, claudin-1 and ZO-1 was significantly increased (P<0.05). The level of IL-8 and TNF-α in the serum was significantly decreased (P<0.01). Therefore, the application of Gn overexpression vector can ameliorate the intestinal inflammatory injury and repair the mucosal barrier in colitis mice, which further suggests the clinical therapeutic potential of GC-C agonists in IBD.


Asunto(s)
Colitis Ulcerosa/sangre , Colitis Ulcerosa/tratamiento farmacológico , Activadores de Enzimas/administración & dosificación , Hormonas Gastrointestinales/administración & dosificación , Vectores Genéticos/administración & dosificación , Péptidos Natriuréticos/administración & dosificación , Receptores de Enterotoxina/metabolismo , Animales , Colitis Ulcerosa/inducido químicamente , Colon/metabolismo , Citocinas/sangre , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Hormonas Gastrointestinales/genética , Mucosa Intestinal/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , Ligandos , Masculino , Mesalamina/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Péptidos Natriuréticos/genética , Permeabilidad/efectos de los fármacos , Plásmidos/genética , Transducción de Señal/efectos de los fármacos , Proteínas de Uniones Estrechas/metabolismo
4.
Biochem Pharmacol ; 171: 113723, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31756326

RESUMEN

The current study has determined the ability of exendin-4 to augment the antidiabetic benefits of the recently characterised GIP/xenin hybrid, (DAla2)GIP/xenin-8-Gln. As such, combined activation of metabolic pathways linked to various gut derived hormones has been shown to exert complementary beneficial metabolic effects in diabetes. (DAla2)GIP/xenin-8-Gln and exendin-4 were administered twice daily to high fat fed (HFF) or db/db mice for 28 days and antidiabetic benefits assessed. Persistence of beneficial metabolic effects in HFF mice was also examined. Twice-daily injection of (DAla2)GIP/xenin-8-Gln for 28 days in HFF mice significantly reduced energy intake, body weight, circulating glucose, HbA1c and improved glucose tolerance and insulin sensitivity. Overall pancreatic islet, alpha- and beta-cell areas were reduced, with concurrent reduction in alpha- and beta-cell proliferation that was more apparent in the combined treatment group. Addition of exendin-4 to (DAla2)GIP/xenin-8-Gln therapy did not significantly improve metabolic control. Remarkably, beneficial effects were still evident 14 days following complete cessation of peptide administration. Thus, circulating glucose and insulin, HbA1c concentrations and glucose tolerance were still significantly improved when compared to control HFF mice on day 42, with minimal changes to pancreatic islet architecture. In contrast to HFF mice, combined treatment of db/db mice with (DAla2)GIP/xenin-8-Gln plus exendin-4 was required to induce beneficial effects on key metabolic parameters, which were not observed with either treatment alone. This included improvements in glucose tolerance and insulin sensitivity, but no effect on pancreatic architecture. These studies highlight the clear, and persistent, metabolic advantages of sustained activation of GLP-1 receptors, alongside concurrent activation of related GIP and xenin cell signalling pathways, in diabetes.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético/efectos de los fármacos , Exenatida/administración & dosificación , Polipéptido Inhibidor Gástrico/administración & dosificación , Neurotensina/administración & dosificación , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 2/prevención & control , Dieta Alta en Grasa/efectos adversos , Encefalinas/administración & dosificación , Hormonas Gastrointestinales/administración & dosificación , Hemoglobina Glucada/metabolismo , Hipoglucemiantes/administración & dosificación , Insulina/sangre , Ratones , Resultado del Tratamiento
5.
Expert Opin Drug Discov ; 14(11): 1151-1159, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31355685

RESUMEN

Introduction: Obesity is a global pandemic and new pharmacotherapies which combine weight loss efficacy, long-term safety, and reversal of metabolic co-morbidities are sorely needed. Gut hormones play key roles in regulating satiety and metabolism, and are natural candidates for therapeutic development. Areas covered: The authors discuss recent drug developments for the treatment of obesity using gut hormones. The review was based on a search of PubMed using keywords 'obesity' AND ('therapy' OR 'pharmacotherapy' OR 'gut hormones' OR 'analogues'), limited to the last 10 years. Expert opinion: Analogues of glucagon-like peptide (GLP-1) have been developed for obesity but so far do not provide enough weight loss. Bariatric surgery increases the post-prandial secretion of multiple gut hormones, leading to beneficial effects on weight loss and metabolism. This recognition has led to poly-agonist strategies: GLP-1/glucagon or GLP-1/glucose-dependent insulinotropic peptide (GIP) dual agonism, or even GLP-1/glucagon/GIP triple agonism. New delivery approaches include peptide-conjugate therapies that target key metabolic tissues. Practicable methods for oral delivery of peptide gut hormones are also close to market, expanding the potential market for these treatments. Anti-obesity therapy is therefore poised for an exciting phase, and it will be interesting to see which of these will eventually prevail.


Asunto(s)
Fármacos Antiobesidad/farmacología , Descubrimiento de Drogas/métodos , Obesidad/tratamiento farmacológico , Animales , Fármacos Antiobesidad/administración & dosificación , Sistemas de Liberación de Medicamentos , Desarrollo de Medicamentos/métodos , Hormonas Gastrointestinales/administración & dosificación , Hormonas Gastrointestinales/metabolismo , Hormonas Gastrointestinales/farmacología , Humanos , Pérdida de Peso/efectos de los fármacos
6.
Glia ; 66(10): 2137-2157, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30277602

RESUMEN

Astrocyte reactivity is disease- and stimulus-dependent, adopting either a proinflammatory A1 phenotype or a protective, anti-inflammatory A2 phenotype. Recently, we demonstrated, using cell culture, animal models and human brain samples, that dopaminergic neurons produce and secrete higher levels of the chemokine-like signaling protein Prokineticin-2 (PK2) as a compensatory protective response against neurotoxic stress. As astrocytes express a high level of PK2 receptors, herein, we systematically characterize the role of PK2 in astrocyte structural and functional properties. PK2 treatment greatly induced astrocyte migration, which was accompanied by a shift in mitochondrial energy metabolism, a reduction in proinflammatory factors, and an increase in the antioxidant genes Arginase-1 and Nrf2. Overexpression of PK2 in primary astrocytes or in the in vivo mouse brain induced the A2 astrocytic phenotype with upregulation of key protective genes and A2 reactivity markers including Arginase-1 and Nrf2, PTX3, SPHK1, and TM4SF1. A small-molecule PK2 agonist, IS20, not only mimicked the protective effect of PK2 in primary cultures, but also increased glutamate uptake by upregulating GLAST. Notably, IS20 blocked not only MPTP-induced reductions in the A2 phenotypic markers SPHK1 and SCL10a6 but also elevation of the of A1 marker GBP2. Collectively, our results reveal that PK2 regulates a novel neuron-astrocyte signaling mechanism by promoting an alternative A2 protective phenotype in astrocytes, which could be exploited for development of novel therapeutic strategies for PD and other related chronic neurodegenerative diseases. PK2 signals through its receptors on astrocytes and promotes directed chemotaxis. PK2-induced astrocyte reactivity leads to an increase in antioxidant and anti-inflammatory proteins while increasing glutamate uptake, along with decreased inflammatory factors. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Astrocitos/metabolismo , Quimiotaxis/fisiología , Hormonas Gastrointestinales/metabolismo , Neuropéptidos/metabolismo , Animales , Arginasa/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Hormonas Gastrointestinales/administración & dosificación , Regulación de la Expresión Génica , Ácido Glutámico/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Dinámicas Mitocondriales/fisiología , Factor 2 Relacionado con NF-E2/metabolismo , Neuropéptidos/administración & dosificación , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes/administración & dosificación , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
7.
Annu Rev Pharmacol Toxicol ; 57: 585-605, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-28061687

RESUMEN

Antiobesity medical management has shown unsatisfactory results to date in terms of efficacy, safety, and long-term maintenance of weight loss. This poor performance could be attributed to the complexity of appetite regulation mechanisms; the serious drug side effects; and, crucially, the lack of profile-matching treatment strategies and individualized, multidisciplinary follow-up. Nevertheless, antiobesity pharmacotherapy remains a challenging, exciting field of intensive scientific interest. According to the latest studies, the future of bariatric medicine lies in developing drugs acting at multiple levels of the brain-gut axis. Currently, research is focused on the generation of combination treatments based on gut hormones in a way that mimics changes underlying surgically induced weight loss, in addition to centrally acting agents; these aim to restore energy balance disruptions and enhance energy expenditure. Collectively, the pharmacological resolution of obesity could potentially be achieved with combination regimens targeting different molecules and levels of the energy homeostasis system, in parallel with matching patients' needs, resulting in a favorable metabolic profile.


Asunto(s)
Fármacos Antiobesidad/metabolismo , Hormonas Gastrointestinales/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Animales , Fármacos Antiobesidad/administración & dosificación , Regulación del Apetito/efectos de los fármacos , Regulación del Apetito/fisiología , Sistemas de Liberación de Medicamentos/métodos , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Hormonas Gastrointestinales/administración & dosificación , Humanos , Hormonas Peptídicas/administración & dosificación , Hormonas Peptídicas/metabolismo , Resultado del Tratamiento
8.
J Neuroendocrinol ; 28(9)2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27203571

RESUMEN

Peripheral anorectic hormones, such as glucagon-like peptide (GLP)-1, cholecystokinin (CCK)-8 and leptin, suppress food intake. The newly-identified anorectic neuropeptide, nesfatin-1, is synthesised in both peripheral tissues and the central nervous system, particularly by various nuclei in the hypothalamus and brainstem. In the present study, we examined the effects of i.p. administration of GLP-1 and CCK-8 and co-administrations of GLP-1 and leptin at subthreshold doses as confirmed by measurement of food intake, on nesfatin-1-immunoreactive (-IR) neurones in the hypothalamus and brainstem of rats by Fos immunohistochemistry. Intraperitoneal administration of GLP-1 (100 µg/kg) caused significant increases in the number of nesfatin-1-IR neurones expressing Fos-immunoreactivity in the supraoptic nucleus (SON), the area postrema (AP) and the nucleus tractus solitarii (NTS) but not in the paraventricular nucleus (PVN), the arcuate nucleus (ARC) or the lateral hypothalamic area (LHA). On the other hand, i.p. administration of CCK-8 (50 µg/kg) resulted in marked increases in the number of nesfatin-1-IR neurones expressing Fos-immunoreactivity in the SON, PVN, AP and NTS but not in the ARC or LHA. No differences in the percentage of nesfatin-1-IR neurones expressing Fos-immunoreactivity in the nuclei of the hypothalamus and brainstem were observed between rats treated with saline, GLP-1 (33 µg/kg) or leptin. However, co-administration of GLP-1 (33 µg/kg) and leptin resulted in significant increases in the number of nesfatin-1-IR neurones expressing Fos-immunoreactivity in the AP and the NTS. Furthermore, decreased food intake induced by GLP-1, CCK-8 and leptin was attenuated significantly by pretreatment with i.c.v. administration of antisense nesfatin-1. These results indicate that nesfatin-1-expressing neurones in the brainstem may play an important role in sensing peripheral levels of GLP-1 and leptin in addition to CCK-8, and also suppress food intake in rats.


Asunto(s)
Tronco Encefálico/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN/metabolismo , Conducta Alimentaria , Hormonas Gastrointestinales/fisiología , Hipotálamo/metabolismo , Leptina/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Animales , Ingestión de Alimentos , Hormonas Gastrointestinales/administración & dosificación , Péptido 1 Similar al Glucagón/administración & dosificación , Péptido 1 Similar al Glucagón/sangre , Péptido 1 Similar al Glucagón/fisiología , Inyecciones Intraperitoneales , Leptina/administración & dosificación , Leptina/sangre , Masculino , Nucleobindinas , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas Wistar , Sincalida/administración & dosificación , Sincalida/fisiología , Núcleo Supraóptico/metabolismo
9.
Physiol Rep ; 4(10)2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27207785

RESUMEN

Glucagon-like peptide-1 (GLP-1), peptide YY (3-36) [PYY(3-36)], amylin, ghrelin, insulin, and leptin are thought to act as hormonal signals from periphery to brain to control food intake. Here, we determined the effects of solid-phase extraction of plasma in measuring these hormones in blood of lean and diet-induced obese rats. Individual enzyme-linked immunoassays and a multiplex assay were used to measure active GLP-1, total PYY, active amylin, active ghrelin, insulin, leptin, and total GIP in response to (1) addition of known amounts of the peptides to lean and obese plasma, (2) a large meal in lean and obese rats, and (3) intravenous infusions of anorexigenic doses of GLP-1, PYY(3-36), amylin, and leptin in lean rats. Extraction of lean and obese plasma prior to assays produced consistent recoveries across assays for GLP-1, PYY, amylin, ghrelin, and insulin, reflecting losses inherent to the extraction procedure. Plasma extraction prior to assays generally revealed larger meal-induced changes in plasma GLP-1, PYY, amylin, ghrelin, and insulin in lean and obese rats. Plasma extraction and the multiplex assay were used to compare plasma levels of GLP-1, PYY, and amylin after a large meal with plasma levels produced by IV infusions of anorexigenic doses of GLP-1, PYY(3-36), and amylin. Infusions produced dose-dependent increases in plasma peptide levels, which were well above their postprandial levels. These results do not support the hypothesis that postprandial plasma levels of GLP-1, PYY(3-36), and amylin are sufficient to decrease food intake by an endocrine mechanism.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos/fisiología , Ayuno/sangre , Hormonas Gastrointestinales/sangre , Obesidad/sangre , Extracción en Fase Sólida/métodos , Delgadez/sangre , Animales , Ingestión de Alimentos/efectos de los fármacos , Hormonas Gastrointestinales/administración & dosificación , Infusiones Intravenosas , Masculino , Obesidad/inducido químicamente , Ratas
10.
Int J Neuropsychopharmacol ; 18(5)2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25522396

RESUMEN

BACKGROUND: Central serotonergic pathways influence brain areas involved in vagal cardiovascular regulation and, thereby, influence sympathetic efferent activity. Selective serotonin reuptake inhibitors (SSRIs) affect multiple serotonergic pathways, including central autonomic pathways. However, only a few studies have assessed SSRI-mediated effects on autonomic reactivity in healthy individuals using heart rate variability (HRV). METHODS: The present study assessed the influence of long-term treatment with escitalopram (ESC) on autonomic reactivity to an intravenous application of 50 µg cholecystokinin tetrapeptide (CCK-4) in 30 healthy young men using a double-blind, placebo (PLA)-controlled, randomized, within-subject cross-over design. Main outcome measures were time- and frequency-domain HRV parameters, assessed at both baseline and immediately after CCK-4 application. RESULTS: Results showed substantial effects for the treatment × CCK-4 challenge interaction with respect to heart rate (p < 0.001; pη(2) = 0.499), SDNN (p < 0.001; pη(2) = 576), RMSSD (p = 0.015; pη(2) = 194), NN50% (p = 0.008; pη(2) = 0.224), and LF% (p = 0.014; pη(2) = 0.196), and moderate effects with respect HF% (p = 0.099; pη(2) = 0.094), with PLA subjects showing a higher increase in HR and SDNN and a higher decrease in RMSSD, NN50, LF and HF than subjects in the ESC condition. Thus, ESC treatment significantly blunted the autonomic reactivity to CCK-4. Secondary analysis indicated no effect of the 5-HTTLPR polymorphism on CCK-4-induced autonomic response. CONCLUSIONS: Our results support findings suggesting an effect of SSRI treatment on autonomic regulation and provide evidence that ESC treatment is associated with blunted autonomic reactivity in healthy men.


Asunto(s)
Sistema Nervioso Autónomo/efectos de los fármacos , Citalopram/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Administración Intravenosa , Adulto , Sistema Nervioso Autónomo/fisiología , Citalopram/administración & dosificación , Estudios Cruzados , Método Doble Ciego , Hormonas Gastrointestinales/administración & dosificación , Hormonas Gastrointestinales/farmacología , Voluntarios Sanos , Humanos , Masculino , Distribución Aleatoria , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Tetragastrina/administración & dosificación , Tetragastrina/farmacología , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
11.
Br J Pharmacol ; 168(2): 403-10, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22935107

RESUMEN

BACKGROUND AND PURPOSE: Prokineticin 2 (PK2) has recently been shown to acutely reduce food intake in rodents. We aimed to determine the CNS sites and receptors that mediate the anorectic effects of peripherally administered PK2 and its chronic effects on glucose and energy homeostasis. EXPERIMENTAL APPROACH: We investigated neuronal activation following i.p. administration of PK2 using c-Fos-like immunoreactivity (CFL-IR). The anorectic effect of PK2 was examined in mice with targeted deletion of either prokineticin receptor 1 (PKR1) or prokineticin receptor 2 (PKR2), and in wild-type mice following administration of the PKR1 antagonist, PC1. The effect of IP PK2 administration on glucose homeostasis was investigated. Finally, the effect of long-term administration of PK2 on glucose and energy homeostasis in diet-induced obese (DIO) mice was determined. KEY RESULTS: I.p. PK2 administration significantly increased CFL-IR in the dorsal motor vagal nucleus of the brainstem. The anorectic effect of PK2 was maintained in mice lacking the PKR2 but abolished in mice lacking PKR1 and in wild-type mice pre-treated with PC1. DIO mice treated chronically with PK2 had no changes in glucose levels but significantly reduced food intake and body weight compared to controls. CONCLUSIONS AND IMPLICATIONS: Together, our data suggest that the anorectic effects of peripherally administered PK2 are mediated via the brainstem and this effect requires PKR1 but not PKR2 signalling. Chronic administration of PK2 reduces food intake and body weight in a mouse model of human obesity, suggesting that PKR1-selective agonists have potential to be novel therapeutics for the treatment of obesity.


Asunto(s)
Fármacos Antiobesidad/administración & dosificación , Tronco Encefálico/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Hormonas Gastrointestinales/administración & dosificación , Neuropéptidos/administración & dosificación , Receptores Acoplados a Proteínas G/fisiología , Animales , Glucemia/análisis , Peso Corporal/efectos de los fármacos , Tronco Encefálico/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Obesidad/tratamiento farmacológico , Obesidad/fisiopatología , Proteínas Proto-Oncogénicas c-fos/metabolismo
12.
Int J Pharm ; 440(1): 48-62, 2013 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-22503954

RESUMEN

Diabetes is a metabolic disease and is a major cause of mortality and morbidity in epidemic proportions. A type I diabetic patient is dependent on daily injections of insulin, for survival and also to maintain a normal life, which is uncomfortable, painful and also has deleterious effects. Extensive efforts are being made worldwide for developing noninvasive drug delivery systems, especially via oral route. Oral route is the most widely accepted means of administration. However it is not feasible for direct delivery of peptide and protein drugs. To overcome the gastro-intestinal barriers various types of formulations such as polymeric micro/nanoparticles, liposomes, etc. are investigated. In the recent years lot of advances have taken place in developing and understanding the oral peptide delivery systems. Simultaneously, the development and usage of other peptides having anti-diabetic potentials are also considered for diabetes therapy. In this review we are focusing on the advances reported during the past decade in the field of oral insulin delivery along with the possibility of other peptidic incretin hormones such as GLP-1, exendin-4, for diabetes therapy.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hormonas Gastrointestinales/administración & dosificación , Hipoglucemiantes/administración & dosificación , Insulina/administración & dosificación , Péptidos/administración & dosificación , Ponzoñas/administración & dosificación , Administración Oral , Animales , Exenatida , Humanos , Incretinas/administración & dosificación
13.
Ann Clin Psychiatry ; 23(3): 193-201, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21808751

RESUMEN

BACKGROUND: This randomized, open-label trial aimed to compare the metabolic effects of olanzapine orally disintegrating tablets (ODT) and solid oral tablets (SOT) in bipolar depressed and mixed outpatients. METHODS: Participants were openly randomized to receive olanzapine ODT (n = 13) or SOT (n = 10), 10 to 20 mg, once daily. Weight, body mass index (BMI), Food Craving Inventory (FCI), and Three-Factor Eating Questionnaire (3-FEQ) scores were assessed at baseline and at weeks 1, 2, 4, 6, and 8. Fasting glucose and lipid levels were assessed at baseline and at week 8. Insulin and leptin concentrations were measured just prior to olanzapine baseline dosing, 1 and 2 hours following administration of baseline dose, and at weeks 4 and 8. RESULTS: Patients showed significant increases in weight, BMI, and leptin area under the concentration-time curve (AUC), but not in FCI or 3-FEQ scores, over 8 weeks of treatment with olanzapine ODT and SOT. However, no significant differences between olanzapine formulations (ODT vs SOT) were observed in any of the measures assessed, except for a significantly lower triglyceride concentration in the ODT group at week 8. CONCLUSIONS: There was no consistent difference in metabolic profile between olanzapine ODT and SOT formulations during short-term treatment of bipolar depressed patients. Potential differences related to effects on triglyceride concentration warrant further confirmation.


Asunto(s)
Antipsicóticos/administración & dosificación , Benzodiazepinas/administración & dosificación , Trastorno Bipolar/tratamiento farmacológico , Peso Corporal/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Índice Glucémico/efectos de los fármacos , Administración Oral , Adulto , Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico , Benzodiazepinas/farmacología , Benzodiazepinas/uso terapéutico , Esquema de Medicación , Femenino , Hormonas Gastrointestinales/administración & dosificación , Hormonas Gastrointestinales/sangre , Hormonas Gastrointestinales/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Olanzapina , Comprimidos/administración & dosificación
14.
J Endocrinol ; 207(1): 87-93, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20631047

RESUMEN

Recently, glucagon-like peptide 1 (GLP1) and glucose-dependent insulinotropic polypeptide (GIP) have received much attention regarding possible roles in aetiology and treatment of type 2 diabetes. However, peptides co-secreted from the same enteroendocrine cells are less well studied. The present investigation was designed to characterise the in vitro and in vivo effects of xenin, a peptide co-secreted with GIP from intestinal K-cells. We examined the enzymatic stability, insulin-releasing activity and associated cAMP production capability of xenin in vitro. In addition, the effects of xenin on satiety, glucose homoeostasis and insulin secretion were examined in vivo. Xenin was time dependently degraded (t(1/2)=162±6 min) in plasma in vitro. In clonal BRIN-BD11 cells, xenin stimulated insulin secretion at 5.6 mM (P<0.05) and 16.7 mM (P<0.05 to P<0.001) glucose levels compared to respective controls. Xenin also exerted an additive effect on GIP, GLP1 and neurotensin-mediated insulin secretion. In clonal ß-cells, xenin did not stimulate cellular cAMP production, alter membrane potential or elevate intra-cellular Ca(2)(+). In normal mice, xenin exhibited a short-acting (P<0.01) satiety effect at high dosage (500 nmol/kg). In overnight fasted mice, acute injection of xenin enhanced glucose-lowering and elevated insulin secretion when injected concomitantly or 30 min before glucose. These effects were not observed when xenin was administered 60 min before the glucose challenge, reflecting the short half-life of the native peptide in vivo. Overall, these data demonstrate that xenin may have significant metabolic effects on glucose control, which merit further study.


Asunto(s)
Glucemia/metabolismo , Hormonas Gastrointestinales/farmacología , Hormonas Gastrointestinales/fisiología , Insulina/metabolismo , Neurotensina/farmacología , Neurotensina/fisiología , Respuesta de Saciedad/efectos de los fármacos , Respuesta de Saciedad/fisiología , Animales , Línea Celular , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Hormonas Gastrointestinales/administración & dosificación , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ratones , Neurotensina/administración & dosificación , Estabilidad Proteica , Transducción de Señal/efectos de los fármacos
15.
Curr Mol Pharmacol ; 2(3): 285-92, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20021465

RESUMEN

Guanylyl cyclase C (GCC) is the receptor specifically expressed by intestinal cells for the paracrine hormones guanylin and uroguanylin and diarrheagenic bacterial heat-stable enterotoxins. This tissue-specific receptor coordinates lineage-dependent regulation of epithelial homeostasis, and its disruption contributes to intestinal tumorigenesis. It coordinates regenerative and metabolic circuits by restricting the cell cycle and proliferation and programming metabolic transitions central to organizing the dynamic crypt-surface axis. Further, mice deficient in GCC signaling are more susceptible to colon cancer induced by Apc mutations or the carcinogen azoxymethane. Moreover, guanylin and uroguanylin are gene products most commonly lost, early, in colon cancer in animals and humans. The role of GCC as a tumor suppressing receptor regulating proliferation and metabolism, together with the universal loss of guanylin and uroguanylin in tumorigenesis, suggests a model in which colorectal cancer is a paracrine hormone deficiency syndrome. In that context, activation of GCC reverses the tumorigenic phenotype by limiting growth of colorectal cancer cells by restricting progression through the G1/S transition and reprogramming metabolic circuits from glycolysis to oxidative phosphorylation, limiting bioenergetic support for rapid proliferation. These observations suggest a pathophysiological hypothesis in which GCC is a lineage-dependent tumor suppressing receptor coordinating proliferative homeostasis whose dysregulation through hormone loss contributes to neoplasia. The correlative therapeutic hypothesis suggests that colorectal cancer is a disease of hormone insufficiency that can be prevented or treated by oral supplementation with GCC ligands.


Asunto(s)
Neoplasias Colorrectales/prevención & control , Hormonas Gastrointestinales/uso terapéutico , Guanilato Ciclasa/genética , Terapia de Reemplazo de Hormonas , Péptidos Natriuréticos/uso terapéutico , Receptores de Péptidos/genética , Administración Oral , Animales , Proliferación Celular , Transformación Celular Neoplásica , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/fisiopatología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Hormonas Gastrointestinales/administración & dosificación , Hormonas Gastrointestinales/metabolismo , Guanilato Ciclasa/metabolismo , Humanos , Interfase , Intestino Grueso/metabolismo , Intestino Grueso/patología , Intestino Grueso/fisiopatología , Ratones , Péptidos Natriuréticos/administración & dosificación , Péptidos Natriuréticos/metabolismo , Especificidad de Órganos , Receptores de Enterotoxina , Receptores Acoplados a la Guanilato-Ciclasa , Receptores de Péptidos/metabolismo
18.
Nutr Clin Care ; 7(3): 114-21, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15624543

RESUMEN

Short bowel syndrome (SBS) is the predominant cause of intestinal failure and is associated with a high degree of morbidity and mortality. One of the reasons this occurs is because of the dramatic reduction in nutrient absorptive capacity. There are many causes for the development of SBS, but the most common is extensive surgical resection. The bowel has an impressive ability to adapt both morphologically and functionally. This adaptation becomes critical in decreasing morbidity and mortality. It is also critical for the improvement of quality of life and long-term outcomes. Provision of sufficient nutritional and fluid support is essential to the management of SBS patients. The primary goal is to prevent or eliminate the need for total parenteral nutrition. Specific diets and hormonal therapy may enhance the adaptation process depending on the characteristics of the remaining bowel. For those patients who experience both intestinal failure and life-threatening complications from total parenteral nutrition, bowel transplantation may be indicated.


Asunto(s)
Dieta con Restricción de Grasas , Apoyo Nutricional/métodos , Síndrome del Intestino Corto/terapia , Adaptación Fisiológica , Transporte Biológico , Terapia Combinada , Hormonas Gastrointestinales/administración & dosificación , Hormonas Gastrointestinales/fisiología , Humanos , Absorción Intestinal , Intestinos/fisiología , Intestinos/cirugía , Intestinos/trasplante , Nutrición Parenteral Total/efectos adversos , Nutrición Parenteral Total/métodos , Calidad de Vida , Síndrome del Intestino Corto/dietoterapia
19.
Diabetes Care ; 24(4): 720-5, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11315837

RESUMEN

OBJECTIVE: The incretin hormone glucagon-like peptide-1 (GLP-1) reduces plasma glucose in type 2 diabetic patients by stimulating insulin secretion and inhibiting glucagon secretion. The biguanide metformin is believed to lower plasma glucose without affecting insulin secretion. We conducted this study to investigate the effect of a combination therapy with GLP-1 and metformin, which could theoretically be additive, in type 2 diabetic patients. RESEARCH DESIGN AND METHODS: In a semiblinded randomized crossover study, seven patients received treatment with metformin (1,500 mg daily orally) alternating with GLP-1 (continuous subcutaneous infusion of 2.4 pmol x kg(-1) x min(-1)) alternating with a combination of metformin and GLP-1 for 48 h. Under fixed energy intake, we examined the effects on plasma glucose, insulin, C-peptide, glucagon, and appetite. RESULTS: Fasting plasma glucose (day 2) decreased from 13.9 +/- 1 (no treatment) to 11.2 +/- 0.4 (metformin) and 11.5 +/- 0.5 (GLP-1) and further decreased to 9.4 +/- 0.7 (combination therapy) (P = 0.0005, no difference between monotherapy with GLP-1 and metformin). The 24-h mean plasma glucose (day 2) decreased from 11.8 +/- 0.5 (metformin) and 11.7 +/- 0.8 (GLP-1) to 9.8 +/- 0.5 (combination) (P = 0.02, no difference between GLP-1 and metformin). Insulin levels were similar between the three regimens, but glucagon levels were significantly reduced with GLP-1 compared with metformin (P = 0.0003). Combination therapy had no additional effect on appetite scores. CONCLUSIONS: Monotherapy with GLP-1 and metformin have equal effects on plasma glucose and additive effects upon combination.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hormonas Gastrointestinales/uso terapéutico , Metformina/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Glucemia/metabolismo , Péptido C/sangre , Péptido C/metabolismo , Estudios Cruzados , Diabetes Mellitus Tipo 2/sangre , Esquema de Medicación , Quimioterapia Combinada , Femenino , Hormonas Gastrointestinales/administración & dosificación , Glucagón/sangre , Glucagón/metabolismo , Péptido 1 Similar al Glucagón , Péptidos Similares al Glucagón , Hemoglobina Glucada/análisis , Humanos , Infusiones Parenterales , Insulina/sangre , Insulina/metabolismo , Secreción de Insulina , Cinética , Masculino , Persona de Mediana Edad , Fragmentos de Péptidos/administración & dosificación , Placebos , Proyectos de Investigación
20.
Ugeskr Laeger ; 163(3): 287-91, 2001 Jan 15.
Artículo en Danés | MEDLINE | ID: mdl-11219107

RESUMEN

We report here that glucagon-like peptide 2(GLP-2) and its receptor constitute a distinct projection system connecting the nucleus of the solitary tract with the dorsomedial hypothalamic nucleus (DMH). The DMH contains a dense plexus of GLP-2 immunoreactive fibres and is the only hypothalamic nucleus expressing GLP-2 receptor mRNA. Consistent with this, central application of GLP-2 activates the expression of neurones solely in the DMH. Furthermore, central administration of GLP-2 causes a dose-related, a pharmacologically and behaviourally specific inhibition of food intake in rats. Surprisingly, the alleged GLP-1 receptor antagonist, Exending (9-39), proved a functional antagonist of centrally applied GLP-2. These data implicate GLP-2 as an important neurotransmitter in the regulation of food intake and likely bodyweight. Our data therefore point to the DMH as a crossroad for endocrine and visceral information affecting feeding behaviour.


Asunto(s)
Regulación del Apetito/fisiología , Hormonas Gastrointestinales/fisiología , Neurotransmisores/fisiología , Péptidos/fisiología , Receptores de Glucagón/fisiología , Animales , Regulación del Apetito/efectos de los fármacos , Tronco Encefálico/inmunología , Tronco Encefálico/metabolismo , Núcleo Hipotalámico Dorsomedial/inmunología , Núcleo Hipotalámico Dorsomedial/metabolismo , Hormonas Gastrointestinales/administración & dosificación , Hormonas Gastrointestinales/inmunología , Expresión Génica , Péptido 1 Similar al Glucagón , Péptido 2 Similar al Glucagón , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Neurotransmisores/administración & dosificación , Neurotransmisores/inmunología , Péptidos/administración & dosificación , Péptidos/inmunología , ARN Mensajero/genética , Ratas , Ratas Wistar , Receptores de Glucagón/genética , Receptores de Glucagón/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...