Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Mol Pharm ; 17(12): 4572-4588, 2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33125243

RESUMEN

Neurotensin (NTS)-polyplex is a multicomponent nonviral vector that enables gene delivery via internalization of the neurotensin type 1 receptor (NTSR1) to dopaminergic neurons and cancer cells. An approach to improving its therapeutic safety is replacing the viral karyophilic component (peptide KPSV40; MAPTKRKGSCPGAAPNKPK), which performs the nuclear import activity, by a shorter synthetic peptide (KPRa; KMAPKKRK). We explored this issue and the mechanism of plasmid DNA translocation through the expression of the green fluorescent protein or red fluorescent protein fused with KPRa and internalization assays and whole-cell patch-clamp configuration experiments in a single cell together with importin α/ß pathway blockers. We showed that KPRa electrostatically bound to plasmid DNA increased the transgene expression compared with KPSV40 and enabled nuclear translocation of KPRa-fused red fluorescent proteins and plasmid DNA. Such translocation was blocked with ivermectin or mifepristone, suggesting importin α/ß pathway mediation. KPRa also enabled NTS-polyplex-mediated expression of reporter or physiological genes such as human mesencephalic-derived neurotrophic factor (hMANF) in dopaminergic neurons in vivo. KPRa is a synthetic monopartite peptide that showed nuclear import activity in NTS-polyplex vector-mediated gene delivery. KPRa could also improve the transfection of other nonviral vectors used in gene therapy.


Asunto(s)
Portadores de Fármacos/síntesis química , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Neurotensina/administración & dosificación , Fragmentos de Péptidos/síntesis química , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Núcleo Celular/metabolismo , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Terapia Genética/métodos , Vectores Genéticos/genética , Masculino , Ratones , Modelos Animales , Nanopartículas/química , Neurotensina/genética , Neurotensina/farmacocinética , Técnicas de Placa-Clamp , Plásmidos/genética , Ratas , Receptores de Neurotensina/metabolismo , Análisis de la Célula Individual , Técnicas Estereotáxicas
2.
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
3.
Eur J Pharmacol ; 848: 80-87, 2019 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-30707956

RESUMEN

Opioid and neurotensin (NT) receptors are expressed in both central and peripheral nervous systems where they modulate nociceptive responses. Nowadays, opioid analgesics like morphine remain the most prescribed drugs for the treatment of moderate to severe pain. However, despite their daily used, opioids can produce life-threatening side effects, such as constipation or respiratory depression. Besides, NT analogs exert strong opioid-independent analgesia. Here, we thus hypothesized that the combined use of opioid and NT agonists would require lower doses to produce significant analgesic effects, hence decreasing opioid-induced adverse effects. We used isobologram analyses to determine if the combination of a NT brain-penetrant analog, An2-NT(8-13) with morphine results in an inhibitory, synergistic or additive analgesic response. We found that intravenous administration of An2-NT(8-13) reduced by 90% the nocifensive behaviors induced by formalin injection, at the dose of 0.018 mg/kg. Likewise, subcutaneous morphine reduced pain by 90% at 1.8 mg/kg. Importantly, isobologram analyses revealed that the co-injection of An2-NT(8-13) with morphine induced an additive analgesic response. We finally assessed the effects of morphine and An2-NT(8-13) on the gastrointestinal tract motility using the charcoal meal test. As opposed to morphine which significantly reduced the intestinal motility at the analgesic effective dose of 1.8 mg/kg, An2-NT(8-13) did not affect the charcoal meal intestinal transit at 0.018 mg/kg. Interestingly, at the dose providing 90% pain relief, the co-administration of morphine with An2-NT(8-13) had a reduced effect on constipation. Altogether, these results suggest that combining NT agonists with morphine may improve its analgesic/adverse effect ratio.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Neurotensina/administración & dosificación , Dimensión del Dolor/efectos de los fármacos , Fragmentos de Péptidos/administración & dosificación , Péptidos/administración & dosificación , Receptores de Neurotensina/agonistas , Receptores Opioides mu/agonistas , Animales , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Masculino , Morfina/administración & dosificación , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley , Receptores de Neurotensina/metabolismo , Receptores Opioides mu/metabolismo
4.
Mol Pharm ; 15(8): 3093-3100, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-29889537

RESUMEN

Several studies have suggested that neurotensin receptors (NTRs) and neurotensin (NT) greatly affect the growth and survival of pancreatic ductal adenocarcinoma (PDAC). Developing NTR-targeted PET probes could therefore be important for the management of a pancreatic cancer patient by providing key information on the NTR expression profile noninvasively. Despite the initial success on the synthesis of 18F-labeled NT PET probes, the labeling procedure generally requires lengthy steps including azeotropic drying of 18F. Using a straightforward chelation method, here we report the simple preparation of aluminum-18F-NOTA-NT starting from aqueous 18F. The cell binding test demonstrated that [19F]AlF-NOTA-NT maintained high receptor-binding affinity to NTR1. This probe was then further evaluated in NTR1 positive pancreatic tumor models (AsPC-1 and PANC-1). After the administration of [18F]AlF-NOTA-NT, small animal PET studies showed a high contrast between tumor and background in both models at 1 and 4 h time points. A blocking experiment was performed to demonstrate the receptor specificity: the tumor uptake in AsPC1 without and with blocking agent was 1.0 ± 0.2 and 0.1 ± 0.0%ID/g, respectively, at 4 h post injection. In summary, a NTR specific PET agent, [18F]AlF-NOTA-NT, was prepared through the simple chelation method. This NTR-targeted PET probe may not only be used to detect NTR1 positive pancreatic tumors (diagnosis), but also it may be fully integrated to NTR target therapy leading to personalized medicine (theranostic).


Asunto(s)
Carcinoma Ductal Pancreático/diagnóstico por imagen , Neoplasias Pancreáticas/diagnóstico , Radiofármacos/administración & dosificación , Receptores de Neurotensina/metabolismo , Aluminio/administración & dosificación , Aluminio/química , Animales , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Flúor/administración & dosificación , Flúor/química , Radioisótopos de Flúor/administración & dosificación , Radioisótopos de Flúor/química , Compuestos Heterocíclicos con 1 Anillo/administración & dosificación , Compuestos Heterocíclicos con 1 Anillo/química , Humanos , Ratones , Ratones Desnudos , Imagen Molecular/métodos , Terapia Molecular Dirigida/métodos , Neurotensina/administración & dosificación , Neurotensina/química , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Tomografía de Emisión de Positrones/métodos , Radiofármacos/química , Receptores de Neurotensina/antagonistas & inhibidores , Nanomedicina Teranóstica/métodos , Microtomografía por Rayos X/métodos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Eur J Pharm Biopharm ; 128: 147-155, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29680483

RESUMEN

Silk fibroin (SF) films containing a peptide, neurotensin (NT), stimulated by iontophoresis were developed aiming to modulate the inflammatory process and prevent the growth of microorganisms typical of wounds. NT-loaded SF films composition shows predominance of ß-sheet structures that conferred adequate mechanical properties, transparency, moderate roughness and low swelling index to fibroin films. Infrared spectroscopy and thermal analysis suggested the presence of non-covalent interactions between NT and fibroin. Using the MALDI imaging technique, it was possible to visualize the homogeneous NT distribution throughout the film surface, in addition to its prolonged release for up to 72 h. In vitro studies in E. coli liposaccharide-stimulated macrophages showed a significant reduction of interleukins production after NT-loaded film application, whereas NT solution did not reduce them. Bi-laminated NT-loaded fibroin films containing silver electrodes provided a burst release of NT when anodic iontophoresis was applied, enabling a rapid onset of drug action. In addition, anodic iontophoresis presented a bacteriostatic effect against gram-positive microorganisms. Different iontophoresis densities, from 0.2 to 0.6 mA/cm2, did not significantly reduce fibroblast viability after 30 min of application. In conclusion, iontophoretic-stimulated peptide-loaded fibroin films could be a promising platform for the treatment of wounds.


Asunto(s)
Antibacterianos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Fibroínas/química , Neurotensina/administración & dosificación , Cicatrización de Heridas/efectos de los fármacos , Animales , Antibacterianos/química , Bacterias/efectos de los fármacos , Bacterias/metabolismo , Bombyx/química , Supervivencia Celular , Fibroblastos , Interleucinas/metabolismo , Iontoforesis/métodos , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Pruebas de Sensibilidad Microbiana , Neurotensina/química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
6.
PLoS One ; 13(2): e0192441, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29466430

RESUMEN

We previously demonstrated that infusion of an intestinal peptide called xenin-25 (Xen) amplifies the effects of glucose-dependent insulinotropic polypeptide (GIP) on insulin secretion rates (ISRs) and plasma glucagon levels in humans. However, these effects of Xen, but not GIP, were blunted in humans with type 2 diabetes. Thus, Xen rather than GIP signaling to islets fails early during development of type 2 diabetes. The current crossover study determines if cholinergic signaling relays the effects of Xen on insulin and glucagon release in humans as in mice. Fasted subjects with impaired glucose tolerance were studied. On eight separate occasions, each person underwent a single graded glucose infusion- two each with infusion of albumin, Xen, GIP, and GIP plus Xen. Each infusate was administered ± atropine. Heart rate and plasma glucose, insulin, C-peptide, glucagon, and pancreatic polypeptide (PP) levels were measured. ISRs were calculated from C-peptide levels. All peptides profoundly increased PP responses. From 0 to 40 min, peptide(s) infusions had little effect on plasma glucose concentrations. However, GIP, but not Xen, rapidly and transiently increased ISRs and glucagon levels. Both responses were further amplified when Xen was co-administered with GIP. From 40 to 240 min, glucose levels and ISRs continually increased while glucagon concentrations declined, regardless of infusate. Atropine increased resting heart rate and blocked all PP responses but did not affect ISRs or plasma glucagon levels during any of the peptide infusions. Thus, cholinergic signaling mediates the effects of Xen on insulin and glucagon release in mice but not humans.


Asunto(s)
Glucagón/metabolismo , Intolerancia a la Glucosa/sangre , Insulina/metabolismo , Neurotensina/farmacología , Polipéptido Pancreático/metabolismo , Receptores Colinérgicos/metabolismo , Transducción de Señal , Adulto , Atropina/administración & dosificación , Atropina/farmacología , Glucemia/metabolismo , Estudios Cruzados , Femenino , Polipéptido Inhibidor Gástrico/administración & dosificación , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Secreción de Insulina , Masculino , Persona de Mediana Edad , Neurotensina/administración & dosificación
7.
Peptides ; 100: 202-211, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29412820

RESUMEN

Combined modulation of peptide hormone receptors including, glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP) and xenin, have established benefits for the treatment of diabetes. The present study has assessed the biological actions and therapeutic efficacy of a novel exendin-4/xenin-8-Gln hybrid peptide, both alone and in combination with the GIP receptor agonist (DAla2)GIP. Exendin-4/xenin-8-Gln was enzymatically stable and exhibited enhanced insulin secretory actions when compared to its parent peptides. Exendin-4/xenin-8-Gln also possessed ability to potentiate the in vitro actions of GIP. Acute administration of exendin-4/xenin-8-Gln in mice induced appetite suppressive effects, as well as significant and protracted glucose-lowering and insulin secretory actions. Twice daily administration of exendin-4/xenin-8-Gln, alone or in combination with (DAla2)GIP, for 21-days significantly reduced non-fasting glucose and increased circulating insulin levels in high fat fed mice. In addition, all exendin-4/xenin-8-Gln treated mice displayed improved glucose tolerance, insulin sensitivity and metabolic responses to GIP. Combination therapy with (DAla2)GIP did not result in any obvious further benefits. Metabolic improvements in all treatment groups were accompanied by reduced pancreatic beta-cell area and insulin content, suggesting reduced insulin demand. Interestingly, body weight, food intake, circulating glucagon, metabolic rate and amylase activity were unaltered by the treatment regimens. However, all treatment groups, barring (DAla2)GIP alone, exhibited marked reductions in total- and LDL-cholesterol. Furthermore, exendin-4 therapy also reduced circulating triacylglycerol. This study highlights the positive antidiabetic effects of exendin-4/xenin-8-Gln, and suggests that combined modulation of GLP-1 and xenin related signalling pathways represents an exciting treatment option for type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Polipéptido Inhibidor Gástrico/administración & dosificación , Péptido 1 Similar al Glucagón/administración & dosificación , Hipoglucemiantes/administración & dosificación , Animales , Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Combinación de Medicamentos , Exenatida/administración & dosificación , Exenatida/química , Polipéptido Inhibidor Gástrico/química , Glucagón/química , Glucagón/metabolismo , Péptido 1 Similar al Glucagón/química , Glucosa/metabolismo , Humanos , Hipoglucemiantes/química , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Neurotensina/administración & dosificación , Neurotensina/química
8.
Mol Cancer Res ; 15(7): 929-941, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28360038

RESUMEN

We examined the impact of crosstalk between the insulin receptor and G protein-coupled receptor (GPCR) signaling pathways on the regulation of Yes-associated protein (YAP) localization, phosphorylation, and transcriptional activity in the context of human pancreatic ductal adenocarcinoma (PDAC). Stimulation of PANC-1 or MiaPaCa-2 cells with insulin and neurotensin, a potent mitogenic combination of agonists for these cells, promoted striking YAP nuclear localization and decreased YAP phosphorylation at Ser127 and Ser397 Challenging PDAC cells with either insulin or neurotensin alone modestly induced the expression of YAP/TEAD-regulated genes, including connective tissue growth factor (CTGF), cysteine-rich angiogenic inducer 61 (CYR61), and CXCL5, whereas the combination of neurotensin and insulin induced a marked increase in the level of expression of these genes. In addition, siRNA-mediated knockdown of YAP/TAZ prevented the increase in the expression of these genes. A small-molecule inhibitor (A66), selective for the p110α subunit of PI3K, abrogated the increase in phosphatidylinositol 3,4,5-trisphosphate production and the expression of CTGF, CYR61, and CXCL5 induced by neurotensin and insulin. Furthermore, treatment of PDAC cells with protein kinase D (PKD) family inhibitors (CRT0066101 or kb NB 142-70) or with siRNAs targeting the PKD family prevented the increase of CTGF, CYR61, and CXCL5 mRNA levels in response to insulin and neurotensin stimulation. Thus, PI3K and PKD mediate YAP activation in response to insulin and neurotensin in pancreatic cancer cells.Implications: Inhibitors of PI3K or PKD disrupt crosstalk between insulin receptor and GPCR signaling systems by blocking YAP/TEAD-regulated gene expression in pancreatic cancer cells. Mol Cancer Res; 15(7); 929-41. ©2017 AACR.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Carcinoma Ductal Pancreático/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Neoplasias Pancreáticas/genética , Fosfoproteínas/genética , Proteína Quinasa C/genética , Antígenos CD/genética , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Quimiocina CXCL5/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Proteína 61 Rica en Cisteína/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Insulina/administración & dosificación , Insulina/metabolismo , Neurotensina/administración & dosificación , Neurotensina/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Fosforilación/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Pirimidinas/administración & dosificación , Receptor de Insulina/genética , Receptores Acoplados a Proteínas G/genética , Factores de Transcripción , Proteínas Señalizadoras YAP
9.
Adv Healthc Mater ; 5(23): 3016-3026, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27805786

RESUMEN

Delivery efficiency with gene transfection is a pivotal point in achieving maximized therapeutic efficacy and has been an important challenge with central nervous system (CNS) diseases. In this study, neurotensin (NT, a neuro-specific peptide)-conjugated polyethylenimine (PEI)-modified reduced graphene oxide (rGO) nanoparticles with precisely controlled two-stage near-infrared (NIR)-laser photothermal treatment to enhance the ability to target neurons and achieve high gene transfection in neurons. First-stage NIR laser irradiation on the cells with nanoparticles attached on the surface can increase the permeability of the cell membrane, resulting in an apparent increase in cellular uptake compared to untreated cells. In addition, second-stage NIR laser irradiation on the cells with nanoparticles inside can further induce endo/lysosomal cavitation, which not only helps nanoparticles escape from endo/lysosomes but also prevents plasmid DNA (pDNA) from being digested by DNase I. At least double pDNA amount can be released from rGO-PEI-NT/pDNA under NIR laser trigger release compared to natural release. Moreover, in vitro differentiated PC-12 cell and in vivo mice (C57BL/6) brain transfection experiments have demonstrated the highest transfection efficiency occurring when NT modification is combined with external multi-stage stimuli-responsive NIR laser treatment. The combination of neuro-specific targeting peptide and external NIR-laser-triggered aid provides a nanoplatform for gene therapy in CNS diseases.


Asunto(s)
Grafito/administración & dosificación , Enfermedades Neurodegenerativas/tratamiento farmacológico , Neuronas/efectos de los fármacos , Neurotensina/administración & dosificación , Óxidos/administración & dosificación , Animales , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/efectos de los fármacos , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Grafito/química , Ratones , Ratones Endogámicos C57BL , Nanopartículas/administración & dosificación , Nanopartículas/química , Neurotensina/química , Óxidos/química , Plásmidos/metabolismo , Polietileneimina/química , Ratas , Espectroscopía Infrarroja Corta/métodos , Transfección/métodos
10.
Endocrinology ; 157(9): 3482-92, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27580810

RESUMEN

Neurotensin (NT) is a peptide expressed in the brain and in the gastrointestinal tract. Brain NT inhibits food intake, but the effects of peripheral NT are less investigated. In this study, peripheral NT decreased food intake in both mice and rats, which was abolished by a NT antagonist. Using c-Fos immunohistochemistry, we found that peripheral NT activated brainstem and hypothalamic regions. The anorexigenic effect of NT was preserved in vagotomized mice but lasted shorter than in sham-operated mice. This in combination with a strong increase in c-Fos activation in area postrema after ip administration indicates that NT acts both through the blood circulation and the vagus. To improve the pharmacokinetics of NT, we developed a pegylated NT peptide, which presumably prolonged the half-life, and thus, the effect on feeding was extended compared with native NT. On a molecular level, the pegylated NT peptide increased proopiomelanocortin mRNA in the arcuate nucleus. We also investigated the importance of NT for the decreased food intake after gastric bypass surgery in a rat model of Roux-en-Y gastric bypass (RYGB). NT was increased in plasma and in the gastrointestinal tract in RYGB rats, and pharmacological antagonism of NT increased food intake transiently in RYGB rats. Taken together, our data suggest that NT is a metabolically active hormone, which contributes to the regulation of food intake.


Asunto(s)
Regulación del Apetito/efectos de los fármacos , Derivación Gástrica , Neurotensina/administración & dosificación , Animales , Ingestión de Alimentos/efectos de los fármacos , Femenino , Masculino , Ratones Endogámicos C57BL , Neurotensina/antagonistas & inhibidores , Neurotensina/sangre , Ratas Sprague-Dawley , Sacarosa , Vagotomía
11.
PLoS One ; 11(6): e0156852, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27304975

RESUMEN

UNLABELLED: Peripheral muscarinic acetylcholine receptors regulate insulin and glucagon release in rodents but their importance for similar roles in humans is unclear. Bethanechol, an acetylcholine analogue that does not cross the blood-brain barrier, was used to examine the role of peripheral muscarinic signaling on glucose homeostasis in humans with normal glucose tolerance (NGT; n = 10), impaired glucose tolerance (IGT; n = 11), and type 2 diabetes mellitus (T2DM; n = 9). Subjects received four liquid meal tolerance tests, each with a different dose of oral bethanechol (0, 50, 100, or 150 mg) given 60 min before a meal containing acetaminophen. Plasma pancreatic polypeptide (PP), glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide-1 (GLP-1), glucose, glucagon, C-peptide, and acetaminophen concentrations were measured. Insulin secretion rates (ISRs) were calculated from C-peptide levels. Acetaminophen and PP concentrations were surrogate markers for gastric emptying and cholinergic input to islets. The 150 mg dose of bethanechol increased the PP response 2-fold only in the IGT group, amplified GLP-1 release in the IGT and T2DM groups, and augmented the GIP response only in the NGT group. However, bethanechol did not alter ISRs or plasma glucose, glucagon, or acetaminophen concentrations in any group. Prior studies showed infusion of xenin-25, an intestinal peptide, delays gastric emptying and reduces GLP-1 release but not ISRs when normalized to plasma glucose levels. Analysis of archived plasma samples from this study showed xenin-25 amplified postprandial PP responses ~4-fold in subjects with NGT, IGT, and T2DM. Thus, increasing postprandial cholinergic input to islets augments insulin secretion in mice but not humans. TRIAL REGISTRATION: ClinicalTrials.gov NCT01434901.


Asunto(s)
Betanecol/farmacología , Diabetes Mellitus Tipo 2/sangre , Hormonas/sangre , Administración Oral , Adulto , Betanecol/administración & dosificación , Glucemia/metabolismo , Péptido C/sangre , Estudios Cruzados , Diabetes Mellitus Tipo 2/fisiopatología , Relación Dosis-Respuesta a Droga , Femenino , Vaciamiento Gástrico/efectos de los fármacos , Polipéptido Inhibidor Gástrico/sangre , Glucagón/sangre , Péptido 1 Similar al Glucagón/sangre , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/fisiopatología , Humanos , Insulina/sangre , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Masculino , Persona de Mediana Edad , Agonistas Muscarínicos/administración & dosificación , Agonistas Muscarínicos/farmacología , Neurotensina/administración & dosificación , Neurotensina/farmacología , Ensayos Clínicos Controlados no Aleatorios como Asunto , Polipéptido Pancreático/sangre , Periodo Posprandial
12.
Peptides ; 82: 76-84, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27288245

RESUMEN

Xenin-25 (Xen) is a neurotensin-related peptide secreted by a subset of enteroendocrine cells located in the proximal small intestine. Many effects of Xen are mediated by neurotensin receptor-1 on neurons. In healthy humans with normal glucose tolerance (NGT), Xen administration causes diarrhea and inhibits postprandial glucagon-like peptide-1 (GLP-1) release but not insulin secretion. This study determines (i) if Xen has similar effects in humans with Roux-en-Y gastric bypass (RYGB) and (ii) whether neural pathways potentially mediate effects of Xen on glucose homeostasis. Eight females with RYGB and no history of type 2 diabetes received infusions with 0, 4 or 12pmol Xen/kg/min with liquid meals on separate occasions. Plasma glucose and gastrointestinal hormone levels were measured and insulin secretion rates calculated. Pancreatic polypeptide and neuropeptide Y levels were surrogate markers for parasympathetic input to islets and sympathetic tone, respectively. Responses were compared to those in well-matched non-surgical participants with NGT from our earlier study. Xen similarly increased pancreatic polypeptide and neuropeptide Y responses in patients with and without RYGB. In contrast, the ability of Xen to inhibit GLP-1 release and cause diarrhea was severely blunted in patients with RYGB. With RYGB, Xen had no statistically significant effect on glucose, insulin secretory, GLP-1, glucose-dependent insulinotropic peptide, and glucagon responses. However, insulin and glucose-dependent insulinotropic peptide secretion preceded GLP-1 release suggesting circulating GLP-1 does not mediate exaggerated insulin release after RYGB. Thus, Xen has unmasked neural circuits to the distal gut that inhibit GLP-1 secretion, cause diarrhea, and are altered by RYGB.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diarrea/metabolismo , Insulina/metabolismo , Neurotensina/administración & dosificación , Adolescente , Adulto , Anciano , Glucemia , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/patología , Diarrea/inducido químicamente , Diarrea/fisiopatología , Femenino , Derivación Gástrica/métodos , Polipéptido Inhibidor Gástrico/metabolismo , Glucagón/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Humanos , Secreción de Insulina , Masculino , Persona de Mediana Edad , Neuropéptido Y/metabolismo , Neurotensina/efectos adversos , Neurotensina/metabolismo , Polipéptido Pancreático/metabolismo
13.
Acta Diabetol ; 52(3): 461-71, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25374384

RESUMEN

AIMS: Xenin-25 is co-secreted with glucose-dependent insulinotropic polypeptide (GIP) from intestinal K-cells following a meal. Xenin-25 is believed to play a key role in glucose homoeostasis and potentiate the insulinotropic effect of GIP. METHODS: This study investigated the effects of sub-chronic administration of the stable and longer-acting xenin-25 analogue, xenin-25[Lys(13)PAL] (25 nmol/kg), in diabetic mice fed with a high-fat diet. RESULTS: Initial studies confirmed the significant persistent glucose-lowering (p < 0.05) and insulin-releasing (p < 0.05) actions of xenin-25[Lys(13)PAL] compared with native xenin-25. Interestingly, xenin-25 retained significant glucose-lowering activity in GIP receptor knockout mice. Twice-daily intraperitoneal (i.p.) injection of xenin-25[Lys(13)PAL] for 14 days had no significant effect on food intake or body weight in high-fat-fed mice. Non-fasting glucose and insulin levels were also unchanged, but overall glucose levels during an i.p. glucose tolerance and oral nutrient challenge were significantly (p < 0.05) lowered by xenin-25[Lys(13)PAL] treatment. These changes were accompanied by significant improvements in i.p. (p < 0.05) and oral (p < 0.001) nutrient-stimulated insulin concentrations. No appreciable changes in insulin sensitivity were observed between xenin-25[Lys(13)PAL] and saline-treated high-fat mice. However, xenin-25[Lys(13)PAL] treatment restored notable sensitivity to the biological actions of exogenous GIP injection. Consumption of O2, production of CO2, respiratory exchange ratio and energy expenditure were not altered by 14-day twice-daily treatment with xenin-25[Lys(13)PAL]. In contrast, ambulatory activity was significantly (p < 0.05 to p < 0.001) increased during the dark phase in xenin-25[Lys(13)PAL] mice compared with high-fat controls. CONCLUSIONS: These data indicate that sustained administration of a stable analogue of xenin-25 exerts a spectrum of beneficial metabolic effects in high-fat-fed mice.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Hipoglucemiantes/administración & dosificación , Neurotensina/administración & dosificación , Acetilación , Animales , Glucemia/metabolismo , Modelos Animales de Enfermedad , Metabolismo Energético , Humanos , Hipoglucemiantes/química , Masculino , Ratones , Neurotensina/química , Oxígeno/metabolismo
14.
Am J Physiol Gastrointest Liver Physiol ; 306(4): G301-9, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24356886

RESUMEN

Xenin-25 (Xen) is a neurotensin-related peptide secreted by a subset of glucose-dependent insulinotropic polypeptide (GIP)-producing enteroendocrine cells. In animals, Xen regulates gastrointestinal function and glucose homeostasis, typically by initiating neural relays. However, little is known about Xen action in humans. This study determines whether exogenously administered Xen modulates gastric emptying and/or insulin secretion rates (ISRs) following meal ingestion. Fasted subjects with normal (NGT) or impaired (IGT) glucose tolerance and Type 2 diabetes mellitus (T2DM; n = 10-14 per group) ingested a liquid mixed meal plus acetaminophen (ACM; to assess gastric emptying) at time zero. On separate occasions, a primed-constant intravenous infusion of vehicle or Xen at 4 (Lo-Xen) or 12 (Hi-Xen) pmol · kg(-1) · min(-1) was administered from zero until 300 min. Some subjects with NGT received 30- and 90-min Hi-Xen infusions. Plasma ACM, glucose, insulin, C-peptide, glucagon, Xen, GIP, and glucagon-like peptide-1 (GLP-1) levels were measured and ISRs calculated. Areas under the curves were compared for treatment effects. Infusion with Hi-Xen, but not Lo-Xen, similarly delayed gastric emptying and reduced postprandial glucose levels in all groups. Infusions for 90 or 300 min, but not 30 min, were equally effective. Hi-Xen reduced plasma GLP-1, but not GIP, levels without altering the insulin secretory response to glucose. Intense staining for Xen receptors was detected on PGP9.5-positive nerve fibers in the longitudinal muscle of the human stomach. Thus Xen reduces gastric emptying in humans with and without T2DM, probably via a neural relay. Moreover, endogenous GLP-1 may not be a major enhancer of insulin secretion in healthy humans under physiological conditions.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Vaciamiento Gástrico/efectos de los fármacos , Hipoglucemiantes/uso terapéutico , Neurotensina/uso terapéutico , Periodo Posprandial , Adulto , Biomarcadores/sangre , Glucemia/metabolismo , Péptido C/sangre , Estudios Cruzados , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/fisiopatología , Esquema de Medicación , Femenino , Glucagón/sangre , Péptido 1 Similar al Glucagón/sangre , Humanos , Hipoglucemiantes/administración & dosificación , Infusiones Intravenosas , Insulina/sangre , Masculino , Persona de Mediana Edad , Missouri , Neurotensina/administración & dosificación , Receptores de Neurotensina/efectos de los fármacos , Receptores de Neurotensina/metabolismo , Factores de Tiempo , Resultado del Tratamiento
15.
Biomed Res Int ; 2013: 941764, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24000330

RESUMEN

Diabetic foot ulcers (DFUs) are characterized by an unsatisfactory inflammatory and migratory response. Skin inflammation involves the participation of many cells and particularly macrophages. Macrophage function can be modulated by neuropeptides; however, little is known regarding the role of neurotensin (NT) as a modulator of macrophages under inflammatory and hyperglycemic conditions. RAW 264.7 cells were maintained at 10/30 mM glucose, stimulated with/without LPS (1 µg/mL), and treated with/without NT(10 nM). The results show that NT did not affect macrophage viability. However, NT reverted the hyperglycemia-induced impair in the migration of macrophages. The expression of IL-6 and IL-1ß was significantly increased under 10 mM glucose in the presence of NT, while IL-1ß and IL-12 expression significantly decreased under inflammatory and hyperglycemic conditions. More importantly, high glucose modulates NT and NT receptor expression under normal and inflammatory conditions. These results highlight the effect of NT on cell migration, which is strongly impaired under hyperglycemic conditions, as well as its effect in decreasing the proinflammatory status of macrophages under hyperglycemic and inflammatory conditions. These findings provide new insights into the potential therapeutic role of NT in chronic wounds, such as in DFU, characterized by a deficit in the migratory properties of cells and a chronic proinflammatory status.


Asunto(s)
Citocinas/inmunología , Hiperglucemia/inmunología , Inflamación/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Neurotensina/administración & dosificación , Animales , Línea Celular , Movimiento Celular , Hiperglucemia/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Ratones
17.
Diabetes ; 61(7): 1793-800, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22522617

RESUMEN

Glucose-dependent insulinotropic polypeptide (GIP) potentiates glucose-stimulated insulin secretion (GSIS). This response is blunted in type 2 diabetes (T2DM). Xenin-25 is a 25-amino acid neurotensin-related peptide that amplifies GIP-mediated GSIS in hyperglycemic mice. This study determines if xenin-25 amplifies GIP-mediated GSIS in humans with normal glucose tolerance (NGT), impaired glucose tolerance (IGT), or T2DM. Each fasting subject received graded glucose infusions to progressively raise plasma glucose concentrations, along with vehicle alone, GIP, xenin-25, or GIP plus xenin-25. Plasma glucose, insulin, C-peptide, and glucagon levels and insulin secretion rates (ISRs) were determined. GIP amplified GSIS in all groups. Initially, this response was rapid, profound, transient, and essentially glucose independent. Thereafter, ISRs increased as a function of plasma glucose. Although magnitudes of insulin secretory responses to GIP were similar in all groups, ISRs were not restored to normal in subjects with IGT and T2DM. Xenin-25 alone had no effect on ISRs or plasma glucagon levels, but the combination of GIP plus xenin-25 transiently increased ISR and plasma glucagon levels in subjects with NGT and IGT but not T2DM. Since xenin-25 signaling to islets is mediated by a cholinergic relay, impaired islet responses in T2DM may reflect defective neuronal, rather than GIP, signaling.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Polipéptido Inhibidor Gástrico/administración & dosificación , Intolerancia a la Glucosa/sangre , Insulina/metabolismo , Neurotensina/administración & dosificación , Adulto , Glucemia/análisis , Péptido C/sangre , Femenino , Glucagón/sangre , Glucosa , Prueba de Tolerancia a la Glucosa , Hemoglobina Glucada/análisis , Humanos , Insulina/sangre , Secreción de Insulina , Masculino , Persona de Mediana Edad
18.
Brain Res ; 1433: 47-55, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-22138429

RESUMEN

Neurotensin (NT) is a 13 amino acid neuropeptide that is identical in mice and humans and is released from and acts upon a number of social brain regions. Recent work indicates NT neurotransmission may be altered in postpartum females and support the onset of some maternal behaviors. In a recent study, we highlighted how virgin and postpartum brains from mice selected for high offspring protection differ in response to injected NT (0.1 µg) relative to vehicle when examining c-Fos profiles across the CNS. In this companion study we use a second marker for brain activity, Egr-1, and evaluate multiple brain regions. Common significant increased Egr-1 responses to NT (relative to vehicle) were found in both female groups only in ventromedial hypothalamus. In lateral periaqueductal gray, virgin mice showed a significant Egr-1 increase with NT (relative to vehicle), but maternal mice did not. When comparing NT injections, virgin (relative to maternal) mice had significantly higher activity in five regions, including anterior hypothalamus, lateral hypothalamus, somatosensory cortex, paraventricular nucleus, and zona incerta; no regions were higher in maternal mice. A Principal Components Analysis was also used for data mining and in virgin mice, greater changes in activity hubs were found with NT (relative to vehicle) than for maternal mice. Overall, a lower sensitivity to NT in terms of Egr-1 reactivity in the maternal state was highlighted and this is consistent with previous c-Fos results. These findings provide additional insight into the mechanisms by which NT functions in the CNS.


Asunto(s)
Química Encefálica/fisiología , Encéfalo/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Conducta Materna/fisiología , Neurotensina/administración & dosificación , Periodo Posparto/metabolismo , Animales , Encéfalo/efectos de los fármacos , Química Encefálica/efectos de los fármacos , Femenino , Inyecciones Intraventriculares , Conducta Materna/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Periodo Posparto/efectos de los fármacos , Embarazo , Proteínas Proto-Oncogénicas c-fos/metabolismo
19.
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
20.
Peptides ; 31(1): 150-4, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19799951

RESUMEN

UNLABELLED: GIP is a major physiological component of the enteroinsular axis. Several researchers have pointed to a neural regulation of GIP secretion. We have previously studied the effect of intracerebroventricular (icv) infusion of insulin, NPY and bombesin in the regulation of GIP secretion. The aim of the present study is to evaluate a possible role of neurotensin in neural regulation of GIP secretion. Thirty-two adult dogs were used in this study. In a dose-response study (experiment 1) we used 3 different doses of neurotensin (25, 50 and 100microg) in a bolus icv infusion. In experiment 2 the animals received a bolus icv infusion of 50microg neurotensin and an equivalent amount of artificial cerebrospinal fluid (aCSF) at 1-week interval. In experiment 3 the animals received a continuous icv infusion of neurotensin at a constant rate of 1microg/kg/h and aCSF over a 3-h period, at 1-week interval. In experiment 4 the experiment of group 3 was repeated with a simultaneous intraduodenal infusion of a glucose load through the Mann-Bollman fistula. Plasma levels of glucose, insulin and GIP were assayed. RESULTS: Bolus and continuous icv infusion of neurotensin produced a significant increase in glucose, GIP and insulin levels. In the 4th experiment icv infusion of neurotensin produced a more prominent increase of glucose and insulin levels compare to infusion of aCSF. GIP levels were lower after infusion of neurotensin compared to aCSF. CONCLUSIONS: Our data suggest a differential effect of neurotensin on GIP secretion, dependent on the energy load.


Asunto(s)
Perros , Polipéptido Inhibidor Gástrico/metabolismo , Neurotensina , Animales , Glucemia/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Inyecciones Intraventriculares , Insulina/sangre , Neurotensina/administración & dosificación , Neurotensina/farmacología , Distribución Aleatoria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA