Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Endocrinol Metab ; 322(1): E10-E23, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34779255

RESUMEN

Cholecystokinin (CCK) increases core body temperature via CCK2 receptors when administered intracerebroventricularly (icv). The mechanisms of CCK-induced hyperthermia are unknown, and it is also unknown whether CCK contributes to the fever response to systemic inflammation. We studied the interaction between central CCK signaling and the cyclooxygenase (COX) pathway. Body temperature was measured in adult male Wistar rats pretreated with intraperitoneal infusion of the nonselective COX enzyme inhibitor metamizol (120 mg/kg) or a selective COX-2 inhibitor, meloxicam, or etoricoxib (10 mg/kg for both) and, 30 min later, treated with intracerebroventricular CCK (1.7 µg/kg). In separate experiments, CCK-induced neuronal activation (with and without COX inhibition) was studied in thermoregulation- and feeding-related nuclei with c-Fos immunohistochemistry. CCK increased body temperature by ∼0.4°C from 10 min postinfusion, which was attenuated by metamizol. CCK reduced the number of c-Fos-positive cells in the median preoptic area (by ∼70%) but increased it in the dorsal hypothalamic area and in the rostral raphe pallidus (by ∼50% in both); all these changes were completely blocked with metamizol. In contrast, CCK-induced satiety and neuronal activation in the ventromedial hypothalamus were not influenced by metamizol. CCK-induced hyperthermia was also completely blocked with both selective COX-2 inhibitors studied. Finally, the CCK2 receptor antagonist YM022 (10 µg/kg icv) attenuated the late phases of fever induced by bacterial lipopolysaccharide (10 µg/kg; intravenously). We conclude that centrally administered CCK causes hyperthermia through changes in the activity of "classical" thermoeffector pathways and that the activation of COX-2 is required for the development of this response.NEW & NOTEWORTHY An association between central cholecystokinin signaling and the cyclooxygenase-prostaglandin E pathway has been proposed but remained poorly understood. We show that the hyperthermic response to the central administration of cholecystokinin alters the neuronal activity within efferent thermoeffector pathways and that these effects are fully blocked by the inhibition of cyclooxygenase. We also show that the activation of cyclooxygenase-2 is required for the hyperthermic effect of cholecystokinin and that cholecystokinin is a modulator of endotoxin-induced fever.


Asunto(s)
Temperatura Corporal/efectos de los fármacos , Colecistoquinina/administración & dosificación , Ciclooxigenasa 2/metabolismo , Hipertermia/inducido químicamente , Hipertermia/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Anorexia/inducido químicamente , Benzodiazepinas/administración & dosificación , Regulación de la Temperatura Corporal/efectos de los fármacos , Colecistoquinina/efectos adversos , Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Fiebre/inducido químicamente , Fiebre/tratamiento farmacológico , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Lipopolisacáridos/efectos adversos , Masculino , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar , Receptor de Colecistoquinina B/antagonistas & inhibidores , Resultado del Tratamiento
2.
Mol Pharm ; 18(6): 2285-2297, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33998814

RESUMEN

Cholecystokinin-2 receptor (CCK2R) has been proven to be a specific biomarker for colorectal malignancies. Immunotoxins are a valuable class of immunotherapy agents consisting of a targeting element and a bacterial or plant toxin. Previous work demonstrated that targeting CCK2R is a good therapeutic strategy for the treatment of colorectal cancer (CRC). In the present study, we developed a new version of CCK2R-targeting immunotoxin GD9P using a targeted peptide, GD9, as the binding motif and a truncated Pseudomonas exotoxin A (PE38) as the cytokiller. BALB/c nude mice were treated with different doses of GD9P, and pharmacodynamics, pharmacokinetic, and toxicological data were obtained throughout this study. Compared to the parental immunotoxin rCCK8PE38, GD9P exhibited about 1.5-fold yield, higher fluorescence intensity, and increased antitumor activity against human CRC in vitro and in vivo. The IC50 values of GD9P in vitro ranged from 1.61 to 4.55 nM. Pharmacokinetic studies were conducted in mice with a T1/2 of 69.315 min. When tumor-bearing nude mice were treated with GD9P at doses ≥2 mg/kg for five doses, a rapid shrinkage in tumor volume and, in some cases, complete remission was observed. A preliminary safety evaluation demonstrated a good safety profile of GD9P as a Pseudomonas exotoxin A-based immunotherapy. The therapy in combination with oxaliplatin can increase the antitumor efficacy and reduce the toxic side effects caused by chemotherapy. In conclusion, the data support the use of GD9P as a promising immunotherapy targeting CCK2R-expressing colorectal malignancies.


Asunto(s)
ADP Ribosa Transferasas/farmacología , Antineoplásicos/farmacología , Toxinas Bacterianas/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Exotoxinas/farmacología , Receptor de Colecistoquinina B/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/farmacología , Factores de Virulencia/farmacología , ADP Ribosa Transferasas/genética , ADP Ribosa Transferasas/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Toxinas Bacterianas/genética , Toxinas Bacterianas/uso terapéutico , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Exotoxinas/genética , Exotoxinas/uso terapéutico , Humanos , Ratones , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/uso terapéutico , Distribución Tisular , Pruebas de Toxicidad Aguda , Factores de Virulencia/genética , Factores de Virulencia/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Exotoxina A de Pseudomonas aeruginosa
3.
Biol Pharm Bull ; 44(5): 701-706, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33952826

RESUMEN

We previously reported that exposure of human colon adenocarcinoma (Caco-2) cells to the bitter substance phenylthiocarbamide (PTC) rapidly enhanced the transport function of P-glycoprotein (P-gp). In this study, we investigated the short-term effect of etoposide, another bitter-tasting P-gp substrate, on P-gp transport function in the same cell line. We found that etoposide exposure significantly increased both the P-gp protein level in the plasma membrane fraction and the efflux rate of rhodamine123 (Rho123) in Caco-2 cells within 10 min. The efflux ratio (ratio of the apparent permeability coefficient in the basal-to-apical direction to that in the apical-to-basal direction) of Rho123 in etoposide-treated cells was also significantly increased compared with the control. These results indicated that etoposide rapidly enhances P-gp function in Caco-2 cells. In contrast, P-gp expression in whole cells at both the mRNA and protein level was unchanged by etoposide exposure, compared with the levels in non-treated cells. Furthermore, etoposide increased the level of phosphorylated ezrin, radixin and moesin (P-ERM) proteins in the plasma membrane fraction of Caco-2 cells within 10 min. P-gp functional changes were blocked by YM022, an inhibitor of cholecystokinin (CCK) receptor. These results suggest that etoposide induces release of CCK, causing activation of the CCK receptor followed by phosphorylation of ERM proteins, which recruit intracellular P-gp for trafficking to the gastrointestinal membrane, thereby increasing the functional activity of P-gp.


Asunto(s)
Etopósido/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Benzodiazepinas/farmacología , Células CACO-2 , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Colecistoquinina/metabolismo , Proteínas del Citoesqueleto/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Fosforilación/efectos de los fármacos , Receptor de Colecistoquinina B/antagonistas & inhibidores , Receptor de Colecistoquinina B/metabolismo
4.
Molecules ; 26(4)2021 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-33572353

RESUMEN

The cholecystokinin-2 receptor (CCK-2R) is overexpressed in several human cancers but displays limited expression in normal tissues. For this reason, it is a suitable target for developing specific radiotracers. In this study, a nastorazepide-based ligand functionalized with a 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) chelator (IP-001) was synthesized and labelled with indium-111. The radiolabeling process yielded >95% with a molar activity of 10 MBq/nmol and a radiochemical purity of >98%. Stability studies have shown a remarkable resistance to degradation (>93%) within 120 h of incubation in human blood. The in vitro uptake of [111In]In-IP-001 was assessed for up to 24 h on a high CCK-2R-expressing tumor cell line (A549) showing maximal accumulation after 4 h of incubation. Biodistribution and single photon emission tomography (SPECT)/CT imaging were evaluated on BALB/c nude mice bearing A549 xenograft tumors. Implanted tumors could be clearly visualized after only 4 h post injection (2.36 ± 0.26% ID/cc), although a high amount of radiotracer was also found in the liver, kidneys, and spleen (8.25 ± 2.21%, 6.99 ± 0.97%, and 3.88 ± 0.36% ID/cc, respectively). Clearance was slow by both hepatobiliary and renal excretion. Tumor retention persisted for up to 24 h, with the tumor to organs ratio increasing over-time and ending with a tumor uptake (1.52 ± 0.71% ID/cc) comparable to liver and kidneys.


Asunto(s)
Benzodiazepinas/química , Radioisótopos de Indio/farmacocinética , Neoplasias Pulmonares/metabolismo , Radiofármacos/farmacocinética , Receptor de Colecistoquinina B/antagonistas & inhibidores , Animales , Apoptosis , Proliferación Celular , Femenino , Humanos , Radioisótopos de Indio/administración & dosificación , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/radioterapia , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Radiofármacos/administración & dosificación , Receptor de Colecistoquinina B/metabolismo , Distribución Tisular , Tomografía Computarizada de Emisión de Fotón Único , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Dig Dis Sci ; 65(1): 189-203, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31297627

RESUMEN

BACKGROUND AND AIMS: Nonalcoholic steatohepatitis (NASH) is a common inflammatory liver condition that may lead to cirrhosis and hepatocellular carcinoma (HCC). Risk factors for NASH include a saturated fat diet, altered lipid metabolism, and genetic and epigenetic factors, including microRNAs. Serum levels of cholecystokinin (CCK) are elevated in mice and humans that consume a high-saturated fat diet. CCK receptors (CCK-Rs) have been reported on fibroblasts which when activated can induce fibrosis; however, their role in hepatic fibrosis remains unknown. We hypothesized that elevated levels of CCK acting on the CCK-Rs play a role in the development of NASH and in NASH-associated HCC. METHODS: We performed a NASH Prevention study and Reversal study in mice fed a saturated fat 75% choline-deficient-ethionine-supplemented (CDE) diet for 12 or 18 weeks. In each study, half of the mice received untreated drinking water, while the other half received water supplemented with the CCK-R antagonist proglumide. CCK-R expression was evaluated in mouse liver and murine HCC cells. RESULTS: CCK receptor antagonist treatment not only prevented NASH but also reversed hepatic inflammation, fibrosis, and steatosis and normalized hepatic transaminases after NASH was established. Thirty-five percent of the mice on the CDE diet developed HCC compared with none in the proglumide-treated group. We found that CCK-BR expression was markedly upregulated in mouse CDE liver and HCC cells compared with normal hepatic parenchymal cells, and this expression was epigenetically regulated by microRNA-148a. CONCLUSION: These results support the novel role of CCK receptors in the pathogenesis of NASH and HCC.


Asunto(s)
Carcinoma Hepatocelular/prevención & control , Antagonistas de Hormonas/farmacología , Neoplasias Hepáticas/prevención & control , Hígado/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Proglumida/farmacología , Receptor de Colecistoquinina B/antagonistas & inhibidores , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Deficiencia de Colina/complicaciones , Modelos Animales de Enfermedad , Epigénesis Genética , Etionina , Femenino , Regulación Neoplásica de la Expresión Génica , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Receptor de Colecistoquinina B/genética , Receptor de Colecistoquinina B/metabolismo , Transducción de Señal
6.
Mol Carcinog ; 58(10): 1908-1918, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31313401

RESUMEN

Gastrin signaling mediated through cholecystokinin-2 receptor (CCK2R) and its downstream molecules is altered in pancreatic cancer. CCK2R antagonists, YF476 (netazepide) and JNJ-26070109, were tested systematically for their effect on pancreatic intraepithelial neoplasia (PanIN) progression to pancreatic ductal adenocarcinoma (PDAC) in KrasG12D mice. After dose selection using wild-type mice, six-week-old p48Cre/+ -LSL-KrasG12D (22-24 per group) genetically engineered mice (GEM) were fed AIN-76A diets containing 0, 250, or 500 ppm JNJ-26070109 or YF-476 for 38 weeks. At termination, pancreata were collected, weighed, and evaluated for PanINs and PDAC. Results demonstrated that control-diet-fed mice showed 69% (males) and 33% (females) incidence of PDAC. Administration of low and high dose JNJ-26070109 inhibited the incidence of PDAC by 88% and 71% (P < .004) in male mice and by 100% and 24% (P > .05) in female mice, respectively. Low and high dose YF476 inhibited the incidence of PDAC by 74% (P < .02) and 69% (P < .02) in male mice and by 45% and 33% (P > .05) in female mice, respectively. Further, transcriptome analysis showed downregulation of Cldn1, Sstr1, Apod, Gkn1, Siglech, Cyp2c44, Bnc1, Fmo2, 623169, Kcne4, Slc27a6, Cma1, Rho GTPase activating protein 18, and Gpr85 genes in JNJ-26070109-treated mice compared with untreated mice. YF476-treated mouse pancreas showed downregulation of Riks, Zpbp, Ntf3, Lrrn4, Aass, Skint3, Kcnb1, Dgkb, Ddx60, and Aspn gene expressions compared with untreated mouse pancreas. Overall, JNJ-26070109 showed better chemopreventive efficacy than YF476. However, caution is recommended when selecting doses, as the agents appeared to exhibit gender-specific effects.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Receptor de Colecistoquinina B/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Benzodiazepinonas/farmacología , Carcinoma in Situ , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Quimioprevención/métodos , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Compuestos de Fenilurea/farmacología , Proteínas Proto-Oncogénicas p21(ras)/genética , Quinoxalinas/farmacología , Receptor de Colecistoquinina B/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología
7.
Artículo en Inglés | MEDLINE | ID: mdl-31051262

RESUMEN

Urocortin-3 (UCN3) as a brain-gut peptide inhibits food intake of animal, but the underlying mechanism is not clear. To explore the appetite mechanism about the action of UCN3 in fish, intraperitoneal injection of UCN3 with CCK8, Lorglumide (CCK1R antagonist) or LY225910 (CCK2R antagonist) were conducted. Siberian sturgeon administrated with UCN3 and CCK8 showed a drastic reduction in food intake. The anorectic effect of UCN3 was significantly blocked by LY225910, but not affected by Lorglumide. Furthermore, LY225910 could effectively reverse appetite factor mRNA expressions, including cck, pyy, cart, npy, ucn3, apelin and nucb2 in the whole brain, stomach and intestinum valvula, but Lorglumide could only partially reverse these effects, suggesting the anorectic effect of UCN3 may be primarily mediated CCK2R in Siberian sturgeon. This study indicates for the first time in fish that UCN3 may inhibit food intake in coordination with CCK and CCK2R.


Asunto(s)
Ingestión de Alimentos/genética , Peces/fisiología , Receptor de Colecistoquinina B/genética , Urocortinas/genética , Animales , Peces/genética , Proglumida/análogos & derivados , Proglumida/farmacología , Receptor de Colecistoquinina B/antagonistas & inhibidores , Urocortinas/antagonistas & inhibidores
8.
Eur J Clin Pharmacol ; 73(2): 129-139, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27796466

RESUMEN

PURPOSE: Hypergastrinaemia induced by proton pump inhibitor (PPI) therapy may cause ECL-cell and parietal-cell hyperplasia and rebound hyperacidity and dyspepsia after PPI withdrawal. The aim of the study was to assess the effect of different dosage-regimens of netazepide, a gastrin/CCK2 receptor antagonist, on PPI-induced hypergastrinaemia and elevated chromogranin A (CgA). METHODS: Six groups of eight healthy subjects participated in a randomised, double-blind study of esomeprazole 40 mg daily for 28 days, in combination with netazepide 1, 5 or 25 mg or placebo, daily, during the last 14 days of esomeprazole or during 14 days after treatment withdrawal. Fasting serum gastrin and plasma CgA were measured during treatment and after withdrawal, as biomarkers of acid suppression and ECL-cell activity, respectively. Dyspepsia was monitored throughout the study. RESULTS: Esomeprazole increased gastrin and CgA. Netazepide increased gastrin, but not CgA, and inhibited dose dependently the CgA response to esomeprazole. Gastrin and CgA returned to baseline within 2-3 days of esomeprazole withdrawal; netazepide did not shorten that time. There was no rebound dyspepsia after esomeprazole withdrawal. CONCLUSIONS: Esomeprazole and netazepide each increase gastrin, consistent with a secondary effect of gastric acid suppression, but by different mechanisms. Esomeprazole-induced hypergastrinaemia stimulates ECL cells and thereby increases CgA. Netazepide-induced hypergastrinaemia does not increase CgA, because netazepide blocks gastrin/CCK2 receptors on ECL cells. Co-administration of netazepide 5 mg abolishes the effect of esomeprazole-induced hypergastrinaemia on ECL cells. The quick return to baseline of gastrin and CgA and absence of dyspepsia after esomeprazole withdrawal do not support the concept of rebound hyperacidity.


Asunto(s)
Antiulcerosos/efectos adversos , Benzodiazepinonas/uso terapéutico , Esomeprazol/efectos adversos , Gastrinas/sangre , Compuestos de Fenilurea/uso terapéutico , Inhibidores de la Bomba de Protones/efectos adversos , Receptor de Colecistoquinina B/antagonistas & inhibidores , Adulto , Anciano , Cromogranina A/sangre , Método Doble Ciego , Dispepsia/prevención & control , Femenino , Determinación de la Acidez Gástrica , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
9.
J Neurosci ; 35(38): 13160-70, 2015 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-26400945

RESUMEN

Somatodendritically released peptides alter synaptic function through a variety of mechanisms, including autocrine actions that liberate retrograde transmitters. Cholecystokinin (CCK) is a neuropeptide expressed in neurons in the dorsomedial hypothalamic nucleus (DMH), a region implicated in satiety and stress. There are clear demonstrations that exogenous CCK modulates food intake and neuropeptide expression in the DMH, but there is no information on how endogenous CCK alters synaptic properties. Here, we provide the first report of somatodendritic release of CCK in the brain in male Sprague Dawley rats. CCK is released from DMH neurons in response to repeated postsynaptic depolarizations, and acts in an autocrine fashion on CCK2 receptors to enhance postsynaptic NMDA receptor function and liberate the retrograde transmitter, nitric oxide (NO). NO subsequently acts presynaptically to enhance GABA release through a soluble guanylate cyclase-mediated pathway. These data provide the first demonstration of synaptic actions of somatodendritically released CCK in the hypothalamus and reveal a new form of retrograde plasticity, depolarization-induced potentiation of inhibition. Significance statement: Somatodendritic signaling using endocannabinoids or nitric oxide to alter the efficacy of afferent transmission is well established. Despite early convincing evidence for somatodendritic release of neurohypophysial peptides in the hypothalamus, there is only limited evidence for this mode of release for other peptides. Here, we provide the first evidence for somatodendritic release of the satiety peptide cholecystokinin (CCK) in the brain. We also reveal a new form of synaptic plasticity in which postsynaptic depolarization results in enhancement of inhibition through the somatodendritic release of CCK.


Asunto(s)
Colecistoquinina/metabolismo , Núcleo Hipotalámico Dorsomedial/citología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo , Animales , Animales Recién Nacidos , Colecistoquinina/farmacología , GABAérgicos/farmacología , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacología , Técnicas In Vitro , Masculino , Técnicas de Placa-Clamp , Péptidos/farmacología , Proglumida/análogos & derivados , Proglumida/farmacología , Quinazolinonas/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Colecistoquinina B/antagonistas & inhibidores , Receptor de Colecistoquinina B/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína 25 Asociada a Sinaptosomas/antagonistas & inhibidores , Proteína 25 Asociada a Sinaptosomas/metabolismo , Tionucleótidos/farmacología , Ácido gamma-Aminobutírico/farmacología
10.
Mol Pain ; 122016.
Artículo en Inglés | MEDLINE | ID: mdl-27573516

RESUMEN

Burn injury is a cause of significant mortality and morbidity worldwide and is frequently associated with severe and long-lasting pain that remains difficult to manage throughout recovery. We characterised a mouse model of burn-induced pain using pharmacological and transcriptomic approaches. Mechanical allodynia elicited by burn injury was partially reversed by meloxicam (5 mg/kg), gabapentin (100 mg/kg) and oxycodone (3 and 10 mg/kg), while thermal allodynia and gait abnormalities were only significantly improved by amitriptyline (3 mg/kg) and oxycodone (10 mg/kg). The need for relatively high opioid doses to elicit analgesia suggested a degree of opioid resistance, similar to that shown clinically in burn patients. We thus assessed the gene expression changes in dorsal root ganglion neurons and pathophysiological mechanisms underpinning burn injury-induced pain using a transcriptomic approach. Burn injury was associated with significantly increased expression of genes associated with axon guidance, neuropeptide signalling, behavioural defence response and extracellular signalling, confirming a mixed neuropathic and inflammatory aetiology. Notably, among the pain-related genes that were upregulated post-injury was the cholecystokinin 2 receptor (Cckbr), a G protein-coupled receptor known as a pain target involved in reducing opioid effectiveness. Indeed, the clinically used cholecystokinin receptor antagonist proglumide (30 mg/kg) was effective at reversing mechanical allodynia, with additional analgesia evident in combination with low-dose oxycodone (1 mg/kg), including significant reversal of thermal allodynia. These findings highlight the complex pathophysiological mechanisms underpinning burn injury-induced pain and suggest that cholecystokinin-2 receptor antagonists may be useful clinically as adjuvants to decrease opioid requirements and improve analgesic management.


Asunto(s)
Trastornos Neurológicos de la Marcha/etiología , Regulación de la Expresión Génica/fisiología , Hiperalgesia/etiología , Dolor , Receptor de Colecistoquinina B/metabolismo , Transcriptoma , Aminas/farmacología , Aminas/uso terapéutico , Amitriptilina/farmacología , Amitriptilina/uso terapéutico , Animales , Ácidos Ciclohexanocarboxílicos/farmacología , Ácidos Ciclohexanocarboxílicos/uso terapéutico , Modelos Animales de Enfermedad , Gabapentina , Trastornos Neurológicos de la Marcha/tratamiento farmacológico , Ganglios Espinales/citología , Regulación de la Expresión Génica/efectos de los fármacos , Hiperalgesia/tratamiento farmacológico , Masculino , Metacarpo/patología , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Oxicodona/farmacología , Oxicodona/uso terapéutico , Dolor/complicaciones , Dolor/tratamiento farmacológico , Dolor/metabolismo , Umbral del Dolor/efectos de los fármacos , Proglumida/farmacología , Proglumida/uso terapéutico , Receptor de Colecistoquinina B/antagonistas & inhibidores , Receptor de Colecistoquinina B/genética , Células Receptoras Sensoriales/efectos de los fármacos , Soporte de Peso/fisiología , Ácido gamma-Aminobutírico/farmacología , Ácido gamma-Aminobutírico/uso terapéutico
11.
Scand J Gastroenterol ; 51(11): 1398-405, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27309188

RESUMEN

OBJECTIVES: To review the presentation, treatment and outcome of patients with type 1 gastric carcinoid tumours. MATERIAL AND METHODS: We retrospectively reviewed medical records and re-evaluated histopathological specimens of 26 patients with type 1 gastric carcinoids treated at a single tertiary referral centre from 1993 to 2013, with median time of follow-up 52.5 months (IQR 90.8). RESULTS: Seven patients (27%) had single tumours and 19 patients (73%) multiple tumours at the time of diagnosis. The median number of tumours and median diameter of largest tumour were 2.5 (IQR 3.2) and 6.0 mm (IQR 9.5) respectively. Median serum gastrin was 321.0 pmol/l (IQR 604.0) and median serum chromogranin A 7.7 nmol/l (IQR 5.3). Three patients had metastatic disease at the time of diagnosis and two developed metastases during follow-up. Patients with metastatic disease had larger primary tumours than the others (20.0 mm (IQR 28.5) vs. 5.0 mm (IQR 5.5), p = 0.04). There was a positive correlation between age and tumour size (r = 0.44, p = 0.03) and between serum chromogranin A and serum gastrin at diagnosis (r = 0.76, p = 0.001). Patients were either treated with surgery (n = 8 (31%)), a long-acting somatostatin analogue and/or gastrin receptor antagonist (n = 10 (39%)) for a period of time, or were observed without treatment (n = 8 (31%) with close endoscopic follow up. CONCLUSIONS: Although gastric carcinoids have an overall good prognosis, a significant proportion develops metastatic disease. As partial and total gastrectomy is associated with major side effects, treatment with long-acting a somatostatin analogue or gastrin antagonist should be considered.


Asunto(s)
Tumor Carcinoide/patología , Tumor Carcinoide/terapia , Células Similares a las Enterocromafines/patología , Neoplasias Gástricas/patología , Neoplasias Gástricas/terapia , Anciano , Antineoplásicos Hormonales/uso terapéutico , Tumor Carcinoide/mortalidad , Cromogranina A/sangre , Comorbilidad , Femenino , Estudios de Seguimiento , Gastrectomía , Mucosa Gástrica/patología , Gastrinas/sangre , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Noruega , Octreótido/uso terapéutico , Receptor de Colecistoquinina B/antagonistas & inhibidores , Receptor de Colecistoquinina B/uso terapéutico , Estudios Retrospectivos , Somatostatina/análogos & derivados , Somatostatina/uso terapéutico , Neoplasias Gástricas/mortalidad , Centros de Atención Terciaria , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
12.
Arch Pharm (Weinheim) ; 349(6): 456-65, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27142294

RESUMEN

4-Amino-2(5H)-furanones were synthesized in high yields over two synthetic steps from readily available mucochloric acid. These 5-alkyloxy-4-amino-2(5H)-furanones were screened in a ([125]) I-CCK-8 radioligand receptor binding assay for CCK2 affinity and novel active ligands in the nanomolar range were identified. SAR was optimized leading to the cyclohexyl derivative 25 with an IC50 of 27 nM. Furanone 18 was obtained as a stable crystalline material with an IC50 of 85 nM, but had a higher CCK2 selectivity. It was subsequently tested in the isolated guinea pig ileum assay with sulfated CCK8 , and the CCK antagonizing properties of the ligand were confirmed. The CCK2 selective antagonist 18 was found to potentiate analgesia in the tail flick assay in mice, for the strong opiate morphine, the partial opiate agonist tramadol and the tricyclic antidepressant desimipramine.


Asunto(s)
Furanos/química , Furanos/farmacología , Dimensión del Dolor/efectos de los fármacos , Receptor de Colecistoquinina B/antagonistas & inhibidores , Animales , Desipramina/farmacología , Sinergismo Farmacológico , Furanos/síntesis química , Cobayas , Ratones , Morfina/farmacología , Ensayo de Unión Radioligante , Relación Estructura-Actividad , Tramadol/farmacología
13.
J Neurosci ; 34(11): 3878-87, 2014 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-24623766

RESUMEN

Decreased medial prefrontal cortex (mPFC) neuronal activity is associated with social defeat-induced depression- and anxiety-like behaviors in mice. However, the molecular mechanisms underlying the decreased mPFC activity and its prodepressant role remain unknown. We show here that induction of the transcription factor ΔFosB in mPFC, specifically in the prelimbic (PrL) area, mediates susceptibility to stress. ΔFosB induction in PrL occurred selectively in susceptible mice after chronic social defeat stress, and overexpression of ΔFosB in this region, but not in the nearby infralimbic (IL) area, enhanced stress susceptibility. ΔFosB produced these effects partly through induction of the cholecystokinin (CCK)-B receptor: CCKB blockade in mPFC induces a resilient phenotype, whereas CCK administration into mPFC mimics the anxiogenic- and depressant-like effects of social stress. We previously found that optogenetic stimulation of mPFC neurons in susceptible mice reverses several behavioral abnormalities seen after chronic social defeat stress. Therefore, we hypothesized that optogenetic stimulation of cortical projections would rescue the pathological effects of CCK in mPFC. After CCK infusion in mPFC, we optogenetically stimulated mPFC projections to basolateral amygdala or nucleus accumbens, two subcortical structures involved in mood regulation. Stimulation of corticoamygdala projections blocked the anxiogenic effect of CCK, although no effect was observed on other symptoms of social defeat. Conversely, stimulation of corticoaccumbens projections reversed CCK-induced social avoidance and sucrose preference deficits but not anxiogenic-like effects. Together, these results indicate that social stress-induced behavioral deficits are mediated partly by molecular adaptations in mPFC involving ΔFosB and CCK through cortical projections to distinct subcortical targets.


Asunto(s)
Trastornos de Ansiedad/fisiopatología , Colecistoquinina/fisiología , Trastorno Depresivo/fisiopatología , Corteza Prefrontal/fisiología , Proteínas Proto-Oncogénicas c-fos/fisiología , Receptor de Colecistoquinina B/fisiología , Animales , Ansiolíticos/farmacología , Trastornos de Ansiedad/patología , Mapeo Encefálico , Enfermedad Crónica , Trastorno Depresivo/patología , Indoles/farmacología , Sistema Límbico/citología , Sistema Límbico/efectos de los fármacos , Sistema Límbico/fisiología , Masculino , Meglumina/análogos & derivados , Meglumina/farmacología , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/fisiología , Corteza Prefrontal/citología , Corteza Prefrontal/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/genética , Receptor de Colecistoquinina B/antagonistas & inhibidores , Receptor de Colecistoquinina B/genética , Predominio Social , Estrés Psicológico/patología , Estrés Psicológico/fisiopatología
14.
Br J Clin Pharmacol ; 79(5): 744-55, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25335860

RESUMEN

AIMS: To compare gastric acid suppression by netazepide, a gastrin/CCK2 receptor antagonist, with that by a proton pump inhibitor (PPI), and to determine if netazepide can prevent the trophic effects of PPI-induced hypergastrinaemia. METHODS: Thirty healthy subjects completed a double-blind, randomized, parallel group trial of oral netazepide and rabeprazole, alone and combined, once daily for 6 weeks. Primary end points were: basal and pentagastrin-stimulated gastric acid and 24 h circulating gastrin and chromogranin A (CgA) at baseline, start and end of treatment, gastric biopsies at baseline and end of treatment and basal and pentagastrin-stimulated gastric acid and dyspepsia questionnaire after treatment withdrawal. RESULTS: All treatments similarly inhibited pentagastrin-stimulated gastric acid secretion. All treatments increased serum gastrin, but the combination and rabeprazole did so more than netazepide alone. The combination also reduced basal acid secretion. Rabeprazole increased plasma CgA, whereas netazepide and the combination reduced it. None of the biopsies showed enterochromaffin-like (ECL) cell hyperplasia. Withdrawal of treatments led neither to rebound hyperacidity nor dyspepsia. CONCLUSIONS: Netazepide suppressed pentagastrin-stimulated gastric acid secretion as effectively as did rabeprazole. The reduction in basal acid secretion and greater increase in serum gastrin by the combination is consistent with more effective acid suppression. Despite our failure to show rabeprazole-induced ECL cell hyperplasia and rebound hyperacidity, the increase in plasma CgA after rabeprazole is consistent with a trophic effect on ECL cells, which netazepide prevented. Thus, netazepide is a potential treatment for the trophic effects of hypergastrinaemia and, with or without a PPI, is a potential treatment for acid-related conditions.


Asunto(s)
Benzodiazepinonas/farmacología , Ácido Gástrico/metabolismo , Compuestos de Fenilurea/farmacología , Rabeprazol/farmacología , Receptor de Colecistoquinina B/antagonistas & inhibidores , Estómago , Adulto , Anciano , Benzodiazepinonas/administración & dosificación , Benzodiazepinonas/efectos adversos , Benzodiazepinonas/sangre , Método Doble Ciego , Quimioterapia Combinada , Femenino , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/metabolismo , Mucosa Gástrica/ultraestructura , Gastrinas/sangre , Voluntarios Sanos , Humanos , Hiperplasia/inducido químicamente , Hiperplasia/prevención & control , Masculino , Persona de Mediana Edad , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/efectos adversos , Compuestos de Fenilurea/sangre , Rabeprazol/administración & dosificación , Rabeprazol/efectos adversos , Rabeprazol/sangre , Estómago/efectos de los fármacos , Estómago/patología , Adulto Joven
15.
Scand J Gastroenterol ; 50(5): 550-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25665655

RESUMEN

OBJECTIVE: Gastric carcinoids (neuroendocrine tumors) arise from enterochromaffin-like cells in the gastric mucosa. Most are caused by hypergastrinemia. The objectives were to determine if their prevalence in Europe, USA and Japan meets the criteria for an orphan disease and to justify treatment with a gastrin/CCK2 receptor antagonist. METHODS: We obtained data from European and USA cancer registries, and searched PubMed. RESULTS: Prevalence per 10,000 population obtained from cancer registries was: median 0.32 (range 0.09-0.92) for Europe; and 0.17 for the USA, equivalent to 4812 for the whole population. A PubMed search for gastric carcinoids yielded prevalence for Japan only, which was 0.05 per 10,000 population, equivalent to 665 for the entire population. A further search for gastric carcinoids in patients with pernicious anemia (PA) or autoimmune chronic atrophic gastritis (CAG), two presentations of about 80% of gastric carcinoids, produced prevalence rates of 5.2-11%. Prevalence of PA itself was 0.12-1.9%. Data on CAG epidemiology were sparse. CONCLUSION: Prevalence of gastric carcinoids varied widely. All sources probably underestimate prevalence. However, prevalence was below the limits required for recognition by drug regulatory authorities as an orphan disease: 5 per 10,000 population of Europe; 200,000 for the whole population of the USA; and 50,000 for the whole population of Japan. Because gastric carcinoids are an orphan disease, and nonclinical and healthy volunteer studies support treatment with netazepide, a gastrin/CCK2 antagonist, netazepide has been designated an orphan medicinal product in Europe and the USA for development as targeted treatment for gastric carcinoids.


Asunto(s)
Benzodiazepinonas/uso terapéutico , Tumor Carcinoide/tratamiento farmacológico , Tumor Carcinoide/epidemiología , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/epidemiología , Compuestos de Fenilurea/uso terapéutico , Receptor de Colecistoquinina B/antagonistas & inhibidores , Anciano , Anciano de 80 o más Años , Anemia Perniciosa/epidemiología , Células Similares a las Enterocromafines/metabolismo , Europa (Continente)/epidemiología , Femenino , Mucosa Gástrica/metabolismo , Gastritis Atrófica/epidemiología , Humanos , Japón/epidemiología , Masculino , Prevalencia , Enfermedades Raras , Distribución por Sexo , Estados Unidos/epidemiología
16.
J Am Chem Soc ; 135(7): 2560-73, 2013 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-23323542

RESUMEN

Seven-transmembrane receptors (7TMRs), also termed G protein-coupled receptors (GPCRs), form the largest class of cell surface membrane receptors, involving several hundred members in the human genome. Nearly 30% of marketed pharmacological agents target 7TMRs. 7TMRs adopt multiple conformations upon agonist binding. Biased agonists, in contrast to non-biased agonists, are believed to stabilize conformations preferentially activating either G-protein- or ß-arrestin-dependent signaling pathways. However, proof that cognate conformations of receptors display structural differences within their binding site where biased agonism initiates, are still lacking. Here, we show that a non-biased agonist, cholecystokinin (CCK) induces conformational states of the CCK2R activating Gq-protein-dependent pathway (CCK2R(G)) or recruiting ß-arrestin2 (CCK2R(ß)) that are pharmacologically and structurally distinct. Two structurally unrelated antagonists competitively inhibited both pathways. A third ligand (GV150013X) acted as a high affinity competitive antagonist on CCK2R(G) but was nearly inefficient as inhibitor of CCK2R(ß). Several structural elements on both GV150013X and in CCK2R binding cavity, which hinder binding of GV150013X only to the CCK2R(ß) were identified. At last, proximity between two conserved amino acids from transmembrane helices 3 and 7 interacting through sulfur-aromatic interaction was shown to be crucial for selective stabilization of the CCK2R(ß) state. These data establish structural evidence for distinct conformations of a 7TMR associated with ß-arrestin-2 recruitment or G-protein coupling and validate relevance of the design of biased ligands able to selectively target each functional conformation of 7TMRs.


Asunto(s)
Arrestinas/química , Receptor de Colecistoquinina B/química , Fosfolipasas de Tipo C/metabolismo , Adamantano/análogos & derivados , Adamantano/farmacología , Sitios de Unión , Activación Enzimática/efectos de los fármacos , Humanos , Microscopía Confocal , Modelos Moleculares , Estructura Molecular , Mutación , Compuestos de Fenilurea/farmacología , Conformación Proteica , Receptor de Colecistoquinina B/antagonistas & inhibidores , Receptor de Colecistoquinina B/genética , Transducción de Señal , Fosfolipasas de Tipo C/química , Regulación hacia Arriba , Arrestina beta 2 , beta-Arrestinas
17.
Gastroenterology ; 142(4): 834-843.e3, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22245844

RESUMEN

BACKGROUND & AIMS: The duodenum senses nutrients to maintain energy and glucose homeostasis, but little is known about the signaling and neuronal mechanisms involved. We tested whether duodenal activation of adenosine 3',5'-cyclic monophosphate (cAMP)-dependent protein kinase A (PKA) is sufficient and necessary for cholecystokinin (CCK) signaling to trigger vagal afferent firing and regulate glucose production. METHODS: In rats, we selectively activated duodenal PKA and evaluated changes in glucose kinetics during the pancreatic (basal insulin) pancreatic clamps and vagal afferent firing. The requirement of duodenal PKA signaling in glucose regulation was evaluated by inhibiting duodenal activation of PKA in the presence of infusion of the intraduodenal PKA agonist (Sp-cAMPS) or CCK1 receptor agonist (CCK-8). We also assessed the involvement of a neuronal network and the metabolic impact of duodenal PKA activation in rats placed on high-fat diets. RESULTS: Intraduodenal infusion of Sp-cAMPS activated duodenal PKA and lowered glucose production, in association with increased vagal afferent firing in control rats. The metabolic and neuronal effects of duodenal Sp-cAMPS were negated by coinfusion with either the PKA inhibitor H89 or Rp-CAMPS. The metabolic effect was also negated by coinfusion with tetracaine, molecular and pharmacologic inhibition of NR1-containing N-methyl-d-aspartate (NMDA) receptors within the dorsal vagal complex, or hepatic vagotomy in rats. Inhibition of duodenal PKA blocked the ability of duodenal CCK-8 to reduce glucose production in control rats, whereas duodenal Sp-cAMPS bypassed duodenal CCK resistance and activated duodenal PKA and lowered glucose production in rats on high-fat diets. CONCLUSIONS: We identified a neural glucoregulatory function of duodenal PKA signaling.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Duodeno/enzimología , Duodeno/inervación , Glucosa/metabolismo , Hígado/inervación , Hígado/metabolismo , Nervio Vago/fisiología , Animales , Colecistoquinina/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Dieta Alta en Grasa , Duodeno/efectos de los fármacos , Activación Enzimática , Activadores de Enzimas/farmacología , Técnica de Clampeo de la Glucosa , Homeostasis , Antagonistas de Hormonas/farmacología , Masculino , Páncreas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Receptor de Colecistoquinina B/antagonistas & inhibidores , Receptor de Colecistoquinina B/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal , Vagotomía , Nervio Vago/efectos de los fármacos
18.
Cell Physiol Biochem ; 31(4-5): 542-54, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23571358

RESUMEN

BACKGROUND/AIMS: In this study, we studied the effects of cholecystokinin (CCK) on pacemaker potentials in cultured interstitial cells of Cajal (ICCs) from mouse small intestine using the whole cell patch clamp technique. METHODS: ICCs are pacemaker cells that exhibit periodic spontaneous depolarization, which is responsible for the production of slow waves in gastrointestinal smooth muscle, and generate periodic pacemaker potentials in current-clamp mode. RESULTS: Exposure to CCK (100 nM-5 µM) decreased the amplitudes of pacemaker potentials and depolarized resting membrane potentials. To identify the type of CCK receptors involved in ICCs, we examined the effects of CCK agonists and found that the addition of CCK1 agonist (A-71323, 1 µM) depolarized resting membrane potentials, whereas exposure to CCK2 agonist (gastrin, 1 µM) had no effect on pacemaker potentials. To confirm these results, we examined the effects of CCK antagonists and found that pretreatment with CCK1 antagonist (SR 27897, 1 µM) blocked CCK-induced effects. However, pretreatment with CCK2 antagonist (LY 225910, 1 µM) did not. Furthermore, intracellular GDPßS suppressed CCK-induced effects. To investigate the involvements of phospholipase C (PLC), protein kinase C (PKC), and protein kinase A (PKA) in the effects of CCK in cultured ICCs, we used U-73122 (an active PLC inhibitor), chelerythrine (a PKC inhibitor), SQ-22536 (an inhibitor of adenylate cyclase), or mPKAI (an inhibitor of myristoylated PKA). All inhibitors blocked the CCK-mediated effects on pacemaker potentials. In addition, we found that transient receptor potential classical 5 (TRPC5) channel was involved in CCK-activated currents in cultured ICCs. CONCLUSION: These results suggest that the CCK induced depolarization of pacemaking activity occurs in a G-protein-, PLC-, PKC-, and PKA-dependent manner via CCK1 receptor and TRPC5 channel is a candidate for CCK-activated currents in cultured ICCs in murine small intestine. Therefore, the ICCs are targets for CCK and their interaction can affect intestinal motility.


Asunto(s)
Colecistoquinina/farmacología , Células Intersticiales de Cajal/metabolismo , Intestino Delgado/fisiología , Potenciales de la Membrana/efectos de los fármacos , Animales , Células Cultivadas , Quimiocinas CC , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Estrenos/farmacología , Gastrinas/farmacología , Ácidos Indolacéticos/farmacología , Células Intersticiales de Cajal/citología , Intestino Delgado/citología , Ratones , Ratones Endogámicos BALB C , Técnicas de Placa-Clamp , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Pirrolidinonas/farmacología , Quinazolinonas/farmacología , Receptor de Colecistoquinina B/agonistas , Receptor de Colecistoquinina B/antagonistas & inhibidores , Receptor de Colecistoquinina B/metabolismo , Receptores de Colecistoquinina/agonistas , Receptores de Colecistoquinina/antagonistas & inhibidores , Receptores de Colecistoquinina/metabolismo , Canales Catiónicos TRPC/metabolismo , Tiazoles/farmacología , Fosfolipasas de Tipo C/antagonistas & inhibidores , Fosfolipasas de Tipo C/metabolismo
19.
Br J Clin Pharmacol ; 76(5): 680-8, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23432415

RESUMEN

AIM: To administer repeated oral doses of netazepide to healthy subjects for the first time, to assess safety, tolerability, pharmacokinetics and effect on 24 h gastric pH and plasma gastrin. METHOD: We did two randomized, double-blind, parallel group studies. The first compared netazepide 25 and 100 mg 12 hourly, omeprazole 20 mg once daily and placebo for 7 days. On day 7 only, we measured pH and assayed plasma gastrin. The second study compared netazepide 5, 10 and 25 mg and placebo once daily for 14 days. We measured pH on days 1, 7 and 14 and assayed plasma gastrin on days 1 and 14. We compared treatments by time gastric pH ≥ 4 during 0-4, 4-9, 9-13 and 13-24 h after the morning dose, and by plasma gastrin. P < 0.05 was significant. RESULTS: Netazepide was well tolerated. On day 7 of the first study, netazepide increased pH significantly only during 9-13 h after the 100 mg dose, whereas omeprazole raised pH significantly during all periods. Both netazepide and omeprazole increased plasma gastrin significantly. Netazepide had linear pharmacokinetics. In the second study, netazepide caused dose-dependent, sustained increases in pH on day 1, but as in the first study, netazepide had little effect on pH on days 7 and 14. Again, netazepide increased plasma gastrin significantly. CONCLUSION: Although repeated doses of netazepide led to tolerance to its effect on pH, the accompanying increase in plasma gastrin is consistent with continued inhibition of acid secretion, via gastrin receptor antagonism and gene up-regulation.


Asunto(s)
Benzodiazepinonas/farmacología , Gastrinas/sangre , Omeprazol/farmacología , Compuestos de Fenilurea/farmacología , Receptor de Colecistoquinina B/antagonistas & inhibidores , Adulto , Benzodiazepinonas/administración & dosificación , Benzodiazepinonas/efectos adversos , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Tolerancia a Medicamentos , Femenino , Determinación de la Acidez Gástrica , Humanos , Concentración de Iones de Hidrógeno , Masculino , Omeprazol/administración & dosificación , Omeprazol/efectos adversos , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/efectos adversos , Inhibidores de la Bomba de Protones/administración & dosificación , Inhibidores de la Bomba de Protones/efectos adversos , Inhibidores de la Bomba de Protones/farmacología , Factores de Tiempo , Adulto Joven
20.
Br J Clin Pharmacol ; 76(5): 689-98, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23432534

RESUMEN

AIMS: To confirm by means of pentagastrin, a synthetic gastrin agonist, that netazepide is a gastrin/CCK2 receptor antagonist in healthy subjects, and that antagonism persists during repeated dosing. METHODS: We did two studies in which we infused pentagastrin (0.6 µg kg(-1) h(-1) intravenously), aspirated gastric secretion and measured the volume, pH and H(+) secretion rate of the gastric aspirate. First, we did a double-blind, five-way crossover study (n = 10) to assess the effect of single oral doses of netazepide (1, 5, 25 and 100 mg) and placebo on the response to pentagastrin. Then, we did a single-blind, placebo-controlled study (n = 8) to assess the effect of the first and last oral doses of netazepide (100 mg) twice daily for 13 doses on the response to pentagastrin. RESULTS: Netazepide was well tolerated. After placebo, pentagastrin increased the volume and H(+) secretion rate and reduced the pH of gastric aspirate. Compared with placebo, single doses of netazepide caused dose-dependent inhibition of the pentagastrin response (P < 0.02); netazepide (100 mg) abolished the response. After 13 doses, the reduction in volume and H(+) secretion rate persisted (P < 0.001), but the pH effect was mostly lost. CONCLUSIONS: Netazepide is an orally active, potent, competitive antagonist of human gastrin/CCK2 receptors. Antagonism is dose dependent and persists during repeated dosing, despite tolerance to the effect on pH. Further studies are required to explain that tolerance. Netazepide is a tool to study the physiology and pharmacology of gastrin, and merits studies in patients to assess its potential to treat gastric acid-related conditions and the trophic effects of hypergastrinaemia.


Asunto(s)
Benzodiazepinonas/farmacología , Gastrinas/metabolismo , Pentagastrina/farmacología , Compuestos de Fenilurea/farmacología , Receptor de Colecistoquinina B/antagonistas & inhibidores , Administración Oral , Adulto , Benzodiazepinonas/administración & dosificación , Benzodiazepinonas/efectos adversos , Estudios Cruzados , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Tolerancia a Medicamentos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Masculino , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/efectos adversos , Método Simple Ciego , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA