Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Oncol ; 18(6): 1486-1509, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38375974

RESUMEN

Inter-alpha-trypsin inhibitor heavy chain 5 (ITIH5) has been identified as a metastasis suppressor gene in pancreatic cancer. Here, we analyzed ITIH5 promoter methylation and protein expression in The Cancer Genome Atlas (TCGA) dataset and three tissue microarray cohorts (n = 618), respectively. Cellular effects, including cell migration, focal adhesion formation and protein tyrosine kinase activity, induced by forced ITIH5 expression in pancreatic cancer cell lines were studied in stable transfectants. ITIH5 promoter hypermethylation was associated with unfavorable prognosis, while immunohistochemistry demonstrated loss of ITIH5 in the metastatic setting and worsened overall survival. Gain-of-function models showed a significant reduction in migration capacity, but no alteration in proliferation. Focal adhesions in cells re-expressing ITIH5 exhibited a smaller and more rounded phenotype, typical for slow-moving cells. An impressive increase of acetylated alpha-tubulin was observed in ITIH5-positive cells, indicating more stable microtubules. In addition, we found significantly decreased activities of kinases related to focal adhesion. Our results indicate that loss of ITIH5 in pancreatic cancer profoundly affects its molecular profile: ITIH5 potentially interferes with a variety of oncogenic signaling pathways, including the PI3K/AKT pathway. This may lead to altered cell migration and focal adhesion formation. These cellular alterations may contribute to the metastasis-inhibiting properties of ITIH5 in pancreatic cancer.


Asunto(s)
Adhesión Celular , Movimiento Celular , Neoplasias Pancreáticas , Transducción de Señal , Humanos , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Movimiento Celular/genética , Adhesión Celular/genética , Línea Celular Tumoral , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas/genética , Adhesiones Focales/metabolismo , Adhesiones Focales/genética , Metilación de ADN/genética , Regiones Promotoras Genéticas/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Inhibidoras de Proteinasas Secretoras
2.
Cancers (Basel) ; 15(3)2023 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-36765639

RESUMEN

In pancreatic cancer treatment, tumor stage-dependent chemotherapies are used to prolong overall survival. By measuring DNA promoter hypermethylation in the plasma of patients with stage IV pancreatic cancer, it was recently shown that promoter DNA methylation of the tumor suppressor gene SFRP1 has a high value for predicting failure of drug treatment with gemcitabine. In this study, we therefore aimed to identify as precisely as possible the region in the SFRP1 promoter that is frequently hypermethylated in pancreatic cancer tissue. First, we used the TCGA data set to define CpG-rich regions flanking the SFRP1 transcription start site that were significantly more methylated in pancreatic cancer compared to normal pancreatic acinar tissue. A core CpG island was identified that exhibited abundant tumor DNA methylation and anti-correlation of SFRP1 mRNA expression. To validate our in silico results, we performed bisulfide conversion followed by DNA pyrosequencing of 28 matched formalin-fixed, paraffin-embedded (FFPE) pancreatic cancer cases and six pancreatic cancer cell lines. A defined block of seven CpG sites within the core CpG island was identified, which confirmed our in silico results by showing significantly higher SFRP1 methylation in pancreatic cancer specimens than in normal pancreatic tissue. By selecting this core CpG island, we were able to determine a median overall survival benefit for the low SFRP1 methylation group compared to the high SFRP1 methylation group (702 versus 517 days, p = 0.01) in the TCGA pancreatic cancer cohort. We propose a compact pyrosequencing assay that can be used in the future to further investigate the prognostic value of SFRP1 promoter hypermethylation in predicting pancreatic cancer chemoresistance. Therefore, instead of DNA analysis from blood (liquid biopsy), DNA easily extractable from cancer tissue blocks (FFPE material) could be used.

3.
Early Hum Dev ; 94: 25-30, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26894665

RESUMEN

BACKGROUND: Very low birth weight (VLBW) infants are at risk for postnatal growth restriction due to inadequate nutrient delivery and concomitant illness. Integrated clinical pathways or protocols can improve growth outcomes by decreasing practice variability. METHODS: A comprehensive nutrition bundle comprising standardized recommendations for initiating, advancing, and fortifying enteral feedings, and timely discontinuation of central lines was implemented in July 2012. Eligible were infants with a birth weight of <1500 g and <34 weeks gestation who were born over a 1-year period pre- and post-intervention, respectively. The primary aim was to determine if the intervention improved anthropometric parameter delta z scores at 36 weeks PMA. Secondary aims included time to first and full enteral feedings, central line-days, and rates of necrotizing enterocolitis (NEC) and sepsis/sepsis-like episodes. RESULTS: A total of 299 infants were included, of which 156 received the proposed intervention (Nutrition bundle group), and 143 received non-standardized nutrition practices (Conventional group). Median delta z scores for length (-1.2 versus -1.71; p=0.01) and head circumference (-0.73 versus -1.21; p=0.03) but not weight at 36 weeks PMA (-1.42 versus -1.58; p=0.74) were significantly higher in the Nutrition bundle group as compared to the Conventional group. Fewer infants in the intervention group had severe growth restriction. Time to first feed, full feeds, and central line duration were significantly shorter in the intervention period. The incidence of NEC and sepsis/sepsis-like episodes decreased with the intervention. CONCLUSIONS: A strategy using a comprehensive nutrition bundle improved linear and head circumference growth, reduced postnatal growth restriction, and decreased comorbidities in VLBW infants.


Asunto(s)
Nutrición Enteral/métodos , Enterocolitis Necrotizante/prevención & control , Adhesión a Directriz , Fenómenos Fisiológicos Nutricionales del Lactante , Recién Nacido de muy Bajo Peso/fisiología , Cuidado Intensivo Neonatal/métodos , Sepsis/prevención & control , Estudios de Casos y Controles , Femenino , Humanos , Recién Nacido , Recién Nacido de muy Bajo Peso/crecimiento & desarrollo , Masculino
4.
Bioorg Med Chem ; 21(22): 7064-73, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24094437

RESUMEN

Peptide agonists of the glucagon-like peptide 1 (GLP-1) receptor (GLP1R) are rapidly gaining favor as antidiabetic agents, since in addition to increasing glucose-dependent insulin secretion, they also cause weight loss. Oxyntomodulin (OXM), a natural peptide with sequence homology to both glucagon and GLP-1, has glucose-lowering activity in rodents and anorectic activity in rodents and humans, but its clinical utility is limited by a short circulatory half-life due to rapid renal clearance and degradation by dipeptidyl peptidase IV (DPP-IV). Here, we describe the development of a novel DPP-IV-resistant, long-acting GLP1R agonist, based on derivatization of a suitably chosen OXM analog with high molecular weight polyethylene glycol (PEG) ('PEGylation'). PEG-OXM exerts an anti-hyperglycemic effect in diet-induced obese (DIO) mice in a glucose-dependent manner, with a maximally efficacious dose of 0.1mg/kg, and reduces food intake and body weight with a minimally efficacious dose of 1mg/kg. If this pharmacology is recapitulated in patients with type 2 diabetes, these results indicate PEG-OXM as a potential novel once-weekly GLP-1 mimetic with both glucose-lowering activity and weight loss efficacy.


Asunto(s)
Depresores del Apetito/química , Hipoglucemiantes/química , Oxintomodulina/química , Polietilenglicoles/química , Receptores de Glucagón/agonistas , Animales , Depresores del Apetito/síntesis química , Depresores del Apetito/farmacocinética , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón , Prueba de Tolerancia a la Glucosa , Semivida , Hipoglucemiantes/síntesis química , Hipoglucemiantes/farmacocinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Primates , Receptores de Glucagón/metabolismo
5.
Am J Physiol Endocrinol Metab ; 303(2): E265-71, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22621866

RESUMEN

Glucagon-like peptide-1 (GLP-1) and oxyntomodulin (OXM) are peptide hormones secreted postprandially from the gut that stimulate insulin secretion in a glucose-dependent manner. OXM activates both the GLP-1 receptor (GLP1R) and the glucagon receptor (GCGR). It has been suggested that OXM acutely modulates glucose metabolism solely through GLP1R agonism. Because OXM activates the GLP1R with lower affinity than GLP-1, we generated a peptide analog (Q→E, OXMQ3E) that does not exhibit glucagon receptor agonist activity but retains the same affinity as OXM for GLP1R. We compared the effects of OXM and OXMQ3E in a glucose tolerance test and, to better characterize the effect on glucose metabolism, we performed controlled infusions of OXM or OXMQ3E during a hyperglycemic clamp performed in wild-type, Glp1r(-/-), and Gcgr(-/-) mice. Our findings show that OXM, but not OXMQ3E, activates the GCGR in vivo. Second, OXM and OXMQ3E improve glucose tolerance following an acute glucose challenge and during a hyperglycemic clamp in mice. Finally, OXM infusion during a glucose clamp reduces the glucose infusion rate (GIR) despite a simultaneous increase in insulin levels in Glp1r(-/-) mice, whereas OXM and OXMQ3E increase GIR to a similar extent in Gcgr(-/-) mice. In conclusion, activation of the GCGR seems to partially attenuate the acute beneficial effects on glucose and contributes to the insulinotropic action of oxyntomodulin.


Asunto(s)
Péptido 1 Similar al Glucagón/farmacología , Glucosa/metabolismo , Oxintomodulina/farmacología , Animales , Glucemia/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón , Técnica de Clampeo de la Glucosa , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Glucagón/genética , Receptores de Glucagón/metabolismo
6.
PLoS One ; 7(4): e35240, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22506074

RESUMEN

Short-chain fatty acids (SCFAs), primarily acetate, propionate, and butyrate, are metabolites formed by gut microbiota from complex dietary carbohydrates. Butyrate and acetate were reported to protect against diet-induced obesity without causing hypophagia, while propionate was shown to reduce food intake. However, the underlying mechanisms for these effects are unclear. It was suggested that SCFAs may regulate gut hormones via their endogenous receptors Free fatty acid receptors 2 (FFAR2) and 3 (FFAR3), but direct evidence is lacking. We examined the effects of SCFA administration in mice, and show that butyrate, propionate, and acetate all protected against diet-induced obesity and insulin resistance. Butyrate and propionate, but not acetate, induce gut hormones and reduce food intake. As FFAR3 is the common receptor activated by butyrate and propionate, we examined these effects in FFAR3-deficient mice. The effects of butyrate and propionate on body weight and food intake are independent of FFAR3. In addition, FFAR3 plays a minor role in butyrate stimulation of Glucagon-like peptide-1, and is not required for butyrate- and propionate-dependent induction of Glucose-dependent insulinotropic peptide. Finally, FFAR3-deficient mice show normal body weight and glucose homeostasis. Stimulation of gut hormones and food intake inhibition by butyrate and propionate may represent a novel mechanism by which gut microbiota regulates host metabolism. These effects are largely intact in FFAR3-deficient mice, indicating additional mediators are required for these beneficial effects.


Asunto(s)
Butiratos/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Hormonas Gastrointestinales/metabolismo , Obesidad/metabolismo , Propionatos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Butiratos/farmacología , Dieta , Polipéptido Inhibidor Gástrico/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/prevención & control , Propionatos/farmacología
7.
Obesity (Silver Spring) ; 20(8): 1566-71, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22421924

RESUMEN

Oxyntomodulin (OXM) is a peptide secreted postprandially from the L-cells of the gut that has a weak affinity for both the glucagon-like peptide-1 receptor (GLP1R) and the glucagon receptor (GCGR). Peripheral administration of OXM in humans and rodents causes weight loss reducing food intake and increasing energy expenditure. It has been suggested that OXM modulates energy intake solely through GLP1R agonism. Because glucagon decreases food intake in rodents and humans, we examined whether activation of the GCGR is involved in the body weight-lowering effects of OXM. We identified an equipotent GLP1R-selective peptide agonist that differs from OXM by only one residue (Q3→E, OXMQ3E), but has no significant GCGR agonist activity in vitro and ~100-fold reduced ability to stimulate liver glycogenolysis. Chronic treatment of obese mice with OXM and OXMQ3E demonstrated that OXM exhibits superior weight loss and lipid-lowering efficacy, and antihyperglycemic activity that is comparable to the corresponding GLP1R-selective agonist. Studies in Glp1r(-/-) mice and coadministration of OXM and a GCGR antagonist revealed that the antiobesity effect of OXM requires activation of both GLP1R and GCGR. Our data provide new insight into the mechanism of action of OXM and suggest that activation of GCGR is involved in the body weight-lowering action of OXM.


Asunto(s)
Fármacos Antiobesidad/farmacología , Ingestión de Energía/fisiología , Glucagón/metabolismo , Obesidad/metabolismo , Oxintomodulina/metabolismo , Receptores de Glucagón/agonistas , Pérdida de Peso/efectos de los fármacos , Animales , Ingestión de Energía/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón , Glucogenólisis/efectos de los fármacos , Humanos , Hipoglucemiantes/farmacología , Hipolipemiantes/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad/tratamiento farmacológico , Oxintomodulina/farmacología , Receptores de Glucagón/metabolismo
8.
Endocrinology ; 152(7): 2644-54, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21586559

RESUMEN

Neuromedin U (NMU) is a highly conserved peptide reported to modulate energy homeostasis. Pharmacological studies have shown that centrally administered NMU inhibits food intake, reduces body weight, and increases energy expenditure. NMU-deficient mice develop obesity, whereas transgenic mice overexpressing NMU become lean and hypophagic. Two high-affinity NMU receptors, NMUR1 and NMUR2, have been identified. NMUR1 is found primarily in the periphery and NMUR2 primarily in the brain, where it mediates the anorectic effects of centrally administered NMU. Given the broad expression pattern of NMU, we evaluated whether peripheral administration of NMU has effects on energy homeostasis. We observed that acute and chronic peripheral administration of NMU in rodents dose-dependently reduced food intake and body weight and that these effects required NMUR1. The anorectic effects of NMU appeared to be partly mediated by vagal afferents. NMU treatment also increased core body temperature and metabolic rate in mice, suggesting that peripheral NMU modulates energy expenditure. Additionally, peripheral administration of NMU significantly improved glucose excursion. Collectively, these data suggest that NMU functions as a peripheral regulator of energy and glucose homeostasis and the development of NMUR1 agonists may be an effective treatment for diabetes and obesity.


Asunto(s)
Metabolismo Energético , Intolerancia a la Glucosa/prevención & control , Neuropéptidos/fisiología , Animales , Regulación del Apetito , Metabolismo Basal , Regulación de la Temperatura Corporal , Diabetes Mellitus/tratamiento farmacológico , Preferencias Alimentarias , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Terapia Molecular Dirigida , Neuropéptidos/administración & dosificación , Obesidad/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley , Receptores de Neurotransmisores/genética , Receptores de Neurotransmisores/metabolismo , Pérdida de Peso
9.
J Pharmacol Exp Ther ; 336(2): 356-64, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21036912

RESUMEN

Bombesin receptor subtype-3 (BRS-3) is an orphan G protein-coupled receptor implicated in the regulation of energy homeostasis. Here, we report the biologic effects of a highly optimized BRS-3 agonist, (2S)-1,1,1-trifluoro-2-[4-(1H-pyrazol-1-yl)phenyl]-3-(4-{[1-(trifluoromethyl)cyclopropyl]methyl}-1H-imidazol-2-yl)propan-2-ol (MK-5046). Single oral doses of MK-5046 inhibited 2-h and overnight food intake and increased fasting metabolic rate in wild-type but not Brs3 knockout mice. Upon dosing for 14 days, MK-5046 at 25 mg · kg(-1) · day(-1) reduced body weight of diet-induced obese mouse by 9% compared with vehicle-dosed controls. In mice, 50% brain receptor occupancy was achieved at a plasma concentration of 0.34 ± 0.23 µM. With chronic dosing, effects on metabolic rate, rather than food intake, seem to be the predominant mechanism for weight reduction by MK-5046. The compound also effectively reduced body weight in rats and caused modest increases in body temperature, heart rate, and blood pressure. These latter effects on temperature, heart rate, and blood pressure were transient in nature and desensitized with continued dosing. MK-5046 is the first BRS-3 agonist with properties suitable for use in larger mammals. In dogs, MK-5046 treatment produced statistically significant and persistent weight loss, which was initially accompanied by increases in body temperature and heart rate that abated with continued dosing. Our results demonstrate antiobesity efficacy for MK-5046 in rodents and dogs and further support BRS-3 agonism as a new approach to the treatment of obesity.


Asunto(s)
Fármacos Antiobesidad/farmacología , Imidazoles/farmacología , Pirazoles/farmacología , Receptores de Bombesina/agonistas , Animales , Presión Sanguínea/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Perros , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Receptores de Bombesina/análisis
10.
ACS Med Chem Lett ; 2(1): 43-7, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-24900253

RESUMEN

We report the development and characterization of compound 22 (MK-5046), a potent, selective small molecule agonist of BRS-3 (bombesin receptor subtype-3). In pharmacological testing using diet-induced obese mice, compound 22 caused mechanism-based, dose-dependent reductions in food intake and body weight.

11.
Cell Metab ; 11(2): 101-12, 2010 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-20096642

RESUMEN

Bombesin receptor subtype 3 (BRS-3) is a G protein coupled receptor whose natural ligand is unknown. We developed potent, selective agonist (Bag-1, Bag-2) and antagonist (Bantag-1) ligands to explore BRS-3 function. BRS-3-binding sites were identified in the hypothalamus, caudal brainstem, and several midbrain nuclei that harbor monoaminergic cell bodies. Antagonist administration increased food intake and body weight, whereas agonists increased metabolic rate and reduced food intake and body weight. Prolonged high levels of receptor occupancy increased weight loss, suggesting a lack of tachyphylaxis. BRS-3 agonist effectiveness was absent in Brs3(-/Y) (BRS-3 null) mice but was maintained in Npy(-/-)Agrp(-/-), Mc4r(-/-), Cnr1(-/-), and Lepr(db/db) mice. In addition, Brs3(-/Y) mice lost weight upon treatment with either a MC4R agonist or a CB1R inverse agonist. These results demonstrate that BRS-3 has a role in energy homeostasis that complements several well-known pathways and that BRS-3 agonists represent a potential approach to the treatment of obesity.


Asunto(s)
Fármacos Antiobesidad/uso terapéutico , Obesidad/tratamiento farmacológico , Péptidos/uso terapéutico , Receptores de Bombesina/agonistas , Receptores de Bombesina/metabolismo , Animales , Fármacos Antiobesidad/farmacocinética , Peso Corporal/efectos de los fármacos , Encéfalo/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Humanos , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Péptidos/farmacocinética , Ratas , Ratas Sprague-Dawley , Receptores de Bombesina/antagonistas & inhibidores
12.
Endocrinology ; 150(7): 3101-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19324999

RESUMEN

Neuromedin U (NMU) and neuromedin S (NMS) are structurally related neuropeptides that have been reported to modulate energy homeostasis. Pharmacological data have shown that NMU and NMS inhibit food intake when administered centrally and that NMU increases energy expenditure. Additionally, NMU-deficient mice develop obesity, whereas transgenic mice overexpressing NMU are lean and hypophagic. Two high-affinity NMU/NMS receptors, NMUR1 and NMUR2, have been identified. NMUR1 is predominantly expressed in the periphery, whereas NMUR2 is predominantly expressed in the brain, suggesting that the effects of centrally administered NMU and NMS are mediated by NMUR2. To evaluate the role of NMUR2 in the regulation of energy homeostasis, we characterized NMUR2-deficient (Nmur2(-/-)) mice. Nmur2(-/-) mice exhibited a modest resistance to diet-induced obesity that was at least in part due to reduced food intake. Acute central administration of NMU and NMS reduced food intake in wild-type but not in Nmur2(-/-) mice. The effects on activity and core temperature induced by centrally administered NMU were also absent in Nmur2(-/-) mice. Moreover, chronic central administration of NMU and NMS evoked significant reductions in body weight and sustained reductions in food intake in mice. In contrast, Nmur2(-/-) mice were largely resistant to these effects. Collectively, these data demonstrate that the anorectic and weight-reducing actions of centrally administered NMU and NMS are mediated predominantly by NMUR2, suggesting that NMUR2-selective agonists may be useful for the treatment of obesity.


Asunto(s)
Neuropéptidos/farmacología , Receptores de Neurotransmisores/fisiología , Animales , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/fisiología , Aseo Animal/efectos de los fármacos , Ratones , Ratones Obesos , Actividad Motora/efectos de los fármacos , Neuropéptidos/fisiología , Obesidad/prevención & control , Receptores de Neurotransmisores/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA