Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Medicinas Complementárias
Tipo del documento
Intervalo de año de publicación
1.
Genes (Basel) ; 12(8)2021 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-34440457

RESUMEN

The inclusion of fat in livestock diets represents a valuable and cost-effective way to increase the animal's caloric intake. Beyond their caloric value, fatty acids can be understood in terms of their bioactivity, via the modulation of the ligand-dependent nuclear peroxisome proliferator-activated receptors (PPAR). Isotypes of PPAR regulate important metabolic processes in both monogastric and ruminant animals, including the metabolism of fatty acids (FA), the production of milk fat, and the immune response; however, information on the modulation of bovine PPAR by fatty acids is limited. The objective of this study was to expand our understanding on modulation of bovine PPAR by FA, both when used individually and in combination, in an immortalized cell culture model of bovine liver. Of the 10 FA included in the study, the greatest activation of the PPAR reporter was detected with saturated FA C12:0, C16:0, and C18:0, as well as phytanic acid, and the unsaturated FA C16:1 and C18:1. When supplemented in mixtures of 2 FA, the most effective combination was C12:0 + C16:0, while in mixtures of 3 FA, the greatest activation was caused by combinations of C12:0 with C16:0 and either C18:0, C16:1, or C18:1. Some mixtures display a synergistic effect that leads to PPAR activation greater than the sum of their parts, which may be explained by structural dynamics within the PPAR ligand-binding pocket. Our results provide fundamental information for the development of tailored dietary plans that focus on the use of FA mixtures for nutrigenomic purposes.


Asunto(s)
Ingestión de Energía/genética , Ácidos Grasos/metabolismo , Hígado/metabolismo , Receptores Activados del Proliferador del Peroxisoma/genética , Tejido Adiposo/metabolismo , Alimentación Animal , Animales , Bovinos , Ácidos Grasos/genética , Ácidos Grasos/farmacología , Femenino , Inmunidad/genética , Lactancia/efectos de los fármacos , Lactancia/genética , Leche/metabolismo , Nutrigenómica , Receptores Activados del Proliferador del Peroxisoma/metabolismo
2.
Behav Processes ; 170: 103981, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31682870

RESUMEN

Convergent evidence in literature shows that rapid disruption of maternal care and breastfeeding due to an early weaning protocol changes the development of several neurobehavioral patterns in rodents, including the circadian pattern of feeding. The serotoninergic system has been associated with the control of feeding patterns. Therefore, we aim to evaluate the patterns of feeding, the mRNA expression of 5 H T-1b, 5 H T-2c, and SERT on the hypothalamus, brainstem, and the body weight of female juvenile Wistar rats, submitted to early (PND15) or regular (PND30) weaning. The results demonstrate that early weaning promotes an increase in food intake in a 24 -h period, in the dark phase of the circadian cycle and in the four-hour time intervals at the beginning of the dark and light phases. Also, early weaning decreases the mRNA expression of 5 H T-1b, 5 H T-2c, and SERT on the hypothalamus, but increases it on the brainstem. Additionally, early weaning promotes an increase in body weight. Therefore, the present data demonstrate that early weaning changes the patterns of feeding in juvenile female rats and suggests that this behavioral modification is due to the modulations promoted in the 5 H T-system.


Asunto(s)
Conducta Alimentaria/fisiología , Serotonina/fisiología , Destete , Animales , Peso Corporal/genética , Encéfalo/anatomía & histología , Tronco Encefálico/metabolismo , Ritmo Circadiano , Ingestión de Alimentos/fisiología , Ingestión de Energía/genética , Femenino , Hipotálamo/metabolismo , Conducta Materna , Tamaño de los Órganos/genética , ARN Mensajero/biosíntesis , Proteínas de Unión al ARN/genética , Ratas , Ratas Wistar
3.
Sci Rep ; 8(1): 17984, 2018 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-30573851

RESUMEN

The effect of short-term caloric restriction on gene expression in critically ill patients has not been studied. In this sub-study of the PermiT trial (Permissive Underfeeding or Standard Enteral Feeding in Critically Ill Adults Trial- ISRCTN68144998), we examined gene expression patterns in peripheral white blood cells (buffy coat) associated with moderate caloric restriction (permissive underfeeding) in critically ill patients compared to standard feeding. Blood samples collected on study day 1 and 14 were subjected to total RNA extraction and gene expression using microarray analysis. We enrolled 50 patients, 25 in each group. Among 1751 tested genes, 332 genes in 12 pathways were found to be significantly upregulated or downregulated between study day 1 and 14 (global p value for the pathway ≤ 0.05). Using the heatmap, the differential expression of genes from day 1 to 14 in the permissive underfeeding group was compared to the standard feeding group. We further compared gene expression signal intensity in permissive underfeeding compared standard feeding by constructing univariate and multivariate linear regression models on individual patient data. We found differential expression of several genes with permissive underfeeding, most notably those related to metabolism, autophagy and other cellular functions, indicating that moderate differences in caloric intake trigger different cellular pathways.


Asunto(s)
Enfermedad Crítica , Ingestión de Energía/genética , Leucocitos/metabolismo , Desnutrición/genética , Terapia Nutricional/métodos , Transcriptoma , Adulto , Anciano , Restricción Calórica , Cuidados Críticos/métodos , Cuidados Críticos/normas , Enfermedad Crítica/terapia , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Desnutrición/sangre , Persona de Mediana Edad , Estudios Retrospectivos , Nivel de Atención , Adulto Joven
4.
Discov Med ; 24(131): 95-106, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28972878

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease in the world. The nutrients play important roles in the development and progression of NAFLD. High-calorie diet, especially the diet rich in saturated fatty acids and cholesterol, as well as sugary drinks with high fructose content, induces hepatic steatosis and triggers progression of steatohepatitis, fibrosis, and even hepatocellular carcinoma. Disordered micronutrient status and gut microbiota are also involved in the pathogenesis of NAFLD. Nutrients related NAFLD could be aggravated by a genetic predisposition, for instance, genetic mutations in patatinlike phospholipase domain-containing 3 (PNPLA3) and transmembrane 6 superfamily member 2 (TM6SF2). Reduction of caloric intake through lifestyle interventions and use of dietary supplements such as omega-3 fatty acids, vitamins, and probiotics may help alleviate liver injury in NAFLD.


Asunto(s)
Carbohidratos de la Dieta/efectos adversos , Grasas de la Dieta/efectos adversos , Ingestión de Energía/genética , Microbioma Gastrointestinal , Lipasa/genética , Proteínas de la Membrana/genética , Enfermedad del Hígado Graso no Alcohólico , Polimorfismo Genético , Animales , Humanos , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/microbiología
5.
J Dairy Sci ; 100(7): 5501-5514, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28478005

RESUMEN

The objective of the present study was to estimate genetic parameters across lactation for measures of energy balance (EB) and a range of feed efficiency variables as well as to quantify the genetic inter-relationships between them. Net energy intake (NEI) from pasture and concentrate intake was estimated up to 8 times per lactation for 2,481 lactations from 1,274 Holstein-Friesian cows. A total of 8,134 individual feed intake measurements were used. Efficiency traits were either ratio based or residual based; the latter were derived from least squares regression models. Residual energy intake (REI) was defined as NEI minus predicted energy requirements [e.g., net energy of lactation (NEL), maintenance, and body tissue anabolism] or supplied from body tissue mobilization; residual energy production was defined as the difference between actual NEL and predicted NEL based on NEI, maintenance, and body tissue anabolism/catabolism. Energy conversion efficiency was defined as NEL divided by NEI. Random regression animal models were used to estimate residual, additive genetic, and permanent environmental (co)variances across lactation. Heritability across lactation stages varied from 0.03 to 0.36 for all efficiency traits. Within-trait genetic correlations tended to weaken as the interval between lactation stages compared lengthened for EB, REI, residual energy production, and NEI. Analysis of eigenvalues and associated eigenfunctions for EB and the efficiency traits indicate the ability to genetically alter the profile of these lactation curves to potentially improve dairy cow efficiency differently at different stages of lactation. Residual energy intake and EB were moderately to strongly genetically correlated with each other across lactation (genetic correlations ranged from 0.45 to 0.90), indicating that selection for lower REI alone (i.e., deemed efficient cows) would favor cows with a compromised energy status; nevertheless, selection for REI within a holistic breeding goal could be used to overcome such antagonisms. The smallest (8.90% of genetic variance) and middle (11.22% of genetic variance) eigenfunctions for REI changed sign during lactation, indicating the potential to alter the shape of the REI lactation profile. Results from the present study suggest exploitable genetic variation exists for a range of efficiency traits, and the magnitude of this variation is sufficiently large to justify consideration of the feed efficiency complex in future dairy breeding goals. Moreover, it is possible to alter the trajectories of the efficiency traits to suit a particular breeding objective, although this relies on very precise across-parity genetic parameter estimates, including genetic correlations with health and fertility traits (as well as other traits).


Asunto(s)
Ingestión de Energía/genética , Metabolismo Energético/genética , Herbivoria/genética , Lactancia/genética , Alimentación Animal/estadística & datos numéricos , Animales , Cruzamiento , Bovinos , Industria Lechera , Femenino , Embarazo
6.
Mol Cell Endocrinol ; 450: 113-125, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28479374

RESUMEN

We evaluated the effect of cafeteria diet (CAF) on the mRNA levels and DNA methylation state of feeding-related neuropeptides, and neurosteroidogenic enzymes in discrete hypothalamic nuclei. Besides, the expression of steroid hormone receptors was analyzed. Female rats fed with CAF from weaning increased their energy intake, body weight, and fat depots, but did not develop metabolic syndrome. The increase in energy intake was related to an orexigenic signal of paraventricular (PVN) and ventromedial (VMN) nuclei, given principally by upregulation of AgRP and NPY. This was mildly counteracted by the arcuate nucleus, with decreased AgRP expression and increased POMC and kisspeptin expression. CAF altered the transcription of neurosteroidogenic enzymes in PVN and VMN, and epigenetic mechanisms associated with differential promoter methylation were involved. The changes observed in the hypothalamic nuclei studied could add information about their differential role in food intake control and how their action is disrupted in obesity.


Asunto(s)
Metilación de ADN/genética , Dieta , Ingestión de Alimentos/genética , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Tejido Adiposo/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Sitios de Unión , Peso Corporal , Simulación por Computador , Ingestión de Energía/genética , Femenino , Prueba de Tolerancia a la Glucosa , Neuropéptidos/genética , Neuropéptidos/metabolismo , Tamaño de los Órganos/genética , Núcleo Hipotalámico Paraventricular/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , Receptores Citoplasmáticos y Nucleares/metabolismo , Esteroides/metabolismo , Transcripción Genética , Núcleo Hipotalámico Ventromedial/metabolismo
7.
Sci Rep ; 7: 44444, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28294152

RESUMEN

Melanocortin-3 receptors (MC3R) have a contextual role in appetite control that is amplified with hypocaloric conditioning. C57BL/6J (B6) mice subjected to hypocaloric feeding schedules (HFS) exhibit compulsive behavioral responses involving food anticipatory activity (FAA) and caloric loading following food access. These homeostatic responses to calorie-poor environs are attenuated in B6 mice in which Mc3r transcription is suppressed by a lox-stop-lox sequence in the 5'UTR (Mc3rTB/TB). Here, we report that optimization of caloric loading in B6 mice subject to HFS, characterized by increased meal size and duration, is not observed in Mc3rTB/TB mice. Analysis of hypothalamic and neuroendocrine responses to HFS throughout the light-dark cycle suggests uncoupling of hypothalamic responses involving appetite-stimulating fasting-responsive hypothalamic neurons expressing agouti-related peptide (AgRP) and neuropeptide Y (Npy). Rescuing Mc3rs expression in Nkx2.1(+ve) neurons is sufficient to restore normal hypothalamic responses to negative energy balance. In addition, Mc3rs expressed in Nkx2.1(+ve) neurons are also sufficient to restore FAA and caloric loading of B6 mice subjected to HFS. In summary, MC3Rs expressed in Nkx2.1(+ve) neurons are sufficient to coordinate hypothalamic response and expression of compulsive behavioral responses involving meal anticipation and consumption of large meals during situations of prolonged negative energy balance.


Asunto(s)
Proteína Relacionada con Agouti/genética , Metabolismo Energético/genética , Neuropéptido Y/genética , Receptor de Melanocortina Tipo 3/genética , Animales , Apetito/genética , Ingestión de Energía/genética , Homeostasis , Hipotálamo/metabolismo , Ratones , Neuronas/metabolismo , Fotoperiodo , Factor Nuclear Tiroideo 1/genética
8.
Pharmacogenomics J ; 13(1): 80-93, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21969101

RESUMEN

Several genetic diseases are triggered by nonsense mutations leading to the formation of truncated and defective proteins. Aminoglycosides have the capability to mediate a bypass of stop mutations during translation thus resulting in a rescue of protein expression. So far no attention has been directed to obesity-associated stop mutations as targets for nonsense suppression. Herein, we focus on the characterization of the melanocortin-4-receptor (MC4R) nonsense allele W16X identified in obese subjects. Cell culture assays revealed a loss-of-function of Mc4r(X16) characterized by impaired surface expression and defect signaling. The aminoglycoside G-418 restored Mc4r(X16) function in vitro demonstrating that Mc4r(X16) is susceptible to nonsense suppression. For the evaluation of nonsense suppression in vivo, we generated a Mc4r(X16) knock-in mouse line by gene targeting. Mc4r(X16) knock-in mice developed hyperphagia, impaired glucose tolerance, severe obesity and an increased body length demonstrating that this new mouse model resembles typical characteristics of Mc4r deficiency. In a first therapeutic trial, the aminoglycosides gentamicin and amikacin induced no amelioration of obesity. Further experiments with Mc4r(X16) knock-in mice will be instrumental to establish nonsense suppression for Mc4r as an obesity-associated target gene expressed in the central nervous system.


Asunto(s)
Codón sin Sentido , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Aminoglicósidos/genética , Aminoglicósidos/metabolismo , Animales , Composición Corporal/genética , Temperatura Corporal/genética , Peso Corporal/genética , Células COS , Línea Celular , Chlorocebus aethiops , Ingestión de Energía/genética , Expresión Génica/genética , Células HEK293 , Humanos , Hipotálamo/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/genética , Obesidad/metabolismo
9.
Chin J Integr Med ; 18(1): 23-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22231706

RESUMEN

OBJECTIVE: To study the influence of Radix aconiti lateralis preparata and Rhizoma zingiberis, two species of Chinese medicinal herbs with hot property, on energy metabolism and gene expression spectrum, and to analyze the possible mechanism of their effects. METHODS: Forty-eight specific pathogen free Wistar rats were randomly divided into a Radix aconiti lateralis preparata group, a Rhizoma zingiberis group, and a control group. They were intragastrically treated with concentrated decoction of Radix aconiti lateralis preparata, Rhizoma zingiberis and normal saline respectively for 20 days. Toe temperature (TT), energy intake (EI), digestible energy (DE), and metabolizable energy (ME) were measured. The content of adenosine triphosphate (ATP) and energy charge (EC) in hepatic tissue were measured with high performance liquid chromatography (HPLC). The activity of ATPase and succinate dehydrogenase (SDH) in the liver were detected with chemical colorimetry. The gene expression in the liver was detected with Illumina's rat Ref-12 gene array. The differential expression genes were selected, annotated and classified based on Gene Ontology (GO). Real-time quantitative reverse-transcriptase PCR (Q-RT-PCR) was used to test the accuracy of results. RESULTS: Compared with the control group, the TT on the 10(th) day after the beginning of administration and ATP in the Radix aconiti lateralis preparata and Rhizoma zingiberis groups increased significantly (P<0.05). EI/body mass (BM), DE/BM, ME/BM, the hepatic EC and the activity of Na(+)-K(+)-ATPase, Ca(2+)-Mg(2+)-ATPase and SDH of liver increased significantly only in the Radix aconiti lateralis preparata group (P<0.05). There were 592 differential expression genes in the Radix aconiti lateralis preparata group and 1 159 in the Rhizoma zingiberis group compared with the control group. Among the differential expression genes, genes related to metabolic processes were the most significant based on GO analysis. There were 337 strips of gene differential expression in common in both Radix aconiti lateralis preparata and Rhizoma zingiberis groups compared with the control group. CONCLUSIONS: Herbs with hot property such as Radix aconiti lateralis preparata and Rhizoma zingiberis could improve the energy metabolism in rats, through influencing the metabolic process of sugar, lipid, and amino acid. It could also promote the production, storage, and utilization of energy by regulating the gene expression related to metabolism, which may be the main molecular mechanism of warming yang and dispelling cold for the treatment of the cold syndrome according to Chinese medicine theory.


Asunto(s)
Aconitum/química , Medicamentos Herbarios Chinos/farmacología , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Regulación de la Expresión Génica/efectos de los fármacos , Rizoma/química , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Temperatura Corporal/efectos de los fármacos , Medicamentos Herbarios Chinos/administración & dosificación , Ingestión de Energía/efectos de los fármacos , Ingestión de Energía/genética , Femenino , Perfilación de la Expresión Génica , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Reproducibilidad de los Resultados , Succinato Deshidrogenasa/metabolismo
10.
Proc Nutr Soc ; 70(1): 135-43, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21092367

RESUMEN

Taste is often cited as the factor of greatest significance in food choice, and has been described as the body's 'nutritional gatekeeper'. Variation in taste receptor genes can give rise to differential perception of sweet, umami and bitter tastes, whereas less is known about the genetics of sour and salty taste. Over twenty-five bitter taste receptor genes exist, of which TAS2R38 is one of the most studied. This gene is broadly tuned to the perception of the bitter-tasting thiourea compounds, which are found in brassica vegetables and other foods with purported health benefits, such as green tea and soya. Variations in this gene contribute to three thiourea taster groups of people: supertasters, medium tasters and nontasters. Differences in taster status have been linked to body weight, alcoholism, preferences for sugar and fat levels in food and fruit and vegetable preferences. However, genetic predispositions to food preferences may be outweighed by environmental influences, and few studies have examined both. The Tastebuddies study aimed at taking a holistic approach, examining both genetic and environmental factors in children and adults. Taster status, age and gender were the most significant influences in food preferences, whereas genotype was less important. Taster perception was associated with BMI in women; nontasters had a higher mean BMI than medium tasters or supertasters. Nutrient intakes were influenced by both phenotype and genotype for the whole group, and in women, the AVI variation of the TAS2R38 gene was associated with a nutrient intake pattern indicative of healthy eating.


Asunto(s)
Disgeusia/genética , Preferencias Alimentarias/fisiología , Variación Genética , Percepción del Gusto/genética , Gusto/genética , Factores de Edad , Índice de Masa Corporal , Disgeusia/complicaciones , Ingestión de Energía/genética , Genotipo , Humanos , Fenotipo , Factores Sexuales
11.
Int J Obes (Lond) ; 35(2): 226-35, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20548306

RESUMEN

BACKGROUND: αMUPA mice carry as a transgene the cDNA encoding urokinase-type plasminogen activator, a member of the plasminogen/plasmin system that functions in fibrinolysis and extracellular proteolysis. These mice spontaneously consume less food when fed ad libitum and live longer compared with wild-type (WT) control mice. αMUPA mice are obesity resistant and they share many similarities with calorically restricted animals. However, extensive metabolic characterization of this unique transgenic model has never been performed. METHOD: Metabolism of αMUPA mice was analyzed by measuring hormone, lipid and glucose levels in the serum, as well as gene and protein expression levels in the liver, hypothalamus and brainstem. RESULTS: αMUPA mice were found to be leaner than WT mice mainly because of reduced fat depots. Serum analyses showed that αMUPA mice have high levels of the anorexigenic hormones insulin and leptin, and low levels of the orexigenic hormone ghrelin. Analyses of brain neuropeptides showed that the transcript of the anorexigenic neuropeptide Pomc is highly expressed in the brainstem, whereas the expression of the orexigenic neuropeptides Npy, Orexin and Mch is blunted in the hypothalamus of αMUPA mice. In addition, adenosine monophosphate (AMP)-activated protein kinase (AMPK) levels were higher in the liver and lower in the hypothalamus, thus promoting simultaneously central reduction in appetite and peripheral loss of fat. The levels of SIRT1 were low in the liver, but high in the hypothalamus, a feature that αMUPA mice share with calorically restricted animals. CONCLUSION: Taken together, αMUPA mice exhibit a unique metabolic phenotype of low-calorie intake and high leptin levels, and could serve as a model for both spontaneous calorie restriction and resistance to obesity.


Asunto(s)
Ingestión de Energía/fisiología , Metabolismo Energético/fisiología , Conducta Alimentaria/fisiología , Leptina/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/genética , Animales , Glucemia/análisis , Glucemia/metabolismo , Tronco Encefálico/metabolismo , Ingestión de Energía/genética , Metabolismo Energético/genética , Femenino , Ghrelina/sangre , Hipotálamo/metabolismo , Insulina/sangre , Leptina/genética , Lípidos/sangre , Hígado/metabolismo , Longevidad/fisiología , Ratones , Ratones Obesos , Ratones Transgénicos , Neuropéptidos/metabolismo , Delgadez/genética , Delgadez/metabolismo
12.
Endocrinology ; 151(10): 4756-64, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20685869

RESUMEN

Maternal obesity during pregnancy increases the risk of obesity in the offspring. Obesity, arising from an imbalance of energy intake and expenditure, can be driven by the ingestion of palatable [high fat (HF), high sugar], energy-dense foods. Dopamine and opioid circuitry are neural substrates associated with reward that can affect animals' preference for palatable foods. Using a mouse model, the long-term effect of maternal consumption of a HF diet on dopamine and opioid gene expression within the mesocorticolimbic reward circuitry and hypothalamus of the offspring was investigated. Mice from dams fed a HF diet during pregnancy and lactation showed an increased preference for sucrose and fat. Gene expression, measured using quantitative real-time PCR, revealed a significant approximately 3- to 10-fold up-regulation of dopamine reuptake transporter (DAT) in the ventral tegmental area, nucleus accumbens, and prefrontal cortex and a down-regulation of DAT in the hypothalamus. Additionally, expression of both µ-opioid receptor (MOR) and preproenkephalin (PENK) was increased in nucleus accumbens, prefrontal cortex, and hypothalamus of mice from dams that consumed the HF diet. Epigenetic mechanisms have been associated with long-term programming of gene expression after various in utero insults. We observed global and gene-specific (DAT, MOR, and PENK) promoter DNA hypomethylation in the brains of offspring from dams that consumed the HF diet. These data demonstrate that maternal consumption of a HF diet can change the offsprings' epigenetic marks (DNA hypomethylation) in association with long-term alterations in gene expression (dopamine and opioids) and behavior (preference for palatable foods).


Asunto(s)
Metilación de ADN/efectos de los fármacos , Grasas de la Dieta/farmacología , Dopamina/genética , Péptidos Opioides/genética , Receptores Opioides/genética , Animales , Animales Recién Nacidos , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Ingestión de Energía/genética , Femenino , Preferencias Alimentarias/fisiología , Expresión Génica/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Fenómenos Fisiologicos Nutricionales Maternos/efectos de los fármacos , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Intercambio Materno-Fetal/efectos de los fármacos , Intercambio Materno-Fetal/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Péptidos Opioides/metabolismo , Embarazo , Receptores Opioides/metabolismo , Recompensa
13.
Physiol Behav ; 101(3): 331-43, 2010 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-20600198

RESUMEN

We examined sugar-induced obesity in mouse strains polymorphic for Tas1r3, a gene that codes for the T1R3 sugar taste receptor. The T1R3 receptor in the FVB and B6 strains has a higher affinity for sugars than that in the AKR and 129P3 strains. In Experiment 1, mice had 40days of access to lab chow plus water, sucrose (10 or 34%), or fructose (10 or 34%) solutions. The strains consumed more of the sucrose than isocaloric fructose solutions. The pattern of strain differences in caloric intake from the 10% sugar solutions was FVB>129P3=B6>AKR; and that from the 34% sugar solutions was FVB>129P3>B6>/=AKR. Despite consuming more sugar calories, the FVB mice resisted obesity altogether. The AKR and 129P3 mice became obese exclusively on the 34% sucrose diet, while the B6 mice did so on the 34% sucrose and 34% fructose diets. In Experiment 2, we compared total caloric intake from diets containing chow versus chow plus 34% sucrose. All strains consumed between 11 and 25% more calories from the sucrose-supplemented diet. In Experiment 3, we compared the oral acceptability of the sucrose and fructose solutions, using lick tests. All strains licked more avidly for the 10% sucrose solutions. The results indicate that in mice (a) Tas1r3 genotype does not predict sugar-induced hyperphagia or obesity; (b) sucrose solutions stimulate higher daily intakes than isocaloric fructose solutions; and (c) susceptibility to sugar-induced obesity varies with strain, sugar concentration and sugar type.


Asunto(s)
Ingestión de Energía/genética , Fructosa , Hiperfagia/inducido químicamente , Obesidad/genética , Receptores Acoplados a Proteínas G/genética , Sacarosa , Administración Oral , Análisis de Varianza , Animales , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Ingestión de Energía/fisiología , Femenino , Preferencias Alimentarias/fisiología , Fructosa/administración & dosificación , Fructosa/metabolismo , Hiperfagia/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos AKR , Ratones Endogámicos C57BL , Obesidad/metabolismo , Distribución Aleatoria , Receptores Acoplados a Proteínas G/fisiología , Especificidad de la Especie , Sacarosa/administración & dosificación , Sacarosa/metabolismo , Gusto/efectos de los fármacos , Gusto/genética , Gusto/fisiología , Percepción del Gusto/genética , Factores de Tiempo , Aumento de Peso/efectos de los fármacos , Aumento de Peso/genética , Aumento de Peso/fisiología
14.
Diabetes ; 59(4): 907-15, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20103704

RESUMEN

OBJECTIVE: Because adipose tissue is highly vascularized, modifying adipose tissue vasculature may provide a novel method for reducing body fat. A peptide sequence that elicits apoptosis of endothelium in white fat potently reduced body weight. We sought to determine how inhibiting adipose tissue vasculature changes key aspects of energy balance regulation and the neuroendocrine system that maintains energy balance. RESEARCH DESIGN AND METHODS: Lean and obese mice or rats were treated with proapoptotic peptide for 4 or 27 days. Daily energy intake and expenditure were measured in mice on a low- (LFD) or high-fat diet (HFD) and in rats on a HFD. A conditioned taste aversion test was performed to assess whether proapoptotic peptide produces visceral illness. Hypothalamic neuropeptide Y, agouti-related peptide, and proopiomelanocoritin (POMC) mRNA expression and plasma leptin levels were evaluated. RESULTS: Proapoptotic peptide completely reversed HFD-induced obesity in mice and reduced body weight in mice and rats on a HFD but not in those on a LFD. Fat loss occurred with no change of energy expenditure but reduced food intake that occurred without signs of illness and despite reduced circulating leptin and reduced hypothalamic POMC gene expression, indicating that the decrease in food intake is independent of the action of leptin. CONCLUSIONS: These experiments provide compelling evidence for a previously unknown relationship between the status of adipose tissue vasculature and the regulation of food intake.


Asunto(s)
Tejido Adiposo/irrigación sanguínea , Apoptosis/fisiología , Grasas de la Dieta/farmacología , Células Endoteliales/fisiología , Péptidos/farmacología , Tejido Adiposo/efectos de los fármacos , Proteína Relacionada con Agouti/genética , Animales , Apoptosis/efectos de los fármacos , Composición Corporal , Peso Corporal/efectos de los fármacos , Dieta con Restricción de Grasas , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Células Endoteliales/efectos de los fármacos , Ingestión de Energía/efectos de los fármacos , Ingestión de Energía/genética , Hipotálamo/fisiología , Leptina/sangre , Leptina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Neovascularización Fisiológica , Neuropéptido Y/genética , Obesidad/etiología , Obesidad/genética , Obesidad/metabolismo , Fragmentos de Péptidos/farmacología , Proopiomelanocortina/genética , ARN Mensajero/genética , Ratas
15.
Biol Pharm Bull ; 32(8): 1462-8, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19652391

RESUMEN

The aim of this research was to investigate differential gene expression of cyclin-dependent kinase inhibitors (CKIs) in white adipose tissue (WAT) and liver from high-fat fed male Wistar rats with or without vitamin C (VC) supplementation (750 mg/kg of body weight). After 56 d of experimentation, animals fed on a cafeteria diet increased significantly body weights and total body fat. Reverse transcription-polymerase chain reaction (RT-PCR) studies showed that cafeteria diet decreased p21 and p57 mRNA expression in subcutaneous WAT and increased p21 mRNA in liver. Overall, these data provide new information about the role of high fat intake on mRNA levels of several CKIs with implications in adipogenesis, cell metabolism and weight homeostasis. Interestingly, VC supplementation partially prevented diet-induced adiposity and increased p27 mRNA in liver without any changes in the other tissues and genes analyzed. Thus, hepatic mRNA changes induced by ascorbic acid indicate a possible role of these genes in diet-induced oxidative stress processes.


Asunto(s)
Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/genética , Grasas de la Dieta/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Obesidad/genética , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Antioxidantes/uso terapéutico , Ácido Ascórbico/uso terapéutico , Peso Corporal/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Grasas de la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Ingestión de Energía/efectos de los fármacos , Ingestión de Energía/genética , Leptina/sangre , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Malondialdehído/metabolismo , Obesidad/etiología , Obesidad/prevención & control , Tamaño de los Órganos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , ARN Mensajero/genética , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Int J Obes (Lond) ; 33(4): 481-9, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19188926

RESUMEN

OBJECTIVE: Obese patients respond differently to weight loss interventions. No efficient diagnostic tool exists to separate obese patients into subtypes as a means to improve prediction of response to interventions. We aimed to separate obese subjects into distinct subgroups using microarray technology to identify gene expression-based subgroups to predict weight loss. DESIGN: A total of 72 obese men and women without family history of diabetes were enrolled in the study; 52 were treated with ephedra and caffeine (E+C) and 20 with placebo for 8 weeks. Adipose and skeletal muscle tissue biopsies were performed at baseline. RNA sample pairs were labeled and hybridized to oligonucleotide microarrays. Quantile normalization and gene shaving were performed, and a clustering algorithm was then applied to cluster subjects based on their gene expression profile. Clusters were visualized using heat maps and related to weight changes. RESULTS: Cluster analysis of gene expression data revealed two distinct subgroups of obesity and predicted weight loss in response to the treatment with E+C. One cluster ('red') decreased to 96.87+/-2.35% body weight, and the second cluster ('green') decreased to 95.59+/-2.75% body weight (P<0.05). 'Red' cluster had less visceral adipose tissue mass (2.77+/-1.08 vs 3.43+/-1.49 kg; P<0.05) and decreased size of the very large fat cells (1.45+/-0.61 vs 2.16+/-1.74 microl; P<0.05) compared to 'green' cluster. Gene expression for both skeletal muscle and adipose tissue was also different between clusters. CONCLUSIONS: Our study provides the first evidence that the combined approach of gene expression profiling and cluster analysis can identify discrete subtypes of obesity, these subtypes have different physiological characteristics and respond differently to an adrenergic weight loss therapy. This brings us that into an era of personalized treatment in the obesity clinic.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Grasa Intraabdominal/fisiología , Obesidad/genética , Pérdida de Peso/genética , Adulto , Algoritmos , Antropometría , Cafeína/uso terapéutico , Análisis por Conglomerados , Dieta , Ingestión de Energía/genética , Ephedra , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Obesidad/clasificación , Obesidad/tratamiento farmacológico , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Valor Predictivo de las Pruebas , Adulto Joven
17.
Obesity (Silver Spring) ; 17(5): 980-4, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19247273

RESUMEN

Oxytocin (Oxt) is secreted both peripherally and centrally and is involved in several functions including parturition, milk let-down reflex, social behavior, and food intake. Recently, it has been shown that mice deficient in Oxt receptor develop late-onset obesity. In this study, we characterized a murin model deficient in Oxt peptide (Oxt(-/-)) to evaluate food intake and body weight, glucose tolerance and insulin tolerance, leptin and adrenaline levels. We found that Oxt(-/-) mice develop late-onset obesity and hyperleptinemia without any alterations in food intake in addition to having a decreased insulin sensitivity and glucose intolerance. The lack of Oxt in our murin model also results in lower adrenalin levels which led us to hypothesize that the metabolic changes observed are associated with a decreased sympathetic nervous tone. It has been shown that Oxt neurons in the paraventricular nucleus (PVN) are a component of a leptin-sensitive signaling circuit between the hypothalamus and caudal brain stem for the regulation of food intake and energy homeostasis. Nevertheless, the lack of Oxt in these mice does not have a direct impact on feeding behavior whose regulation is probably dependent on the complex interplay of several factors. The lack of hyperphagia evident in the Oxt(-/-) mice may, in part, be attributed to the developmental compensation of other satiety factors such as cholecystokinin or bombesin-related peptides which merits further investigation. These findings identify Oxt as an important central regulator of energy homeostasis.


Asunto(s)
Ingestión de Energía/genética , Leptina/fisiología , Obesidad/fisiopatología , Oxitocina/deficiencia , Sistema Nervioso Simpático/fisiopatología , Tejido Adiposo/anatomía & histología , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Peso Corporal , Tronco Encefálico/fisiología , Tronco Encefálico/fisiopatología , Cruzamientos Genéticos , Intolerancia a la Glucosa/genética , Hipotálamo/fisiología , Hipotálamo/fisiopatología , Insulina/fisiología , Insulina/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Transducción de Señal/fisiología , Estómago/anatomía & histología
18.
Nutr Rev ; 65(12 Pt 1): 517-34, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18236691

RESUMEN

In the course of evolution, the need to eat has powerfully shaped biological structure and function. As a result, nutrient-depletion signals strongly activate neural mechanisms that orchestrate foraging, appetitive, and ingestive behaviors and at the same time instigate an internal energy savings mode through autonomic, endocrine, and peripheral cellular mechanisms. Although the hypothalamus and brainstem play crucial roles in the initiation and coordination of these responses, it is the integrated action of a much more complex and distributed neural system that is engaged in this fundamental survival reflex. In our modern, media-driven and mechanized environment of plenty, it is particularly important to recognize the neural systems responsible for learning and memory, reward, emotions and mood, decision-making and choice, and dealing with stress. These neural systems appear to powerfully assist the hypothalamic regulator in defending the lower limits of body weight, but they do little in overcoming its inherent weakness to defend over-nutrition and the upper limits of body weight and adiposity. The challenge is to define the role of these extrahypothalamic brain structures involved in the cognitive, rewarding, and emotional aspects of ingestive and physical activity behaviors and their relationship to the homeostatic regulator, and to assess the capacity of these mechanisms in predisposing to obesity.


Asunto(s)
Evolución Biológica , Encéfalo/fisiología , Metabolismo Energético/fisiología , Hipotálamo/fisiología , Obesidad/genética , Apetito/genética , Apetito/fisiología , Ingestión de Energía/genética , Ingestión de Energía/fisiología , Metabolismo Energético/genética , Homeostasis , Humanos , Vías Nerviosas/fisiología , Obesidad/etiología
19.
Herz ; 26(3): 178-84, 2001 May.
Artículo en Alemán | MEDLINE | ID: mdl-11413797

RESUMEN

BACKGROUND: The deposition of excess amounts of energy in adipose tissue is enhanced by high-fat diets and lack of physical activity. Furthermore, the existence of a specific genetic predisposition towards the development of obesity becomes evident by marked interindividual differences in the response to caloric oversupply. GENETIC DEFECTS AND HORMONES: In recent years, numerous genes and genetic defects with importance for human obesity were identified, especially through studies in animal models. The adipocyte-derived hormone leptin and its hypothalamic receptor play a premier role, as they interact with a network of proteins and neuropeptides within the regulation of food intake and energy expenditure. CONCLUSION: The search for the key molecular mechanisms in the pathogenesis of obesity will not only improve our understanding of energy metabolism, but may ultimately also lead to the development of new treatment strategies for obese patients.


Asunto(s)
Enfermedades Cardiovasculares/genética , Obesidad/genética , Receptores de Superficie Celular , Tejido Adiposo/fisiopatología , Animales , Proteínas Portadoras/genética , Ingestión de Energía/genética , Metabolismo Energético/genética , Regulación de la Expresión Génica/fisiología , Humanos , Hipotálamo/fisiología , Leptina/genética , Receptores de Leptina , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA