Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Physiol Rev ; 103(2): 1423-1485, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36422994

RESUMEN

The cephalic phase insulin response (CPIR) is classically defined as a head receptor-induced early release of insulin during eating that precedes a postabsorptive rise in blood glucose. Here we discuss, first, the various stimuli that elicit the CPIR and the sensory signaling pathways (sensory limb) involved; second, the efferent pathways that control the various endocrine events associated with eating (motor limb); and third, what is known about the central integrative processes linking the sensory and motor limbs. Fourth, in doing so, we identify open questions and problems with respect to the CPIR in general. Specifically, we consider test conditions that allow, or may not allow, the stimulus to reach the potentially relevant taste receptors and to trigger a CPIR. The possible significance of sweetness and palatability as crucial stimulus features and whether conditioning plays a role in the CPIR are also discussed. Moreover, we ponder the utility of the strict classical CPIR definition based on what is known about the effects of vagal motor neuron activation and thereby acetylcholine on the ß-cells, together with the difficulties of the accurate assessment of insulin release. Finally, we weigh the evidence of the physiological and clinical relevance of the cephalic contribution to the release of insulin that occurs during and after a meal. These points are critical for the interpretation of the existing data, and they support a sharper focus on the role of head receptors in the overall insulin response to eating rather than relying solely on the classical CPIR definition.


Asunto(s)
Insulina , Papilas Gustativas , Humanos , Insulina/metabolismo , Gusto/fisiología , Glucemia/metabolismo , Transducción de Señal
2.
Physiol Rev ; 102(2): 689-813, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34486393

RESUMEN

During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.


Asunto(s)
Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Hipotálamo/fisiología , Neuronas/fisiología , Animales , Homeostasis/fisiología , Humanos , Transducción de Señal/fisiología
3.
Diabetes Obes Metab ; 24(2): 268-280, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34658116

RESUMEN

Peripheral glucagon-like peptide-1 (GLP-1) and cholecystokinin (CCK) are secreted from enteroendocrine cells, and their plasma concentrations increase in response to eating. While the satiating effect of gut-derived CCK on food-intake control is well documented, the effect of peripheral GLP-1 is less clear. There is evidence that native GLP-1 can inhibit food intake only in the fed state but not in the fasting state. We therefore hypothesized that other gut peptides released during a meal might influence the subsequent effect of endogenous GLP-1 and investigated whether CCK could do so. We found that intraperitoneal injection of CCK in food-restricted mice inhibited food intake during the first 30-minute segment of a 1-hour session of ad libitum chow intake and that mice compensated by increasing their intake during the second half of the session. Importantly, this compensatory behaviour was abolished by an intraperitoneal injection of GLP-1 administered following an intraperitoneal injection of CCK and prior to the 1-hour session. In vivo activation of the free fatty acid 1 (FFA1) receptor with orally administered TAK875 increased plasma CCK concentration and, consistent with the effect of exogenous CCK, we found that prior oral administration of TAK875 increased the eating inhibitory effect of peripherally administered GLP-1. To examine the role of the vagus nerve in this effect, we utilized a saporin-based lesioning procedure to selectively ablate the CCK receptor-expressing gastrointestinal vagal afferent neurones (VANs). We found that the combined anorectic effect of TAK875 and GLP-1 was significantly attenuated in the absence of CCK receptor expressing VANs. Taken together, our results indicate that endogenous CCK interacts with GLP-1 to promote satiation and that activation of the FFA1 receptor can initiate this interaction by stimulating the release of CCK.


Asunto(s)
Colecistoquinina , Péptido 1 Similar al Glucagón , Animales , Ingestión de Alimentos , Humanos , Ratones , Receptores de Colecistoquinina , Saciedad/fisiología , Nervio Vago/fisiología
4.
Int J Behav Med ; 28(5): 641-646, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33721233

RESUMEN

BACKGROUND: Weight gain is common as women approach mid-life. Reduced levels of leptin, an anorexigenic hormone, may facilitate this. Studies in middle-aged women with obesity have shown that dysfunctional eating behaviour, such as restrained eating, is linked to lower leptin. Furthermore, states of low oestradiol signalling, as are found in post-menopause or anorexia nervosa, have been found to impact leptin levels. The aim of this study was to investigate, for the first time, how different aspects of dysfunctional eating, menopausal status, and a history of anorexia nervosa relate to leptin levels in normal-weight middle-aged women. METHODS: A total of N = 57 women were recruited. Thirty-one were post-menopausal, and 27 had a history of anorexia nervosa. Dysfunctional eating behaviour was measured by the Three-Factor Eating Questionnaire, which contains three subscales: susceptibility/responsiveness to hunger, restraint, and disinhibition. Body composition was assessed by bioelectrical impedance analysis. A fasting blood sample was obtained to determine leptin. RESULTS: Controlling for age, body mass index, and fat mass, susceptibility/responsiveness to hunger was positively associated with leptin (ß = 0.267, p = 0.031), whereas restrained eating (ß = - 0.183, p = 0.079) and a history of anorexia nervosa (ß = - 0.221, p = 0.059) were, by trend, negatively associated with leptin. Neither disinhibited eating nor menopausal status was related to leptin. CONCLUSIONS: Leptin may decline as a response to repeated states of a negative energy balance. A possible implication is that mid-life weight management should avoid extreme changes in eating behaviour and instead focus on the macronutrient composition of diet and physical activity. Further, longitudinal enquiries are warranted to investigate these relationships.

5.
J Neurosci ; 38(7): 1634-1647, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29326171

RESUMEN

Reduced activity of vagal efferents has long been implicated in schizophrenia and appears to be responsible for diminished parasympathetic activity and associated peripheral symptoms such as low heart rate variability and cardiovascular complications in affected individuals. In contrast, only little attention has been paid to the possibility that impaired afferent vagal signaling may be relevant for the disorder's pathophysiology as well. The present study explored this hypothesis using a model of subdiaphragmatic vagal deafferentation (SDA) in male rats. SDA represents the most complete and selective vagal deafferentation method existing to date as it leads to complete disconnection of all abdominal vagal afferents while sparing half of the abdominal vagal efferents. Using next-generation mRNA sequencing, we show that SDA leads to brain transcriptional changes in functional networks annotating with schizophrenia. We further demonstrate that SDA induces a hyperdopaminergic state, which manifests itself as increased sensitivity to acute amphetamine treatment and elevated accumbal levels of dopamine and its major metabolite, 3,4-dihydroxyphenylacetic acid. Our study also shows that SDA impairs sensorimotor gating and the attentional control of associative learning, which were assessed using the paradigms of prepulse inhibition and latent inhibition, respectively. These data provide converging evidence suggesting that the brain transcriptome, dopamine neurochemistry, and behavioral functions implicated in schizophrenia are subject to visceral modulation through abdominal vagal afferents. Our findings may encourage the further establishment and use of therapies for schizophrenia that are based on vagal interventions.SIGNIFICANCE STATEMENT The present work provides a better understanding of how disrupted vagal afferent signaling can contribute to schizophrenia-related brain and behavioral abnormalities. More specifically, it shows that subdiaphragmatic vagal deafferentation (SDA) in rats leads to (1) brain transcriptional changes in functional networks related to schizophrenia, (2) increased sensitivity to dopamine-stimulating drugs and elevated dopamine levels in the nucleus accumbens, and (3) impairments in sensorimotor gating and the attentional control of associative learning. These findings may encourage the further establishment of novel therapies for schizophrenia that are based on vagal interventions.


Asunto(s)
Abdomen/inervación , Química Encefálica/genética , Neuronas Aferentes/fisiología , Esquizofrenia/genética , Transcriptoma , Nervio Vago/fisiología , Anfetamina/farmacología , Animales , Aprendizaje por Asociación , Atención/efectos de los fármacos , Desnervación , Dopamina/metabolismo , Dopaminérgicos/farmacología , Masculino , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto , Filtrado Sensorial
6.
Am J Physiol Regul Integr Comp Physiol ; 314(5): R724-R733, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29341824

RESUMEN

Intestinal lymph supposedly provides a readout for the secretion of intestinal peptides. We here assessed how mesenteric lymph duct (MLD) lymph levels of glucagon-like peptide (GLP-1), insulin, and metabolites [glucose and triglycerides (TG)] evolve after isocaloric high- and low-fat diet (HFD and LFD) meals and how they compare with hepatic portal vein (HPV) plasma levels. Moreover, we examined the effects of intraperitoneally administered GLP-1 (1 or 10 nmol/kg) on these parameters. At 20 min after the HFD meal onset, GLP-1 levels were higher in MLD lymph than in HPV plasma. No such difference occurred with the LFD meal. Intraperitoneal injections of 10 nmol/kg GLP-1 before meals enhanced the meal-induced increases in MLD lymph and HPV plasma GLP-1 levels except for the MLD lymph levels after the HFD meal. Intraperitoneal injection of 1 nmol/kg GLP-1 only increased HPV plasma GLP-1 levels at 60 min after the HFD meal. GLP-1 injections did not increase the MLD lymph or HPV plasma GLP-1 concentrations beyond the physiological range, suggesting that intraperitoneal GLP-1 injections can recapitulate the short-term effects of endogenous GLP-1. Dipeptidyl peptidase IV (DPP-IV) activity in MLD lymph was lower than in HPV plasma, which presumably contributed to the higher levels of GLP-1 in lymph than in plasma. Insulin and glucose showed similar profiles in MLD lymph and HPV plasma, whereas TG levels were higher in lymph than in plasma. These results indicate that intestinal lymph provides a sensitive readout of intestinal peptide release and potential action, in particular when fat-rich diets are consumed.


Asunto(s)
Péptido 1 Similar al Glucagón/metabolismo , Secreciones Intestinales/metabolismo , Linfa/metabolismo , Vasos Linfáticos/metabolismo , Periodo Posprandial , Animales , Biomarcadores/metabolismo , Dieta con Restricción de Grasas , Dieta Alta en Grasa , Dipeptidil Peptidasa 4/metabolismo , Ingestión de Energía , Péptido 1 Similar al Glucagón/administración & dosificación , Péptido 1 Similar al Glucagón/sangre , Glucosa/metabolismo , Inyecciones Intraperitoneales , Insulina/metabolismo , Masculino , Ratas Sprague-Dawley , Vías Secretoras , Factores de Tiempo , Triglicéridos/metabolismo
7.
Am J Physiol Regul Integr Comp Physiol ; 315(4): R708-R720, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29847161

RESUMEN

Endogenous intestinal glucagon-like peptide-1 (GLP-1) controls satiation and glucose metabolism via vagal afferent neurons (VANs). Recently, VANs have received increasing attention for their role in brown adipose tissue (BAT) thermogenesis. It is, however, unclear whether VAN GLP-1 receptor (GLP-1R) signaling affects BAT thermogenesis and energy expenditure (EE) and whether this VAN mechanism contributes to energy balance. First, we tested the effect of the GLP-1R agonist exendin-4 (Ex4, 0.3 µg/kg ip) on EE and BAT thermogenesis and whether these effects require VAN GLP-1R signaling using a rat model with a selective Glp1r knockdown (kd) in VANs. Second, we examined the role of VAN GLP-1R in energy balance during chronic high-fat diet (HFD) feeding in VAN Glp1r kd rats. Finally, we used viral transsynaptic tracers to identify the possible neuronal substrates of such a gut-BAT interaction. VAN Glp1r kd attenuated the acute suppressive effects of Ex4 on EE and BAT thermogenesis. Consistent with this finding, the VAN Glp1r kd increased EE and BAT activity, diminished body weight gain, and improved insulin sensitivity compared with HFD-fed controls. Anterograde transsynaptic viral tracing of VANs infected major hypothalamic and hindbrain areas involved in BAT sympathetic regulation. Moreover, retrograde tracing from BAT combined with laser capture microdissection revealed that a population of VANs expressing Glp1r is synaptically connected to the BAT. Our findings reveal a novel role of VAN GLP-1R signaling in the regulation of EE and BAT thermogenesis and imply that through this gut-brain-BAT connection, intestinal GLP-1 plays a role in HFD-induced metabolic syndrome.


Asunto(s)
Tejido Adiposo Pardo/inervación , Sistema Nervioso Autónomo/metabolismo , Encéfalo/metabolismo , Metabolismo Energético , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Intestinos/inervación , Termogénesis , Animales , Sistema Nervioso Autónomo/efectos de los fármacos , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Exenatida/farmacología , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/genética , Incretinas/farmacología , Masculino , Vías Nerviosas/metabolismo , Neuronas Aferentes/metabolismo , Ratas Sprague-Dawley , Transducción de Señal , Termogénesis/efectos de los fármacos
8.
J Cell Physiol ; 232(1): 167-75, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27061934

RESUMEN

The small intestine is the main organ involved in the digestion and absorption of nutrients. It is in an ideal position to sense the availability of energy in the lumen in addition to its absorptive function. Consumption of a high-fat diet (HFD) influences the metabolic characteristics of the small intestine. Therefore, to better understand the metabolic features of the small intestine and their changes in response to dietary fat, we characterized the metabolism of duodenal, jejunal, and hepatic cell lines and assessed the metabolic changes in the enterocytes and the liver after short-term (3 days) or medium-term (14 days) HFD feeding in mice. Experiments with immortalized enterocytes indicated a higher glycolytic capacity in the duodenal cell line compared to the other two cell lines, whereas the jejunal cell line exhibited a high oxidative metabolism. Short-term HFD feeding induced changes in the expression of glucose and lipid metabolism-related genes in the duodenum and the jejunum of mice, but not in the liver. When focusing on fatty acid oxidation both, short- and medium-term HFD feeding induced an upregulation of 3-hydroxy-3-methylglutaryl-coenzyme A, the key enzyme of ketogenesis, at the protein level in the intestinal epithelial cells, but not in the liver. These results suggest that HFD feeding induces an early adaptation of the small intestine rather than the liver in response to a substantial fat load. This highlights the importance of the small intestine in the adaptation of the body to the metabolic changes induced by HFD exposure. J. Cell. Physiol. 232: 167-175, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Dieta Alta en Grasa , Enterocitos/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Obesidad/metabolismo , Adaptación Fisiológica/efectos de los fármacos , Composición Corporal/efectos de los fármacos , Composición Corporal/fisiología , Grasas de la Dieta/metabolismo , Enterocitos/metabolismo , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Factores de Tiempo
9.
Neurobiol Learn Mem ; 142(Pt B): 190-199, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28499738

RESUMEN

Vagal afferents are a crucial neuronal component of the gut-brain axis and mediate the information flow from the viscera to the central nervous system. Based on the findings provided by experiments involving vagus nerve stimulation, it has been suggested that vagal afferent signaling may influence various cognitive functions such as recognition memory and cognitive flexibility. Here, we examined this hypothesis using a rat model of subdiaphragmatic vagal deafferentation (SDA), the most complete and selective abdominal vagal deafferentation method existing to date. We found that SDA did not affect working memory in a nonspatial alternation task, nor did it influence short-, intermediate-, and long-term object recognition memory. SDA did also not affect the acquisition of positively reinforced left-right discrimination learning, but it facilitated the subsequent reversal left-right discrimination learning. The SDA-induced effects on reversal learning emerged in the absence of concomitant changes in motivation towards the positive reinforcer, indicating selective effects on cognitive flexibility. Taken together, these findings suggest that the relative contribution of vagal afferent signaling to cognitive functions is limited. At the same time, our study demonstrates that cognitive flexibility, at least in the domains of positively reinforced learning, is subjected to visceral modulation through abdominal vagal afferents.


Asunto(s)
Vías Aferentes/fisiología , Memoria a Corto Plazo/fisiología , Reconocimiento en Psicología/fisiología , Aprendizaje Inverso/fisiología , Nervio Vago/fisiología , Abdomen/inervación , Vías Aferentes/cirugía , Animales , Conducta Animal/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Nervio Vago/cirugía
10.
Front Neuroendocrinol ; 38: 1-11, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25527432

RESUMEN

The gastrointestinal hormone peptide tyrosine tyrosine 3-36 (PYY(3-36)) has attained broad recognition with respect to its involvement in energy homeostasis and the control of food intake. It is mainly secreted by distal intestinal enteroendocrine L-cells in response to eating and exerts neurally mediated, paracrine and endocrine effects on various target organs. In addition to its gastrointestinal effects, PYY(3-36) has long been known to inhibit food intake. Recent closer examination of the effects of PYY(3-36) revealed that this gut-derived peptide also influences a wide spectrum of behavioral and cognitive functions that are pivotal for basic processes of perception and judgment, including central information processing, salience learning, working memory, and behavioral responding to novelty. Here, we review the effects of PYY(3-36) that go beyond food intake and provide a conceptual framework suggesting that several apparently unrelated behavioral actions of PYY(3-36) may actually reflect different manifestations of modulating the central dopamine system.


Asunto(s)
Dopamina/metabolismo , Ingestión de Alimentos/fisiología , Homeostasis/fisiología , Péptido YY/metabolismo , Animales , Absorción Gastrointestinal/fisiología , Humanos
11.
Am J Physiol Regul Integr Comp Physiol ; 311(3): R618-27, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27488889

RESUMEN

Thylakoids reduce body weight gain and body fat accumulation in rodents. This study investigated whether an enhanced oxidation of dietary fat-derived fatty acids in the intestine contributes to the thylakoid effects. Male Sprague-Dawley rats were fed a high-fat diet with (n = 8) or without thylakoids (n = 8) for 2 wk. Body weight, food intake, and body fat were measured, and intestinal mucosa was collected and analyzed. Quantitative real-time PCR was used to measure gene expression levels of key enzymes involved in fatty acid transport, fatty acid oxidation, and ketogenesis. Another set of thylakoid-treated (n = 10) and control rats (n = 10) went through indirect calorimetry. In the first experiment, thylakoid-treated rats (n = 8) accumulated 25% less visceral fat than controls. Furthermore, fatty acid translocase (Fat/Cd36), carnitine palmitoyltransferase 1a (Cpt1a), and mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase 2 (Hmgcs2) genes were upregulated in the jejunum of the thylakoid-treated group. In the second experiment, thylakoid-treated rats (n = 10) gained 17.5% less weight compared with controls and their respiratory quotient was lower, 0.86 compared with 0.91. Thylakoid-intake resulted in decreased food intake and did not cause steatorrhea. These results suggest that thylakoids stimulated intestinal fatty acid oxidation and ketogenesis, resulting in an increased ability of the intestine to handle dietary fat. The increased fatty acid oxidation and the resulting reduction in food intake may contribute to the reduced fat accumulation in thylakoid-treated animals.


Asunto(s)
Dieta Alta en Grasa , Ácidos Grasos/metabolismo , Mucosa Intestinal/metabolismo , Grasa Intraabdominal/fisiología , Tilacoides/metabolismo , Regulación hacia Arriba/fisiología , Animales , Masculino , Tamaño de los Órganos/fisiología , Oxidación-Reducción , Ratas , Ratas Sprague-Dawley , Tilacoides/química
12.
J Neurosci ; 34(21): 7067-76, 2014 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-24849343

RESUMEN

Vagal afferents are an important neuronal component of the gut-brain axis allowing bottom-up information flow from the viscera to the CNS. In addition to its role in ingestive behavior, vagal afferent signaling has been implicated modulating mood and affect, including distinct forms of anxiety and fear. Here, we used a rat model of subdiaphragmatic vagal deafferentation (SDA), the most complete and selective vagal deafferentation method existing to date, to study the consequences of complete disconnection of abdominal vagal afferents on innate anxiety, conditioned fear, and neurochemical parameters in the limbic system. We found that compared with Sham controls, SDA rats consistently displayed reduced innate anxiety-like behavior in three procedures commonly used in preclinical rodent models of anxiety, namely the elevated plus maze test, open field test, and food neophobia test. On the other hand, SDA rats exhibited increased expression of auditory-cued fear conditioning, which specifically emerged as attenuated extinction of conditioned fear during the tone re-exposure test. The behavioral manifestations in SDA rats were associated with region-dependent changes in noradrenaline and GABA levels in key areas of the limbic system, but not with functional alterations in the hypothalamus-pituitary-adrenal grand stress. Our study demonstrates that innate anxiety and learned fear are both subjected to visceral modulation through abdominal vagal afferents, possibly via changing limbic neurotransmitter systems. These data add further weight to theories emphasizing an important role of afferent visceral signals in the regulation of emotional behavior.


Asunto(s)
Ansiedad/fisiopatología , Condicionamiento Clásico/fisiología , Miedo/fisiología , Tracto Gastrointestinal/inervación , Nervio Vago/fisiología , Animales , Condicionamiento Clásico/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Extinción Psicológica/efectos de los fármacos , Extinción Psicológica/fisiología , Miedo/psicología , Lateralidad Funcional , Tracto Gastrointestinal/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Neurotransmisores/metabolismo , Ratas , Ratas Sprague-Dawley , Sincalida/farmacología , Nervio Vago/efectos de los fármacos
13.
Rev Geophys ; 53(2): 323-361, 2015 06.
Artículo en Inglés | MEDLINE | ID: mdl-27478878

RESUMEN

Regional climate modeling using convection-permitting models (CPMs; horizontal grid spacing <4 km) emerges as a promising framework to provide more reliable climate information on regional to local scales compared to traditionally used large-scale models (LSMs; horizontal grid spacing >10 km). CPMs no longer rely on convection parameterization schemes, which had been identified as a major source of errors and uncertainties in LSMs. Moreover, CPMs allow for a more accurate representation of surface and orography fields. The drawback of CPMs is the high demand on computational resources. For this reason, first CPM climate simulations only appeared a decade ago. In this study, we aim to provide a common basis for CPM climate simulations by giving a holistic review of the topic. The most important components in CPMs such as physical parameterizations and dynamical formulations are discussed critically. An overview of weaknesses and an outlook on required future developments is provided. Most importantly, this review presents the consolidated outcome of studies that addressed the added value of CPM climate simulations compared to LSMs. Improvements are evident mostly for climate statistics related to deep convection, mountainous regions, or extreme events. The climate change signals of CPM simulations suggest an increase in flash floods, changes in hail storm characteristics, and reductions in the snowpack over mountains. In conclusion, CPMs are a very promising tool for future climate research. However, coordinated modeling programs are crucially needed to advance parameterizations of unresolved physics and to assess the full potential of CPMs.

14.
Am J Physiol Regul Integr Comp Physiol ; 309(1): R1-R12, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25924881

RESUMEN

Prenatal immunological adversities such as maternal infection have been widely acknowledged to contribute to an increased risk of neurodevelopmental brain disorders. In recent years, epidemiological and experimental evidence has accumulated to suggest that prenatal exposure to immune challenges can also negatively affect various physiological and metabolic functions beyond those typically associated with primary defects in CNS development. These peripheral changes include excessive accumulation of adipose tissue and increased body weight, impaired glycemic regulation and insulin resistance, altered myeloid lineage development, increased gut permeability, hyperpurinergia, and changes in microbiota composition. Experimental work in animal models further suggests that at least some of these peripheral abnormalities could directly contribute to CNS dysfunctions, so that normalization of peripheral pathologies could lead to an amelioration of behavioral deficits. Hence, seemingly unrelated central and peripheral effects of prenatal infection could represent interrelated pathological entities that emerge in response to a common developmental stressor. Targeting peripheral abnormalities may thus represent a valuable strategy to improve the wide spectrum of behavioral abnormalities that can emerge in subjects with prenatal infection histories.


Asunto(s)
Sistema Nervioso Central , Enfermedades Transmisibles/complicaciones , Enfermedades Transmisibles/microbiología , Complicaciones Infecciosas del Embarazo , Efectos Tardíos de la Exposición Prenatal , Adiposidad , Factores de Edad , Animales , Bacterias/inmunología , Bacterias/patogenicidad , Peso Corporal , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/microbiología , Sistema Nervioso Central/fisiopatología , Enfermedades Transmisibles/inmunología , Enfermedades Transmisibles/fisiopatología , Disbiosis , Metabolismo Energético , Femenino , Tracto Gastrointestinal/microbiología , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/microbiología , Complicaciones Infecciosas del Embarazo/fisiopatología , Factores de Riesgo , Transducción de Señal , Factores de Tiempo
15.
Am J Physiol Regul Integr Comp Physiol ; 308(2): R131-7, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25427767

RESUMEN

Hepatic fatty acid oxidation (FAO) has long been implicated in the control of eating. Nevertheless, direct evidence for a causal relationship between changes in hepatic FAO and changes in food intake is still missing. Here we tested whether increasing hepatic FAO via adenovirus-mediated expression of a mutated form of the key regulatory enzyme of mitochondrial FAO carnitine palmitoyltransferase 1A (CPT1mt), which is active but insensitive to inhibition by malonyl-CoA, affects eating and metabolism in mice. CPT1mt expression increased hepatocellular CPT1 protein levels. This resulted in an increase in circulating ketone body levels in fasted CPT1mt-expressing mice, suggesting an increase in hepatic FAO. These mice did not show any significant changes in cumulative food intake, energy expenditure, or respiratory quotient after 4-h food deprivation. After 24-h food deprivation, however, the CPT1mt-expressing mice displayed increased food intake. Thus expression of CPT1mt in the liver increases hepatic FAO capacity, but does not inhibit eating. Rather, it may even stimulate eating after prolonged food deprivation. These data do not support the hypothesis that an increase in hepatic FAO decreases food intake.


Asunto(s)
Carnitina O-Palmitoiltransferasa/metabolismo , Ingestión de Alimentos/fisiología , Ácidos Grasos/metabolismo , Metabolismo de los Lípidos/fisiología , Hígado/metabolismo , Mitocondrias/metabolismo , Animales , Metabolismo Energético/fisiología , Privación de Alimentos/fisiología , Masculino , Ratones Endogámicos C57BL , Modelos Animales , Oxidación-Reducción
16.
Proc Natl Acad Sci U S A ; 109(29): 11711-6, 2012 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-22753483

RESUMEN

A low fat oxidative capacity has been linked to muscle diacylglycerol (DAG) accumulation and insulin resistance. Alternatively, a low fat oxidation rate may stimulate glucose oxidation, thereby enhancing glucose disposal. Here, we investigated whether an ethyl-2-[6-(4-chlorophenoxy)hexyl]-oxirane-2-carboxylate (etomoxir)-induced inhibition of fat oxidation leads to muscle fat storage and insulin resistance. An intervention in healthy male subjects was combined with studies in human primary myotubes. Furthermore, muscle DAG and triacylglycerol (TAG), mitochondrial function, and insulin signaling were examined in etomoxir-treated C57bl6 mice. In humans, etomoxir administration increased glucose oxidation at the expense of fat oxidation. This effect was accompanied by an increased abundance of GLUT4 at the sarcolemma and a lowering of plasma glucose levels, indicative of improved glucose homeostasis. In mice, etomoxir injections resulted in accumulation of muscle TAG and DAG, yet improved insulin-stimulated GLUT4 translocation. Also in human myotubes, insulin signaling was improved by etomoxir, in the presence of increased intramyocellular lipid accumulation. These insulin-sensitizing effects in mice and human myotubes were accompanied by increased phosphorylation of AMP-activated protein kinase (AMPK). Our results show that a reduction in fat oxidation leading to accumulation of muscle DAG does not necessarily lead to insulin resistance, as the reduction in fat oxidation may activate AMPK.


Asunto(s)
Diglicéridos/metabolismo , Compuestos Epoxi/farmacología , Ácidos Grasos/metabolismo , Resistencia a la Insulina/fisiología , Mitocondrias/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Triglicéridos/metabolismo , Análisis de Varianza , Animales , Western Blotting , Técnica del Anticuerpo Fluorescente , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Sarcolema/metabolismo
18.
Am J Physiol Regul Integr Comp Physiol ; 307(2): R167-78, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24829501

RESUMEN

The endogenous lipid messenger oleoylethanolamide (OEA) inhibits eating and modulates fat metabolism supposedly through the activation of peroxisome proliferator-activated receptor-α (PPARα) and vagal sensory fibers. We tested in adult male rats whether OEA stimulates fatty acid oxidation (FAO) and ketogenesis and whether it increases plasma levels of the satiating gut peptides glucagon-like peptide-1 (GLP-1) and peptide YY (PYY). We also explored whether OEA still inhibits eating after subdiaphragmatic vagal deafferentation (SDA). We found that intraperitoneally injected OEA (10 mg/kg body wt) reduced (P < 0.05) food intake mainly by increasing meal latency and that this effect was stronger in rats fed a 60% high-fat diet (HFD) than in chow-fed rats. OEA increased (P < 0.05) postprandial plasma nonesterified fatty acids and ß-hydroxybutyrate (BHB) in the hepatic portal vein (HPV) and vena cava (VC) 30 min after injection, which was more pronounced in HFD- than in chow-fed rats. OEA also increased the protein expression of the key ketogenetic enzyme, mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase, in the jejunum of HFD-fed rats, but not in the liver or duodenum of either diet group. Furthermore, OEA decreased GLP-1 and PYY concentrations (P < 0.05) in the HPV and VC 30 min after administration. Finally, OEA reduced food intake in SDA and sham-operated rats similarly. Our findings indicate that neither intact abdominal vagal afferents nor prandial increases in GLP-1 or


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Tracto Gastrointestinal/efectos de los fármacos , Metabolismo de los Lípidos , Ácidos Oléicos/farmacología , Saciedad/efectos de los fármacos , Animales , Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos/fisiología , Endocannabinoides , Tracto Gastrointestinal/inervación , Tracto Gastrointestinal/metabolismo , Péptido 1 Similar al Glucagón/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/fisiología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ácidos Oléicos/administración & dosificación , Ratas , Ratas Sprague-Dawley , Saciedad/fisiología , Nervio Vago/cirugía
19.
Int J Neuropsychopharmacol ; 17(3): 407-19, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24131590

RESUMEN

The gastrointestinal hormone PYY(3-36) is a preferential Y2 neuropeptide Y (NPY) receptor agonist. Recent evidence indicates that PYY(3-36) acts on central dopaminergic pathways, but its influence on dopamine-dependent behaviours remains largely unknown. We therefore explored the effects of peripheral PYY(3-36) treatment on the behavioural responses to novelty and to dopamine-activating drugs in mice. In addition, we examined whether PYY(3-36) administration may activate distinct dopamine and γ-aminobutyric acid (GABA) cell populations in the mesoaccumbal and nigrostriatal pathways. We found that i.p. PYY(3-36) injection led to a dose-dependent increase in novel object exploration. The effective dose of PYY(3-36) (1 µg/100 g body weight) also potentiated the locomotor reaction to the indirect dopamine receptor agonist amphetamine and increased stereotyped climbing/leaning responses following administration of the direct dopamine receptor agonist apomorphine. PYY(3-36) administration did not affect activity of midbrain dopaminergic cells as evaluated by double immuno-enzyme staining of the neuronal early gene product c-Fos with tyrosine hydroxylase. PYY(3-36) did, however, lead to a marked increase in the number of cells co-expressing c-Fos with glutamic acid decarboxylase in the nucleus accumbens and caudate putamen, indicating activation of GABAergic cells in dorsal and ventral striatal areas. Our results support the hypothesis that acute administration of the preferential Y2 receptor agonist PYY(3-36) modulates dopamine-dependent behaviours. These effects do not seem to involve direct activation of midbrain dopamine cells but instead are associated with neuronal activation in the major input areas of the mesoaccumbal and nigrostriatal pathways.


Asunto(s)
Dopaminérgicos/farmacología , Conducta Exploratoria/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Péptido YY/farmacología , Receptores de Neuropéptido Y/agonistas , Anfetamina/farmacología , Análisis de Varianza , Animales , Apomorfina/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Glutamato Descarboxilasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Conducta Estereotipada/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo
20.
J Lipid Res ; 54(5): 1369-84, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23449193

RESUMEN

Acyl CoA:diacylglycerol acyltransferase-1 (DGAT-1) catalyzes the final step in triacylglycerol (TAG) synthesis and is highly expressed in the small intestine. Because DGAT-1 knockout mice are resistant to diet-induced obesity, we investigated the acute effects of intragastric (IG) infusion of a small molecule diacylglycerol acyltransferase-1 inhibitor (DGAT-1i) on eating, circulating fat metabolites, indirect calorimetry, and hepatic and intestinal expression of key fat catabolism enzymes in male rats adapted to an 8 h feeding-16 h deprivation schedule. Also, the DGAT-1i effect on fatty acid oxidation (FAO) was investigated in enterocyte cell culture models. IG DGAT-1i infusions reduced energy intake compared with vehicle in high-fat diet (HFD)-fed rats, but scarcely in chow-fed rats. IG DGAT-1i also blunted the postprandial increase in serum TAG and increased ß-hydroxybutyrate levels only in HFD-fed rats, in which it lowered the respiratory quotient and increased intestinal, but not hepatic, protein levels of Complex III of the mitochondrial respiratory chain and of mitochondrial hydroxymethylglutaryl-CoA synthase. Finally, the DGAT-1i enhanced FAO in CaCo2 (EC50 = 0.3494) and HuTu80 (EC50 = 0.00762) cells. Thus, pharmacological DGAT-1 inhibition leads to an increase in intestinal FAO and ketogenesis when dietary fat is available. This may contribute to the observed eating-inhibitory effect.


Asunto(s)
Diacilglicerol O-Acetiltransferasa/metabolismo , Ácidos Grasos/metabolismo , Mucosa Intestinal/metabolismo , Oxidación-Reducción , Acilcoenzima A/metabolismo , Animales , Diacilglicerol O-Acetiltransferasa/antagonistas & inhibidores , Dieta Alta en Grasa , Complejo III de Transporte de Electrones/metabolismo , Ingestión de Energía , Humanos , Intestinos/enzimología , Hígado/enzimología , Hígado/metabolismo , Masculino , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA