Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Med ; 20: 179-90, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24687160

RESUMEN

Chronic stress has deleterious effects on immune function, which can lead to adverse health outcomes. However, studies investigating the impact of stress reduction interventions on immunity in clinical research have yielded divergent results, potentially stemming from differences in study design and genetic heterogeneity, among other clinical research challenges. To test the hypothesis that reducing glucocorticoid levels enhances certain immune functions, we administered influenza vaccine once (prime) or twice (boost) to mice housed in either standard control caging or environmental enrichment (EE) caging. We have shown that this approach reduces mouse corticosterone production. Compared with controls, EE mice had significantly lower levels of fecal corticosterone metabolites (FCMs) and increased splenic B and T lymphocyte numbers. Corticosterone levels were negatively associated with the numbers of CD19(+) (r(2) = 0.43, p = 0.0017), CD4(+) (r(2) = 0.28, p = 0.0154) and CD8(+) cells (r(2) = 0.20, p = 0.0503). Vaccinated mice showed nonsignificant differences in immunoglobulin G (IgG) titer between caging groups, although EE mice tended to exhibit larger increases in titer from prime to boost than controls; the interaction between the caging group (control versus EE) and vaccine group (prime versus boost) showed a strong statistical trend (cage-group*vaccine-group, F = 4.27, p = 0.0555), suggesting that there may be distinct effects of EE caging on primary versus secondary IgG vaccine responses. Vaccine-stimulated splenocytes from boosted EE mice had a significantly greater frequency of interleukin 5 (IL-5)-secreting cells than boosted controls (mean difference 7.7, IL-5 spot-forming units/10(6) splenocytes, 95% confidence interval 0.24-135.1, p = 0.0493) and showed a greater increase in the frequency of IL-5-secreting cells from prime to boost. Our results suggest that corticosterone reduction via EE caging was associated with enhanced secondary vaccine responses, but had little effect on primary responses in mice. These findings help identify differences in primary and secondary vaccine responses in relationship to stress mediators that may be relevant in clinical studies.


Asunto(s)
Linfocitos B/inmunología , Corticosterona/metabolismo , Citocinas/metabolismo , Vacunas contra la Influenza/inmunología , Bazo/inmunología , Linfocitos T/inmunología , Animales , Ambiente Controlado , Inmunización Secundaria , Masculino , Ratones , Ratones Endogámicos BALB C
3.
Mol Med ; 18: 606-17, 2012 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-22398685

RESUMEN

Chronic stress is associated with negative health outcomes and is linked with neuroendocrine changes, deleterious effects on innate and adaptive immunity, and central nervous system neuropathology. Although stress management is commonly advocated clinically, there is insufficient mechanistic understanding of how decreasing stress affects disease pathogenesis. Therefore, we have developed a "calm mouse model" with caging enhancements designed to reduce murine stress. Male BALB/c mice were divided into four groups: control (Cntl), standard caging; calm (Calm), large caging to reduce animal density, a cardboard nest box for shelter, paper nesting material to promote innate nesting behavior, and a polycarbonate tube to mimic tunneling; control exercise (Cntl Ex), standard caging with a running wheel, known to reduce stress; and calm exercise (Calm Ex), calm caging with a running wheel. Calm, Cntl Ex and Calm Ex animals exhibited significantly less corticosterone production than Cntl animals. We also observed changes in spleen mass, and in vitro splenocyte studies demonstrated that Calm Ex animals had innate and adaptive immune responses that were more sensitive to acute handling stress than those in Cntl. Calm animals gained greater body mass than Cntl, although they had similar food intake, and we also observed changes in body composition, using magnetic resonance imaging. Together, our results suggest that the Calm mouse model represents a promising approach to studying the biological effects of stress reduction in the context of health and in conjunction with existing disease models.


Asunto(s)
Ratones , Modelos Animales , Estrés Psicológico , Animales , Conducta Animal , Composición Corporal , Peso Corporal , Corticosterona/biosíntesis , Modelos Animales de Enfermedad , Ingestión de Energía , Ambiente , Masculino , Ratones Endogámicos BALB C , Hormonas Peptídicas/sangre , Condicionamiento Físico Animal
4.
Psychoneuroendocrinology ; 36(10): 1513-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21906885

RESUMEN

Chronically stressed rodents who are allowed to eat calorie-dense "comfort" food develop greater mesenteric fat, which in turn dampens hypothalamic-pituitary-adrenocortical (HPA) axis activity. We tested whether similar relations exist in humans, at least cross-sectionally. Fifty-nine healthy premenopausal women were exposed to a standard laboratory stressor to examine HPA response to acute stress and underwent diurnal saliva sampling for basal cortisol and response to dexamethasone administration. Based on perceived stress scores, women were divided into extreme quartiles of low versus high stress categories. We found as hypothesized that the high stress group had significantly greater BMI and sagittal diameter, and reported greater emotional eating. In response to acute lab stressor, the high stress group showed a blunted cortisol response, lower diurnal cortisol levels, and greater suppression in response to dexamethasone. These cross-sectional findings support the animal model, which suggests that long-term adaptation to chronic stress in the face of dense calories result in greater visceral fat accumulation (via ingestion of calorie-dense food), which in turn modulates HPA axis response, resulting in lower cortisol levels.


Asunto(s)
Adaptación Psicológica/fisiología , Ingestión de Alimentos/psicología , Alimentos , Red Nerviosa/fisiología , Estrés Psicológico/prevención & control , Estrés Psicológico/psicología , Adulto , Índice de Masa Corporal , Cuidadores/psicología , Niño , Enfermedad Crónica , Depresión/psicología , Femenino , Humanos , Sistema Hipotálamo-Hipofisario/fisiología , Persona de Mediana Edad , Sistema Hipófiso-Suprarrenal/fisiología , Índice de Severidad de la Enfermedad , Estrés Psicológico/fisiopatología
5.
Reprod Sci ; 17(11): 1024-8, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20713970

RESUMEN

The metabolic state has long been shown to affect reproduction. Peripheral signals and hormones from the reproductive organs are also known to regulate energy metabolism and feeding and energy expenditure. Much attention has been paid to determine the signaling flow from key hypothalamic neuronal populations, including those producing the anorexigenic proopiomelanocortin (POMC) derivate, α-melanocyte stimulating hormone (α-MSH), to the medial preoptic area gonadotropin-releasing hormone (GnRH) neurons, cells that are the drivers of ovulation and reproduction in general. In this study, the authors explored whether a reverse signaling modality may also exist. Specifically, the authors analyzed GnRH efferents in the arcuate nucleus with particular emphasis on their anatomical proximity to arcuate nucleus melanocortin perikarya. Using correlated light and electron microscopy, the authors observed direct apposition between GnRH-containing axon terminals and POMC cell bodies. These data provide the first experimental evidence to suggest that GnRH may have a direct influence on feeding, energy expenditure, and glucose homeostasis, independent of the activity of the gonadal axis.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/ultraestructura , Hormona Liberadora de Gonadotropina/metabolismo , Fibras Nerviosas/metabolismo , Neuronas/metabolismo , Neuronas/ultraestructura , Proopiomelanocortina/metabolismo , Animales , Femenino , Ratones , Fibras Nerviosas/ultraestructura , Terminales Presinápticos/metabolismo , Terminales Presinápticos/ultraestructura
6.
Stress ; 13(3): 248-57, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20392196

RESUMEN

The hypothalamic-pituitary-adrenal (HPA) axis self-regulates through a glucocorticoid negative feedback mechanism that is stereotypically slow and long lasting. Rapid (seconds to minutes) glucocorticoid feedback, however, inhibits stress-induced adrenocorticotropic hormone (ACTH) secretion too quickly to result from classic transcriptional effects of the occupied glucocorticoid receptor. Cannabinoids may act as rapid intermediary messengers between glucocorticoids and HPA activation via retroactive inhibition of afferent glutamate stimulation of the corticotropin-releasing factor neurons in the paraventricular nucleus. We demonstrated fast feedback effects of GR stimulation and blockade and observed the effect of cannabinoid receptor (CB1) antagonist AM251 on HPA axis reactivity in vivo. Rats were injected intraperitoneally with varying doses of the specific GR agonist RU28362, the GR antagonist RU486, or AM251 2 min before restraint. Blood was collected at predetermined times and corticosterone and ACTH concentrations were measured. RU28362 blunted stress-induced ACTH secretion while RU486 and AM251 significantly increased stress-induced ACTH release 15 min after restraint onset. Next, we injected AM251 58 min before RU28362, 2 min before restraint, to determine if inhibition of ACTH by RU28362 was contingent on CB1 activation. Unexpectedly, CB1 blockade failed to prevent glucocorticoid negative feedback and instead enhanced it. These studies not only establish an in vivo fast feedback model but show that rapid glucococorticoid negative feedback is similarly altered by GR and CB1 blockade. Although the hormonal consequences of acute AM251 treatment were strikingly similar to those of RU486 treatment, we are unable to draw conclusions about the serial nature of the interaction between GR activation and CB release from these results.


Asunto(s)
Cannabinoides/metabolismo , Glucocorticoides/metabolismo , Sistema Hipotálamo-Hipofisario/fisiología , Sistema Hipófiso-Suprarrenal/fisiología , Estrés Fisiológico/fisiología , Hormona Adrenocorticotrópica/metabolismo , Androstanoles/farmacología , Animales , Relación Dosis-Respuesta a Droga , Retroalimentación Fisiológica/efectos de los fármacos , Retroalimentación Fisiológica/fisiología , Antagonistas de Hormonas/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Mifepristona/farmacología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Piperidinas/farmacología , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/metabolismo , Pirazoles/farmacología , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB2/metabolismo , Receptores de Glutamato/metabolismo , Restricción Física , Estrés Fisiológico/efectos de los fármacos
7.
Trends Endocrinol Metab ; 21(3): 159-65, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19926299

RESUMEN

Stress and emotional brain networks foster eating behaviors that can lead to obesity. The neural networks underlying the complex interactions among stressors, body, brain and food intake are now better understood. Stressors, by activating a neural stress-response network, bias cognition toward increased emotional activity and degraded executive function. This causes formed habits to be used rather than a cognitive appraisal of responses. Stress also induces secretion of glucocorticoids, which increases motivation for food, and insulin, which promotes food intake and obesity. Pleasurable feeding then reduces activity in the stress-response network, reinforcing the feeding habit. These effects of stressors emphasize the importance of teaching mental reappraisal techniques to restore responses from habitual to thoughtful, thus battling stress-induced obesity.


Asunto(s)
Conducta Alimentaria/psicología , Obesidad/etiología , Obesidad/psicología , Estrés Psicológico/fisiopatología , Animales , Glucocorticoides/metabolismo , Humanos , Insulina/metabolismo , Sistema Nervioso/metabolismo
8.
Am J Physiol Regul Integr Comp Physiol ; 296(5): R1366-75, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19279289

RESUMEN

Corticosterone and insulin play complex roles in the amount and composition of calories ingested, and the utilization and deposition of this energy. Understanding the interplay of these two hormones is complicated because increasing concentrations of corticosterone dose-dependently increase circulating insulin levels. We addressed individual contributions of each hormone by controlling, at steady-state levels, corticosterone (by adrenalectomy and exogenous replacement) and insulin (by streptozotocin-induced destruction of pancreatic beta-cells and exogenous replacement) across a spectrum of concentrations in rats, creating 8 hormonal combinations. For 5 days after surgery, all rats received chow. At day 5, they were subdivided into those that continued to receive chow and those that had a choice between chow, lard, and 32% sucrose for a further 5 days. During the choice/chow period, total calories ingested were stimulated by corticosterone and choice diet, and subject to a corticosterone-insulin interaction. Sucrose, but not lard, intake was stimulated by insulin. Body weight was increased by insulin, decreased by high corticosterone, and unaffected by diet. White adipose tissue depot weights were stimulated by insulin, corticosterone, and diet. Plasma triglycerides, free fatty acids, total ketone bodies, glucose, and glycerol were all significantly increased by corticosterone and the choice diet but inhibited by insulin. In contrast, plasma leptin was only increased by insulin and diet, plasma glucagon and liver glycogen was only affected by insulin and liver triglycerides, and arcuate nucleus proopiomelanocortin mRNA was only influenced by diet. Collectively, these data show that corticosterone and insulin determine the intake, form, and compartmentalization of energy both independently and interactively.


Asunto(s)
Corticosterona/fisiología , Ingestión de Energía/fisiología , Metabolismo Energético/fisiología , Insulina/fisiología , Adrenalectomía , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Peso Corporal/fisiología , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Glucógeno/metabolismo , Leptina/metabolismo , Hígado/metabolismo , Masculino , Neuropéptido Y/metabolismo , Proopiomelanocortina/metabolismo , Ratas , Ratas Sprague-Dawley , Estreptozocina , Triglicéridos/metabolismo
9.
Ann Surg ; 249(2): 269-76, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19212181

RESUMEN

OBJECTIVE: We sought to examine insulin-sensitive food intake behavior and neuroendocrine and metabolic variables of rats that had undergone a duodenal-jejunal bypass (DJB). SUMMARY OF BACKGROUND DATA: A DJB that circumvents the duodenum and proximal jejunum while leaving the stomach unperturbed rapidly improves insulin sensitivity in type 2 diabetic rats. This segment of proximal small intestine is innervated by the gastroduodenal branch of the vagus nerve, the transection of which influences food intake choices in streptozotocin-diabetic rats. METHODS: Rats were first placed on a choice of chow and lard for 7 days and additionally provided with an enriched liquid diet for another 7 days before surgery and were allowed only the liquid diet for 7 days after either a sham or DJB operation. RESULTS: After surgery, DJB-operated rats initially consumed less than the sham-operated counterparts. When the rats were subsequently provided with the choice of chow and lard for 7 days, there were no differences in intake between the DJB and sham-operated groups. Similarly, the majority of metabolic and neuroendocrine variables measured were unchanged. However, DJB-operated rats exhibited greater mesenteric white adipose tissue weight, fecal output, arcuate nucleus neuropeptide Y mRNA expression, plasma corticosterone, and glucagon levels together with reduced plasma leptin concentrations. CONCLUSIONS: DJB surgery does not produce significant differences in food intake choices after a period of recovery; however, there are enduring metabolic and neuroendocrine changes, which are collectively important to understanding the beneficial outcomes of the operation.


Asunto(s)
Derivación Yeyunoileal , Obesidad Mórbida/metabolismo , Obesidad Mórbida/cirugía , Animales , Modelos Animales de Enfermedad , Ingestión de Alimentos , Resistencia a la Insulina/fisiología , Intestino Delgado/metabolismo , Intestino Delgado/fisiopatología , Masculino , Sistemas Neurosecretores/fisiopatología , Periodo Posoperatorio , Ratas , Ratas Sprague-Dawley
10.
Physiol Behav ; 96(4-5): 651-61, 2009 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-19162053

RESUMEN

Previous studies have shown that the successive negative contrast procedure, in which food-restricted rats entrained to once daily, brief presentations of 32% sucrose are unexpectedly shifted to a 4% solution, results in an adrenocortical response on the second, but not the first postshift day. We attempted to generalize that finding in our own procedure. In Experiment 1, two groups of rats were given a 32% sucrose solution once daily in their home cages for 14 days before being shifted to a 4% solution. One group was killed 10 min after the first 4% solution and one was killed after the second 4% solution. In addition, two groups receiving either 32% or 4% sucrose throughout the experiment served as unshifted controls. In contrast to previous findings, both shifted groups exhibited prominent adrenocorticotropin hormone (ACTH) and adrenocortical (B) responses on both postshift days compared to unshifted controls, which did not differ from one another. Experiment 2a employed distinctive contexts to test if the lack of generality of the delayed HPA axis response was due to suppressive effects of S(+) on the first postshift day. Rats were given once daily 32% sucrose in S(+) and equal exposure time in S(-). Half of these rats were shifted to 4% sucrose in S(+) and half were shifted in S(-). These two groups were compared to home cage controls. Half of each group was killed after their first 4% sucrose, and half after the second 4% sucrose. All rats showed ACTH and B responses comparable to shifted rats in Experiment 1. S(+) failed to suppress the HPA axis, and the stress response was higher on the first compared to the second day of the shift. Experiment 2b established that distinctive contexts predicting sucrose, S(+), or not predicting sucrose, S(-), controlled behavioral choice and contextual discrimination. Thus, there was no evidence that issues of stimulus control could explain the lack of generality of previous findings. The data indicate that thwarting sucrose expectancies is stressful, and that this stress response habituates across days.


Asunto(s)
Hormona Adrenocorticotrópica/sangre , Condicionamiento Operante/fisiología , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Estrés Psicológico/sangre , Adaptación Fisiológica , Adaptación Psicológica , Corteza Suprarrenal/fisiología , Análisis de Varianza , Animales , Aprendizaje por Asociación/efectos de los fármacos , Aprendizaje por Asociación/fisiología , Condicionamiento Operante/efectos de los fármacos , Discriminación en Psicología/efectos de los fármacos , Discriminación en Psicología/fisiología , Relación Dosis-Respuesta a Droga , Ambiente , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Masculino , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Aprendizaje por Probabilidad , Ratas , Ratas Sprague-Dawley , Sacarosa/administración & dosificación , Edulcorantes/administración & dosificación , Factores de Tiempo , Incertidumbre
11.
Endocrinology ; 150(5): 2325-33, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19106219

RESUMEN

Previous studies have shown reduced hypothalamo-pituitary-adrenal responses to both acute and chronic restraint stressors in rats allowed to ingest highly palatable foods (32% sucrose +/- lard) prior to restraint. In this study we tested the effects of prior access (7 d) to chow-only, sucrose/chow, lard/chow, or sucrose/lard/chow diets on central corticotropin-releasing factor (CRF) expression in rats studied in two experiments, 15 and 240 min after onset of restraint. Fat depot, particularly intraabdominal fat, weights were increased by prior access to palatable food, and circulating leptin concentrations were elevated in all groups. Metabolite concentrations were appropriate for values obtained after stressors. For unknown reasons, the 15-min experiment did not replicate previous results. In the 240-min experiment, ACTH and corticosterone responses were inhibited, as previously, and CRF mRNA in the hypothalamus and oval nucleus of the bed nuclei of the stria terminalis were reduced by palatable foods, suggesting strongly that both neuroendocrine and autonomic outflows are decreased by increased caloric deposition and palatable food. In the central nucleus of the amygdala, CRF was increased in the sucrose-drinking group and decreased in the sucrose/lard group, suggesting that the consequence of ingestion of sucrose uses different neural networks from the ingestion of lard. The results suggest strongly that ingestion of highly palatable foods reduces activity in the central stress response network, perhaps reducing the feeling of stressors.


Asunto(s)
Hormona Adrenocorticotrópica/sangre , Corticosterona/sangre , Hormona Liberadora de Corticotropina/sangre , Metabolismo Energético/fisiología , Alimentos , Estrés Fisiológico/fisiología , Gusto/fisiología , Tejido Adiposo/crecimiento & desarrollo , Animales , Peso Corporal/fisiología , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Ingestión de Energía/fisiología , Hipotálamo/metabolismo , Insulina/sangre , Leptina/sangre , Masculino , Hueso Paladar/fisiología , Ratas , Ratas Sprague-Dawley , Restricción Física/fisiología , Restricción Física/psicología
12.
Am J Physiol Endocrinol Metab ; 294(1): E190-200, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17971508

RESUMEN

The common hepatic branch of the vagus nerve negatively regulates lard intake in rats with streptozotocin (STZ)-induced, insulin-dependent diabetes. However, this branch consists of two subbranches: the hepatic branch proper, which serves the liver, and the gastroduodenal branch, which serves the distal stomach, pancreas, and duodenum. The aim of this study was to determine whether the gastroduodenal branch specifically regulates voluntary lard intake. We performed a gastroduodenal branch vagotomy (GV) on nondiabetic, STZ-diabetic, and STZ-diabetic insulin-treated groups of rats and compared them with sham-operated counterparts. All rats had high steady-state corticosterone levels to maximize lard intake. Five days after surgery, all rats were provided with the choice of chow or lard to eat for another 5 days. STZ-diabetes resulted in a reduction in lard intake that was partially rescued by either GV or insulin treatment. Patterns of white adipose tissue (WAT) deposition differed after GV- and insulin-induced lard intake, with subcutaneous WAT increasing exclusively after the former and mesenteric WAT increasing exclusively in the latter. GV also prevented the insulin-induced reduction in the STZ-elevated plasma glucagon, triglycerides, free fatty acids, and total ketone bodies but did not alter the effect of insulin-induced reduction of plasma glucose levels. These data suggest that the gastroduodenal branch of the vagus inhibits lard intake and regulates WAT deposition and plasma metabolite levels in STZ-diabetic rats.


Asunto(s)
Distribución de la Grasa Corporal , Diabetes Mellitus Experimental/metabolismo , Hígado/inervación , Hígado/metabolismo , Nervio Vago/fisiología , Animales , Glucemia/metabolismo , Corticosterona/sangre , Diabetes Mellitus Experimental/tratamiento farmacológico , Grasas de la Dieta/farmacología , Ingestión de Alimentos/fisiología , Ácidos Grasos/sangre , Polipéptido Inhibidor Gástrico/sangre , Glucagón/sangre , Glucógeno/metabolismo , Hipoglucemiantes/sangre , Hipoglucemiantes/farmacología , Insulina/sangre , Insulina/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Vagotomía , Nervio Vago/anatomía & histología , Pérdida de Peso/fisiología
13.
Behav Neurosci ; 121(4): 764-78, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17663601

RESUMEN

Previous findings (N. Pecoraro, J. Chou-Green, & M. F. Dallman, 2003; N. Pecoraro & M. F. Dallman, 2005) indicated that unexpected reductions in sucrose concentration in once daily meals result in a febrile response on the 1st, but not the 2nd day of a concentration shift. This study shows that this day-specific fever is blocked by adrenalectomy accompanied by constant low corticosterone replacement. Rats implanted with telemetry probes were adrenalectomized and given low-corticosterone pellets or were sham operated. Food-restricted rats were given 2 rounds of sucrose concentration downshifts, as follows: 32% sucrose (14 days), 4% sucrose (6 days), 32% sucrose (4 days), and 4% sucrose (4 days). Intact rats showed more pronounced anticipation of the sucrose than did rats having low, clamped corticosterone. Only intact rats showed a 4-hr, postshift temperature burst on the 1st, but not the 2nd day of the shift to 4% sucrose, during both rounds of shifting. Increased activity accompanied the fever. These data confirm previous findings, show them to be dependent on high corticosterone, and appear to be related to a host of day-specific alterations in other motor outflows following unexpected downward shifts in palatable sucrose concentrations.


Asunto(s)
Temperatura Corporal/efectos de los fármacos , Corticosterona/farmacología , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Recompensa , Sacarosa/administración & dosificación , Adrenalectomía/métodos , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Temperatura Corporal/fisiología , Peso Corporal/efectos de los fármacos , Corticosterona/sangre , Relación Dosis-Respuesta a Droga , Conducta de Ingestión de Líquido/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Masculino , Ratas , Ratas Sprague-Dawley
16.
J Physiol ; 583(Pt 2): 431-6, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17556388

RESUMEN

Glucocorticoids act primarily in a feed-forward fashion on brain to activate CNS pathways that implement wanting appropriate to physiological needs. Thus, depending on the available conditions, elevated glucocorticoids may augment the behavioural want to run, fight or feed. Although glucocorticoids stimulate intake of chow, fat and sucrose, insulin appears to sculpt calorie-associated desires toward foods high in fat, acting through hepatic branch afferents of the vagus nerve. Both conditions of reduced food allowance and chronic stress excite glucocorticoid-augmented central neural networks that may lead toward ultimate abdominal obesity.


Asunto(s)
Encéfalo/metabolismo , Ingestión de Energía , Conducta Alimentaria , Glucocorticoides/metabolismo , Insulina/metabolismo , Obesidad/metabolismo , Adrenalectomía , Animales , Encéfalo/fisiopatología , Grasas de la Dieta/metabolismo , Sacarosa en la Dieta/metabolismo , Retroalimentación Fisiológica , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/fisiopatología , Hígado/inervación , Hígado/metabolismo , Obesidad/fisiopatología , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipófiso-Suprarrenal/fisiopatología , Nervio Vago/fisiopatología
17.
J Physiol ; 583(Pt 2): 455-67, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17584842

RESUMEN

The common hepatic branch of the vagus nerve is a two-way highway of communication between the brain and the liver, duodenum, stomach and pancreas that regulates many aspects of food intake and metabolism. In this study, we utilized the afferent-specific neurotoxin capsaicin to examine if common hepatic vagal sensory afferents regulate lard intake. Rats implanted with a corticosterone pellet were made diabetic using streptozotocin (STZ) and a subset received steady-state exogenous insulin replacement into the superior mesenteric vein. These were compared with non-diabetic counterparts. Each group was then subdivided into those whose common hepatic branch of the vagus was treated with vehicle or capsaicin. Five days after surgery, the rats were offered the choice of chow and lard to consume for a further 5 days. The STZ-diabetic rats ate significantly less lard than the non-diabetic rats. Capsaicin treatment restored lard intake to that of the insulin-replaced, STZ-diabetic rats, but modified neither chow nor total caloric intake. This increased lard intake led to selective fat deposition into the mesenteric white adipose tissue depot, as opposed to an increase in all visceral fat pad depots evident after insulin replacement-induced lard intake. Capsaicin treatment also increased the levels of circulating glucose and triglycerides and negated the actions of insulin on these and free fatty acids and ketone bodies. Collectively, these data suggest that afferent signalling through the common hepatic branch of the vagus inhibits lard, but not chow, intake, directs fat deposition and regulates plasma metabolite levels.


Asunto(s)
Biomarcadores/sangre , Diabetes Mellitus Experimental/fisiopatología , Grasas de la Dieta/metabolismo , Conducta Alimentaria , Hígado/inervación , Nervio Vago/fisiopatología , Tejido Adiposo Blanco/patología , Glándulas Suprarrenales/patología , Vías Aferentes/fisiopatología , Animales , Glucemia/metabolismo , Peso Corporal , Capsaicina/farmacología , Corticosterona/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Grasas de la Dieta/administración & dosificación , Conducta Alimentaria/efectos de los fármacos , Preferencias Alimentarias , Glucagón/sangre , Glucógeno/metabolismo , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Insulina/sangre , Insulina/farmacología , Insulina/uso terapéutico , Leptina/sangre , Lípidos/sangre , Hígado/metabolismo , Masculino , Tamaño de los Órganos , Ratas , Ratas Sprague-Dawley , Bazo/patología , Timo/patología , Factores de Tiempo , Nervio Vago/efectos de los fármacos
18.
Endocrinology ; 148(7): 3288-98, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17412812

RESUMEN

Although high insulin concentrations reduce food intake, low insulin concentrations promote lard intake over chow, possibly via an insulin-derived, liver-mediated signal. To investigate the role of the hepatic vagus in voluntary lard intake, streptozotocin-diabetic rats with insulin or vehicle replaced into either the superior mesenteric or jugular veins received a hepatic branch vagotomy (HV) or a sham operation. All rats received a pellet of corticosterone that clamped the circulating steroid at moderately high concentrations to enhance lard intake. After 5 d of recovery, rats were offered the choice of lard and chow for 5 d. In streptozotocin-diabetic rats, HV, like insulin replacement, restored lard intake to nondiabetic levels. Consequently, this reduced chow intake without affecting total caloric intake, and insulin site-specifically increased white adipose tissue weight. HV also ablated the effects of insulin on reducing circulating glucose levels and attenuated the streptozotocin-induced weight loss in most groups. Collectively, these data suggest that the hepatic vagus normally inhibits lard intake and can influence glucose homeostasis and the pattern of white adipose tissue deposition. These actions may be modulated by insulin acting both centrally and peripherally.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Diabetes Mellitus Experimental/terapia , Grasas de la Dieta/metabolismo , Insulina/farmacología , Vagotomía/métodos , Tejido Adiposo Blanco/efectos de los fármacos , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Corticosterona/administración & dosificación , Corticosterona/farmacología , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/fisiopatología , Grasas de la Dieta/administración & dosificación , Ingestión de Energía/efectos de los fármacos , Ácidos Grasos no Esterificados/sangre , Glucógeno/metabolismo , Hipoglucemiantes/farmacología , Insulina/sangre , Cuerpos Cetónicos/sangre , Masculino , Ratas , Ratas Sprague-Dawley , Estreptozocina , Factores de Tiempo , Triglicéridos/sangre , Nervio Vago/fisiopatología
19.
Curr Alzheimer Res ; 4(2): 199-204, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17430247

RESUMEN

In mammals, glucocorticoid actions appear to have evolved to maintain and enhance energy stores to be used for life-saving gluconeogenesis. They act on the brain to stimulate search behaviors, palatable feeding and emotionally relevant memories, and they act on the body to mobilize stored peripheral energy and direct it to central depots that serve the substrate needs of the liver. Our work in rats shows that searching and intake of palatable foods (sucrose, saccharin and lard) are stimulated by corticosterone in a dose-related fashion. Adrenalectomized rats gain weight poorly, have low fat content, increased sympathetic neural and hypothalamo-pituitary-adrenal outflow, and altered behaviors. Replacement with corticosterone reverses these effects. Surprisingly, when such rats are provided with 30% sucrose to drink, in addition to saline, all of the usual effects of adrenalectomy are corrected without corticosterone. We hypothesize that there is a metabolic feedback system that decreases stress-responsiveness. Although we have not yet identified the signal associated with sucrose drinking, the weight of mesenteric fat correlates inversely with hypothalamic corticotropin-releasing factor (CRF). When rats eat lard and sucrose ad libitum, fat stores increase and CRF, ACTH and corticosterone responses are reduced. During stress, chow intake decreases but intake of lard and sucrose does not. Our current working model suggests that palatability signals and neural signals from fat stores act on brain to reduce activity in the central stress response system. Correlative results from a clinical study support the powerful role of small changes in glucocorticoids in type 2 diabetes.


Asunto(s)
Glucocorticoides/metabolismo , Enfermedades Metabólicas/etiología , Obesidad/etiología , Animales , Humanos , Insulina/metabolismo , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Modelos Biológicos , Obesidad/metabolismo , Obesidad/patología , Factores de Riesgo
20.
Prog Neurobiol ; 79(5-6): 247-340, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16982128

RESUMEN

The hypothalamo-pituitary-adrenal (HPA) axis is the critical mediator of the vertebrate stress response system, responding to environmental stressors by maintaining internal homeostasis and coupling the needs of the body to the wants of the mind. The HPA axis has numerous complex drivers and highly flexible operating characterisitics. Major drivers include two circadian drivers, two extra-hypothalamic networks controlling top-down (psychogenic) and bottom-up (systemic) threats, and two intra-hypothalamic networks coordinating behavioral, autonomic, and neuroendocrine outflows. These various networks jointly and flexibly control HPA axis output of periodic (oscillatory) functions and a range of adventitious systemic or psychological threats, including predictable daily cycles of energy flow, actual metabolic deficits over many time scales, predicted metabolic deficits, and the state-dependent management of post-prandial responses to feeding. Evidence is provided that reparation of metabolic derangement by either food or glucocorticoids results in a metabolic signal that inhibits HPA activity. In short, the HPA axis is intimately involved in managing and remodeling peripheral energy fluxes, which appear to provide an unidentified metabolic inhibitory feedback signal to the HPA axis via glucocorticoids. In a complementary and perhaps a less appreciated role, adrenocortical hormones also act on brain to provide not only feedback, but feedforward control over the HPA axis itself and its various drivers, as well as coordinating behavioral and autonomic outflows, and mounting central incentive and memorial networks that are adaptive in both appetitive and aversive motivational modes. By centrally remodeling the phenotype, the HPA axis provides ballistic and predictive control over motor outflows relevant to the type of stressor. Evidence is examined concerning the global hypothesis that the HPA axis comprehensively induces integrative phenotypic plasticity, thus remodeling the body and its governor, the brain, to yoke the needs of the body to the wants of the mind. Adverse side effects of this yoking under conditions of glucocorticoid excess are discussed.


Asunto(s)
Sistema Hipotálamo-Hipofisario/fisiología , Motivación , Fenotipo , Sistema Hipófiso-Suprarrenal/fisiología , Animales , Glucocorticoides/fisiología , Humanos , Sistema Hipotálamo-Hipofisario/anatomía & histología , Modelos Biológicos , Red Nerviosa/fisiología , Vías Nerviosas/fisiología , Sistema Hipófiso-Suprarrenal/anatomía & histología , Estrés Fisiológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA