Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Physiol ; 13: 841935, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35557971

RESUMO

Obesogenic diets can produce hippocampal insulin resistance and impairments to hippocampal-dependent cognition. This study investigated the effect of disrupted insulin signaling in Neuropeptide Y (NPY) neurons on diet-induced deficits in hippocampal-dependent memory. Wild-type mice and mice that had a targeted knockout of insulin receptors on NPY cells (IRlox/lox;NPYCre/+) were given ad libitum access to a high-fat diet (high fat; HF), 10% sucrose solution (high sugar; HS), both high-fat diet and sucrose solution (high fat, high sugar; HFHS), or a normal fat control chow for 12 weeks. Mice were tested in the Morris Water Maze (MWM), a hippocampal-dependent spatial memory task. Glucose homeostasis was assessed via a glucose tolerance test. Independent of genotype, consumption of HF, but not HS, diet increased energy intake, body weight, and plasma leptin, and impaired glucose tolerance. Disrupted insulin signaling in NPY cells and dietary interventions did not significantly affect the ability of mice to learn the location of the platform in the MWM. However, for IRlox/lox control mice, consumption of HF, but not HS, diet resulted in reduced time spent in the target quadrant during the probe trial, suggesting a hippocampal-dependent memory deficit. IRlox/lox;NPYCre/+ mice had poor performance in the probe trial regardless of diet, suggesting a floor effect. This study did not find adverse effects of chronic sucrose intake on metabolic outcomes or hippocampal-dependent memory. These data also suggest that the effects of HF diet on hippocampal-dependent memory may be dependent on insulin signaling in hippocampal NPY cells.

2.
J Integr Neurosci ; 21(1): 6, 2022 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-35164442

RESUMO

Insulin is known to act in the central nervous system to regulate several physiological and behavioural outcomes, including energy balance, glucose homeostasis and cognitive functioning. However, the neuronal populations through which insulin enhances cognitive performance remain unidentified. Insulin receptors are found in neuropeptide-Y (NPY) expressing neurons, which are abundant in the hypothalamus and hippocampus; regions involved in feeding behaviour and spatial memory, respectively. Here we show that mice with a tissue specific knockout of insulin receptors in NPY expressing neurons (IRl⁢o⁢x/l⁢o⁢x; NPYC⁢r⁢e⁣/+) display an impaired performance in the probe trial of the Morris Water Maze compared with control mice at both the 6 and the 12, but not at the 24 months time point, consistent with a crucial role of insulin and NPY in cognitive functioning. By 24 months of age all groups demonstrated similar reductions in spatial memory performance. Together, these data suggest that the mechanisms through which insulin influences cognitive functioning are, at least in part, via insulin receptor signaling in NPY expressing neurons. These results also highlight that cognitive impairments observed in aging may be due to impaired insulin signaling.


Assuntos
Envelhecimento/fisiologia , Disfunção Cognitiva , Hipocampo , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Receptor de Insulina/fisiologia , Envelhecimento/metabolismo , Animais , Comportamento Animal/fisiologia , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/fisiopatologia , Modelos Animais de Doenças , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Insulina/deficiência , Memória Espacial/fisiologia
4.
Front Endocrinol (Lausanne) ; 12: 682726, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34149621

RESUMO

Obesity is a growing health problem worldwide. The renin-angiotensin system (RAS) is present in adipose tissue, and evidence suggests that it is involved in both diet-induced obesity and the inflammation associated with obesity. The present experiments determined the effect of (1) different angiotensin-converting enzyme (ACE) inhibitors (captopril, perindopril, enalapril) and angiotensin receptor blockers (ARBs: telmisartan, losartan) on adiposity of mice fed a high-fat diet for 28 days (2); acute treatment with the ACE-inhibitor captopril on gene expression of inflammatory markers in mice fed a high-fat diet (HFD); and (3) short-term (2 days) and chronic (28 days) treatment of ACE-inhibition on energy expenditure (EE) and energy balance in mice fed HFD ad libitum (AL), as well as receiving HFD limited to the amount of calories eaten by controls (pair-fed (PF) group). Body weight, food intake, adiposity and plasma leptin were lower in ACE inhibitor or ARB-treated groups over 28 days compared with HFD untreated mice. Short-term treatment with captopril led to increased EE relative to the level in the PF group. After 28 days, EE was lower in both captopril-treated and PF mice compared with AL, but the effect was greater in the captopril-treated group. Adiponectin was elevated in captopril-treated mice, but not in PF mice, after both 2 and 28 days. Additionally, acute RAS blockade in HFD-fed mice reduced mRNA expression for MCP-1, IL-6, TLR4, and leptin in adipose tissue relative to values in untreated groups. These data demonstrate that ACE inhibition and angiotensin receptor blockade reduce food intake to produce weight loss and suggest that the anti-inflammatory effects of ACE inhibition may be independent of weight loss.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Antagonistas de Receptores de Angiotensina/farmacologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Mediadores da Inflamação/metabolismo , Adiponectina/sangue , Tecido Adiposo/metabolismo , Animais , Composição Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos , Metabolismo Energético/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Leptina/sangue , Masculino , Camundongos Endogâmicos C57BL , Obesidade/sangue , Obesidade/tratamento farmacológico , Obesidade/genética , Obesidade/metabolismo , Sistema Renina-Angiotensina
5.
J Neuroendocrinol ; 33(4): e12952, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33656205

RESUMO

Food intake and energy expenditure are regulated by peripheral signals providing feedback on nutrient status and adiposity to the central nervous system. One of these signals is the pancreatic hormone, insulin. Unlike peripheral administration of insulin, which often causes weight gain, central administration of insulin leads to a reduction in food intake and body weight when administered long-term. This is a result of feedback processes in regions of the brain that regulate food intake. Within the hypothalamus, the arcuate nucleus (ARC) contains subpopulations of neurones that produce orexinergic neuropeptides agouti-related peptide (AgRP)/neuropeptide Y (NPY) and anorexigenic neuropeptides, pro-opiomelanocortin (POMC)/cocaine- and amphetamine-regulated transcript (CART). Intracerebroventricular infusion of insulin down-regulates the expression of AgRP/NPY at the same time as up-regulating expression of POMC/CART. Recent evidence suggests that insulin activity within the amygdala may play an important role in regulating energy balance. Insulin infusion into the central nucleus of the amygdala (CeA) can decrease food intake, possibly by modulating activity of NPY and other neurone subpopulations. Insulin signalling within the CeA can also influence stress-induced obesity. Overall, it is evident that the CeA is a critical target for insulin signalling and the regulation of energy balance.


Assuntos
Encéfalo/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Retroalimentação Fisiológica/efeitos dos fármacos , Insulina/farmacologia , Proteína Relacionada com Agouti/metabolismo , Animais , Encéfalo/metabolismo , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Retroalimentação Fisiológica/fisiologia , Humanos , Insulina/metabolismo , Neuropeptídeo Y/metabolismo , Pró-Opiomelanocortina/metabolismo
6.
Endocrinology ; 162(5)2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33522579

RESUMO

Polycystic ovary syndrome (PCOS) is a prevalent endocrine condition characterized by a range of endocrine, reproductive, and metabolic abnormalities. At present, management of women with PCOS is suboptimal as treatment is only symptomatic. Clinical and experimental advances in our understanding of PCOS etiology support a pivotal role for androgen neuroendocrine actions in PCOS pathogenesis. Hyperandrogenism is a key PCOS trait and androgen actions play a role in regulating the kisspeptin-/neurokinin B-/dynorphin (KNDy) system. This study aimed to investigate if targeted antagonism of neurokinin B signaling through the neurokinin 3 receptor (NK3R) would reverse PCOS traits in a dihydrotestosterone (DHT)-induced mouse model of PCOS. After 3 months, DHT exposure induced key reproductive PCOS traits of cycle irregularity and ovulatory dysfunction, and PCOS-like metabolic traits including increased body weight; white and brown fat pad weights; fasting serum triglyceride and glucose levels, and blood glucose incremental area under the curve. Treatment with a NK3R antagonist (MLE4901) did not impact the observed reproductive defects. In contrast, following NK3R antagonist treatment, PCOS-like females displayed decreased total body weight, adiposity, and adipocyte hypertrophy, but increased respiratory exchange ratio, suggesting NK3R antagonism altered the metabolic status of the PCOS-like females. NK3R antagonism did not improve circulating serum triglyceride or fasted glucose levels. Collectively, these findings demonstrate that NK3R antagonism may be beneficial in the treatment of adverse metabolic features associated with PCOS and support neuroendocrine targeting in the development of novel therapeutic strategies for PCOS.


Assuntos
Lectinas/administração & dosagem , Proteínas de Membrana/administração & dosagem , Síndrome do Ovário Policístico/tratamento farmacológico , Receptores da Neurocinina-3/antagonistas & inibidores , Androgênios/sangue , Animais , Glicemia/metabolismo , Di-Hidrotestosterona/efeitos adversos , Modelos Animais de Doenças , Feminino , Humanos , Hiperandrogenismo/genética , Hiperandrogenismo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Receptores da Neurocinina-3/genética , Receptores da Neurocinina-3/metabolismo , Triglicerídeos/sangue
7.
Front Aging Neurosci ; 12: 19, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32184716

RESUMO

Aging results in decreased fluid intake following dehydration and other dipsogenic stimuli; similar reductions in sodium intake have also been observed with aging. Given that cyclooxygenase (COX)-derived prostanoids are elevated in aged rats in the midbrain and proinflammatory prostanoids are known to decrease fluid intake in dehydrated rats, the aim of this study was to determine if the reductions of fluid intake and sodium intake in aging are mediated by proinflammatory eicosanoid signaling. Therefore, we examined the effect of acute COX inhibition in adult (4 months-old) and aged (30 months-old) rats prior to ingestive behavior challenges. COX inhibition, using acetylsalicylic acid (ASA), increased fluid intake in aged, but not adult, rats in response to 24-h dehydration. ASA had no effect on salt intake following sodium depletion and ASA did not change basal fluid or sodium consumption in either age group. Hypothalamic COX-1 and -2, prostaglandin E synthase (PGES) and inducible nitric oxide synthase (iNOS) mRNA expression were all elevated in aged animals, leading to elevated PGE2 levels. COX expression in the hypothalamus was reduced by ASA treatment in rats of both ages resulting in reduced PGE2 levels in aged ASA treated animals. These data indicate that the reduced fluid intake that occurs in aging is due to increased COX-PGE2-mediated inflammation. However, the reduced sodium intake in these animals appears to occur via an alternate mechanism.

8.
Psychoneuroendocrinology ; 104: 7-17, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30784904

RESUMO

Research with predominantly male samples supports primary and secondary developmental pathways to psychopathy that are phenotypically indistinguishable on aggressive and antisocial behavior. The aim of this study was to examine whether female variants of psychopathy show divergent endocrine (i.e., cortisol, dehydroepiandrosterone [DHEA], testosterone, and their ratios) and psychophysiological (i.e., heart rate variability [HRV]) reactivity to social provocation. We also tested whether variants differed on reactive aggression when performing a competitive reaction time task against the fictitious participant who previously insulted them. Latent profile analyses on 101 undergraduate women oversampled for high psychopathic traits identified a high-anxious, maltreated secondary variant (n=64) and a low-anxious primary variant (n=37). Although variants did not differ on aggression, secondary variants showed higher cortisol, testosterone, cortisol-to-DHEA ratios, and HRV following social provocation relative to primary variants. Findings suggest that the neurobiological mechanisms underpinning aggression in psychopathy may differ between women on primary versus secondary developmental pathways.


Assuntos
Agressão/fisiologia , Transtorno da Personalidade Antissocial/metabolismo , Transtornos Psicofisiológicos/metabolismo , Adulto , Agressão/psicologia , Ira/fisiologia , Transtorno da Personalidade Antissocial/patologia , Ansiedade , Transtorno da Conduta/fisiopatologia , Desidroepiandrosterona/análise , Sistema Endócrino/metabolismo , Sistema Endócrino/fisiologia , Feminino , Frequência Cardíaca/fisiologia , Humanos , Hidrocortisona/análise , Comportamento Impulsivo/fisiologia , Transtornos Psicofisiológicos/patologia , Autorrelato , Testosterona/análise , Adulto Jovem
9.
Physiol Behav ; 193(Pt B): 218-222, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29577941

RESUMO

Despite the ability of some gastrointestinal hormones to reliably reduce meal size when administered prior to a meal, it is not understood why the repeated administration or genetic knockout of these hormones appear largely ineffective in reducing food intake and body weight. Here, we review evidence that the ability of GI peptides such as cholecystokinin (CCK) to elicit satiation is a consequence of prior learning. Evidence includes first, that the ability of some of these signals to modify food intake depends upon past experience and is malleable with new experience. Additionally, the ability of CCK and other gut signals to reduce food intake may not be hard-wired; i.e., any so-called "satiation" signal that reduces food intake in a single-meal situation may not continue to do so over repeated trials. The individual will respond to the signal only so long as it provides reliable information about caloric content. If a particular signal becomes unreliable, the individual will rely on other signals to end meals. Thus, gut peptides/hormones have important metabolic effects such as mediating absorption, digestion, and many aspects of the distribution of ingested nutrients throughout the body; and, if they have been reliably associated with natural stimuli that mediate satiation, they also inform behavior.


Assuntos
Ingestão de Alimentos/fisiologia , Hormônios Gastrointestinais/metabolismo , Peptídeos/metabolismo , Saciação/fisiologia , Animais , Humanos
10.
Artigo em Inglês | MEDLINE | ID: mdl-30619085

RESUMO

Obesity and high fat diet consumption contribute to the development of metabolic disorders, insulin resistance, neuroinflammation, and cognitive impairments. CNS administration of insulin into the brain can attenuate these cognitive impairments. The present study investigated whether hippocampal-dependent spatial memory impairments in a dietary induced mouse model of obesity could be improved by the direct administration of insulin into the hippocampus and whether this was associated with markers of hippocampal inflammation. C57Bl/6J mice consumed a low fat or high fat diet for 16 weeks and continuous intrahippocampal saline or insulin infusion for the final 4 weeks, during a period of behavioral testing, before gene expression analysis was performed. The high fat diet group demonstrated poorer spatial memory performance in the Morris water maze and Y-maze, supporting the hypothesis that high fat diet leads to hippocampal dependent cognitive impairment. Insulin infusion into the hippocampus reversed the deficit of high fat diet consumption on both of the tasks. Increased expression of inflammatory markers was detected in the hippocampus in the high fat diet group and expression of these markers was ameliorated in insulin infused mice. This demonstrates that CNS insulin can improve hippocampal-dependent memory and that hippocampal inflammation may be a factor in the development of cognitive deficits associated with diet-induced obesity. Furthermore, these data suggest that insulin may act to attenuate high fat diet induced cognitive deficits by reducing neuroinflammation.

11.
Physiol Behav ; 178: 28-34, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28267585

RESUMO

During heat waves, significant mortality and morbidity occurs in elderly populations due to heat-stress and dehydration. The dehydration is primarily attributable to inadequate water intake caused by dysfunction of the central nervous system mechanisms controlling thirst. The phenomenon of a reduced thirst in response to dehydration in aging was first observed decades ago and has been examined extensively since. The reduced thirst and ingestive behavior have been reported consistently in response to hyperosmotic stimuli, hypovolemic stimuli and dehydration in both elderly humans and animal models of aging. There are also data to suggest that sodium appetite is reduced in aged rats, potentially indicating a common etiology. Accompanying the behavioral changes in water and sodium intake that occur with aging there are also alterations to a number of hormonal systems involved in body fluid and electrolyte homeostasis. These changes include reductions in activity of the renin-angiotensin system and increases in circulating atrial natriuretic peptide and arginine vasopressin. While there is substantial evidence to suggest that the behavioral and physiological mechanisms responsible for body fluid and sodium homeostasis are impaired in aging, the precise etiology of reduced thirst remain to be determined.


Assuntos
Envelhecimento/fisiologia , Sede/fisiologia , Equilíbrio Hidroeletrolítico/fisiologia , Animais , Desidratação/fisiopatologia , Humanos , Sódio na Dieta/metabolismo
12.
Physiol Behav ; 178: 172-178, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-27923718

RESUMO

The cerebrospinal fluid (CSF) offers a window into the workings of the brain and blood-brain barrier (BBB). Molecules that enter into the central nervous system (CNS) by passive diffusion or receptor-mediated transport through the choroid plexus often appear in the CSF prior to acting within the brain. Other molecules enter the CNS by passing through the BBB into the brain's interstitial fluid prior to appearing in the CSF. This pattern is also often observed for molecules synthesized by neurons or glia within the CNS. The CSF is therefore an important conduit for the entry and clearance of molecules into/from the CNS and thereby constitutes an important window onto brain activity and barrier function. Assessing the CSF basally, under experimental conditions, or in the context of challenges or metabolic diseases can provide powerful insights about brain function. Here, we review important findings made by our labs, as influenced by the late Randall Sakai, by interrogating the CSF.


Assuntos
Líquido Cefalorraquidiano/metabolismo , Comportamento Alimentar/fisiologia , Animais , Encéfalo/metabolismo , Humanos , Insulina/metabolismo
13.
Physiol Behav ; 165: 392-7, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27570192

RESUMO

Food intake occurs in bouts or meals, and numerous meal-generated signals have been identified that act to limit the size of ongoing meals. Hormones such as cholecystokinin (CCK) are secreted from the intestine as ingested food is being processed, and in addition to aiding the digestive process, they provide a signal to the brain that contributes to satiation, limiting the size of the meal. The potency of CCK to elicit satiation is enhanced by elevated levels of adiposity signals such as insulin. In the present experiments we asked whether CCK and insulin interact at the level of the blood-brain barrier (BBB). We first isolated rat brain capillary endothelial cells that comprise the BBB and found that they express the mRNA for both the CCK1R and the insulin receptor, providing a basis for a possible interaction. We then administered insulin intraperitoneally to another group of rats and 15min later administered CCK-8 intraperitoneally to half of those rats. After another 15min, CSF and blood samples were obtained and assayed for immunoreactive insulin. Plasma insulin was comparably elevated above baseline in both the CCK-8 and control groups, indicating that the CCK had no effect on circulating insulin levels given these parameters. In contrast, rats administered CCK had CSF-insulin levels that were more than twice as high as those of control rats. We conclude that circulating CCK greatly facilitates the transport of insulin into the brain, likely by acting directly at the BBB. These findings imply that in circumstances in which the plasma levels of both CCK and insulin are elevated, such as during and soon after meals, satiation is likely to be due, in part, to this newly-discovered synergy between CCK and insulin.


Assuntos
Encéfalo/anatomia & histologia , Insulina/metabolismo , Microvasos/efeitos dos fármacos , Receptor de Colecistocinina A/metabolismo , Sincalida/farmacologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas In Vitro , Masculino , Transporte Proteico/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Long-Evans , Receptor de Colecistocinina A/genética
14.
Curr Top Behav Neurosci ; 27: 15-34, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26323244

RESUMO

Although food intake is necessary to provide energy for all bodily activities, considering food intake as a motivated behavior is complex. Rather than being a simple unconditioned reflex to energy need, eating is mediated by diverse factors. These include homeostatic signals such as those related to body fat stores, to food available and being eaten, and to circulating energy-rich compounds like glucose and fatty acids. Eating is also greatly influenced by non-homeostatic signals that convey information related to learning and experience, hedonics, stress, the social situation, opportunity, and many other factors. Recent developments identifying the intricate nature of the relationships between homeostatic and non-homeostatic influences significantly add to the complexity underlying the neural basis of the motivation to eat. The future of research in the field of food intake would seem to lie in the identification of the neural circuitry and interactions between homeostatic and non-homeostatic influences.


Assuntos
Encéfalo/fisiopatologia , Comportamento Alimentar/fisiologia , Transtornos da Alimentação e da Ingestão de Alimentos/fisiopatologia , Motivação/fisiologia , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/fisiologia , Encéfalo/metabolismo , Encéfalo/fisiologia , Colecistocinina/metabolismo , Transtornos da Alimentação e da Ingestão de Alimentos/metabolismo , Homeostase , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Insulina/metabolismo , Leptina/metabolismo , Saciação/fisiologia , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia
15.
Mol Metab ; 4(10): 692-705, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26500841

RESUMO

OBJECTIVE: Melanocortin-4 receptors (MC4Rs) are highly expressed by dopamine-secreting neurons of the mesolimbic tract, but their functional role has not been fully resolved. Voluntary wheel running (VWR) induces adaptations in the mesolimbic dopamine system and has a myriad of long-term beneficial effects on health. In the present experiments we asked whether MC4R function regulates the effects of VWR, and whether VWR ameliorates MC4R-associated symptoms of the metabolic syndrome. METHODS: Electrically evoked dopamine release was measured in slice preparations from sedentary wild-type and MC4R-deficient Mc4r (K314X) (HOM) rats. VWR was assessed in wild-type and HOM rats, and in MC4R-deficient loxTB (Mc4r) mice, wild-type mice body weight-matched to loxTB (Mc4r) mice, and wild-type mice with intracerebroventricular administration of the MC4R antagonist SHU9119. Mesolimbic dopamine system function (gene/protein expression) and metabolic parameters were examined in wheel-running and sedentary wild-type and HOM rats. RESULTS: Sedentary obese HOM rats had increased electrically evoked dopamine release in several ventral tegmental area (VTA) projection sites compared to wild-type controls. MC4R loss-of-function decreased VWR, and this was partially independent of body weight. HOM wheel-runners had attenuated markers of intracellular D1-type dopamine receptor signaling despite increased dopamine flux in the VTA. VWR increased and decreased ΔFosB levels in the nucleus accumbens (NAc) of wild-type and HOM runners, respectively. VWR improved metabolic parameters in wild-type wheel-runners. Finally, moderate voluntary exercise corrected many aspects of the metabolic syndrome in HOM runners. CONCLUSIONS: Central dopamine dysregulation during VWR reinforces the link between MC4R function and molecular and behavioral responding to rewards. The data also suggest that exercise can be a successful lifestyle intervention in MC4R-haploinsufficient individuals despite reduced positive reinforcement during exercise training.

16.
Cell Metab ; 22(3): 348-51, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26331600

RESUMO

The ability of hormones such as insulin, leptin, and cholecystokinin to alter food intake is influenced by intricate interactions between homeostatic and non-homeostatic factors. Consequently, when administered exogenously, the likelihood of these hormones influencing food intake is probabilistic, leading to difficulties replicating previously reported outcomes both within and between labs.


Assuntos
Ingestão de Alimentos , Homeostase , Animais , Regulação do Apetite , Colecistocinina/metabolismo , Humanos , Insulina/metabolismo , Leptina/metabolismo , Reprodutibilidade dos Testes
17.
Vitam Horm ; 98: 229-48, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25817871

RESUMO

The pancreatic hormone insulin plays a well-described role in the periphery, based principally on its ability to lower circulating glucose levels via activation of glucose transporters. However, insulin also acts within the central nervous system (CNS) to alter a number of physiological outcomes ranging from energy balance and glucose homeostasis to cognitive performance. Insulin is transported into the CNS by a saturable receptor-mediated process that is proposed to be dependent on the insulin receptor. Transport of insulin into the brain is dependent on numerous factors including diet, glycemia, a diabetic state and notably, obesity. Obesity leads to a marked decrease in insulin transport from the periphery into the CNS and the biological basis of this reduction of transport remains unresolved. Despite decades of research into the effects of central insulin on a wide range of physiological functions and its transport from the periphery to the CNS, numerous questions remain unanswered including which receptor is responsible for transport and the precise mechanisms of action of insulin within the brain.


Assuntos
Transporte Biológico/fisiologia , Encéfalo/metabolismo , Insulina/metabolismo , Receptor de Insulina/metabolismo , Animais , Glicemia/metabolismo , Barreira Hematoencefálica , Diabetes Mellitus/metabolismo , Humanos , Insulina/líquido cefalorraquidiano , Obesidade/metabolismo , Fatores de Risco
18.
Diabetes ; 64(7): 2457-66, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25667307

RESUMO

Insulin detemir (DET) reduces glycemia comparably to other long-acting insulin formulations but causes less weight gain. Insulin signaling in the brain is catabolic, reducing food intake. We hypothesized that DET reduces weight gain, relative to other insulins, owing to increased transport into the central nervous system and/or increased catabolic action within the brain. Transport of DET and NPH insulin into the cerebrospinal fluid (CSF) was compared over several hours and after the administration of different doses peripherally in rats. DET and NPH had comparable saturable, receptor-mediated transport into the CSF. CSF insulin remained elevated significantly longer after intraperitoneal DET than after NPH. When administered acutely into the 3rd cerebral ventricle, both DET and NPH insulin reduced food intake and body weight at 24 h, and both food intake and body weight remained lower after DET than after NPH after 48 h. In direct comparison with another long-acting insulin, insulin glargine (GLAR), DET led to more prolonged increases in CSF insulin despite a shorter plasma half-life in both rats and mice. Additionally, peripheral DET administration reduced weight gain and increased CSF insulin compared with saline or GLAR in mice. Overall, these data support the hypothesis that DET has distinct effects on energy balance through enhanced and prolonged centrally mediated reduction of food intake.


Assuntos
Depressores do Apetite/farmacologia , Encéfalo/efeitos dos fármacos , Hipoglicemiantes/farmacocinética , Insulina Isófana/farmacocinética , Insulina de Ação Prolongada/farmacocinética , Animais , Transporte Biológico , Composição Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Injeções Intraperitoneais , Insulina/sangue , Insulina/líquido cefalorraquidiano , Insulina Detemir , Insulina Isófana/administração & dosagem , Insulina Isófana/farmacologia , Insulina de Ação Prolongada/administração & dosagem , Insulina de Ação Prolongada/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Wistar
19.
Am J Physiol Endocrinol Metab ; 307(11): E1065-72, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25315695

RESUMO

Vertical sleeve gastrectomy (VSG) is currently one of the most effective treatments for obesity. Despite recent developments, the underlying mechanisms that contribute to the metabolic improvements following bariatric surgery remain unresolved. VSG reduces postprandial intestinal triglyceride (TG) production, but whether the effects of VSG on intestinal metabolism are related to metabolic outcomes has yet to be established. The lipid synthesis enzyme acyl CoA:monoacylglycerol acyltransferase-2 (Mogat2; MGAT2) plays a crucial role in the assimilation of dietary fat in the intestine and in regulation of adiposity stores as well. Given the phenotypic similarities between VSG-operated and MGAT2-deficient animals, we reasoned that this enzyme could also have a key role in mediating the metabolic benefits of VSG. However, VSG reduced body weight and fat mass and improved glucose metabolism similarly in whole body MGAT2-deficient (Mogat2(-/-)) mice and wild-type littermates. Furthermore, along with an increase in energy expenditure, surgically naive Mogat2(-/-) mice had altered macronutrient preference, shifting preference away from fat and toward carbohydrates, and increased locomotor activity. Collectively, these data suggest that the beneficial effects of VSG on body weight and glucose metabolism are independent of MGAT2 activity and rather that they are separate from the effects of MGAT2 deficiency. Because MGAT2 inhibitors are proposed as a pharmacotherapeutic option for obesity, our data suggest that, in addition to increasing energy expenditure, shifting macronutrient preference away from fat could be another important mechanism by which these compounds could contribute to weight loss.


Assuntos
Gastrectomia , N-Acetilglucosaminiltransferases/deficiência , Animais , Composição Corporal , Peso Corporal , Dieta com Restrição de Gorduras , Ingestão de Alimentos , Preferências Alimentares , Masculino , Camundongos , Camundongos Knockout , Obesidade/genética
20.
Diabetes ; 63(11): 3798-804, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24898144

RESUMO

Uroguanylin is a gastrointestinal hormone primarily involved in fluid and electrolyte handling. It has recently been reported that prouroguanylin, secreted postprandially, is converted to uroguanylin in the brain and activates the receptor guanylate cyclase-C (GC-C) to reduce food intake and prevent obesity. We tested central nervous system administration of two GC-C agonists and found no significant reduction of food intake. We also carefully phenotyped mice lacking the GC-C receptor and found them to have normal body weight, adiposity, and glucose tolerance. Interestingly, uroguanylin knockout mice had a small but significant increase in body weight and adiposity that was accompanied by glucose intolerance. Our data indicate that the modest effects of uroguanylin on energy and glucose homeostasis are not mediated by central GC-C receptors.


Assuntos
Glucose/metabolismo , Guanilato Ciclase/metabolismo , Animais , GMP Cíclico/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Masculino , Camundongos , Peptídeos Natriuréticos/farmacologia , Ratos , Receptores de Enterotoxina , Receptores Acoplados a Guanilato Ciclase/metabolismo , Receptores de Peptídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...