Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Physiol Rev ; 97(1): 411-463, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28003328

RESUMO

The efficacy of Roux-en-Y gastric-bypass (RYGB) and other bariatric surgeries in the management of obesity and type 2 diabetes mellitus and novel developments in gastrointestinal (GI) endocrinology have renewed interest in the roles of GI hormones in the control of eating, meal-related glycemia, and obesity. Here we review the nutrient-sensing mechanisms that control the secretion of four of these hormones, ghrelin, cholecystokinin (CCK), glucagon-like peptide-1 (GLP-1), and peptide tyrosine tyrosine [PYY(3-36)], and their contributions to the controls of GI motor function, food intake, and meal-related increases in glycemia in healthy-weight and obese persons, as well as in RYGB patients. Their physiological roles as classical endocrine and as locally acting signals are discussed. Gastric emptying, the detection of specific digestive products by small intestinal enteroendocrine cells, and synergistic interactions among different GI loci all contribute to the secretion of ghrelin, CCK, GLP-1, and PYY(3-36). While CCK has been fully established as an endogenous endocrine control of eating in healthy-weight persons, the roles of all four hormones in eating in obese persons and following RYGB are uncertain. Similarly, only GLP-1 clearly contributes to the endocrine control of meal-related glycemia. It is likely that local signaling is involved in these hormones' actions, but methods to determine the physiological status of local signaling effects are lacking. Further research and fresh approaches are required to better understand ghrelin, CCK, GLP-1, and PYY(3-36) physiology; their roles in obesity and bariatric surgery; and their therapeutic potentials.


Assuntos
Colecistocinina/metabolismo , Derivação Gástrica , Grelina/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptídeo YY/metabolismo , Glicemia/metabolismo , Ingestão de Alimentos/fisiologia , Humanos , Obesidade/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(44): 27516-27527, 2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33077601

RESUMO

Multiple sclerosis (MS) is an autoimmune disease of the central nervous system. The etiology of MS is multifactorial, with disease risk determined by genetics and environmental factors. An emerging risk factor for immune-mediated diseases is an imbalance in the gut microbiome. However, the identity of gut microbes associated with disease risk, their mechanisms of action, and the interactions with host genetics remain obscure. To address these questions, we utilized the principal autoimmune model of MS, experimental autoimmune encephalomyelitis (EAE), together with a genetically diverse mouse model representing 29 unique host genotypes, interrogated by microbiome sequencing and targeted microbiome manipulation. We identified specific gut bacteria and their metabolic functions associated with EAE susceptibility, implicating short-chain fatty acid metabolism as a key element conserved across multiple host genotypes. In parallel, we used a reductionist approach focused on two of the most disparate phenotypes identified in our screen. Manipulation of the gut microbiome by transplantation and cohousing demonstrated that transfer of these microbiomes into genetically identical hosts was sufficient to modulate EAE susceptibility and systemic metabolite profiles. Parallel bioinformatic approaches identified Lactobacillus reuteri as a commensal species unexpectedly associated with exacerbation of EAE in a genetically susceptible host, which was functionally confirmed by bacterial isolation and commensal colonization studies. These results reveal complex interactions between host genetics and gut microbiota modulating susceptibility to CNS autoimmunity, providing insights into microbiome-directed strategies aimed at lowering the risk for autoimmune disease and underscoring the need to consider host genetics and baseline gut microbiome composition.


Assuntos
Encefalomielite Autoimune Experimental/genética , Microbioma Gastrointestinal/imunologia , Predisposição Genética para Doença , Interações entre Hospedeiro e Microrganismos/imunologia , Esclerose Múltipla/genética , Animais , Autoimunidade/genética , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/microbiologia , Feminino , Variação Genética , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Limosilactobacillus reuteri/imunologia , Masculino , Camundongos , Esclerose Múltipla/imunologia , Esclerose Múltipla/microbiologia
3.
Appetite ; 146: 104467, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31557496

RESUMO

Flavor-consequence learning refers to learned associations between flavor stimuli and post-oral consequences of food that affect food selection, amount eaten and affect. Forms of flavor-consequence learning include flavor aversions, flavor avoidance, conditioned satiety, expected satiety and appetition. Roux-en-Y gastric bypass surgery (RYGB) and other bariatric procedures alter gastrointestinal processing of food in a number of ways. Thus, it is plausible that these procedures alter post-oral unconditioned stimuli that support flavor-consequence learning, leading to altered food selection, amount eaten, and affect. Surprisingly, however, there is almost no research on the role of flavor-consequence learning in the effects of bariatric surgery on appetite. This issue urgently warrants investigation.


Assuntos
Aprendizagem por Associação , Cirurgia Bariátrica , Aromatizantes/análise , Preferências Alimentares/psicologia , Obesidade Mórbida/psicologia , Apetite , Derivação Gástrica , Humanos , Obesidade Mórbida/cirurgia , Período Pós-Operatório
4.
Am J Physiol Endocrinol Metab ; 316(4): E568-E577, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30753113

RESUMO

Deletion of the leptin receptor from vagal afferent neurons (VAN) using a conditional deletion (Nav1.8/LepRfl/fl) results in an obese phenotype with increased food intake and lack of exogenous cholecystokinin (CCK)-induced satiation in male mice. Female mice are partially protected from weight gain and increased food intake in response to ingestion of high-fat (HF) diets. However, whether the lack of leptin signaling in VAN leads to an obese phenotype or disruption of hypothalamic-pituitary-gonadal axis function in female mice is unclear. Here, we tested the hypothesis that leptin signaling in VAN is essential to maintain estrogen signaling and control of food intake, energy expenditure, and adiposity in female mice. Female Nav1.8/LepRfl/fl mice gained more weight, had increased gonadal fat mass, increased meal number in the dark phase, and increased total food intake compared with wild-type controls. Resting energy expenditure was unaffected. The decrease in food intake produced by intraperitoneal injection of CCK (3 µg/kg body wt) was attenuated in female Nav1.8/LepRfl/fl mice compared with wild-type controls. Intraperitoneal injection of ghrelin (100 µg/kg body wt) increased food intake in Nav1.8/LepRfl/fl mice but not in wild-type controls. Ovarian steroidogenesis was suppressed, resulting in decreased plasma estradiol, which was accompanied by decreased expression of estrogen receptor-1 (Esr1) in VAN but not in the hypothalamic arcuate nucleus. These data suggest that the absence of leptin signaling in VAN is accompanied by disruption of estrogen signaling in female mice, leading to an obese phenotype possibly via altered control of feeding behavior.


Assuntos
Ingestão de Alimentos/genética , Comportamento Alimentar/fisiologia , Neurônios Aferentes/metabolismo , Obesidade/genética , Receptores para Leptina/genética , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal/genética , Colecistocinina/farmacologia , Dieta Hiperlipídica , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético , Estradiol/metabolismo , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Comportamento Alimentar/efeitos dos fármacos , Feminino , Grelina/farmacologia , Camundongos , Obesidade/metabolismo , Saciação , Nervo Vago/citologia , Aumento de Peso/genética
5.
Appetite ; 131: 94-99, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30118785

RESUMO

We used a novel rat model to investigate the physiological bases of early satiation after Roux-en-Y gastric bypass surgery (RYGB). Female rats were subjected to RYGB or sham surgery. Chronic infusion catheters were placed in the Roux limb of RYGB rats and the corresponding anatomical locus of the jejuna of sham-RYGB rats. Rats were also ovariectomized and chronically treated with either estradiol (E2; 2 µg each 4th day SC) or the oil vehicle. Testing was begun 10-12 wk after surgery. Intrajejunal lipid infusions (10 min, 4.4 mL, 8.8 kcal) were performed just before test meals of a low-energy artificially sweetened gel diet (0.1 kcal/g) that RYGB rats ingest avidly. Intrajejunal lipid infusions reduced test-meal size more in RYGB rats than sham-operated rats, indicating that, at least after prolonged adaptation to surgery, the satiating actions of lipids acting intra- or post-jejunally are increased by RYGB and that accelerated meal appearance in the intestines after RYGB is not necessary for this effect. The satiating effects of intrajejunal lipid infusions were similar in E2-and oil-treated rats, suggesting that the effect was not dependent on an activational effect of estrogens. In a second experiment, pretreatment with the cholecystokinin A-receptor antagonist devazepide reduced the satiating effect of intrajejunal lipid infusions in E2-treated RYGB rats. Although these data are preliminary due to the smaller numbers of rats than in the first experiment, they suggest that cholecystokinin-mediated jejunal satiation contributes to early satiation after RYGB in ovariectomized rats with peri-ovulatory levels of estradiol. The results of these experiments may be relevant to understanding RYGB outcome in pre- and postmenopausal women.


Assuntos
Derivação Gástrica , Jejuno , Lipídeos/administração & dosagem , Saciação/fisiologia , Animais , Peso Corporal , Estradiol/administração & dosagem , Feminino , Ovariectomia , Ratos , Ratos Long-Evans
6.
Int J Eat Disord ; 50(6): 624-635, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28230907

RESUMO

Because binge eating and emotional eating vary through the menstrual cycle in human females, we investigated cyclic changes in binge-like eating in female rats and their control by estrogens. Binge-like eating was elicited by three cycles of 4 days of food restriction and 4 days of free feeding followed by a single frustrative nonreward-stress episode (15 min visual and olfactory exposure to a familiar palatable food) immediately before presentation of the palatable food. Intact rats showed binge-like eating during the diestrous and proestrous phases of the ovarian cycle, but not during the estrous (periovulatory) phase. Ovariectomized (OVX) rats not treated with estradiol (E2) displayed binge-like eating, whereas E2-treated OVX rats did not. The procedure did not increase signs of anxiety in an open-field test. OVX rats not treated with E2 that were subjected to food restriction and sacrificed immediately after frustrative nonreward had increased numbers of cells expressing phosphorylated extracellular signal-regulated kinases (ERK) in the central nucleus of the amygdala (CeA), paraventricular nucleus of hypothalamus (PVN), and dorsal and ventral bed nuclei of the stria terminalis (BNST) compared with nonrestricted or E2-treated rats. These data suggest that this female rat model is appropriate for mechanistic studies of some aspects of menstrual-cycle effects on emotional and binge eating in human females, that anxiety is not a sufficient cause of binge-like eating, and that the PVN, CeA, and BNST may contribute to information processing underlying binge-like eating.


Assuntos
Transtorno da Compulsão Alimentar/complicações , Estrogênios/metabolismo , Privação de Alimentos/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Ratos , Ratos Sprague-Dawley , Estresse Fisiológico
7.
Appetite ; 98: 133-41, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26707654

RESUMO

Weight re-gain within 2 y after Roux-en-Y gastric bypass (RYGB) is significantly associated with increased intake of and cravings for sweet foods. Here we describe a novel model of this late increase in sweet appetite. Ovariectomized RYGB and Sham-operated rats, with or without estradiol treatment, were maintained on Ensure liquid diet and offered a low-energy, artificially sweetened diet (ASD) 2 h/d. First, we tested rats more than six months after RYGB. ASD meals were larger in RYGB than Sham rats, whereas Ensure meals were smaller. General physical activity increased during ASD meals in RYGB rats, but not during Ensure meals. Second, new rats were adapted to ASD before surgery, and were then offered ASD again during 4-10 wk following surgery. Estradiol-treated RYGB rats lost the most weight and progressively increased ASD intake to >20 g/2 h in wk 9-10 vs. ∼3 g/2 h in Sham rats. Finally, the same rats were then treated with leptin or saline for 8 d. Leptin did not affect body weight, Ensure intake, or activity during meals, but slightly reduced ASD intake in estradiol-treated RYGB rats. Food-anticipatory activity was increased in estradiol-treated RYGB rats during the saline-injection tests. Because increased meal-related physical activity together with larger meals is evidence of hunger in rats, these data suggest that (1) RYGB can increase hunger for a low-energy sweet food in rats and (2) low leptin levels contribute to this hunger, but are not its only cause. This provides a unique rat model for the increased avidity for sweets that is significantly associated with weight recidivism late after RYGB.


Assuntos
Peso Corporal , Derivação Gástrica , Adoçantes não Calóricos/administração & dosagem , Animais , Sacarose Alimentar/administração & dosagem , Ingestão de Energia , Feminino , Alimentos Formulados , Fome , Leptina/sangue , Atividade Motora , Ovariectomia , Ratos , Ratos Long-Evans , Aumento de Peso , Redução de Peso
8.
Eur J Neurosci ; 40(7): 3055-66, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25040689

RESUMO

Amylin reduces meal size by activating noradrenergic neurons in the area postrema (AP). Neurons in the AP also mediate the eating-inhibitory effects of salmon calcitonin (sCT), a potent amylin agonist, but the phenotypes of the neurons mediating its effect are unknown. Here we investigated whether sCT activates similar neuronal populations to amylin, and if its anorectic properties also depend on AP function. Male rats underwent AP lesion (APX) or sham surgery. Meal patterns were analysed under ad libitum and post-deprivation conditions. The importance of the AP in mediating the anorectic action of sCT was examined in feeding experiments of dose-response effects of sCT in APX vs. sham rats. The effect of sCT to induce Fos expression was compared between surgery groups, and relative to amylin. The phenotype of Fos-expressing neurons in the brainstem was examined by testing for the co-expression of dopamine beta hydroxylase (DBH) or tryptophan hydroxylase (TPH). By measuring the apposition of vesicular glutamate transporter-2 (VGLUT2)-positive boutons, potential glutamatergic input to amylin- and sCT-activated AP neurons was compared. Similar to amylin, an intact AP was necessary for sCT to reduce eating. Further, co-expression between Fos activation and DBH after amylin or sCT did not differ markedly, while co-localization of Fos and TPH was minor. Approximately 95% of neurons expressing Fos and DBH after amylin or sCT treatment were closely apposed to VGLUT2-positive boutons. Our study suggests that the hindbrain pathways engaged by amylin and sCT share many similarities, including the mediation by AP neurons.


Assuntos
Área Postrema/fisiologia , Calcitonina/fisiologia , Ingestão de Alimentos/fisiologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/fisiologia , Neurônios/metabolismo , Animais , Área Postrema/efeitos dos fármacos , Área Postrema/metabolismo , Calcitonina/farmacologia , Dopamina beta-Hidroxilase/análise , Ingestão de Alimentos/efeitos dos fármacos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/farmacologia , Masculino , Neurônios/efeitos dos fármacos , Fenótipo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Triptofano Hidroxilase/análise , Proteína Vesicular 2 de Transporte de Glutamato/análise
9.
Gastroenterology ; 143(2): 325-7.e2, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22609384

RESUMO

Despite the fact that ∼85% of bariatric operations are performed in women, the effects of the reproductive axis function on outcome of bariatric surgery remain to be determined. Here we developed the first published model of Roux-en-Y gastric bypass (RYGB) in female rats. We show in ovariectomized rats receiving estradiol or control treatment that (1) RYGB-induced body weight loss and (2) the satiating efficacy of endogenous glucagon-like peptide-1 and cholecystokinin satiation were significantly increased in estradiol-treated rats. These data are relevant to the care of obese women, in particular perimenopausal women, undergoing bariatric surgery.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Derivação Gástrica , Obesidade/cirurgia , Saciação/efeitos dos fármacos , Redução de Peso/efeitos dos fármacos , Animais , Biomarcadores/metabolismo , Colecistocinina/metabolismo , Estradiol/administração & dosagem , Estrogênios/administração & dosagem , Comportamento Alimentar , Feminino , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Menopausa , Modelos Animais , Obesidade/metabolismo , Ovariectomia , Ratos , Resultado do Tratamento
10.
Am J Physiol Regul Integr Comp Physiol ; 305(11): R1215-67, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23904103

RESUMO

Hypothalamic-pituitary-gonadal (HPG) axis function fundamentally affects the physiology of eating. We review sex differences in the physiological and pathophysiological controls of amounts eaten in rats, mice, monkeys, and humans. These controls result from interactions among genetic effects, organizational effects of reproductive hormones (i.e., permanent early developmental effects), and activational effects of these hormones (i.e., effects dependent on hormone levels). Male-female sex differences in the physiology of eating involve both organizational and activational effects of androgens and estrogens. An activational effect of estrogens decreases eating 1) during the periovulatory period of the ovarian cycle in rats, mice, monkeys, and women and 2) tonically between puberty and reproductive senescence or ovariectomy in rats and monkeys, sometimes in mice, and possibly in women. Estrogens acting on estrogen receptor-α (ERα) in the caudal medial nucleus of the solitary tract appear to mediate these effects in rats. Androgens, prolactin, and other reproductive hormones also affect eating in rats. Sex differences in eating are mediated by alterations in orosensory capacity and hedonics, gastric mechanoreception, ghrelin, CCK, glucagon-like peptide-1 (GLP-1), glucagon, insulin, amylin, apolipoprotein A-IV, fatty-acid oxidation, and leptin. The control of eating by central neurochemical signaling via serotonin, MSH, neuropeptide Y, Agouti-related peptide (AgRP), melanin-concentrating hormone, and dopamine is modulated by HPG function. Finally, sex differences in the physiology of eating may contribute to human obesity, anorexia nervosa, and binge eating. The variety and physiological importance of what has been learned so far warrant intensifying basic, translational, and clinical research on sex differences in eating.


Assuntos
Ingestão de Alimentos/fisiologia , Caracteres Sexuais , Animais , Regulação do Apetite/fisiologia , Comportamento Alimentar/fisiologia , Feminino , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Humanos , Leptina/metabolismo , Masculino
11.
Endocrinology ; 164(1)2022 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-36423205

RESUMO

To better understand the physiological basis of obesity in women, we investigated whether obesity or menstrual cycle phase affects laboratory test-meal size or meal-stimulated plasma cholecystokinin (CCK) concentration. Women with healthy weight (body mass index [BMI] of 18.5-24.9 kg/m2, N = 16) or obesity (BMI 30-39.9 kg/m2, N = 20) were tested once in the late-follicular or peri-ovulatory phase (LF/PO) and once in the mid-luteal phase (ML). Meals of ham sandwiches were offered and blood was sampled. Menstrual cycle phases were verified with participants' reports of menses and measurements of progesterone and luteinizing hormone (LH) concentrations. Women with obesity ate significantly larger meals than women with healthy weight, (mean, 711 [95% CI, 402-1013] kJ, P = 0.001, during the LF/PO and 426 [105-734] kJ, P = 0.027, larger during the ML). Women with healthy weight ate smaller meals during LF/PO than ML (decrease, 510 [192-821 kJ], P = 0.008), but women with obesity did not (decrease, 226 [-87-542] kJ, P = 0.15). CCK concentrations 18 to 30 minutes after meal onset were lower in women with obesity than in women with healthy weight during LF/PO (3.6 [3.1-4.1] vs 6.1 [4.5-7.7] pmol/L; P = 0.004), but not during ML, with a significant interaction effect (1.8 [1.2-2.4] pmol/L, P = 0.048). Women with obesity consumed larger meals than women with healthy weight but displayed reduced meal-stimulated plasma CCK concentrations. These data are consistent with the hypothesis that a defect in CCK secretion compromises satiation in obese women and contributes to the development or maintenance of obesity.


Assuntos
Colecistocinina , Refeições , Obesidade , Feminino , Humanos , Colecistocinina/sangue , Obesidade/sangue , Obesidade/fisiopatologia , Refeições/fisiologia , Índice de Massa Corporal , Ciclo Menstrual
12.
Front Neurosci ; 16: 800976, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35250448

RESUMO

BACKGROUND: The importance of menstrual cycle physiology in appetite and obesity is poorly understood. We investigated the effects of body mass index (BMI), menstrual cycle phase and sweet and salty taste on monetary valuation of snack foods. METHODS: We recruited 72 women and after the application of in- and exclusion criteria 31 participants with healthy weight and 25 with obesity remained. The participants completed a willingness to pay (WTP) task to measure subjective value of 30 snack food items in the pre-ovulatory and mid-luteal cycle phases. RESULTS: Generalized linear mixed model (GLMM) analysis revealed that BMI, cycle phase and snack taste interacted to influence WTP (-0.15 [-0.22, -0.03], p = 0.002). Hence, WTP was inversely related to BMI, but the strength of the relation depended on cycle phase and taste. The WTP of participants with healthy weight for salty taste changed across cycle phase but the WTP for sweet taste was not affected by cycle phase. Moreover, the cycle effect for the salty snacks ceased in participants with obesity. CONCLUSION: The inverse effect of BMI on WTP valuation of snack foods contrasts with the positive effect of BMI on pleasantness ratings for milkshakes by the same women that we previously reported. This indicates that the two measures reflect different aspects of food-related valuative processing in obesity. Furthermore, the WTP data suggest that the selection of salty snacks may differ from that of sweet snacks in the pre-ovulatory phase of the menstrual cycle for individuals of healthy weight. The cycle phase does not seem to affect food valuation of participants with obesity. These findings are relevant to understanding and treating obesity in women.

13.
Endocrinology ; 149(4): 1609-17, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18096668

RESUMO

17beta-estradiol (E2), acting via estrogen receptor (ER)-alpha, inhibits feeding in animals. One mechanism apparently involves an increase in the satiating potency of cholecystokinin (CCK) released from the small intestine by ingested food. For example, the satiating potency of intraduodenal lipid infusions is increased by E2 in ovariectomized rats; this increased satiation is dependent on CCK, and it is accompanied by increases in the numbers of ERalpha-positive cells that express c-Fos in a subregion of the caudal nucleus tractus solitarius (cNTS) that receives abdominal vagal afferent projections. To test whether direct administration of E2 to this area of the hindbrain is sufficient to inhibit food intake, we first implanted 0.2 microg estradiol benzoate (EB) in cholesterol or cholesterol alone either sc or onto the surface of the hindbrain over the cNTS. Food intake was significantly reduced after hindbrain EB implants but not after sc EB implants. Next we verified that equimolar hindbrain implants of E2 and EB had similar feeding-inhibitory effects and determined that only small amounts of E2 reached brain areas outside the dorsal caudal hindbrain after hindbrain implants of (3)H-labeled E2. Neither plasma estradiol concentration nor plasma inflammatory cytokine concentration was increased by either hindbrain or sc EB implants. Finally, hindbrain EB implants, but not sc implants, increased c-Fos in ERalpha-positive cells in the cNTS after ip injection of 4 microg/kg CCK-8. We conclude that E2, acting via ERalpha in cNTS neurons, including neurons stimulated by ip CCK, is sufficient to inhibit feeding.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Estradiol/farmacologia , Receptor alfa de Estrogênio/análise , Rombencéfalo/efeitos dos fármacos , Núcleo Solitário/efeitos dos fármacos , Animais , Peso Corporal , Colecistocinina/farmacologia , Citocinas/sangue , Estradiol/sangue , Feminino , Ovariectomia , Proteínas Proto-Oncogênicas c-fos/análise , Ratos , Ratos Long-Evans , Núcleo Solitário/química
14.
Endocrinology ; 148(12): 5656-66, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17823256

RESUMO

Part of the mechanism through which estradiol, acting via estrogen receptor (ERalpha) signaling, inhibits feeding in rats and mice is increasing the satiating potency of cholecystokinin (CCK) acting on peripheral CCK-1 receptors. Ingested lipid is a principal secretagogue of intestinal CCK, and intraduodenal lipid infusions elicit CCK-mediated satiation in animals and humans. Here we tested whether estradiol affects the satiating potency of intraduodenal lipid infusions in ovariectomized rats and, using c-Fos immunocytochemistry, searched for potential brain sites of ERalpha involved. Food-deprived ovariectomized rats with open gastric cannulas sham fed 0.8 m sucrose 2 d after estradiol (estradiol benzoate, 10 mug, sc) or vehicle injection. Estradiol markedly increased the satiating potency of intraduodenal infusions of Intralipid but not the satiating potency of L-phenylalanine (10 min infusions, 0.44 ml/min, 0.13 kcal/ml), which in male rats satiates via a CCK-independent mechanism. Estradiol had no significant effect in rats pretreated with the CCK-1 receptor antagonist Devazepide (1 mg/kg, ip). The effect of estradiol on intraduodenal Intralipid-induced satiation was mirrored by selective increases in the number of cells expressing c-Fos immunoreactivity in a circumscribed region of the nucleus tractus solitarius (NTS), just caudal to the area postrema (cNTS) but not elsewhere in the NTS or the hypothalamic paraventricular or arcuate nuclei. In addition, a significant proportion of cNTS c-Fos-positive cells also expressed ERalpha. These data provide behavioral and cellular evidence that estradiol-ERalpha signaling in cNTS neurons increases the satiating potency of endogenous CCK released in response to ingested lipid.


Assuntos
Colecistocinina/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Ovariectomia , Saciação/efeitos dos fármacos , Núcleo Solitário/metabolismo , Animais , Colecistocinina/antagonistas & inibidores , Devazepida/farmacologia , Estradiol/administração & dosagem , Comportamento Alimentar/efeitos dos fármacos , Feminino , Imuno-Histoquímica , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Lipídeos , Masculino , Fenilalanina/administração & dosagem , Fenilalanina/farmacologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Núcleo Solitário/efeitos dos fármacos
15.
Pharmacol Biochem Behav ; 86(3): 493-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17307246

RESUMO

Peripheral administration of bacterial lipopolysaccharide (LPS) elicits anorexia in several species, including rats and mice. There is strong evidence that antagonism of serotonergic activity at 2C receptors (5-HT(2C)R) attenuates LPS anorexia in rats. Here we used pharmacological and genetic approaches to examine the role of the 5-HT(2C)R in LPS anorexia in mice. In Experiment 1, SB 242084, a potent and selective 5-HT(2C) antagonist (0.3 mg/kg) was injected intraperitoneally 15 min before intraperitoneal LPS (2 microg/kg) injections just prior to dark onset in c57BL/6 mice. Food intake was recorded 1, 2 and 4 h after LPS administration. In Experiment 2, we recorded 2, 4 and 24 h food intake following dark onset intraperitoneal LPS (0.125, 0.25, 0.5, 1 and 2 microg/kg) injections in mice with a genetic deletion of 5-HT(2C)R and their WT controls. Our pharmacological results suggest that at least part of the anorexia following peripheral LPS administration is mediated by an increase in 5-HT-ergic activity at the 5-HT(2C)R. Our genetic data, in contrast, suggest that 5-HT(2C)R is not a necessary part of LPS anorexia.


Assuntos
Anorexia/prevenção & controle , Lipopolissacarídeos/toxicidade , Antagonistas do Receptor 5-HT2 de Serotonina , Aminopiridinas/farmacologia , Animais , Animais Congênicos , Anorexia/induzido quimicamente , Anorexia/fisiopatologia , Anorexia/psicologia , Sequência de Bases , Primers do DNA/genética , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Indóis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Receptor 5-HT2C de Serotonina/deficiência , Receptor 5-HT2C de Serotonina/genética , Antagonistas da Serotonina/farmacologia
16.
Hum Reprod Update ; 23(3): 300-321, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28333235

RESUMO

BACKGROUND: Obesity is caused by an imbalance between energy intake, i.e. eating and energy expenditure (EE). Severe obesity is more prevalent in women than men worldwide, and obesity pathophysiology and the resultant obesity-related disease risks differ in women and men. The underlying mechanisms are largely unknown. Pre-clinical and clinical research indicate that ovarian hormones may play a major role. OBJECTIVE AND RATIONALE: We systematically reviewed the clinical and pre-clinical literature on the effects of ovarian hormones on the physiology of adipose tissue (AT) and the regulation of AT mass by energy intake and EE. SEARCH METHODS: Articles in English indexed in PubMed through January 2016 were searched using keywords related to: (i) reproductive hormones, (ii) weight regulation and (iii) central nervous system. We sought to identify emerging research foci with clinical translational potential rather than to provide a comprehensive review. OUTCOMES: We find that estrogens play a leading role in the causes and consequences of female obesity. With respect to adiposity, estrogens synergize with AT genes to increase gluteofemoral subcutaneous AT mass and decrease central AT mass in reproductive-age women, which leads to protective cardiometabolic effects. Loss of estrogens after menopause, independent of aging, increases total AT mass and decreases lean body mass, so that there is little net effect on body weight. Menopause also partially reverses women's protective AT distribution. These effects can be counteracted by estrogen treatment. With respect to eating, increasing estrogen levels progressively decrease eating during the follicular and peri-ovulatory phases of the menstrual cycle. Progestin levels are associated with eating during the luteal phase, but there does not appear to be a causal relationship. Progestins may increase binge eating and eating stimulated by negative emotional states during the luteal phase. Pre-clinical research indicates that one mechanism for the pre-ovulatory decrease in eating is a central action of estrogens to increase the satiating potency of the gastrointestinal hormone cholecystokinin. Another mechanism involves a decrease in the preference for sweet foods during the follicular phase. Genetic defects in brain α-melanocycte-stimulating hormone-melanocortin receptor (melanocortin 4 receptor, MC4R) signaling lead to a syndrome of overeating and obesity that is particularly pronounced in women and in female animals. The syndrome appears around puberty in mice with genetic deletions of MC4R, suggesting a role of ovarian hormones. Emerging functional brain-imaging data indicates that fluctuations in ovarian hormones affect eating by influencing striatal dopaminergic processing of flavor hedonics and lateral prefrontal cortex processing of cognitive inhibitory controls of eating. There is a dearth of research on the neuroendocrine control of eating after menopause. There is also comparatively little research on the effects of ovarian hormones on EE, although changes in ovarian hormone levels during the menstrual cycle do affect resting EE. WIDER IMPLICATIONS: The markedly greater obesity burden in women makes understanding the diverse effects of ovarian hormones on eating, EE and body adiposity urgent research challenges. A variety of research modalities can be used to investigate these effects in women, and most of the mechanisms reviewed are accessible in animal models. Therefore, human and translational research on the roles of ovarian hormones in women's obesity and its causes should be intensified to gain further mechanistic insights that may ultimately be translated into novel anti-obesity therapies and thereby improve women's health.


Assuntos
Tecido Adiposo/fisiologia , Ingestão de Alimentos/psicologia , Estrogênios/metabolismo , Obesidade/metabolismo , Progesterona/metabolismo , Fatores Etários , Animais , Feminino , Humanos , Menopausa/fisiologia , Ciclo Menstrual/fisiologia , Obesidade/etiologia , Maturidade Sexual , Saúde da Mulher
17.
Physiol Rep ; 5(21)2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29138359

RESUMO

In contrast to the many studies of the effects of individual amino acids (AAs) on eating, no studies have compared the effects of different AAs on eating and underlying preabsorptive gastrointestinal mechanisms. To compare the effects of intraduodenal infusions of l-tryptophan (TRP), l-leucine (LEU), l-phenylalanine (PHE) and l-glutamine (GLN) on appetite, gastrointestinal hormone responses (including ghrelin, cholecystokinin (CCK), peptide YY (PYY) and glucagon-like peptide-1 [GLP-1]), glycemia (glucagon, insulin and glucose) and test meal size in healthy males, we retrospectively analyzed data from four published independent, randomized, double-blind, placebo-controlled studies of 90-min intraduodenal infusions of the individual AAs. The designs of the studies were identical, except the dose of TRP (0.15 kcal/min) was lower than that of the other AAs (0.45 kcal/min) because higher doses of this AA were not well tolerated. TRP and LEU decreased intake more than PHE (reductions relative to control, ~219 ± 68, ~170 ± 48 and ~12 ± 57 kcal, respectively), and TRP decreased intake more than GLN (~31 ± 82 kcal). These effects of TRP and LEU versus GLN, but not versus PHE, were paralleled by greater decreases in plasma ghrelin, and increases in CCK, concentrations. TRP increased PYY more than GLN or LEU, but not PHE. LEU increased PYY less than PHE. No significant differences were detected for GLP-1. PHE increased glucagon more than TRP or LEU, and increased insulin more than TRP. Under our experimental conditions, intraduodenal TRP and LEU were more satiating than PHE and GLN. The greater satiating efficacy of LEU versus PHE was significantly dissociated from the effects of these AAs on PYY, while the greater satiating potency of TRP versus PHE was significantly dissociated from the effects of these AAs on insulin and glucagon. In contrast, ghrelin and CCK, and potentially other mechanisms, including central sensing of individual AAs, appear to be stronger candidate mechanisms for the relative satiating effects obtained.


Assuntos
Aminoácidos/farmacologia , Depressores do Apetite/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , Hormônios Gastrointestinais/sangue , Adolescente , Adulto , Aminoácidos/administração & dosagem , Apetite/efeitos dos fármacos , Depressores do Apetite/administração & dosagem , Glicemia/metabolismo , Colecistocinina/sangue , Método Duplo-Cego , Ingestão de Energia/efeitos dos fármacos , Grelina/sangue , Peptídeo 1 Semelhante ao Glucagon/sangue , Hormônios/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Peptídeo YY/sangue , Ensaios Clínicos Controlados Aleatórios como Assunto , Adulto Jovem
19.
Physiol Behav ; 82(2-3): 251-61, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15276786

RESUMO

Lipopolysaccharide (LPS) derived from the cell walls of gram-negative bacteria causes a robust acute phase response (APR) that includes fever, anorexia, and many other elements. Because immune system function, including some models of illness anorexia, is sexually differentiated, we investigated the sexual differentiation of the anorexia induced by intraperitoneal LPS injections in rats. Cycling female Long-Evans rats tested either during diestrus or estrus ate less following 6.25 microg/kg LPS than did intact males. Following 12.5 microg/kg LPS, females in estrus ate less than either females during diestrus or males. Similarly, a more pronounced anorexia occurred following 12.5, 25, and 50 microg/kg LPS in ovariectomized females that received cyclic estradiol treatment and were tested on the day modeling estrus than in untreated ovariectomized rats. LPS also increased the length of the rats' ovarian cycles, usually by a day, especially when injected during diestrus. As in male rats, when LPS injections were repeated in the same rats, both estradiol-treated and untreated rats failed to display any significant anorexia. The inhibitory effects of LPS on eating in intact and ovariectomized rats were expressed solely as decreases in spontaneous meal frequency, without significant alteration of spontaneous meal size. These data indicate that anorexia following peripheral LPS administration is sexually differentiated and that estradiol is sufficient to produce this response. The mechanism of the pathophysiological effect of estradiol on meal frequency appears to be different from the physiological effect of estradiol on food intake because the latter is expressed solely as a change in meal size.


Assuntos
Anorexia/fisiopatologia , Regulação do Apetite/fisiologia , Estradiol/fisiologia , Comportamento Alimentar/fisiologia , Reação de Fase Aguda/induzido quimicamente , Análise de Variância , Animais , Anorexia/induzido quimicamente , Relação Dose-Resposta a Droga , Ciclo Estral/fisiologia , Feminino , Injeções Intraperitoneais , Lipopolissacarídeos/administração & dosagem , Masculino , Ratos , Ratos Long-Evans , Fatores Sexuais
20.
Horm Mol Biol Clin Investig ; 19(3): 163-92, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25390024

RESUMO

Abstract Eating is a simple behavior with complex functions. The unconscious neuroendocrine process that stops eating and brings a meal to its end is called satiation. Energy homeostasis is mediated accomplished through the control of meal size via satiation. It involves neural integrations of phasic negative-feedback signals related to ingested food and tonic signals, such as those related to adipose tissue mass. Energy homeostasis is accomplished through adjustments in meal size brought about by changes in these satiation signals. The best understood meal-derived satiation signals arise from gastrointestinal nutrient sensing. Gastrointestinal hormones secreted during the meal, including cholecystokinin, glucagon-like peptide 1, and PYY, mediate most of these. Other physiological signals arise from activation of metabolic-sensing neurons, mainly in the hypothalamus and caudal brainstem. We review both classes of satiation signal and their integration in the brain, including their processing by melanocortin, neuropeptide Y/agouti-related peptide, serotonin, noradrenaline, and oxytocin neurons. Our review is not comprehensive; rather, we discuss only what we consider the best-understood mechanisms of satiation, with a special focus on normally operating physiological mechanisms.


Assuntos
Sistemas Neurossecretores/fisiologia , Saciação/fisiologia , Animais , Encéfalo/fisiologia , Hormônios Gastrointestinais/metabolismo , Trato Gastrointestinal/metabolismo , Humanos , Fenômenos Fisiológicos da Nutrição/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA