Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Diabetes Obes Metab ; 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38720199

RESUMEN

Interventions aimed at weight control often have limited effectiveness in combating obesity. This review explores how obesity-induced dysfunction in white (WAT) and brown adipose tissue (BAT), skeletal muscle, and the brain blunt weight loss, leading to retention of stored fat. In obesity, increased adrenergic stimulation and inflammation downregulate ß-adrenoreceptors and impair catecholaminergic signalling in adipocytes. This disrupts adrenergic-mediated lipolysis, diminishing lipid oxidation in both white and brown adipocytes, lowering thermogenesis and blunting fat loss. Emerging evidence suggests that WAT fibrosis is associated with worse weight loss outcomes; indeed, limiting collagen and laminin-α4 deposition mitigates WAT accumulation, enhances browning, and protects against high-fat-diet-induced obesity. Obesity compromises mitochondrial oxidative capacity and lipid oxidation in skeletal muscle, impairing its ability to switch between glucose and lipid metabolism in response to varying nutrient levels and exercise. This dysfunctional phenotype in muscle is exacerbated in the presence of obesity-associated sarcopenia. Additionally, obesity suppresses sarcolipin-induced sarcoplasmic reticulum calcium ATPase (SERCA) activation, resulting in reduced oxidative capacity, diminished energy expenditure, and increased adiposity. In the hypothalamus, obesity and overnutrition impair insulin and leptin signalling. This blunts central satiety signals, favouring a shift in energy balance toward energy conservation and body fat retention. Moreover, both obese animals and humans demonstrate impaired dopaminergic signalling and diminished responses to nutrient intake in the striatum, which tend to persist after weight loss. This may result in enduring inclinations toward overeating and a sedentary lifestyle. Collectively, the tissue adaptations described pose significant challenges to effectively achieving and sustaining weight loss in obesity.

2.
Nature ; 546(7656): 107-112, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28538730

RESUMEN

Menopause is associated with bone loss and enhanced visceral adiposity. A polyclonal antibody that targets the ß-subunit of the pituitary hormone follicle-stimulating hormone (Fsh) increases bone mass in mice. Here, we report that this antibody sharply reduces adipose tissue in wild-type mice, phenocopying genetic haploinsufficiency for the Fsh receptor gene Fshr. The antibody also causes profound beiging, increases cellular mitochondrial density, activates brown adipose tissue and enhances thermogenesis. These actions result from the specific binding of the antibody to the ß-subunit of Fsh to block its action. Our studies uncover opportunities for simultaneously treating obesity and osteoporosis.


Asunto(s)
Tejido Adiposo/metabolismo , Adiposidad , Hormona Folículo Estimulante de Subunidad beta/antagonistas & inhibidores , Termogénesis , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo Beige/efectos de los fármacos , Tejido Adiposo Beige/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Adiposidad/efectos de los fármacos , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Dieta Alta en Grasa/efectos adversos , Femenino , Hormona Folículo Estimulante de Subunidad beta/inmunología , Haploinsuficiencia , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/prevención & control , Osteoporosis/tratamiento farmacológico , Ovariectomía , Consumo de Oxígeno/efectos de los fármacos , Receptores de HFE/antagonistas & inhibidores , Receptores de HFE/genética , Receptores de HFE/metabolismo , Termogénesis/efectos de los fármacos , Proteína Desacopladora 1/biosíntesis
3.
Int J Obes (Lond) ; 46(5): 918-925, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35017714

RESUMEN

BACKGROUND: Obesity is often associated with hyperinsulinemia due to insulin resistance. In mice models of hyperinsulinemia, adenovirus-derived E4orf1 protein promotes glucose disposal via insulin-independent pathway, and reduces insulin response to glucose load, described as its "Insulin Sparing Action". This is likely because less insulin is needed for disposing glucose in presence of E4orf1, however, there are other potential possibilities. This study determined if E4orf1 reduces insulin response to glucose load because it a) suppresses the ability of pancreatic ß-cells to secret insulin, or b) upregulates glucagon production by the pancreas. METHODS: C57BL/6J wild type (control) and transgenic C57BL/6J (E4orf1) mice that express E4orf1 protein in adipose tissue upon doxycycline feeding, were used. Post-doxycycline feeding, insulin and glucagon secretion in response to glibenclamide or phenylephrine were compared between the two groups. The pancreases were examined for histological changes. RESULTS: In response to glibenclamide, E4orf1 mice secreted more insulin and exhibited lower blood glucose compared to control (47.4 ± 4.4 vs 27.4 ± 3.7 mg/dl, p < 0.003), but showed no difference in glucagon secretion. Post-phenylephrine injection, no differences were observed between the two groups for glucagon or insulin, except E4orf1 mice had a lower blood glucose rise after 10-min of injection compared to the control (39.7 ± 4.7 vs. 58.3 ± 7.5 mg/dl, p < 0.05). E4orf1 mice had significantly larger pancreatic islets and higher number of islets per mm2 tissue area. Neither the size nor the number of islets met the criteria of hypertrophy or hyperplasia. CONCLUSIONS/INTERPRETATION: E4orf1 retains and may enhance the ability of the pancreases to secret insulin in response to insulin secretagogue. Glucagon does not seem to play a role in the Insulin Sparing Action of E4orf1. Overall, the histology studies support better pancreatic islet health in presence of E4orf1, compared to that in control mice. The "insulin-independent" role of E4orf1 has potential therapeutic implications in addressing hyperinsulinemia in obesity.


Asunto(s)
Proteínas E4 de Adenovirus , Hiperinsulinismo , Células Secretoras de Insulina , Islotes Pancreáticos , Proteínas E4 de Adenovirus/metabolismo , Animales , Glucemia/metabolismo , Doxiciclina , Glucagón , Glucosa/metabolismo , Gliburida , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Fenilefrina
4.
Int J Obes (Lond) ; 45(2): 383-395, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32951009

RESUMEN

INTRODUCTION: The hypothalamo-pituitary-adrenal (HPA) axis is perturbed in obesity. We previously reported presence of leptin resistance in the brainstem and uncoupling between central noradrenergic tone and the HPA axis in obesity-prone (DIO) rats. Metformin is shown to lower body weight and adiposity, but the underlying mechanism is unclear. We hypothesized that this is associated with restored HPA axis function. METHODS: Adult male DIO rats were placed on either a regular chow or HF diet for 7 weeks. Starting week 4, the animals were given either a low dose (60 mg/kg) or high dose (300 mg/kg) of metformin in drinking water. In addition to body weight and feeding, we examined different arms of the HPA axis to test if metformin can reinstate its function and coupling. To understand potential mechanisms, leptin signaling in the brainstem and circulating free fatty acid levels were also assessed. RESULTS: Metformin treatment lowered weight gain, fat mass, caloric intake, and serum leptin levels. HPA axis activity as determined by corticotropin-releasing hormone in the median eminence and serum corticosterone was decreased by metformin in a dose-dependent manner, and so was norepinephrine (NE) in the paraventricular nucleus. Importantly, metformin completely normalized the NE-HPA axis uncoupling. While brainstem pSTAT-3 and SOCS-3, key markers of leptin signaling, were not different between groups, circulating saturated and unsaturated free fatty acids were reduced in HF-fed, metformin-treated animals. CONCLUSIONS: These findings suggest that oral metformin can successfully correct HPA axis dysfunction that is associated with lowered circulating free fatty acids in DIO rats, thereby uncovering a novel effect of metformin in the treatment of obesity.


Asunto(s)
Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Metformina/farmacología , Obesidad/metabolismo , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Corticosterona/sangre , Dieta Alta en Grasa , Masculino , Ratas
5.
Int J Mol Sci ; 22(24)2021 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-34947993

RESUMEN

Type 1 diabetes (T1D) is characterized by hyperphagia, hyperglycemia and activation of the hypothalamic-pituitary-adrenal (HPA) axis. We have reported previously that daily leptin injections help to alleviate these symptoms. Therefore, we hypothesized that leptin gene therapy could help to normalize the neuroendocrine dysfunction seen in T1D. Adult male Sprague Dawley rats were injected i.v. with a lentiviral vector containing the leptin gene or green fluorescent protein. Ten days later, they were injected with the vehicle or streptozotocin (STZ). HPA function was assessed by measuring norepinephrine (NE) levels in the paraventricular nucleus (PVN) and serum corticosterone (CS). Treatment with the leptin lentiviral vector (Lepvv) increased leptin and insulin levels in non-diabetic rats, but not in diabetic animals. There was a significant reduction in blood glucose levels in diabetic rats due to Lepvv treatment. Both NE levels in the PVN and serum CS were reduced in diabetic rats treated with Lepvv. Results from this study provide evidence that leptin gene therapy in STZ-induced diabetic rats was able to partially normalize some of the neuroendocrine abnormalities, but studies with higher doses of the Lepvv are needed to develop this into a viable option for treating T1D.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Vectores Genéticos/administración & dosificación , Leptina/genética , Animales , Corticosterona/sangre , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animales de Enfermedad , Terapia Genética , Inyecciones Intravenosas , Lentivirus/genética , Masculino , Norepinefrina/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas , Ratas Sprague-Dawley
6.
Mediators Inflamm ; 2020: 8862034, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33281501

RESUMEN

BACKGROUND: Obesity is associated with the gut microbiota and decreased micronutrient status. Bariatric surgery is a recommended therapy for obesity. It can positively affect the composition of the gut bacteria but also disrupt absorption of nutrients. Low levels of micronutrients can affect metabolic processes, like glycolysis, TCA cycle, and oxidative phosphorylation, that are associated with the immune system also known as immunometabolism. METHODS: MEDLINE, PUBMED, and Google Scholar were searched. Articles involving gut microbiome, micronutrient deficiency, gut-targeted therapies, transcriptome analysis, micronutrient supplementation, and bariatric surgery were included. RESULTS: Studies show that micronutrients play a pivotal role in the intestinal immune system and regulating immunometabolism. Research demonstrates that gut-targeting therapies may improve the microbiome health for bariatric surgery populations. There is limited research that examines the role of micronutrients in modulating the gut microbiota among the bariatric surgery population. CONCLUSIONS: Investigations are needed to understand the influence that micronutrient deficiencies have on the gut, particularly immunometabolism. Nutritional transcriptomics shows great potential in providing this type of analysis to develop gut-modulating therapies as well as more personalized nutrition recommendations for bariatric surgery patients.


Asunto(s)
Cirugía Bariátrica/métodos , Microbioma Gastrointestinal , Micronutrientes , Obesidad Mórbida/cirugía , Ciclo del Ácido Cítrico , Femenino , Ácido Fólico/metabolismo , Alimentos Funcionales , Glucólisis , Humanos , Sistema Inmunológico , Intestinos/patología , Hierro/metabolismo , Masculino , Desnutrición , Estado Nutricional , Obesidad Mórbida/inmunología , Obesidad Mórbida/microbiología , Fosforilación Oxidativa , Probióticos , Tiamina/metabolismo , Transcriptoma , Vitamina B 12/metabolismo , Vitamina D/metabolismo
7.
Appetite ; 146: 104340, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31265857

RESUMEN

With behavioral and pharmacological interventions continuously failing to tackle the obesity epidemic, bariatric surgery has been hailed as the most effective treatment strategy. Current literature suggests that bariatric surgery successfully decreases body weight and excess fat mass through targeting both variables of the energy homeostasis - energy intake and energy expenditure. Here we review current knowledge on changes in caloric consumption, an important arm in the energy balance equation, in rodent models of bariatric surgery. In particular, circadian feeding dynamics, post-surgical caloric intake at both "rapid weight loss" phase and "weight maintenance" phase, as well as meal pattern analysis will be the subject of this review. Considering that different types of bariatric surgery may trigger differential energy intake dynamics resulting in variable weight loss outcomes, the effects of most popular surgeries - vertical sleeve gastrectomy (VSG), Roux-en-Y gastric bypass (RYGB), and gastric banding (GB) - are elaborated. Potential candidate mechanisms underlying alterations in food intake and meal patterns following different bariatric procedures are briefly discussed at the end.


Asunto(s)
Cirugía Bariátrica/métodos , Ingestión de Energía/fisiología , Metabolismo Energético/fisiología , Comidas/fisiología , Obesidad/cirugía , Animales , Modelos Animales de Enfermedad , Conducta Alimentaria , Ratones , Periodo Posoperatorio , Ratas , Resultado del Tratamiento , Pérdida de Peso
8.
Int J Obes (Lond) ; 43(11): 2143-2150, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30718818

RESUMEN

OBJECTIVES: Dieting often fails because weight loss triggers strong counter-regulatory biological responses such as increased hunger and hypometabolism that are thought to be critically dependent on the master fuel sensor in the mediobasal hypothalamus (MBH). Because prolonged starvation has been shown to increase AgRP and NPY, the expression level of these two orexigenic genes has been taken as an experimental readout for the presence or absence of hunger. Roux-en-Y gastric bypass (RYGB) surgery leads to a significant weight loss without inducing the associated hunger, indicating possible changes in hypothalamic neuropeptides and/or signaling. Our goal was to assess key genes in the MBH involved in regulating body weight, appetite, and inflammation/oxidative stress after RYGB surgery in mice. METHODS: Obese mice on a high-fat diet were subjected to either sham or RYGB surgery, or caloric restriction to match the weight of RYGB group. Chow-fed mice without surgery served as an additional control group. After 2 or 12 weeks post-surgery, hypothalamic genes were analyzed by real-time qPCR. RESULTS: During the rapid weight loss phase at 2 weeks after RYGB surgery, hypothalamic AgRP and NPY gene expression was not increased compared to mice with sham surgery, indicating that the mice are not hungry. In contrast, the same weight loss induced by caloric restriction promptly triggered increased AgRP and NPY expression. This differential effect of RYGB and caloric restriction was no longer observed during the weight-maintenance phase at 12 weeks after surgery. A similar differential effect was observed for ObRb, but not for POMC and CART expression. Furthermore, RAGE and IBA-1, two markers for inflammation/oxidative stress, were significantly suppressed after RYGB compared to caloric restriction at 2 weeks post-surgery. CONCLUSIONS: These findings suggest that RYGB prevents the biologically adaptive hunger response triggered by undernutrition and weight loss, and suppresses weight loss-induced hypothalamic inflammation markers.


Asunto(s)
Proteína Relacionada con Agouti/análisis , Restricción Calórica , Dieta Alta en Grasa , Derivación Gástrica , Hipotálamo/química , Neuropéptido Y/análisis , Animales , Ratones
10.
Alzheimers Dement ; 12(8): 851-61, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26928090

RESUMEN

INTRODUCTION: Epidemiologic studies have demonstrated an association between diabetes and dementia. Insulin signaling within the brain, in particular within the hypothalamus regulates carbohydrate, lipid, and branched chain amino acid (BCAA) metabolism in peripheral organs such as the liver and adipose tissue. We hypothesized that cerebral amyloidosis impairs central nervous system control of metabolism through disruption of insulin signaling in the hypothalamus, which dysregulates glucose and BCAA homeostasis resulting in increased susceptibility to diabetes. METHODS: We examined whether APP/PS1 mice exhibit increased susceptibility to aging or high-fat diet (HFD)-induced metabolic impairment using metabolic phenotyping and insulin-signaling studies. RESULTS: APP/PS1 mice were more susceptible to high-fat feeding and aging-induced metabolic dysregulation including disrupted BCAA homeostasis and exhibited impaired hypothalamic insulin signaling. DISCUSSION: Our data suggest that AD pathology increases susceptibility to diabetes due to impaired hypothalamic insulin signaling, and that plasma BCAA levels could serve as a biomarker of hypothalamic insulin action in patients with AD.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Aminoácidos de Cadena Ramificada/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Enfermedades Metabólicas/etiología , Transducción de Señal/fisiología , Enfermedad de Alzheimer/genética , Aminoácidos de Cadena Ramificada/sangre , Precursor de Proteína beta-Amiloide/genética , Animales , Peso Corporal/genética , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Ingestión de Alimentos/genética , Regulación de la Expresión Génica/genética , Humanos , Hipotálamo/fisiopatología , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Transgénicos , Mutación/genética , Presenilina-1/genética , Triglicéridos/metabolismo
11.
Nutr Diabetes ; 14(1): 40, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38844453

RESUMEN

BACKGROUND: High-protein diets are often enriched with branched-chain amino acids (BCAAs) known to enhance protein synthesis and provide numerous physiological benefits, but recent studies reveal their association with obesity and diabetes. In support of this, protein or BCAA supplementation is shown to disrupt glucose metabolism while restriction improves it. However, it is not clear if these are primary, direct effects of BCAAs or secondary to other physiological changes during chronic manipulation of dietary BCAAs. METHODS: Three-month-old C57Bl/6 mice were acutely treated with either vehicle/BCAAs or BT2, a BCAA-lowering compound, and detailed in vivo metabolic phenotyping, including frequent sampling and pancreatic clamps, were conducted. RESULTS: Using a catheter-guided frequent sampling method in mice, here we show that a single infusion of BCAAs was sufficient to acutely elevate blood glucose and plasma insulin. While pre-treatment with BCAAs did not affect glucose tolerance, a constant infusion of BCAAs during hyperinsulinemic-euglycemic clamps impaired whole-body insulin sensitivity. Similarly, a single injection of BT2 was sufficient to prevent BCAA rise during fasting and markedly improve glucose tolerance in high-fat-fed mice, suggesting that abnormal glycemic control in obesity may be causally linked to high circulating BCAAs. We further show that chemogenetic over-activation of AgRP neurons in the hypothalamus, as present in obesity, significantly impairs glucose tolerance that is completely normalized by acute BCAA reduction. Interestingly, most of these effects were demonstrated only in male, but not in female mice. CONCLUSION: These findings suggest that BCAAs per se can acutely impair glucose homeostasis and insulin sensitivity, thus offering an explanation for how they may disrupt glucose metabolism in the long-term as observed in obesity and diabetes. Our findings also reveal that AgRP neuronal regulation of blood glucose is mediated through BCAAs, further elucidating a novel mechanism by which brain controls glucose homeostasis.


Asunto(s)
Proteína Relacionada con Agouti , Aminoácidos de Cadena Ramificada , Glucemia , Resistencia a la Insulina , Ratones Endogámicos C57BL , Neuronas , Animales , Resistencia a la Insulina/fisiología , Proteína Relacionada con Agouti/metabolismo , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Masculino , Ratones , Glucemia/metabolismo , Femenino , Aminoácidos de Cadena Ramificada/metabolismo , Insulina/sangre , Insulina/metabolismo , Técnica de Clampeo de la Glucosa , Dieta Alta en Grasa , Obesidad/metabolismo
12.
Antioxidants (Basel) ; 12(2)2023 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-36829974

RESUMEN

Alzheimer's disease (AD) is the most common type of dementia that affects millions of individuals worldwide. It is an irreversible neurodegenerative disorder that is characterized by memory loss, impaired learning and thinking, and difficulty in performing regular daily activities. Despite nearly two decades of collective efforts to develop novel medications that can prevent or halt the disease progression, we remain faced with only a few options with limited effectiveness. There has been a recent growth of interest in the role of nutrition in brain health as we begin to gain a better understanding of what and how nutrients affect hormonal and neural actions that not only can lead to typical cardiovascular or metabolic diseases but also an array of neurological and psychiatric disorders. Vitamins and minerals, also known as micronutrients, are elements that are indispensable for functions including nutrient metabolism, immune surveillance, cell development, neurotransmission, and antioxidant and anti-inflammatory properties. In this review, we provide an overview on some of the most common vitamins and minerals and discuss what current studies have revealed on the link between these essential micronutrients and cognitive performance or AD.

13.
Nutrients ; 15(7)2023 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-37049555

RESUMEN

Type 2 diabetes (T2D) is a challenging health concern worldwide. A lifestyle intervention to treat T2D is difficult to adhere, and the effectiveness of approved medications such as metformin, thiazolidinediones (TZDs), and sulfonylureas are suboptimal. On the other hand, bariatric procedures such as Roux-en-Y gastric bypass (RYGB) are being recognized for their remarkable ability to achieve diabetes remission, although the underlying mechanism is not clear. Recent evidence points to branched-chain amino acids (BCAAs) as a potential contributor to glucose impairment and insulin resistance. RYGB has been shown to effectively lower plasma BCAAs in insulin-resistant or T2D patients that may help improve glycemic control, but the underlying mechanism for BCAA reduction is not understood. Hence, we attempted to explore the mechanism by which RYGB reduces BCAAs. To this end, we randomized diet-induced obese (DIO) mice into three groups that underwent either sham or RYGB surgery or food restriction to match the weight of RYGB mice. We also included regular chow-diet-fed healthy mice as an additional control group. Here, we show that compared to sham surgery, RYGB in DIO mice markedly lowered serum BCAAs most likely by rescuing BCAA breakdown in both liver and white adipose tissues. Importantly, the restored BCAA metabolism following RYGB was independent of caloric intake. Fasting insulin and HOMA-IR were decreased as expected, and serum valine was strongly associated with insulin resistance. While gut hormones such as glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) are postulated to mediate various surgery-induced metabolic benefits, mice lacking these hormonal signals (GLP-1R/Y2R double KO) were still able to effectively lower plasma BCAAs and improve glucose tolerance, similar to mice with intact GLP-1 and PYY signaling. On the other hand, mice deficient in fibroblast growth factor 21 (FGF21), another candidate hormone implicated in enhanced glucoregulatory action following RYGB, failed to decrease plasma BCAAs and normalize hepatic BCAA degradation following surgery. This is the first study using an animal model to successfully recapitulate the RYGB-led reduction of circulating BCAAs observed in humans. Our findings unmasked a critical role of FGF21 in mediating the rescue of BCAA metabolism following surgery. It would be interesting to explore the possibility of whether RYGB-induced improvement in glucose homeostasis is partly through decreased BCAAs.


Asunto(s)
Diabetes Mellitus Tipo 2 , Derivación Gástrica , Resistencia a la Insulina , Humanos , Ratones , Animales , Obesidad/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Aminoácidos de Cadena Ramificada , Insulina , Péptido 1 Similar al Glucagón/metabolismo , Glucosa , Glucemia/metabolismo
14.
Ann Surg ; 255(2): 294-301, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22202582

RESUMEN

OBJECTIVE: To determine the role of the common hepatic branch of the abdominal vagus on the beneficial effects of Roux-en-Y gastric bypass (RYGB) on weight loss, food intake, food choice, and energy expenditure in a rat model. BACKGROUND: Although changes in gut hormone patterns are the leading candidates in RYGB's effects on appetite, weight loss, and reversal of diabetes, a potential role for afferent signaling through the vagal hepatic branch potentially sensing glucose levels in the hepatic portal vein has recently been suggested in a mouse model of RYGB. METHODS: Male Sprague-Dawley rats underwent either RYGB alone (RYGB; n = 7), RYGB + common hepatic branch vagotomy (RYGB + HV; n = 6), or sham procedure (sham; n = 9). Body weight, body composition, meal patterns, food choice, energy expenditure, and fecal energy loss were monitored up to 3 months after intervention. RESULTS: Both RYGB and RYGB + HV significantly reduced body weight, adiposity, meal size, and fat preference, and increased satiety, energy expenditure, and respiratory exchange rate compared with sham procedure, and there were no significant differences in these effects between RYGB and RYGB + HV rats. CONCLUSIONS: Integrity of vagal nerve supply to the liver, hepatic portal vein, and the proximal duodenum provided by the common hepatic branch is not necessary for RYGB to reduce food intake and body weight or increase energy expenditure. Specifically, it is unlikely that a hepatic portal vein glucose sensor signaling RYGB-induced increased intestinal gluconeogenesis to the brain depends on vagal afferent fibers.


Asunto(s)
Regulación del Apetito/fisiología , Metabolismo Energético/fisiología , Derivación Gástrica , Hígado/inervación , Obesidad Mórbida/cirugía , Vena Porta/inervación , Nervio Vago/fisiología , Pérdida de Peso/fisiología , Animales , Modelos Animales de Enfermedad , Venas Hepáticas/inervación , Hígado/irrigación sanguínea , Masculino , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento , Vagotomía , Nervio Vago/cirugía
15.
J Mol Med (Berl) ; 100(5): 665-676, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35286401

RESUMEN

Chronic exposure to high levels of particulate matter (PM) is correlated to a higher prevalence of cardio-metabolic disturbances. Adipose tissue represents a pivotal regulator of metabolic homeostasis, and its dysfunction is associated with health issues in PM-exposed models. This review discusses the adaptive changes of white (WAT) and brown (BAT) adipose tissue in response to fine particulate matter (PM2.5), investigating the underlying pathophysiology. In exposed models, PM2.5 increases oxidative stress and impairs mitochondria functionality and biogenesis in WAT and BAT. Chronic exposure also upregulates the main apoptotic/pro-inflammatory pathways and promotes the infiltration of monocytes and the accumulation of activated macrophages. Oxidative stress and inflammation are responsible for the inhibition of insulin signal transduction and glucose uptake in both the adipose tissues. The increased inflammatory status also suppresses the metabolic activity of brown adipocytes, promoting the whitening. Altogether, this evidence suggests the shift of WAT and BAT toward an inflammatory and metabolic dysfunctional phenotype. Although the underlying mechanisms remain to be clarified, the development of inflammation in lungs, gut, and hypothalamus seems to have a pivotal role in the alteration of adipose tissue homeostasis. The potential consequences on systemic cardio-metabolic health render the relationship PM-adipose tissue a key issue to investigate.


Asunto(s)
Tejido Adiposo Pardo , Resistencia a la Insulina , Tejido Adiposo/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Humanos , Inflamación/metabolismo , Resistencia a la Insulina/genética , Material Particulado/efectos adversos , Material Particulado/metabolismo
16.
Trends Endocrinol Metab ; 33(11): 737-740, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36175280

RESUMEN

Exposure to particulate matter ≤2.5 µm in diameter (PM2.5) alters cardiometabolic homeostasis. The reduced oxidative capacity in brown adipocytes and the development of inflammation and insulin resistance in white adipose tissue (WAT) can account for the dysmetabolic setting on PM2.5 exposure. In this forum article, we discuss relevant evidence to highlight a causal connection between PM2.5-induced adipose tissue dysfunction and cardiometabolic disturbances.


Asunto(s)
Enfermedades Cardiovasculares , Resistencia a la Insulina , Humanos , Material Particulado/efectos adversos , Material Particulado/metabolismo , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo , Enfermedades Cardiovasculares/metabolismo
17.
Cells ; 11(21)2022 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-36359919

RESUMEN

Alzheimer's disease (AD) is an irreversible neurodegenerative disorder with a complex pathophysiology. Type 2 diabetes (T2D) is a strong risk factor for AD that shares similar abnormal features including metabolic dysregulation and brain pathology such as amyloid and/or Tau deposits. Emerging evidence suggests that circulating branched-chain amino acids (BCAAs) are associated with T2D. While excess BCAAs are shown to be harmful to neurons, its connection to AD is poorly understood. Here we show that individuals with AD have elevated circulating BCAAs and their metabolites compared to healthy individuals, and that a BCAA metabolite is correlated with the severity of dementia. APPSwe mouse model of AD also displayed higher plasma BCAAs compared to controls. In pursuit of understanding a potential causality, BCAA supplementation to HT-22 neurons was found to reduce genes critical for neuronal health while increasing phosphorylated Tau. Moreover, restricting BCAAs from diet delayed cognitive decline and lowered AD-related pathology in the cortex and hippocampus in APP/PS1 mice. BCAA restriction for two months was sufficient to correct glycemic control and increased/restored dopamine that were severely reduced in APP/PS1 controls. Treating 5xFAD mice that show early brain pathology with a BCAA-lowering compound recapitulated the beneficial effects of BCAA restriction on brain pathology and neurotransmitters including norepinephrine and serotonin. Collectively, this study reveals a positive association between circulating BCAAs and AD. Our findings suggest that BCAAs impair neuronal functions whereas BCAA-lowering alleviates AD-related pathology and cognitive decline, thus establishing a potential causal link between BCAAs and AD progression.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Diabetes Mellitus Tipo 2 , Ratones , Animales , Aminoácidos de Cadena Ramificada/metabolismo , Enfermedad de Alzheimer/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Cognición
18.
Am J Physiol Regul Integr Comp Physiol ; 300(6): R1266-77, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21411768

RESUMEN

Given the unabated obesity problem, there is increasing appreciation of expressions like "my eyes are bigger than my stomach," and recent studies in rodents and humans suggest that dysregulated brain reward pathways may be contributing not only to drug addiction but also to increased intake of palatable foods and ultimately obesity. After describing recent progress in revealing the neural pathways and mechanisms underlying food reward and the attribution of incentive salience by internal state signals, we analyze the potentially circular relationship between palatable food intake, hyperphagia, and obesity. Are there preexisting individual differences in reward functions at an early age, and could they be responsible for development of obesity later in life? Does repeated exposure to palatable foods set off a cascade of sensitization as in drug and alcohol addiction? Are reward functions altered by secondary effects of the obese state, such as increased signaling through inflammatory, oxidative, and mitochondrial stress pathways? Answering these questions will significantly impact prevention and treatment of obesity and its ensuing comorbidities as well as eating disorders and drug and alcohol addiction.


Asunto(s)
Hiperfagia/psicología , Obesidad/psicología , Recompensa , Animales , Conducta Adictiva/fisiopatología , Conducta Adictiva/psicología , Humanos , Hiperfagia/fisiopatología , Modelos Animales , Vías Nerviosas/fisiopatología , Obesidad/fisiopatología , Transducción de Señal/fisiología
19.
Am J Physiol Regul Integr Comp Physiol ; 301(5): R1267-80, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21849633

RESUMEN

Cross-sectional studies in both humans and animals have demonstrated associations between obesity and altered reward functions at the behavioral and neural level, but it is unclear whether these alterations are cause or consequence of the obese state. Reward behaviors were quantified in male, outbred Sprague-Dawley (SD) and selected line obesity-prone (OP) and obesity-resistant (OR) rats after induction of obesity by high-fat diet feeding and after subsequent loss of excess body weight by chronic calorie restriction. As measured by the brief access lick and taste-reactivity paradigms, both obese SD and OP rats "liked" low concentrations of sucrose and corn oil less, but "liked" the highest concentrations more, compared with lean rats, and this effect was fully reversed by weight loss in SD rats. Acute food deprivation was unable to change decreased responsiveness to low concentrations but eliminated increased responsiveness to high concentrations in obese SD rats, and leptin administration in weight-reduced SD rats shifted concentration-response curves toward that seen in the obese state in the brief access lick test. "Wanting" and reinforcement learning as assessed in the incentive runway and progressive ratio lever-pressing paradigms was paradoxically decreased in both obese (compared with lean SD rats) and OP (compared with OR rats). Thus, reversible, obesity-associated, reduced "liking" and "wanting" of low-calorie foods in SD rats suggest a role for secondary effects of the obese state on reward functions, while similar differences between select lines of OP and OR rats before induction of obesity indicate a genetic component.


Asunto(s)
Aceite de Maíz , Conducta Alimentaria , Preferencias Alimentarias , Leptina/metabolismo , Obesidad/psicología , Sacarosa , Gusto/genética , Pérdida de Peso , Adiposidad/genética , Animales , Restricción Calórica , Grasas de la Dieta , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Masculino , Motivación , Obesidad/etiología , Obesidad/genética , Obesidad/metabolismo , Obesidad/fisiopatología , Fenotipo , Ratas , Ratas Sprague-Dawley , Refuerzo en Psicología , Recompensa
20.
Am J Physiol Regul Integr Comp Physiol ; 300(3): R693-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21178126

RESUMEN

Estrogens are known to cause hyperprolactinemia, most probably by acting on the tuberoinfundibular dopaminergic (TIDA) system of the hypothalamus. Dopamine (DA) produced by TIDA neurons directly inhibits prolactin secretion and, therefore, to stimulate prolactin secretion, estrogens inhibit TIDA neurons to decrease DA production. However, the mechanism by which estrogen produces this effect is not clear. In the present study, we used a paradigm involving chronic exposure to low levels of estradiol-17ß (E(2)) to mimic prolonged exposures to environmental and endogenous estrogens. We hypothesized that chronic exposure to low levels of E(2) induces oxidative stress in the arcuate nucleus (AN) of the hypothalamus that contains TIDA neurons and causes nitration of tyrosine hydroxylase (TH), the rate-limiting enzyme in the synthesis of DA. This results in a significant decrease in DA and consequently, hyperprolactinemia. To investigate this, adult, intact female cycling rats were implanted with slow-release E(2) pellets (20 ng/day) for 30, 60, or 90 days and were compared with old (16-18 mo old) constant estrous (OCE) rats. Chronic E(2) exposure significantly increased the expression of glial fibrillary acidic protein and the concentrations of interleukin-1ß (IL-1ß) and nitrate in the AN that contains perikarya of TIDA neurons and increased nitration of TH in the median eminence (ME) that contains the terminals. These levels were comparable to those seen in OCE rats. We observed a significant decrease in DA concentrations in the ME and hyperprolactinemia in an exposure-dependent manner similar to that seen in OCE rats. It was concluded that chronic exposure to low levels of E(2) evokes oxidative stress in the AN to inhibit TIDA neuronal function, most probably leading to hyperprolactinemia.


Asunto(s)
Dopamina/metabolismo , Estradiol/toxicidad , Hiperprolactinemia/inducido químicamente , Hipotálamo/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Prolactina/sangre , Factores de Edad , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Regulación hacia Abajo , Implantes de Medicamentos , Estradiol/administración & dosificación , Estradiol/sangre , Estro , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Hiperprolactinemia/metabolismo , Hipotálamo/metabolismo , Interleucina-1beta/metabolismo , Óxido Nítrico/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Tirosina 3-Monooxigenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA