Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 274
Filtrar
Más filtros

Intervalo de año de publicación
1.
Am J Physiol Endocrinol Metab ; 326(2): E134-E147, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-38117265

RESUMEN

Vertical sleeve gastrectomy (VSG) restores glucose homeostasis in obese mice and humans. In addition, the increased fibroblast growth factor (FGF)15/19 circulating level postsurgery has been implicated in this effect. However, the impact of FGF15/19 on pancreatic islets remains unclear. Using a diet-induced obese mice model, we demonstrate that VSG attenuates insulin hypersecretion in isolated pancreatic islets, likely due to morphological alterations in the endocrine pancreas such as reduction in islet, ß-cell, and α-cell mass. In addition, VSG relieves gene expression of endoplasmic reticulum (ER) stress and inflammation markers in islets from obese mice. Incubation of INS-1E ß-cells with serum from obese mice induced dysfunction and cell death, whereas these conditions were not induced with serum from obese mice submitted to VSG, implicating the involvement of a humoral factor. Indeed, VSG increased FGF15 circulating levels in obese mice, as well as the expression of FGF receptor 1 (Fgfr1) and its coreceptor ß-klotho (Klb), both in pancreatic islets from VSG mice and in INS-1E cells treated with the serum from these mice. Moreover, exposing INS-1E cells to an FGFR inhibitor abolished the effects of VSG serum on insulin secretion and cell death. Also, recombinant FGF19 prevents INS-1E cells from dysfunction and death induced by serum from obese mice. These findings indicate that the amelioration of glucose-insulin homeostasis promoted by VSG is mediated, at least in part, by FGF15/19. Therefore, approaches promoting FGF15/19 release or action may restore pancreatic islet function in obesity.NEW & NOTEWORTHY Vertical sleeve gastrectomy (VSG) decreases insulin secretion, endoplasmic reticulum (ER) stress, and inflammation in pancreatic islets from obese mice. In addition, VSG increased fibroblast growth factor (FGF)15 circulating levels in obese mice, as well as the expression of FGF receptor 1 (Fgfr1) and its coreceptor ß-klotho (Klb), both in pancreatic islets from VSG mice and in INS-1E ß-cells treated with the serum from these mice. Serum from operated mice protects INS-1E cells from dysfunction and apoptosis, which was mediated by FGF15/19.


Asunto(s)
Células Secretoras de Insulina , Insulina , Ratones , Humanos , Animales , Insulina/metabolismo , Ratones Obesos , Células Secretoras de Insulina/metabolismo , Glucosa/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Gastrectomía , Inflamación/metabolismo , Homeostasis
2.
Am J Physiol Endocrinol Metab ; 326(5): E681-E695, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38597829

RESUMEN

Hypothalamic proopiomelanocortin (POMC) neurons are sensors of signals that reflect the energy stored in the body. Inducing mild stress in proopiomelanocortin neurons protects them from the damage promoted by the consumption of a high-fat diet, mitigating the development of obesity; however, the cellular mechanisms behind these effects are unknown. Here, we induced mild stress in a proopiomelanocortin neuron cell line by inhibiting Crif1. In proopiomelanocortin neurons exposed to high levels of palmitate, the partial inhibition of Crif1 reverted the defects in mitochondrial respiration and ATP production; this was accompanied by improved mitochondrial fusion/fission cycling. Furthermore, the partial inhibition of Crif1 resulted in increased reactive oxygen species production, increased fatty acid oxidation, and reduced dependency on glucose for mitochondrial respiration. These changes were dependent on the activity of CPT-1. Thus, we identified a CPT-1-dependent metabolic shift toward greater utilization of fatty acids as substrates for respiration as the mechanism behind the protective effect of mild stress against palmitate-induced damage of proopiomelanocortin neurons.NEW & NOTEWORTHY Saturated fats can damage hypothalamic neurons resulting in positive energy balance, and this is mitigated by mild cellular stress; however, the mechanisms behind this protective effect are unknown. Using a proopiomelanocortin cell line, we show that under exposure to a high concentration of palmitate, the partial inhibition of the mitochondrial protein Crif1 results in protection due to a metabolic shift warranted by the increased expression and activity of the mitochondrial fatty acid transporter CPT-1.


Asunto(s)
Carnitina O-Palmitoiltransferasa , Proteínas de Ciclo Celular , Ácidos Grasos , Mitocondrias , Animales , Ratones , Carnitina O-Palmitoiltransferasa/metabolismo , Carnitina O-Palmitoiltransferasa/genética , Línea Celular , Ácidos Grasos/metabolismo , Hipotálamo/metabolismo , Hipotálamo/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Proopiomelanocortina/genética , Especies Reactivas de Oxígeno/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo
3.
Am J Physiol Endocrinol Metab ; 324(2): E154-E166, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36598900

RESUMEN

Maternal obesity is an important risk factor for obesity, cardiovascular, and metabolic diseases in the offspring. Studies have shown that it leads to hypothalamic inflammation in the progeny, affecting the function of neurons regulating food intake and energy expenditure. In adult mice fed a high-fat diet, one of the hypothalamic abnormalities that contribute to the development of obesity is the damage of the blood-brain barrier (BBB) at the median eminence-arcuate nucleus (ME-ARC) interface; however, how the hypothalamic BBB is affected in the offspring of obese mothers requires further investigation. Here, we used confocal and transmission electron microscopy, transcript expression analysis, glucose tolerance testing, and a cross-fostering intervention to determine the impact of maternal obesity and breastfeeding on BBB integrity at the ME-ARC interface. The offspring of obese mothers were born smaller; conversely, at weaning, they presented larger body mass and glucose intolerance. In addition, maternal obesity-induced structural and functional damage of the offspring's ME-ARC BBB. By a cross-fostering intervention, some of the defects in barrier integrity and metabolism seen during development in an obesogenic diet were recovered. The offspring of obese dams breastfed by lean dams presented a reduction of body mass and glucose intolerance as compared to the offspring continuously exposed to an obesogenic environment during intrauterine and perinatal life; this was accompanied by partial recovery of the anatomical structure of the ME-ARC interface, and by the normalization of transcript expression of genes coding for hypothalamic neurotransmitters involved in energy balance and BBB integrity. Thus, maternal obesity promotes structural and functional damage of the hypothalamic BBB, which is, in part, reverted by lactation by lean mothers.NEW & NOTEWORTHY Maternal dietary habits directly influence offspring health. In this study, we aimed at determining the impact of maternal obesity on BBB integrity. We show that DIO offspring presented a leakier ME-BBB, accompanied by changes in the expression of transcripts encoding for endothelial and tanycytic proteins, as well as of hypothalamic neuropeptides. Breastfeeding in lean dams was sufficient to protect the offspring from ME-BBB disruption, providing a preventive strategy of nutritional intervention during early life.


Asunto(s)
Intolerancia a la Glucosa , Obesidad Materna , Humanos , Femenino , Animales , Ratones , Embarazo , Barrera Hematoencefálica/metabolismo , Eminencia Media/metabolismo , Obesidad Materna/metabolismo , Madres , Intolerancia a la Glucosa/metabolismo , Obesidad/metabolismo , Hipotálamo/metabolismo , Dieta Alta en Grasa/efectos adversos , Fenómenos Fisiologicos Nutricionales Maternos
4.
Am J Physiol Endocrinol Metab ; 324(3): E226-E240, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36724126

RESUMEN

Obesity is one of the leading noncommunicable diseases in the world. Despite intense efforts to develop strategies to prevent and treat obesity, its prevalence continues to rise worldwide. A recent study has shown that the tricarboxylic acid intermediate succinate increases body energy expenditure by promoting brown adipose tissue thermogenesis through the activation of uncoupling protein-1; this has generated interest surrounding its potential usefulness as an approach to treat obesity. It is currently unknown how succinate impacts brown adipose tissue protein expression, and how exogenous succinate impacts body mass reduction promoted by a drug approved to treat human obesity, the glucagon-like-1 receptor agonist, liraglutide. In the first part of this study, we used bottom-up shotgun proteomics to determine the acute impact of exogenous succinate on the brown adipose tissue. We show that succinate rapidly affects the expression of 177 brown adipose tissue proteins, which are mostly associated with mitochondrial structure and function. In the second part of this study, we performed a short-term preclinical pharmacological intervention, treating diet-induced obese mice with a combination of exogenous succinate and liraglutide. We show that the combination was more efficient than liraglutide alone in promoting body mass reduction, food energy efficiency reduction, food intake reduction, and an increase in body temperature. Using serum metabolomics analysis, we showed that succinate, but not liraglutide, promoted a significant increase in the blood levels of several medium and long-chain fatty acids. In conclusion, exogenous succinate promotes rapid changes in brown adipose tissue mitochondrial proteins, and when used in association with liraglutide, increases body mass reduction.NEW & NOTEWORTHY Exogenous succinate induces major changes in brown adipose tissue protein expression affecting particularly mitochondrial respiration and structural proteins. When given exogenously in drinking water, succinate mitigates body mass gain in a rodent model of diet-induced obesity; in addition, when given in association with the glucagon-like peptide-1 receptor agonist, liraglutide, succinate increases body mass reduction promoted by liraglutide alone.


Asunto(s)
Tejido Adiposo Pardo , Liraglutida , Animales , Ratones , Tejido Adiposo Pardo/metabolismo , Metabolismo Energético , Liraglutida/farmacología , Liraglutida/uso terapéutico , Obesidad/metabolismo , Proteoma/metabolismo , Ácido Succínico/farmacología , Ácido Succínico/metabolismo , Ácido Succínico/uso terapéutico , Termogénesis , Proteína Desacopladora 1/metabolismo
5.
Mol Genet Metab ; 138(4): 107552, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36889041

RESUMEN

BACKGROUND AND AIMS: Low-density lipoprotein (LDL) plasma concentration decline is a biomarker for acute inflammatory diseases, including coronavirus disease-2019 (COVID-19). Phenotypic changes in LDL during COVID-19 may be equally related to adverse clinical outcomes. METHODS: Individuals hospitalized due to COVID-19 (n = 40) were enrolled. Blood samples were collected on days 0, 2, 4, 6, and 30 (D0, D2, D4, D6, and D30). Oxidized LDL (ox-LDL), and lipoprotein-associated phospholipase A2 (Lp-PLA2) activity were measured. In a consecutive series of cases (n = 13), LDL was isolated by gradient ultracentrifugation from D0 and D6 and was quantified by lipidomic analysis. Association between clinical outcomes and LDL phenotypic changes was investigated. RESULTS: In the first 30 days, 42.5% of participants died due to Covid-19. The serum ox-LDL increased from D0 to D6 (p < 0.005) and decreased at D30. Moreover, individuals who had an ox-LDL increase from D0 to D6 to over the 90th percentile died. The plasma Lp-PLA2 activity also increased progressively from D0 to D30 (p < 0.005), and the change from D0 to D6 in Lp-PLA2 and ox-LDL were positively correlated (r = 0.65, p < 0.0001). An exploratory untargeted lipidomic analysis uncovered 308 individual lipids in isolated LDL particles. Paired-test analysis from D0 and D6 revealed higher concentrations of 32 lipid species during disease progression, mainly represented by lysophosphatidyl choline and phosphatidylinositol. In addition, 69 lipid species were exclusively modulated in the LDL particles from non-survivors as compared to survivors. CONCLUSIONS: Phenotypic changes in LDL particles are associated with disease progression and adverse clinical outcomes in COVID-19 patients and could serve as a potential prognostic biomarker.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa , COVID-19 , Humanos , Lipoproteínas LDL , Biomarcadores , Lisofosfatidilcolinas
6.
Eur J Nucl Med Mol Imaging ; 50(8): 2432-2440, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36988710

RESUMEN

PURPOSE: The aim of this study was to compare [18F]FDG and [68Ga]Ga-PSMA-11 PET/CT image findings in patients with multiple myeloma (MM). METHODS: Twenty consecutive patients with symptomatic biopsy-proven MM were submitted to whole body [18F]FDG and [68Ga]Ga-PSMA-11 PET/CT with a time interval of 1-8 days between procedures. All lesions were counted and had their maximum SUV (SUVmax) measured. Intra-class correlation (ICC) was used to assess the agreement between [18F]FDG and [68Ga]Ga-PSMA-11 PET/CT findings. RESULTS: A total of 266 lesions were detected in 19/20 patients. [18F]FDG detected 223/266 (84%) lesions in 17 patients and [68Ga]Ga-PSMA-11 190/266 (71%) lesions in 19 patients. Both procedures did not identify any active lesion in 1 patient. Forty-three (16%) lesions were detected only by [68Ga]Ga-PSMA-11 and 76 (29%) only by [18F]FDG. Both tracers identified 147 (55%) lesions. Intralesional mismatch of FDG-PSMA uptake was identified in 25 of these 147 lesions, found in 8 different patients. Different lesions with uptake of only [18F]FDG or [68Ga]Ga-PSMA-11 in the same patient were found in 4 patients. The highest SUVmax of [18F]FDG and [68Ga]Ga-PSMA-11 had a median (min-max) SUVmax of 6.5 (2.0-37.8) and 5.5 (1.7-51.3), respectively. [18F]FDG and [68Ga]Ga-PSMA-11 respectively identified 18 and 19 soft tissue lesions. False-positive [18F]FDG findings had minimal or no uptake of [68Ga]Ga-PSMA-11. Good reliability (ICC ≥ 0.75) was found for number of lesions, number of soft tissue lesions and highest SUVmax in each patient. CONCLUSION: [18F]FDG or [68Ga]Ga-PSMA-11 alone can detect most MM lesions. Almost half of the lesions take up only one of the tracers, reflecting increased glycolysis or angiogenesis in specific lesions, and suggesting their possible complementary role in MM. The marked [68Ga]Ga-PSMA-11 uptake in some cases raises the possibility of a theranostic approach in selected patients.


Asunto(s)
Radioisótopos de Galio , Mieloma Múltiple , Humanos , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Fluorodesoxiglucosa F18 , Mieloma Múltiple/diagnóstico por imagen , Reproducibilidad de los Resultados
7.
Proc Natl Acad Sci U S A ; 117(38): 23932-23941, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32900951

RESUMEN

DICER is a key enzyme in microRNA (miRNA) biogenesis. Here we show that aerobic exercise training up-regulates DICER in adipose tissue of mice and humans. This can be mimicked by infusion of serum from exercised mice into sedentary mice and depends on AMPK-mediated signaling in both muscle and adipocytes. Adipocyte DICER is required for whole-body metabolic adaptations to aerobic exercise training, in part, by allowing controlled substrate utilization in adipose tissue, which, in turn, supports skeletal muscle function. Exercise training increases overall miRNA expression in adipose tissue, and up-regulation of miR-203-3p limits glycolysis in adipose under conditions of metabolic stress. We propose that exercise training-induced DICER-miR-203-3p up-regulation in adipocytes is a key adaptive response that coordinates signals from working muscle to promote whole-body metabolic adaptations.


Asunto(s)
Tejido Adiposo/metabolismo , ARN Helicasas DEAD-box/metabolismo , Ejercicio Físico/fisiología , Ribonucleasa III/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Adaptación Fisiológica/fisiología , Adipocitos/metabolismo , Animales , Células Cultivadas , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , Femenino , Glucólisis , Humanos , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Condicionamiento Físico Animal , Ribonucleasa III/deficiencia , Ribonucleasa III/genética
8.
J Neurosci ; 41(48): 10004-10022, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34675088

RESUMEN

Nescient helix-loop-helix 2 (NHLH2) is a hypothalamic transcription factor that controls the expression of prohormone convertase 1/3, therefore having an impact on the processing of proopiomelanocortin and thus on energy homeostasis. Studies have shown that KO of Nhlh2 results in increased body mass, reduced physical activity, and hypogonadism. In humans, a polymorphism of the NHLH2 gene is associated with obesity; and in Prader-Willi syndrome, a condition characterized by obesity, hypogonadism and behavioral abnormalities, the expression of NHLH2 is reduced. Despite clinical and experimental evidence suggesting that NHLH2 could be a good target for the treatment of obesity, no previous study has evaluated the impact of NHLH2 overexpression in obesity. Here, in mice fed a high-fat diet introduced right after the arcuate nucleus intracerebroventricular injection of a lentivirus that promoted 40% increase in NHLH2, there was prevention of the development of obesity by a mechanism dependent on the reduction of caloric intake. When hypothalamic overexpression of NHLH2 was induced in previously obese mice, the beneficial impact on obesity-associated phenotype was even greater; thus, there was an 80% attenuation in body mass gain, reduced whole-body adiposity, increased brown adipose tissue temperature, reduced hypothalamic inflammation, and reduced liver steatosis. In this setting, the beneficial impact of hypothalamic overexpression of NHLH2 was a result of combined effects on caloric intake, energy expenditure, and physical activity. Moreover, the hypothalamic overexpression of NHLH2 reduced obesity-associated anxiety/depression behavior. Thus, we provide an experimental proof of concept supporting that hypothalamic NHLH2 is a good target for the treatment of obesity.SIGNIFICANCE STATEMENT Obesity is a highly prevalent medical condition that lacks an effective treatment. The main advance provided by this study is the demonstration of the beneficial metabolic and behavioral outcomes resulting from the overexpression of NHLH2 in the hypothalamus. When NHLH2 was overexpressed simultaneously with the introduction of a high-fat diet, there was prevention of obesity by a mechanism dependent on reduced caloric intake. Conversely, when NHLH2 was overexpressed in previously obese mice, there was reduction of the obese phenotype because of a combination of reduced caloric intake, increased physical activity, and increased thermogenesis. In addition, the overexpression of NHLH2 reduced anxiety/depression-like behavior. Thus, NHLH2 emerges as a potential target for the combined treatment of obesity and its associated anxiety/depression-like behavior.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Obesidad/metabolismo , Animales , Ansiedad/metabolismo , Índice de Masa Corporal , Depresión/metabolismo , Dieta Alta en Grasa/efectos adversos , Femenino , Masculino , Ratones , Obesidad/psicología
9.
J Neuroinflammation ; 19(1): 136, 2022 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-35681242

RESUMEN

Brain macrophages and microglia are centrally involved in immune surveillance of the central nervous system. Upon inflammatory stimuli, they become reactive and release key molecules to prevent further damage to the neuronal network. In the hypothalamic area, perivascular macrophages (PVMs) are the first line of host defence against pathogenic organisms, particles and/or substances from the blood. They are distributed throughout the circumventricular organ median eminence, wrapping endothelial cells from fenestrated portal capillaries and in the hypothalamic vascular network, where they are localised in the perivascular space of the blood-brain barrier (BBB). Some studies have indicated that PVMs from the hypothalamus increase the expression of inducible nitric oxide synthase and vascular endothelial growth factor upon feeding for a long time on a high-fat diet. This adaptive response contributes to the impairment of glucose uptake, facilitates BBB leakage and leads to increased lipid and inflammatory cell influx towards the hypothalamic parenchyma. Despite these early findings, there is still a lack of studies exploring the mechanisms by which PVMs contribute to the development of obesity-related hypothalamic dysfunction, particularly at the early stages when there is chemotaxis of peripheral myeloid cells into the mediobasal hypothalamus. Here, we reviewed the studies involving the ontogeny, hallmarks and main features of brain PVMs in vascular homeostasis, inflammation and neuroendocrine control. This review provides a framework for understanding the potential involvement of PVMs in diet-induced hypothalamic inflammation.


Asunto(s)
Dieta Alta en Grasa , Células Endoteliales , Dieta Alta en Grasa/efectos adversos , Células Endoteliales/metabolismo , Humanos , Hipotálamo/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Int J Neurosci ; : 1-13, 2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35659180

RESUMEN

INTRODUCTION: Obesity presents with structural and functional hypothalamic dysfunction. However, it is unclear whether weight loss can lead to hypothalamic changes. We therefore aimed to conduct a systematic review and meta-analysis to determine the effect of body mass reduction in obese individuals on hypothalamic structure and function. METHODS: PubMed, Embase and Cochrane databases were searched for studies that reported the change in hypothalamic structure and function after weight loss. Qualitative and quantitative analyses were performed on magnetic resonance imaging techniques, medio-basal hypothalamus T2-relaxation time, blood oxygen level dependent (BOLD) contrast, voxel-based morphometry (VBM) and biomarkers including glucose, insulin, leptin, ghrelin and inflammatory markers of interleukins. Mean differences between pre- and post-weight loss and 95% confidence intervals (CIs) were pooled using random-effects models. RESULTS: Thirteen pre-post studies were included, of which six accounted for the meta-analysis. Studies showed a favorable decrease in T2-relaxation time (n = 1), favorable change in hypothalamic activity after weight loss on BOLD contrast (n = 4), with higher peak activities after surgical weight loss (n = 2). No differences were found in the gray matter density of the hypothalamus on VBM (n = 1). Pooled mean differences between pre- and post-surgical weight loss revealed a decrease of 8.53 mg/dl (95% CI: 5.17, 11.9) in glucose, 7.73 pmol/l (95% CI: 5.07, 10.4) in insulin, 15.5 ng/ml (95% CI: 9.40, 21.6) in leptin, 142.9 pg/ml (95% CI: 79.0, 206.8) in ghrelin and 9.43 pg/ml (95% CI: -6.89, 25.7) in IL-6 level. CONCLUSIONS: Our study showed weight reduction in obesity led to limited structural change and significant functional changes in the hypothalamus.

11.
Am J Physiol Endocrinol Metab ; 320(2): E326-E332, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33284086

RESUMEN

Interoceptive signals from gut and adipose tissue and sensory cues from the environment are integrated by hubs in the brain to regulate feeding behavior and maintain homeostatic control of body weight. In vivo neural recordings have revealed that these signals control the activity of multiple layers of hunger neurons and eating is not only the result of feedback correction to a set point, but can also be under the influence of anticipatory regulations. A series of recent technical developments have revealed how peripheral and sensory signals, in particular, from the gut are conveyed to the brain to integrate neural circuits. Here, we describe the mechanisms involved in gastrointestinal stimulation by nutrients and how these signals act on the hindbrain to generate motivated behaviors. We also consider the organization of multidirectional intra- and extrahypothalamic circuits and how this has created a framework for understanding neural control of feeding.


Asunto(s)
Regulación del Apetito , Encéfalo/fisiología , Tracto Gastrointestinal/fisiología , Animales , Peso Corporal/fisiología , Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Microbioma Gastrointestinal , Homeostasis/fisiología , Humanos , Hambre/fisiología , Transducción de Señal/fisiología
12.
J Neuroinflammation ; 18(1): 192, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34465367

RESUMEN

BACKGROUND: Interleukin-6 (IL6) produced in the context of exercise acts in the hypothalamus reducing obesity-associated inflammation and restoring the control of food intake and energy expenditure. In the hippocampus, some of the beneficial actions of IL6 are attributed to its neurogenesis-inducing properties. However, in the hypothalamus, the putative neurogenic actions of IL6 have never been explored, and its potential to balance energy intake can be an approach to prevent or attenuate obesity. METHODS: Wild-type (WT) and IL6 knockout (KO) mice were employed to study the capacity of IL6 to induce neurogenesis. We used cell labeling with Bromodeoxyuridine (BrdU), immunofluorescence, and real-time PCR to determine the expression of markers of neurogenesis and neurotransmitters. We prepared hypothalamic neuroprogenitor cells from KO that were treated with IL6 in order to provide an ex vivo model to further characterizing the neurogenic actions of IL6 through differentiation assays. In addition, we analyzed single-cell RNA sequencing data and determined the expression of IL6 and IL6 receptor in specific cell types of the murine hypothalamus. RESULTS: IL6 expression in the hypothalamus is low and restricted to microglia and tanycytes, whereas IL6 receptor is expressed in microglia, ependymocytes, endothelial cells, and astrocytes. Exogenous IL6 reduces diet-induced obesity. In outbred mice, obesity-resistance is accompanied by increased expression of IL6 in the hypothalamus. IL6 induces neurogenesis-related gene expression in the hypothalamus and in neuroprogenitor cells, both from WT as well as from KO mice. CONCLUSION: IL6 induces neurogenesis-related gene expression in the hypothalamus of WT mice. In KO mice, the neurogenic actions of IL6 are preserved; however, the appearance of new fully differentiated proopiomelanocortin (POMC) and neuropeptide Y (NPY) neurons is either delayed or disturbed.


Asunto(s)
Hipotálamo/metabolismo , Interleucina-6/genética , Neurogénesis/genética , Neuronas/metabolismo , Obesidad/genética , Animales , Metabolismo Energético/fisiología , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/metabolismo , Hipotálamo/efectos de los fármacos , Interleucina-6/metabolismo , Interleucina-6/farmacología , Masculino , Ratones , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/metabolismo , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Obesidad/metabolismo , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo
13.
Int J Obes (Lond) ; 44(1): 178-185, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31201362

RESUMEN

BACKGROUND/OBJECTIVES: Hypothalamic neurons play a major role in the control of body mass. Obese subjects present radiologic signs of gliosis in the hypothalamus, which may reflect the damage or loss of neurons involved in whole-body energy homeostasis. It is currently unknown if hypothalamic gliosis (1) differs between obese nondiabetic (ND) and obese diabetic subjects (T2D) or (2) is modified by extensive body mass reduction via Roux-n-Y gastric bypass (RYGB). SUBJECTS/METHODS: Fifty-five subjects (all female) including lean controls (CT; n = 13), ND (n = 28), and T2D (n = 14) completed at least one study visit. Subjects underwent anthropometrics and a multi-echo MRI sequence to measure mean bilateral T2 relaxation time in the mediobasal hypothalamus (MBH) and two reference regions (amygdala and putamen). The obese groups underwent RYGB and were re-evaluated 9 months later. Analyses were by linear mixed models. RESULTS: Analyses of T2 relaxation time at baseline showed a group by region interaction only in the MBH (P < 0.0001). T2D had longer T2 relaxation times compared to either CT or ND groups. To examine the effects of RYGB on hypothalamic gliosis a three-way (group by region by time) mixed effects model adjusted for age was executed. Group by region (P < 0.0001) and region by time (P = 0.0005) interactions were significant. There was a reduction in MBH relaxation time by RYGB, and, although the T2D group still had higher T2 relaxation time overall compared to the ND group, the T2D group had significantly lower T2 relaxation time after surgery and the ND group showed a trend. The degree of reduction in MBH T2 relaxation time by RYGB was unrelated to clinical outcomes. CONCLUSION: T2 relaxation times, a marker of hypothalamic gliosis, are higher in obese women with T2D and are reduced by RYGB-induced weight loss.


Asunto(s)
Cirugía Bariátrica , Diabetes Mellitus Tipo 2/complicaciones , Gliosis , Hipotálamo , Obesidad , Femenino , Gliosis/diagnóstico por imagen , Gliosis/patología , Humanos , Hipotálamo/diagnóstico por imagen , Hipotálamo/patología , Imagen por Resonancia Magnética , Obesidad/complicaciones , Obesidad/cirugía , Resultado del Tratamiento
14.
Brain Behav Immun ; 87: 272-285, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31863824

RESUMEN

Interleukin-17 (IL-17) is expressed in the intestine in response to changes in the gut microbiome landscape and plays an important role in intestinal and systemic inflammatory diseases. There is evidence that dietary factors can also modify the expression of intestinal IL-17. Here, we hypothesized that, similar to several other gut-produced factors, IL-17 may act in the hypothalamus to modulate food intake. We confirm that food intake increases IL-17 expression in the mouse ileum and human blood. There is no expression of IL-17 in the hypothalamus; however, IL-17 receptor A is expressed in both pro-opiomelanocortin (POMC) and agouti-related peptide (AgRP) neurons. Upon systemic injection, IL-17 promoted a rapid increase in hypothalamic POMC expression, which was followed by a late increase in the expression of AgRP. Both systemic and intracerebroventricular injections of IL-17 reduced calorie intake without affecting whole-body energy expenditure. Systemic but not intracerebroventricular injection of IL-17 increase brown adipose tissue temperature. Thus, IL-17 is a gut-produced factor that is controlled by diet and modulates food intake by acting in the hypothalamus. Our findings provide the first evidence of a cytokine that is acutely regulated by food intake and plays a role in the regulation of eating.


Asunto(s)
Hipotálamo , Interleucina-17 , Proteína Relacionada con Agouti/metabolismo , Animales , Ingestión de Alimentos , Humanos , Hipotálamo/metabolismo , Ratones , Proopiomelanocortina/metabolismo
15.
Neuroendocrinology ; 110(1-2): 92-104, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31104058

RESUMEN

In outbred mice, susceptibility or resistance to diet-induced obesity is associated with rapid changes in hypothalamic proopiomelanocortin (POMC) levels. Here, we evaluated 3 hypotheses that potentially explain the development of the different obesity phenotypes in outbred Swiss mice. First, rapid and differential changes in the gut microbiota in obesity-prone (OP) and obesity-resistant (OR) mice fed on a high-fat diet (HFD) might cause differential efficiencies in fatty acid harvesting leading to changes in systemic fatty acid concentrations that in turn affect POMC expression and processing. Second, independently of the gut microbiota, OP mice might have increased blood fatty acid levels after the introduction of a HFD, which could affect POMC expression and processing. Third, fatty acids might act directly in the hypothalamus to differentially regulate POMC expression and/or processing in OP and OR mice. We evaluated OP and OR male Swiss mice using 16S rRNA sequencing for the determination of gut microbiota; gas chromatography for blood lipid determination; and immunoblot and real-time polymerase chain reaction for protein and transcript determination and indirect calorimetry. Some experiments were performed with human pluripotent stem cells differentiated into hypothalamic neurons. We did not find evidence supporting the first 2 hypotheses. However, we found that in OP but not in OR mice, palmitate induces a rapid increase in hypothalamic POMC, which is followed by increased expression of proprotein convertase subtilisin/kexin type 1 PC1/3. Lentiviral inhibition of hypothalamic PC1/3 increased caloric intake and body mass in both OP and OR mice. In human stem cell-derived hypothalamic cells, we found that palmitate potently suppressed the production of POMC-derived peptides. Palmitate directly regulates PC1/3 in OP mice and likely has a functional impact on POMC processing.


Asunto(s)
Microbioma Gastrointestinal , Hipotálamo/metabolismo , Inflamación/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Palmitatos/farmacología , Proopiomelanocortina/metabolismo , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Humanos , Ácido Linoleico/farmacología , Masculino , Ratones , Obesidad/sangre , Obesidad/etiología , Células Madre Pluripotentes , ARN Ribosómico 16S
16.
J Cell Biochem ; 120(10): 18186-18192, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31144370

RESUMEN

Obesity and aging lead to abnormal transforming growth factor-ß1 (TGF-ß1) signaling in the hypothalamus, triggering the imbalance on glucose metabolism and energy homeostasis. Here, we determine the effect of acute exercise on TGF-ß1 expression in the hypothalamus of two models of obesity in mice. The bioinformatics analysis was performed to evaluate the correlation between hypothalamic Tgf-ß1 messenger RNA (mRNA) and genes related to thermogenesis in the brown adipose tissue (BAT) by using a large panel of isogenic BXD mice. Thereafter, leptin-deficient (ob/ob) mice and obese C57BL/6 mice fed on a high-fat diet (HFD) were submitted to the acute exercise protocol. Transcriptomic analysis by using BXD mouse reference population database revealed that hypothalamic Tgf-ß1 mRNA is negatively correlated with genes related to thermogenesis in brown adipose tissue of BXD mice, such as peroxisome proliferator-activated receptor gamma coactivator and is positively correlated with respiratory exchange ratio. In agreement with these results, leptin-deficient (ob/ob) and HFD-fed mice displayed high levels of Tgf-ß1 mRNA in the hypothalamus and reduction of Pgc1α mRNA in BAT. Interestingly, an acute exercise session reduced TGF-ß1 expression in the hypothalamus, increased Pgc1α mRNA in the BAT and reduced food consumption in obese mice. Our results demonstrated that acute physical exercise suppressed hypothalamic TGF-ß1 expression, increasing Pgc1α mRNA in BAT in obese mice.


Asunto(s)
Regulación hacia Abajo , Hipotálamo/metabolismo , Obesidad/genética , Condicionamiento Físico Animal/fisiología , Factor de Crecimiento Transformador beta1/genética , Tejido Adiposo Pardo/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/genética , Perfilación de la Expresión Génica/métodos , Leptina/deficiencia , Leptina/genética , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/etiología , Obesidad/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Termogénesis/genética , Factor de Crecimiento Transformador beta1/metabolismo
17.
Int J Obes (Lond) ; 43(12): 2361-2369, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31548571

RESUMEN

The characterization of the hypothalamic neuronal network, that controls food intake and energy expenditure, has provided great advances in the understanding of the pathophysiology of obesity. Most of the advances in this field were obtained thanks to the development of a number of genetic and nongenetic animal models that, at least in part, overtook the anatomical constraints that impair the study of the human hypothalamus. Despite the undisputed differences between human and rodent physiology, most seminal studies undertaken in rodents that have unveiled details of the neural regulation of energy homeostasis were eventually confirmed in humans; thus, placing experimental studies in the forefront of obesity research. During the last 15 years, researchers have provided extensive experimental proof that supports the existence of hypothalamic dysfunction, which leads to a progressive whole-body positive energy balance, and thus, to obesity. Here, we review the experimental work that unveiled the mechanisms behind hypothalamic dysfunction in obesity.


Asunto(s)
Hipotálamo , Neuronas/fisiología , Obesidad/fisiopatología , Animales , Modelos Animales de Enfermedad , Humanos , Hipotálamo/citología , Hipotálamo/fisiopatología , Inflamación/fisiopatología , Ratones
18.
Brain Behav Immun ; 78: 78-90, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30660601

RESUMEN

Obesity-associated hypothalamic inflammation plays an important role in the development of defective neuronal control of whole body energy balance. Because dietary fats are the main triggers of hypothalamic inflammation, we hypothesized that CD1, a lipid-presenting protein, may be involved in the hypothalamic inflammatory response in obesity. Here, we show that early after the introduction of a high-fat diet, CD1 expressing cells gradually appear in the mediobasal hypothalamus. The inhibition of hypothalamic CD1 reduces diet-induced hypothalamic inflammation and rescues the obese and glucose-intolerance phenotype of mice fed a high-fat diet. Conversely, the chemical activation of hypothalamic CD1 further increases diet-induced obesity and hypothalamic inflammation. A bioinformatics analysis revealed that hypothalamic CD1 correlates with transcripts encoding for proteins known to be involved in diet-induced hypothalamic abnormalities in obesity. Thus, CD1 is involved in at least part of the hypothalamic inflammatory response in diet-induced obesity and its modulation affects the body mass phenotype of mice.


Asunto(s)
Antígenos CD1/metabolismo , Hipotálamo/inmunología , Obesidad/metabolismo , Animales , Antígenos CD1/inmunología , Biología Computacional/métodos , Dieta Alta en Grasa , Grasas de la Dieta , Metabolismo Energético , Inflamación/metabolismo , Linfocitos/metabolismo , Masculino , Ratones , Obesidad/inmunología
19.
Aesthet Surg J ; 39(6): NP202-NP212, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-30265289

RESUMEN

BACKGROUND: Laboratory reports on adipose tissue suggest that fat grafting to the breast may pose an oncologic risk. One possible reason for this is the theoretic chronic inflammation due to adipokynes released by grafted white adipose tissue (WAT). OBJECTIVES: The aim of this study was to analyze inflammatory activity in lipofilled breast through the use of proinflammatory markers. METHODS: Fifty-four paired-breasts of female rats were divided into 4 groups: control, sham, and breasts grafted with either autologous subcutaneous (SC) WAT or autologous omentum (OM). The WAT was prepared through centrifugation, and the grafting was performed with the use of 0.9-mm blunt-tip cannula. The rats were killed 8 weeks postoperatively, and their breasts were harvested for immunohistochemical staining for CD68-expressing macrophages, gene expression (real-time PCR) for monocyte chemoattractant protein 1 (MCP-1), F4/80, Cox-2, and IL-6. RESULTS: The weights of the rats that underwent a procedure differed from those of the unmanipulated control group (P < 0.01). The macrophage counts of CD68 differed only between breasts lipofilled with OM and control (P < 0.01). MCP-1, F4/80, and Cox-2 were similarly expressed among the groups (P = 0.422, P = 0.143, and P = 0.209, respectively). The expression of IL-6 differed between breast samples grafted with SC and OM WAT (P = 0.015), but not between samples of control and OM (P = 0.752), and control and SC (P = 0.056). CONCLUSIONS: No inflammation activity was identified in the microenvironment of lipofilled breasts, indicating that chronic inflammation does not seem to be triggered by the breast lipofilling procedure.


Asunto(s)
Grasa Abdominal/trasplante , Mama/patología , Grasa Subcutánea/trasplante , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Recuento de Células , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Femenino , Inmunohistoquímica , Inyecciones Subcutáneas , Interleucina-6/genética , Interleucina-6/metabolismo , Macrófagos/metabolismo , Modelos Animales , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa
20.
J Cell Physiol ; 233(12): 9426-9436, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30063084

RESUMEN

Hypothalamic sphingosine-1-phosphate receptor 1 (S1PR1), the G protein-coupled receptor 1 of sphingosine-1-phosphate, has been described as a modulator in the control of energy homeostasis in rodents. However, this mechanism is still unclear. Here, we evaluate the role of interleukin 6 (IL-6) associated with acute physical exercise in the control of the hypothalamic S1PR1-signal transducer and activator of transcription 3 (STAT3) axis. Acute exercise session and an intracerebroventricular IL-6 injection increased S1PR1 protein content and STAT3 phosphorylation in the hypothalamus of lean and obese mice accompanied by a reduction in food consumption. Transcriptome analysis indicated a strong positive correlation between Il-6 and S1pr1 messenger RNA in several tissues of genetically diverse BXD mice strains and humans, including in the hypothalamus. Interestingly, exercise failed to stimulate the S1PR1-STAT3 axis in IL-6 knockout mice and the disruption of hypothalamic-specific IL-6 action blocked the anorexigenic effects of exercise. Taken together, our results indicate that physical exercise modulates the S1PR1 protein content in the hypothalamus, through the central action of IL-6.


Asunto(s)
Hipotálamo/metabolismo , Interleucina-6/metabolismo , Condicionamiento Físico Animal , Receptores de Lisoesfingolípidos/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Animales , Humanos , Inyecciones Intraventriculares , Interleucina-6/administración & dosificación , Interleucina-6/genética , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Lisoesfingolípidos/genética , Receptores de Esfingosina-1-Fosfato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA