Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brain Behav Immun ; 117: 330-346, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38309640

RESUMO

Nutrient composition in obesogenic diets may influence the severity of disorders associated with obesity such as insulin-resistance and chronic inflammation. Here we hypothesized that obesogenic diets rich in fat and varying in fatty acid composition, particularly in omega 6 (ω6) to omega 3 (ω3) ratio, have various effects on energy metabolism, neuroinflammation and behavior. Mice were fed either a control diet or a high fat diet (HFD) containing either low (LO), medium (ME) or high (HI) ω6/ω3 ratio. Mice from the HFD-LO group consumed less calories and exhibited less body weight gain compared to other HFD groups. Both HFD-ME and HFD-HI impaired glucose metabolism while HFD-LO partly prevented insulin intolerance and was associated with normal leptin levels despite higher subcutaneous and perigonadal adiposity. Only HFD-HI increased anxiety and impaired spatial memory, together with increased inflammation in the hypothalamus and hippocampus. Our results show that impaired glucose metabolism and neuroinflammation are uncoupled, and support that diets with a high ω6/ω3 ratio are associated with neuroinflammation and the behavioral deterioration coupled with the consumption of diets rich in fat.


Assuntos
Insulinas , Doenças Neuroinflamatórias , Animais , Camundongos , Obesidade/metabolismo , Dieta Hiperlipídica/efeitos adversos , Ácidos Graxos/metabolismo , Inflamação , Glucose
2.
Pain ; 165(2): 470-486, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-37733484

RESUMO

ABSTRACT: Lipid-rich diet is the major cause of obesity, affecting 13% of the worldwide adult population. Obesity is a major risk factor for metabolic syndrome that includes hyperlipidemia and diabetes mellitus. The early phases of metabolic syndrome are often associated with hyperexcitability of peripheral small diameter sensory fibers and painful diabetic neuropathy. Here, we investigated the effect of high-fat diet-induced obesity on the activity of dorsal root ganglion (DRG) sensory neurons and pain perception. We deciphered the underlying cellular mechanisms involving the acid-sensing ion channel 3 (ASIC3). We show that mice made obese through consuming high-fat diet developed the metabolic syndrome and prediabetes that was associated with heat pain hypersensitivity, whereas mechanical sensitivity was not affected. Concurrently, the slow conducting C fibers in the skin of obese mice showed increased activity on heating, whereas their mechanosensitivity was not altered. Although ASIC3 knockout mice fed with high-fat diet became obese, and showed signs of metabolic syndrome and prediabetes, genetic deletion, and in vivo pharmacological inhibition of ASIC3, protected mice from obesity-induced thermal hypersensitivity. We then deciphered the mechanisms involved in the heat hypersensitivity of mice and found that serum from high-fat diet-fed mice was enriched in lysophosphatidylcholine (LPC16:0, LPC18:0, and LPC18:1). These enriched lipid species directly increased the activity of DRG neurons through activating the lipid sensitive ASIC3 channel. Our results identify ASIC3 channel in DRG neurons and circulating lipid species as a mechanism contributing to the hyperexcitability of nociceptive neurons that can cause pain associated with lipid-rich diet consumption and obesity.


Assuntos
Síndrome Metabólica , Estado Pré-Diabético , Animais , Camundongos , Canais Iônicos Sensíveis a Ácido/metabolismo , Dieta Hiperlipídica/efeitos adversos , Gânglios Espinais/metabolismo , Lipídeos , Síndrome Metabólica/metabolismo , Obesidade , Dor , Estado Pré-Diabético/metabolismo , Células Receptoras Sensoriais/metabolismo
3.
Neurobiol Dis ; 191: 106393, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38154608

RESUMO

Phosphodiesterase 2 A (PDE2A) is an enzyme involved in the homeostasis of cAMP and cGMP and is the most highly expressed PDE in human brain regions critical for socio-cognitive behavior. In cerebral cortex and hippocampus, PDE2A expression level is upregulated in Fmr1-KO mice, a model of the Fragile X Syndrome (FXS), the most common form of inherited intellectual disability (ID) and autism spectrum disorder (ASD). Indeed, PDE2A translation is negatively modulated by FMRP, whose functional absence causes FXS. While the pharmacological inhibition of PDE2A has been associated to its pro-cognitive role in normal animals and in models of ID and ASD, homozygous PDE2A mutations have been identified in patients affected by ID, ASD and epilepsy. To clarify this apparent paradox about the role of PDE2A in brain development, we characterized here Pde2a+/- mice (homozygote animals being not viable) at the behavioral, cellular, molecular and electrophysiological levels. Pde2a+/- females display a milder form of the disorder with reduced cognitive performance in adulthood, conversely males show severe socio-cognitive deficits throughout their life. In males, these phenotypes are associated with microglia activation, elevated glutathione levels and increased externalization of Glutamate receptor (GluR1) in CA1, producing reduced mGluR-dependent Long-term Depression. Overall, our results reveal molecular targets of the PDE2A-dependent pathway underlying socio-cognitive performance. These results clarify the mechanism of action of pro-cognitive drugs based on PDE2A inactivation, which have been shown to be promising therapeutic approaches for Alzheimer's disease, schizophrenia, FXS as well as other forms of ASD.


Assuntos
Transtorno do Espectro Autista , Síndrome do Cromossomo X Frágil , Animais , Feminino , Humanos , Masculino , Camundongos , Cognição , Proteína do X Frágil de Retardo Mental/genética , Camundongos Knockout , Microglia/metabolismo , Diester Fosfórico Hidrolases/metabolismo
4.
FEBS J ; 2022 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-35880408

RESUMO

Maintaining energy balance is essential for survival and health. This physiological function is controlled by the brain, which adapts food intake to energy needs. Indeed, the brain constantly receives a multitude of biological signals that are derived from digested foods or that originate from the gastrointestinal tract, energy stores (liver and adipose tissues) and other metabolically active organs (muscles). These signals, which include circulating nutrients, hormones and neuronal inputs from the periphery, collectively provide information on the overall energy status of the body. In the brain, several neuronal populations can specifically detect these signals. Nutrient-sensing neurons are found in discrete brain areas and are highly enriched in the hypothalamus. In turn, specialized brain circuits coordinate homeostatic responses acting mainly on appetite, peripheral metabolism, activity and arousal. Accumulating evidence shows that hypothalamic microglial cells located at the vicinity of these circuits can influence the brain control of energy balance. However, microglial cells could have opposite effects on energy balance, that is homeostatic or detrimental, and the conditions for this shift are not totally understood yet. One hypothesis relies on the extent of microglial activation, and nutritional lipids can considerably change it.

5.
Antioxid Redox Signal ; 37(4-6): 349-369, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35166124

RESUMO

Aims: Although prebiotics, probiotics, and fecal transplantation can alter the sensation of hunger and/or feeding behavior, the role of the constitutive gut microbiota in the short-term regulation of food intake during normal physiology is still unclear. Results: An antibiotic-induced microbiota depletion study was designed to compare feeding behavior in conventional and microbiota-depleted mice. Tissues were sampled to characterize the time profile of microbiota-derived signals in mice during consumption of either standard or high-fat food for 1 h. Pharmacological and genetic tools were used to evaluate the contribution of postprandial endotoxemia and inflammatory responses in the short-term regulation of food intake. We observed constitutive microbial and macronutrient-dependent control of food intake at the time scale of a meal; that is, within 1 h of food introduction. Specifically, microbiota depletion increased food intake, and the microbiota-derived anorectic effect became significant during the consumption of high-fat but not standard food. This anorectic effect correlated with a specific postprandial microbial metabolic signature, and did not require postprandial endotoxemia or an NOD-, LRR-, and Pyrin domain-containing protein 3-inflammasome-mediated inflammatory response. Innovation and Conclusion: These findings show that the gut microbiota controls host appetite at the time scale of a meal under normal physiology. Interestingly, a microbiota-derived anorectic effect develops specifically with a high-fat meal, indicating that gut microbiota activity is involved in the satietogenic properties of foods. Antioxid. Redox Signal. 37, 349-369.


Assuntos
Depressores do Apetite , Endotoxemia , Microbiota , Animais , Ingestão de Alimentos , Peptídeo 1 Semelhante ao Glucagon , Inflamação , Camundongos , Camundongos Endogâmicos NOD , Estresse Oxidativo
6.
Nutrients ; 13(8)2021 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-34444945

RESUMO

Anorexia nervosa (AN) is a severe eating disorder where caloric restriction, excessive physical activity and metabolic alterations lead to life-threatening situations. Despite weight restoration after treatment, a significant part of patients experience relapses. In this translational study, we combined clinical and preclinical approaches. We describe preliminary data about the effect of weight gain on the symptomatology of patients suffering from acute AN (n = 225) and partially recovered (n = 41). We measured more precisely physical activity with continuous cardiac monitoring in a sub-group (n = 68). Using a mouse model, we investigated whether a long-term food restriction followed by nutritional recovery associated or not with physical activity may differentially impact peripheral and central homeostatic regulation. We assessed the plasma concentration of acyl ghrelin, desacyl ghrelin and leptin and the mRNA expression of hypothalamic neuropeptides and their receptors. Our data show an effect of undernutrition history on the level of physical activity in AN. The preclinical model supports an important role of physical activity in the recovery process and points out the leptin system as one factor that can drive a reliable restoration of metabolic variables through the hypothalamic regulation of neuropeptides involved in feeding behavior.


Assuntos
Anorexia Nervosa/metabolismo , Anorexia Nervosa/reabilitação , Exercício Físico , Adolescente , Adulto , Animais , Anorexia Nervosa/sangue , Índice de Massa Corporal , Peso Corporal , Comportamento Alimentar , Feminino , Grelina/análogos & derivados , Grelina/sangue , Grelina/metabolismo , Frequência Cardíaca , Humanos , Hipotálamo/metabolismo , Leptina/sangue , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Neuropeptídeos/metabolismo , RNA Mensageiro/metabolismo , Recidiva , Aumento de Peso , Adulto Jovem
7.
Pharmacol Res ; 167: 105539, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33737242

RESUMO

Deterioration of insulin secretion and pancreatic beta-cell mass by inflammatory attacks is one of the main pathophysiological features of type 2 diabetes (T2D). Therefore, preserving beta-cell mass and stimulating insulin secretion only in response to glucose for avoiding the hypoglycemia risks, are the most state-of-the-art option for the treatment of T2D. In this study we tested two correlated hypothesis that 1/ the endogenous peptide released from sortilin, known as PE, that stimulates insulin secretion only in response to glucose, protects beta-cells against death induced by cytokines, and 2/ Spadin and Mini-Spadin, two synthetic peptides derived from PE, that mimic the effects of PE in insulin secretion, also provide beneficial effect on beta-cells survival. We show that PE and its derivatives by inducing a rise of intracellular calcium concentration by depolarizing the membrane protect beta-cells against death induced by Interleukin-1ß. Using biochemical, confocal imaging and cell biology techniques, we reveal that the protective effects of PE and its derivatives rely on the activation of the CaM-Kinase pathway, and on the phosphorylation and activation of the transcription factor CREB. In addition, Mini-Spadin promotes beta-cell proliferation, suggesting its possible regenerative effect. This study highlights new possible roles of PE in pancreatic beta-cell survival and its derivatives as pharmacological tools against diabetes.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Peptídeos/farmacologia , Proteínas Adaptadoras de Transporte Vesicular/química , Animais , Linhagem Celular , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Peptídeos/química , Ratos , Transdução de Sinais/efeitos dos fármacos
8.
Glia ; 69(1): 42-60, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32659044

RESUMO

In humans, obesity is associated with brain inflammation, glial reactivity, and immune cells infiltration. Studies in rodents have shown that glial reactivity occurs within 24 hr of high-fat diet (HFD) consumption, long before obesity development, and takes place mainly in the hypothalamus (HT), a crucial brain structure for controlling body weight. Here, we sought to characterize the postprandial HT inflammatory response to 1, 3, and 6 hr of exposure to either a standard diet (SD) or HFD. HFD exposure increased gene expression of astrocyte and microglial markers (glial fibrillary acidic protein [GFAP] and Iba1, respectively) compared to SD-treated mice and induced morphological modifications of microglial cells in HT. This remodeling was associated with higher expression of inflammatory genes and differential regulation of hypothalamic neuropeptides involved in energy balance regulation. DREADD and PLX5622 technologies, used to modulate GFAP-positive or microglial cells activity, respectively, showed that both glial cell types are involved in hypothalamic postprandial inflammation, with their own specific kinetics and reactiveness to ingested foods. Thus, recurrent exacerbated postprandial inflammation in the brain might promote obesity and needs to be characterized to address this worldwide crisis.


Assuntos
Gorduras na Dieta , Microglia , Animais , Dieta Hiperlipídica/efeitos adversos , Proteína Glial Fibrilar Ácida , Hipotálamo , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade
9.
Cell Rep ; 30(9): 3067-3078.e5, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32130907

RESUMO

Mechanistic studies in rodents evidenced synaptic remodeling in neuronal circuits that control food intake. However, the physiological relevance of this process is not well defined. Here, we show that the firing activity of anorexigenic POMC neurons located in the hypothalamus is increased after a standard meal. Postprandial hyperactivity of POMC neurons relies on synaptic plasticity that engages pre-synaptic mechanisms, which does not involve structural remodeling of synapses but retraction of glial coverage. These functional and morphological neuroglial changes are triggered by postprandial hyperglycemia. Chemogenetically induced glial retraction on POMC neurons is sufficient to increase POMC activity and modify meal patterns. These findings indicate that synaptic plasticity within the melanocortin system happens at the timescale of meals and likely contributes to short-term control of food intake. Interestingly, these effects are lost with a high-fat meal, suggesting that neuroglial plasticity of POMC neurons is involved in the satietogenic properties of foods.


Assuntos
Hiperglicemia/fisiopatologia , Hipotálamo/metabolismo , Refeições , Neuroglia/patologia , Plasticidade Neuronal , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Animais , Glicemia/metabolismo , Fenômenos Eletrofisiológicos , Comportamento Alimentar , Hiperglicemia/sangue , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Período Pós-Prandial , Sinapses/metabolismo
10.
Artigo em Inglês | MEDLINE | ID: mdl-28855891

RESUMO

The hypothalamus is a key brain region in the regulation of energy balance as it controls food intake and both energy storage and expenditure through integration of humoral, neural, and nutrient-related signals and cues. Many years of research have focused on the regulation of energy balance by hypothalamic neurons, but the most recent findings suggest that neurons and glial cells, such as microglia and astrocytes, in the hypothalamus actually orchestrate together several metabolic functions. Because glial cells have been described as mediators of inflammatory processes in the brain, the existence of a causal link between hypothalamic inflammation and the deregulations of feeding behavior, leading to involuntary weight loss or obesity for example, has been suggested. Several inflammatory pathways that could impair the hypothalamic control of energy balance have been studied over the years such as, among others, toll-like receptors and canonical cytokines. Yet, less studied so far, chemokines also represent interesting candidates that could link the aforementioned pathways and the activity of hypothalamic neurons. Indeed, chemokines, in addition to their role in attracting immune cells to the inflamed site, have been suggested to be capable of neuromodulation. Thus, they could disrupt cellular activity together with synthesis and/or secretion of multiple neurotransmitters/mediators involved in the maintenance of energy balance. This review discusses the different inflammatory pathways that have been identified so far in the hypothalamus in the context of feeding behavior and body weight control impairments, with a particular focus on chemokines signaling that opens a new avenue in the understanding of the major role played by inflammation in obesity.

11.
Health Psychol ; 36(11): 1059-1064, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28650196

RESUMO

OBJECTIVE: Given the impact of individuals' habits on health, it is important to study how behaviors can become habitual. Cortisol has been well documented to have a role in habit formation. This study aimed to elucidate the influence of the circadian rhythm of cortisol on habit formation in a real-life setting. METHOD: Forty-eight students were followed for 90 days during which they attempted to adopt a health behavior (psoas iliac stretch). They were randomly assigned to perform the stretch either upon waking in the morning, when cortisol concentrations are high, or before evening bedtime, when cortisol levels approach the nadir. A smartphone application was used to assess the Self-Report Behavioural Automaticity Index every day and to provide reminders for salivary measurements every 30 days. The speed of the health habit formation process was calculated by modeling the learning curves. RESULTS: Extrapolation of the curves indicated that the morning group achieved automaticity at an earlier time point (105.95 days) than did the evening group (154.01 days). In addition, the cortisol level during the performance of the health behavior was identified as a significant mediator of the time point when the health behavior became habitual. CONCLUSION: The present findings suggest that the time course of the development of healthy habits depends on the time of the day and that the effect is mediated through diurnal variation in cortisol levels. Future studies are now needed to determine to what extent cortisol rhythmicity can help individuals to adopt new health behaviors. (PsycINFO Database Record


Assuntos
Comportamentos Relacionados com a Saúde , Hidrocortisona/metabolismo , Adulto , Ritmo Circadiano , Terapia por Exercício , Feminino , Hábitos , Humanos , Masculino , Memória , Exercícios de Alongamento Muscular , Saliva/metabolismo , Adulto Jovem
12.
Neuroendocrinology ; 105(4): 372-383, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28006784

RESUMO

Although the short-term effects of fasting or energy deficit on hypothalamic neuropeptide circuitries are now better understood, the effects of long-term energy deficit and refeeding remain to be elucidated. We showed that after a long-term energy deficit, mice exhibited persistent hypoleptinemia following the refeeding period despite restoration of fat mass, ovarian activity, and feeding behavior. We aimed to examine the hypothalamic adaptations after 10 weeks of energy deficit and after 10 further weeks of nutritional recovery. To do so, we assessed the mRNA levels of the leptin receptor and the main orexigenic and anorexigenic peptides, and their receptors regulated by leptin. Markers of hypothalamic inflammation were assessed as leptin can also participate in this phenomenon. Long-term time-restricted feeding and separation induced significant increase in mRNA levels of hypothalamic orexigenic peptides, while both Y1 and Y5 receptor mRNAs were downregulated. No changes occurred in the mRNA levels of orexin (OX), melanin-concentrating hormone, pro-opiomelanocortin, 26RFa (26-amino acid RF-amide peptide), and their receptors despite an increase in the expression of melanocortin receptors (MC3-R and MC4-R) and OXR1 (OX receptor 1). The refeeding period induced an overexpression of leptin receptor mRNA in the hypothalamus. The other assessed mRNA levels were normalized except for Y2, Y5, MC3-R, and MC4-R, which remained upregulated. No convincing changes were observed in neuroinflammatory markers, even if interleukin-1ß mRNA levels were increased in parallel with those of Iba1 (ionized calcium-binding adaptor molecule 1), a marker of microglial activation. Normalization of leptin-regulated functions and hypothalamic gene expressions in refed mice with low plasma leptin levels could be sustained by recalibration of hypothalamic sensitivity to leptin.


Assuntos
Modelos Animais de Doenças , Ingestão de Alimentos/fisiologia , Hipolipoproteinemias/patologia , Hipotálamo/metabolismo , Leptina/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Peso Corporal/fisiologia , Citocinas/genética , Citocinas/metabolismo , Feminino , Hipolipoproteinemias/sangue , Hormônios Hipotalâmicos , Melaninas , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeo Y/metabolismo , Neuropeptídeos/metabolismo , Orexinas/genética , Orexinas/metabolismo , Hormônios Hipofisários , RNA Mensageiro/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo
13.
Bio Protoc ; 7(16): e2521, 2017 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34541181

RESUMO

The hypothalamus is a primary brain area which, in mammals, regulates several physiological functions that are all related to maintaining general homeostasis, by linking the central nervous system (CNS) and the periphery. The hypothalamus itself can be considered an endocrine brain region of some sort as it hosts in its different nuclei several kinds of neuropeptide-producing and -secreting neurons. These neuropeptides have specific roles and participate in the regulation of homeostasis in general, which includes the regulation of energy metabolism, feeding behavior, water intake and body core temperature for example. As previously mentioned, in order to exert their effects, these peptides have to be produced but also, and mostly, to be secreted. In this context, it is of great importance to be able to assess how certain conditions, diseases, or treatments can actually influence the secretion of neuropeptides, thus the function of the different neuropeptidergic circuits. One method to assess this is the perifusion of hypothalamic explants followed by quantification of peptides within the collected fractions. Here, we explain step-by-step how to collect fractions during ex vivo perifusion of hypothalamic explants in which one can determine quantitatively neuropeptide/neurohormone release from these viable isolated tissues. Hypothalami perifusion has two great advantages over other existing assays: (1) it allows pharmacological manipulation to dissect out signaling mechanisms underlying release of different neuropeptides/neurohormones in the hypothalamic explants and, (2) it allows simultaneous experiments with different conditions on multiple hypothalami preparations, (3) it is, to our knowledge, the only method that permits the study of neuropeptide secretion in basal conditions and under repeated stimulations with the same hypothalami explants.

14.
EMBO Rep ; 17(12): 1738-1752, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27733491

RESUMO

Sickness behavior defines the endocrine, autonomic, behavioral, and metabolic responses associated with infection. While inflammatory responses were suggested to be instrumental in the loss of appetite and body weight, the molecular underpinning remains unknown. Here, we show that systemic or central lipopolysaccharide (LPS) injection results in specific hypothalamic changes characterized by a precocious increase in the chemokine ligand 2 (CCL2) followed by an increase in pro-inflammatory cytokines and a decrease in the orexigenic neuropeptide melanin-concentrating hormone (MCH). We therefore hypothesized that CCL2 could be the central relay for the loss in body weight induced by the inflammatory signal LPS. We find that central delivery of CCL2 promotes neuroinflammation and the decrease in MCH and body weight. MCH neurons express CCL2 receptor and respond to CCL2 by decreasing both electrical activity and MCH release. Pharmacological or genetic inhibition of CCL2 signaling opposes the response to LPS at both molecular and physiologic levels. We conclude that CCL2 signaling onto MCH neurons represents a core mechanism that relays peripheral inflammation to sickness behavior.


Assuntos
Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Hormônios Hipotalâmicos/metabolismo , Hipotálamo/metabolismo , Inflamação/metabolismo , Melaninas/metabolismo , Neurônios/metabolismo , Hormônios Hipofisários/metabolismo , Transdução de Sinais , Animais , Quimiocina CCL2/deficiência , Quimiocina CCL2/imunologia , Citocinas/biossíntese , Citocinas/genética , Citocinas/imunologia , Hormônios Hipotalâmicos/genética , Hormônios Hipotalâmicos/imunologia , Comportamento de Doença , Lipopolissacarídeos/imunologia , Melaninas/genética , Melaninas/imunologia , Camundongos , Neurônios/imunologia , Hormônios Hipofisários/genética , Hormônios Hipofisários/imunologia , Receptores CCR2/metabolismo , Redução de Peso
15.
Biol Aujourdhui ; 210(4): 211-225, 2016.
Artigo em Francês | MEDLINE | ID: mdl-28327280

RESUMO

The hypothalamus is a key brain region in the regulation of energy balance. It especially controls food intake and both energy storage and expenditure through integration of humoral, neural and nutrient-related signals and cues. Hypothalamic neurons and glial cells act jointly to orchestrate, both spatially and temporally, regulated metabolic functions of the hypothalamus. Thus, the existence of a causal link between hypothalamic inflammation and deregulations of feeding behavior, such as involuntary weight-loss or obesity, has been suggested. Among the inflammatory mediators that could induce deregulations of hypothalamic control of the energy balance, chemokines represent interesting candidates. Indeed, chemokines, primarily known for their chemoattractant role of immune cells to the inflamed site, have also been suggested capable of neuromodulation. Thus, chemokines could disrupt cellular activity together with synthesis and/or secretion of multiple neurotransmitters/mediators that are involved in the maintenance of energy balance. Here, we relate, on one hand, recent results showing the primary role of the central chemokinergic signaling CCL2/CCR2 for metabolic and behavioral adaptation to high-grade inflammation, especially loss of appetite and weight, through its activity on hypothalamic neurons producing the orexigenic peptide Melanin-Concentrating Hormone (MCH) and, on the other hand, results that suggest that chemokines could also deregulate hypothalamic neuropeptidergic circuits to induce an opposite phenotype and eventually participate in the onset/development of obesity. In more details, we will emphasize a study recently showing, in a model of high-grade acute inflammation of LPS injection in mice, that central CCL2/CCR2 signaling is of primary importance for several aspects explaining weight loss associated with inflammation: after LPS injection, animals lose weight, reduce their food intake, increase their fat oxidation (thus energy consumption from fat storage)...These inflammation-induced metabolic and behavioral changes are reduced when central CCR2 signaling is disrupted either pharmacologically (by a specific inhibitor of CCR2) or genetically (in mice deficient for CCR2). This underlines the importance of this signaling in inflammation-related weight loss. We further determined that the LPS-induced and CCR2-mediated weight loss depends on the direct effect of CCR2 activation on MCH neurons activity. Indeed, the MCH neurons express CCR2, and the application of CCL2 on brain slices revealed that activation of CCR2 actually depolarizes MCH neurons and induces delays and/or failures of action potential emission. Furthermore, CCL2 is able to reduce KCl-evoked MCH secretion from hypothalamic explants. Taken together, these results demonstrate the role of the central CCL2/CCR2 signaling in metabolic and behavioral adaptation to inflammation. On the other hand, this first description of how the chemokinergic system can actually modulate the activity of the hypothalamic regulation of energy balance, but also some less advanced studies and some unpublished data, suggest that some other chemokines, such as CCL5, could participate in the development of the opposite phenotype, that is to say obesity.


Assuntos
Quimiocinas/fisiologia , Doenças Hipotalâmicas/complicações , Doenças Hipotalâmicas/metabolismo , Doenças Metabólicas/etiologia , Animais , Metabolismo Energético/fisiologia , Humanos , Hipotálamo/metabolismo , Hipotálamo/patologia , Inflamação , Camundongos , Obesidade/etiologia , Obesidade/imunologia , Obesidade/metabolismo , Redução de Peso/genética , Redução de Peso/imunologia
16.
Ann N Y Acad Sci ; 1351: 127-40, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26251227

RESUMO

Inflammation is an innate mechanism that defends organisms against harmful stimuli. Inflammation leads to the production and secretion of proinflammatory mediators that activate and recruit immune cells to damaged tissues, including the brain, to resolve the cause of inflammation. In the central nervous system, inflammation is referred to as neuroinflammation, which occurs in various pathological conditions of the brain. The primary role of neuroinflammation is to protect the brain. However, prolonged and/or inappropriate inflammation can be harmful for the brain, from individual cells to the whole tissue. This review focuses on a particular type of inflammatory mediator, chemokines, and describes their complex effects both under physiological and pathophysiological conditions of the brain. The clinical relevance of the multiple characters of chemokines is highlighted with respect to acute and chronic inflammation of the brain, including their actions in stroke and Alzheimer's disease, respectively.


Assuntos
Doença de Alzheimer/fisiopatologia , Encéfalo/imunologia , Quimiocinas/imunologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Acidente Vascular Cerebral/fisiopatologia , Doença de Alzheimer/imunologia , Encéfalo/citologia , Humanos , Hipóxia-Isquemia Encefálica/imunologia , Inflamação/imunologia , Mediadores da Inflamação/imunologia , Neuroimunomodulação/imunologia , Receptores de Quimiocinas/imunologia , Acidente Vascular Cerebral/imunologia
17.
Surg Endosc ; 28(2): 592-602, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24196540

RESUMO

BACKGROUND: Sleeve gastrectomy (SG) has become a popular bariatric procedure. The mechanisms responsible for weight loss and improvement of metabolic disturbances have still not been completely elucidated. We investigated the effect of SG on body weight, adipose tissue depots, glucose tolerance, and liver steatosis independent of reduced caloric intake in high-fat-diet-induced obese mice. METHODS: C57BI/6 J mice fed a high fat diet (45 %) for 33 weeks were divided into three groups: sleeve gastrectomy (SG, 13 mice), sham-operated ad libitum fed (SALF, 13 mice) and sham-operated pair fed (PFS, 13 mice). The animals were humanely killed 23 days after surgery. RESULTS: In SG mice, food intake was reduced transiently, but weight loss was significant and persistent compared to controls (SG vs. PFS, P < 0.05; PFS vs. SALF, P < 0.05). SG mice showed improved glucose tolerance and lower levels of liver steatosis compared with controls (area under the curve, SG vs. PFS, P < 0.01; PFS vs. SALF, P < 0.05) (liver steatosis, SG vs. PFS, P < 0.05; PFS vs. SALF, P < 0.01). This was associated with a decrease in the ratios of the weight of pancreas, epididymal and inguinal adipose tissues to body weight, and an increase in the ratio of brown adipose tissue weight to body weight. Epididymal adipose tissue was also infiltrated by fewer activated T cells and by more anti-inflammatory regulatory T cells. Serum levels of fasting acyl ghrelin were still significantly decreased 3 weeks after surgery in SG mice compared to PFS mice (P < 0.05). CONCLUSIONS: Reduced white adipose tissue inflammation, modification of adipose tissue development (brown vs. white adipose tissue), and ectopic fat are potential mechanisms that may account for the reduced caloric intake independent effects of SG.


Assuntos
Tecido Adiposo Branco/patologia , Ingestão de Energia/fisiologia , Gastrectomia/métodos , Gastroplastia/métodos , Inflamação/patologia , Obesidade/cirurgia , Redução de Peso/fisiologia , Animais , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia
18.
Nat Neurosci ; 16(7): 845-7, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23708141
19.
Am J Clin Nutr ; 94(2): 450-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21677057

RESUMO

BACKGROUND: Obesity is considered a low-grade inflammatory state that improves with weight loss. In addition to acute-phase proteins, other cytokines might contribute to systemic inflammation. OBJECTIVE: Our objective was to compare serum concentrations of a large panel of inflammation-related factors in obese and normal-weight subjects and to determine kinetic changes induced by caloric restriction. DESIGN: The cohort comprised 14 normal-weight women and 51 obese women who were followed over 2 y after Roux-en-Y gastric bypass. Multiplexed proteomics were used to simultaneously assay 27 cytokines and growth factors in serum. RESULTS: Concentrations of interleukin (IL)-9, IL-1-receptor antagonist, IL-10, interferon-γ-inducible protein 10, macrophage inflammatory protein 1ß, monocyte chemoattractant protein 1, IL-8, RANTES (regulated upon activation, normal T cell expressed and secreted), monokine induced by interferon-γ, and vascular endothelial growth factor were found to be elevated in obesity. IL-10 was further elevated in diabetic obese patients, whereas eotaxin was found to be higher only in diabetic subjects. After surgery, many factors showed a biphasic pattern of variation, decreasing sharply at month 3 before rising back to presurgical values at month 6; these changes closely tracked similar kinetic changes in calorie and carbohydrate intake. After 1 y, an overall reduction in cytokines accompanied the reduction in body mass index and an amelioration in metabolic status. CONCLUSIONS: Obesity is associated with elevated circulating concentrations of a large panel of cytokines. Coordinated kinetic changes during weight loss suggest an early influence of calorie and carbohydrate intakes, whereas a longer-term reduction in corpulence might prevail in regulating circulating cytokine concentrations. This trial is registered at clincaltrials.gov as NCT00476658.


Assuntos
Citocinas/sangue , Carboidratos da Dieta/administração & dosagem , Ingestão de Energia , Obesidade/imunologia , Procedimentos Cirúrgicos Operatórios , Redução de Peso , Adulto , Diabetes Mellitus Tipo 2/imunologia , Feminino , Derivação Gástrica , Humanos , Pessoa de Meia-Idade , Obesidade/cirurgia
20.
Front Neuroendocrinol ; 32(1): 10-24, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20624414

RESUMO

Chemokines are small secreted proteins that chemoattract and activate immune and non-immune cells. Their role in the immune system is well-known, and it has recently been suggested that they may also play a role in the central nervous system (CNS). Indeed, they do not only act as immunoinflammatory mediators in the brain but they also act as potential modulators in neurotransmission. Although we are only beginning to be aware of the implication of chemokines in neuroendocrine functions, this review aims at summarizing what is known in that booming field of research. First we describe the expression of chemokines and their receptors in the CNS with a focus on the hypothalamo-pituitary system. Secondly, we present what is known on some chemokines in the regulation of neuroendocrine functions such as cell migration, stress, thermoregulation, drinking and feeding as well as anterior pituitary functions. We suggest that chemokines provide a fine modulatory tuning system of neuroendocrine regulations.


Assuntos
Quimiocinas/fisiologia , Sistemas Neurossecretores/fisiologia , Receptores de Quimiocinas/fisiologia , Animais , Quimiocinas/genética , Quimiocinas/metabolismo , Humanos , Modelos Biológicos , Sistemas Neurossecretores/metabolismo , Adeno-Hipófise/metabolismo , Adeno-Hipófise/fisiologia , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...