Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Mol Metab ; 47: 101186, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33571700

RESUMEN

OBJECTIVE: The ventromedial nucleus of the hypothalamus (VMH) is a critical component of the forebrain pathways that regulate energy homeostasis. It also plays an important role in the metabolic response to fasting. However, the mechanisms contributing to these physiological processes remain elusive. Autophagy is an evolutionarily conserved mechanism that maintains cellular homeostasis by turning over cellular components and providing nutrients to the cells during starvation. Here, we investigated the importance of the autophagy-related gene Atg7 in Sf1-expressing neurons of the VMH in control and fasted conditions. METHODS: We generated Sf1-Cre; Atg7loxP/loxP mice and examined their metabolic and cellular response to fasting. RESULTS: Fasting induces autophagy in the VMH, and mice lacking Atg7 in Sf1-expressing neurons display altered leptin sensitivity and impaired energy expenditure regulation in response to fasting. Moreover, loss of Atg7 in Sf1 neurons causes alterations in the central response to fasting. Furthermore, alterations in mitochondria morphology and activity are observed in mutant mice. CONCLUSION: Together, these data show that autophagy is nutritionally regulated in VMH neurons and that VMH autophagy participates in the control of energy homeostasis during fasting.


Asunto(s)
Autofagia , Ayuno , Mitocondrias/metabolismo , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/metabolismo , Animales , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/metabolismo , Metabolismo Energético , Femenino , Homeostasis , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Transcriptoma
2.
Cell Rep ; 30(9): 3067-3078.e5, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32130907

RESUMEN

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.


Asunto(s)
Hiperglucemia/fisiopatología , Hipotálamo/metabolismo , Comidas , Neuroglía/patología , Plasticidad Neuronal , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Animales , Glucemia/metabolismo , Fenómenos Electrofisiológicos , Conducta Alimentaria , Hiperglucemia/sangre , Ratones Endogámicos C57BL , Ratones Transgénicos , Periodo Posprandial , Sinapsis/metabolismo
3.
Mol Metab ; 20: 166-177, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30553770

RESUMEN

OBJECTIVE: Hypothalamic glucose sensing (HGS) initiates insulin secretion (IS) via a vagal control, participating in energy homeostasis. This requires mitochondrial reactive oxygen species (mROS) signaling, dependent on mitochondrial fission, as shown by invalidation of the hypothalamic DRP1 protein. Here, our objectives were to determine whether a model with a HGS defect induced by a short, high fat-high sucrose (HFHS) diet in rats affected the fission machinery and mROS signaling within the mediobasal hypothalamus (MBH). METHODS: Rats fed a HFHS diet for 3 weeks were compared with animals fed a normal chow. Both in vitro (calcium imaging) and in vivo (vagal nerve activity recordings) experiments to measure the electrical activity of isolated MBH gluco-sensitive neurons in response to increased glucose level were performed. In parallel, insulin secretion to a direct glucose stimulus in isolated islets vs. insulin secretion resulting from brain glucose stimulation was evaluated. Intra-carotid glucose load-induced hypothalamic DRP1 translocation to mitochondria and mROS (H2O2) production were assessed in both groups. Finally, compound C was intracerebroventricularly injected to block the proposed AMPK-inhibited DRP1 translocation in the MBH to reverse the phenotype of HFHS fed animals. RESULTS: Rats fed a HFHS diet displayed a decreased HGS-induced IS. Responses of MBH neurons to glucose exhibited an alteration of their electrical activity, whereas glucose-induced insulin secretion in isolated islets was not affected. These MBH defects correlated with a decreased ROS signaling and glucose-induced translocation of the fission protein DRP1, as the vagal activity was altered. AMPK-induced inhibition of DRP1 translocation increased in this model, but its reversal through the injection of the compound C, an AMPK inhibitor, failed to restore HGS-induced IS. CONCLUSIONS: A hypothalamic alteration of DRP1-induced fission and mROS signaling in response to glucose was observed in HGS-induced IS of rats exposed to a 3 week HFHS diet. Early hypothalamic modifications of the neuronal activity could participate in a primary defect of the control of IS and ultimately, the development of diabetes.


Asunto(s)
Glucemia/metabolismo , Dinaminas/metabolismo , Hipotálamo/metabolismo , Mitocondrias/metabolismo , Células Receptoras Sensoriales/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Arterias Carótidas/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Proteínas Quinasas/metabolismo , Transporte de Proteínas , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
4.
Diabetes ; 66(2): 314-324, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27899482

RESUMEN

The mediobasal hypothalamus (MBH) contains neurons capable of directly detecting metabolic signals such as glucose to control energy homeostasis. Among them, glucose-excited (GE) neurons increase their electrical activity when glucose rises. In view of previous work, we hypothesized that transient receptor potential canonical type 3 (TRPC3) channels are involved in hypothalamic glucose detection and the control of energy homeostasis. To investigate the role of TRPC3, we used constitutive and conditional TRPC3-deficient mouse models. Hypothalamic glucose detection was studied in vivo by measuring food intake and insulin secretion in response to increased brain glucose level. The role of TRPC3 in GE neuron response to glucose was studied by using in vitro calcium imaging on freshly dissociated MBH neurons. We found that whole-body and MBH TRPC3-deficient mice have increased body weight and food intake. The anorectic effect of intracerebroventricular glucose and the insulin secretory response to intracarotid glucose injection are blunted in TRPC3-deficient mice. TRPC3 loss of function or pharmacological inhibition blunts calcium responses to glucose in MBH neurons in vitro. Together, the results demonstrate that TRPC3 channels are required for the response to glucose of MBH GE neurons and the central effect of glucose on insulin secretion and food intake.


Asunto(s)
Peso Corporal/genética , Ingestión de Alimentos/genética , Metabolismo Energético/genética , Glucosa/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Neuronas/metabolismo , Canales Catiónicos TRPC/genética , Animales , Western Blotting , Ayuno , Prueba de Tolerancia a la Glucosa , Homeostasis , Hipotálamo/citología , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Canales Catiónicos TRPC/metabolismo
5.
Cell Metab ; 23(2): 324-34, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26621107

RESUMEN

The composition of gut microbiota has been associated with host metabolic phenotypes, but it is not known if gut bacteria may influence host appetite. Here we show that regular nutrient provision stabilizes exponential growth of E. coli, with the stationary phase occurring 20 min after nutrient supply accompanied by bacterial proteome changes, suggesting involvement of bacterial proteins in host satiety. Indeed, intestinal infusions of E. coli stationary phase proteins increased plasma PYY and their intraperitoneal injections suppressed acutely food intake and activated c-Fos in hypothalamic POMC neurons, while their repeated administrations reduced meal size. ClpB, a bacterial protein mimetic of α-MSH, was upregulated in the E. coli stationary phase, was detected in plasma proportional to ClpB DNA in feces, and stimulated firing rate of hypothalamic POMC neurons. Thus, these data show that bacterial proteins produced after nutrient-induced E. coli growth may signal meal termination. Furthermore, continuous exposure to E. coli proteins may influence long-term meal pattern.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/crecimiento & desarrollo , Tracto Gastrointestinal/microbiología , Respuesta de Saciedad , Adenosina Trifosfato/biosíntesis , Amígdala del Cerebelo/metabolismo , Animales , Fenómenos Electrofisiológicos , Endopeptidasa Clp , Escherichia coli/metabolismo , Conducta Alimentaria , Femenino , Péptido 1 Similar al Glucagón/metabolismo , Proteínas de Choque Térmico/metabolismo , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Péptido YY/metabolismo , Proopiomelanocortina/metabolismo , Proteómica , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Sprague-Dawley , Ratas Wistar
6.
Cell Rep ; 6(3): 438-44, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24485657

RESUMEN

The reversible phosphorylation of the α subunit of eukaryotic initiation factor 2 (eIF2α) is a highly conserved signal implicated in the cellular adaptation to numerous stresses such as the one caused by amino acid limitation. In response to dietary amino acid deficiency, the brain-specific activation of the eIF2α kinase GCN2 leads to food intake inhibition. We report here that GCN2 is rapidly activated in the mediobasal hypothalamus (MBH) after consumption of a leucine-deficient diet. Furthermore, knockdown of GCN2 in this particular area shows that MBH GCN2 activity controls the onset of the aversive response. Importantly, pharmacological experiments demonstrate that the sole phosphorylation of eIF2α in the MBH is sufficient to regulate food intake. eIF2α signaling being at the crossroad of stress pathways activated in several pathological states, our study indicates that hypothalamic eIF2α phosphorylation could play a critical role in the onset of anorexia associated with certain diseases.


Asunto(s)
Ingestión de Alimentos/fisiología , Factor 2 Eucariótico de Iniciación/metabolismo , Hipotálamo/metabolismo , Transducción de Señal , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Técnicas de Silenciamiento del Gen , Leucina/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo
7.
Antioxid Redox Signal ; 20(4): 557-73, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23879244

RESUMEN

AIMS: We have previously demonstrated that central apelin is implicated in the control of peripheral glycemia, and its action depends on nutritional (fast versus fed) and physiological (normal versus diabetic) states. An intracerebroventricular (icv) injection of a high dose of apelin, similar to that observed in obese/diabetic mice, increase fasted glycemia, suggesting (i) that apelin contributes to the establishment of a diabetic state, and (ii) the existence of a hypothalamic to liver axis. Using pharmacological, genetic, and nutritional approaches, we aim at unraveling this system of regulation by identifying the hypothalamic molecular actors that trigger the apelin effect on liver glucose metabolism and glycemia. RESULTS: We show that icv apelin injection stimulates liver glycogenolysis and gluconeogenesis via an over-activation of the sympathetic nervous system (SNS), leading to fasted hyperglycemia. The effect of central apelin on liver function is dependent of an increased production of hypothalamic reactive oxygen species (ROS). These data are strengthened by experiments using lentiviral vector-mediated over-expression of apelin in hypothalamus of mice that present over-activation of SNS associated to an increase in hepatic glucose production. Finally, we report that mice fed a high-fat diet present major alterations of hypothalamic apelin/ROS signaling, leading to activation of glycogenolysis. INNOVATION/CONCLUSION: These data bring compelling evidence that hypothalamic apelin is one master switch that participates in the onset of diabetes by directly acting on liver function. Our data support the idea that hypothalamic apelin is a new potential therapeutic target to treat diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Hígado/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Adipoquinas , Animales , Apelina , Sistema Nervioso Autónomo , Glucemia , Gluconeogénesis , Glucogenólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Transducción de Señal
8.
J Cereb Blood Flow Metab ; 34(2): 339-46, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24301293

RESUMEN

Hypothalamic glucose detection participates in maintaining glycemic balance, food intake, and thermogenesis. Although hypothalamic neurons are the executive cells involved in these responses, there is increasing evidence that astrocytes participate in glucose sensing (GS); however, it is unknown whether astroglial networking is required for glucose sensitivity. Astroglial connexins 30 and 43 (Cx30 and Cx43) form hexameric channels, which are apposed in gap junctions, allowing for the intercellular transfer of small molecules such as glucose throughout the astroglial networks. Here, we hypothesized that hypothalamic glucose sensitivity requires these connexins. First, we showed that both Cxs are enriched in the rat hypothalamus, with highly concentrated Cx43 expression around blood vessels of the mediobasal hypothalamus (MBH). Both fasting and high glycemic levels rapidly altered the protein levels of MBH astroglial connexins, suggesting cross talk within the MBH between glycemic status and the connexins' ability to dispatch glucose. Finally, the inhibition of MBH Cx43 (by transient RNA interference) attenuated hypothalamic glucose sensitivity in rats, which was demonstrated by a pronounced decreased insulin secretion in response to a brain glucose challenge. These results illustrate that astroglial connexins contribute to hypothalamic GS.


Asunto(s)
Astrocitos/metabolismo , Conexina 43/metabolismo , Conexinas/metabolismo , Glucosa/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Astrocitos/citología , Conexina 30 , Conexina 43/genética , Conexinas/genética , Ayuno/metabolismo , Glucosa/genética , Hipotálamo/citología , Secreción de Insulina , Masculino , Proteínas del Tejido Nervioso/genética , Interferencia de ARN , Ratas , Ratas Wistar
9.
PLoS One ; 8(9): e74021, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24040150

RESUMEN

Variations in plasma fatty acid (FA) concentrations are detected by FA sensing neurons in specific brain areas such as the hypothalamus. These neurons play a physiological role in the control of food intake and the regulation of hepatic glucose production. Le Foll et al. previously showed in vitro that at least 50% of the FA sensing in ventromedial hypothalamic (VMH) neurons is attributable to the interaction of long chain FA with FA translocase/CD36 (CD36). The present work assessed whether in vivo effects of hypothalamic FA sensing might be partly mediated by CD36 or intracellular events such as acylCoA synthesis or ß-oxidation. To that end, a catheter was implanted in the carotid artery toward the brain in male Wistar rats. After 1 wk recovery, animals were food-deprived for 5 h, then 10 min infusions of triglyceride emulsion, Intralipid +/- heparin (IL, IL(H), respectively) or saline/heparin (SH) were carried out and food intake was assessed over the next 5 h. Experimental groups included: 1) Rats previously injected in ventromedian nucleus (VMN) with shRNA against CD36 or scrambled RNA; 2) Etomoxir (CPT1 inhibitor) or saline co-infused with IL(H)/S(H); and 3) Triacsin C (acylCoA synthase inhibitor) or saline co-infused with IL(H)/S(H). IL(H) significantly lowered food intake during refeeding compared to S(H) (p<0.001). Five hours after refeeding, etomoxir did not affect this inhibitory effect of IL(H) on food intake while VMN CD36 depletion totally prevented it. Triacsin C also prevented IL(H) effects on food intake. In conclusion, the effect of FA to inhibit food intake is dependent on VMN CD36 and acylCoA synthesis but does not required FA oxidation.


Asunto(s)
Antígenos CD36/metabolismo , Ácidos Grasos/metabolismo , Conducta Alimentaria/fisiología , Hipotálamo/fisiología , Animales , Antígenos CD36/genética , Ingestión de Alimentos , Emulsiones/administración & dosificación , Proteínas de Transporte de Ácidos Grasos/genética , Ácidos Grasos/sangre , Expresión Génica , Masculino , Modelos Biológicos , Fosfolípidos/administración & dosificación , Proteínas Proto-Oncogénicas c-fos/genética , Ratas , Aceite de Soja/administración & dosificación
10.
PLoS One ; 8(8): e72029, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23967273

RESUMEN

The hypothalamus plays a crucial role in the control of the energy balance and also retains neurogenic potential into adulthood. Recent studies have reported the severe alteration of the cell turn-over in the hypothalamus of obese animals and it has been proposed that a neurogenic deficiency in the hypothalamus could be involved in the development of obesity. To explore this possibility, we examined hypothalamic cell renewal during the homeostatic response to dietary fat in mice, i.e., at the onset of diet-induced obesity. We found that switching to high-fat diet (HFD) accelerated cell renewal in the hypothalamus through a local, rapid and transient increase in cell proliferation, peaking three days after introducing the HFD. Blocking HFD-induced cell proliferation by central delivery of an antimitotic drug prevented the food intake normalization observed after HFD introduction and accelerated the onset of obesity. This result showed that HFD-induced dividing brain cells supported an adaptive anorectic function. In addition, we found that the percentage of newly generated neurons adopting a POMC-phenotype in the arcuate nucleus was increased by HFD. This observation suggested that the maturation of neurons in feeding circuits was nutritionally regulated to adjust future energy intake. Taken together, these results showed that adult cerebral cell renewal was remarkably responsive to nutritional conditions. This constituted a physiological trait required to prevent severe weight gain under HFD. Hence this report highlighted the amazing plasticity of feeding circuits and brought new insights into our understanding of the nutritional regulation of the energy balance.


Asunto(s)
Hipotálamo/metabolismo , Neuronas/metabolismo , Obesidad/etiología , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/metabolismo , Astrocitos/metabolismo , Proliferación Celular , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Hipotálamo/citología , Masculino , Ratones , Proopiomelanocortina/metabolismo , Aumento de Peso
11.
PLoS One ; 8(7): e68709, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23894333

RESUMEN

AIMS: Hypoglycemia is a severe side effect of intensive insulin therapy. Recurrent hypoglycemia (RH) impairs the counter-regulatory response (CRR) which restores euglycemia. During hypoglycemia, ventromedial hypothalamus (VMH) production of nitric oxide (NO) and activation of its receptor soluble guanylyl cyclase (sGC) are critical for the CRR. Hypoglycemia also increases brain reactive oxygen species (ROS) production. NO production in the presence of ROS causes protein S-nitrosylation. S-nitrosylation of sGC impairs its function and induces desensitization to NO. We hypothesized that during hypoglycemia, the interaction between NO and ROS increases VMH sGC S-nitrosylation levels and impairs the CRR to subsequent episodes of hypoglycemia. VMH ROS production and S-nitrosylation were quantified following three consecutive daily episodes of insulin-hypoglycemia (RH model). The CRR was evaluated in rats in response to acute insulin-induced hypoglycemia or via hypoglycemic-hyperinsulinemic clamps. Pretreatment with the anti-oxidant N-acetyl-cysteine (NAC) was used to prevent increased VMH S-nitrosylation. RESULTS: Acute insulin-hypoglycemia increased VMH ROS levels by 49±6.3%. RH increased VMH sGC S-nitrosylation. Increasing VMH S-nitrosylation with intracerebroventricular injection of the nitrosylating agent S-nitroso-L-cysteine (CSNO) was associated with decreased glucagon secretion during hypoglycemic clamp. Finally, in RH rats pre-treated with NAC (0.5% in drinking water for 9 days) hypoglycemia-induced VMH ROS production was prevented and glucagon and epinephrine production was not blunted in response to subsequent insulin-hypoglycemia. CONCLUSION: These data suggest that NAC may be clinically useful in preventing impaired CRR in patients undergoing intensive-insulin therapy.


Asunto(s)
Hipoglucemia/metabolismo , Hipoglucemia/fisiopatología , Hipotálamo/metabolismo , Hipotálamo/fisiopatología , Acetilcisteína/administración & dosificación , Animales , Glucosa/metabolismo , Hipoglucemia/inducido químicamente , Hipotálamo/efectos de los fármacos , Insulina/efectos adversos , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo
12.
Neurosci Lett ; 534: 75-9, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23201632

RESUMEN

Hypothalamic detection of nutrients is involved in the control of energy metabolism and is altered in metabolic disorders. Although hypothalamic detection of blood lactate lowers hepatic glucose production and food intake, it is unknown whether it also modulates insulin secretion. To address this, a lactate injection via the right carotid artery (cephalad) was performed in Wistar rats. This triggered a transient increase in insulin secretion. Rats made hyperglycemic for 48h exhibited prolonged insulin secretion in response to a glucose injection via the carotid artery, but lactate injection induced two types of responses: half of the HG rats showed no difference compared to controls and the other half had markedly decreased insulin secretion. Astroglial monocarboxylates transporters MCT1 and MCT4 isoforms transfer lactate from blood to astrocytes and release lactate to the extracellular space, whilst the neuronal MCT2 isoform permits neuronal lactate uptake. We found that astroglial MCT1 and MCT4, and neuronal MCT2 protein levels in the medio-basal hypothalamus (MBH) were not modified by 48h-hyperglycemia. Together, these results indicate that hypothalamic sensing of circulating lactate triggers insulin secretion. Both glucose and lactate sensing are altered in a model of hyperglycemia, without alteration of MBH MCTs protein levels.


Asunto(s)
Glucosa/metabolismo , Hiperglucemia/metabolismo , Hipotálamo/metabolismo , Ácido Láctico/sangre , Animales , Glucosa/farmacología , Hipotálamo/irrigación sanguínea , Insulina/metabolismo , Secreción de Insulina , Ácido Láctico/farmacología , Masculino , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/metabolismo , Ratas , Ratas Wistar , Simportadores/metabolismo , Factores de Tiempo
13.
J Neurosci ; 32(48): 17097-107, 2012 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-23197703

RESUMEN

Leptin is an adipocyte-derived hormone that controls energy balance by acting primarily in the CNS, but its action is lost in common forms of obesity due to central leptin resistance. One potential mechanism for such leptin resistance is an increased hypothalamic expression of Suppressor of cytokine signaling 3 (Socs3), a feedback inhibitor of the Jak-Stat pathway that prevents Stat3 activation. Ample studies have confirmed the important role of Socs3 in leptin resistance and obesity. However, the degree to which Socs3 participates in the regulation of energy homeostasis in nonobese conditions remains largely undetermined. In this study, using adult mice maintained under standard diet, we demonstrate that Socs3 deficiency in the mediobasal hypothalamus (MBH) reduces food intake, protects against body weight gain, and limits adiposity, suggesting that Socs3 is necessary for normal body weight maintenance. Mechanistically, MBH Socs3-deficient mice display increased hindbrain sensitivity to endogenous, meal-related satiety signals, mediated by oxytocin signaling. Thus, oxytocin signaling likely mediates the effect of hypothalamic leptin on satiety circuits of the caudal brainstem. This provides an anatomical substrate for the effect of leptin on meal size, and more generally, a mechanism for how the brain controls short-term food intake as a function of the energetic stores available in the organism to maintain energy homeostasis. Any dysfunction in this pathway could potentially lead to overeating and obesity.


Asunto(s)
Hipotálamo/metabolismo , Oxitocina/metabolismo , Rombencéfalo/metabolismo , Respuesta de Saciedad/fisiología , Transducción de Señal/fisiología , Proteínas Supresoras de la Señalización de Citocinas/genética , Animales , Devazepida/farmacología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Antagonistas de Hormonas/farmacología , Hipotálamo/efectos de los fármacos , Leptina/metabolismo , Ratones , Receptores de Colecistoquinina/antagonistas & inhibidores , Rombencéfalo/efectos de los fármacos , Respuesta de Saciedad/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
14.
PLoS One ; 7(7): e39087, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22802935

RESUMEN

OBJECTIVE: This study aims at exploring the effects of sodium tungstate treatment on hypothalamic plasticity, which is known to have an important role in the control of energy metabolism. METHODS: Adult lean and high-fat diet-induced obese mice were orally treated with sodium tungstate. Arcuate and paraventricular nuclei and lateral hypothalamus were separated and subjected to proteomic analysis by DIGE and mass spectrometry. Immunohistochemistry and in vivo magnetic resonance imaging were also performed. RESULTS: Sodium tungstate treatment reduced body weight gain, food intake, and blood glucose and triglyceride levels. These effects were associated with transcriptional and functional changes in the hypothalamus. Proteomic analysis revealed that sodium tungstate modified the expression levels of proteins involved in cell morphology, axonal growth, and tissue remodeling, such as actin, CRMP2 and neurofilaments, and of proteins related to energy metabolism. Moreover, immunohistochemistry studies confirmed results for some targets and further revealed tungstate-dependent regulation of SNAP25 and HPC-1 proteins, suggesting an effect on synaptogenesis as well. Functional test for cell activity based on c-fos-positive cell counting also suggested that sodium tungstate modified hypothalamic basal activity. Finally, in vivo magnetic resonance imaging showed that tungstate treatment can affect neuronal organization in the hypothalamus. CONCLUSIONS: Altogether, these results suggest that sodium tungstate regulates proteins involved in axonal and glial plasticity. The fact that sodium tungstate could modulate hypothalamic plasticity and networks in adulthood makes it a possible and interesting therapeutic strategy not only for obesity management, but also for other neurodegenerative illnesses like Alzheimer's disease.


Asunto(s)
Axones/efectos de los fármacos , Hipotálamo/fisiología , Neuroglía/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Obesidad/tratamiento farmacológico , Compuestos de Tungsteno/uso terapéutico , Animales , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Proteínas del Tejido Nervioso/efectos de los fármacos , Procesamiento Proteico-Postraduccional
15.
Antioxid Redox Signal ; 14(3): 519-30, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20977349

RESUMEN

Mitochondrial reactive oxygen species (mROS) have emerged as signaling molecules in physiology primarily as a result of studies of uncoupling mechanisms in mitochondrial respiration. The discovery that this mechanism negatively regulates mROS generation in many cell types has drawn the attention of the scientific community to the pathological consequences of excess mROS production. From reports of the energetic fluxes in cells grown under normal conditions, the hypothesis that mROS are an integrated physiological signal of the metabolic status of the cell has emerged. Here, we consider recent studies that support this point of view in two key nutrient sensors of the body, beta cells and the hypothalamus, which are the main coordinators of endocrine and nervous controls of energy metabolism and adipose tissue, which is of paramount importance in controlling body weight and, therefore, the development of obesity and type 2 diabetes. In this context, finely balanced mROS production may be at the core of proper metabolic maintenance, and unbalanced mROS production, which is largely documented, might be an important trigger of metabolic disorders.


Asunto(s)
Mitocondrias/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Adipogénesis/fisiología , Animales , Glucemia/metabolismo , Humanos , Hipotálamo/metabolismo , Células Secretoras de Insulina/metabolismo , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Neuronas/metabolismo , Proteína Desacopladora 2
16.
Am J Physiol Endocrinol Metab ; 298(5): E1078-87, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20179244

RESUMEN

The sugar transporter GLUT2, present in several tissues of the gut-brain axis, has been reported to be involved in the control of food intake. GLUT2 is a sugar transporter sustaining energy production in the cell, but it can also function as a receptor for extracellular glucose. A glucose-signaling pathway is indeed triggered, independently of glucose metabolism, through its large cytoplasmic loop domain. However, the contribution of the receptor function over the transporter function of GLUT2 in the control of food intake remains to be determined. Thus, we generated transgenic mice that express a GLUT2-loop domain, blocking the detection of glucose but leaving GLUT2-dependent glucose transport unaffected. Inhibiting GLUT2-mediated glucose detection augmented daily food intake by a mechanism that increased the meal size but not the number of meals. Peripheral hormones (ghrelin, insulin, leptin) were unaffected, leading to a focus on central aspects of feeding behavior. We found defects in c-Fos activation by glucose in the arcuate nucleus and changes in the amounts of TRH and orexin neuropeptide mRNA, which are relevant to poorly controlled meal size. Our data provide evidence that glucose detection by GLUT2 contributes to the control of food intake by the hypothalamus. The sugar transporter receptor, i.e., "transceptor" GLUT2, may constitute a drug target to treat eating disorders and associated metabolic diseases, particularly by modulating its receptor function without affecting vital sugar provision by its transporter function.


Asunto(s)
Ingestión de Alimentos/fisiología , Transportador de Glucosa de Tipo 2/metabolismo , Glucosa/metabolismo , Hipotálamo/metabolismo , Análisis de Varianza , Animales , Transporte Biológico/fisiología , Peso Corporal/fisiología , Recuento de Células , Metabolismo Energético , Conducta Alimentaria/fisiología , Ghrelina/sangre , Transportador de Glucosa de Tipo 2/genética , Homeostasis/fisiología , Inmunohistoquímica , Insulina/sangre , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leptina/sangre , Ratones , Ratones Transgénicos , Neuropéptidos/genética , Neuropéptidos/metabolismo , Orexinas , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Estadísticas no Paramétricas , Hormona Liberadora de Tirotropina/genética , Hormona Liberadora de Tirotropina/metabolismo
17.
Diabetes ; 58(10): 2189-97, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19581415

RESUMEN

OBJECTIVE: Recent data demonstrated that glucose sensing in different tissues is initiated by an intracellular redox signaling pathway in physiological conditions. However, the relevance of such a mechanism in metabolic disease is not known. The aim of the present study was to determine whether brain glucose hypersensitivity present in obese Zücker rats is related to an alteration in redox signaling. RESEARCH DESIGN AND METHODS: Brain glucose sensing alteration was investigated in vivo through the evaluation of electrical activity in arcuate nucleus, changes in reactive oxygen species levels, and hypothalamic glucose-induced insulin secretion. In basal conditions, modifications of redox state and mitochondrial functions were assessed through oxidized glutathione, glutathione peroxidase, manganese superoxide dismutase, aconitase activities, and mitochondrial respiration. RESULTS: Hypothalamic hypersensitivity to glucose was characterized by enhanced electrical activity of the arcuate nucleus and increased insulin secretion at a low glucose concentration, which does not produce such an effect in normal rats. It was associated with 1) increased reactive oxygen species levels in response to this low glucose load, 2) constitutive oxidized environment coupled with lower antioxidant enzyme activity at both the cellular and mitochondrial level, and 3) overexpression of several mitochondrial subunits of the respiratory chain coupled with a global dysfunction in mitochondrial activity. Moreover, pharmacological restoration of the glutathione hypothalamic redox state by reduced glutathione infusion in the third ventricle fully reversed the cerebral hypersensitivity to glucose. CONCLUSIONS: The data demonstrated that obese Zücker rats' impaired hypothalamic regulation in terms of glucose sensing is linked to an abnormal redox signaling, which originates from mitochondria dysfunction.


Asunto(s)
Glucosa/farmacología , Hipotálamo/fisiopatología , Obesidad/fisiopatología , Aconitato Hidratasa/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Encéfalo/fisiopatología , Homeostasis , Hipersensibilidad/metabolismo , Hipersensibilidad/fisiopatología , Hipotálamo/efectos de los fármacos , Hipotálamo/fisiología , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Obesidad/genética , Oxidación-Reducción , Fosforilación Oxidativa/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Ratas , Ratas Zucker , Transducción de Señal
18.
Diabetes ; 58(7): 1544-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19389827

RESUMEN

OBJECTIVE: Insulin plays an important role in the hypothalamic control of energy balance, especially by reducing food intake. Emerging data point to a pivotal role of reactive oxygen species (ROS) in energy homeostasis regulation, but their involvement in the anorexigenic effect of insulin is unknown. Furthermore, ROS signal derived from NADPH oxidase activation is required for physiological insulin effects in peripheral cells. In this study, we investigated the involvement of hypothalamic ROS and NADPH oxidase in the feeding behavior regulation by insulin. RESEARCH DESIGN AND METHODS: We first measured hypothalamic ROS levels and food intake after acute intracerebroventricular injection of insulin. Second, effect of pretreatment with a ROS scavenger or an NADPH oxidase inhibitor was evaluated. Third, we examined the consequences of two nutritional conditions of central insulin unresponsiveness (fasting or short-term high-fat diet) on the ability of insulin to modify ROS level and food intake. RESULTS: In normal chow-fed mice, insulin inhibited food intake. At the same dose, insulin rapidly and transiently increased hypothalamic ROS levels by 36%. The pharmacological suppression of this insulin-stimulated ROS elevation, either by antioxidant or by an NADPH oxidase inhibitor, abolished the anorexigenic effect of insulin. Finally, in fasted and short-term high-fat diet-fed mice, insulin did not promote elevation of ROS level and food intake inhibition, likely because of an increase in hypothalamic diet-induced antioxidant defense systems. CONCLUSIONS: A hypothalamic ROS increase through NADPH oxidase is required for the anorexigenic effect of insulin.


Asunto(s)
Ventrículos Cerebrales/fisiología , Ingestión de Energía/fisiología , Hipotálamo/fisiología , Insulina/farmacología , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Glucemia/metabolismo , Ventrículos Cerebrales/efectos de los fármacos , Ingestión de Energía/efectos de los fármacos , Metabolismo Energético , Glutatión/metabolismo , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Insulina/administración & dosificación , Insulina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL
19.
J Neurosci Methods ; 178(2): 301-7, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19150628

RESUMEN

Different roles of mitochondria in brain function according to brain area are now clearly emerging. Unfortunately, no technique is yet described to investigate mitochondria function in specific brain area. In this article, we provide a complete description of a procedure to analyze the mitochondrial function in rat brain biopsies. Our two-step method consists in a saponin permeabilization of fresh brain tissues in combination with high-resolution respirometry to acquire the integrated respiratory rate of the biopsy. In the first part, we carefully checked the mitochondria integrity after permeabilization, defined experimental conditions to determine the respiratory control ratio (RCR), and tested the reproducibility of this technique. In the second part, we applied our method to test its sensitivity. As a result, this method was sensitive enough to reveal region specificity of mitochondrial respiration within the brain. Moreover, we detected physiopathological modulation of the mitochondrial function in the hypothalamus. Thus this new technique that takes all cell types into account, and does not discard or select any mitochondria sub-population is very suitable to analyze the integrated mitochondrial respiration of brain biopsies.


Asunto(s)
Técnicas de Laboratorio Clínico , Hipotálamo/fisiología , Mitocondrias/fisiología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Respiración de la Célula , Ayuno/fisiología , Hiperglucemia/fisiopatología , Hipotálamo/efectos de los fármacos , Hipotálamo/ultraestructura , Masculino , Microscopía Electrónica , Mitocondrias/ultraestructura , Consumo de Oxígeno , Permeabilidad/efectos de los fármacos , Fosforilación , Ratas , Ratas Wistar , Ratas Zucker , Saponinas/farmacología , Estrés Fisiológico
20.
Diabetes ; 58(3): 673-81, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19073765

RESUMEN

OBJECTIVE: Insulin secretion involves complex events in which the mitochondria play a pivotal role in the generation of signals that couple glucose detection to insulin secretion. Studies on the mitochondrial generation of reactive oxygen species (ROS) generally focus on chronic nutrient exposure. Here, we investigate whether transient mitochondrial ROS production linked to glucose-induced increased respiration might act as a signal for monitoring insulin secretion. RESEARCH DESIGN AND METHODS: ROS production in response to glucose was investigated in freshly isolated rat islets. ROS effects were studied using a pharmacological approach and calcium imaging. RESULTS: Transient glucose increase from 5.5 to 16.7 mmol/l stimulated ROS generation, which was reversed by antioxidants. Insulin secretion was dose dependently blunted by antioxidants and highly correlated with ROS levels. The incapacity of beta-cells to secrete insulin in response to glucose with antioxidants was associated with a decrease in ROS production and in contrast to the maintenance of high levels of ATP and NADH. Then, we investigated the mitochondrial origin of ROS (mROS) as the triggering signal. Insulin release was mimicked by the mitochondrial-complex blockers, antimycin and rotenone, that generate mROS. The adding of antioxidants to mitochondrial blockers or to glucose was used to lower mROS reversed insulin secretion. Finally, calcium imaging on perifused islets using glucose stimulation or mitochondrial blockers revealed that calcium mobilization was completely reversed using the antioxidant trolox and that it was of extracellular origin. No toxic effects were present using these pharmacological approaches. CONCLUSIONS: Altogether, these complementary results demonstrate that mROS production is a necessary stimulus for glucose-induced insulin secretion.


Asunto(s)
Glucosa/farmacología , Insulina/metabolismo , Islotes Pancreáticos/fisiología , Mitocondrias/fisiología , Especies Reactivas de Oxígeno/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Cromanos/farmacología , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Cinética , Masculino , Mitocondrias/efectos de los fármacos , NAD/metabolismo , Ratas , Ratas Wistar , Transducción de Señal , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Tapsigargina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA