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










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38798316

RESUMEN

Glucose-inhibited (GI) neurons of the ventromedial hypothalamus (VMH) depend on neuronal nitric oxide synthase (nNOS) and AMP-activated protein kinase (AMPK) for activation in low glucose. The Lopez laboratory has shown that the effects of estrogen on brown fat thermogenesis and white fat browning require inhibition of VMH AMPK. This effect of estrogen was mediated by downstream lateral hypothalamus (LH) orexin neurons 1,2 . We previously showed that estrogen inhibits activation of GI neurons in low glucose by inhibiting AMPK 3 . Thus, we hypothesized that VMH AMPK- and nNOS-dependent GI neurons project to and inhibit orexin neurons. Estrogen inhibition of AMPK in GI neurons would then disinhibit orexin neurons and stimulate brown fat thermogenesis and white fat browning, leading to decreased body weight. To test this hypothesis, we reduced VMH nNOS expression using nNOS shRNA in female mice and measured body weight, adiposity, body temperature, white and brown fat uncoupling protein (UCP1; an index of thermogenesis and browning), locomotor activity, and blood glucose levels. Surprisingly, we saw no effect of reduced VMH nNOS expression on body temperature or UCP1. Instead, body weight and adiposity increased by 30% over 2 weeks post injection of nNOS shRNA. This was associated with increased blood glucose levels and decreased locomotor activity. We also found that VMH nNOS-GI neurons project to the LH. However, stimulation of VMH-LH projections increased excitatory glutamate input onto orexin neurons. Thus, our data do not support our original hypothesis. Excitation of orexin neurons has previously been shown to increase physical activity, leading to decreased blood glucose and body weight 4 . We now hypothesize that VMH nNOS-GI neurons play a role in this latter function of orexin neurons.

2.
J Neuroendocrinol ; 35(12): e13344, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37857383

RESUMEN

OBJECTIVES: Intensive insulin therapy provides optimal glycemic control in patients with diabetes. However, intensive insulin therapy causes so-called iatrogenic hypoglycemia as a major adverse effect. The ventromedial hypothalamus (VMH) has been described as the primary brain area initiating the counter-regulatory response (CRR). Nevertheless, the VMH receives projections from other brain areas which could participate in the regulation of the CRR. In particular, studies suggest a potential role of the serotonin (5-HT) network. Thus, the objective of this study was to determine the contribution of 5-HT neurons in CRR control. METHODS: Complementary approaches have been used to test this hypothesis in quantifying the level of 5-HT in several brain areas by HPLC in response to insulin-induced hypoglycemia, measuring the electrical activity of dorsal raphe (DR) 5-HT neurons in response to insulin or decreased glucose level by patch-clamp electrophysiology; and measuring the CRR hormone glucagon as an index of the CRR to the modulation of the activity of 5-HT neurons using pharmacological or pharmacogenetic approaches. RESULTS: HPLC measurements show that the 5HIAA/5HT ratio is increased in several brain regions including the VMH in response to insulin-induced hypoglycemia. Patch-clamp electrophysiological recordings show that insulin, but not decreased glucose level, increases the firing frequency of DR 5-HT neurons in the DR. In vivo, both the pharmacological inhibition of 5-HT neurons by intraperitoneal injection of the 5-HT1A receptor agonist 8-OH-DPAT or the chemogenetic inhibition of these neurons reduce glucagon secretion, suggesting an impaired CRR. CONCLUSION: Taken together, these data highlight a new neuronal network involved in the regulation of the CRR. In particular, this study shows that DR 5-HT neurons detect iatrogenic hypoglycemia in response to the increased insulin level and may play an important role in the regulation of CRR.


Asunto(s)
Glucagón , Hipoglucemia , Humanos , Neuronas Serotoninérgicas , Serotonina/farmacología , Hipoglucemia/inducido químicamente , Insulina/farmacología , Glucosa , Enfermedad Iatrogénica
3.
Mol Metab ; 76: 101788, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37536499

RESUMEN

OBJECTIVE: The present study tests the hypothesis that changes in the glucose sensitivity of lateral hypothalamus (LH) hypocretin/orexin glucose-inhibited (GI) neurons following weight loss leads to glutamate plasticity on ventral tegmental area (VTA) dopamine neurons and drives food seeking behavior. METHODS: C57BL/6J mice were calorie restricted to a 15% body weight loss and maintained at that body weight for 1 week. The glucose sensitivity of LH hypocretin/orexin GI and VTA dopamine neurons was measured using whole cell patch clamp recordings in brain slices. Food seeking behavior was assessed using conditioned place preference (CPP). RESULTS: 1-week maintenance of calorie restricted 15% body weight loss reduced glucose inhibition of hypocretin/orexin GI neurons resulting in increased neuronal activation with reduced glycemia. The effect of decreased glucose on hypocretin/orexin GI neuronal activation was blocked by pertussis toxin (inhibitor of G-protein coupled receptor subunit Gαi/o) and Rp-cAMP (inhibitor of protein kinase A, PKA). This suggests that glucose sensitivity is mediated by the Gαi/o-adenylyl cyclase-cAMP-PKA signaling pathway. The excitatory effect of the hunger hormone, ghrelin, on hcrt/ox neurons was also blocked by Rp-cAMP suggesting that hormonal signals of metabolic status may converge on the glucose sensing pathway. Food restriction and weight loss increased glutamate synaptic strength (indexed by increased AMPA/NMDA receptor current ratio) on VTA dopamine neurons and the motivation to seek food (indexed by CPP). Chemogenetic inhibition of hypocretin/orexin neurons during caloric restriction and weight loss prevented these changes in glutamate plasticity and food seeking behavior. CONCLUSIONS: We hypothesize that this change in the glucose sensitivity of hypocretin/orexin GI neurons may drive, in part, food seeking behavior following caloric restriction.


Asunto(s)
Área Hipotalámica Lateral , Neuropéptidos , Ratones , Animales , Orexinas/metabolismo , Área Hipotalámica Lateral/metabolismo , Neuropéptidos/metabolismo , Restricción Calórica , Glucosa/metabolismo , Ratones Endogámicos C57BL , Neuronas Dopaminérgicas/metabolismo , Glutamatos/metabolismo , Glutamatos/farmacología
4.
Appetite ; 189: 106621, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37311483

RESUMEN

Orexin neurons in the Lateral Hypothalamus (LH) play an important role in food seeking behavior. Approximately 60 percent of LH orexin neurons are inhibited by elevated extracellular glucose. It has been shown that elevated LH glucose decreases conditioned place preference for a food associated chamber. However, it has never been shown how modulation of LH extracellular glucose effects a rat's motivation to work for food. In this experiment we used reverse microdialysis to modulate extracellular glucose levels in LH during an operant task. Results from a progressive ratio task demonstrated that 4 mM glucose perfusion significantly decreased the animal's motivation to work for sucrose pellets while not effecting the hedonic value of the pellets. In a second experiment we demonstrated that 4 mM but not 2.5 mM glucose perfusion was sufficient to significantly decrease the number of sucrose pellets earned. Finally, we showed that modulating LH extracellular glucose mid-session from 0.7 mM to 4 mM did not affect behavior. This indicates that once feeding behavior has begun the animal becomes unresponsive to changes in extracellular glucose levels in LH. Taken together these experiments indicate that LH glucose sensing neurons play an important role in motivation to initiate feeding. However, once consumption has begun it is likely that feeding is controlled by brain regions downstream of LH.


Asunto(s)
Área Hipotalámica Lateral , Sacarosa , Ratas , Animales , Área Hipotalámica Lateral/metabolismo , Orexinas/metabolismo , Orexinas/farmacología , Sacarosa/farmacología , Recompensa , Conducta Alimentaria/fisiología
5.
Diabetes ; 72(8): 1144-1153, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-36525384

RESUMEN

Perifornical hypothalamus (PFH) orexin glucose-inhibited (GI) neurons that facilitate arousal have been implicated in hypoglycemia awareness. Mice lacking orexin exhibit narcolepsy, and orexin mediates the effect of the antinarcolepsy drug modafinil. Thus, hypoglycemia awareness may require a certain level of arousal for awareness of the sympathetic symptoms of hypoglycemia (e.g., tremors, anxiety). Recurrent hypoglycemia (RH) causes hypoglycemia unawareness. We hypothesize that RH impairs the glucose sensitivity of PFH orexin GI neurons and that modafinil normalizes glucose sensitivity of these neurons and restores hypoglycemia awareness after RH. Using patch-clamp recording, we found that RH enhanced glucose inhibition of PFH orexin GI neurons in male mice, thereby blunting activation of these neurons in low-glucose conditions. We then used a modified conditioned place preference behavioral test to demonstrate that modafinil reversed hypoglycemia unawareness in male mice after RH. Similarly, modafinil restored normal glucose sensitivity to PFH orexin GI neurons. We conclude that impaired glucose sensitivity of PFH orexin GI neurons plays a role in hypoglycemia unawareness and that normalizing their glucose sensitivity after RH is associated with restoration of hypoglycemia awareness. This suggests that the glucose sensitivity of PFH orexin GI neurons is a therapeutic target for preventing hypoglycemia unawareness.


Asunto(s)
Complicaciones de la Diabetes , Hipoglucemia , Ratones , Masculino , Animales , Orexinas/farmacología , Modafinilo/farmacología , Hipoglucemia/tratamiento farmacológico , Glucosa/farmacología , Neuronas
6.
J Neuroendocrinol ; 33(4): e12937, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33507001

RESUMEN

Subsequent to the discovery of insulin 100 years ago, great strides have been made in understanding its function, especially in the brain. It is now clear that insulin is a critical regulator of the neuronal circuitry controlling energy balance and glucose homeostasis. This review focuses on the effects of insulin and diabetes on the activity and glucose sensitivity of hypothalamic glucose-sensing neurones. We highlight the role of electrophysiological data in understanding how insulin regulates glucose-sensing neurones. A brief introduction describing the benefits and limitations of the major electrophysiological techniques used to investigate glucose-sensing neurones is provided. The mechanisms by which hypothalamic neurones sense glucose are discussed with an emphasis on those glucose-sensing neurones already shown to be modulated by insulin. Next, the literature pertaining to how insulin alters the activity and glucose sensitivity of these hypothalamic glucose-sensing neurones is described. In addition, the effects of impaired insulin signalling during diabetes and the ramifications of insulin-induced hypoglycaemia on hypothalamic glucose-sensing neurones are covered. To the extent that it is known, we present hypotheses concerning the mechanisms underlying the effects of these insulin-related pathologies. To conclude, electrophysiological data from the hippocampus are evaluated aiming to provide clues regarding how insulin might influence neuronal plasticity in glucose-sensing neurones. Although much has been accomplished subsequent to the discovery of insulin, the work described in our review suggests that the regulation of central glucose sensing by this hormone is both important and understudied.


Asunto(s)
Glucosa/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Neuronas/metabolismo , Animales , Humanos , Hipotálamo/efectos de los fármacos , Insulina/farmacología , Neuronas/efectos de los fármacos
7.
J Neuroendocrinol ; 32(1): e12773, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31329314

RESUMEN

The ventromedial hypothalamus (VMH) plays a complex role in glucose and energy homeostasis. The VMH is necessary for the counter-regulatory response to hypoglycaemia (CRR) that increases hepatic gluconeogenesis to restore euglycaemia. On the other hand, the VMH also restrains hepatic glucose production during euglycaemia and stimulates peripheral glucose uptake. The VMH is also important for the ability of oestrogen to increase energy expenditure. This latter function is mediated by VMH modulation of the lateral/perifornical hypothalamic area (lateral/perifornical hypothalamus) orexin neurones. Activation of VMH AMP-activated protein kinase (AMPK) is necessary for the CRR. By contrast, VMH AMPK inhibition favours decreased basal glucose levels and is required for oestrogen to increase energy expenditure. Specialised VMH glucose-sensing neurones confer the ability to sense and respond to changes in blood glucose levels. Glucose-excited (GE) neurones increase and glucose-inhibited (GI) neurones decrease their activity as glucose levels rise. VMH GI neurones, in particular, appear to be important in the CRR, although a role for GE neurones cannot be discounted. AMPK mediates glucose sensing in VMH GI neurones suggesting that, although activation of these neurones is important for the CRR, it is necessary to silence them to lower basal glucose levels and enable oestrogen to increase energy expenditure. In support of this, we found that oestrogen reduces activation of VMH GI neurones in low glucose by inhibiting AMPK. In this review, we present the evidence underlying the role of the VMH in glucose and energy homeostasis. We then discuss the role of VMH glucose-sensing neurones in mediating these effects, with a strong emphasis on oestrogenic regulation of glucose sensing and how this may affect glucose and energy homeostasis.


Asunto(s)
Glucemia/metabolismo , Metabolismo Energético/fisiología , Homeostasis/fisiología , Neuronas/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Humanos
8.
Brain Res ; 1731: 145808, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29787770

RESUMEN

Glucose inhibits ∼60% of lateral hypothalamic (LH) orexin neurons. Fasting increases the activation of LH orexin glucose-inhibited (GI) neurons in low glucose. Increases in spontaneous glutamate excitatory postsynaptic currents (sEPSCs) onto putative VTA DA neurons in low glucose are orexin dependent (Sheng et al., 2014). VTA DA neurons modulate reward-based feeding. We tested the hypothesis that increased activation of LH orexin-GI neurons in low glucose increases glutamate signaling onto VTA DA neurons and contributes to reward-based feeding in food restricted animals. N-methyl-D-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) currents on putative VTA DA neurons were measured using whole cell voltage clamp recording in horizontal brain slices containing the LH and VTA. Decreased glucose increased the NMDA receptor current for at least one hour after returning glucose to basal levels (P < 0.05; N = 8). The increased current was blocked by an orexin 1 receptor antagonist (P < 0.05; N = 5). Low glucose caused a similar persistent enhancement of AMPA receptor currents (P < 0.05; N = 7). An overnight fast increased the AMPA/NMDA receptor current ratio, an in vivo index of glutamate plasticity, on putative VTA DA neurons. Conditioned place preference (CPP) for palatable food was measured during LH dialysis with glucose. CPP score was negatively correlated with increasing LH glucose (P < 0.05; N = 20). These data suggest that increased activation of LH orexin-GI neurons in low glucose after weight loss, leads to enhanced glutamate signaling on VTA DA neurons, increases the drive to eat rewarding food, and may contribute to weight regain.


Asunto(s)
Conducta Alimentaria/fisiología , Ácido Glutámico/fisiología , Área Hipotalámica Lateral/fisiología , Neuronas/fisiología , Recompensa , Transmisión Sináptica , Área Tegmental Ventral/fisiología , Animales , Glucosa/administración & dosificación , Glucosa/fisiología , Masculino , Ratones Endogámicos C57BL , Orexinas/fisiología , Ratas Sprague-Dawley , Receptores AMPA/fisiología , Receptores de N-Metil-D-Aspartato/fisiología
9.
Front Physiol ; 9: 192, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29593556

RESUMEN

Hypoglycemia is a profound threat to the brain since glucose is its primary fuel. As a result, glucose sensors are widely located in the central nervous system and periphery. In this perspective we will focus on the role of hypothalamic glucose-inhibited (GI) neurons in sensing and correcting hypoglycemia. In particular, we will discuss GI neurons in the ventromedial hypothalamus (VMH) which express neuronal nitric oxide synthase (nNOS) and in the perifornical hypothalamus (PFH) which express orexin. The ability of VMH nNOS-GI neurons to depolarize in low glucose closely parallels the hormonal response to hypoglycemia which stimulates gluconeogenesis. We have found that nitric oxide (NO) production in low glucose is dependent on oxidative status. In this perspective we will discuss the potential relevance of our work showing that enhancing the glutathione antioxidant system prevents hypoglycemia associated autonomic failure (HAAF) in non-diabetic rats whereas VMH overexpression of the thioredoxin antioxidant system restores hypoglycemia counterregulation in rats with type 1 diabetes.We will also address the potential role of the orexin-GI neurons in the arousal response needed for hypoglycemia awareness which leads to behavioral correction (e.g., food intake, glucose administration). The potential relationship between the hypothalamic sensors and the neurocircuitry in the hindbrain and portal mesenteric vein which is critical for hypoglycemia correction will then be discussed.

10.
Neuropsychopharmacology ; 43(3): 607-616, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28857071

RESUMEN

Long-chain fatty acids (FAs) act centrally to decrease food intake and hepatic glucose production and alter hypothalamic neuronal activity in a manner that depends on FA type and cellular transport proteins. However, it is not known whether FAs are sensed by ventral tegmental area (VTA) dopamine (DA) neurons to control food-motivated behavior and DA neurotransmission. We investigated the impact of the monounsaturated FA oleate in the VTA on feeding, locomotion, food reward, and DA neuronal activity and DA neuron expression of FA-handling proteins and FA uptake. A single intra-VTA injection of oleate, but not of the saturated FA palmitate, decreased food intake and increased locomotor activity. Furthermore, intra-VTA oleate blunted the rewarding effects of high-fat/sugar food in an operant task and inhibited DA neuronal firing. Using sorted DA neuron preparations from TH-eGFP mice we found that DA neurons express FA transporter and binding proteins, and are capable of intracellular transport of long-chain FA. Finally, we demonstrate that a transporter blocker attenuates FA uptake into DA neurons and blocks the effects of intra-VTA oleate to decrease food-seeking and DA neuronal activity. Together, these results suggest that DA neurons detect FA and that oleate has actions in the VTA to suppress DA neuronal activity and food seeking following cellular incorporation. These findings highlight the capacity of DA neurons to act as metabolic sensors by responding not only to hormones but also to FA nutrient signals to modulate food-directed behavior.


Asunto(s)
Dopamina/metabolismo , Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Ácido Oléico/metabolismo , Recompensa , Área Tegmental Ventral/metabolismo , Animales , Conducta Apetitiva/fisiología , Células Cultivadas , Condicionamiento Operante/fisiología , Neuronas Dopaminérgicas/metabolismo , Ingestión de Alimentos/psicología , Conducta Alimentaria/psicología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/fisiología , Ratas Wistar
11.
Diabetes ; 67(1): 120-130, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29079703

RESUMEN

We previously showed that the glutathione precursor, N-acetylcysteine (NAC), prevented hypoglycemia-associated autonomic failure (HAAF) and impaired activation of ventromedial hypothalamus (VMH) glucose-inhibited (GI) neurons by low glucose after recurrent hypoglycemia (RH) in nondiabetic rats. However, NAC does not normalize glucose sensing by VMH GI neurons when RH occurs during diabetes. We hypothesized that recruiting the thioredoxin (Trx) antioxidant defense system would prevent HAAF and normalize glucose sensing after RH in diabetes. To test this hypothesis, we overexpressed Trx-1 (cytosolic form of Trx) in the VMH of rats with streptozotocin (STZ)-induced type 1 diabetes. The counterregulatory response (CRR) to hypoglycemia in vivo and the activation of VMH GI neurons in low glucose using membrane potential sensitive dye in vitro was measured before and after RH. VMH Trx-1 overexpression normalized both the CRR and glucose sensing by VMH GI neurons in STZ rats. VMH Trx-1 overexpression also lowered the insulin requirement to prevent severe hyperglycemia in STZ rats. However, like NAC, VMH Trx-1 overexpression did not prevent HAAF or normalize activation of VMH GI neurons by low glucose in STZ rats after RH. We conclude that preventing HAAF in type 1 diabetes may require the recruitment of both antioxidant systems.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Hipoglucemia/metabolismo , Hipotálamo/metabolismo , Tiorredoxinas/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/genética , Glucosa/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Tiorredoxinas/genética , Núcleo Hipotalámico Ventromedial/efectos de los fármacos
12.
Diabetes ; 66(3): 587-597, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27797912

RESUMEN

GLUT4 in muscle and adipose tissue is important in maintaining glucose homeostasis. However, the role of insulin-responsive GLUT4 in the central nervous system has not been well characterized. To assess its importance, a selective knockout of brain GLUT4 (BG4KO) was generated by crossing Nestin-Cre mice with GLUT4-floxed mice. BG4KO mice had a 99% reduction in GLUT4 protein expression throughout the brain. Despite normal feeding and fasting glycemia, BG4KO mice were glucose intolerant, demonstrated hepatic insulin resistance, and had reduced glucose uptake in the brain. In response to hypoglycemia, BG4KO mice had impaired glucose sensing, noted by impaired epinephrine and glucagon responses and impaired c-fos activation in the hypothalamic paraventricular nucleus. Moreover, in vitro glucose sensing of glucose-inhibitory neurons from the ventromedial hypothalamus was impaired in BG4KO mice. In summary, BG4KO mice are glucose intolerant, insulin resistant, and have impaired glucose sensing, indicating a critical role for brain GLUT4 in sensing and responding to changes in blood glucose.


Asunto(s)
Glucemia/metabolismo , Encéfalo/metabolismo , Intolerancia a la Glucosa/genética , Hipoglucemia/genética , Resistencia a la Insulina/genética , Animales , Western Blotting , Dieta Alta en Grasa , Epinefrina/metabolismo , Glucagón/metabolismo , Glucosa/metabolismo , Técnica de Clampeo de la Glucosa , Prueba de Tolerancia a la Glucosa , Transportador de Glucosa de Tipo 4 , Homeostasis/genética , Hipotálamo/citología , Hipotálamo/metabolismo , Técnicas In Vitro , Indinavir/farmacología , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley
13.
J Neurosci ; 36(45): 11469-11481, 2016 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-27911750

RESUMEN

Signals of energy homeostasis interact closely with neural circuits of motivation to control food intake. An emerging hypothesis is that the transition to maladaptive feeding behavior seen in eating disorders or obesity may arise from dysregulation of these interactions. Focusing on key brain regions involved in the control of food intake (ventral tegmental area, striatum, hypothalamus, and thalamus), we describe how activity of specific cell types embedded within these regions can influence distinct components of motivated feeding behavior. We review how signals of energy homeostasis interact with these regions to influence motivated behavioral output and present evidence that experience-dependent neural adaptations in key feeding circuits may represent cellular correlates of impaired food intake control. Future research into mechanisms that restore the balance of control between signals of homeostasis and motivated feeding behavior may inspire new treatment options for eating disorders and obesity.


Asunto(s)
Regulación del Apetito/fisiología , Encéfalo/fisiología , Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Homeostasis/fisiología , Motivación/fisiología , Animales , Peso Corporal/fisiología , Humanos
14.
Mol Metab ; 5(10): 823-833, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27688996

RESUMEN

OBJECTIVE: Brain regulation of glucose homeostasis is sexually dimorphic; however, the impact sex hormones have on specific neuronal populations within the ventromedial hypothalamic nucleus (VMN), a metabolically sensitive brain region, has yet to be fully characterized. Glucose-excited (GE) and -inhibited (GI) neurons are located throughout the VMN and may play a critical role in glucose and energy homeostasis. Within the ventrolateral portion of the VMN (VL-VMN), glucose sensing neurons and estrogen receptor (ER) distributions overlap. We therefore tested the hypothesis that VL-VMN glucose sensing neurons were sexually dimorphic and regulated by 17ß-estradiol (17ßE). METHODS: Electrophysiological recordings of VL-VMN glucose sensing neurons in brain slices isolated from age- and weight-matched female and male mice were performed in the presence and absence of 17ßE. RESULTS: We found a new class of VL-VMN GI neurons whose response to low glucose was transient despite continued exposure to low glucose. Heretofore, we refer to these newly identified VL-VMN GI neurons as 'adapting' or AdGI neurons. We found a sexual dimorphic response to low glucose, with male nonadapting GI neurons, but not AdGI neurons, responding more robustly to low glucose than those from females. 17ßE blunted the response of both nonadapting GI and AdGI neurons to low glucose in both males and females, which was mediated by activation of estrogen receptor ß and inhibition of AMP-activated kinase. In contrast, 17ßE had no impact on GE or non-glucose sensing neurons in either sex. CONCLUSION: These data suggest sex differences and estrogenic regulation of VMN hypothalamic glucose sensing may contribute to the sexual dimorphism in glucose homeostasis.

15.
Physiol Behav ; 167: 248-254, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27666162

RESUMEN

OBJECTIVE: 17ß-Estradiol (17ßE) regulates glucose homeostasis in part by centrally mediated mechanisms. In female rodents, the influence of the ovarian cycle on hypoglycemia counterregulation and glucose tolerance is unclear. We found previously that in prepubertal females, 17ßE modulates glucose sensing in nonadapting glucose-inhibited (GI) and adapting GI (AdGI) neurons within the ventrolateral portion of the ventromedial nucleus (VL-VMN). Nonadapting GI neurons persistently decrease their activity as glucose increases while AdGI neurons transiently respond to a glucose increase. To begin to understand if endogenous fluctuations in estrogen levels across the estrous cycle impact hypothalamic glucose sensing and glucose homeostasis, we assessed whether hypoglycemia counterregulation and glucose tolerance differed across the phases of the estrous cycle. We hypothesized that the response to insulin-induced hypoglycemia (IIH) and/or glucose tolerance would vary throughout the estrous cycle according to changes in 17ßE availability. Moreover, that these changes would correlate with estrous-dependent changes in the glucose sensitivity of VL-VMN glucose-sensing neurons (GSNs). METHODS: These hypotheses were tested in female mice by measuring the response to IIH, glucose tolerance and the glucose sensitivity of VL-VMN GSNs during each phase of the estrous cycle. Furthermore, a physiological brain concentration of 17ßE seen during proestrus was acutely applied to brain slices isolated on the day of diestrous and the response to low glucose in VL-VMN GSNs was assayed. RESULTS: The response to IIH was strongest during diestrous. The response of nonadapting GI and AdGI neurons to a glucose decrease from 2.5 to 0.5mM also peaked during diestrous; an effect which was blunted by the addition of 17ßE. In contrast, the glucose sensitivity of the subpopulation of GSNs which are excited by glucose (GE) was not affected by estrous phase or exogenous 17ßE application. CONCLUSION: These data suggest that physiological fluctuations in circulating 17ßE levels across the estrous cycle lead to changes in hypothalamic glucose sensing and the response to IIH.


Asunto(s)
Ciclo Estral/fisiología , Homeostasis/fisiología , Neuronas/fisiología , Núcleo Hipotalámico Ventromedial/citología , Núcleo Hipotalámico Ventromedial/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Análisis de Varianza , Animales , Glucemia/efectos de los fármacos , Glucemia/fisiología , Estradiol/farmacología , Ciclo Estral/efectos de los fármacos , Ayuno , Femenino , Glucosa/farmacología , Prueba de Tolerancia a la Glucosa , Homeostasis/efectos de los fármacos , Técnicas In Vitro , Insulina/farmacología , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Núcleo Hipotalámico Ventromedial/efectos de los fármacos
16.
Brain Res ; 1648(Pt A): 181-192, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27473896

RESUMEN

A population of Neuropeptide Y (NPY) neurons which co-express Agouti-related peptide (AgRP) in the arcuate nucleus of the hypothalamus (ARC) are inhibited at physiological levels of brain glucose and activated when glucose levels decline (e.g. glucose-inhibited or GI neurons). Fasting enhances the activation of NPY/AgRP-GI neurons by low glucose. In the present study we tested the hypothesis that lipopolysaccharide (LPS) inhibits the enhanced activation of NPY/AgRP-GI neurons by low glucose following a fast. Mice which express green fluorescent protein (GFP) on their NPY promoter were used to identify NPY/AgRP neurons. Fasting for 24h and LPS injection decreased blood glucose levels. As we have found previously, fasting increased c-fos expression in NPY/AgRP neurons and increased the activation of NPY/AgRP-GI neurons by decreased glucose. As we predicted, LPS blunted these effects of fasting at the 24h time point. Moreover, the inflammatory cytokine tumor necrosis factor alpha (TNFα) blocked the activation of NPY/AgRP-GI neurons by decreased glucose. These data suggest that LPS and TNFα may alter glucose and energy homeostasis, in part, due to changes in the glucose sensitivity of NPY/AgRP neurons. Interestingly, our findings also suggest that NPY/AgRP-GI neurons use a distinct mechanism to sense changes in extracellular glucose as compared to our previous studies of GI neurons in the adjacent ventromedial hypothalamic nucleus.


Asunto(s)
Glucosa/metabolismo , Lipopolisacáridos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína Relacionada con Agouti/antagonistas & inhibidores , Proteína Relacionada con Agouti/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Ayuno , Homeostasis , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leptina/farmacología , Lipopolisacáridos/farmacología , Lipopolisacáridos/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuropéptido Y/antagonistas & inhibidores , Neuropéptido Y/metabolismo , Factor de Necrosis Tumoral alfa/uso terapéutico
17.
Diab Vasc Dis Res ; 12(1): 2-12, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25268022

RESUMEN

In Type 1 and advanced Type 2 diabetes mellitus, elevation of plasma epinephrine plays a key role in normalizing plasma glucose during hypoglycaemia. However, recurrent hypoglycaemia blunts this elevation of plasma epinephrine. To determine whether recurrent hypoglycaemia affects peripheral components of the sympatho-adrenal system responsible for epinephrine release, male rats were administered subcutaneous insulin daily for 3 days. These recurrent hypoglycaemic animals showed a smaller elevation of plasma epinephrine than saline-injected controls when subjected to insulin-induced hypoglycaemia. Electrical stimulation of an adrenal branch of the splanchnic nerve in recurrent hypoglycaemic animals elicited less release of epinephrine and norepinephrine than in controls, without a change in adrenal catecholamine content. Responsiveness of isolated, perfused adrenal glands to acetylcholine and other acetylcholine receptor agonists was also unchanged. These results indicate that recurrent hypoglycaemia compromised the efficacy with which peripheral neuronal activity stimulates adrenal catecholamine release and demonstrate that peripheral components of the sympatho-adrenal system were directly affected by recurrent hypoglycaemia.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Epinefrina/metabolismo , Hipoglucemia/fisiopatología , Norepinefrina/metabolismo , Acetilcolina/metabolismo , Glándulas Suprarrenales/efectos de los fármacos , Glándulas Suprarrenales/inervación , Animales , Glucemia/análisis , Agonistas Colinérgicos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Estimulación Eléctrica , Epinefrina/sangre , Hipoglucemia/sangre , Hipoglucemia/metabolismo , Masculino , Agonistas Muscarínicos/farmacología , Nicotina/farmacología , Norepinefrina/sangre , Perfusión , Pilocarpina/farmacología , Ratas Sprague-Dawley , Receptores Colinérgicos/química , Receptores Colinérgicos/metabolismo , Recurrencia , Nervios Esplácnicos/metabolismo
18.
Front Syst Neurosci ; 8: 236, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25540613

RESUMEN

The neuroendocrine system governs essential survival and homeostatic functions. For example, growth is needed for development, thermoregulation maintains optimal core temperature in a changing environment, and reproduction ensures species survival. Stress and immune responses enable an organism to overcome external and internal threats while the circadian system regulates arousal and sleep such that vegetative and active functions do not overlap. All of these functions require a significant portion of the body's energy. As the integrator of the neuroendocrine system, the hypothalamus carefully assesses the energy status of the body in order to appropriately partition resources to provide for each system without compromising the others. While doing so the hypothalamus must ensure that adequate glucose levels are preserved for brain function since glucose is the primary fuel of the brain. To this end, the hypothalamus contains specialized glucose sensing neurons which are scattered throughout the nuclei controlling distinct neuroendocrine functions. We hypothesize that these neurons play a key role in enabling the hypothalamus to partition energy to meet these peripheral survival needs without endangering the brain's glucose supply. This review will first describe the varied mechanisms underlying glucose sensing in neurons within discrete hypothalamic nuclei. We will then evaluate the way in which peripheral energy status regulates glucose sensitivity. For example, during energy deficit such as fasting specific hypothalamic glucose sensing neurons become sensitized to decreased glucose. This increases the gain of the information relay when glucose availability is a greater concern for the brain. Finally, changes in glucose sensitivity under pathological conditions (e.g., recurrent insulin-hypoglycemia, diabetes) will be addressed. The overall goal of this review is to place glucose sensing neurons within the context of hypothalamic control of neuroendocrine function.

19.
Mol Cell Neurosci ; 62: 30-41, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25107627

RESUMEN

Lateral hypothalamic area (LHA) orexin neurons modulate reward-based feeding by activating ventral tegmental area (VTA) dopamine (DA) neurons. We hypothesize that signals of peripheral energy status influence reward-based feeding by modulating the glucose sensitivity of LHA orexin glucose-inhibited (GI) neurons. This hypothesis was tested using electrophysiological recordings of LHA orexin-GI neurons in brain slices from 4 to 6week old male mice whose orexin neurons express green fluorescent protein (GFP) or putative VTA-DA neurons from C57Bl/6 mice. Low glucose directly activated ~60% of LHA orexin-GFP neurons in both whole cell and cell attached recordings. Leptin indirectly reduced and ghrelin directly enhanced the activation of LHA orexin-GI neurons by glucose decreases from 2.5 to 0.1mM by 53±12% (n=16, P<0.001) and 41±24% (n=8, P<0.05), respectively. GABA or neurotensin receptor blockade prevented leptin's effect on glucose sensitivity. Fasting increased activation of LHA orexin-GI neurons by decreased glucose, as would be predicted by these hormonal effects. We also evaluated putative VTA-DA neurons in a novel horizontal slice preparation containing the LHA and VTA. Decreased glucose increased the frequency of spontaneous excitatory post-synaptic currents (sEPSCs; 125 ± 40%, n=9, P<0.05) and action potentials (n=9; P<0.05) in 45% (9/20) of VTA DA neurons. sEPSCs were completely blocked by AMPA and NMDA glutamate receptor antagonists (CNQX 20 µM, n=4; APV 20µM, n=4; respectively), demonstrating that these sEPSCs were mediated by glutamatergic transmission onto VTA DA neurons. Orexin-1 but not 2 receptor antagonism with SB334867 (10µM; n=9) and TCS-OX2-29 (2µM; n=5), respectively, blocks the effects of decreased glucose on VTA DA neurons. Thus, decreased glucose increases orexin-dependent excitatory glutamate neurotransmission onto VTA DA neurons. These data suggest that the glucose sensitivity of LHA orexin-GI neurons links metabolic state and reward-based feeding.


Asunto(s)
Glucosa/metabolismo , Área Hipotalámica Lateral/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mesencéfalo/metabolismo , Neuronas/metabolismo , Neuropéptidos/metabolismo , Recompensa , Potenciales de Acción/fisiología , Animales , Benzoxazoles/farmacología , Área Hipotalámica Lateral/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Naftiridinas , Neuronas/efectos de los fármacos , Orexinas , Transmisión Sináptica/fisiología , Urea/análogos & derivados , Urea/farmacología , Área Tegmental Ventral/metabolismo
20.
J Vis Exp ; (81)2013 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-24326343

RESUMEN

Studies of neuronal activity are often performed using neurons from rodents less than 2 months of age due to the technical difficulties associated with increasing connective tissue and decreased neuronal viability that occur with age. Here, we describe a methodology for the dissociation of healthy hypothalamic neurons from adult-aged mice. The ability to study neurons from adult-aged mice allows the use of disease models that manifest at a later age and might be more developmentally accurate for certain studies. Fluorescence imaging of dissociated neurons can be used to study the activity of a population of neurons, as opposed to using electrophysiology to study a single neuron. This is particularly useful when studying a heterogeneous neuronal population in which the desired neuronal type is rare such as for hypothalamic glucose sensing neurons. We utilized membrane potential dye imaging of adult ventromedial hypothalamic neurons to study their responses to changes in extracellular glucose. Glucose sensing neurons are believed to play a role in central regulation of energy balance. The ability to study glucose sensing in adult rodents is particularly useful since the predominance of diseases related to dysfunctional energy balance (e.g. obesity) increase with age.


Asunto(s)
Colorantes Fluorescentes/química , Glucosa/metabolismo , Neuronas/fisiología , Espectrometría de Fluorescencia/métodos , Coloración y Etiquetado/métodos , Núcleo Hipotalámico Ventromedial/fisiología , Animales , Masculino , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/metabolismo , Núcleo Hipotalámico Ventromedial/citología , Núcleo Hipotalámico Ventromedial/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA