Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Am J Physiol Regul Integr Comp Physiol ; 326(5): R383-R400, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38105761

RESUMEN

The hormone leptin reduces food intake through actions in the peripheral and central nervous systems, including in the hindbrain nucleus of the solitary tract (NTS). The NTS receives viscerosensory information via vagal afferents, including information from the gastrointestinal tract, which is then relayed to other central nervous system (CNS) sites critical for control of food intake. Leptin receptors (lepRs) are expressed by a subpopulation of NTS neurons, and knockdown of these receptors increases both food intake and body weight. Recently, we demonstrated that leptin increases vagal activation of lepR-expressing neurons via increased NMDA receptor (NMDAR) currents, thereby potentiating vagally evoked firing. Furthermore, chemogenetic activation of these neurons was recently shown to inhibit food intake. However, the vagal inputs these neurons receive had not been characterized. Here we performed whole cell recordings in brain slices taken from lepRCre × floxedTdTomato mice and found that lepR neurons of the NTS are directly activated by monosynaptic inputs from C-type afferents sensitive to the transient receptor potential vanilloid type 1 (TRPV1) agonist capsaicin. CCK administered onto NTS slices stimulated spontaneous glutamate release onto lepR neurons and induced action potential firing, an effect mediated by CCKR1. Interestingly, NMDAR activation contributed to the current carried by spontaneous excitatory postsynaptic currents (EPSCs) and enhanced CCK-induced firing. Peripheral CCK also increased c-fos expression in these neurons, suggesting they are activated by CCK-sensitive vagal afferents in vivo. Our results indicate that the majority of NTS lepR neurons receive direct inputs from CCK-sensitive C vagal-type afferents, with both peripheral and central CCK capable of activating these neurons and NMDARs able to potentiate these effects.


Asunto(s)
Receptores de N-Metil-D-Aspartato , Núcleo Solitario , Animales , Ratones , Leptina/metabolismo , Fibras Nerviosas Amielínicas/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Núcleo Solitario/metabolismo , Nervio Vago/fisiología
2.
Am J Physiol Regul Integr Comp Physiol ; 325(3): R229-R237, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37424401

RESUMEN

To investigate the role of glial cells in the regulation of glucoprivic responses in rats, a chemogenetic approach was used to activate astrocytes neighboring catecholamine (CA) neurons in the ventromedial medulla (VLM) where A1 and C1 CA cell groups overlap (A1/C1). Previous results indicate that activation of CA neurons in this region is necessary and sufficient for feeding and corticosterone release in response to glucoprivation. However, it is not known whether astrocyte neighbors of CA neurons contribute to glucoregulatory responses. Hence, we made nanoinjections of AAV5-GFAP-hM3D(Gq)-mCherry to selectively transfect astrocytes in the A1/C1 region with the excitatory designer receptor exclusively activated by designer drugs (DREADDs), hM3D(Gq). After allowing time for DREADD expression, we evaluated the rats for increased food intake and corticosterone release in response to low systemic doses of the antiglycolytic agent, 2-deoxy-d-glucose (2DG), alone and in combination with the hM3D(Gq) activator clozapine-n-oxide (CNO). We found that DREADD-transfected rats ate significantly more food when 2DG and CNO were coadministered than when either 2DG or CNO was injected alone. We also found that CNO significantly enhanced 2DG-induced FOS expression in the A1/C1 CA neurons, and that corticosterone release also was enhanced when CNO and 2DG were administered together. Importantly, CNO-induced activation of astrocytes in the absence of 2DG did not trigger food intake or corticosterone release. Our results indicate that during glucoprivation, activation of VLM astrocytes cells markedly increases the sensitivity or responsiveness of neighboring A1/C1 CA neurons to glucose deficit, suggesting a potentially important role for VLM astrocytes in glucoregulation.


Asunto(s)
Astrocitos , Corticosterona , Ratas , Animales , Astrocitos/metabolismo , Desoxiglucosa/farmacología , Ratas Sprague-Dawley , Bulbo Raquídeo/metabolismo , Glucosa/metabolismo , Catecolaminas/metabolismo
3.
J Neurosci ; 40(37): 7054-7064, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32817248

RESUMEN

Leptin signaling within the nucleus of the solitary tract (NTS) contributes to the control of food intake, and injections of leptin into the NTS reduce meal size and increase the efficacy of vagus-mediated satiation signals. Leptin receptors (LepRs) are expressed by vagal afferents as well as by a population of NTS neurons. However, the electrophysiological properties of LepR-expressing NTS neurons have not been well characterized, and it is unclear how leptin might act on these neurons to reduce food intake. To address this question, we recorded from LepR-expressing neurons in horizontal brain slices containing the NTS from male and female LepR-Cre X Rosa-tdTomato mice. We found that the vast majority of NTS LepR neurons received monosynaptic innervation from vagal afferent fibers and LepR neurons exhibited large synaptic NMDA receptor (NMDAR)-mediated currents compared with non-LepR neurons. During high-frequency stimulation of vagal afferents, leptin increased the size of NMDAR-mediated currents, but not AMPAR-mediated currents. Leptin also increased the size of evoked EPSPs and the ability of low-intensity solitary tract stimulation to evoke action potentials in LepR neurons. These effects of leptin were blocked by bath applying a competitive NMDAR antagonist (DCPP-ene) or by an NMDAR channel blocker applied through the recording pipette (MK-801). Last, feeding studies using male rats demonstrate that intra-NTS injections of DCPP-ene attenuate reduction of overnight food intake following intra-NTS leptin injection. Our results suggest that leptin acts in the NTS to reduce food intake by increasing NMDAR-mediated currents, thus enhancing NTS sensitivity to vagal inputs.SIGNIFICANCE STATEMENT Leptin is a hormone that critically impacts food intake and energy homeostasis. The nucleus of the solitary tract (NTS) is activated by vagal afferents from the gastrointestinal tract, which promotes termination of a meal. Injection of leptin into the NTS inhibits food intake, while knockdown of leptin receptors (LepRs) in NTS neurons increases food intake. However, little was known about how leptin acts in the NTS neurons to inhibit food intake. We found that leptin increases the sensitivity of LepR-expressing neurons to vagal inputs by increasing NMDA receptor-mediated synaptic currents and that NTS NMDAR activation contributes to leptin-induced reduction of food intake. These findings suggest a novel mechanism by which leptin, acting in the NTS, could potentiate gastrointestinal satiation signals.


Asunto(s)
Potenciales Postsinápticos Excitadores , Leptina/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Núcleo Solitario/metabolismo , Nervio Vago/metabolismo , Animales , Maleato de Dizocilpina/farmacología , Ingestión de Alimentos , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/fisiología , Prolina/análogos & derivados , Prolina/farmacología , Piridinas/farmacología , Ratas , Núcleo Solitario/citología , Núcleo Solitario/fisiología , Sinapsis/metabolismo , Sinapsis/fisiología , Nervio Vago/citología , Nervio Vago/fisiología
4.
Am J Physiol Regul Integr Comp Physiol ; 316(4): R406-R416, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30726118

RESUMEN

Neuropeptide Y (NPY), peptide YY (PYY), and their cognate receptors (YR) are expressed by subpopulations of central and peripheral nervous system neurons. Intracerebroventricular injections of NPY or PYY increase food intake, and intrahypothalamic NPY1 or NPY5 receptor agonist injections also increase food intake. In contrast, injection of PYY in the periphery reduces food intake, apparently by activating peripheral Y2R. The dorsal vagal complex (DVC) of the hindbrain is the site where vagal afferents relay gut satiation signals to the brain. While contributions of the DVC are increasingly investigated, a role for DVC YR in control of food intake has not been examined systematically. We used in situ hybridization to confirm expression of Y1R and Y2R, but not Y5R, in the DVC and vagal afferent neurons. We found that nanoinjections of a Y2R agonist, PYY-(3-36), into the DVC significantly increased food intake over a 4-h period in satiated male rats. PYY-(3-36)-evoked food intake was prevented by injection of a selective Y2R antagonist. Injection of a Y1R/Y5R-preferring agonist into the DVC failed to increase food intake at doses reported to increase food intake following hypothalamic injection. Finally, injection of PYY-(3-36) into the DVC prevented reduction of 30-min food intake following intraperitoneal injection of cholecystokinin (CCK). Our results indicate that activation of DVC Y2R, unlike hypothalamic or peripheral Y2R, increases food intake. Furthermore, in the context of available electrophysiological observations, our results are consistent with the hypothesis that DVC Y2R control food intake by dampening vagally mediated satiation signals in the DVC.


Asunto(s)
Colecistoquinina/farmacología , Ingestión de Alimentos/efectos de los fármacos , Receptores de Neuropéptido Y/agonistas , Saciedad/efectos de los fármacos , Nervio Vago/efectos de los fármacos , Animales , Inyecciones , Masculino , Péptido YY/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Neuropéptido Y/antagonistas & inhibidores , Receptores de Neuropéptido Y/efectos de los fármacos
5.
J Neurosci ; 34(38): 12636-45, 2014 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-25232103

RESUMEN

Injection of the melanocortin-3/4 receptor agonist melanotan-II (MTII) into the nucleus of the solitary tract (NTS) produces rapid and sustained reduction of food intake. Melanocortin-4 receptors (MC4Rs) are expressed by vagal afferent endings in the NTS, but it is not known whether these endings participate in MTII-induced reduction of food intake. In experiments described here, we evaluated the contribution of central vagal afferent endings in MTII-induced reduction of food intake. Examination of rat hindbrain sections revealed that neuronal processes expressing immunoreactivity for the endogenous MC4R agonist α-melanoctyte-stimulating hormone course parallel and wrap around anterogradely labeled vagal afferent endings in the NTS and thus are aptly positioned to activate vagal afferent MC4Rs. Furthermore, MTII and endogenous MC4R agonists increased protein kinase A (PKA)-catalyzed phosphorylation of synapsin I in vagal afferent endings, an effect known to increase synaptic strength by enhancing neurotransmitter release in other neural systems. Hindbrain injection of a PKA inhibitor, KT5720, significantly attenuated MTII-induced reduction of food intake and the increase in synapsin I phosphorylation. Finally, unilateral nodose ganglion removal, resulting in degeneration of vagal afferent endings in the ipsilateral NTS, abolished MTII-induced synapsin I phosphorylation ipsilateral to nodose ganglion removal. Moreover, reduction of food intake following MTII injection into the NTS ipsilateral to nodose ganglion removal was significantly attenuated, whereas the response to MTII was not diminished when injected into the contralateral NTS. Altogether, our results suggest that reduction of food intake following hindbrain MC4R activation is mediated by central vagal afferent endings.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Terminaciones Nerviosas/efectos de los fármacos , Neuronas Aferentes/fisiología , Péptidos Cíclicos/farmacología , Receptor de Melanocortina Tipo 4/agonistas , Núcleo Solitario/efectos de los fármacos , Nervio Vago/efectos de los fármacos , alfa-MSH/análogos & derivados , Animales , Carbazoles/administración & dosificación , Carbazoles/farmacología , Masculino , Microinyecciones , Terminaciones Nerviosas/fisiología , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/metabolismo , Ganglio Nudoso/fisiología , Péptidos Cíclicos/administración & dosificación , Péptidos Cíclicos/antagonistas & inhibidores , Fosforilación , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Pirroles/administración & dosificación , Pirroles/farmacología , Ratas , Receptor de Melanocortina Tipo 4/metabolismo , Núcleo Solitario/fisiología , Sinapsinas/metabolismo , Nervio Vago/fisiología , alfa-MSH/administración & dosificación , alfa-MSH/antagonistas & inhibidores , alfa-MSH/metabolismo , alfa-MSH/farmacología
6.
J Physiol ; 593(1): 111-25, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25281729

RESUMEN

Hindbrain NMDA receptors play important roles in reflexive and behavioural responses to vagal activation. NMDA receptors have also been shown to contribute to the synaptic responses of neurons in the nucleus of the solitary tract (NTS), but their exact role remains unclear. In this study we used whole cell patch-clamping techniques in rat horizontal brain slice to investigate the role of NMDA receptors in the fidelity of transmission across solitary tract afferent-NTS neuron synapses. Results show that NMDA receptors contribute up to 70% of the charge transferred across the synapse at high (>5 Hz) firing rates, but have little contribution at lower firing frequencies. Results also show that NMDA receptors critically contribute to the fidelity of transmission across these synapses during high frequency (>5 Hz) afferent discharge rates. This novel role of NMDA receptors may explain in part how primary visceral afferents, including vagal afferents, can maintain fidelity of transmission across a broad range of firing frequencies. Neurons within the nucleus of the solitary tract (NTS) receive vagal afferent innervations that initiate gastrointestinal and cardiovascular reflexes. Glutamate is the fast excitatory neurotransmitter released in the NTS by vagal afferents, which arrive there via the solitary tract (ST). ST stimulation elicits excitatory postsynaptic currents (EPSCs) in NTS neurons mediated by both AMPA- and NMDA-type glutamate receptors (-Rs). Vagal afferents exhibit a high probability of vesicle release and exhibit robust frequency-dependent depression due to presynaptic vesicle depletion. Nonetheless, synaptic throughput is maintained even at high frequencies of afferent activation. Here we test the hypothesis that postsynaptic NMDA-Rs are essential in maintaining throughput across ST-NTS synapses. Using patch clamp electrophysiology in horizontal brainstem slices, we found that NMDA-Rs, including NR2B subtypes, carry up to 70% of the charge transferred across the synapse during high frequency stimulations (>5 Hz). In contrast, their relative contribution to the ST-EPSC is much less during low (<2 Hz) frequency stimulations. Afferent-driven activation of NMDA-Rs produces a sustained depolarization during high, but not low, frequencies of stimulation as a result of relatively slow decay kinetics. Hence, NMDA-Rs are critical for maintaining action potential generation at high firing rates. These results demonstrate a novel role for NMDA-Rs enabling a high probability of release synapse to maintain the fidelity of synaptic transmission during high frequency firing when glutamate release and AMPA-R responses are reduced. They also suggest why NMDA-Rs are critical for responses that may depend on high rates of afferent discharge.


Asunto(s)
Receptores de N-Metil-D-Aspartato/fisiología , Núcleo Solitario/fisiología , Animales , Potenciales Postsinápticos Excitadores/fisiología , Técnicas In Vitro , Masculino , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley
7.
Am J Physiol Regul Integr Comp Physiol ; 308(1): R1-9, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25394828

RESUMEN

Hindbrain injection of a melanocortin-3/4 receptor agonist, MTII, reduces food intake primarily by reducing meal size. Our previously reported results indicate that N-methyl-D-aspartate-type glutamate receptors (NMDAR) in the nucleus of the solitary tract (NTS) play an important role in the control of meal size and food intake. Therefore, we hypothesized that activation of NTS NMDARs contribute to reduction of food intake in response to fourth ventricle or NTS injection of MTII. We found that coinjection of a competitive NMDAR antagonist (d-CPP-ene) with MTII into the fourth ventricle or directly into the NTS of adult male rats attenuated MTII-induced reduction of food intake. Hindbrain NMDAR antagonism also attenuated MTII-induced ERK1/2 phosphorylation in NTS neurons and prevented synapsin I phosphorylation in central vagal afferent endings, both of which are cellular mechanisms previously shown to participate in hindbrain melanocortinergic reduction of food intake. Together, our results indicate that NMDAR activation significantly contributes to reduction of food intake following hindbrain melanocortin receptor activation, and it participates in melanocortinergic signaling in NTS neural circuits that mediate reduction of food intake.


Asunto(s)
Regulación del Apetito/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Receptores de Melanocortina/agonistas , Receptores de N-Metil-D-Aspartato/metabolismo , Rombencéfalo/efectos de los fármacos , alfa-MSH/análogos & derivados , Animales , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Inyecciones Intraventriculares , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Fosforilación , Ratas Sprague-Dawley , Receptores de Melanocortina/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Rombencéfalo/metabolismo , Saciedad , Transducción de Señal/efectos de los fármacos , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/metabolismo , Sinapsinas/metabolismo , Factores de Tiempo , Nervio Vago/efectos de los fármacos , Nervio Vago/metabolismo , alfa-MSH/administración & dosificación
8.
Endocrinology ; 165(5)2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38368624

RESUMEN

Glucoprivic feeding is one of several counterregulatory responses (CRRs) that facilitates restoration of euglycemia following acute glucose deficit (glucoprivation). Our previous work established that glucoprivic feeding requires ventrolateral medullary (VLM) catecholamine (CA) neurons that coexpress neuropeptide Y (NPY). However, the connections by which VLM CA/NPY neurons trigger increased feeding are uncertain. We have previously shown that glucoprivation, induced by an anti-glycolygic agent 2-deoxy-D-glucose (2DG), activates perifornical lateral hypothalamus (PeFLH) neurons and that expression of NPY in the VLM CA/NPY neurons is required for glucoprivic feeding. We therefore hypothesized that glucoprivic feeding and possibly other CRRs require NPY-sensitive PeFLH neurons. To test this, we used the ribosomal toxin conjugate NPY-saporin (NPY-SAP) to selectively lesion NPY receptor-expressing neurons in the PeFLH of male rats. We found that NPY-SAP destroyed a significant number of PeFLH neurons, including those expressing orexin, but not those expressing melanin-concentrating hormone. The PeFLH NPY-SAP lesions attenuated 2DG-induced feeding but did not affect 2DG-induced increase in locomotor activity, sympathoadrenal hyperglycemia, or corticosterone release. The 2DG-induced feeding response was also significantly attenuated in NPY-SAP-treated female rats. Interestingly, PeFLH NPY-SAP lesioned male rats had reduced body weights and decreased dark cycle feeding, but this effect was not seen in female rats. We conclude that a NPY projection to the PeFLH is necessary for glucoprivic feeding, but not locomotor activity, hyperglycemia, or corticosterone release, in both male and female rats.


Asunto(s)
Conducta Alimentaria , Hipotálamo , Neuronas , Neuropéptido Y , Ratas Sprague-Dawley , Animales , Femenino , Masculino , Ratas , Desoxiglucosa/farmacología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Conducta Alimentaria/efectos de los fármacos , Glucosa/metabolismo , Área Hipotalámica Lateral/metabolismo , Área Hipotalámica Lateral/efectos de los fármacos , Hormonas Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Hipotálamo/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Melaninas/metabolismo , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Neuropéptido Y/metabolismo , Neuropéptido Y/farmacología , Neuropéptidos/metabolismo , Orexinas/metabolismo , Hormonas Hipofisarias/metabolismo , Receptores de Neuropéptido Y/metabolismo , Receptores de Neuropéptido Y/genética , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Saporinas/farmacología
9.
Am J Physiol Regul Integr Comp Physiol ; 302(2): R264-73, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22031786

RESUMEN

Previous reports suggest that glucagon-like peptide (GLP-1), a peptide secreted from the distal small intestine, is an endocrine satiation signal. Nevertheless, there are conflicting reports regarding the site where circulating GLP-1 acts to reduce food intake. To test the hypothesis that vagal afferents are necessary for reduction of food intake by circulating GLP-1, we measured intake of 15% sucrose during intravenous GLP-1 infusion in intact, vagotomized, and capsaicin-treated rats. We also measured sucrose intake during intravenous infusion of cholecystokinin, a peptide known to reduce food intake via abdominal vagal afferents. We found that reduction of intake by GLP-1 was not diminished by capsaicin treatment or vagotomy. In fact, reduction of sucrose intake by our highest GLP-1 dose was enhanced in vagotomized and capsaicin-treated rats. Intravenous GLP-1 induced comparable increases of hindbrain c-Fos immunoreactivity in intact, capsaicin-treated, and vagotomized rats. Plasma concentrations of active GLP-1 in capsaicin-treated rats did not differ from those of controls during the intravenous infusions. Finally, capsaicin treatment was not associated with altered GLP-1R mRNA in the brain, but nodose ganglia GLP-1R mRNA was significantly reduced in capsaicin-treated rats. Although reduction of food intake by intraperitoneal cholecystokinin was abolished in vagotomized and capsaicin-treated rats, reduction of intake by intravenous cholecystokinin was only partially attenuated. These results indicate that vagal or capsaicin-sensitive neurons are not necessary for reduction of food intake by circulating (endocrine) GLP-1, or cholecystokinin. Vagal participation in satiation by these peptides may be limited to paracrine effects exerted near the sites of their secretion.


Asunto(s)
Capsaicina/farmacología , Ingestión de Alimentos/efectos de los fármacos , Péptido 1 Similar al Glucagón/farmacología , Sincalida/farmacología , Nervio Vago/fisiología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ingestión de Alimentos/fisiología , Péptido 1 Similar al Glucagón/sangre , Masculino , Ganglio Nudoso/efectos de los fármacos , Ganglio Nudoso/metabolismo , Ratas , Ratas Sprague-Dawley , Sincalida/sangre , Vagotomía , Nervio Vago/efectos de los fármacos
10.
Br J Pharmacol ; 179(11): 2589-2609, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35023154

RESUMEN

BACKGROUND AND PURPOSE: 'Food addiction' is the subject of intense public and research interest. However, this nosology based on neurobehavioural similarities among obese individuals, patients with eating disorders and those with substance use disorders (drug addiction) remains controversial. We thus sought to determine which aspects of disordered eating are causally linked to preclinical models of drug addiction. We hypothesized that extensive drug histories, known to cause addiction-like brain changes and drug motivation in rats, would also cause addiction-like food motivation. EXPERIMENTAL APPROACH: Rats underwent extensive cocaine, alcohol, caffeine or obesogenic diet histories and were subsequently tested for punishment-resistant food self-administration or 'compulsive appetite', as a measure of addiction-like food motivation. KEY RESULTS: Extensive cocaine and alcohol (but not caffeine) histories caused compulsive appetite that persisted long after the last drug exposure. Extensive obesogenic diet histories also caused compulsive appetite, although neither cocaine nor alcohol histories caused excess calorie intake and bodyweight during abstinence. Hence, compulsive appetite and obesity appear to be dissociable, with the former sharing common mechanisms with preclinical drug addiction models. CONCLUSION AND IMPLICATIONS: Compulsive appetite, as seen in subsets of obese individuals and patients with binge-eating disorder and bulimia nervosa (eating disorders that do not necessarily result in obesity), appears to epitomize 'food addiction'. Because different drug and obesogenic diet histories caused compulsive appetite, overlapping dysregulations in the reward circuits, which control drug and food motivation independently of energy homeostasis, may offer common therapeutic targets for treating addictive behaviours across drug addiction, eating disorders and obesity.


Asunto(s)
Conducta Adictiva , Cocaína , Adicción a la Comida , Trastornos Relacionados con Sustancias , Animales , Apetito , Conducta Alimentaria , Alimentos , Adicción a la Comida/complicaciones , Humanos , Obesidad/etiología , Preparaciones Farmacéuticas , Ratas
11.
Am J Physiol Regul Integr Comp Physiol ; 301(2): R448-55, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21562094

RESUMEN

Intraperitoneal injection of CCK reduces food intake and triggers a behavioral pattern similar to natural satiation. Reduction of food intake by CCK is mediated by vagal afferents that innervate the stomach and small intestine. These afferents synapse in the hindbrain nucleus of the solitary tract (NTS) where gastrointestinal satiation signals are processed. Previously, we demonstrated that intraperitoneal (IP) administration of either competitive or noncompetitive N-methyl-d-aspartate (NMDA) receptor antagonists attenuates reduction of food intake by CCK. However, because vagal afferents themselves express NMDA receptors at both central and peripheral endings, our results did not speak to the question of whether NMDA receptors in the brain play an essential role in reduction of feeding by CCK. We hypothesized that activation of NMDA receptors in the NTS is necessary for reduction of food intake by CCK. To test this hypothesis, we measured food intake following IP CCK, subsequent to NMDA receptor antagonist injections into the fourth ventricle, directly into the NTS or subcutaneously. We found that either fourth-ventricle or NTS injection of the noncompetitive NMDA receptor antagonist MK-801 was sufficient to inhibit CCK-induced reduction of feeding, while the same antagonist doses injected subcutaneously did not. Similarly fourth ventricle injection of d-3-(2-carboxypiperazin-4-yl)-1-propenyl-1-phosphoric acid (d-CPPene), a competitive NMDA receptor antagonist, also blocked reduction of food intake following IP CCK. Finally, d-CPPene injected into the fourth ventricle attenuated CCK-induced expression of nuclear c-Fos immunoreactivity in the dorsal vagal complex. We conclude that activation of NMDA receptors in the hindbrain is necessary for the reduction of food intake by CCK. Hindbrain NMDA receptors could comprise a critical avenue for control and modulation of satiation signals to influence food intake and energy balance.


Asunto(s)
Colecistoquinina/farmacología , Ingestión de Alimentos/efectos de los fármacos , Receptores de Glutamato/metabolismo , Rombencéfalo/efectos de los fármacos , Animales , Maleato de Dizocilpina/administración & dosificación , Maleato de Dizocilpina/farmacología , Regulación de la Expresión Génica/fisiología , Genes fos/fisiología , Inyecciones Intraventriculares , Masculino , Piperazinas/administración & dosificación , Piperazinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato/clasificación , Rombencéfalo/fisiología , Saciedad
12.
J Comp Neurol ; 496(6): 877-85, 2006 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-16628619

RESUMEN

Most vagal afferent neurons in rat nodose ganglia express mRNA coding for the NR1 subunit of the heteromeric N-methyl-D-aspartate (NMDA) receptor ion channel. NMDA receptor subunit immunoreactivity has been detected on axon terminals of vagal afferents in the dorsal hindbrain, suggesting a role for presynaptic NMDA receptors in viscerosensory function. Although NMDA receptor subunits (NR1, NR2B, NR2C, and NR2D) have been linked to distinct neuronal populations in the brain, the NMDA receptor subunit phenotype of vagal afferent neurons has not been determined. Therefore, we examined NMDA receptor subunit (NR1, NR2B, NR2C, and NR2D) immunoreactivity in vagal afferent neurons. We found that, although the left nodose contained significantly more neurons (7,603), than the right (5,978), the proportions of NMDA subunits expressed in the left and right nodose ganglia were not significantly different. Immunoreactivity for NMDA NR1 subunit was present in 92.3% of all nodose neurons. NR2B immunoreactivity was present in 56.7% of neurons; NR2C-expressing nodose neurons made up 49.4% of the total population; NR2D subunit immunoreactivity was observed in just 13.5% of all nodose neurons. Double labeling revealed that 30.2% of nodose neurons expressed immunoreactivity to both NR2B and NR2C, whereas NR2B and NR2D immunoreactivities were colocalized in 11.5% of nodose neurons. NR2C immunoreactivity colocalized with NR2D in 13.1% of nodose neurons. Our results indicate that most vagal afferent neurons express NMDA receptor ion channels composed of NR1, NR2B, and NR2C subunits and that a minority phenotype that expresses NR2D also expresses NR1, NR2B, and NR2C.


Asunto(s)
Neuronas Aferentes/metabolismo , Ganglio Nudoso/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Nervio Vago/metabolismo , Animales , Recuento de Células , Núcleo Celular/ultraestructura , Técnica del Anticuerpo Fluorescente , Masculino , Neuronas Aferentes/citología , Ganglio Nudoso/citología , Fenotipo , Subunidades de Proteína/clasificación , Subunidades de Proteína/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/clasificación
13.
Brain Res ; 1119(1): 86-93, 2006 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-16989781

RESUMEN

Previous reports suggest that NMDA receptors participate in control of food intake via vagal afferent neurons that innervate the upper gastrointestinal (GI) tract. While messenger RNA coding for the NR1 NMDA receptor subunit is present in a majority of vagal afferent neurons of nodose ganglia (NG), immunoreactivity for other NMDA receptor subunits (NR2B, NR2C and NR2D) are expressed in more limited subpopulations of vagal afferents. To determine whether vagal afferent neurons that project to the stomach or duodenum exhibit distinct NMDA receptor subunit phenotypes, we examined immunoreactivity (IR) for NMDA receptor NR1, NR2B, NR2C and NR2D subunits in NG neurons that were labeled by injections of the retrograde tracer Fast Blue (FB) into the wall of the stomach or duodenum. FB injections into the fundus or corpus of the stomach labeled comparable numbers of neurons in both the left and right NG, while proximal duodenal injections labeled only neurons of left NG. NR1-IR expression was observed in most neurons innervating the upper GI tract (fundus, 97%; corpus, 95%; duodenum, 98%). Likewise, most neurons that innervated the upper GI tract expressed NR2B-IR (fundus, 98%; corpus, 85%; duodenum, 81%). NR2C-IR was observed in only 52%, 46% and 32% of FB-positive neurons projecting to the fundus, corpus or duodenum respectively, while NR2D-IR occurred in an even more restricted FB-labeled subpopulation (fundus, 13%; corpus, 26%; and duodenum, 18%). Our observations indicate that different subpopulations of vagal afferents express distinct NMDA receptor subunit phenotypes. However, the neuronal distribution of NMDA receptor subunits is not correlated with innervation of either the stomach or duodenum.


Asunto(s)
Tracto Gastrointestinal/inervación , Neuronas Aferentes/metabolismo , Subunidades de Proteína/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Nervio Vago/metabolismo , Aferentes Viscerales/metabolismo , Amidinas , Animales , Recuento de Células , Duodeno/inervación , Duodeno/fisiología , Tracto Gastrointestinal/fisiología , Inmunohistoquímica , Masculino , Neuronas Aferentes/citología , Ganglio Nudoso/citología , Ganglio Nudoso/metabolismo , Fenotipo , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/metabolismo , Estómago/inervación , Estómago/fisiología , Nervio Vago/citología , Aferentes Viscerales/citología
14.
Physiol Behav ; 89(4): 477-85, 2006 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-16872644

RESUMEN

The gut-peptide, cholecystokinin (CCK), reduces food intake by acting at CCK-1 receptors on vagal afferent neurons, whereas the feeding effects of the adipokine hormone, leptin, are associated primarily with its action on receptors (ObRb) in the hypothalamus. Recently, however, ObRb mRNA has been reported in vagal afferent neurons, some of which also express CCK-1 receptor, suggesting that leptin, alone or in cooperation with CCK, might activate vagal afferent neurons, and influence food intake via a vagal route. To evaluate these possibilities we have been examining the cellular and behavioral effects of leptin and CCK on vagal afferent neurons. In cultured vagal afferent neurons leptin and CCK evoked short latency, transient depolarizations, often leading to action potentials, and increases in cytosolic calcium. There was a much higher prevalence of CCK and leptin sensitivity amongst cultured vagal afferent neurons that innervate stomach or duodenum than there was in the overall vagal afferent population. Furthermore, almost all leptin-responsive gastric and duodenal vagal afferents also were sensitive to CCK. Leptin, infused into the upper GI tract arterial supply, reduced meal size, and enhanced satiation evoked by CCK. These results indicate that vagal afferent neurons are activated by leptin, and that this activation is likely to participate in meal termination, perhaps by enhancing vagal sensitivity to CCK. Our findings are consistent with the view that leptin and CCK exert their influence on food intake by accessing multiple neural systems (viscerosensory, motivational, affective and motor) at multiple points along the neuroaxis.


Asunto(s)
Vías Aferentes/fisiología , Regulación del Apetito/fisiología , Colecistoquinina/fisiología , Leptina/fisiología , Nervio Vago/fisiología , Animales , Ingestión de Alimentos/fisiología , Tracto Gastrointestinal/inervación , Tracto Gastrointestinal/fisiología , Humanos , Neuronas/fisiología , Saciedad/fisiología , Transducción de Señal/fisiología , Nervio Vago/citología
15.
Brain Res ; 1051(1-2): 155-63, 2005 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-16005445

RESUMEN

Many of the actions of cholecystokinin (CCK) are mediated by CCK-1 receptors, expressed by enteric and vagal afferent neurons. Otsuka Long-Evans Tokushima Fatty rats (OLETF) do not express CCK-1 receptors, and do not exhibit the vagally mediated responses to CCK. To determine whether the OLETF rat's failure to respond to CCK is correlated with failure of CCK to activate enteric and vagal neurons, we quantified neuronal Fos immunoreactivity in the dorsal vagal complex of the hindbrain, the nodose ganglia, and the ganglia of the myenteric and submucosal plexuses of the duodenum following intraperitoneal injection of CCK-8 (20 microg/kg). Compared to vehicle injection, CCK administration resulted in significant increases in the number of Fos-immunopositive neurons in the nucleus of the solitary tract, area postrema, and dorsal vagal motor nucleus of control, LETO rats. In OLETF rats, however, CCK did not increase numbers of Fos-immunoreactive neurons in any of these brain structures. CCK also induced significantly larger numbers of Fos-immunoreactive neuronal nuclei in the nodose ganglia of LETO rats, but not in the nodose ganglia of OLETF rats. Finally, LETO, but not OLETF rats exhibited striking increases in the number of Fos-immunoreactive nuclei of myenteric and submucosal neurons, following CCK injection. Absence of CCK-induced Fos expression in OLETF rats is consistent with attenuation of ingestive and gastrointestinal responses to CCK in the CCK-1 receptor deficient rats. These results also suggest that CCK-induced Fos expression in enteric and vagal sensory neurons of rats can be accounted for entirely by activation of CCK-1 receptors and is not due to occupation of CCK-2 (gastrin) receptors, which also are expressed in the intestine and by some vagal afferent neurons.


Asunto(s)
Colecistoquinina/fisiología , Neuronas Aferentes/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptor de Colecistoquinina A/fisiología , Animales , Duodeno/inervación , Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/metabolismo , Neuronas Aferentes/citología , Ganglio Nudoso/citología , Ganglio Nudoso/metabolismo , Ratas , Ratas Endogámicas OLETF , Ratas Long-Evans , Receptor de Colecistoquinina A/deficiencia , Rombencéfalo/citología , Rombencéfalo/metabolismo , Nervio Vago/citología , Nervio Vago/metabolismo
16.
Physiol Behav ; 86(1-2): 128-35, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16098547

RESUMEN

Intestinal infusion of nutrients, such as glucose and oleic acid, increase Fos-like immunoreactivity (Fos-LI) in both the enteric nervous system and neurons of the dorsal vagal complex (DVC) of the hindbrain. To test the hypothesis that increased Fos-LI in enteric neurons and the DVC, following intestinal nutrient infusions is mediated by cholecystokinin(1) receptors (CCK(1)), we counted enteric and DVC neurons that expressed Fos-LI following intestinal infusion of oleate or glucose, with and without pretreatment with the CCK(1) receptor antagonist, lorglumide. Both oleate and glucose infusions increased Fos-LI in the DVC. Oleate also increased Fos-LI in the myenteric and submucosal plexuses of the duodenum and the jejunum, but not the ileum, while glucose only increased Fos-LI in the submucosal plexus of the ileum. The CCK(1) receptor antagonist, lorglumide, abolished Fos-LI in the DVC following infusions of either oleate or glucose. In addition, lorglumide attenuated oleate-induced Fos-LI in the myenteric and submucosal plexuses of the duodenum and jejunum. However, lorglumide failed to attenuate glucose-induced Fos-LI in the submucosal plexus of the ileum. These data confirm previous reports indicating that CCK(1) receptors mediate increased DVC Fos-LI following intestinal infusion of oleate or glucose. CCK(1) receptors also contribute to increased Fos-LI in enteric neurons following intestinal oleate infusion. However, failure of lorglumide to attenuate the increase of Fos-LI in the ileal submucosal plexus following intestinal glucose suggests that some intestinal nutrients trigger Fos-LI induction via CCK(1) receptor-independent pathways.


Asunto(s)
Plexo Mientérico/citología , Neuronas/efectos de los fármacos , Ácido Oléico/administración & dosificación , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de Colecistoquinina/fisiología , Animales , Recuento de Células/métodos , Diagnóstico por Imagen/métodos , Interacciones Farmacológicas , Antagonistas de Hormonas/farmacología , Inmunohistoquímica/métodos , Intestinos/citología , Intestinos/efectos de los fármacos , Masculino , Plexo Mientérico/efectos de los fármacos , Plexo Mientérico/metabolismo , Neuronas/metabolismo , Proglumida/análogos & derivados , Proglumida/farmacología , Ratas , Ratas Sprague-Dawley
17.
Peptides ; 24(2): 237-44, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12668208

RESUMEN

Cholecystokinin (CCK) is a peptide hormone released from the I-cells of the upper small intestine. CCK evokes a variety of physiological responses, such as stimulation of pancreatic secretion, reduction of food intake and inhibition of gastric emptying. Previously, we reported that CCK activates enteric neurons in the rat. However the specific subpopulations of enteric neurons activated by CCK have not been identified. In the work reported here, we utilized immunohistochemical detection of nuclear Fos, a marker for neuronal activation, and selected phenotypic markers to identify some of the neuronal subpopulations activated by CCK. The phenotypic markers that we examined were: nitric oxide synthase (NOS), neurokinin-1 receptor (NK-1R), calbindin (Cal), Calretinin (Calr), and neurofilament-M (NF-M). We found that in the myenteric plexus of the rat duodenum and jejunum, CCK activated NOS immunoreactive neurons. In the submucosal plexus of duodenum and jejunum, CCK activated Cal, Calr and NF-M immunoreactive neurons. CCK failed to activate NK-1R immunoreactive neurons in either plexus. Our results indicate that CCK activates distinct enteric neurons in the rat upper small intestine. Furthermore the fact that NOS immunoreactive neurons were activated suggests that CCK modulates the activity of inhibitory motor neurons in the myenteric plexus. Expression of Fos immunoreactivity in Calr and Cal immunoreactive neurons is consistent with a role for CCK in modulation of intrinsic sensory and/or secretomotor neuronal activity in the submucosal plexus.


Asunto(s)
Colecistoquinina/farmacología , Intestino Delgado/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Calbindina 2 , Calbindinas , Núcleo Celular/química , Núcleo Celular/efectos de los fármacos , Femenino , Inmunohistoquímica , Intestino Delgado/química , Proteínas de Neurofilamentos/análisis , Neuronas/química , Óxido Nítrico Sintasa/análisis , Proteínas Proto-Oncogénicas c-fos/análisis , Ratas , Ratas Sprague-Dawley , Receptores de Neuroquinina-1/análisis , Proteína G de Unión al Calcio S100/análisis , Sincalida/farmacología
18.
Anat Rec A Discov Mol Cell Evol Biol ; 271(1): 209-16, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12552637

RESUMEN

Characterization of the enteric neurons is vital for understanding their physiological role. We have used single and dual label fluorescence and peroxidase-based immunohistochemistry in myenteric and submucosal whole mounts from the rat small intestine to evaluate the morphology and distribution of enteric neurons immunoreactive for the following phenotypic antigens: neuronal nitric oxide synthase (NOS), neurokinin-1 receptor (NK-1R), calretinin (Calr), calbindin (Cal), and neurofilament-M (NF-M). NOS-immunoreactive neurons had Dogiel type I morphology, were abundant in the myenteric plexus compared to the submucosal plexus, and never coexpressed NK-1R immunoreactivity. NK-1R- and Calr-immunoreactive neurons had Dogiel type II morphology and were distributed comparably in both plexuses. NK-1R and Calr-immunoreactivity were coexpressed in many of the same neurons. Calbindin-immunoreactive neurons exhibited four distinct morphologies: small and large Dogiel type II neurons, Dogiel type I neurons, and small elongated neurons. These neurons were significantly fewer in number in the myenteric plexus compared to the submucosal plexus. Neurofilament-M-immunoreactive neurons had three morphologies, Dogiel type II neurons, small Dogiel type II neurons, and a less common subpopulation of small, elongated, multipolar neurons. These neurons were also fewer in number in the myenteric plexus compared to the submucosal plexus. The distribution of these phenotypic markers may assist future work that elucidates the functional activities of these enteric neurons such as control of intestinal motility and adaptation to the entry of gastric contents.


Asunto(s)
Intestino Delgado/inervación , Neuronas/metabolismo , Animales , Calbindina 2 , Calbindinas , Masculino , Plexo Mientérico/química , Plexo Mientérico/citología , Proteínas de Neurofilamentos/metabolismo , Óxido Nítrico Sintasa/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Neuroquinina-1/metabolismo , Proteína G de Unión al Calcio S100/metabolismo , Plexo Submucoso/química , Plexo Submucoso/citología , Distribución Tisular
19.
Physiol Behav ; 81(2): 249-73, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15159171

RESUMEN

Satiation for food comprises the physiological processes that result in the termination of eating. Satiation is evoked by physical and chemical qualities of ingested food, which trigger afferent signals to the brain from multiple sites in the GI tract, including the stomach, the proximal small intestine, the distal small intestine and the colon. The physiological nature of each signal's contribution to satiation and overall control of food intake is likely to vary, depending on the level of the GI tract from which the signal arises. This article is a critical, though non-exhaustive, review of our current understanding of the mechanisms and adaptive value of satiation signals from the stomach and intestine.


Asunto(s)
Fenómenos Fisiológicos del Sistema Digestivo , Alimentos , Saciedad/fisiología , Animales , Humanos , Intestinos/fisiología , Transducción de Señal
20.
Auton Neurosci ; 115(1-2): 54-63, 2004 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-15507406

RESUMEN

Nutrients entering the small intestine trigger a variety of neural and endocrine reflexes that involve specific afferents, efferents and interneurons, many of which are located within the enteric nervous system (ENS). We hypothesized that intestinal nutrient stimuli might activate specific subpopulations of these neurons. To test this hypothesis, we utilized immunohistochemical detection of nuclear c-fos expression in the myenteric and submucosal plexuses of the rat small intestine following intraintestinal infusions of oleate or glucose. Additionally, we used dual label methods to detect both Fos-immunoreactivity and immunoreactivity for five phenotypic neuronal markers: neurokinin-1 receptor (NK-1R), neurofilament-M (NF-M), neuronal nitric oxide synthase (NOS), calbindin (Cal) and calretinin (Calr), to characterize neurons that were activated by intestinal infusion of oleate and glucose. We found that oleate and glucose activated myenteric neurons in the duodenum and jejunum, but not the ileum. Oleate and glucose infusions significantly increased the number of Fos-immunoreactive nuclei in the submucosal plexus of the duodenum and jejunum, however, only glucose increased Fos-immunoreactivity in the ileum. Oleate and glucose infusions were associated with a small increase in Fos-immunoreactivity in NOS-immunoreactive neurons in the myenteric plexus. In the submucosal plexus, the majority of neurons activated by intestinal infusion of oleate or glucose were immunoreactive to Cal and Calr. In the rat, many of these neurons have Dogiel Type II-like morphology, which is consistent with the possibility that these neurons function as mucosal afferents in reflexes activated by nutrient stimuli.


Asunto(s)
Sistema Nervioso Entérico/efectos de los fármacos , Glucosa/administración & dosificación , Intestino Delgado/efectos de los fármacos , Ácido Oléico/administración & dosificación , Animales , Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/metabolismo , Infusiones Parenterales/métodos , Intestino Delgado/citología , Intestino Delgado/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA