Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
J Neurosci ; 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38811166

RESUMEN

Neurons in the caudal nucleus of the solitary tract (cNTS) and intermediate reticular nucleus (IRt) that express the glucagon gene (Gcg) give rise to GLP1-immunopositive axons in the spinal cord and many subcortical brain regions. Central GLP1 receptor signaling contributes to motivated behavior and stress responses in rats and mice, in which hindbrain GLP1 neurons are activated to express cFos in a metabolic state-dependent manner. The present study examined whether GLP1 inputs to distinct brain regions arise from distinct subsets of Gcg-expressing neurons, and mapped the collective distribution of axon collaterals arising from projection-defined GLP1 neural populations. Using our Gcg-Cre knock-in rat model, Cre-dependent adeno-associated virus (AAV1) tracing was conducted in adult male and female rats to compare axonal projections of IRt vs. cNTS GLP1 neurons. Overlapping axonal projections were observed in all brain regions that receive GLP1 input, with the caveat that cNTS injections produced Cre-dependent labeling of some IRt neurons, and vice-versa. In additional experiments, specific diencephalic or limbic forebrain nuclei were microinjected with Cre-dependent retrograde AAVs (AAVrg) expressing reporters that fully labeled the axon collaterals of transduced GLP1 neural populations. AAVrg injected into each forebrain site labeled Gcg-expressing neurons in both the cNTS and IRt. The collective axon collaterals of these labeled neurons entered the spinal cord and every brain region previously reported to contain GLP1-positive axons. These results indicate that axons arising from populations of GLP1 neurons that innervate the thalamic PVT, hypothalamic PVH, and/or limbic forebrain BST collectively innervate all central regions that receive GLP1 axonal input.Significance statement Our novel anatomical findings indicate that target-defined populations of forebrain-projecting GLP1 neurons collectively project to downstream target regions in a widespread sprinkler-type manner, although collateralized axons arising from individual GLP1 projection neurons remain to be defined. Considered together with results from studies investigating the role of central GLP1 receptor signaling pathways in physiology and behavior, these findings support our emerging view that hindbrain Gcg-expressing neurons are positioned to simultaneously modulate synaptic transmission in widespread spinal cord, brainstem, hypothalamic, and limbic forebrain circuits in a metabolic state-dependent manner.

2.
Neuroendocrinology ; 113(5): 535-548, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36566746

RESUMEN

INTRODUCTION: Interoceptive feedback to the brain regarding the body's physiological state plays an important role in guiding motivated behaviors. For example, a state of negative energy balance tends to increase exploratory/food-seeking behaviors while reducing avoidance behaviors. We recently reported that overnight food deprivation reduces conditioned passive avoidance behavior in male (but not female) rats. Since fasting increases circulating levels of ghrelin, we hypothesized that ghrelin signaling contributes to the ability of fasting to reduce conditioned avoidance. METHODS: Ad libitum-fed male rats were trained in a passive avoidance procedure using mild footshock. Later, following overnight food deprivation, the same rats were pretreated with ghrelin receptor antagonist (GRA) or saline vehicle 30 min before avoidance testing. RESULTS: GRA restored passive avoidance in fasted rats as measured by both latency to enter and time spent in the shock-paired context. In addition, compared to vehicle-injected fasted rats, fasted rats that received GRA before reexposure to the shock-paired context displayed more cFos activation of prolactin-releasing peptide (PrRP)-positive noradrenergic (NA) neurons in the caudal nucleus of the solitary tract, accompanied by more cFos activation in downstream target sites of PrRP neurons (i.e., bed nucleus of the stria terminalis and paraventricular nucleus of the hypothalamus). DISCUSSION: These results support the view that ghrelin signaling contributes to the inhibitory effect of fasting on learned passive avoidance behavior, perhaps by suppressing recruitment of PrRP-positive NA neurons and their downstream hypothalamic and limbic forebrain targets.


Asunto(s)
Ghrelina , Receptores de Ghrelina , Ratas , Masculino , Animales , Ghrelina/farmacología , Ratas Sprague-Dawley , Ayuno , Núcleo Hipotalámico Paraventricular
3.
J Neurosci ; 39(14): 2649-2663, 2019 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-30683681

RESUMEN

The anterior lateral bed nucleus of the stria terminalis (alBST) expresses glucagon-like peptide-1 receptors (GLP1Rs) and receives input from caudal brainstem GLP1 neurons. GLP1 administered centrally reduces food intake and increases anxiety-like behavior and plasma corticosterone (cort) levels in rats, whereas central GLP1R antagonism has opposite effects. Anxiogenic threats and other stressors robustly activate c-fos expression in both GLP1-producing neurons and also in neurons within alBST subregions expressing GLP1R. To examine the functional role of GLP1R signaling within the alBST, adult male Sprague Dawley rats received bilateral alBST-targeted injections of an adeno-associated virus (AAV) vector expressing short hairpin RNA (shRNA) to knock down the translation of GLP1R mRNA (GLP1R-KD rats), or similar injections of a control AAV (CTRL rats). In situ hybridization revealed that GLP1R mRNA is expressed in a subset of GABAergic alBST neurons, and quantitative real-time PCR confirmed that GLP1R-KD rats displayed a significant 60% reduction in translatable GLP1R mRNA. Compared with CTRL rats, GLP1R-KD rats gained more body weight over time and displayed less anxiety-like behavior, including a loss of light-enhanced acoustic startle and less stress-induced hypophagia. Conversely, while baseline plasma cort levels were similar in GLP1R-KD and CTRL rats, GLP1R-KD rats displayed a prolonged stress-induced elevation of plasma cort levels. GLP1R-KD and CTRL rats displayed similar home cage food intake and a similar hypophagic response to systemic Exendin-4, a GLP1R agonist that crosses the blood-brain barrier. We conclude that GLP1R expressed within the alBST contributes to multiple behavioral responses to anxiogenic threats, yet also serves to limit the plasma cort response to acute stress.SIGNIFICANCE STATEMENT Anxiety is an affective and physiological state that supports threat avoidance. Identifying the neural bases of anxiety-like behaviors in animal models is essential for understanding mechanisms that contribute to normative and pathological anxiety in humans. In rats, anxiety/avoidance behaviors can be elicited or enhanced by visceral or cognitive threats that increase glucagon-like peptide-1 (GLP1) signaling from the caudal brainstem to the hypothalamus and limbic forebrain. Data reported here support a role for limbic GLP1 receptor signaling to enhance anxiety-like behavior and to attenuate stress-induced elevations in plasma cort levels in rats. Improved understanding of central GLP1 neural pathways that impact emotional responses to stress could expand potential therapeutic options for anxiety and other stress-related disorders in humans.


Asunto(s)
Ansiedad/metabolismo , Regulación del Apetito/fisiología , Corticosterona/sangre , Receptor del Péptido 1 Similar al Glucagón/metabolismo , ARN Mensajero/metabolismo , Núcleos Septales/metabolismo , Estrés Psicológico/sangre , Animales , Ansiedad/prevención & control , Ansiedad/psicología , Regulación del Apetito/efectos de los fármacos , Biomarcadores/sangre , Receptor del Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Receptor del Péptido 1 Similar al Glucagón/genética , Masculino , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/fisiología , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética , ARN Interferente Pequeño/administración & dosificación , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/fisiología , Estrés Psicológico/genética , Estrés Psicológico/psicología
4.
J Neurosci ; 39(49): 9767-9781, 2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31666353

RESUMEN

Stress responses are coordinated by widespread neural circuits. Homeostatic and psychogenic stressors activate preproglucagon (PPG) neurons in the caudal nucleus of the solitary tract (cNTS) that produce glucagon-like peptide-1; published work in rodents indicates that these neurons play a crucial role in stress responses. While the axonal targets of PPG neurons are well established, their afferent inputs are unknown. Here we use retrograde tracing with cholera toxin subunit b to show that the cNTS in male and female mice receives axonal inputs similar to those reported in rats. Monosynaptic and polysynaptic inputs specific to cNTS PPG neurons were revealed using Cre-conditional pseudorabies and rabies viruses. The most prominent sources of PPG monosynaptic input include the lateral (LH) and paraventricular (PVN) nuclei of the hypothalamus, parasubthalamic nucleus, lateral division of the central amygdala, and Barrington's nucleus (Bar). Additionally, PPG neurons receive monosynaptic vagal sensory input from the nodose ganglia and spinal sensory input from the dorsal horn. Sources of polysynaptic input to cNTS PPG neurons include the hippocampal formation, paraventricular thalamus, and prefrontal cortex. Finally, cNTS-projecting neurons within PVN, LH, and Bar express the activation marker cFOS in mice after restraint stress, identifying them as potential sources of neurogenic stress-induced recruitment of PPG neurons. In summary, cNTS PPG neurons in mice receive widespread monosynaptic and polysynaptic input from brain regions implicated in coordinating behavioral and physiological stress responses, as well as from vagal and spinal sensory neurons. Thus, PPG neurons are optimally positioned to integrate signals of homeostatic and psychogenic stress.SIGNIFICANCE STATEMENT Recent research has indicated a crucial role for glucagon-like peptide-1-producing preproglucagon (PPG) neurons in regulating both appetite and behavioral and autonomic responses to acute stress. Intriguingly, the central glucagon-like peptide-1 system defined in rodents is conserved in humans, highlighting the translational importance of understanding its anatomical organization. Findings reported here indicate that PPG neurons receive significant monosynaptic and polysynaptic input from brain regions implicated in autonomic and behavioral responses to stress, as well as direct input from vagal and spinal sensory neurons. Improved understanding of the neural pathways underlying the recruitment of PPG neurons may facilitate the development of novel therapies for the treatment of stress-related disorders.


Asunto(s)
Neuronas/fisiología , Proglucagón/fisiología , Sinapsis/fisiología , Nervio Vago/fisiología , Animales , Axones/fisiología , Femenino , Hipotálamo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/fisiología , Neuronas Aferentes/fisiología , Células del Asta Posterior/fisiología , Reflejo Monosináptico/fisiología , Restricción Física , Núcleo Solitario/citología , Núcleo Solitario/fisiología , Estrés Psicológico/fisiopatología , Tálamo/fisiología
5.
Am J Physiol Regul Integr Comp Physiol ; 318(5): R1014-R1023, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32292065

RESUMEN

In rats, overnight fasting reduces the ability of systemic cholecystokinin-8 (CCK) to suppress food intake and to activate cFos in the caudal nucleus of the solitary tract (cNTS), specifically within glucagon-like peptide-1 (GLP-1) and noradrenergic (NA) neurons of the A2 cell group. Systemic CCK increases vagal sensory signaling to the cNTS, an effect that is amplified by leptin and reduced by ghrelin. Since fasting reduces plasma leptin and increases plasma ghrelin levels, we hypothesized that peripheral leptin administration and/or antagonism of ghrelin receptors in fasted rats would rescue the ability of CCK to activate GLP-1 neurons and a caudal subset of A2 neurons that coexpress prolactin-releasing peptide (PrRP). To test this, cFos expression was examined in ad libitum-fed and overnight food-deprived (DEP) rats after intraperitoneal CCK, after coadministration of leptin and CCK, or after intraperitoneal injection of a ghrelin receptor antagonist (GRA) before CCK. In fed rats, CCK activated cFos in ~60% of GLP-1 and PrRP neurons. Few or no GLP-1 or PrRP neurons expressed cFos in DEP rats treated with CCK alone, CCK combined with leptin, or GRA alone. However, GRA pretreatment increased the ability of CCK to activate GLP-1 and PrRP neurons and also enhanced the hypophagic effect of CCK in DEP rats. Considered together, these new findings suggest that reduced behavioral sensitivity to CCK in fasted rats is at least partially due to ghrelin-mediated suppression of hindbrain GLP-1 and PrRP neural responsiveness to CCK.


Asunto(s)
Regulación del Apetito/efectos de los fármacos , Colecistoquinina/administración & dosificación , Ingestión de Alimentos/efectos de los fármacos , Ayuno/metabolismo , Conducta Alimentaria/efectos de los fármacos , Ghrelina/sangre , Neuronas/efectos de los fármacos , Rombencéfalo/efectos de los fármacos , Animales , Péptido 1 Similar al Glucagón/metabolismo , Leptina/sangre , Masculino , Neuronas/metabolismo , Hormona Liberadora de Prolactina/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Sprague-Dawley , Receptores de Ghrelina/metabolismo , Rombencéfalo/metabolismo , Transducción de Señal
6.
J Neurosci ; 37(2): 362-370, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-28077715

RESUMEN

Cisplatin chemotherapy is commonly used to treat cancer despite severe energy balance side effects. In rats, cisplatin activates nucleus tractus solitarius (NTS) projections to the lateral parabrachial nucleus (lPBN) and calcitonin-gene related peptide (CGRP) projections from the lPBN to the central nucleus of the amygdala (CeA). We demonstrated previously that CeA glutamate receptor signaling mediates cisplatin-induced anorexia and body weight loss. Here, we used neuroanatomical tracing, immunofluorescence, and confocal imaging to demonstrate that virtually all NTS→lPBN and lPBN→CeA CGRP projections coexpress vesicular glutamate transporter 2 (VGLUT2), providing evidence that excitatory projections mediate cisplatin-induced energy balance dysregulation. To test whether lPBN→CeA projection neurons are required for cisplatin-induced anorexia and weight loss, we inhibited these neurons chemogenetically using a retrograde Cre-recombinase-expressing canine adenovirus-2 in combination with Cre-dependent inhibitory Designer Receptors Exclusive Activated by Designer Drugs (DREADDs) before cisplatin treatment. Inhibition of lPBN→CeA neurons attenuated cisplatin-induced anorexia and body weight loss significantly. Using a similar approach, we additionally demonstrated that inhibition of NTS→lPBN neurons attenuated cisplatin-induced anorexia and body weight loss significantly. Together, our data support the view that excitatory hindbrain-forebrain projections are necessary for cisplatin's untoward effects on energy intake, elucidating a key neuroanatomical circuit driving pathological anorexia and weight loss that accompanies chemotherapy treatment. SIGNIFICANCE STATEMENT: Chemotherapy treatments are commonly used to treat cancers despite accompanying anorexia and weight loss that may limit treatment adherence and reduce patient quality of life. Strikingly, we lack a neural understanding of, and effective treatments for, chemotherapy-induced anorexia and weight loss. The current data characterize the excitatory nature of neural projections activated by cisplatin in rats and reveal the necessity of specific hindbrain-forebrain projections for cisplatin-induced anorexia and weight loss. Together, these findings help to characterize the neural mechanisms mediating cisplatin-induced anorexia, advancing opportunities to develop better-tolerated chemotherapies and adjuvant therapies to prevent anorexia and concurrent nutritional deficiencies during cancer treatment.


Asunto(s)
Amígdala del Cerebelo/fisiología , Anorexia/inducido químicamente , Cisplatino/toxicidad , Núcleos Parabraquiales/fisiología , Núcleo Solitario/fisiología , Pérdida de Peso/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Anorexia/fisiopatología , Antineoplásicos/toxicidad , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Masculino , Núcleos Parabraquiales/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Núcleo Solitario/efectos de los fármacos , Pérdida de Peso/efectos de los fármacos
7.
Int J Neuropsychopharmacol ; 20(10): 855-860, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28977522

RESUMEN

Background: The ventral subiculum is known to be activated by the presentation of novel stressors. It has been hypothesized that neuronal ensembles at the ventral aspect of the hippocampal formation are involved in context-dependent processing and can guide the learning of appropriate action selections in response to threatening contexts. Artificial activation of the ventral subiculum can excite medium spiny neurons of the nucleus accumbens and can increase the excitability of mesolimbic dopamine neurons via a polysynaptic pathway through the basal ganglia. However, it remains unknown whether this circuit can be activated by aversive experience, and if so, whether ventral subiculum engages nucleus accumbens monosynaptically. Methods: To address this, the retrograde tracer fluorogold was used in rats to label neurons projecting to the caudomedial nucleus accumbens. One to 2 weeks later, the same rats were exposed to psychogenic stress (i.e., acute restraint in a novel test room) or served as nonhandled controls, followed by dual immunocytochemical localization of retrogradely transported tracer and nuclear Fos. Results: Compared with controls, rats exposed to psychogenic stress displayed more fluorogold-positive ventral subiculum neurons that were double-labeled for Fos. Conclusion: This study establishes that the direct pathway from ventral subiculum to the caudomedial nucleus accumbens is activated by stressful experience.


Asunto(s)
Hipocampo/metabolismo , Neuronas/metabolismo , Núcleo Accumbens/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Estrés Psicológico/metabolismo , Animales , Modelos Animales de Enfermedad , Hipocampo/patología , Inmunohistoquímica , Masculino , Vías Nerviosas/metabolismo , Vías Nerviosas/patología , Técnicas de Trazados de Vías Neuroanatómicas , Neuronas/patología , Núcleo Accumbens/patología , Ratas Sprague-Dawley , Restricción Física , Estrés Psicológico/patología
8.
J Neurosci ; 35(30): 10701-14, 2015 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-26224855

RESUMEN

Previous reports indicate that caloric restriction attenuates anxiety and other behavioral responses to acute stress, and blunts the ability of stress to increase anterior pituitary release of adrenocorticotropic hormone. Since hindbrain glucagon-like peptide-1 (GLP-1) neurons and noradrenergic prolactin-releasing peptide (PrRP) neurons participate in behavioral and endocrine stress responses, and are sensitive to the metabolic state, we examined whether overnight food deprivation blunts stress-induced recruitment of these neurons and their downstream hypothalamic and limbic forebrain targets. A single overnight fast reduced anxiety-like behavior assessed in the elevated-plus maze and acoustic startle test, including marked attenuation of light-enhanced startle. Acute stress [i.e., 30 min restraint (RES) or 5 min elevated platform exposure] robustly activated c-Fos in GLP-1 and PrRP neurons in fed rats, but not in fasted rats. Fasting also significantly blunted the ability of acute stress to activate c-Fos expression within the anterior ventrolateral bed nucleus of the stria terminalis (vlBST). Acute RES stress suppressed dark-onset food intake in rats that were fed ad libitum, whereas central infusion of a GLP-1 receptor antagonist blocked RES-induced hypophagia, and reduced the ability of RES to activate PrRP and anterior vlBST neurons in ad libitum-fed rats. Thus, an overnight fast "silences" GLP-1 and PrRP neurons, and reduces both anxiety-like and hypophagic responses to acute stress. The partial mimicking of these fasting-induced effects in ad libitum-fed rats after GLP-1 receptor antagonism suggests a potential mechanism by which short-term negative energy balance attenuates neuroendocrine and behavioral responses to acute stress. SIGNIFICANCE STATEMENT: The results from this study reveal a potential central mechanism for the "metabolic tuning" of stress responsiveness. A single overnight fast, which markedly reduces anxiety-like behavior in rats, reduces or blocks the ability of acute stress to activate hindbrain neurons that are immunoreactive for either prolactin-releasing peptide or glucagon-like peptide 1, and attenuates the activation of their stress-sensitive projection targets in the limbic forebrain. In nonfasted rats, central antagonism of glucagon-like peptide 1 receptors partially mimics the effect of an overnight fast by blocking the ability of acute stress to inhibit food intake, and by attenuating stress-induced activation of hindbrain and limbic forebrain neurons. We propose that caloric restriction attenuates behavioral and physiological responses to acute stress by "silencing" central glucagon-like peptide 1 signaling pathways.


Asunto(s)
Privación de Alimentos/fisiología , Péptido 1 Similar al Glucagón/biosíntesis , Rombencéfalo/metabolismo , Transducción de Señal/fisiología , Estrés Psicológico/metabolismo , Animales , Ansiedad/metabolismo , Restricción Calórica , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Aprendizaje por Laberinto/fisiología , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Ratas , Ratas Sprague-Dawley
9.
Am J Physiol Regul Integr Comp Physiol ; 310(10): R906-16, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26936779

RESUMEN

Published research supports a role for central glucagon-like peptide 1 (GLP-1) signaling in suppressing food intake in rodent species. However, it is unclear whether GLP-1 neurons track food intake and contribute to satiety, and/or whether GLP-1 signaling contributes to stress-induced hypophagia. To examine whether GLP-1 neurons track intake volume, rats were trained to consume liquid diet (LD) for 1 h daily until baseline intake stabilized. On test day, schedule-fed rats consumed unrestricted or limited volumes of LD or unrestricted volumes of diluted (calorically matched to LD) or undiluted Ensure. Rats were perfused after the test meal, and brains processed for immunolocalization of cFos and GLP-1. The large majority of GLP-1 neurons expressed cFos in rats that consumed satiating volumes, regardless of diet type, with GLP-1 activation proportional to intake volume. Since GLP-1 signaling may limit intake only when such large proportions of GLP-1 neurons are activated, a second experiment examined the effect of central GLP-1 receptor (R) antagonism on 2 h intake in schedule-fed rats. Compared with baseline, intracerebroventricular vehicle (saline) suppressed Ensure intake by ∼11%. Conversely, intracerebroventricular injection of vehicle containing GLP-1R antagonist increased intake by ∼14% compared with baseline, partly due to larger second meals. We conclude that GLP-1 neural activation effectively tracks liquid diet intake, that intracerebroventricular injection suppresses intake, and that central GLP-1 signaling contributes to this hypophagic effect. GLP-1 signaling also may contribute to satiety after large volumes have been consumed, but this potential role is difficult to separate from a role in the hypophagic response to intracerebroventricular injection.


Asunto(s)
Ingestión de Alimentos/fisiología , Péptido 1 Similar al Glucagón/metabolismo , Neuronas/fisiología , Fragmentos de Péptidos/farmacología , Rombencéfalo/citología , Estrés Fisiológico , Aclimatación , Animales , Sacarosa en la Dieta , Alimentos Formulados , Receptor del Péptido 1 Similar al Glucagón , Comidas , Ratas , Factores de Tiempo
10.
Nicotine Tob Res ; 18(9): 1920-3, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27178831

RESUMEN

INTRODUCTION: Numerous studies have shown that nicotine (NIC) can enhance the reinforcing effects of non-NIC stimuli through a nonassociative mechanism. To date, it is unclear whether NIC reinforcement enhancement serves to increase behaviors motivated by rewarding stimuli only, or whether NIC potentiates behavior motivated by all stimuli, regardless of valence. METHODS: The current study used a place conditioning procedure to examine whether acute NIC injection modulates avoidance of an environment previously associated with an aversive stimulus. Separate groups of rats underwent place conditioning using either lithium chloride (125mg/kg/ml, i.p.) or footshock (0.75 mA) as the aversive stimulus. Other rats served as nonconditioned controls. The magnitude of place avoidance was assessed after acute NIC (0.1 or 0.4mg/kg/ml, s.c.) or saline. RESULTS: Rats avoided chambers previously paired with either lithium chloride or footshock, and conditioned place avoidance was significantly enhanced by NIC pre-treatment. CONCLUSIONS: These results demonstrate that the ability of NIC to enhance motivated behavior extends to behaviors elicited by aversive stimuli, evidence that NIC affects behavior motivated by a broader range of stimuli than previously appreciated. IMPLICATIONS: The current study examined whether the reinforcement enhancement properties of NIC apply to aversive stimuli by testing NIC enhancement of conditioned place avoidance in rats. The results demonstrate that NIC enhances the motivational impact of these distinct aversive stimuli, providing novel evidence that NIC affects behavior motivated by a broader range of stimuli than has previously been demonstrated.


Asunto(s)
Condicionamiento Operante/efectos de los fármacos , Alimentos , Cloruro de Litio , Nicotina/farmacología , Fumar , Animales , Aprendizaje por Asociación/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Infusiones Parenterales , Masculino , Modelos Animales , Nicotina/administración & dosificación , Ratas , Ratas Sprague-Dawley
11.
Stress ; 18(4): 381-99, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26303312

RESUMEN

This manuscript summarizes the proceedings of the symposium entitled, "Stress, Palatable Food and Reward", that was chaired by Drs. Linda Rinaman and Yvonne Ulrich-Lai at the 2014 Neurobiology of Stress Workshop held in Cincinnati, OH. This symposium comprised research presentations by four neuroscientists whose work focuses on the biological bases for complex interactions among stress, food intake and emotion. First, Dr Ulrich-Lai describes her rodent research exploring mechanisms by which the rewarding properties of sweet palatable foods confer stress relief. Second, Dr Stephanie Fulton discusses her work in which excessive, long-term intake of dietary lipids, as well as their subsequent withdrawal, promotes stress-related outcomes in mice. Third, Dr Mark Wilson describes his group's research examining the effects of social hierarchy-related stress on food intake and diet choice in group-housed female rhesus macaques, and compared the data from monkeys to results obtained in analogous work using rodents. Finally, Dr Gorica Petrovich discusses her research program that is aimed at defining cortical-amygdalar-hypothalamic circuitry responsible for curbing food intake during emotional threat (i.e. fear anticipation) in rats. Their collective results reveal the complexity of physiological and behavioral interactions that link stress, food intake and emotional state, and suggest new avenues of research to probe the impact of genetic, metabolic, social, experiential and environmental factors on these interactions.


Asunto(s)
Conducta de Elección , Ingestión de Alimentos/psicología , Emociones , Conducta Alimentaria/psicología , Estrés Psicológico/psicología , Animales , Dieta , Grasas de la Dieta , Sacarosa en la Dieta , Ingestión de Alimentos/fisiología , Femenino , Sistema Hipotálamo-Hipofisario/metabolismo , Macaca mulatta , Ratones , Sistema Hipófiso-Suprarrenal/metabolismo , Ratas , Recompensa , Estrés Psicológico/metabolismo
12.
Am J Physiol Regul Integr Comp Physiol ; 306(8): R576-85, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24523344

RESUMEN

Leptin released peripherally acts within the central nervous system (CNS) to modulate numerous physiological and behavioral functions. Histochemical identification of leptin-responsive CNS cells can reveal the specific cellular phenotypes and neural circuits through which leptin signaling modulates these functions. Leptin signaling elicits phosphorylation of signal transducer and activator of transcription 3 (pSTAT3), making pSTAT3-immunoreactivity (ir) a useful proxy for identifying leptin-responsive cells. Relatively low systemic doses of leptin (i.e., 10-130 µg/kg body wt) are sufficient to decrease food intake, inhibit gastric emptying, and increase sympathetic activity, but there are no histological reports of central pSTAT3-ir following leptin doses within this range. Considering this, we quantified central pSTAT3-ir in rats after intraperitoneal injections of leptin at doses ranging from 50 to 800 µg/kg body wt. Tissue sections were processed to identify pSTAT3-ir alone or in combination with immunolabeling for cocaine- and amphetamine-regulated transcript (CART), glucagon-like peptide-1 (GLP-1), prolactin-releasing peptide (PrRP), or dopamine-ß-hydroxylase (DßH). Leptin doses as low as 50, 100, and 200 µg/kg body wt significantly increased the number of pSTAT3-ir cells in the arcuate nucleus of the hypothalamus (ARC), nucleus of the solitary tract (NTS), and ventromedial nucleus of the hypothalamus, respectively, and also led to robust pSTAT3 labeling in neural processes. The differential dose-dependent increases in pSTAT3-ir across brain regions provide new information regarding central leptin sensitivity. Within the ARC, CART-ir and pSTAT3-ir were often colocalized, consistent with evidence of leptin sensitivity in this neural population. Conversely, within the NTS, pSTAT3 only rarely colocalized with PrRP and/or DßH, and never with GLP-1.


Asunto(s)
Hipotálamo/efectos de los fármacos , Leptina/farmacología , Neuronas/efectos de los fármacos , Rombencéfalo/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Hipotálamo/metabolismo , Masculino , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Rombencéfalo/metabolismo , Transducción de Señal/efectos de los fármacos , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/metabolismo
13.
Am J Physiol Regul Integr Comp Physiol ; 306(5): R341-51, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24430885

RESUMEN

Signals from the vestibular system, area postrema, and forebrain elicit nausea and vomiting, but gastrointestinal (GI) vagal afferent input arguably plays the most prominent role in defense against food poisoning. It is difficult to determine the contribution of GI vagal afferent input on emesis because various agents (e.g., chemotherapy) often act on multiple sensory pathways. Intragastric copper sulfate (CuSO4) potentially provides a specific vagal emetic stimulus, but its actions are not well defined in musk shrews (Suncus murinus), a primary small animal model used to study emesis. The aims of the current study were 1) to investigate the effects of subdiaphragmatic vagotomy on CuSO4-induced emesis and 2) to conduct preliminary transneuronal tracing of the GI-brain pathways in musk shrews. Vagotomy failed to inhibit the number of emetic episodes produced by optimal emetic doses of CuSO4 (60 and 120 mg/kg ig), but the effects of lower doses were dependent on an intact vagus (20 and 40 mg/kg). Vagotomy also failed to affect emesis produced by motion (1 Hz, 10 min) or nicotine administration (5 mg/kg sc). Anterograde transport of the H129 strain of herpes simplex virus-1 from the ventral stomach wall identified the following brain regions as receiving inputs from vagal afferents: the nucleus of the solitary tract, area postrema, and lateral parabrachial nucleus. These data indicate that the contribution of vagal pathways to intragastric CuSO4-induced emesis is dose dependent in musk shrews. Furthermore, the current neural tracing data suggest brain stem anatomical circuits that are activated by GI signaling in the musk shrew.


Asunto(s)
Sulfato de Cobre/toxicidad , Eméticos/toxicidad , Musarañas/fisiología , Nervio Vago/fisiología , Vómitos/inducido químicamente , Animales , Femenino , Herpesvirus Humano 1/clasificación , Herpesvirus Humano 1/fisiología , Masculino , Mareo por Movimiento , Nicotina/toxicidad , Ratas , Ratas Sprague-Dawley , Estómago/inervación , Estómago/virología , Vagotomía
14.
bioRxiv ; 2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38645069

RESUMEN

Background: Visceral feedback from the body is often subconscious, but plays an important role in guiding motivated behaviors. Vagal sensory neurons relay "gut feelings" to noradrenergic (NA) neurons in the caudal nucleus of the solitary tract (cNTS), which in turn project to the anterior ventrolateral bed nucleus of the stria terminalis (vlBNST) and other hypothalamic-limbic forebrain regions. Prior work supports a role for these circuits in modulating memory consolidation and extinction, but a potential role in retrieval of conditioned avoidance remains untested. Results: To examine this, adult male rats underwent passive avoidance conditioning. We then lesioned gut-sensing vagal afferents by injecting cholecystokinin-conjugated saporin toxin (CSAP) into the vagal nodose ganglia (Experiment 1), or lesioned NA inputs to the vlBNST by injecting saporin toxin conjugated to an antibody against dopamine-beta hydroxylase (DSAP) into the vlBNST (Experiment 2). When avoidance behavior was later assessed, rats with vagal CSAP lesions or NA DSAP lesions displayed significantly increased conditioned passive avoidance. Conclusions: These new findings support the view that a gut vagal afferent-to-cNTSNA-to-vlBNST circuit plays a role in modulating the expression/retrieval of learned passive avoidance. Overall, our data suggest a dynamic modulatory role of vagal sensory feedback to the limbic forebrain in integrating interoceptive signals with contextual cues that elicit conditioned avoidance behavior.

15.
Obesity (Silver Spring) ; 32(8): 1425-1440, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39010249

RESUMEN

In April 2023, the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), in partnership with the National Institute of Child Health and Human Development, the National Institute on Aging, and the Office of Behavioral and Social Sciences Research, hosted a 2-day online workshop to discuss neural plasticity in energy homeostasis and obesity. The goal was to provide a broad view of current knowledge while identifying research questions and challenges regarding neural systems that control food intake and energy balance. This review includes highlights from the meeting and is intended both to introduce unfamiliar audiences with concepts central to energy homeostasis, feeding, and obesity and to highlight up-and-coming research in these areas that may be of special interest to those with a background in these fields. The overarching theme of this review addresses plasticity within the central and peripheral nervous systems that regulates and influences eating, emphasizing distinctions between healthy and disease states. This is by no means a comprehensive review because this is a broad and rapidly developing area. However, we have pointed out relevant reviews and primary articles throughout, as well as gaps in current understanding and opportunities for developments in the field.


Asunto(s)
Dieta , Metabolismo Energético , Plasticidad Neuronal , Obesidad , Humanos , Metabolismo Energético/fisiología , Plasticidad Neuronal/fisiología , Obesidad/fisiopatología , Obesidad/metabolismo , Homeostasis/fisiología , Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Animales
16.
Eur J Neurosci ; 37(8): 1340-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23368289

RESUMEN

The α2 adrenergic receptor antagonist yohimbine (YO) increases transmitter release from noradrenergic (NA) terminals in cortical and subcortical brain regions, including the bed nucleus of the stria terminalis (BST). YO activates the hypothalamic-pituitary-adrenal (HPA) stress axis and is potently anxiogenic in rats and humans. We previously reported that hindbrain NA neurons within the caudal nucleus of the solitary tract (NST-A2/C2) and ventrolateral medulla (VLM-A1/C1) that innervate the anterior ventrolateral (vl)BST contribute to the ability of YO to activate the HPA stress axis in rats. To determine whether the same NA pathway also contributes to YO-induced anxiogenesis in the elevated plus maze (EPMZ), a selective saporin ribotoxin conjugate (dopamine beta hydroxylase conjugated to saporin toxin, DSAP) was microinjected bilaterally into the anterior vlBST to destroy its NA inputs. Sham-lesioned controls were microinjected with vehicle. Two experiments were conducted to determine DSAP lesion effects on EPMZ behavior. DSAP lesions did not alter maze behavior in rats after intraperitoneal saline, and did not alter the significant effect of prior maze experience to reduce exploratory and open arm maze activities. However, in maze-naïve rats, DSAP lesions abolished YO anxiogenesis in the EPMZ. Post-mortem immunocytochemical analyses confirmed that DSAP consistently ablated caudal NST-A2/C2 and VLM-A1/C1 neurons that innervate the anterior vlBST. DSAP lesions did not destroy non-NA inputs to the anterior vlBST, and produced inconsistent cell loss within the pontine locus coeruleus (A6 cell group) that was unrelated to YO anxiogenesis. Thus, the ability of YO to increase anxiety-like behavior in the EPMZ depends on hindbrain NA neurons that target the anterior vlBST.


Asunto(s)
Neuronas Adrenérgicas/fisiología , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Ansiedad/fisiopatología , Rombencéfalo/fisiología , Yohimbina/farmacología , Neuronas Adrenérgicas/citología , Neuronas Adrenérgicas/efectos de los fármacos , Animales , Ansiedad/inducido químicamente , Sistema Hipotálamo-Hipofisario/metabolismo , Inmunohistoquímica , Masculino , Sistema Hipófiso-Suprarrenal/metabolismo , Ratas , Ratas Sprague-Dawley , Rombencéfalo/citología , Rombencéfalo/efectos de los fármacos
17.
Br J Pharmacol ; 179(4): 642-658, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34050926

RESUMEN

Neuroendocrine, behavioural and autonomic responses to stressful stimuli are orchestrated by complex neural circuits. The caudal nucleus of the solitary tract (cNTS) in the dorsomedial hindbrain is uniquely positioned to integrate signals of both interoceptive and psychogenic stress. Within the cNTS, glucagon-like peptide-1 (GLP-1) and prolactin-releasing peptide (PrRP) neurons play crucial roles in organising neural responses to a broad range of stressors. In this review we discuss the anatomical and functional overlap between PrRP and GLP-1 neurons. We outline their co-activation in response to stressful stimuli and their importance as mediators of behavioural and physiological stress responses. Finally, we review evidence that PrRP neurons are downstream of GLP-1 neurons and outline unexplored areas of the research field. Based on the current state-of-knowledge, PrRP and GLP-1 neurons may be compelling targets in the treatment of stress-related disorders. LINKED ARTICLES: This article is part of a themed issue on GLP1 receptor ligands (BJP 75th Anniversary). To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.4/issuetoc.


Asunto(s)
Péptido 1 Similar al Glucagón , Núcleo Solitario , Neuronas/metabolismo , Hormona Liberadora de Prolactina/metabolismo , Núcleo Solitario/metabolismo , Estrés Fisiológico/fisiología
18.
Mol Metab ; 66: 101631, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36368622

RESUMEN

OBJECTIVE: The glucagon gene (Gcg) encodes preproglucagon, which is cleaved to form glucagon-like peptide 1 (GLP1) and other mature signaling molecules implicated in metabolic functions. To date there are no transgenic rat models available for precise manipulation of GLP1-expressing cells in the brain and periphery. METHODS: To visualize and manipulate Gcg-expressing cells in rats, CRISPR/Cas9 was used to express iCre under control of the Gcg promoter. Gcg-Cre rats were bred with tdTomato reporter rats to tag Gcg-expressing cells. Cre-dependent AAVs and RNAscope in situ hybridization were used to evaluate the specificity of iCre expression by GLP1 neurons in the caudal nucleus of the solitary tract (cNTS) and intermediate reticular nucleus (IRt), and by intestinal and pancreatic secretory cells. Food intake was assessed in heterozygous (Het) Gcg-Cre rats after chemogenetic stimulation of cNTS GLP1 neurons expressing an excitatory DREADD. RESULTS: While genotype has minimal effect on body weight or composition in chow-fed Gcg-Cre rats, homozygous (Homo) rats have lower plasma glucose levels. In neonatal and adult Gcg-Cre/tdTom rats, reporter-labeled cells are present in the cNTS and IRt, and in additional brain regions (e.g., basolateral amygdala, piriform cortex) that lack detectable Gcg mRNA in adults but display transient developmental or persistently low Gcg expression. Compared to wildtype (WT) rats, hindbrain Gcg mRNA and GLP1 protein in brain and plasma are markedly reduced in Homo Gcg-Cre rats. Chemogenetic stimulation of cNTS GLP1 neurons reduced overnight chow intake in males but not females, the effect in males was blocked by antagonism of central GLP1 receptors, and hypophagia was enhanced when combined with a subthreshold dose of cholecystokinin-8 to stimulate gastrointestinal vagal afferents. CONCLUSIONS: Gcg-Cre rats are a novel and valuable experimental tool for analyzing the development, anatomy, and function of Gcg-expressing cells in the brain and periphery. In addition, Homo Gcg-Cre rats are a unique model for assessing the role of Gcg-encoded proteins in glucose homeostasis and energy metabolism.


Asunto(s)
Células Secretoras de Glucagón , Glucagón , Masculino , Animales , Ratas , Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Péptido 1 Similar al Glucagón/genética , Péptido 1 Similar al Glucagón/metabolismo , Núcleo Solitario/metabolismo , ARN Mensajero/metabolismo
19.
Am J Physiol Regul Integr Comp Physiol ; 300(2): R222-35, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20962208

RESUMEN

Central noradrenergic (NA) signaling is broadly implicated in behavioral and physiological processes related to attention, arousal, motivation, learning and memory, and homeostasis. This review focuses on the A2 cell group of NA neurons, located within the hindbrain dorsal vagal complex (DVC). The intra-DVC location of A2 neurons supports their role in vagal sensory-motor reflex arcs and visceral motor outflow. A2 neurons also are reciprocally connected with multiple brain stem, hypothalamic, and limbic forebrain regions. The extra-DVC connections of A2 neurons provide a route through which emotional and cognitive events can modulate visceral motor outflow and also a route through which interoceptive feedback from the body can impact hypothalamic functions as well as emotional and cognitive processing. This review considers some of the hallmark anatomical and chemical features of A2 neurons, followed by presentation of evidence supporting a role for A2 neurons in modulating food intake, affective behavior, behavioral and physiological stress responses, emotional learning, and drug dependence. Increased knowledge about the organization and function of the A2 cell group and the neural circuits in which A2 neurons participate should contribute to a better understanding of how the brain orchestrates adaptive responses to the various threats and opportunities of life and should further reveal the central underpinnings of stress-related physiological and emotional dysregulation.


Asunto(s)
Sistema Nervioso Autónomo/fisiología , Conducta Animal/fisiología , Cognición/fisiología , Células Neuroendocrinas/fisiología , Neuronas/fisiología , Norepinefrina/metabolismo , Rombencéfalo/citología , Rombencéfalo/fisiología , Animales , Neuronas/citología , Roedores
20.
Neurobiol Stress ; 15: 100363, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34277897

RESUMEN

We previously reported that GABAergic neurons within the ventral anterior lateral bed nucleus of the stria terminalis (alBST) express glucagon-like peptide 1 receptor (GLP1R) in rats, and that virally-mediated "knock-down" of GLP1R expression in the alBST prolongs the hypothalamic-pituitary-adrenal axis response to acute stress. Given other evidence that a GABAergic projection pathway from ventral alBST serves to limit stress-induced activation of the HPA axis, we hypothesized that GLP1 signaling promotes activation of GABAergic ventral alBST neurons that project directly to the paraventricular nucleus of the hypothalamus (PVN). After PVN microinjection of fluorescent retrograde tracer followed by preparation of ex vivo rat brain slices, whole-cell patch clamp recordings were made in identified PVN-projecting neurons within the ventral alBST. Bath application of Exendin-4 (a specific GLP1R agonist) indirectly depolarized PVN-projecting neurons in the ventral alBST and adjacent hypothalamic parastrial nucleus (PS) through a network-dependent increase in excitatory synaptic inputs, coupled with a network-independent reduction in inhibitory inputs. Additional retrograde tracing experiments combined with in situ hybridization confirmed that PVN-projecting neurons within the ventral alBST/PS are GABAergic, and do not express GLP1R mRNA. Conversely, GLP1R mRNA is expressed by a subset of neurons that project into the ventral alBST and were likely contained within coronal ex vivo slices, including GABAergic neurons within the oval subnucleus of the dorsal alBST and glutamatergic neurons within the substantia innominata. Our novel findings reveal potential GLP1R-mediated mechanisms through which the alBST exerts inhibitory control over the endocrine HPA axis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA