Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Physiol Rev ; 103(2): 1423-1485, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36422994

RESUMEN

The cephalic phase insulin response (CPIR) is classically defined as a head receptor-induced early release of insulin during eating that precedes a postabsorptive rise in blood glucose. Here we discuss, first, the various stimuli that elicit the CPIR and the sensory signaling pathways (sensory limb) involved; second, the efferent pathways that control the various endocrine events associated with eating (motor limb); and third, what is known about the central integrative processes linking the sensory and motor limbs. Fourth, in doing so, we identify open questions and problems with respect to the CPIR in general. Specifically, we consider test conditions that allow, or may not allow, the stimulus to reach the potentially relevant taste receptors and to trigger a CPIR. The possible significance of sweetness and palatability as crucial stimulus features and whether conditioning plays a role in the CPIR are also discussed. Moreover, we ponder the utility of the strict classical CPIR definition based on what is known about the effects of vagal motor neuron activation and thereby acetylcholine on the ß-cells, together with the difficulties of the accurate assessment of insulin release. Finally, we weigh the evidence of the physiological and clinical relevance of the cephalic contribution to the release of insulin that occurs during and after a meal. These points are critical for the interpretation of the existing data, and they support a sharper focus on the role of head receptors in the overall insulin response to eating rather than relying solely on the classical CPIR definition.


Asunto(s)
Insulina , Papilas Gustativas , Humanos , Insulina/metabolismo , Gusto/fisiología , Glucemia/metabolismo , Transducción de Señal
2.
Physiol Rev ; 102(2): 689-813, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34486393

RESUMEN

During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.


Asunto(s)
Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Hipotálamo/fisiología , Neuronas/fisiología , Animales , Homeostasis/fisiología , Humanos , Transducción de Señal/fisiología
3.
Proc Natl Acad Sci U S A ; 116(16): 8018-8027, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30923123

RESUMEN

Control of multiple life-critical physiological and behavioral functions requires the hypothalamus. Here, we provide a comprehensive description and rigorous analysis of mammalian intrahypothalamic network architecture. To achieve this at the gray matter region (macroscale) level, macroscale connection (macroconnection) data for the rat hypothalamus were extracted from the primary literature. The dataset indicated the existence of 7,982 (of 16,770 possible) intrahypothalamic macroconnections. Network analysis revealed that the intrahypothalamic macroconnection network (its macroscale subconnectome) is divided into two identical top-level subsystems (or subnetworks), each composed of two nested second-level subsystems. At the top-level, this suggests a deeply integrated network; however, regional grouping of the two second-level subsystems suggested a partial separation between control of physiological functions and behavioral functions. Furthermore, inclusion of four candidate hubs (dominant network nodes) in the second-level subsystem that is associated prominently with physiological control suggests network primacy with respect to this function. In addition, comparison of network analysis with expression of gene markers associated with inhibitory (GAD65) and excitatory (VGLUT2) neurotransmission revealed a significant positive correlation between measures of network centrality (dominance) and the inhibitory marker. We discuss these results in relation to previous understandings of hypothalamic organization and provide, and selectively interrogate, an updated hypothalamus structure-function network model to encourage future hypothesis-driven investigations of identified hypothalamic subsystems.


Asunto(s)
Conectoma , Hipotálamo , Vías Nerviosas , Animales , Biología Computacional , Hipotálamo/anatomía & histología , Hipotálamo/fisiología , Masculino , Modelos Neurológicos , Vías Nerviosas/anatomía & histología , Vías Nerviosas/fisiología , Ratas , Ratas Sprague-Dawley
4.
Am J Physiol Regul Integr Comp Physiol ; 314(6): R811-R823, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29384699

RESUMEN

Hindbrain catecholamine neurons convey gut-derived metabolic signals to an interconnected neuronal network in the hypothalamus and adjacent forebrain. These neurons are critical for short-term glycemic control, glucocorticoid and glucoprivic feeding responses, and glucagon-like peptide 1 (GLP-1) signaling. Here we investigate whether these pathways also contribute to long-term energy homeostasis by controlling obesogenic sensitivity to a high-fat/high-sucrose choice (HFSC) diet. We ablated hindbrain-originating catecholaminergic projections by injecting anti-dopamine-ß-hydroxylase-conjugated saporin (DSAP) into the paraventricular nucleus of the hypothalamus (PVH) of male rats fed a chow diet for up to 12 wk or a HFSC diet for 8 wk. We measured the effects of DSAP lesions on food choices; visceral adiposity; plasma glucose, insulin, and leptin; and indicators of long-term ACTH and corticosterone secretion. We also determined lesion effects on the number of carbohydrate or fat calories required to increase visceral fat. Finally, we examined corticotropin-releasing hormone levels in the PVH and arcuate nucleus expression of neuropeptide Y ( Npy), agouti-related peptide ( Agrp), and proopiomelanocortin ( Pomc). DSAP-injected chow-fed rats slowly increase visceral adiposity but quickly develop mild insulin resistance and elevated blood glucose. DSAP-injected HFSC-fed rats, however, dramatically increase food intake, body weight, and visceral adiposity beyond the level in control HFSC-fed rats. These changes are concomitant with 1) a reduction in the number of carbohydrate calories required to generate visceral fat, 2) abnormal Npy, Agrp, and Pomc expression, and 3) aberrant control of insulin secretion and glucocorticoid negative feedback. Long-term metabolic adaptations to high-carbohydrate diets, therefore, require intact forebrain catecholamine projections. Without them, animals cannot alter forebrain mechanisms to restrain increased visceral adiposity.


Asunto(s)
Catecolaminas/metabolismo , Red Nerviosa/fisiopatología , Obesidad/fisiopatología , Prosencéfalo/fisiopatología , Animales , Glucemia/metabolismo , Dieta , Dopamina beta-Hidroxilasa/antagonistas & inhibidores , Ingestión de Energía , Insulina/sangre , Grasa Intraabdominal/efectos de los fármacos , Leptina/sangre , Masculino , Vías Nerviosas/fisiopatología , Núcleo Hipotalámico Paraventricular , Ratas , Ratas Sprague-Dawley , Saporinas/farmacología
5.
Am J Physiol Regul Integr Comp Physiol ; 315(4): R708-R720, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29847161

RESUMEN

Endogenous intestinal glucagon-like peptide-1 (GLP-1) controls satiation and glucose metabolism via vagal afferent neurons (VANs). Recently, VANs have received increasing attention for their role in brown adipose tissue (BAT) thermogenesis. It is, however, unclear whether VAN GLP-1 receptor (GLP-1R) signaling affects BAT thermogenesis and energy expenditure (EE) and whether this VAN mechanism contributes to energy balance. First, we tested the effect of the GLP-1R agonist exendin-4 (Ex4, 0.3 µg/kg ip) on EE and BAT thermogenesis and whether these effects require VAN GLP-1R signaling using a rat model with a selective Glp1r knockdown (kd) in VANs. Second, we examined the role of VAN GLP-1R in energy balance during chronic high-fat diet (HFD) feeding in VAN Glp1r kd rats. Finally, we used viral transsynaptic tracers to identify the possible neuronal substrates of such a gut-BAT interaction. VAN Glp1r kd attenuated the acute suppressive effects of Ex4 on EE and BAT thermogenesis. Consistent with this finding, the VAN Glp1r kd increased EE and BAT activity, diminished body weight gain, and improved insulin sensitivity compared with HFD-fed controls. Anterograde transsynaptic viral tracing of VANs infected major hypothalamic and hindbrain areas involved in BAT sympathetic regulation. Moreover, retrograde tracing from BAT combined with laser capture microdissection revealed that a population of VANs expressing Glp1r is synaptically connected to the BAT. Our findings reveal a novel role of VAN GLP-1R signaling in the regulation of EE and BAT thermogenesis and imply that through this gut-brain-BAT connection, intestinal GLP-1 plays a role in HFD-induced metabolic syndrome.


Asunto(s)
Tejido Adiposo Pardo/inervación , Sistema Nervioso Autónomo/metabolismo , Encéfalo/metabolismo , Metabolismo Energético , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Intestinos/inervación , Termogénesis , Animales , Sistema Nervioso Autónomo/efectos de los fármacos , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Exenatida/farmacología , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/genética , Incretinas/farmacología , Masculino , Vías Nerviosas/metabolismo , Neuronas Aferentes/metabolismo , Ratas Sprague-Dawley , Transducción de Señal , Termogénesis/efectos de los fármacos
6.
Am J Physiol Regul Integr Comp Physiol ; 312(3): R324-R337, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28077392

RESUMEN

The brain networks connected to the sympathetic motor and sensory innervations of brown (BAT) and white (WAT) adipose tissues were originally described using two transneuronally transported viruses: the retrogradely transported pseudorabies virus (PRV), and the anterogradely transported H129 strain of herpes simplex virus-1 (HSV-1 H129). Further complexity was added to this network organization when combined injections of PRV and HSV-1 H129 into either BAT or WAT of the same animal generated sets of coinfected neurons in the brain, spinal cord, and sympathetic and dorsal root ganglia. These neurons are well positioned to act as sensorimotor links in the feedback circuits that control each fat pad. We have now determined the extent of sensorimotor crosstalk between interscapular BAT (IBAT) and inguinal WAT (IWAT). PRV152 and HSV-1 H129 were each injected into IBAT or IWAT of the same animal: H129 into IBAT and PRV152 into IWAT. The reverse configuration was applied in a different set of animals. We found single-labeled neurons together with H129+PRV152 coinfected neurons in multiple brain sites, with lesser numbers in the sympathetic and dorsal root ganglia that innervate IBAT and IWAT. We propose that these coinfected neurons mediate sensory-sympathetic motor crosstalk between IBAT and IWAT. Comparing the relative numbers of coinfected neurons between the two injection configurations showed a bias toward IBAT-sensory and IWAT-sympathetic motor feedback loops. These coinfected neurons provide a neuroanatomical framework for functional interactions between IBAT thermogenesis and IWAT lipolysis that occurs with cold exposure, food restriction/deprivation, exercise, and more generally with alterations in adiposity.


Asunto(s)
Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/inervación , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/inervación , Corteza Sensoriomotora/citología , Sistema Nervioso Simpático/citología , Tejido Adiposo Pardo/fisiología , Tejido Adiposo Blanco/fisiología , Animales , Cricetinae , Retroalimentación Sensorial , Masculino , Vías Nerviosas/citología , Vías Nerviosas/fisiología , Neuronas/citología , Neuronas/fisiología , Phodopus , Receptor Cross-Talk , Corteza Sensoriomotora/fisiología , Sistema Nervioso Simpático/fisiología
7.
Am J Physiol Regul Integr Comp Physiol ; 310(11): R1177-85, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27030665

RESUMEN

The consensus view of the ventromedial nucleus of the hypothalamus (VMH) is that it is a key node in the rodent brain network controlling sympathoadrenal counterregulatory responses to hypoglycemia. To identify the location of hypoglycemia-responsive neurons in the VMH, we performed a high spatial resolution Fos analysis in the VMH of rats made hypoglycemic with intraperitoneal injections of insulin. We examined Fos expression in the four constituent parts of VMH throughout its rostrocaudal extent and determined their relationship to blood glucose concentrations. Hypoglycemia significantly decreased Fos expression only in the dorsomedial and central parts of the VMH, but not its anterior or ventrolateral parts. Moreover, the number of Fos-expressing neurons was significantly and positively correlated in the two responsive regions with terminal blood glucose concentrations. We also measured Fos responses in the paraventricular nucleus of the hypothalamus (PVH) and in several levels of the periaqueductal gray (PAG), which receives strong projections from the VMH. We found the expected and highly significant increase in Fos in the neuroendocrine PVH, which was negatively correlated to terminal blood glucose concentrations, but no significant differences were seen in any part of the PAG. Our results show that there are distinct populations of VMH neurons whose Fos expression is suppressed by hypoglycemia, and their numbers correlate with blood glucose. These findings support a clear division of glycemic control functions within the different parts of the VMH.


Asunto(s)
Glucemia/metabolismo , Hipoglucemia/fisiopatología , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo , Animales , Progresión de la Enfermedad , Regulación hacia Abajo , Masculino , Especificidad de Órganos , Ratas , Ratas Wistar , Distribución Tisular
8.
Physiology (Bethesda) ; 29(5): 314-24, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25180261

RESUMEN

Hypoglycemia poses a serious threat to the integrity of the brain, owing to its reliance on blood glucose as a fuel. Protecting against hypoglycemia is an extended network of glucose sensors located within the brain and in the periphery that serve to mediate responses restoring euglycemia, i.e., counterregulatory responses. This review examines the various glucose sensory loci involved in hypoglycemic detection, with a particular emphasis on peripheral glucose sensory loci and their contribution to hypoglycemic counterregulation.


Asunto(s)
Glucemia/análisis , Glucosa/metabolismo , Hipoglucemia/diagnóstico , Hipoglucemia/metabolismo , Hipoglucemiantes/uso terapéutico , Animales , Encéfalo/metabolismo , Humanos , Neuronas/metabolismo
9.
Curr Diab Rep ; 14(3): 465, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24510608

RESUMEN

It is now widely accepted that the brain makes important contributions to the dysregulated glucose metabolism, altered feeding behaviors, and the obesity often seen in type 2 diabetes (T2D). Although studies focusing on genetic, cellular, and molecular regulatory elements in pancreas, liver, adipose tissue etc provide a good understanding of how these processes relate to T2D, our knowledge of how brain wiring patterns are organized is much less developed. This article discusses animal studies that illustrate the importance of understanding the network organization of those brain regions most closely implicated in T2D. It will describe the brain networks, as well as the methodologies used to explore them. To illustrate some of the gaps in our knowledge, we will discuss the connectional network of the ventromedial nucleus and its adjacent cell groups in the hypothalamus; structures that are widely recognized as key elements in the brain's ability to control glycemia, feeding, and body weight.


Asunto(s)
Encéfalo/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Red Nerviosa/fisiopatología , Animales , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Red Nerviosa/metabolismo , Red Nerviosa/patología , Núcleo Hipotalámico Ventromedial/metabolismo , Núcleo Hipotalámico Ventromedial/patología , Núcleo Hipotalámico Ventromedial/fisiopatología
10.
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
11.
Am J Physiol Endocrinol Metab ; 305(1): E78-88, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23651844

RESUMEN

Blood glucose concentration is tightly regulated by the rate of insulin secretion and clearance, a process partially controlled by sensory neurons serving as metabolic sensors in relevant tissues. The activity of these neurons is regulated by the products of metabolism which regulate transmitter release, and recent evidence suggests that neuronally expressed ion channels of the transient receptor potential (TRP) family function in this critical process. Here, we report the novel finding that the cold and menthol-gated channel TRPM8 is necessary for proper insulin homeostasis. Mice lacking TRPM8 respond normally to a glucose challenge while exhibiting prolonged hypoglycemia in response to insulin. Additionally, Trpm8-/- mice have increased rates of insulin clearance compared with wild-type animals and increased expression of insulin-degrading enzyme in the liver. TRPM8 channels are not expressed in the liver, but TRPM8-expressing sensory afferents innervate the hepatic portal vein, suggesting a TRPM8-mediated neuronal control of liver insulin clearance. These results demonstrate that TRPM8 is a novel regulator of serum insulin and support the role of sensory innervation in metabolic homeostasis.


Asunto(s)
Glucemia/metabolismo , Hipoglucemia/genética , Insulina/metabolismo , Células Receptoras Sensoriales/metabolismo , Canales Catiónicos TRPM/genética , Animales , Plásmidos de Bacteriocinas , Diabetes Mellitus Experimental/metabolismo , Homeostasis/fisiología , Hipoglucemia/metabolismo , Células Secretoras de Insulina/metabolismo , Hígado/irrigación sanguínea , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vena Porta/inervación , Ratas , Canales Catiónicos TRPM/metabolismo
12.
J Neuroendocrinol ; 35(11): e13251, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37053148

RESUMEN

Throughout the 20th Century, regulatory peptide discovery advanced from the identification of gut hormones to the extraction and characterization of hypothalamic hypophysiotropic factors, and to the isolation and cloning of multiple brain neuropeptides. These discoveries were followed by the discovery of G-protein-coupled and other membrane receptors for these peptides. Subsequently, the systems physiology associated with some of these multiple regulatory peptides and receptors has been comprehensively elucidated and has led to improved therapeutics and diagnostics and their approval by the US Food and Drug Administration. In light of this wealth of information and further potential, it is truly a time of renaissance for regulatory peptides. In this perspective, we review what we have learned from the pioneers in exemplified fields of gut peptides, such as cholecystokinin, enterochromaffin-like-cell peptides, and glucagon, from the trailblazing studies on the key stress hormone, corticotropin-releasing factor, as well as from more recently characterized relaxin-family peptides and receptors. The historical viewpoints are based on our understanding of these topics in light of the earliest phases of research and on subsequent studies and the evolution of knowledge, aiming to sharpen our vision of the current state-of-the-art and those studies that should be prioritized in the future.


Asunto(s)
Neuropéptidos , Relaxina , Hormona Liberadora de Corticotropina , Colecistoquinina , Glucagón
13.
J Neurosci ; 31(50): 18479-91, 2011 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-22171049

RESUMEN

Physiological responses to hypoglycemia, hyperinsulinemia, and hyperglycemia include a critical adrenocortical component that is initiated by hypothalamic control of the anterior pituitary and adrenal cortex. These adrenocortical responses ensure appropriate long-term glucocorticoid-mediated modifications to metabolism. Despite the importance of these mechanisms to disease processes, how hypothalamic afferent pathways engage the intracellular mechanisms that initiate adrenocortical responses to glycemia-related challenges are unknown. This study explores these mechanisms using network- and cellular-level interventions in in vivo and ex vivo rat preparations. Results show that a hindbrain-originating catecholamine afferent system selectively engages a MAP kinase pathway in rat paraventricular hypothalamic CRH (corticotropin-releasing hormone) neuroendocrine neurons shortly after vascular insulin and 2-deoxyglucose challenges. In turn, this MAP kinase pathway can control both neuroendocrine neuronal firing rate and the state of CREB phosphorylation in a reduced ex vivo paraventricular hypothalamic preparation, making this signaling pathway an ideal candidate for coordinating CRH synthesis and release. These results establish the first clear structural and functional relationships linking neurons in known nutrient-sensing regions with intracellular mechanisms in hypothalamic CRH neuroendocrine neurons that initiate the adrenocortical response to various glycemia-related challenges.


Asunto(s)
Catecolaminas/metabolismo , Hipotálamo/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Sistema Hipófiso-Suprarrenal/metabolismo , Rombencéfalo/metabolismo , Animales , Glucemia/metabolismo , Desoxiglucosa/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotálamo/efectos de los fármacos , Insulina/farmacología , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Rombencéfalo/efectos de los fármacos
14.
Front Neuroendocrinol ; 31(1): 32-43, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19836412

RESUMEN

Glucose is the primary fuel for the vast majority of cells, and animals have evolved essential cellular, autonomic, endocrine, and behavioral measures to counteract both hypo- and hyperglycemia. A central component of these counterregulatory mechanisms is the ability of specific sensory elements to detect changes in blood glucose and then use that information to produce appropriate counterregulatory responses. Here we focus on the organization of the neural systems that are engaged by glucosensing mechanisms when blood glucose concentrations fall to levels that pose a physiological threat. We employ a classic sensory-motor integrative schema to describe the peripheral, hindbrain, and hypothalamic components that make up counterregulatory mechanisms in the brain. We propose that models previously developed to describe how the forebrain modulates autonomic reflex loops in the hindbrain offer a reasoned framework for explaining how counterregulatory neural mechanisms in the hypothalamus and hindbrain are structured.


Asunto(s)
Glucemia/metabolismo , Encéfalo , Homeostasis/fisiología , Hipoglucemia , Red Nerviosa , Encéfalo/patología , Encéfalo/fisiopatología , Glucosa , Humanos , Hipoglucemia/fisiopatología , Masculino , Venas Mesentéricas , Red Nerviosa/patología , Red Nerviosa/fisiopatología , Neuronas/fisiología , Vena Porta
16.
eNeuro ; 8(1)2021.
Artículo en Inglés | MEDLINE | ID: mdl-33495245

RESUMEN

Vagal and spinal sensory endings in the wall of the hepatic portal and superior mesenteric veins (PMV) provide the brain with chemosensory information important for energy balance and other functions. To determine their medullary neuronal targets, we injected the transsynaptic anterograde viral tracer HSV-1 H129-772 (H129) into the PMV wall or left nodose ganglion (LNG) of male rats, followed by immunohistochemistry (IHC) and high-resolution imaging. We also determined the chemical phenotype of H129-infected neurons, and potential vagal and spinal axon terminal appositions in the dorsal motor nucleus of the vagus (DMX) and the nucleus of the solitary tract (NTS). PMV wall injections generated H129-infected neurons in both nodose ganglia and in thoracic dorsal root ganglia (DRGs). In the medulla, cholinergic preganglionic parasympathetic neurons in the DMX were virtually the only targets of chemosensory information from the PMV wall. H129-infected terminal appositions were identified on H129-infected somata and dendrites in the DMX, and on H129-infected DMX dendrites that extend into the NTS. Sensory transmission via vagal and possibly spinal routes from the PMV wall therefore reaches DMX neurons via axo-somatic appositions in the DMX and axo-dendritic appositions in the NTS. However, the dearth of H129-infected NTS neurons indicates that sensory information from the PMV wall terminates on DMX neurons without engaging NTS neurons. These previously underappreciated direct sensory routes into the DMX enable a vago-vagal and possibly spino-vagal reflexes that can directly influence visceral function.


Asunto(s)
Venas Mesentéricas , Ganglio Nudoso , Animales , Masculino , Neuronas , Ratas , Núcleo Solitario , Nervio Vago
17.
Am J Physiol Regul Integr Comp Physiol ; 297(6): R1813-21, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19741057

RESUMEN

Anorexia that accompanies cellular dehydration in rats (DE-anorexia) offers a relatively simple model for investigating the functional organization of neural mechanisms that can suppress feeding during dehydration. Previous studies strongly suggest that the inputs that drive ingestive behavior control neurons in the paraventricular nucleus of the hypothalamus (PVH) and lateral hypothalamic area (LHA) remain active during DE-anorexia. Here we examine whether these two regions retain their sensitivity to neuropeptide Y (NPY). NPY is an important component in two major feeding-related inputs from the arcuate nucleus and the hindbrain. We found that intake responses to NPY injections in the LHA and PVH were suppressed in DE-anorexia, but the PVH remained less sensitive to the effects of NPY than the LHA in DE-anorexic animals. Indeed the higher dose of NPY (238 pmol) completely overcame shorter periods of DE-anorexia when injected into the LHA but not the PVH. However, the latency to eat after NPY injections remained unchanged from control animals, regardless of NPY dose, injection location, or intensity of anorexia. Furthermore, the onset and size of the strong and rapidly induced compensatory feeding that follows the return of water to DE-anorexic animals was also unaffected by any NPY injections. These data support the hypothesis that DE-anorexia develops as a consequence of the premature termination of regularly initiated meals, which perhaps involves processes that alter the sensitivity of satiety mechanisms downstream to the PVH and LHA.


Asunto(s)
Anorexia/metabolismo , Deshidratación/metabolismo , Ingestión de Alimentos , Conducta Alimentaria , Área Hipotalámica Lateral/metabolismo , Neuropéptido Y/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Respuesta de Saciedad , Animales , Anorexia/etiología , Anorexia/psicología , Deshidratación/complicaciones , Deshidratación/psicología , Modelos Animales de Enfermedad , Ingestión de Líquidos , Conducta de Ingestión de Líquido , Inyecciones , Masculino , Neuropéptido Y/administración & dosificación , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción , Factores de Tiempo
18.
J Neurosci ; 27(27): 7344-60, 2007 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-17611287

RESUMEN

Paraventricular hypothalamic (PVH) corticotropin-releasing hormone (CRH) neuroendocrine neurons mount neurosecretory and transcriptional responses to glycemic challenges [intravenous 2-deoxyglucose (2-DG) or insulin]. Although these responses require signals from intact afferents originating from hindbrain CA (catecholaminergic) neurons, the identity of these signals and the mechanisms by which they are transduced by PVH neurons during glycemic challenge remain unclear. Here, we tested whether the prototypical catecholamine, norepinephrine (NE), can reproduce PVH neuroendocrine responses to glycemic challenge. Because these responses include phosphorylation of p44/42 mitogen-activated protein (MAP) kinases [extracellular signal-regulated kinases 1/2 (ERK1/2)], we also determined whether NE activates ERK1/2 in PVH neurons and, if so, by what mechanism. We show that systemic insulin and 2-DG, and PVH-targeted NE microinjections, rapidly elevated PVH phospho-ERK1/2 levels. NE increased Crh and c-fos expression, together with circulating ACTH/corticosterone. However, because injections also increased c-Fos mRNA in other brain regions, we used hypothalamic slices maintained in vitro to clarify whether NE activates PVH neurons without contribution of inputs from distal regions. In slices, bath-applied NE triggered robust phospho-ERK1/2 immunoreactivity in PVH (including CRH) neurons, which attenuated markedly in the presence of the alpha1 adrenoceptor antagonist, prazosin, or the MAP kinase kinase (MEK) inhibitor, U0126 (1,4-diamino-2,3-dicyano-1,4-bis[2-aminophenylthio]butadiene). Therefore, at a systems level, local PVH delivery of NE is sufficient to account for hindbrain activation of CRH neuroendocrine neurons during glycemic challenge. At a cellular level, these data provide the first demonstration that MAP kinase signaling cascades (MEK-->ERK) are intracellular transducers of noradrenergic signals in CRH neurons, and implicate this transduction mechanism as an important component of central neuroendocrine responses during glycemic challenge.


Asunto(s)
Catecolaminas/fisiología , Desoxiglucosa/administración & dosificación , Insulina/administración & dosificación , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Neuronas/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Animales , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Neuronas/efectos de los fármacos , Sistemas Neurosecretores/efectos de los fármacos , Sistemas Neurosecretores/enzimología , Sistemas Neurosecretores/fisiología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
19.
J Comp Neurol ; 526(8): 1287-1306, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29424419

RESUMEN

Virtually all rodent neuroendocrine corticotropin-releasing-hormone (CRH) neurons are in the dorsal medial parvicellular (mpd) part of the paraventricular nucleus of the hypothalamus (PVH). They form the final common pathway for adrenocortical stress responses. Their activity is controlled by sets of GABA-, glutamate-, and catecholamine-containing inputs arranged in an interactive pre-motor network. Defining the nature and arrangement of these inputs can help clarify how stressor type and intensity information is conveyed to neuroendocrine neurons. Here we use immunohistochemistry with high-resolution 3-dimensional image analyses to examine the arrangement of single- and co-occurring GABA, glutamate, and catecholamine markers in synaptophysin-defined pre-synaptic terminals in the PVHmpd of unstressed rats and Crh-IRES-Cre;Ai14 transgenic mice: respectively, vesicular glutamate transporter 2 (VGluT2), vesicular GABA transporter (VGAT), dopamine ß-hydroxylase (DBH), and phenylethanolamine n-methyltransferase (PNMT). Just over half of all PVHmpd pre-synaptic terminals contain VGAT, with slightly less containing VGluT2. The vast majority of terminal appositions with mouse CRH neurons occur non-somatically. However, there are significantly more somatic VGAT than VGluT2 appositions. In the rat PVHmpd, about five times as many pre-synaptic terminals contain PNMT than DBH only. However, because epinephrine release has never been detected in the PVH, PNMT terminals may functionally be noradrenergic not adrenergic. PNMT and VGluT2 co-occur in some pre-synaptic terminals indicating the potential for co-transmission of glutamate and norepinephrine. Collectively, these results provide a structural basis for how GABA/glutamate/catecholamine interactions enable adrenocortical responses to fast-onset interosensory stimuli, and more broadly, how combinations of PVH neurotransmitters and neuromodulators interact dynamically to control adrenocortical activity.


Asunto(s)
Neuronas/citología , Neurotransmisores/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Terminales Presinápticos/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Butiratos/metabolismo , Dopamina beta-Hidroxilasa/metabolismo , Femenino , Proteínas Transportadoras de GABA en la Membrana Plasmática , Masculino , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Hormona Liberadora de Corticotropina/genética , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
20.
Mol Metab ; 11: 33-46, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29650350

RESUMEN

OBJECTIVE: Glucagon-like peptide-1 (GLP-1) neurons in the hindbrain densely innervate the dorsomedial hypothalamus (DMH), a nucleus strongly implicated in body weight regulation and the sympathetic control of brown adipose tissue (BAT) thermogenesis. Therefore, DMH GLP-1 receptors (GLP-1R) are well placed to regulate energy balance by controlling sympathetic outflow and BAT function. METHODS: We investigate this possibility in adult male rats by using direct administration of GLP-1 (0.5 ug) into the DMH, knocking down DMH GLP-1R mRNA with viral-mediated RNA interference, and by examining the neurochemical phenotype of GLP-1R expressing cells in the DMH using in situ hybridization. RESULTS: GLP-1 administered into the DMH increased BAT thermogenesis and hepatic triglyceride (TG) mobilization. On the other hand, Glp1r knockdown (KD) in the DMH increased body weight gain and adiposity, with a concomitant reduction in energy expenditure (EE), BAT temperature, and uncoupling protein 1 (UCP1) expression. Moreover, DMH Glp1r KD induced hepatic steatosis, increased plasma TG, and elevated liver specific de-novo lipogenesis, effects that collectively contributed to insulin resistance. Interestingly, DMH Glp1r KD increased neuropeptide Y (NPY) mRNA expression in the DMH. GLP-1R mRNA in the DMH, however, was found in GABAergic not NPY neurons, consistent with a GLP-1R-dependent inhibition of NPY neurons that is mediated by local GABAergic neurons. Finally, DMH Glp1r KD attenuated the anorexigenic effects of the GLP-1R agonist exendin-4, highlighting an important role of DMH GLP-1R signaling in GLP-1-based therapies. CONCLUSIONS: Collectively, our data show that DMH GLP-1R signaling plays a key role for BAT thermogenesis and adiposity.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Adiposidad , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Hipotálamo/metabolismo , Termogénesis , Animales , Exenatida/metabolismo , Neuronas GABAérgicas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/genética , Resistencia a la Insulina , Lipogénesis , Masculino , Neuropéptido Y/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteína Desacopladora 1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA