1
|
Arcon M. The interplay between hypothalamic and brainstem nuclei in homeostatic control of energy balance. Behav Brain Res 2025; 480:115398. [PMID: 39674373 DOI: 10.1016/j.bbr.2024.115398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/22/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Energy balance and body weight are tightly regulated by homeostatic and hedonic systems of the brain. These systems are ultimately finely tuned by hypothalamic and extrahypothalamic neurocircuitry that modulate feeding and the appetite signalling cascade. The hypothalamus has been extensively researched and its role in homeostatic regulation of energy balance is well established. Later on, evidence indicated that extrahypothalamic signalling also has a critical role in regulation of body mass across the lifespan. One of these brain regions was the brainstem and specifically the dorsal vagal complex (DVC), which comprises of the area postrema (AP), nucleus of the solitary tract (NTS) and dorsal motor complex of the vagus (DMV). These brain stem nuclei were shown to also finely tune feeding behaviour through catecholaminergic, glutamatergic, and GABAergic signals. Moreover, these nuclei also receive afferent signals from the viscera through the gut, as well as humoral input from the bloodstream. Therefore, these brain stem nuclei are deemed as the port of entry where initial appetite-related signals are first conveyed and then modulated to the forebrain to hypothalamic and extrahypothalamic regions such as the arcuate nucleus (ARC) and parabrachial nucleus (PBN). Understanding the intricate interactions and projections between hypothalamic and brainstem nuclei is instrumental to comprehend energy balance regulation as a whole and to potentially address metabolic conditions such as diabetes and obesity. Further research in this area may lead to the development of targeted pharmacological and lifestyle intervention strategies that could lead to mitigation of metabolic disorders and/or promote a healthier body mass across the life span.
Collapse
Affiliation(s)
- Matevz Arcon
- Faculty of Health Sciences, University of Primorska, Izola, Slovenia.
| |
Collapse
|
2
|
Watts AG, Kanoski SE, Sanchez-Watts G, Langhans W. The physiological control of eating: signals, neurons, and networks. Physiol Rev 2022; 102:689-813. [PMID: 34486393 PMCID: PMC8759974 DOI: 10.1152/physrev.00028.2020] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
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.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Graciela Sanchez-Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Eidgenössische Technische Hochschule-Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
3
|
Ramirez-Plascencia OD, Saderi N, Cárdenas Romero S, Flores Sandoval O, Báez-Ruiz A, Martínez Barajas H, Salgado-Delgado R. Temporal dysregulation of hypothalamic integrative and metabolic nuclei in rats fed during the rest phase. Chronobiol Int 2022; 39:374-385. [PMID: 34906015 DOI: 10.1080/07420528.2021.2002352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Temporal coordination of organisms according to the daytime allows a better performance of physiological processes. However, modern lifestyle habits, such as food intake during the rest phase, promote internal desynchronization and compromise homeostasis and health. The hypothalamic suprachiasmatic nucleus (SCN) synchronizes body physiology and behavior with the environmental light-dark cycle by transmitting time information to several integrative hypothalamic nuclei, such as the paraventricular nucleus (PVN), dorsomedial hypothalamic nucleus (DMH) and median preoptic area (MnPO). The SCN receives metabolic information mainly via Neuropeptide Y (NPY) inputs from the intergeniculate nucleus of the thalamus (IGL). Nowadays, there is no evidence of the response of the PVN, DMH and MnPO when the animals are subjected to internal desynchronization by restricting food access to the rest phase of the day. To explore this issue, we compared the circadian activity of the SCN, PVN, DMH and MnPO. In addition, we analyzed the daily activity of the satiety centers of the brainstem, the nucleus of the tractus solitarius (NTS) and area postrema (AP), which send metabolic information to the SCN, directly or via the thalamic intergeniculate leaflet (IGL). For that, male Wistar rats were assigned to three meal protocols: fed during the rest phase (Day Fed); fed during the active phase (Night Fed); free access to food (ad libitum). After 21 d, the daily activity patterns of these nuclei were analyzed by c-Fos immunohistochemistry, as well as NPY immunohistochemistry, in the SCN. The results show that eating during the rest period produces a phase advance in the activity of the SCN, changes the daily activity pattern in the MnPO, NTS and AP and flattens the c-Fos rhythm in the PVN and DMH. Altogether, these results validate previous observations of circadian dysregulation that occurs within the central nervous system when meals are consumed during the rest phase, a behavior that is involved in the metabolic alterations described in the literature.
Collapse
Affiliation(s)
- Oscar D Ramirez-Plascencia
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.,Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Nadia Saderi
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | | | - Omar Flores Sandoval
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Adrián Báez-Ruiz
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | | | | |
Collapse
|
4
|
Khan MS, Spann RA, Münzberg H, Yu S, Albaugh VL, He Y, Berthoud HR, Morrison CD. Protein Appetite at the Interface between Nutrient Sensing and Physiological Homeostasis. Nutrients 2021; 13:4103. [PMID: 34836357 PMCID: PMC8620426 DOI: 10.3390/nu13114103] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/04/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
Feeding behavior is guided by multiple competing physiological needs, as animals must sense their internal nutritional state and then identify and consume foods that meet nutritional needs. Dietary protein intake is necessary to provide essential amino acids and represents a specific, distinct nutritional need. Consistent with this importance, there is a relatively strong body of literature indicating that protein intake is defended, such that animals sense the restriction of protein and adaptively alter feeding behavior to increase protein intake. Here, we argue that this matching of food consumption with physiological need requires at least two concurrent mechanisms: the first being the detection of internal nutritional need (a protein need state) and the second being the discrimination between foods with differing nutritional compositions. In this review, we outline various mechanisms that could mediate the sensing of need state and the discrimination between protein-rich and protein-poor foods. Finally, we briefly describe how the interaction of these mechanisms might allow an animal to self-select between a complex array of foods to meet nutritional needs and adaptively respond to changes in either the external environment or internal physiological state.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Christopher D. Morrison
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; (M.S.K.); (R.A.S.); (H.M.); (S.Y.); (V.L.A.); (Y.H.); (H.-R.B.)
| |
Collapse
|
5
|
Minaya DM, Larson RW, Podlasz P, Czaja K. Glutamate-dependent regulation of food intake is altered with age through changes in NMDA receptor phenotypes on vagal afferent neurons. Physiol Behav 2018; 189:26-31. [PMID: 29476874 DOI: 10.1016/j.physbeh.2018.02.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/19/2018] [Accepted: 02/20/2018] [Indexed: 11/25/2022]
Abstract
Compared to younger individuals, older human subjects have significantly lower food intakes and an increased satiety response. N-methyl-d-aspartate (NMDA) receptors expressed by vagal afferent neurons originating from nodose ganglia (NG) are involved in modulating the satiety response. The present study investigated how NMDA receptor subunit phenotypes in NG neurons change with age and how these age-related alterations in food intake are modulated by presynaptic NMDA receptors in the NG of male Sprague Dawley rats (six week-old and sixty week-old). Food intake was measured at 30-, 60-, and 120-min following intraperitoneal administration of cholecystokinin (CCK) or the non-competitive NMDA receptor antagonist MK-801. Immunofluorescence was used to determine NMDA receptor subunit expression (NR1, NR2B, NR2C, and NR2D) in the NG. The results showed that, CCK reduced food intake at 30-, 60-, and 120-min post injection in both young and the middle-age animals, with no statistical difference between the groups at 30- and 60-min. In contrast, MK-801 produced an increase in food intake that was significantly higher in middle-age rats compared to young animals at all time points studied. NR1 subunit was expressed by almost all NG neurons in both age groups. In young rats, NR2B, NR2C, and NR2D subunits were expressed in 56.1%, 49.3%, and 13.9% of NG neurons, respectively. In contrast, only 30.3% of the neuronal population in middle-aged rats expressed NR2B subunit immunoreactivity, NR2C was present in 34.1%, and only 10.6% of total neurons expressed the NR2D subunit. In conclusion, glutamate-dependent regulation of food intake is altered with age and one of the potential mechanisms through which this age-related changes in intake occur is changes in NMDA receptor phenotypes on vagal afferent neurons located in NG.
Collapse
Affiliation(s)
- Dulce M Minaya
- Department of Veterinary Biosciences and Diagnostic Imaging, The University of Georgia, Athens 30602, GA, United States
| | - Rachel Wanty Larson
- Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman 99164-6520, WA, United States
| | - Piotr Podlasz
- Department of Pathophysiology, Forensic Veterinary and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, 10-719 Olsztyn, Poland
| | - Krzysztof Czaja
- Department of Veterinary Biosciences and Diagnostic Imaging, The University of Georgia, Athens 30602, GA, United States.
| |
Collapse
|
6
|
Abstract
OBJECTIVE Emerging preclinical evidence has shown that the bidirectional signaling between the gastrointestinal (GI) tract and the brain, the so-called gut-brain axis, plays an important role in both host metabolism and behavior. In this review, we discuss the potential mechanisms of the brain-gut axis in relation to the pathophysiology of metabolic syndrome. METHODS A selective literature review was conducted to evaluate GI and brain interactions. RESULTS Evidence suggests reduced microbial diversity in obesity and metabolic dysregulation. However, findings of microbiota composition in obese individuals are inconsistent, and the investigation of causality between gut microbiota and energy homeostasis is complex because multiple variables contribute to the gut microbiota composition. The microbial metabolites short chain fatty acids are found to exert numerous physiologic effects, including energy homeostasis through the regulation of GI hormones such as cholecystokinin, glucagon-like peptide 1, peptide tyrosine-tyrosine, and leptin. Preclinical studies show that modifying rodents' microbiota through fecal transplantation results in alterations of these GI hormones and subsequently an altered metabolism and behavior. However, whether and to what extent preclinical findings translate to human metabolism is unclear. CONCLUSIONS One of the major limitations and challenges in this field of research is interindividual variability of the microbiome. Future research needs to combine recent insights gained into tracking the dynamics of the microbiome as well as the metabolic responses. Furthermore, advanced mapping of the human microbiome is required to investigate the metabolic implications of the gut-brain axis to develop targeted interventions for obesity and metabolic syndrome.
Collapse
|
7
|
Guillebaud F, Girardet C, Abysique A, Gaigé S, Barbouche R, Verneuil J, Jean A, Leprince J, Tonon MC, Dallaporta M, Lebrun B, Troadec JD. Glial Endozepines Inhibit Feeding-Related Autonomic Functions by Acting at the Brainstem Level. Front Neurosci 2017; 11:308. [PMID: 28611581 PMCID: PMC5447764 DOI: 10.3389/fnins.2017.00308] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/16/2017] [Indexed: 01/05/2023] Open
Abstract
Endozepines are endogenous ligands for the benzodiazepine receptors and also target a still unidentified GPCR. The endozepine octadecaneuropeptide (ODN), an endoproteolytic processing product of the diazepam-binding inhibitor (DBI) was recently shown to be involved in food intake control as an anorexigenic factor through ODN-GPCR signaling and mobilization of the melanocortinergic signaling pathway. Within the hypothalamus, the DBI gene is mainly expressed by non-neuronal cells such as ependymocytes, tanycytes, and protoplasmic astrocytes, at levels depending on the nutritional status. Administration of ODN C-terminal octapeptide (OP) in the arcuate nucleus strongly reduces food intake. Up to now, the relevance of extrahypothalamic targets for endozepine signaling-mediated anorexia has been largely ignored. We focused our study on the dorsal vagal complex located in the caudal brainstem. This structure is strongly involved in the homeostatic control of food intake and comprises structural similarities with the hypothalamus. In particular, a circumventricular organ, the area postrema (AP) and a tanycyte-like cells forming barrier between the AP and the adjacent nucleus tractus solitarius (NTS) are present. We show here that DBI is highly expressed by ependymocytes lining the fourth ventricle, tanycytes-like cells, as well as by proteoplasmic astrocytes located in the vicinity of AP/NTS interface. ODN staining observed at the electron microscopic level reveals that ODN-expressing tanycyte-like cells and protoplasmic astrocytes are sometimes found in close apposition to neuronal elements such as dendritic profiles or axon terminals. Intracerebroventricular injection of ODN or OP in the fourth ventricle triggers c-Fos activation in the dorsal vagal complex and strongly reduces food intake. We also show that, similarly to leptin, ODN inhibits the swallowing reflex when microinjected into the swallowing pattern generator located in the NTS. In conclusion, we hypothesized that ODN expressing cells located at the AP/NTS interface could release ODN and modify excitability of NTS neurocircuitries involved in food intake control.
Collapse
Affiliation(s)
- Florent Guillebaud
- Laboratoire Physiologie et Physiopathologie du Système Nerveux Somato-Moteur et Neurovégétatif EA 4674, Faculté des Sciences et Techniques de St Jérôme, Université Aix-MarseilleMarseille, France
| | - Clémence Girardet
- Laboratoire Physiologie et Physiopathologie du Système Nerveux Somato-Moteur et Neurovégétatif EA 4674, Faculté des Sciences et Techniques de St Jérôme, Université Aix-MarseilleMarseille, France
| | - Anne Abysique
- Laboratoire Physiologie et Physiopathologie du Système Nerveux Somato-Moteur et Neurovégétatif EA 4674, Faculté des Sciences et Techniques de St Jérôme, Université Aix-MarseilleMarseille, France
| | - Stéphanie Gaigé
- Laboratoire Physiologie et Physiopathologie du Système Nerveux Somato-Moteur et Neurovégétatif EA 4674, Faculté des Sciences et Techniques de St Jérôme, Université Aix-MarseilleMarseille, France
| | - Rym Barbouche
- Laboratoire Physiologie et Physiopathologie du Système Nerveux Somato-Moteur et Neurovégétatif EA 4674, Faculté des Sciences et Techniques de St Jérôme, Université Aix-MarseilleMarseille, France
| | - Jérémy Verneuil
- Laboratoire Physiologie et Physiopathologie du Système Nerveux Somato-Moteur et Neurovégétatif EA 4674, Faculté des Sciences et Techniques de St Jérôme, Université Aix-MarseilleMarseille, France
| | - André Jean
- Laboratoire Physiologie et Physiopathologie du Système Nerveux Somato-Moteur et Neurovégétatif EA 4674, Faculté des Sciences et Techniques de St Jérôme, Université Aix-MarseilleMarseille, France
| | - Jérôme Leprince
- Institut National de la Santé et de la Recherche Médicale U1239, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Institute for Research and Innovation in Biomedicine, University of Rouen NormadieMont-Saint-Aignan, France
| | - Marie-Christine Tonon
- Institut National de la Santé et de la Recherche Médicale U1239, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Institute for Research and Innovation in Biomedicine, University of Rouen NormadieMont-Saint-Aignan, France
| | - Michel Dallaporta
- Laboratoire Physiologie et Physiopathologie du Système Nerveux Somato-Moteur et Neurovégétatif EA 4674, Faculté des Sciences et Techniques de St Jérôme, Université Aix-MarseilleMarseille, France
| | - Bruno Lebrun
- Laboratoire Physiologie et Physiopathologie du Système Nerveux Somato-Moteur et Neurovégétatif EA 4674, Faculté des Sciences et Techniques de St Jérôme, Université Aix-MarseilleMarseille, France
| | - Jean-Denis Troadec
- Laboratoire Physiologie et Physiopathologie du Système Nerveux Somato-Moteur et Neurovégétatif EA 4674, Faculté des Sciences et Techniques de St Jérôme, Université Aix-MarseilleMarseille, France
| |
Collapse
|
8
|
Duodenal GLP-1 signaling regulates hepatic glucose production through a PKC-δ-dependent neurocircuitry. Cell Death Dis 2017; 8:e2609. [PMID: 28182013 PMCID: PMC5386475 DOI: 10.1038/cddis.2017.28] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 12/13/2016] [Accepted: 12/13/2016] [Indexed: 12/25/2022]
Abstract
Intestinal glucagon-like peptide-1 (GLP-1) is a hormone that stimulates insulin secretion and acts as a neuropeptide to control glucose homeostasis, but little is known whether intestinal GLP-1 has any effect in the control of hepatic glucose production (HGP). Here we found that intraduodenal infusion of GLP-1 activated duodenal PKC-δ, lowered HGP and was accompanied by a decrease in hepatic expression of gluconeogenic enzymes and an increase in hepatic insulin signaling in rats. However, gut co-infusion of either the GLP-1 receptor antagonist Ex-9, or the PKC-δ inhibitor rottlerin with GLP-1, negated the ability of gut GLP-1 to lower HGP and to increase hepatic insulin signaling during clamps. The metabolic and molecular signal effects of duodenal GLP-1 were also negated by co-infusion with tetracaine, pharmacologic inhibition of N-methyl-d-aspartate receptors within the dorsalvagal complex, or hepatic vagotomy in rats. In summary, we identified a neural glucoregulatory function of gut GLP-1 signaling.
Collapse
|
9
|
Abstract
Malnutrition is the result of an inadequate balance between energy intake and energy expenditure that ultimately leads to either obesity or undernutrition. Several factors are associated with the onset and preservation of malnutrition. One of these factors is the gut microbiota, which has been recognized as an important pathophysiologic factor in the development and sustainment of malnutrition. However, to our knowledge, the extent to which the microbiota influences malnutrition has yet to be elucidated. In this review, we summarize the mechanisms via which the gut microbiota may influence energy homeostasis in relation to malnutrition. In addition, we discuss potential therapeutic modalities to ameliorate obesity or undernutrition.
Collapse
Affiliation(s)
- Nicolien C de Clercq
- Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, Netherlands;
| | - Albert K Groen
- Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, Netherlands
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Johannes A Romijn
- Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, Netherlands
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, Netherlands; and
- Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
10
|
Campos CA, Ritter RC. NMDA-type glutamate receptors participate in reduction of food intake following hindbrain melanocortin receptor activation. Am J Physiol Regul Integr Comp Physiol 2015; 308:R1-9. [PMID: 25394828 PMCID: PMC4281681 DOI: 10.1152/ajpregu.00388.2014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 11/07/2014] [Indexed: 02/07/2023]
Abstract
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.
Collapse
Affiliation(s)
- Carlos A Campos
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, Washington
| | - Robert C Ritter
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, Washington
| |
Collapse
|
11
|
Zhao H, Peters JH, Zhu M, Page SJ, Ritter RC, Appleyard SM. Frequency-dependent facilitation of synaptic throughput via postsynaptic NMDA receptors in the nucleus of the solitary tract. J Physiol 2014; 593:111-25. [PMID: 25281729 DOI: 10.1113/jphysiol.2013.258103] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 09/15/2014] [Indexed: 12/28/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Huan Zhao
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, 99164, USA
| | | | | | | | | | | |
Collapse
|
12
|
de Lartigue G. Putative roles of neuropeptides in vagal afferent signaling. Physiol Behav 2014; 136:155-69. [PMID: 24650553 DOI: 10.1016/j.physbeh.2014.03.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/23/2014] [Accepted: 03/10/2014] [Indexed: 02/06/2023]
Abstract
The vagus nerve is a major pathway by which information is communicated between the brain and peripheral organs. Sensory neurons of the vagus are located in the nodose ganglia. These vagal afferent neurons innervate the heart, the lung and the gastrointestinal tract, and convey information about peripheral signals to the brain important in the control of cardiovascular tone, respiratory tone, and satiation, respectively. Glutamate is thought to be the primary neurotransmitter involved in conveying all of this information to the brain. It remains unclear how a single neurotransmitter can regulate such an extensive list of physiological functions from a wide range of visceral sites. Many neurotransmitters have been identified in vagal afferent neurons and have been suggested to modulate the physiological functions of glutamate. Specifically, the anorectic peptide transmitters, cocaine and amphetamine regulated transcript (CART) and the orexigenic peptide transmitters, melanin concentrating hormone (MCH) are differentially regulated in vagal afferent neurons and have opposing effects on food intake. Using these two peptides as a model, this review will discuss the potential role of peptide transmitters in providing a more precise and refined modulatory control of the broad physiological functions of glutamate, especially in relation to the control of feeding.
Collapse
Affiliation(s)
- Guillaume de Lartigue
- Dept Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, Davis, CA 95616, USA.
| |
Collapse
|
13
|
Schwartz GJ, Zeltser LM. Functional organization of neuronal and humoral signals regulating feeding behavior. Annu Rev Nutr 2013; 33:1-21. [PMID: 23642202 DOI: 10.1146/annurev-nutr-071812-161125] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Energy homeostasis--ensuring that energy availability matches energy requirements--is essential for survival. One way that energy balance is achieved is through coordinated action of neural and neuroendocrine feeding circuits, which promote energy intake when energy supply is limited. Feeding behavior engages multiple somatic and visceral tissues distributed throughout the body--contraction of skeletal and smooth muscles in the head and along the upper digestive tract required to consume and digest food, as well as stimulation of endocrine and exocrine secretions from a wide range of organs. Accordingly, neurons that contribute to feeding behaviors are localized to central, peripheral, and enteric nervous systems. To promote energy balance, feeding circuits must be able to identify and respond to energy requirements, as well as the amount of energy available from internal and external sources, and then direct appropriate coordinated responses throughout the body.
Collapse
Affiliation(s)
- Gary J Schwartz
- Department of Medicine, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY 10461, USA
| | | |
Collapse
|
14
|
Blouet C, Schwartz GJ. Duodenal lipid sensing activates vagal afferents to regulate non-shivering brown fat thermogenesis in rats. PLoS One 2012; 7:e51898. [PMID: 23251649 PMCID: PMC3522613 DOI: 10.1371/journal.pone.0051898] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 11/08/2012] [Indexed: 11/18/2022] Open
Abstract
Previous evidence indicates that duodenal lipid sensing engages gut-brain neurocircuits to determine food intake and hepatic glucose production, but a potential role for gut-brain communication in the control of energy expenditure remains to be determined. Here, we tested the hypothesis that duodenal lipid sensing activates a gut-brain-brown adipose tissue neuraxis to regulate thermogenesis. We demonstrate that direct administration of lipids into the duodenum increases brown fat temperature. Co-infusion of the local anesthetic tetracaine with duodenal lipids abolished the lipid-induced increase in brown fat temperature. Systemic administration of the CCKA receptor antagonist devazepide blocked the ability of duodenal lipids to increase brown fat thermogenesis. Parenchymal administration of the N-methyl-d-aspartate receptor blocker MK-801 directly into the caudomedial nucleus of the solitary tract also abolished duodenal lipid-induced activation of brown fat thermogenesis. These findings establish that duodenal lipid sensing activates a gut-brain-brown fat axis to determine brown fat temperature, and thereby reveal a previously unappreciated pathway that regulates thermogenesis.
Collapse
Affiliation(s)
- Clémence Blouet
- Departments of Medicine and Neuroscience, Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Gary J. Schwartz
- Departments of Medicine and Neuroscience, Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
15
|
Singru PS, Wittmann G, Farkas E, Zséli G, Fekete C, Lechan RM. Refeeding-activated glutamatergic neurons in the hypothalamic paraventricular nucleus (PVN) mediate effects of melanocortin signaling in the nucleus tractus solitarius (NTS). Endocrinology 2012; 153:3804-14. [PMID: 22700769 PMCID: PMC3404351 DOI: 10.1210/en.2012-1235] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
We previously demonstrated that refeeding after a prolonged fast activates a subset of neurons in the ventral parvocellular subdivision of the paraventricular nucleus (PVNv) as a result of increased melanocortin signaling. To determine whether these neurons contribute to satiety by projecting to the nucleus tractus solitarius (NTS), the retrogradely transported marker substance, cholera toxin-β (CTB), was injected into the dorsal vagal complex of rats that were subsequently fasted and refed for 2 h. By double-labeling immunohistochemistry, CTB accumulation was found in the cytoplasm of the majority of refeeding-activated c-Fos neurons in the ventral parvocellular subdivision of the hypothalamic paraventricular nucleus (PVNv). In addition, a large number of refeeding-activated c-Fos-expressing neurons were observed in the lateral parvocellular subdivision (PVNl) that also contained CTB and were innervated by axon terminals of proopiomelanocortin neurons. To visualize the location of neuronal activation within the NTS by melanocortin-activated PVN neurons, α-MSH was focally injected into the PVN, resulting in an increased number of c-Fos-containing neurons in the PVN and in the NTS, primarily in the medial and commissural parts. All refeeding-activated neurons in the PVNv and PVNl expressed the mRNA of the glutamatergic marker, type 2 vesicular glutamate transporter (VGLUT2), indicating their glutamatergic phenotype, but only rare neurons contained oxytocin. These data suggest that melanocortin-activated neurons in the PVNv and PVNl may contribute to refeeding-induced satiety through effects on the NTS and may alter the sensitivity of NTS neurons to vagal satiety inputs via glutamate excitation.
Collapse
Affiliation(s)
- Praful S Singru
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Tufts Medical Center, no. 268, 800 Washington Street, Boston, Massachusetts 02111, USA
| | | | | | | | | | | |
Collapse
|
16
|
Abstract
Peripheral and central glucose sensing play a major role in the regulation of food intake. Peripheral sensing occurs at duodenal and portal levels, although the importance of these sensing sites is still controversial. The present study aimed to compare the respective influence of these sensing pathways on the eating patterns; plasma concentrations of glucose, insulin and glucagon-like peptide-1 (GLP-1); and brain activity in juvenile pigs. In Experiment 1, we characterised the changes in the microstructure as a result of a 30-min meal in eight conscious animals after duodenal or portal glucose infusion in comparison with saline infusion. In Experiment 2, glucose, insulin and GLP-1 plasma concentrations were measured during 2 h after duodenal or portal glucose infusions in four anaesthetised animals. In Experiment 3, single photon emission computed tomography brain imaging was performed in five anaesthetised animals receiving duodenal or portal glucose or saline infusions. Both duodenal and portal glucose decreased the amount of food consumed, as well as the ingestion speed, although this effect appeared earlier with the portal infusion. Significant differences of glucose and GLP-1 plasma concentrations between treatments were found at the moment of brain imaging. Both duodenal and portal glucose infusions activated the dorsolateral prefrontal cortex and primary somatosensory cortex. Only duodenal glucose infusion was able to induce activation of the prepyriform area, orbitofrontal cortex, caudate and putamen, as well as deactivation of the anterior prefrontal cortex and anterior entorhinal cortex, whereas only portal glucose infusion induced a significant activation of the insular cortex. We demonstrated that duodenal and portal glucose infusions led to the modulation of brain areas that are known to regulate eating behaviour, which probably explains the decrease of food intake after both stimulations. These stimulation pathways induced specific systemic and central responses, suggesting that different brain processing matrices are involved.
Collapse
Affiliation(s)
- J Boubaker
- Institut National de la Recherche Agronomique, ADNC, Saint-Gilles, France
| | | | | | | |
Collapse
|
17
|
Holmes GM. Upper gastrointestinal dysmotility after spinal cord injury: is diminished vagal sensory processing one culprit? Front Physiol 2012; 3:277. [PMID: 22934031 PMCID: PMC3429051 DOI: 10.3389/fphys.2012.00277] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 06/27/2012] [Indexed: 12/12/2022] Open
Abstract
Despite the widely recognized prevalence of gastric, colonic, and anorectal dysfunction after spinal cord injury (SCI), significant knowledge gaps persist regarding the mechanisms leading to post-SCI gastrointestinal (GI) impairments. Briefly, the regulation of GI function is governed by a mix of parasympathetic, sympathetic, and enteric neurocircuitry. Unlike the intestines, the stomach is dominated by parasympathetic (vagal) control whereby gastric sensory information is transmitted via the afferent vagus nerve to neurons of the nucleus tractus solitarius (NTS). The NTS integrates this sensory information with signals from throughout the central nervous system. Glutamatergic and GABAergic NTS neurons project to other nuclei, including the preganglionic parasympathetic neurons of the dorsal motor nucleus of the vagus (DMV). Finally, axons from the DMV project to gastric myenteric neurons, again, through the efferent vagus nerve. SCI interrupts descending input to the lumbosacral spinal cord neurons that modulate colonic motility and evacuation reflexes. In contrast, vagal neurocircuitry remains anatomically intact after injury. This review presents evidence that unlike the post-SCI loss of supraspinal control which leads to colonic and anorectal dysfunction, gastric dysmotility occurs as an indirect or secondary pathology following SCI. Specifically, emerging data points toward diminished sensitivity of vagal afferents to GI neuroactive peptides, neurotransmitters and, possibly, macronutrients. The neurophysiological properties of rat vagal afferent neurons are highly plastic and can be altered by injury or energy balance. A reduction of vagal afferent signaling to NTS neurons may ultimately bias NTS output toward unregulated GABAergic transmission onto gastric-projecting DMV neurons. The resulting gastroinhibitory signal may be one mechanism leading to upper GI dysmotility following SCI.
Collapse
Affiliation(s)
- Gregory M. Holmes
- Neural and Behavioral Sciences, Penn State University College of MedicineHershey, PA, USA
| |
Collapse
|
18
|
Campos CA, Wright JS, Czaja K, Ritter RC. CCK-induced reduction of food intake and hindbrain MAPK signaling are mediated by NMDA receptor activation. Endocrinology 2012; 153:2633-46. [PMID: 22508518 PMCID: PMC3359610 DOI: 10.1210/en.2012-1025] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 03/23/2012] [Indexed: 01/28/2023]
Abstract
The dorsal vagal complex of the hindbrain, including the nucleus of the solitary tract (NTS), receives neural and humoral afferents that contribute to the process of satiation. The gut peptide, cholecystokinin (CCK), promotes satiation by activating gastrointestinal vagal afferents that synapse in the NTS. Previously, we demonstrated that hindbrain administration of N-methyl-D-aspartate (NMDA)-type glutamate receptor antagonists attenuate reduction of food intake after ip CCK-8 injection, indicating that these receptors play a necessary role in control of food intake by CCK. However, the signaling pathways through which hindbrain NMDA receptors contribute to CCK-induced reduction of food intake have not been investigated. Here we report CCK increases phospho-ERK1/2 in NTS neurons and in identified vagal afferent endings in the NTS. CCK-evoked phospho-ERK1/2 in the NTS was attenuated in rats pretreated with capsaicin and was abolished by systemic injection of a CCK1 receptor antagonist, indicating that phosphorylation of ERK1/2 occurs in and is mediated by gastrointestinal vagal afferents. Fourth ventricle injection of a competitive NMDA receptor antagonist, prevented CCK-induced phosphorylation of ERK1/2 in hindbrain neurons and in vagal afferent endings, as did direct inhibition of MAPK kinase. Finally, fourth ventricle administration of either a MAPK kinase inhibitor or NMDA receptor antagonist prevented the reduction of food intake by CCK. We conclude that activation of NMDA receptors in the hindbrain is necessary for CCK-induced ERK1/2 phosphorylation in the NTS and consequent reduction of food intake.
Collapse
Affiliation(s)
- Carlos A Campos
- Department of Veterinary and Comparative Anatomy, Pharmacology, and Physiology and Programs in Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, Washington 99164-6520, USA
| | | | | | | |
Collapse
|
19
|
Niemann H, Kuhla B, Flachowsky G. Perspectives for feed-efficient animal production1. J Anim Sci 2011; 89:4344-63. [DOI: 10.2527/jas.2011-4235] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
20
|
Dean RL, Eyerman D, Todtenkopf MS, Turncliff RZ, Bidlack JM, Deaver DR. Effects of oral loperamide on efficacy of naltrexone, baclofen and AM-251 in blocking ethanol self-administration in rats. Pharmacol Biochem Behav 2011; 100:530-7. [PMID: 22056608 DOI: 10.1016/j.pbb.2011.10.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 10/17/2011] [Indexed: 11/19/2022]
Abstract
Naltrexone is a μ-opioid receptor antagonist that has been extensively studied for its ability to block the rewarding effects of ethanol. Opioid receptors are widely distributed within the gastrointestinal tract (GIT). Typically, naltrexone is administered by parenteral routes in nonclinical studies. We initially tested if opioid receptors within the GIT would influence the ability of oral naltrexone to inhibit ethanol oral self-administration in rats using the co-administration of oral loperamide, a peripherally restricted opioid agonist. As expected, oral naltrexone only had modest effects on ethanol intake, and the response was not dose-dependent. However in rats, treatment with loperamide prior to the administration of naltrexone resulted in a suppression of ethanol intake which approached that observed with naltrexone given by the subcutaneous (SC) route. Importantly, administration of loperamide prior to administration of naltrexone did not alter blood concentrations of naltrexone. We then evaluated if oral loperamide would enhance effects of baclofen (a GABA(B) receptor agonist) and AM-251 (a CB-1 receptor antagonist) and found that pre-treatment with loperamide did potentiate the action of both drugs to reduce ethanol self-administration. Finally, the specific opioid receptor type involved was investigated using selective μ- and κ-receptor antagonists to determine if these would affect the ability of the AM-251 and loperamide combination to block ethanol drinking behavior. The effect of loperamide was blocked by ALKS 37, a peripherally restricted μ-receptor antagonist. These data suggest an important role for opioid receptors within the GIT in modulating central reward pathways and may provide new insights into strategies for treating reward disorders, including drug dependency.
Collapse
MESH Headings
- Administration, Oral
- Alcohol Deterrents/administration & dosage
- Alcohol Deterrents/blood
- Alcohol Deterrents/pharmacokinetics
- Alcohol Deterrents/therapeutic use
- Alcohol Drinking/prevention & control
- Animals
- Animals, Outbred Strains
- Baclofen/administration & dosage
- Baclofen/therapeutic use
- Behavior, Animal/drug effects
- Drug Synergism
- Drug Therapy, Combination
- GABA-B Receptor Agonists/administration & dosage
- GABA-B Receptor Agonists/therapeutic use
- Loperamide/administration & dosage
- Loperamide/antagonists & inhibitors
- Loperamide/therapeutic use
- Male
- Naltrexone/administration & dosage
- Naltrexone/blood
- Naltrexone/pharmacokinetics
- Naltrexone/therapeutic use
- Narcotic Antagonists/blood
- Narcotic Antagonists/pharmacokinetics
- Narcotic Antagonists/pharmacology
- Narcotic Antagonists/therapeutic use
- Piperidines/administration & dosage
- Piperidines/therapeutic use
- Pyrazoles/administration & dosage
- Pyrazoles/therapeutic use
- Rats
- Rats, Wistar
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/antagonists & inhibitors
Collapse
Affiliation(s)
- Reginald L Dean
- Life Sciences and Toxicology, Alkermes, Inc., Waltham, MA 02451, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Transient receptor potential (TRP) A1 activated currents in TRPV1 and cholecystokinin-sensitive cranial visceral afferent neurons. Brain Res 2011; 1383:36-42. [DOI: 10.1016/j.brainres.2011.02.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 01/31/2011] [Accepted: 02/03/2011] [Indexed: 01/06/2023]
|
22
|
Min DK, Tuor UI, Chelikani PK. Gastric distention induced functional magnetic resonance signal changes in the rodent brain. Neuroscience 2011; 179:151-8. [PMID: 21284950 DOI: 10.1016/j.neuroscience.2011.01.051] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 01/14/2011] [Accepted: 01/23/2011] [Indexed: 12/29/2022]
Abstract
Investigating the localization of gastric sensation within the brain is important for understanding the neural correlates of satiety. Previous rodent studies have identified the brain-stem and hypothalamus as key mediators of gastric distention-induced satiation. Although, recent blood oxygen level-dependent functional magnetic resonance imaging (BOLD fMRI) studies in humans have identified a role for higher cortico-limbic structures in mediating the satiation effects of gastric distention, the role of these regions in rodents remains to be characterized. We determined the effects of gastric distention on global spatio-temporal BOLD fMRI signal changes in the rodent brain. Brain images were acquired with a high resolution 9.4 T magnet during gastric distention with continuous monitoring of blood pressure in adult male Sprague Dawley rats (n=8-10). Distention of the stomach with an intragastric balloon, at rates which mimicked the rate of consumption and emptying of a mixed nutrient liquid meal, resulted in robust reduction in food intake and increase in blood pressure. Gastric distention increased BOLD fMRI activity within homeostatic regions such as the hypothalamus and nucleus tractus solitarius, as well as non homeostatic regions including the hippocampus, amygdala, thalamus, cerebellum and the cortex (cingulate, insular, motor and sensory cortices). Further, the increase in BOLD fMRI activity following distention was strongly correlated to an increase in blood pressure. These results indicate that gastric distention, mimicking the rate of intake and emptying of a liquid meal, increases BOLD fMRI activity in both homeostatic and non homeostatic brain circuits which regulate food intake, and that these BOLD fMRI signal changes may in part be attributable to transient increases in blood pressure.
Collapse
Affiliation(s)
- D K Min
- Gastrointestinal Research Group, Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | | | | |
Collapse
|
23
|
Abstract
Low temperature and noxious chemicals activate transient receptor potential (TRP) A1 channel in spinal nociceptive neurons. TRPA1 ligands are found as common ingredients of pungent spices and can trigger autonomic reflexes and alter blood pressure. Recent discovery of TRPA1 on visceral afferent pathway suggests such nonnociceptive responses may be closely related to TRPA1. To understand potential contributions of TRPA1 on autonomic response, we have tested TRPA1 agonist allyl isothiocyanate (AITC) effects on mechanically dispersed nucleus tractus solitarii neurons. AITC of 200 microM increased glutamate release in 58% of tested neurons and TRP channel antagonist ruthenium red (50 microM) blocked the AITC actions. tetrodotoxin and Cd did not eliminate AITC-evoked glutamate release increase. TRPA1 expressed in capsaicin-sensitive and insensitive neurons.
Collapse
|
24
|
|
25
|
Guard DB, Swartz TD, Ritter RC, Burns GA, Covasa M. Blockade of hindbrain NMDA receptors containing NR2 subunits increases sucrose intake. Am J Physiol Regul Integr Comp Physiol 2009; 296:R921-8. [PMID: 19193935 DOI: 10.1152/ajpregu.90456.2008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have previously shown that blockade of N-methyl-d-aspartate (NMDA) receptors in the caudal brain stem delays satiation and increases food intake. NMDA receptors are heterodimers made up of distinct, but different, ion channel subunits. The NR2 subunits of the NMDA receptor contain the binding site for glutamate. About half of vagal afferents express immunoreactivity for NMDA NR2B subunit and about half of the NR2B expressing afferents also express NMDA NR2C or NR2D subunits. This suggests that increased food intake may be evoked by interference with glutamate binding to NMDA channels containing the NR2B subunit. To test this, we measured deprivation-induced intake of 15% sucrose solution following fourth ventricle and intra-nucleus of the solitary tract (intra-NTS) injections of Conantokin G (Con G; NR2B blocker), d-3-(2-carboxypiperazin-4-yl)-1-propenyl-1-phosphoric acid (d-CPPene; NR2B/2A blocker), and (+/-)-cis-1-(phenanthren-2yl-carbonyl)piperazine-2,3-dicarboxylic acid (PPDA; NR2D/C blocker). Fourth ventricular administration of Con G (5, 20, 40, 80 ng), d-CPPene (3.0, 6.25, 12.5, 25, 50, 100 ng), and PPDA (300, 400 ng) increased sucrose intake significantly compared with control. Likewise, injections of Con G (10 ng), d-CPPene (5 ng, 10 ng), and PPDA (0.5, 1.0, 2.5, 5.0 ng) directly into the NTS significantly increased sucrose intake. These results show that hindbrain injection of competitive NMDA antagonists with selectivity or preference for the NMDA receptor NR2B or NR2C subunits increases food intake.
Collapse
Affiliation(s)
- Douglas B Guard
- Department of Nutritional Sciences, College of Health and Human Development, The Pennsylvania State University, Univ. Park, PA 16802, USA
| | | | | | | | | |
Collapse
|
26
|
Babic T, Townsend RL, Patterson LM, Sutton GM, Zheng H, Berthoud HR. Phenotype of neurons in the nucleus of the solitary tract that express CCK-induced activation of the ERK signaling pathway. Am J Physiol Regul Integr Comp Physiol 2009; 296:R845-54. [PMID: 19176891 DOI: 10.1152/ajpregu.90531.2008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The satiating potency of CCK has been well characterized, including its mediation by capsaicin-sensitive vagal primary afferents. We have previously shown that peripherally administered CCK activates the MAPK-signaling cascade in a population of nucleus of the solitary tract (NTS) neurons and that preventing ERK1/2 phosphorylation partly attenuates CCK's satiating potency. The aim of this study was to identify the neurochemical phenotypes of the NTS neurons that exhibit CCK-induced activation of ERK1/2. Using confocal microscopy, we demonstrate that intraperitoneal CCK administration increases the number of neurons that express phosphorylated ERK1/2 (pERK1/2) in the medial and commissural subnuclei of the NTS and that CCK-induced expression of ERK1/2 is increased in tyrosine hydroxylase-immunoreactive neurons. Using Western blot analysis, we show that the robust increase in tyrosine hydroxylase phosphorylation obtained with intraperitoneal CCK is significantly attenuated in rats pretreated with the ERK-pathway blocker U0126 injected into the 4th ventricle. In addition, CCK injections increased pERK1/2 expression in POMC neurons in the NTS. In contrast, only the rare GAD67, neuronal nitric oxide synthase, and leptin-responsive neuron exhibited CCK-induced pERK immunoreactivity. We conclude that activation of POMC-immunoreactive neurons and tyrosine hydroxylase activity via the ERK-signaling pathway in the NTS likely contributes to CCK's satiating effects.
Collapse
Affiliation(s)
- Tanja Babic
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | | | | | | | | | | |
Collapse
|
27
|
Woods SC, D'Alessio DA. Central control of body weight and appetite. J Clin Endocrinol Metab 2008; 93:S37-50. [PMID: 18987269 PMCID: PMC2585760 DOI: 10.1210/jc.2008-1630] [Citation(s) in RCA: 302] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Accepted: 09/08/2008] [Indexed: 12/14/2022]
Abstract
CONTEXT Energy balance is critical for survival and health, and control of food intake is an integral part of this process. This report reviews hormonal signals that influence food intake and their clinical applications. EVIDENCE ACQUISITION A relatively novel insight is that satiation signals that control meal size and adiposity signals that signify the amount of body fat are distinct and interact in the hypothalamus and elsewhere to control energy homeostasis. This review focuses upon recent literature addressing the integration of satiation and adiposity signals and therapeutic implications for treatment of obesity. EVIDENCE SYNTHESIS During meals, signals such as cholecystokinin arise primarily from the GI tract to cause satiation and meal termination; signals secreted in proportion to body fat such as insulin and leptin interact with satiation signals and provide effective regulation by dictating meal size to amounts that are appropriate for body fatness, or stored energy. Although satiation and adiposity signals are myriad and redundant and reduce food intake, there are few known orexigenic signals; thus, initiation of meals is not subject to the degree of homeostatic regulation that cessation of eating is. There are now drugs available that act through receptors for satiation factors and which cause weight loss, demonstrating that this system is amenable to manipulation for therapeutic goals. CONCLUSIONS Although progress on effective medical therapies for obesity has been relatively slow in coming, advances in understanding the central regulation of food intake may ultimately be turned into useful treatment options.
Collapse
Affiliation(s)
- Stephen C Woods
- Department of Psychiatry, University of Cincinnati, Cincinnati, OH 45237, USA.
| | | |
Collapse
|
28
|
Wan S, Browning KN, Coleman FH, Sutton G, Zheng H, Butler A, Berthoud HR, Travagli RA. Presynaptic melanocortin-4 receptors on vagal afferent fibers modulate the excitability of rat nucleus tractus solitarius neurons. J Neurosci 2008; 28:4957-66. [PMID: 18463249 PMCID: PMC2681297 DOI: 10.1523/jneurosci.5398-07.2008] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Revised: 03/10/2008] [Accepted: 03/31/2008] [Indexed: 11/21/2022] Open
Abstract
The nucleus tractus solitarius (NTS) integrates visceral sensory signals with information from the forebrain to control homeostatic functions, including food intake. Melanocortin 3/4 receptor (MC3/4R) ligands administered directly to the caudal brainstem powerfully modulate meal size but not frequency, suggesting the enhancement of visceral satiety signals. Using whole-cell recordings from rat brainstem slices, we examined the effects of melanocortin ligands, alpha-melanocyte-stimulating hormone (alphaMSH) and melanotan II (MTII), on EPSC in NTS neurons. Thirty-two percent of NTS neurons responded to perfusion with MTII or alphaMSH with either an increase (24%) or a decrease (8%) in the frequency, but not amplitude, of spontaneous EPSCs; the effects of MTII were abolished by pretreatment with SHU9119. After surgical vagal deafferentation, only four of 34 (9%) NTS neurons responded to MTII with an increase in EPSC frequency. When EPSCs were evoked by electrical stimulation of the tractus solitarius in Krebs' solution with 2.4 mm Ca(2+)(e), alphaMSH and MTII increased the amplitude in six of the 28 neurons tested, decreased amplitude in 14 with no effect in the remaining eight neurons. In four of six neurons unresponsive to MTII, decreasing Ca(2+)(e) levels to 1.5 mM uncovered an excitatory effect of MTII on EPSC amplitude. Reverse transcription-PCR analysis revealed the presence of MC4R, but not MC3R, in nodose ganglia. These results show that MC4R signaling leads mainly to presynaptic modulation of glutamatergic synaptic transmission and suggest that melanocortinergic-induced decrease of food intake may occur via enhancement of vagal afferent satiation signals from the gastrointestinal tract.
Collapse
Affiliation(s)
- Shuxia Wan
- Key Laboratory of Allergy and Immune-Related Diseases, Department of Physiology, School of Basic Medical Science, Wuhan University, Wuhan 430071, Hubei, China
| | - Kirsteen N. Browning
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808, and
| | - F. Holly Coleman
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808, and
| | - Gregory Sutton
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808, and
| | - Hiyuan Zheng
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808, and
| | - Andrew Butler
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808, and
| | - Hans-Rudolf Berthoud
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808, and
| | - R. Alberto Travagli
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808, and
| |
Collapse
|
29
|
Upper intestinal lipids trigger a gut-brain-liver axis to regulate glucose production. Nature 2008; 452:1012-6. [PMID: 18401341 DOI: 10.1038/nature06852] [Citation(s) in RCA: 215] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Accepted: 02/22/2008] [Indexed: 12/21/2022]
Abstract
Energy and glucose homeostasis are regulated by food intake and liver glucose production, respectively. The upper intestine has a critical role in nutrient digestion and absorption. However, studies indicate that upper intestinal lipids inhibit food intake as well in rodents and humans by the activation of an intestine-brain axis. In parallel, a brain-liver axis has recently been proposed to detect blood lipids to inhibit glucose production in rodents. Thus, we tested the hypothesis that upper intestinal lipids activate an intestine-brain-liver neural axis to regulate glucose homeostasis. Here we demonstrate that direct administration of lipids into the upper intestine increased upper intestinal long-chain fatty acyl-coenzyme A (LCFA-CoA) levels and suppressed glucose production. Co-infusion of the acyl-CoA synthase inhibitor triacsin C or the anaesthetic tetracaine with duodenal lipids abolished the inhibition of glucose production, indicating that upper intestinal LCFA-CoAs regulate glucose production in the preabsorptive state. Subdiaphragmatic vagotomy or gut vagal deafferentation interrupts the neural connection between the gut and the brain, and blocks the ability of upper intestinal lipids to inhibit glucose production. Direct administration of the N-methyl-d-aspartate ion channel blocker MK-801 into the fourth ventricle or the nucleus of the solitary tract where gut sensory fibres terminate abolished the upper-intestinal-lipid-induced inhibition of glucose production. Finally, hepatic vagotomy negated the inhibitory effects of upper intestinal lipids on glucose production. These findings indicate that upper intestinal lipids activate an intestine-brain-liver neural axis to inhibit glucose production, and thereby reveal a previously unappreciated pathway that regulates glucose homeostasis.
Collapse
|
30
|
Thammacharoen S, Lutz TA, Geary N, Asarian L. Hindbrain administration of estradiol inhibits feeding and activates estrogen receptor-alpha-expressing cells in the nucleus tractus solitarius of ovariectomized rats. Endocrinology 2008; 149:1609-17. [PMID: 18096668 PMCID: PMC2276711 DOI: 10.1210/en.2007-0340] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
17beta-estradiol (E2), acting via estrogen receptor (ER)-alpha, inhibits feeding in animals. One mechanism apparently involves an increase in the satiating potency of cholecystokinin (CCK) released from the small intestine by ingested food. For example, the satiating potency of intraduodenal lipid infusions is increased by E2 in ovariectomized rats; this increased satiation is dependent on CCK, and it is accompanied by increases in the numbers of ERalpha-positive cells that express c-Fos in a subregion of the caudal nucleus tractus solitarius (cNTS) that receives abdominal vagal afferent projections. To test whether direct administration of E2 to this area of the hindbrain is sufficient to inhibit food intake, we first implanted 0.2 microg estradiol benzoate (EB) in cholesterol or cholesterol alone either sc or onto the surface of the hindbrain over the cNTS. Food intake was significantly reduced after hindbrain EB implants but not after sc EB implants. Next we verified that equimolar hindbrain implants of E2 and EB had similar feeding-inhibitory effects and determined that only small amounts of E2 reached brain areas outside the dorsal caudal hindbrain after hindbrain implants of (3)H-labeled E2. Neither plasma estradiol concentration nor plasma inflammatory cytokine concentration was increased by either hindbrain or sc EB implants. Finally, hindbrain EB implants, but not sc implants, increased c-Fos in ERalpha-positive cells in the cNTS after ip injection of 4 microg/kg CCK-8. We conclude that E2, acting via ERalpha in cNTS neurons, including neurons stimulated by ip CCK, is sufficient to inhibit feeding.
Collapse
Affiliation(s)
- Sumpun Thammacharoen
- Institute of Animal Science, Physiology and Behaviour Group, ETH-Zürich, Schorenstrasse 16, 8603 Schwerzenbach, Switzerland
| | | | | | | |
Collapse
|
31
|
Ammori JB, Zhang W, Newman EA, Mulholland MW. Glutamate-induced calcium transients in rat neurons of the dorsal motor nucleus of the vagus. J Gastrointest Surg 2007; 11:1016-24. [PMID: 17549578 DOI: 10.1007/s11605-007-0176-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The dorsal motor nucleus of the vagus (DMNV) integrates peripheral and central signals and sends efferent output to the gastrointestinal system. Glutamate, the major excitatory neurotransmitter of the central nervous system, causes increases in intracellular calcium in DMNV neurons. The mechanisms by which glutamate activates calcium signaling in the DMNV were examined. DMNV neurons were isolated from neonatal rat brainstem using microdissection and enzymatic digestion. Exposure to glutamate caused intracellular Ca(2+) increments in greater than 80% of cells. Removal of extracellular Ca(2+) abolished intracellular Ca(2+) transients. Kynurenic acid, a nonspecific glutamate receptor antagonist, abolished intracellular Ca(2+) transients. Exposure to glutamate while blocking AMPA receptors with GYKI 52466 abolished the Ca(2+) response. Exposure to (S)AMPA, an AMPA receptor agonist, caused intracellular Ca(2+) increments in 97% of cells. Activation and antagonism of NMDA and kainate receptors produced no changes compared to control experiments. NiCl, a nonspecific Ca(2+) channel blocker, abolished intracellular Ca(2+) transients. Blocking T-type Ca(2+) channels with mibefradil abolished the Ca(2+) response in 76% of cells. Blockade of L-type and N-type Ca(2+) channels did not affect the Ca(2+) response. Glutamate mediates intracellular Ca(2+) currents in DMNV neurons via the AMPA receptor and T-type Ca(2+) channels, allowing influx of extracellular Ca(2+).
Collapse
Affiliation(s)
- John B Ammori
- Department of Surgery, The University of Michigan Medical Center, 2101 Taubman Center, 1500 East Medical Center Drive, Ann Arbor, Michigan 48109-0346, USA
| | | | | | | |
Collapse
|
32
|
Ohi Y, Ishii Y, Haji A, Noguchi S, Sasaoka T, Fujimori T, Nabeshima YI, Sasahara M, Hattori Y. Platelet-derived growth factor (PDGF)-BB inhibits AMPA receptor-mediated synaptic transmission via PDGF receptor-beta in murine nucleus tractus solitarius. Brain Res 2007; 1159:77-85. [PMID: 17573050 DOI: 10.1016/j.brainres.2007.05.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2007] [Revised: 05/14/2007] [Accepted: 05/14/2007] [Indexed: 11/26/2022]
Abstract
Although platelet-derived growth factor (PDGF)-BB activates PDGF receptor-beta (PDGFR-beta) and, in turn, inhibits the glutamate N-methyl-D-aspartate (NMDA) receptor function, whether PDGF-BB modulates the CNS function mediated by another glutamate receptors, alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptors, remains poorly understood. Here we now report the inhibitory effect of PDGF-BB on the AMPA receptor function in the nucleus tractus solitarius (NTS) by using slice patch-clamp techniques. Excitatory postsynaptic currents (EPSCs) were evoked by electrical stimulation of the tractus solitarius in mouse NTS second-order neurons. EPSCs were nearly completely eliminated by CNQX but not by MK-801, implying mediation through non-NMDA receptors. PDGF-BB significantly decreased the amplitude of EPSCs without affecting the mean decay time constant. This inhibitory effect was transient and reversible after removing PDGF-BB. Furthermore, PDGF-BB significantly reduced the amplitude of AMPA-induced currents in NTS neurons, which showed that PDGF-BB could suppress the AMPA receptor-mediated excitatory input via the postsynaptic mechanism. The inhibitory effect of PDGF-BB on EPSCs was not observed in mutant mice with conditional deletion of the PDGFR-beta gene in neurons. Together, these studies suggest that the PDGF-B/PDGFR-beta axis inhibits the AMPA receptor-mediated synaptic transmission that comprises the major part of the primary afferent to the NTS second-order neuron. The detected inhibitory action may be involved in the CNS regulation of the respiratory response.
Collapse
Affiliation(s)
- Yoshiaki Ohi
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Nunnink M, Davenport RA, Ortega B, Houpt TA. D-Cycloserine enhances conditioned taste aversion learning in rats. Pharmacol Biochem Behav 2007; 87:321-30. [PMID: 17561237 PMCID: PMC2756720 DOI: 10.1016/j.pbb.2007.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2005] [Revised: 05/08/2007] [Accepted: 05/08/2007] [Indexed: 10/23/2022]
Abstract
Conditioned taste aversion (CTA) is a form of associative learning in which the pairing of a taste with a toxin causes an animal to avoid the taste. NMDA receptor mediated neurotransmission has been implicated in CTA, but the role of the NMDA receptor glycine-binding site has not been examined. To examine the effects on CTA of the glycinergic NMDA receptor agonist D-cycloserine, rats received D-cycloserine (15 mg/kg, i.p.) or vehicle 15 min before 10-min access to 0.125% saccharin, followed by a low dose of LiCl (19 mg/kg, i.p.). CTA was measured with 24-h, 2-bottle preference tests between water and saccharin. Vehicle-treated rats formed a mild CTA that rapidly extinguished, while d-cycloserine-treated rats formed a stronger CTA that extinguished slowly. The effect of d-cycloserine was specific to the NMDA receptor glycine-binding site, because pretreatment with HA-966 (6 mg/kg), a partial glycinergic agonist, blocked enhancement by D-cycloserine. Three follow-up experiments suggest that the enhancement of CTA was not due to an aversive effect of D-cycloserine. First, saccharin paired with D-cycloserine (15 mg/kg) alone did not induce a CTA, although a higher dose (30 mg/kg) did significantly lower saccharin preference. Second, pretreatment with D-cycloserine did not increase the duration of "lying-on-belly" behavior induced by LiCl. Third, pretreatment with D-cycloserine did not increase c-Fos induction by either LiCl or vehicle injection in central visceral relays (the nucleus of the solitary tract, the parabrachial nucleus, the central nucleus of the amygdala, the supraoptic nucleus, and the paraventricular nucleus). These results confirm the participation of NMDA receptor, and specifically the glycine-binding site of NMDA receptor, in CTA learning.
Collapse
Affiliation(s)
- Melissa Nunnink
- Department of Biological Sciences, Program in Neuroscience, BRF 252 MC 4340, The Florida State University, Tallahassee, FL 32306, USA
| | | | | | | |
Collapse
|
34
|
Raab M, Neuhuber WL. Glutamatergic functions of primary afferent neurons with special emphasis on vagal afferents. INTERNATIONAL REVIEW OF CYTOLOGY 2007; 256:223-75. [PMID: 17241909 DOI: 10.1016/s0074-7696(07)56007-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Glutamate has been identified as the main transmitter of primary afferent neurons. This was established based on biochemical, electrophysiological, and immunohistochemical data from studies on glutamatergic receptors and their agonists/antagonists. The availability of specific antibodies directed against glutamate and, more recently, vesicular glutamate transporters corroborated this and led to significant new discoveries. In particular, peripheral endings of various classes of afferents contain vesicular glutamate transporters, suggesting vesicular storage in and exocytotic release of glutamate from peripheral afferent endings. This suggests that autocrine mechanisms regulate sensory transduction processes. However, glutamate release from peripheral sensory terminals could also enable afferent neurons to influence various cells associated with them. This may be particularly relevant for vagal intraganglionic laminar endings, which could represent glutamatergic sensor-effector components of intramural reflex arcs in the gastrointestinal tract. Thus, morphological analysis of the relationships of putative glutamatergic primary afferents with associated tissues may direct forthcoming studies on their functions.
Collapse
Affiliation(s)
- Marion Raab
- Institut für Anatomie, Universität Erlangen-Nürnberg, Erlangen, Germany
| | | |
Collapse
|
35
|
Schwartz GJ. Integrative capacity of the caudal brainstem in the control of food intake. Philos Trans R Soc Lond B Biol Sci 2007; 361:1275-80. [PMID: 16874932 PMCID: PMC1642699 DOI: 10.1098/rstb.2006.1862] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The caudal brainstem nucleus of the solitary tract (NTS) is the initial central nervous system (CNS) terminus for a variety of gastrointestinal mechanical, nutrient chemical and gut peptide signals that limit the amount of food consumed during a meal. It receives neuroanatomical projections from gut vagal and non-vagal visceral afferents that mediate the CNS representation of these meal-stimulated gut feedback signals, and is reciprocally connected to a range of hypothalamic and limbic system sites that play significant roles in the neural processing of meal-related stimuli and in determining food consumption. Neurons in the NTS also contains elements of leptinergic and melanocortinergic signalling systems, presenting the possibility that the brainstem actions of these neuropeptides affect both the NTS processing of meal-stimulated gut afferent neural activity and its behavioural potency. Taken together, these features suggest that the NTS is ideally situated to integrate central and peripheral signals that determine meal size. This manuscript will review recent support from molecular genetic, neurophysiological and immunocytochemical studies that begin to identify and characterize the types of integrative functions performed within the NTS, and highlight the extent to which they are consistent with a causal role for NTS integration of peripheral gut and central neuropeptide signals important in the control of food intake.
Collapse
Affiliation(s)
- Gary J Schwartz
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
36
|
Young RL, Page AJ, O'Donnell TA, Cooper NJ, Blackshaw LA. Peripheral versus central modulation of gastric vagal pathways by metabotropic glutamate receptor 5. Am J Physiol Gastrointest Liver Physiol 2007; 292:G501-11. [PMID: 17053158 DOI: 10.1152/ajpgi.00353.2006] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Metabotropic glutamate receptors (mGluR) are classified into group I, II, and III mGluR. Group I (mGluR1, mGluR5) are excitatory, whereas group II and III are inhibitory. mGluR5 antagonism potently reduces triggering of transient lower esophageal sphincter relaxations and gastroesophageal reflux. Transient lower esophageal sphincter relaxations are mediated via a vagal pathway and initiated by distension of the proximal stomach. Here, we determined the site of action of mGluR5 in gastric vagal pathways by investigating peripheral responses of ferret gastroesophageal vagal afferents to graded mechanical stimuli in vitro and central responses of nucleus tractus solitarius (NTS) neurons with gastric input in vivo in the presence or absence of the mGluR5 antagonist 2-methyl-6-(phenylethynyl)pyridine (MPEP). mGluR5 were also identified immunohistochemically in the nodose ganglia and NTS after extrinsic vagal inputs had been traced from the proximal stomach. Gastroesophageal vagal afferents were classified as mucosal, tension, or tension-mucosal (TM) receptors. MPEP (1-10 microM) inhibited responses to circumferential tension of tension and TM receptors. Responses to mucosal stroking of mucosal and TM receptors were unaffected. MPEP (0.001-10 nmol icv) had no major effect on the majority of NTS neurons excited by gastric distension or on NTS neurons inhibited by distension. mGluR5 labeling was abundant in gastric vagal afferent neurons and sparse in fibers within NTS vagal subnuclei. We conclude that mGluR5 play a prominent role at gastroesophageal vagal afferent endings but a minor role in central gastric vagal pathways. Peripheral mGluR5 may prove a suitable target for reducing mechanosensory input from the periphery, for therapeutic benefit.
Collapse
Affiliation(s)
- Richard L Young
- Nerve-Gut Research Laboratory, Level 1 Hanson Institute, Adelaide, SA, Australia.
| | | | | | | | | |
Collapse
|
37
|
Simon SA, de Araujo IE, Gutierrez R, Nicolelis MAL. The neural mechanisms of gustation: a distributed processing code. Nat Rev Neurosci 2007; 7:890-901. [PMID: 17053812 DOI: 10.1038/nrn2006] [Citation(s) in RCA: 186] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Whenever food is placed in the mouth, taste receptors are stimulated. Simultaneously, different types of sensory fibre that monitor several food attributes such as texture, temperature and odour are activated. Here, we evaluate taste and oral somatosensory peripheral transduction mechanisms as well as the multi-sensory integrative functions of the central pathways that support the complex sensations that we usually associate with gustation. On the basis of recent experimental data, we argue that these brain circuits make use of distributed ensemble codes that represent the sensory and post-ingestive properties of tastants.
Collapse
Affiliation(s)
- Sidney A Simon
- Department of Neurobiology, Duke University, Durham, North Carolina 27710, USA.
| | | | | | | |
Collapse
|
38
|
Berthoud HR, Sutton GM, Townsend RL, Patterson LM, Zheng H. Brainstem mechanisms integrating gut-derived satiety signals and descending forebrain information in the control of meal size. Physiol Behav 2006; 89:517-24. [PMID: 16996546 DOI: 10.1016/j.physbeh.2006.08.018] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2005] [Revised: 07/30/2006] [Accepted: 08/15/2006] [Indexed: 12/12/2022]
Abstract
Ingestive behavior is controlled by a complex interplay between signals conveying availability of (1) potentially ingestible food in the environment, (2) digestible food in the alimentary canal, (3) circulating fuels and (4) stored fuels. Each of these four classes of signals interact with specific sensors and neural circuits whose integrated output determines when food intake is initiated and when it is stopped. Because the final common path responsible for oromotor control is contained within complex neural pattern generators within the brainstem and is intimately linked to sensory information from the alimentary canal, at least part of the integration between the four classes of signals is thought to take place at the level of the caudal brainstem. Here we show that CCK, representing a class 2, or direct signal, and MC4-melanocortin receptor activity, representing a second order class 3/4, or indirect signal, converge in the nucleus of the solitary tract where they modulate activity of the mitogen-activated, extracellular-signal regulated kinases 1 and 2 (ERK) pathway to determine the level of satiation. Blockade of this signaling pathway attenuates suppression of deprivation-induced food intake by intraperitoneal CCK and fourth ventricular MTII injection. Additional findings suggest that specific ERK-phosphorylation sites on ion channels and enzymes involved in catecholamine synthesis of NTS neurons may be involved in ERK-mediated satiation and meal termination. Longer-term downstream effects of ERK activation might involve CREB-mediated gene transcription known to produce plasticity changes in neurocircuitry that could determine inter-meal intervals and the size of future meals.
Collapse
Affiliation(s)
- Hans-Rudolf Berthoud
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana 70808, USA.
| | | | | | | | | |
Collapse
|
39
|
Hayes MR, Covasa M. Gastric distension enhances CCK-induced Fos-like immunoreactivity in the dorsal hindbrain by activating 5-HT3 receptors. Brain Res 2006; 1088:120-30. [PMID: 16630589 DOI: 10.1016/j.brainres.2006.03.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2006] [Revised: 03/06/2006] [Accepted: 03/07/2006] [Indexed: 10/24/2022]
Abstract
The combination of gastric distension and cholecystokinin (CCK) enhances both suppression of food intake and induction of c-Fos-like immunoreactivity (Fos-LI) in the dorsal vagal complex (DVC). Previously, we have shown that serotonin type-3 (5-HT3) receptor mediation of suppression of food intake by CCK requires gastric participation. Therefore, we hypothesized that 5-HT3 receptors mediate CCK-induced Fos-LI in the dorsal hindbrain through a mechanism that involves gastric distension. To test this hypothesis, we counted Fos-LI in the DVC of ondansetron (1 mg/kg; 5-HT3 receptor antagonist) and vehicle-treated rats following gastric balloon distension (5 ml), CCK (1 microg/kg) administration, or CCK combined with gastric distension. Ondansetron administration attenuated DVC Fos-LI by CCK administration. Likewise, ondansetron attenuated Fos-LI by gastric distension in the DVC, specifically within the nucleus of the solitary tract (NTS) and area postrema (AP) nuclei. The most pronounced attenuation of distension-induced Fos-LI by ondansetron occurred in the NTS, particularly in the medial and intermedial NTS. When combined, CCK and gastric distension enhanced Fos-LI in the DVC greater than each treatment alone. Furthermore, ondansetron administration attenuated the overall DVC enhanced Fos-LI induced by CCK + gastric distension, in particular at the NTS and AP nuclei. We found that, within the mid-to-caudal regions of the NTS and AP, 5-HT3 receptors most significantly mediate neuronal activation by CCK + distension. In conjunction with previous behavioral data, these results show that gastric distension enhances CCK-induced neuronal activation in the DVC by activating 5-HT3 receptors.
Collapse
Affiliation(s)
- Matthew R Hayes
- Department of Nutritional Sciences, College of Health and Human Development, The Pennsylvania State University, 126 South Henderson, University Park, PA 16802-6504, USA.
| | | |
Collapse
|
40
|
Zheng H, Patterson LM, Morrison C, Banfield BW, Randall JA, Browning KN, Travagli RA, Berthoud HR. Melanin concentrating hormone innervation of caudal brainstem areas involved in gastrointestinal functions and energy balance. Neuroscience 2006; 135:611-25. [PMID: 16111819 DOI: 10.1016/j.neuroscience.2005.06.055] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2005] [Revised: 05/22/2005] [Accepted: 06/09/2005] [Indexed: 12/30/2022]
Abstract
Neural signaling by melanin-concentrating hormone and its receptor (SLC-1) has been implicated in the control of energy balance, but due to the wide distribution of melanin-concentrating hormone-containing fibers throughout the neuraxis, its critical sites of action for a particular effect have not been identified. The present study aimed to anatomically and functionally characterize melanin-concentrating hormone innervation of the rat caudal brainstem, as this brain area plays an important role in the neural control of ingestive behavior and autonomic outflow. Using retrograde tracing we demonstrate that a significant proportion (5-15%) of primarily perifornical and far-lateral hypothalamic melanin-concentrating hormone neurons projects to the dorsal vagal complex. In the caudal brainstem, melanin-concentrating hormone-ir axon profiles are distributed densely in most areas including the nucleus of the solitary tract, dorsal motor nucleus of the vagus, and sympathetic premotor areas in the ventral medulla. Close anatomical appositions can be demonstrated between melanin-concentrating hormone-ir axon profiles and tyrosine hydroxylase, GABA, GLP-1, NOS-expressing, and nucleus of the solitary tract neurons activated by gastric nutrient infusion. In medulla slice preparations, bath application of melanin-concentrating hormone inhibited in a concentration-dependent manner the amplitude of excitatory postsynaptic currents evoked by solitary tract stimulation via a pre-synaptic mechanism. Fourth ventricular administration of melanin-concentrating hormone (10 microg) in freely moving rats decreased core body temperature but did not change locomotor activity and food and water intake. We conclude that the rich hypothalamo-medullary melanin-concentrating hormone projections in the rat are mainly inhibitory to nucleus of the solitary tract neurons, but are not involved in the control of food intake. Projections to ventral medullary sites may play a role in the inhibitory effect of melanin-concentrating hormone on energy expenditure.
Collapse
Affiliation(s)
- H Zheng
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Timofeeva E, Baraboi ED, Richard D. Contribution of the vagus nerve and lamina terminalis to brain activation induced by refeeding. Eur J Neurosci 2005; 22:1489-501. [PMID: 16190902 DOI: 10.1111/j.1460-9568.2005.04330.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Following refeeding, c-fos expression is induced in a particular set of brain regions that include the nucleus of the solitary tract (NTS), parabrachial nucleus (PB), central amygdala (CeA), paraventricular hypothalamic nucleus (PVH), supraoptic nucleus (SON) and the circumventricular organs. Within the PVH, the expression is particularly intense in the magnocellular division of the nucleus and it is as yet not clear how this activation occurs. The respective contribution of the vagus afferents and lamina terminalis, which conveys signals entering the brain through the forebrain circumventricular organs, has been investigated in rats subjected to a unilateral cervical vagotomy (UCV) or a unilateral lesion of the fibres running within the lamina terminalis (ULT) and projecting to the neuroendocrine hypothalamus. UCV significantly decreased postprandial c-fos expression in the NTS, PB, CeA and parvocellular division of the PVH. In contrast, ULT impaired postprandial activation of the magnocellular neurons in the PVH and SON. The present study also characterized the types of neurons activated in the PVH and SON during refeeding. In the magnocellular regions, arginine-vasopressin (AVP) neurons were activated upon refeeding whereas there was no apparent induction of Fos expression in oxytocin cells. In the parvocellular PVH, postprandial Fos was induced only in 30% of the corticotrophin-releasing factor (CRF) and AVP neurons. The results of the present study suggest that the postprandial activation of the brain requires the integrity of both the vagal- and lamina terminalis-associated pathways.
Collapse
Affiliation(s)
- Elena Timofeeva
- Centre de recherche de l'Hôpital Laval et Centre de recherche sur le métabolisme énergétique de l'Université Laval, Québec, Canada
| | | | | |
Collapse
|
42
|
Sutton GM, Duos B, Patterson LM, Berthoud HR. Melanocortinergic modulation of cholecystokinin-induced suppression of feeding through extracellular signal-regulated kinase signaling in rat solitary nucleus. Endocrinology 2005; 146:3739-47. [PMID: 15961554 DOI: 10.1210/en.2005-0562] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Signals from the gut and hypothalamus converge in the caudal brainstem to control ingestive behavior. We have previously shown that phosphorylation of ERK1/2 in the solitary nucleus (NTS) is necessary for food intake suppression by exogenous cholecystokinin (CCK). Here we test whether this intracellular signaling cascade is also involved in the integration of melanocortin-receptor (MCR) mediated inputs to the caudal brainstem. Using fourth ventricular-cannulated rats and Western blotting of NTS tissue, we show that the MC4R agonist melanotan II (MTII) rapidly and dose-dependently increases phosphorylation of both ERK1/2 and cAMP response element-binding protein (CREB). Sequential administration of fourth ventricular MTII and peripheral CCK at doses that alone produced submaximal stimulation of pERK1/2 produced an additive increase. Prior fourth ventricular administration of the MC4R antagonist SHU9119 completely abolished the CCK-induced increases in pERK and pCREB and, in freely feeding rats, SHU9119 significantly increased meal size and satiety ratio. Prior administration of the MAPK kinase inhibitor U0126 abolished the capacity of MTII to suppress 2-h food intake and significantly decreased MTII-induced ERK phosphorylation in the NTS. Furthermore, pretreatment with the cAMP inhibitor, cAMP receptor protein-Rp isomer, significantly attenuated stimulation of pERK induced by either CCK or MTII. The results demonstrate that activation of the ERK pathway is necessary for peripheral CCK and central MTII to suppress food intake. The cAMP-->ERK-->CREB cascade may thus constitute a molecular integrator for converging satiety signals from the gut and adiposity signals from the hypothalamus in the control of meal size and food intake.
Collapse
Affiliation(s)
- Gregory M Sutton
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University, 6400 Perkins Road, Baton Rouge, Louisiana 70808, USA
| | | | | | | |
Collapse
|
43
|
Ao Y, Wu S, Go VLW, Toy N, Yang H. Maintaining euglycemia prevents insulin-induced Fos expression in brain autonomic regulatory circuits. Pancreas 2005; 31:142-7. [PMID: 16025001 DOI: 10.1097/01.mpa.0000172562.96168.59] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Insulin-induced hypoglycemia activates neurons in hypothalamic and brain medullary nuclei involved in central autonomic regulation. We investigated whether these central neuronal activations relates to a deficiency of glucose supply. METHODS Three groups of non-fasted, conscious rats received intravenous (iv) saline infusion (control), a hyperinsulinemic/hypoglycemic clamp, or a hyperinsulinemic/euglycemic clamp for 120 minutes and then the brains were collected for Fos immunohistochemistry. RESULTS The number of Fos positive cells significantly increased in the paraventricular nucleus of the hypothalamus (PVN, 191 +/- 63 versus 66 +/- 18), pontine locus coeruleus (LC, 53 +/- 19 versus 5 +/- 2), brain medullary dorsal motor nucleus of the vagus (DMV, 26 +/- 4 versus 1 +/- 0), and nucleus tractus solitarii (NTS, 38 +/- 3 versus 10 +/- 35) in rats with hyperinsulinemic/hypoglycemic clamp compared with the controls. Maintaining blood glucose levels within physiological range by hyperinsulinemic/euglycemic clamp prevented insulin infusion-induced Fos expression in the PVN, DMV, and NTS. The numbers of Fos positive cells in these nuclei were significantly lower (-87%, -75%, and -51%, respectively) than that in the hypoglycemic rats. CONCLUSION These results indicate that neuronal activation in hypothalamic and medullary autonomic regulatory nuclei induced by insulin administration is caused by hypoglycemia rather than a direct action of insulin. In addition, certain neurons in the medullary DMV and NTS respond to declines in glucose levels within physiological range.
Collapse
Affiliation(s)
- Yan Ao
- Department of Medicine, Division of Digestive Diseases and Brain Research Institute, UCLA, Los Angeles, CA 90073, USA
| | | | | | | | | |
Collapse
|
44
|
Abstract
Decades of research have demonstrated that anorexia nervosa (AN) may be associated with aberrant cognition, yet, its role in maintaining stringent dieting has received relatively little attention from mainstream researchers of eating disorders. The purpose of the present article is to highlight cognitive ('top-down') factors that are considered responsible for anticipatory anxiety of stoutness and frank fat-phobia (laparophobia). A cognitive model proposed departs from the formulation suggesting that phobia of over-eating is superimposed on avoidant tendencies ('environmental autonomy syndrome'), whereas excessive exercising becomes a natural coping strategy with laparophobia, an instrument of reward. AN ideation involves complex neuronal circuitries and multiple neurochemical components that may conceivably represent a mirror image of those underlying obesity. The emphasis on phobia and aberrant membrane excitability akin to channelopathies behoves the clinicians to be aware of potential uses of drugs acting at the gamma-aminobutyric acid and the N-methyl-D-aspartate/AMPA [2-amino-3-(3-hydroxy-5-methylisoxazol-4-yl) propionic acid] receptors sites as the adjuncts to conventional agents in managing AN.
Collapse
|
45
|
Sutton GM, Patterson LM, Berthoud HR. Extracellular signal-regulated kinase 1/2 signaling pathway in solitary nucleus mediates cholecystokinin-induced suppression of food intake in rats. J Neurosci 2005; 24:10240-7. [PMID: 15537896 PMCID: PMC6730197 DOI: 10.1523/jneurosci.2764-04.2004] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Increased food intake is a major factor in the development of obesity, and the control of meal size is a valid approach to reduce food intake in humans. Meal termination, or satiety, is thought to be organized within the caudal brainstem where direct signals from the food handling alimentary canal and long-term signals from the forebrain converge in the solitary nucleus. Cholecystokinin (CCK) released from the gut after ingestion of food has been strongly implicated in nucleus tractus solitarius (NTS)-mediated satiation, but the exact cellular and intracellular signaling events are not understood. Using Western blotting and immunohistochemistry with phosphospecific antibodies, we demonstrate here that peripheral administration of CCK in rats leads to rapid activation of the extracellular signal-regulated kinase (ERK) signaling cascade in NTS neurons and that blockade of ERK signaling with microinfusion of a selective mitogen-activated ERK kinase inhibitor into the fourth ventricle attenuates the capacity of CCK to suppress food intake. In addition, we show that CCK-induced activation of ERK results in phosphorylation of the voltage-dependent potassium channel Kv4.2 and the nuclear transcription factor CREB (cAMP response element-binding protein). The results demonstrate that ERK signaling is necessary for exogenous CCK to suppress food intake in deprived rats and suggest that this pathway may also be involved in natural satiation and the period of satiety between meals through coupling of ERK activation to both cytosolic and nuclear effector mechanisms that have the potential to confer acute and long-term changes in neuronal functioning.
Collapse
Affiliation(s)
- Gregory M Sutton
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana 70808, USA
| | | | | |
Collapse
|
46
|
Liao Z, Li ZS, Lu Y, Wang WZ. Glutamate receptors within the nucleus of solitary tract contribute to pancreatic secretion stimulated by intraduodenal hypertonic saline. Auton Neurosci 2005; 120:62-7. [PMID: 15897013 DOI: 10.1016/j.autneu.2005.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2005] [Revised: 04/13/2005] [Accepted: 04/14/2005] [Indexed: 02/05/2023]
Abstract
It is well known that central transmission of vago-vagal reflex within the nucleus of solitary tract (NST) plays an important role in the regulation of gastrointestinal functions. The present study was designed to assess the role of NST glutamate receptor mechanism in pancreatic secretion evoked by intraduodenal hypertonic saline (HS) in anesthetized rats. Intraduodenal infusion of HS significantly (P<0.01) stimulated pancreatic protein output (from 2.60+/-0.09 to 4.18+/-0.24 mg/15 min). Bilaterally microinjected L-glutamate (5 nmol) into the medial nucleus of solitary tract (mNST) produced a significant increase of pancreatic protein secretion (from 2.65+/-0.12 to 4.80+/-0.34 mg/15 min, P<0.01). Bilateral injection of glutamate receptor antagonist kynurenic acid (KYN, 5 nmol) into the mNST completely abolished the increase of pancreatic protein output stimulated by intraduodenal HS (from 4.28+/-0.21 to 2.83+/-0.19 mg/15 min). Either NMDA receptor antagonist dl-2-amino-5-phosphonopentanoic acid (AP5, 1.5 nmol) or AMPA/Kainate receptor antagonist 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX, 1.5 nmol) injected into the mNST markedly attenuated (P<0.05) the pancreatic protein secretion stimulated by intraduodenal HS. In conclusion, these findings showed that blockade of the NST glutamate receptors, including NMDA and AMPA/Kainate receptors antagonized pancreatic secretion evoked by intraduodenal osmolality factor, and suggested that glutamate receptor mechanism within the NST contributed to the central regulation of pancreatic secretion.
Collapse
Affiliation(s)
- Zhuan Liao
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, 174 Chang-Hai Road, Shanghai 200433, PR China.
| | | | | | | |
Collapse
|
47
|
Zheng H, Patterson LM, Phifer CB, Berthoud HR. Brain stem melanocortinergic modulation of meal size and identification of hypothalamic POMC projections. Am J Physiol Regul Integr Comp Physiol 2005; 289:R247-58. [PMID: 15746303 DOI: 10.1152/ajpregu.00869.2004] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Metabolic, cognitive, and environmental factors processed in the forebrain modulate food intake by changing the potency of direct controls of meal ingestion in the brain stem. Here, we behaviorally and anatomically test the role of the hypothalamic proopiomelanocortin (POMC) system in mediating some of these descending, indirect controls. Melanotan II (MTII), a stable melanocortin 4 receptor (MC4R) and melanocortin 3 receptor (MC3R) agonist injected into the fourth ventricle near the dorsal vagal complex, potently inhibited 14-h food intake by decreasing meal size but not meal frequency; SHU9119, an antagonist, increased food intake by selectively increasing meal size. Furthermore, MTII injected into the fourth ventricle increased and SHU9119 tended to decrease heart rate and body temperature measured telemetrically in freely moving rats. Numerous alpha-melanocyte-stimulating hormone-immunoreactive axons were in close anatomical apposition to nucleus tractus solitarius neurons showing c-Fos in response to gastric distension, expressing neurochemical phenotypes implicated in ingestive control, and projecting to brown adipose tissue. In retrograde tracing experiments, a small percentage of arcuate nucleus POMC neurons was found to project to the dorsal vagal complex. Thus melanocortin signaling in the brain stem is sufficient to alter food intake via changing the potency of satiety signals and to alter sympathetic outflow. Although the anatomical findings support the involvement of hypothalamomedullary POMC projections in mediating part of the descending, indirect signal, they do not rule out involvement of POMC neurons in the nucleus tractus solitarius in mediating part of the direct signal.
Collapse
Affiliation(s)
- Huiyuan Zheng
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University System, 6400 Perkins Rd., Baton Rouge, LA 70808, USA
| | | | | | | |
Collapse
|
48
|
Val-Laillet D, Meurisse M, Tillet Y, Nowak R. Differential c-Fos expression in the newborn lamb nucleus tractus solitarius and area postrema following ingestion of colostrum or saline. Brain Res 2004; 1028:203-12. [PMID: 15527745 DOI: 10.1016/j.brainres.2004.09.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2004] [Indexed: 11/23/2022]
Abstract
Visceral stimuli and the gut-brain axis play a crucial role in the control of ingestion even in the neonate. The aim of this study was to assess the neuronal activation in the nucleus tractus solitarius (NTS) and the area postrema (AP) following nutritional and non-nutritional stimulations. Lambs received a single gastric infusion of colostrum or saline at 5% birth weight or were sham infused. Infusion of either liquid led to c-Fos-like immunoreactivity (c-FLI) in the NTS and AP. Differences were observed along the sections of the NTS rostro-caudal axis according to the nature of the stimulation, suggesting a specificity of certain afferents and/or NTS areas for nutritional or non-nutritional signals. In the AP, the neuronal activation induced by colostrum was much higher than that induced by saline. A higher number of TH-immunoreactive cells were activated following colostrum infusion, suggesting a specific involvement of the catecholaminergic pathway in the treatment of meal-related stimuli. In spite of functional convergence, the two medullary structures observed responded differently according to the stimulation, indicating a complementary role in the integration of visceral signals.
Collapse
Affiliation(s)
- David Val-Laillet
- UMR 6175 CNRS-INRA-Université de Tours-Haras Nationaux, Equipe de Comportement, F-37380 Nouzilly, France
| | | | | | | |
Collapse
|
49
|
Glass MJ, Kruzich PJ, Kreek MJ, Pickel VM. Decreased plasma membrane targeting of NMDA-NR1 receptor subunit in dendrites of medial nucleus tractus solitarius neurons in rats self-administering morphine. Synapse 2004; 53:191-201. [PMID: 15266550 DOI: 10.1002/syn.20049] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Opioid abuse is associated with repeated administration and escalation of dose that can result in profound adaptations in homeostatic processes. Potential cellular mechanisms and neural sites mediating opiate-dependent adaptations may involve NMDA-dependent synaptic plasticity within brain areas participating in behaviors related to consumption of natural reinforcers, as well as affective-autonomic integration, notably the medial nucleus tractus solitarius (mNTS). NMDA-dependent synaptic plasticity may be mediated by changes in the intracellular and surface targeting of NMDA receptors, particularly in postsynaptic sites including spines or small distal dendrites. High-resolution immunogold electron microscopic immunocytochemistry combined with morphometry were used to measure changes in targeting of the NMDA-NR1 (NR1) receptor subunit between intracellular and plasmalemmal sites in dendrites of neurons of the intermediate mNTS of rats self-administering escalating doses of morphine (EMSA). In control and EMSA rats, the density of plasmalemmal and cytosolic gold particles was inversely related to profile size. Collapsed across all NR1-labeled dendrites, rats self-administering morphine had a lower number of plasmalemmal gold particles per unit surface area (7.1 +/- 0.8 vs. 14.4 +/- 1 per 100 microm), but had a higher number of intracellular gold particles per unit cross-sectional area (169 +/- 6.1 vs. 148 +/- 5.1 per 100 microm2) compared to saline self-administering rats. Morphometric analysis showed that the decrease in plasma membrane labeling of NR1 was most robust in small dendritic profiles (<1 microm), where there was a reciprocal increase in the density of intracellular particles. These results indicate that the plasmalemmal distribution of the essential NR1 subunits in distal sites may prominently contribute to NMDA receptor-dependent modulation of neural circuitry regulating homeostatic processes, and targeting of these proteins can be prominently affected by morphine self-administration.
Collapse
Affiliation(s)
- Michael J Glass
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, New York 10021, USA
| | | | | | | |
Collapse
|
50
|
Covasa M, Hung CY, Ritter RC, Burns GA. Intracerebroventricular administration of MK-801 increases food intake through mechanisms independent of gastric emptying. Am J Physiol Regul Integr Comp Physiol 2004; 287:R1462-7. [PMID: 15358605 DOI: 10.1152/ajpregu.00471.2004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Systemic or hindbrain administration of MK-801, a noncompetitive N-methyl-D-aspartate receptor antagonist, increases meal size. To examine whether MK-801 enhances intake by increasing gastric emptying, we administered MK-801 (2.0 microg/3.0 microl) into the fourth ventricle [intracerebroventricular (ICV)] and measured feeding and gastric emptying of 5-ml NaCl or 15% sucrose loads. In a parallel experiment, we examined food intake and gastric emptying following intraperitoneal (IP) injection of MK-801 (100 microg/kg). MK-801, either IP or ICV, increased 30-min sucrose intake compared with control (12.3 +/- 0.7 vs. 9.8 +/- 0.5 and 16.6 +/- 2.0 vs. 10.7 +/- 0.7 ml, for IP and ICV administration, respectively). Also, IP MK-801 increased 5-min gastric emptying of NaCl (4.13 +/- 0.1 ml emptied) and sucrose (3.11 +/- 0.1 ml emptied) compared with control (3.75 +/- 0.2 and 2.28 +/- 0.1 ml emptied for NaCl and sucrose loads, respectively). In contrast, ICV MK-801 did not alter NaCl emptying (3.82 +/- 0.1 ml emptied) compared with control (3.82 +/- 0.3 ml emptied) and actually reduced gastric emptying of sucrose (2.1 +/- 0.2 and 2.94 +/- 0.1 ml emptied, for MK and vehicle, respectively). These data confirm previous results that systemic as well as hindbrain injection of MK-801 increases food intake. However, because ICV MK-801 failed to increase gastric emptying, these results indicate that MK-801 increases food intake through mechanisms independent of altered gastric emptying.
Collapse
Affiliation(s)
- M Covasa
- Department of Nutritional Sciences, College of Health and Human Development, The Pennsylvania State University, 126 South Henderson, University Park, PA 16802, USA.
| | | | | | | |
Collapse
|