1
|
Swan P, Johnson B, le Roux CW, Miras AD. Harnessing the melanocortin system in the control of food intake and glucose homeostasis. Peptides 2024; 179:171255. [PMID: 38834138 DOI: 10.1016/j.peptides.2024.171255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
The central and peripheral melanocortin system, comprising of five receptors and their endogenous ligands, is responsible for a wide array of physiological functions such as skin pigmentation, sexual function and development, and inflammation. A growing body of both clinical and pre-clinical research is demonstrating the relevance of this system in metabolic health. Disruption of hypothalamic melanocortin signalling is the most common cause of monogenic obesity in humans. Setmelanotide, an FDA-approved analogue of alpha-melanocyte stimulating hormone (α-MSH) that functions by restoring central melanocortin signalling, has proven to be a potent pharmacological tool in the treatment of syndromic obesity. As the first effective therapy targeting the melanocortin system to treat metabolic disorders, its approval has sparked research to further harness the links between these melanocortin receptors and metabolic processes. Here, we outline the structure of the central and peripheral melanocortin system, discuss its critical role in the regulation of food intake, and review promising targets that may hold potential to treat metabolic disorders in humans.
Collapse
Affiliation(s)
- Patrick Swan
- Diabetes Research Centre, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom; Diabetes Complications Research Centre, Conway Institute, University College Dublin, Dublin, Ireland.
| | - Brett Johnson
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, United Kingdom
| | - Carel W le Roux
- Diabetes Research Centre, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom; Diabetes Complications Research Centre, Conway Institute, University College Dublin, Dublin, Ireland
| | - Alexander D Miras
- Diabetes Research Centre, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom; Division of Diabetes, Endocrinology and Metabolism, Imperial College London, United Kingdom
| |
Collapse
|
2
|
Giraudier M, Ventura-Bort C, Weymar M. Effects of Transcutaneous Auricular Vagus Nerve Stimulation on the P300: Do Stimulation Duration and Stimulation Type Matter? Brain Sci 2024; 14:690. [PMID: 39061430 PMCID: PMC11274684 DOI: 10.3390/brainsci14070690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Non-invasive transcutaneous auricular vagus nerve stimulation (taVNS) has attracted increasing interest as a neurostimulation tool with potential applications in modulating cognitive processes such as attention and memory, possibly through the modulation of the locus-coeruleus noradrenaline system. Studies examining the P300 brain-related component as a correlate of noradrenergic activity, however, have yielded inconsistent findings, possibly due to differences in stimulation parameters, thus necessitating further investigation. In this event-related potential study involving 61 participants, therefore, we examined how changes in taVNS parameters, specifically stimulation type (interval vs. continuous stimulation) and duration, influence P300 amplitudes during a visual novelty oddball task. Although no effects of stimulation were found over the whole cluster and time window of the P300, cluster-based permutation tests revealed a distinct impact of taVNS on the P300 response for a small electrode cluster, characterized by larger amplitudes observed for easy targets (i.e., stimuli that are easily discernible from standards) following taVNS compared to sham stimulation. Notably, our findings suggested that the type of stimulation significantly modulated taVNS effects on the P300, with continuous stimulation showing larger P300 differences (taVNS vs. sham) for hard targets and standards compared to interval stimulation. We observed no interaction effects of stimulation duration on the target-related P300. While our findings align with previous research, further investigation is warranted to fully elucidate the influence of taVNS on the P300 component and its potential utility as a reliable marker for neuromodulation in this field.
Collapse
Affiliation(s)
- Manon Giraudier
- Department of Biological Psychology and Affective Science, Faculty of Human Sciences, University of Potsdam, Campus Golm, Karl-Liebknecht-Str. 24/25, 14476 Potsdam, Germany; (C.V.-B.); (M.W.)
| | | | | |
Collapse
|
3
|
Seitz J, Lahaye E, Andreani NA, Thomas B, Takhlidjt S, Chartrel N, Herpertz-Dahlmann B, Baines JF, Fetissov SO. Long-Term Dynamics of Serum α-MSH and α-MSH-Binding Immunoglobulins with a Link to Gut Microbiota Composition in Patients with Anorexia Nervosa. Neuroendocrinology 2024; 114:907-920. [PMID: 38852579 PMCID: PMC11460951 DOI: 10.1159/000539316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/06/2024] [Indexed: 06/11/2024]
Abstract
INTRODUCTION Immunoglobulins (Ig) reactive with α-melanocyte-stimulating hormone (α-MSH), an anorexigenic neuropeptide, are present in humans and were previously associated with eating disorders. In this longitudinal study involving patients with anorexia nervosa (AN), we determined whether α-MSH in serum is bound to IgG and analyzed long-term dynamics of both α-MSH peptide and α-MSH-reactive Ig in relation to changes in BMI and gut microbiota composition. METHODS The study included 64 adolescents with a restrictive form of AN, whose serum samples were collected at hospital admission, discharge, and during a 1-year follow-up visit and 41 healthy controls, all females. RESULTS We found that in both study groups, approximately 40% of serum α-MSH was reversibly bound to IgG and that levels of α-MSH-reactive IgG but not of α-MSH peptide in patients with AN were low at hospital admission but recovered 1 year later. Total IgG levels were also low at admission. Moreover, BMI-standard deviation score correlated positively with α-MSH IgG in both groups studied but negatively with α-MSH peptide only in controls. Significant correlations between the abundance of specific bacterial taxa in the gut microbiota and α-MSH peptide and IgG levels were found in both study groups, but they were more frequent in controls. CONCLUSION We conclude that IgG in the blood plays a role as an α-MSH-binding protein, whose characteristics are associated with BMI in both patients with AN and controls. Furthermore, the study suggests that low production of α-MSH-reactive IgG during the starvation phase in patients with AN may be related to altered gut microbiota composition.
Collapse
Affiliation(s)
- Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, University Hospital, RWTH University Aachen, Aachen, Germany
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, LVR-University Hospital, Essen, Germany
| | - Emilie Lahaye
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Nadia Andrea Andreani
- Guest Group Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Section of Evolutionary Medicine, Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Benjamin Thomas
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Saloua Takhlidjt
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Nicolas Chartrel
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Beate Herpertz-Dahlmann
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, University Hospital, RWTH University Aachen, Aachen, Germany
| | - John F. Baines
- Guest Group Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Section of Evolutionary Medicine, Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Sergueï O. Fetissov
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| |
Collapse
|
4
|
Konjusha A, Yu S, Mückschel M, Colzato L, Ziemssen T, Beste C. Auricular Transcutaneous Vagus Nerve Stimulation Specifically Enhances Working Memory Gate Closing Mechanism: A System Neurophysiological Study. J Neurosci 2023; 43:4709-4724. [PMID: 37221097 PMCID: PMC10286950 DOI: 10.1523/jneurosci.2004-22.2023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 04/24/2023] [Accepted: 04/30/2023] [Indexed: 05/25/2023] Open
Abstract
Everyday tasks and goal-directed behavior involve the maintenance and continuous updating of information in working memory (WM). WM gating reflects switches between these two core states. Neurobiological considerations suggest that the catecholaminergic and the GABAergic are likely involved in these dynamics. Both of these neurotransmitter systems likely underlie the effects to auricular transcutaneous vagus nerve stimulation (atVNS). We examine the effects of atVNS on WM gating dynamics and their underlying neurophysiological and neurobiological processes in a randomized crossover study design in healthy humans of both sexes. We show that atVNS specifically modulates WM gate closing and thus specifically modulates neural mechanisms enabling the maintenance of information in WM. WM gate opening processes were not affected. atVNS modulates WM gate closing processes through the modulation of EEG alpha band activity. This was the case for clusters of activity in the EEG signal referring to stimulus information, motor response information, and fractions of information carrying stimulus-response mapping rules during WM gate closing. EEG-beamforming shows that modulations of activity in fronto-polar, orbital, and inferior parietal regions are associated with these effects. The data suggest that these effects are not because of modulations of the catecholaminergic (noradrenaline) system as indicated by lack of modulatory effects in pupil diameter dynamics, in the inter-relation of EEG and pupil diameter dynamics and saliva markers of noradrenaline activity. Considering other findings, it appears that a central effect of atVNS during cognitive processing refers to the stabilization of information in neural circuits, putatively mediated via the GABAergic system.SIGNIFICANCE STATEMENT Goal-directed behavior depends on how well information in short-term memory can be flexibly updated but also on how well it can be shielded from distraction. These two functions were guarded by a working memory gate. We show how an increasingly popular brain stimulation techniques specifically enhances the ability to close the working memory gate to shield information from distraction. We show what physiological and anatomic aspects underlie these effects.
Collapse
Affiliation(s)
- Anyla Konjusha
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden 01307, Germany
| | - Shijing Yu
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden 01307, Germany
| | - Moritz Mückschel
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden 01307, Germany
| | - Lorenza Colzato
- Faculty of Psychology, Shandong Normal University, Jinan 250014, China
| | - Tjalf Ziemssen
- Department of Neurology, Faculty of Medicine, MS Centre, TU Dresden, Dresden 01307, Germany
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden 01307, Germany
- Faculty of Psychology, Shandong Normal University, Jinan 250014, China
| |
Collapse
|
5
|
Giraudier M, Ventura-Bort C, Burger AM, Claes N, D'Agostini M, Fischer R, Franssen M, Kaess M, Koenig J, Liepelt R, Nieuwenhuis S, Sommer A, Usichenko T, Van Diest I, von Leupoldt A, Warren CM, Weymar M. Evidence for a modulating effect of transcutaneous auricular vagus nerve stimulation (taVNS) on salivary alpha-amylase as indirect noradrenergic marker: A pooled mega-analysis. Brain Stimul 2022; 15:1378-1388. [PMID: 36183953 DOI: 10.1016/j.brs.2022.09.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 09/22/2022] [Accepted: 09/22/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Non-invasive transcutaneous auricular vagus nerve stimulation (taVNS) has received tremendous attention as a potential neuromodulator of cognitive and affective functions, which likely exerts its effects via activation of the locus coeruleus-noradrenaline (LC-NA) system. Reliable effects of taVNS on markers of LC-NA system activity, however, have not been demonstrated yet. METHODS The aim of the present study was to overcome previous limitations by pooling raw data from a large sample of ten taVNS studies (371 healthy participants) that collected salivary alpha-amylase (sAA) as a potential marker of central NA release. RESULTS While a meta-analytic approach using summary statistics did not yield any significant effects, linear mixed model analyses showed that afferent stimulation of the vagus nerve via taVNS increased sAA levels compared to sham stimulation (b = 0.16, SE = 0.05, p = 0.001). When considering potential confounders of sAA, we further replicated previous findings on the diurnal trajectory of sAA activity. CONCLUSION(S) Vagal activation via taVNS increases sAA release compared to sham stimulation, which likely substantiates the assumption that taVNS triggers NA release. Moreover, our results highlight the benefits of data pooling and data sharing in order to allow stronger conclusions in research.
Collapse
Affiliation(s)
- Manon Giraudier
- Department of Biological Psychology and Affective Science, Faculty of Human Sciences, University of Potsdam, Potsdam, Germany.
| | - Carlos Ventura-Bort
- Department of Biological Psychology and Affective Science, Faculty of Human Sciences, University of Potsdam, Potsdam, Germany
| | | | - Nathalie Claes
- Research Group Health Psychology, KU Leuven, Leuven, Belgium
| | | | - Rico Fischer
- Department of Psychology, University of Greifswald, Greifswald, Germany
| | | | - Michael Kaess
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland; Department of Child and Adolescent Psychiatry, Centre for Psychosocial Medicine, University of Heidelberg, Heidelberg, Germany
| | - Julian Koenig
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Cologne, Germany
| | - Roman Liepelt
- Department of General Psychology: Judgment, Decision Making, Action, Faculty of Psychology, University of Hagen (FernUniversität in Hagen), Hagen, Germany
| | - Sander Nieuwenhuis
- Institute of Psychology, Leiden University, Netherlands; Leiden Institute for Brain and Cognition, Leiden University, Netherlands
| | - Aldo Sommer
- Department of General Psychology: Judgment, Decision Making, Action, Faculty of Psychology, University of Hagen (FernUniversität in Hagen), Hagen, Germany; Department of Exercise Physiology, German Sport University Cologne, Cologne, Germany
| | - Taras Usichenko
- Department of Anesthesiology, University Medicine of Greifswald, Greifswald, Germany; Department of Anesthesia, McMaster University, Hamilton, Canada
| | - Ilse Van Diest
- Research Group Health Psychology, KU Leuven, Leuven, Belgium
| | | | - Christopher M Warren
- Emma Eccles Jones College of Education and Human Services, Utah State University, United States
| | - Mathias Weymar
- Department of Biological Psychology and Affective Science, Faculty of Human Sciences, University of Potsdam, Potsdam, Germany; Faculty of Health Sciences Brandenburg, University of Potsdam, Potsdam, Germany.
| |
Collapse
|
6
|
Neuhuber WL, Berthoud HR. Functional anatomy of the vagus system: How does the polyvagal theory comply? Biol Psychol 2022; 174:108425. [PMID: 36100134 DOI: 10.1016/j.biopsycho.2022.108425] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/07/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022]
Abstract
Due to its pivotal role in autonomic networks and interoception, the vagus attracts continued interest from both basic scientists and therapists of various clinical disciplines. In particular, the widespread use of heart rate variability as an index of autonomic cardiac control and a proposed central role of the vagus in biopsychological concepts, e.g., the polyvagal theory, provide a good opportunity to recall basic features of vagal anatomy. In addition to the "classical" vagal brainstem nuclei, i.e., dorsal motor nucleus, nucleus ambiguus and nucleus tractus solitarii, the spinal trigeminal and paratrigeminal nuclei come into play as targets of vagal afferents. On the other hand, the nucleus of the solitary tract receives and integrates not only visceral but also somatic afferents.
Collapse
Affiliation(s)
- Winfried L Neuhuber
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität, Krankenhausstrasse 9, Erlangen, Germany.
| | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| |
Collapse
|
7
|
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
|
8
|
Azimzadeh M, Beheshti S. Antagonism of the ghrelin receptor type 1a in the rat brain induces status epilepticus in an electrical kindling model of epilepsy. Psychopharmacology (Berl) 2022; 239:479-487. [PMID: 34845505 DOI: 10.1007/s00213-021-06026-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 11/08/2021] [Indexed: 10/19/2022]
Abstract
Studies have shown the anti-seizure properties of the ghrelin hormone in different models of epilepsy. Nevertheless, the role of the endogenous ghrelin is unknown in the electrical kindling model of epilepsy. In this study, we evaluated the effect of the antagonism of the ghrelin receptors in the brain of fully kindled rats. Adult male Wistar rats weighing 300 g were used. Animals were stereotaxically implanted with two uni-polar electrodes in the skull surface and a tri-polar electrode in the basolateral amygdala, and a guide cannula in the left lateral ventricle. Animals underwent a rapid kindling protocol. After showing three consecutive stages of five seizures, the animals were considered fully kindled. D-Lys-3-GHRP-6 (1, 50, and 100 μg/rat) was injected intracerebroventricularly (i.c.v.) in the kindled animals. Each rat was considered as its control and received a single dose of D-Lys-3-GHRP-6. Seizure parameters including after discharge duration (ADD), seizure stage (SS), stage four latency (S4L), and stage five duration (S5D) were recorded. The paired t test indicated a significant increase in seizure induction. D-Lys-3-GHRP-6 (1 μg/rat; i.c.v.) prolonged ADD in the kindled rats, significantly. D-Lys-3-GHRP-6 (50 and 100 μg/rat; i.c.v.) induced spontaneous seizures, which led to status epilepticus in the kindled rats. The results indicate that the antagonism of the ghrelin functional receptors prolongs seizures and induces status epilepticus in the kindling model of epilepsy, and propose that the endogenous ghrelin signaling has crucial antiepileptic properties.
Collapse
Affiliation(s)
- Mansour Azimzadeh
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Siamak Beheshti
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
9
|
Neuhuber WL, Berthoud HR. Functional anatomy of the vagus system - Emphasis on the somato-visceral interface. Auton Neurosci 2021; 236:102887. [PMID: 34634680 PMCID: PMC8627476 DOI: 10.1016/j.autneu.2021.102887] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/02/2021] [Accepted: 09/21/2021] [Indexed: 11/18/2022]
Abstract
Due to its pivotal role in autonomic networks, the vagus attracts continuous interest from both basic scientists and clinicians. In particular, recent advances in vagus nerve stimulation strategies and their application to pathological conditions beyond epilepsy provide a good opportunity to recall basic features of vagal peripheral and central anatomy. In addition to the "classical" vagal brainstem nuclei, i.e., dorsal motor nucleus, nucleus ambiguus and nucleus tractus solitarii, the spinal trigeminal and paratrigeminal nuclei come into play as targets of vagal afferents. On the other hand, the nucleus of the solitary tract receives and integrates not only visceral but also somatic afferents. Thus, the vagus system participates significantly in what may be defined as "somato-visceral interface".
Collapse
Affiliation(s)
- Winfried L Neuhuber
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University, Krankenhausstrasse 9, Erlangen, Germany.
| | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| |
Collapse
|
10
|
Pitra S, Smith BN. Musings on the wanderer: What's new in our understanding of vago-vagal reflexes? VI. Central vagal circuits that control glucose metabolism. Am J Physiol Gastrointest Liver Physiol 2021; 320:G175-G182. [PMID: 33205998 PMCID: PMC7938771 DOI: 10.1152/ajpgi.00368.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Neurons in the brain stem dorsal vagal complex (DVC) take part in a continuous bidirectional crosstalk, in which they receive and respond to a vast array of signaling molecules, including glucose. Importantly, chronic dysregulation of blood glucose concentration, a hallmark of high prevalence pathologies, such as diabetes and metabolic syndrome, can induce neuroplasticity in DVC neural networks, which is hypothesized to either contribute to or compensate for the glycemic or insulinemic dysregulation observed in these conditions. Here, we revisit the topic of vagal reflexes to review recent research on the importance of DVC function in regulating systemic glucose homeostasis and the neuroplastic changes in this brain region that are associated with systemic glucose alterations. We also discuss the critical connection between these nuclei and the gut and the role of central vagal circuits in the favorable outcomes associated with bariatric surgical procedures for metabolic disorders.
Collapse
Affiliation(s)
- Soledad Pitra
- 1Department of Neuroscience, University of Kentucky, Lexington, Kentucky
| | - Bret N. Smith
- 1Department of Neuroscience, University of Kentucky, Lexington, Kentucky,2Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
11
|
Hypothalamic Pomc Neurons Innervate the Spinal Cord and Modulate the Excitability of Premotor Circuits. Curr Biol 2020; 30:4579-4593.e7. [PMID: 32976803 DOI: 10.1016/j.cub.2020.08.103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 07/30/2020] [Accepted: 08/27/2020] [Indexed: 12/20/2022]
Abstract
Locomotion requires energy, yet animals need to increase locomotion in order to find and consume food in energy-deprived states. While such energy homeostatic coordination suggests brain origin, whether the central melanocortin 4 receptor (Mc4r) system directly modulates locomotion through motor circuits is unknown. Here, we report that hypothalamic Pomc neurons in zebrafish and mice have long-range projections into spinal cord regions harboring Mc4r-expressing V2a interneurons, crucial components of the premotor networks. Furthermore, in zebrafish, Mc4r activation decreases the excitability of spinal V2a neurons as well as swimming and foraging, while systemic or V2a neuron-specific blockage of Mc4r promotes locomotion. In contrast, in mice, electrophysiological recordings revealed that two-thirds of V2a neurons in lamina X are excited by the Mc4r agonist α-MSH, and acute inhibition of Mc4r signaling reduces locomotor activity. In addition, we found other Mc4r neurons in spinal lamina X that are inhibited by α-MSH, which is in line with previous studies in rodents where Mc4r agonists reduced locomotor activity. Collectively, our studies identify spinal V2a interneurons as evolutionary conserved second-order neurons of the central Mc4r system, providing a direct anatomical and functional link between energy homeostasis and locomotor control systems. The net effects of this modulatory system on locomotor activity can vary between different vertebrate species and, possibly, even within one species. We discuss the biological sense of this phenomenon in light of the ambiguity of locomotion on energy balance and the different living conditions of the different species.
Collapse
|
12
|
Dietary patterns interact with the variations of 18q21.23 rs17782313 locus on regulation of hypothalamic-pituitary axis hormones and cardio-metabolic risk factors in obesity. Eat Weight Disord 2020; 25:1447-1459. [PMID: 32016782 DOI: 10.1007/s40519-020-00855-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/21/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Evidence shows the role of polymorphisms in rs17782313 MC4R gene with increased risk of obesity in Asians adult. In the current report, we investigated the interaction between rs17782313 MC4R gene and major dietary patterns on α-melanocyte stimulating hormone (α-MSH), Agouti-related peptide (AgRP), serum lipids and blood pressure among obese individuals. METHODS This cross-sectional study was performed in 288 obese adults between 20 and 50 years of age. Anthropometric measurements and biochemical assays were conducted with standard methods. To evaluate appetite, the Visual Analogue Scale (VAS) was used. Dietary patterns were obtained by principal component analysis (PCA). Genotyping of rs17782313 was assessed by restriction fragment length polymorphism (PCR-RFLP) method. RESULTS Three major dietary patterns were extracted: Prudent Dietary Pattern (PDP), Legume Dietary Pattern (LDP) and Mixed Dietary Pattern (MDP). Higher PDP score was associated with reduced SBP and insulin concentration while highest MDP score was associated with lower TG concentration (P < 0.05). Significant interactions were observed between higher adherence to PDP and rs17782313 CC genotype on increased SBP (PInteraction = 0.04), serum insulin (PInteraction = 0.05) and AgRP (PInteraction = 0.03) and also between higher adherence to MDP and CC genotype of rs17782313 on reduced serum TG (P = 0.03). CONCLUSIONS The findings of the current study showed that being on CC genotype of rs17782313 polymorphism made obese individuals more prone to have higher SBP, insulin and AgRP even in highest adherence to PDP. However, adherence to MDP could attenuate the risky effects of being on CC genotype of rs17782313 by reducing serum TG concentrations. LEVEL OF EVIDENCE Level V, cross-sectional descriptive study.
Collapse
|
13
|
Brain-Derived Neurotrophic Factor Affects mRNA and miRNA Expression of the Appetite Regulating Centre in the Sheep Arcuate Nucleus. ANNALS OF ANIMAL SCIENCE 2020. [DOI: 10.2478/aoas-2020-0015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Abstract
The neuromodulatory effects of brain-derived neurotrophic factor (BDNF) on appetite regulation centre peptide gene activity in the sheep hypothalamus have not been examined yet. The aim of this study was to determine whether BDNF participates in modulation of neuropeptide Y (npy), agouti-related peptide (agrp), cocaine and amphetamine regulated transcript (cart), and proopiomelanocortin (pomc) mRNA expression and selected microRNAs in the sheep hypothalamic arcuate (ARC) nucleus. Animals (Polish Merino sheep, n=24) were divided into three groups. The control group received a central infusion of Ringer-Locke solution (480 µl/day) whereas the experimental groups were treated with BDNF in two doses: 10 or 60 μg/480 µl/day. All sheep received four intracerebroventricular infusions (performed from 08:40 a.m. to 01:30 p.m.; infusion scheme: 4 x 50 min infusions with 30 min intervals between them) on each of three consecutive days. Immediately after the last infusion, the sheep were slaughtered, and selected structures of the hypothalamus were frozen for further real-time qPCR analysis. Central infusion of BDNF evoked dose-dependent changes in npy, agrp, cart, pomc and peptidylglicine alpha-amidating monooxygenase (pam) mRNA expression in the sheep ARC nucleus. An increase in npy, agrp and pomc mRNA expression but also a decrease in cart mRNA expression in the ARC nucleus were detected. Moreover, a decrease in pam (gene encoding an enzyme that converts POMC into α-MSH) mRNA expression, was also noted. Furthermore, after central BDNF administration, changes in miRNA-33a-5p, miRNA-33b-5p, miRNA-377-3p, miRNA-214-3p, miRNA-485 and miRNA-488 expression were observed. Based on the presented results, it can be concluded that BDNF may affect the appetite regulating centre activity through modulation of npy, agrp, cart, pomc and pam mRNA expression in the ARC nucleus. It was also revealed that BDNF modulates miRNA expression in the sheep ARC nucleus.
Collapse
|
14
|
Fortin SM, Chen J, Hayes MR. Hindbrain melanocortin 3/4 receptors modulate the food intake and body weight suppressive effects of the GLP-1 receptor agonist, liraglutide. Physiol Behav 2020; 220:112870. [PMID: 32179053 PMCID: PMC7227776 DOI: 10.1016/j.physbeh.2020.112870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/14/2020] [Accepted: 03/09/2020] [Indexed: 12/19/2022]
Abstract
Simultaneously targeting multiple energy balance control systems is a promising direction for the development of obesity pharmacotherapies. Here, we explore the interaction between the GLP-1 and melanocortin system within the dorsal vagal complex (DVC) of the caudal brainstem. Using a pharmacological approach, we demonstrate that the full anorectic potential of liraglutide, an FDA-approved GLP-1 analog for the treatment of obesity, requires DVC melanocortin 3/4 receptor (MC3/4R) signaling. Specifically, the food intake and body weight suppressive effects of liraglutide were attenuated by DVC administration of the MC3/4R antagonist SHU9119. In contrast, the anorectic effects of liraglutide were enhanced by combined activation of DVC MC3/4Rs using the agonist MTII. Our findings highlight the modulation of liraglutide-induced anorexia by DVC MC3/4R signaling, thereby suggesting a site of action at which two important energy balance control systems interact.
Collapse
Affiliation(s)
- Samantha M Fortin
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States.
| | - Jack Chen
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Matthew R Hayes
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States.
| |
Collapse
|
15
|
Berthoud HR, Neuhuber WL. Vagal mechanisms as neuromodulatory targets for the treatment of metabolic disease. Ann N Y Acad Sci 2019; 1454:42-55. [PMID: 31268181 PMCID: PMC6810744 DOI: 10.1111/nyas.14182] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/23/2019] [Accepted: 06/05/2019] [Indexed: 12/30/2022]
Abstract
With few effective treatments available, the global rise of metabolic diseases, including obesity, type 2 diabetes mellitus, and cardiovascular disease, seems unstoppable. Likely caused by an obesogenic environment interacting with genetic susceptibility, the pathophysiology of obesity and metabolic diseases is highly complex and involves crosstalk between many organs and systems, including the brain. The vagus nerve is in a key position to bidirectionally link several peripheral metabolic organs with the brain and is increasingly targeted for neuromodulation therapy to treat metabolic disease. Here, we review the basics of vagal functional anatomy and its implications for vagal neuromodulation therapies. We find that most existing vagal neuromodulation techniques either ignore or misinterpret the rich functional specificity of both vagal efferents and afferents as demonstrated by a large body of literature. This lack of specificity of manipulating vagal fibers is likely the reason for the relatively poor beneficial long‐term effects of such therapies. For these therapies to become more effective, rigorous validation of all physiological endpoints and optimization of stimulation parameters as well as electrode placements will be necessary. However, given the large number of function‐specific fibers in any vagal branch, genetically guided neuromodulation techniques are more likely to succeed.
Collapse
Affiliation(s)
- Hans-Rudolf Berthoud
- Neurobiology of Nutrition and Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Winfried L Neuhuber
- Institut fur Anatomie und Zellbiologie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
16
|
Hankir MK, Seyfried F, Miras AD, Cowley MA. Brain Feeding Circuits after Roux-en-Y Gastric Bypass. Trends Endocrinol Metab 2018; 29:218-237. [PMID: 29475578 DOI: 10.1016/j.tem.2018.01.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/17/2018] [Accepted: 01/25/2018] [Indexed: 12/12/2022]
Abstract
Metabolic surgical procedures, such as Roux-en-Y gastric bypass (RYGB), uniquely reprogram feeding behavior and body weight in obese subjects. Clinical neuroimaging and animal studies are only now beginning to shed light on some of the underlying central mechanisms. We present here the roles of key brain neurotransmitter/neuromodulator systems in food choice, value, and intake at various stages after RYGB. In doing so, we elaborate on how known signals emanating from the reorganized gut, including peptide hormones and microbiota products, impinge on newly mapped homeostatic and hedonic brain feeding circuits. Continued progress in the rapidly evolving field of metabolic surgery will inform the design of more effective weight-loss compounds.
Collapse
Affiliation(s)
- Mohammed K Hankir
- Department of Experimental Surgery, University Hospital Wuerzburg, Wuerzburg, Bavaria 97080, Germany; German Research Foundation Collaborative Research Center in Obesity Mechanisms, University of Leipzig, Leipzig, Saxony 04103, Germany.
| | - Florian Seyfried
- Department of Experimental Surgery, University Hospital Wuerzburg, Wuerzburg, Bavaria 97080, Germany
| | - Alexander D Miras
- Department of Investigative Science, Imperial College London Academic Healthcare Centre, London W12 0NN, UK
| | - Michael A Cowley
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Physiology, Monash University, Victoria 3800, Australia
| |
Collapse
|
17
|
Understanding melanocortin-4 receptor control of neuronal circuits: Toward novel therapeutics for obesity syndrome. Pharmacol Res 2018; 129:10-19. [DOI: 10.1016/j.phrs.2018.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/08/2018] [Accepted: 01/08/2018] [Indexed: 01/25/2023]
|
18
|
Melanocortin-4 receptor regulation of pain. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2515-2522. [DOI: 10.1016/j.bbadis.2017.05.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 05/18/2017] [Accepted: 05/18/2017] [Indexed: 11/17/2022]
|
19
|
Melanocortins, Melanocortin Receptors and Multiple Sclerosis. Brain Sci 2017; 7:brainsci7080104. [PMID: 28805746 PMCID: PMC5575624 DOI: 10.3390/brainsci7080104] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/07/2017] [Accepted: 08/08/2017] [Indexed: 02/07/2023] Open
Abstract
The melanocortins and their receptors have been extensively investigated for their roles in the hypothalamo-pituitary-adrenal axis, but to a lesser extent in immune cells and in the nervous system outside the hypothalamic axis. This review discusses corticosteroid dependent and independent effects of melanocortins on the peripheral immune system, central nervous system (CNS) effects mediated through neuronal regulation of immune system function, and direct effects on endogenous cells in the CNS. We have focused on the expression and function of melanocortin receptors in oligodendroglia (OL), the myelin producing cells of the CNS, with the goal of identifying new therapeutic approaches to decrease CNS damage in multiple sclerosis as well as to promote repair. It is clear that melanocortin signaling through their receptors in the CNS has potential for neuroprotection and repair in diseases like MS. Effects of melanocortins on the immune system by direct effects on the circulating cells (lymphocytes and monocytes) and by signaling through CNS cells in regions lacking a mature blood brain barrier are clear. However, additional studies are needed to develop highly effective MCR targeted therapies that directly affect endogenous cells of the CNS, particularly OL, their progenitors and neurons.
Collapse
|
20
|
Grabauskas G, Owyang C. Plasticity of vagal afferent signaling in the gut. MEDICINA-LITHUANIA 2017; 53:73-84. [PMID: 28454890 PMCID: PMC6318799 DOI: 10.1016/j.medici.2017.03.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 03/21/2017] [Indexed: 12/13/2022]
Abstract
Vagal sensory neurons mediate the vago-vagal reflex which, in turn, regulates a wide array of gastrointestinal functions including esophageal motility, gastric accommodation and pancreatic enzyme secretion. These neurons also transmit sensory information from the gut to the central nervous system, which then mediates the sensations of nausea, fullness and satiety. Recent research indicates that vagal afferent neurons process non-uniform properties and a significant degree of plasticity. These properties are important to ensure that vagally regulated gastrointestinal functions respond rapidly and appropriately to various intrinsic and extrinsic factors. Similar plastic changes in the vagus also occur in pathophysiological conditions, such as obesity and diabetes, resulting in abnormal gastrointestinal functions. A clear understanding of the mechanisms which mediate these events may provide novel therapeutic targets for the treatment of gastrointestinal disorders due to vago-vagal pathway malfunctions.
Collapse
Affiliation(s)
- Gintautas Grabauskas
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48019, USA.
| | - Chung Owyang
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48019, USA
| |
Collapse
|
21
|
Khlaifia A, Matias I, Cota D, Tell F. Nutritional status-dependent endocannabinoid signalling regulates the integration of rat visceral information. J Physiol 2017; 595:3267-3285. [PMID: 28233325 DOI: 10.1113/jp273484] [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: 09/16/2016] [Accepted: 02/21/2017] [Indexed: 12/27/2022] Open
Abstract
KEY POINTS Vagal sensory inputs transmit information from the viscera to brainstem neurones located in the nucleus tractus solitarii to set physiological parameters. These excitatory synapses exhibit a CB1 endocannabinoid-induced long-term depression (LTD) triggered by vagal fibre stimulation. We investigated the impact of nutritional status on long-term changes in this long-term synaptic plasticity. Food deprivation prevents LTD induction by disrupting CB1 receptor signalling. Short-term refeeding restores the capacity of vagal synapses to express LTD. Ghrelin and cholecystokinin, respectively released during fasting and refeeding, play a key role in the control of LTD via the activation of energy sensing pathways such as AMPK and the mTOR and ERK pathways. ABSTRACT Communication form the viscera to the brain is essential to set physiological homoeostatic parameters but also to drive more complex behaviours such as mood, memory and emotional states. Here we investigated the impact of the nutritional status on long-term changes in excitatory synaptic transmission in the nucleus tractus solitarii, a neural hub integrating visceral signals. These excitatory synapses exhibit a CB1 endocannabinoid (eCB)-induced long-term depression (LTD) triggered by vagal fibre stimulation. Since eCB signalling is known to be an important component of homoeostatic regulation of the body and is regulated during various stressful conditions, we tested the hypothesis that food deprivation alters eCB signalling in central visceral afferent fibres. Food deprivation prevents eCB-LTD induction due to the absence of eCB signalling. This loss was reversed by blockade of ghrelin receptors. Activation of the cellular fuel sensor AMP-activated protein kinase or inhibition of the mechanistic target of rapamycin pathway abolished eCB-LTD in free-fed rats. Signals associated with energy surfeit, such as short-term refeeding, restore eCB-LTD induction, which in turn requires activation of cholecystokinin receptors and the extracellular signal-regulated kinase pathway. These data suggest a tight link between eCB-LTD in the NTS and nutritional status and shed light on the key role of eCB in the integration of visceral information.
Collapse
Affiliation(s)
- Abdessattar Khlaifia
- Aix-Marseille Université, CNRS, CRN2M, UMR 7286, 51 Boulevard Pierre Dramard, 13344, Marseille, France
| | - Isabelle Matias
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000, Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000, Bordeaux, France
| | - Daniela Cota
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000, Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000, Bordeaux, France
| | - Fabien Tell
- Aix-Marseille Université, CNRS, CRN2M, UMR 7286, 51 Boulevard Pierre Dramard, 13344, Marseille, France
| |
Collapse
|
22
|
Navarro M. The Role of the Melanocortin System in Drug and Alcohol Abuse. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 136:121-150. [DOI: 10.1016/bs.irn.2017.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
23
|
Cerritelli S, Hirschberg S, Hill R, Balthasar N, Pickering AE. Activation of Brainstem Pro-opiomelanocortin Neurons Produces Opioidergic Analgesia, Bradycardia and Bradypnoea. PLoS One 2016; 11:e0153187. [PMID: 27077912 PMCID: PMC4831707 DOI: 10.1371/journal.pone.0153187] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 03/14/2016] [Indexed: 11/19/2022] Open
Abstract
Opioids are widely used medicinally as analgesics and abused for hedonic effects, actions that are each complicated by substantial risks such as cardiorespiratory depression. These drugs mimic peptides such as β-endorphin, which has a key role in endogenous analgesia. The β-endorphin in the central nervous system originates from pro-opiomelanocortin (POMC) neurons in the arcuate nucleus and nucleus of the solitary tract (NTS). Relatively little is known about the NTSPOMC neurons but their position within the sensory nucleus of the vagus led us to test the hypothesis that they play a role in modulation of cardiorespiratory and nociceptive control. The NTSPOMC neurons were targeted using viral vectors in a POMC-Cre mouse line to express either opto-genetic (channelrhodopsin-2) or chemo-genetic (Pharmacologically Selective Actuator Modules). Opto-genetic activation of the NTSPOMC neurons in the working heart brainstem preparation (n = 21) evoked a reliable, titratable and time-locked respiratory inhibition (120% increase in inter-breath interval) with a bradycardia (125±26 beats per minute) and augmented respiratory sinus arrhythmia (58% increase). Chemo-genetic activation of NTSPOMC neurons in vivo was anti-nociceptive in the tail flick assay (latency increased by 126±65%, p<0.001; n = 8). All effects of NTSPOMC activation were blocked by systemic naloxone (opioid antagonist) but not by SHU9119 (melanocortin receptor antagonist). The NTSPOMC neurons were found to project to key brainstem structures involved in cardiorespiratory control (nucleus ambiguus and ventral respiratory group) and endogenous analgesia (periaqueductal gray and midline raphe). Thus the NTSPOMC neurons may be capable of tuning behaviour by an opioidergic modulation of nociceptive, respiratory and cardiac control.
Collapse
Affiliation(s)
- Serena Cerritelli
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Stefan Hirschberg
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Rob Hill
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Nina Balthasar
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Anthony E. Pickering
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, United Kingdom
- Department of Anaesthesia, University Hospitals Bristol, Bristol, BS2 8HW, United Kingdom
| |
Collapse
|
24
|
Carmody JS, Ahmad NN, Machineni S, Lajoie S, Kaplan LM. Weight Loss After RYGB Is Independent of and Complementary to Serotonin 2C Receptor Signaling in Male Mice. Endocrinology 2015; 156:3183-91. [PMID: 26066076 PMCID: PMC4541621 DOI: 10.1210/en.2015-1226] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Roux-en-Y gastric bypass (RYGB) typically leads to substantial, long-term weight loss (WL) and diabetes remission, although there is a wide variation in response to RYGB among individual patients. Defining the pathways through which RYGB works should aid in the development of less invasive anti-obesity treatments, whereas identifying weight-regulatory pathways unengaged by RYGB could facilitate the development of therapies that complement the beneficial effects of surgery. Activation of serotonin 2C receptors (5-HT2CR) by serotonergic drugs causes WL in humans and animal models. 5-HT2CR are located on neurons that activate the melanocortin-4 receptors, which are essential for WL after RYGB. We therefore sought to determine whether 5-HT2CR signaling is also essential for metabolic effects of RYGB or whether it is a potentially complementary pathway, the activation of which could extend the benefits of RYGB. Diet-induced obese male mice deficient for the 5-HT2CR and their wild-type littermates underwent RYGB or sham operation. Both groups lost similar amounts of weight after RYGB, demonstrating that the improved metabolic phenotype after RYGB is 5-HT2CR independent. Consistent with this hypothesis, wild-type RYGB-treated mice lost additional weight after the administration of the serotonergic drugs fenfluramine and meta-chlorophenylpiperazine but not the nonserotonergic agent topiramate. The fact that RYGB does not depend on 5-HT2CR signaling suggests that there are important WL mechanisms not fully engaged by surgery that could potentially be harnessed for medical treatment. These results suggest a rational basis for designing medical-surgical combination therapies to optimize clinical outcomes by exploiting complementary physiological mechanisms of action.
Collapse
Affiliation(s)
- Jill S Carmody
- Obesity, Metabolism, and Nutrition Institute and Gastrointestinal Unit (J.S.C., N.N.A., S.M., S.L., L.M.K.), Massachusetts General Hospital, Boston, Massachusetts 02114; and Department of Medicine (J.S.C., N.N.A., S.M., L.M.K.), Harvard Medical School, Boston, Massachusetts 02115
| | - Nadia N Ahmad
- Obesity, Metabolism, and Nutrition Institute and Gastrointestinal Unit (J.S.C., N.N.A., S.M., S.L., L.M.K.), Massachusetts General Hospital, Boston, Massachusetts 02114; and Department of Medicine (J.S.C., N.N.A., S.M., L.M.K.), Harvard Medical School, Boston, Massachusetts 02115
| | - Sriram Machineni
- Obesity, Metabolism, and Nutrition Institute and Gastrointestinal Unit (J.S.C., N.N.A., S.M., S.L., L.M.K.), Massachusetts General Hospital, Boston, Massachusetts 02114; and Department of Medicine (J.S.C., N.N.A., S.M., L.M.K.), Harvard Medical School, Boston, Massachusetts 02115
| | - Scott Lajoie
- Obesity, Metabolism, and Nutrition Institute and Gastrointestinal Unit (J.S.C., N.N.A., S.M., S.L., L.M.K.), Massachusetts General Hospital, Boston, Massachusetts 02114; and Department of Medicine (J.S.C., N.N.A., S.M., L.M.K.), Harvard Medical School, Boston, Massachusetts 02115
| | - Lee M Kaplan
- Obesity, Metabolism, and Nutrition Institute and Gastrointestinal Unit (J.S.C., N.N.A., S.M., S.L., L.M.K.), Massachusetts General Hospital, Boston, Massachusetts 02114; and Department of Medicine (J.S.C., N.N.A., S.M., L.M.K.), Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
25
|
Asarian L, Bächler T. Neuroendocrine control of satiation. Horm Mol Biol Clin Investig 2015; 19:163-92. [PMID: 25390024 DOI: 10.1515/hmbci-2014-0010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 03/17/2014] [Indexed: 01/13/2023]
Abstract
Abstract Eating is a simple behavior with complex functions. The unconscious neuroendocrine process that stops eating and brings a meal to its end is called satiation. Energy homeostasis is mediated accomplished through the control of meal size via satiation. It involves neural integrations of phasic negative-feedback signals related to ingested food and tonic signals, such as those related to adipose tissue mass. Energy homeostasis is accomplished through adjustments in meal size brought about by changes in these satiation signals. The best understood meal-derived satiation signals arise from gastrointestinal nutrient sensing. Gastrointestinal hormones secreted during the meal, including cholecystokinin, glucagon-like peptide 1, and PYY, mediate most of these. Other physiological signals arise from activation of metabolic-sensing neurons, mainly in the hypothalamus and caudal brainstem. We review both classes of satiation signal and their integration in the brain, including their processing by melanocortin, neuropeptide Y/agouti-related peptide, serotonin, noradrenaline, and oxytocin neurons. Our review is not comprehensive; rather, we discuss only what we consider the best-understood mechanisms of satiation, with a special focus on normally operating physiological mechanisms.
Collapse
|
26
|
Mimee A, Ferguson AV. Glycemic state regulates melanocortin, but not nesfatin-1, responsiveness of glucose-sensing neurons in the nucleus of the solitary tract. Am J Physiol Regul Integr Comp Physiol 2015; 308:R690-9. [PMID: 25695291 DOI: 10.1152/ajpregu.00477.2014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 02/02/2015] [Indexed: 01/30/2023]
Abstract
The nucleus of the solitary tract (NTS) is a medullary integrative center with critical roles in the coordinated control of energy homeostasis. Here, we used whole cell current-clamp recordings on rat NTS neurons in slice preparation to identify the presence of physiologically relevant glucose-sensing neurons. The majority of NTS neurons (n = 81) were found to be glucose-responsive, with 35% exhibiting a glucose-excited (GE) phenotype (mean absolute change in membrane potential: 9.5 ± 1.1 mV), and 21% exhibiting a glucose-inhibited (GI) response (mean: 6.3 ± 0.7 mV). Furthermore, we found glucose-responsive cells are preferentially influenced by the anorexigenic peptide α-melanocyte-stimulating hormone (α-MSH), but not nesfatin-1. Accordingly, alterations in glycemic state have profound effects on the responsiveness of NTS neurons to α-MSH, but not to nesfatin-1. Indeed, NTS neurons showed increasing responsiveness to α-MSH as extracellular glucose concentrations were decreased, and in hypoglycemic conditions, all NTS neurons were depolarized by α-MSH (mean 10.6 ± 3.2 mV; n = 8). Finally, decreasing levels of extracellular glucose correlated with a significant hyperpolarization of the baseline membrane potential of NTS neurons, highlighting the modulatory effect of glucose on the baseline excitability of cells in this region. Our findings reveal individual NTS cells are capable of integrating multiple sources of metabolically relevant inputs, highlight the rapid capacity for plasticity in medullary melanocortin circuits, and emphasize the critical importance of physiological recording conditions for electrophysiological studies pertaining to the central control of energy homeostasis.
Collapse
Affiliation(s)
- Andrea Mimee
- Queen's University, Department of Physiology, Kingston, Ontario, Canada
| | | |
Collapse
|
27
|
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
|
28
|
Stocker SD, Gordon KW. Glutamate receptors in the hypothalamic paraventricular nucleus contribute to insulin-induced sympathoexcitation. J Neurophysiol 2014; 113:1302-9. [PMID: 25475355 DOI: 10.1152/jn.00764.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The sympathoexcitatory response to insulin is mediated by neurons in the arcuate nucleus (ARC) and hypothalamic paraventricular nucleus (PVH). Previous studies have reported that stimulation of ARC neurons increases sympathetic nerve activity (SNA) and arterial blood pressure (ABP) through glutamate receptor activation in the PVH. Therefore, the purpose of the present study was to determine whether glutamatergic neurotransmission in the PVH contributes to insulin-induced sympathoexcitation. Male Sprague-Dawley rats (275-400 g) were infused with isotonic saline or insulin (3.75 mU · kg(-1) · min(-1)) plus 50% dextrose to maintain euglycemia. Intravenous infusion of insulin significantly increased lumbar SNA without a significant change in mean ABP, renal SNA, heart rate, or blood glucose. Bilateral PVH injection of the excitatory amino acid antagonist kynurenic acid (KYN) lowered lumbar SNA and ABP of animals infused with insulin. Similarly, a cocktail of the NMDA antagonist DL-2-amino-5-phosphonopentanoic acid (AP5) and non-NMDA antagonist 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) reduced lumbar SNA and mean ABP during infusion of insulin. In a final experiment, bilateral PVH injection of AP5 only, but not CNQX, lowered lumbar SNA and mean ABP of animals infused with insulin. The peak changes in lumbar SNA and mean ABP of insulin-treated animals were not different between KYN, AP5 plus CNQX, or AP5 alone. These drug treatments did not alter any variable in animals infused with saline. Altogether, these findings suggest that glutamatergic NMDA neurotransmission in the PVH contributes to insulin-induced sympathoexcitation.
Collapse
Affiliation(s)
- Sean D Stocker
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania; and Department of Neural and Behavioral Neuroscience, Pennsylvania State College of Medicine, Hershey, Pennsylvania
| | - Kathryn W Gordon
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania; and
| |
Collapse
|
29
|
Bhagat R, Fortna SR, Browning KN. Exposure to a high fat diet during the perinatal period alters vagal motoneurone excitability, even in the absence of obesity. J Physiol 2014; 593:285-303. [PMID: 25556801 DOI: 10.1113/jphysiol.2014.282806] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 10/24/2014] [Indexed: 01/02/2023] Open
Abstract
KEY POINTS Obesity is recognized as being multifactorial in origin, involving both genetic and environmental factors. The perinatal period is known to be critically important in the development of neural circuits responsible for energy homeostasis and the integration of autonomic reflexes. Diet-induced obesity alters the biophysical, pharmacological and morphological properties of vagal neurocircuits regulating upper gastrointestinal tract functions, including satiety. Less information is available, however, regarding the effects of a high fat diet (HFD) itself on the properties of vagal neurocircuits. The present study was designed to test the hypothesis that exposure to a HFD during the perinatal period alters the electrophysiological, pharmacological and morphological properties of vagal efferent motoneurones innervating the stomach. Our data indicate that perinatal HFD decreases the excitability of gastric-projecting dorsal motor nucleus neurones and dysregulates neurotransmitter release from synaptic inputs and that these alterations occur prior to the development of obesity. These findings represent the first direct evidence that exposure to a HFD modulates the processing of central vagal neurocircuits even in the absence of obesity. The perinatal period is critically important to the development of autonomic neural circuits responsible for energy homeostasis. Vagal neurocircuits are vital to the regulation of upper gastrointestinal functions, including satiety. Diet-induced obesity modulates the excitability and responsiveness of both peripheral vagal afferents and central vagal efferents but less information is available regarding the effects of diet per se on vagal neurocircuit functions. The aims of this study were to investigate whether perinatal exposure to a high fat diet (HFD) dysregulated dorsal motor nucleus of the vagus (DMV) neurones, prior to the development of obesity. Whole cell patch clamp recordings were made from gastric-projecting DMV neurones in thin brainstem slices from rats that were exposed to either a control diet or HFD from pregnancy day 13. Our data demonstrate that following perinatal HFD: (i) DMV neurones had decreased excitability and input resistance with a reduced ability to fire action potentials; (ii) the proportion of DMV neurones excited by cholecystokinin (CCK) was unaltered but the proportion of neurones in which CCK increased excitatory glutamatergic synaptic inputs was reduced; (iii) the tonic activation of presynaptic group II metabotropic glutamate receptors on inhibitory nerve terminals was attenuated, allowing modulation of GABAergic synaptic transmission; and (iv) the size and dendritic arborization of gastric-projecting DMV neurones was increased. These results suggest that perinatal HFD exposure compromises the excitability and responsiveness of gastric-projecting DMV neurones, even in the absence of obesity, suggesting that attenuation of vago-vagal reflex signalling may precede the development of obesity.
Collapse
Affiliation(s)
- Ruchi Bhagat
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA 17033, USA
| | | | | |
Collapse
|
30
|
Calik MW, Radulovacki M, Carley DW. A method of nodose ganglia injection in Sprague-Dawley rat. J Vis Exp 2014:e52233. [PMID: 25490160 DOI: 10.3791/52233] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Afferent signaling via the vagus nerve transmits important general visceral information to the central nervous system from many diverse receptors located in the organs of the abdomen and thorax. The vagus nerve communicates information from stimuli such as heart rate, blood pressure, bronchopulmonary irritation, and gastrointestinal distension to the nucleus of solitary tract of the medulla. The cell bodies of the vagus nerve are located in the nodose and petrosal ganglia, of which the majority are located in the former. The nodose ganglia contain a wealth of receptors for amino acids, monoamines, neuropeptides, and other neurochemicals that can modify afferent vagus nerve activity. Modifying vagal afferents through systemic peripheral drug treatments targeted at the receptors on nodose ganglia has the potential of treating diseases such as sleep apnea, gastroesophageal reflux disease, or chronic cough. The protocol here describes a method of injection neurochemicals directly into the nodose ganglion. Injecting neurochemicals directly into the nodose ganglia allows study of effects solely on cell bodies that modulate afferent nerve activity, and prevents the complication of involving the central nervous system as seen in systemic neurochemical treatment. Using readily available and inexpensive equipment, intranodose ganglia injections are easily done in anesthetized Sprague-Dawley rats.
Collapse
Affiliation(s)
- Michael W Calik
- Center for Narcolepsy, Sleep and Health Research, University of Illinois at Chicago; Department of Biobehavioral Health Science, University of Illinois at Chicago;
| | - Miodrag Radulovacki
- Center for Narcolepsy, Sleep and Health Research, University of Illinois at Chicago; Department of Pharmacology, University of Illinois at Chicago
| | - David W Carley
- Center for Narcolepsy, Sleep and Health Research, University of Illinois at Chicago; Department of Biobehavioral Health Science, University of Illinois at Chicago
| |
Collapse
|
31
|
Central vagal afferent endings mediate reduction of food intake by melanocortin-3/4 receptor agonist. J Neurosci 2014; 34:12636-45. [PMID: 25232103 DOI: 10.1523/jneurosci.1121-14.2014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Injection of the melanocortin-3/4 receptor agonist melanotan-II (MTII) into the nucleus of the solitary tract (NTS) produces rapid and sustained reduction of food intake. Melanocortin-4 receptors (MC4Rs) are expressed by vagal afferent endings in the NTS, but it is not known whether these endings participate in MTII-induced reduction of food intake. In experiments described here, we evaluated the contribution of central vagal afferent endings in MTII-induced reduction of food intake. Examination of rat hindbrain sections revealed that neuronal processes expressing immunoreactivity for the endogenous MC4R agonist α-melanoctyte-stimulating hormone course parallel and wrap around anterogradely labeled vagal afferent endings in the NTS and thus are aptly positioned to activate vagal afferent MC4Rs. Furthermore, MTII and endogenous MC4R agonists increased protein kinase A (PKA)-catalyzed phosphorylation of synapsin I in vagal afferent endings, an effect known to increase synaptic strength by enhancing neurotransmitter release in other neural systems. Hindbrain injection of a PKA inhibitor, KT5720, significantly attenuated MTII-induced reduction of food intake and the increase in synapsin I phosphorylation. Finally, unilateral nodose ganglion removal, resulting in degeneration of vagal afferent endings in the ipsilateral NTS, abolished MTII-induced synapsin I phosphorylation ipsilateral to nodose ganglion removal. Moreover, reduction of food intake following MTII injection into the NTS ipsilateral to nodose ganglion removal was significantly attenuated, whereas the response to MTII was not diminished when injected into the contralateral NTS. Altogether, our results suggest that reduction of food intake following hindbrain MC4R activation is mediated by central vagal afferent endings.
Collapse
|
32
|
Wu SW, Fenwick AJ, Peters JH. Channeling satiation: a primer on the role of TRP channels in the control of glutamate release from vagal afferent neurons. Physiol Behav 2014; 136:179-84. [PMID: 25290762 DOI: 10.1016/j.physbeh.2014.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 09/29/2014] [Indexed: 01/07/2023]
Abstract
Obesity results from the chronic imbalance between food intake and energy expenditure. To maintain homeostasis, the brainstem nucleus of the solitary tract (NTS) integrates peripheral information from visceral organs and initiates reflex pathways that control food intake and other autonomic functions. This peripheral-to-central neural communication occurs through activation of vagal afferent neurons which converge to form the solitary tract (ST) and synapse with strong glutamatergic contacts onto NTS neurons. Vagal afferents release glutamate containing vesicles via three distinct pathways (synchronous, asynchronous, and spontaneous) providing multiple levels of control through fast synaptic neurotransmission at ST-NTS synapses. While temperature at the NTS is relatively constant, vagal afferent neurons express an array of thermosensitive ion channels named transient receptor potential (TRP) channels. Here we review the evidence that TRP channels pre-synaptically control quantal glutamate release and examine the potential roles of TRP channels in vagally mediated satiety signaling. We summarize the current literature that TRP channels contribute to asynchronous and spontaneous release of glutamate which can distinctly influence the transfer of information across the ST-NTS synapse. In other words, multiple glutamate vesicle release pathways, guided by afferent TRP channels, provide for robust while adaptive neurotransmission and expand our understanding of vagal afferent signaling.
Collapse
Affiliation(s)
- Shaw-wen Wu
- Dept. of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Axel J Fenwick
- Dept. of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - James H Peters
- Dept. of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA.
| |
Collapse
|
33
|
Agosti F, López Soto EJ, Cabral A, Castrogiovanni D, Schioth HB, Perelló M, Raingo J. Melanocortin 4 receptor activation inhibits presynaptic N-type calcium channels in amygdaloid complex neurons. Eur J Neurosci 2014; 40:2755-65. [PMID: 24943127 DOI: 10.1111/ejn.12650] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 04/30/2014] [Accepted: 05/05/2014] [Indexed: 12/22/2022]
Abstract
The melanocortin 4 receptor (MC4R) is a G protein-coupled receptor involved in food intake and energy expenditure regulation. MC4R activation modifies neuronal activity but the molecular mechanisms by which this regulation occurs remain unclear. Here, we tested the hypothesis that MC4R activation regulates the activity of voltage-gated calcium channels and, as a consequence, synaptic activity. We also tested whether the proposed effect occurs in the amygdala, a brain area known to mediate the anorexigenic actions of MC4R signaling. Using the patch-clamp technique, we found that the activation of MC4R with its agonist melanotan II specifically inhibited 34.5 ± 1.5% of N-type calcium currents in transiently transfected HEK293 cells. This inhibition was concentration-dependent, voltage-independent and occluded by the Gαs pathway inhibitor cholera toxin. Moreover, we found that melanotan II specifically inhibited 25.9 ± 2.0% of native N-type calcium currents and 55.4 ± 14.4% of evoked inhibitory postsynaptic currents in mouse cultured amygdala neurons. In vivo, we found that the MC4R agonist RO27-3225 increased the marker of cellular activity c-Fos in several components of the amygdala, whereas the N-type channel blocker ω conotoxin GVIA increased c-Fos expression exclusively in the central subdivision of the amygdala. Thus, MC4R specifically inhibited the presynaptic N-type channel subtype, and this inhibition may be important for the effects of melanocortin in the central subdivision of the amygdala.
Collapse
Affiliation(s)
- Francina Agosti
- Laboratory of Electrophysiology, Multidisciplinary Institute of Cell Biology (IMBICE), Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), La Plata, Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
34
|
Swartz EM, Browning KN, Travagli RA, Holmes GM. Ghrelin increases vagally mediated gastric activity by central sites of action. Neurogastroenterol Motil 2014; 26:272-82. [PMID: 24261332 PMCID: PMC3907172 DOI: 10.1111/nmo.12261] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 10/19/2013] [Indexed: 12/31/2022]
Abstract
BACKGROUND Vagally dependent gastric reflexes are mediated through vagal afferent fibers synapsing upon neurons of the nucleus tractus solitarius (NTS) which, in turn modulate the preganglionic parasympathetic dorsal motor nucleus of the vagus (DMV) neurons within the medullary dorsal vagal complex (DVC). The expression and transport of ghrelin receptors has been documented for the afferent vagus nerve, and functional studies have confirmed that vagal pathways are integral to ghrelin-induced stimulation of gastric motility. However, the central actions of ghrelin within the DVC have not been explored fully. METHODS We assessed the responses to ghrelin in fasted rats using: (i) in vivo measurements of gastric tone and motility following IVth ventricle application or unilateral microinjection of ghrelin into the DVC and (ii) whole cell recordings from gastric-projecting neurons of the DMV. KEY RESULTS (i) IVth ventricle application or unilateral microinjection of ghrelin into the DVC-elicited contractions of the gastric corpus via excitation of a vagal cholinergic efferent pathway and (ii) ghrelin facilitates excitatory, but not inhibitory, presynaptic transmission to DMV neurons. CONCLUSIONS & INFERENCES Our data indicate that ghrelin acts centrally by activating excitatory synaptic inputs onto DMV neurons, resulting in increased cholinergic drive by way of vagal motor innervation to the stomach.
Collapse
Affiliation(s)
| | | | | | - Gregory M. Holmes
- Corresponding Author: Dr. Gregory M. Holmes, Penn State University College of Medicine, 500 University Dr., H181, Hershey, PA 17033, Tel: +1 717 531-6413, fax; +1 717 531-5184,
| |
Collapse
|
35
|
Acosta A, Camilleri M, Shin A, Carlson P, Burton D, O'Neill J, Eckert D, Zinsmeister AR. Association of melanocortin 4 receptor gene variation with satiation and gastric emptying in overweight and obese adults. GENES AND NUTRITION 2014; 9:384. [PMID: 24458996 DOI: 10.1007/s12263-014-0384-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 01/11/2014] [Indexed: 12/14/2022]
Abstract
Melanocortin 4 receptor (MC4R) has a major role in energy homeostasis. The rs17782313 polymorphism, mapped 188 kb downstream from MC4R, has been associated with satiety, higher body mass index (BMI) and total calorie intake in adults. To assess the association of rs17782313 with gastric functions, satiation, or satiety, we studied 178 predominantly Caucasian overweight and obese people: 120 females, 58 males; mean BMI 33.4 ± 5.3 kg/m(2) (SD); age 37.7 ± 11.2 years. Quantitative traits assessed were gastric emptying (GE) of solids and liquids; fasting and postprandial gastric volume; satiation by maximum tolerated volume and 4 symptoms by 100-mm visual analog scales (VAS); and satiety by ad libitum buffet meal. Associations of genotype and quantitative traits were assessed by analysis of covariance (using gender and BMI as covariates), based on a dominant [TC (n = 72) - CC (n = 12) vs. TT (n = 94)] genetic model. rs17782313(C) was associated with postprandial satiation symptoms (median Δ total VAS 26.5 mm, p = 0.036), reduced proportion of solid GE at 2 h (median Δ 6.7 %, p = 0.008) and 4 h (median Δ 3.2 %, p = 0.006), and longer t ½ (median Δ 6 min, p = 0.034). Associations of rs17782313 with obesity may be explained by reduced satiation and GE. The role of MC4R mechanisms in satiation and gastric function deserves further study.
Collapse
Affiliation(s)
- Andres Acosta
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Charlton 8-110, 200 First St. S.W., Rochester, MN, 55905, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Mimee A, Kuksis M, Ferguson AV. α-MSH exerts direct postsynaptic excitatory effects on NTS neurons and enhances GABAergic signaling in the NTS. Neuroscience 2013; 262:70-82. [PMID: 24370637 DOI: 10.1016/j.neuroscience.2013.12.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 12/06/2013] [Accepted: 12/18/2013] [Indexed: 11/27/2022]
Abstract
The central melanocortin system plays an essential role in the regulation of energy balance. While anorexigenic effects of α-melanocyte-stimulating hormone (α-MSH) acting in the nucleus of the solitary tract (NTS), a critical medullary autonomic control center, have been established, the cellular events underlying these effects are less well characterized. In this study, we used whole-cell patch-clamp electrophysiology to examine firstly whether α-MSH exerts direct postsynaptic effects on the membrane potential of rat NTS neurons in slice preparation, and secondly whether α-MSH influences GABAergic signaling in the NTS. In normal artificial cerebrospinal fluid, perfusion of α-MSH (500 nM) resulted in a depolarization in 39% of cells (n=16, mean 6.14±0.54 mV), and a hyperpolarization in 22% of cells (n=9, -6.79±1.02 mV). Studies using tetrodotoxin to block neuronal communication revealed α-MSH exerts direct depolarizing effects on some NTS neurons, and indirect inhibitory effects on others. A third subset of neurons is simultaneously directly depolarized and indirectly hyperpolarized by α-MSH, resulting in a net lack of effect on membrane potential. The inhibitory inputs influenced by α-MSH were identified as GABAergic, as α-MSH increased the frequency, but not amplitude, of inhibitory postsynaptic currents (IPSCs) in 50% of NTS neurons. α-MSH had no effect on the frequency or amplitude of miniature IPSCs. Furthermore, pharmacological blockade of GABAA and GABAB receptors, and physical removal of all synaptic inputs via cellular dissociation, abolished hyperpolarizations induced by α-MSH. We conclude α-MSH exerts direct, postsynaptic excitatory effects on a subset of NTS neurons. By exciting GABAergic NTS neurons and presynaptically enhancing GABAergic signaling, α-MSH also indirectly inhibits other NTS cells. These findings provide critical insight into the cellular events underlying medullary melanocortin anorexigenic effects, and expand the understanding of the circuitries involved in central melanocortin signaling.
Collapse
Affiliation(s)
- A Mimee
- Queen's University, Department of Biomedical and Molecular Sciences, Botterell Hall Room 435, 18 Stuart Street, Kingston, Ontario K7L 3N6, Canada
| | - M Kuksis
- Queen's University, Department of Biomedical and Molecular Sciences, Botterell Hall Room 435, 18 Stuart Street, Kingston, Ontario K7L 3N6, Canada
| | - A V Ferguson
- Queen's University, Department of Biomedical and Molecular Sciences, Botterell Hall Room 435, 18 Stuart Street, Kingston, Ontario K7L 3N6, Canada.
| |
Collapse
|
37
|
Abstract
Body weight is determined by a balance between food intake and energy expenditure. Multiple neural circuits in the brain have evolved to process information about food, food-related cues and food consumption to control feeding behavior. Numerous gastrointestinal endocrine cells produce and secrete satiety hormones in response to food consumption and digestion. These hormones suppress hunger and promote satiation and satiety mainly through hindbrain circuits, thus governing meal-by-meal eating behavior. In contrast, the hypothalamus integrates adiposity signals to regulate long-term energy balance and body weight. Distinct hypothalamic areas and various orexigenic and anorexigenic neurons have been identified to homeostatically regulate food intake. The hypothalamic circuits regulate food intake in part by modulating the sensitivity of the hindbrain to short-term satiety hormones. The hedonic and incentive properties of foods and food-related cues are processed by the corticolimbic reward circuits. The mesolimbic dopamine system encodes subjective "liking" and "wanting" of palatable foods, which is subjected to modulation by the hindbrain and the hypothalamic homeostatic circuits and by satiety and adiposity hormones. Satiety and adiposity hormones also promote energy expenditure by stimulating brown adipose tissue (BAT) activity. They stimulate BAT thermogenesis mainly by increasing the sympathetic outflow to BAT. Many defects in satiety and/or adiposity hormone signaling and in the hindbrain and the hypothalamic circuits have been described and are believed to contribute to the pathogenesis of energy imbalance and obesity.
Collapse
Affiliation(s)
- Liangyou Rui
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48109-0622, USA,
| |
Collapse
|
38
|
Tsumori T, Oka T, Yokota S, Niu JG, Yasui Y. Intrapancreatic ganglia neurons receive projection fibers from melanocortin-4 receptor-expressing neurons in the dorsal motor nucleus of the vagus nerve of the mouse. Brain Res 2013; 1537:132-42. [PMID: 24028856 DOI: 10.1016/j.brainres.2013.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 08/26/2013] [Accepted: 09/04/2013] [Indexed: 11/19/2022]
Abstract
Melanocortin-4 receptor (MC4R)-expressing neurons are widely distributed in the central nervous system and play a crucial role in a variety of physiological functions including energy and glucose/insulin homeostasis. However, their neural pathways remain to be elucidated. In the present study, we examined a possible pathway from MC4R-expressing neurons in the dorsal motor nucleus of the vagus nerve (DMV) to the intrapancreatic ganglia using transgenic mice that express green fluorescent protein (GFP) under the control of the MC4R-promoter. Using immunofluorescence labeling, we demonstrated that GFP-immunoreactive (ir) nerve fibers were distributed in the intrapancreatic ganglia closely associated with the islets as well as among the acini. These GFP-ir fibers with bouton-like varicosities were frequently observed to surround ganglion cells immunoreactive for vasoactive intestinal polypeptide, a marker for postganglionic parasympathetic neurons. Using the pre-embedding immunoperoxidase method, we clearly showed that GFP-ir terminals formed synapses predominantly with dendrites and additionally with somata of the ganglion cells. Moreover, bilateral subdiaphragmatic vagotomy caused a marked loss of GFP immunoreactivity in the pancreas. Using a combination of retrograde tracing and immunohistochemistry, we finally demonstrated that nearly half of the pancreas-projecting DMV neurons were immunoreactive for GFP. These results suggest that MC4R-expressing DMV neurons may participate in the regulation of glucose/insulin homeostasis through their projections to the intrapancreatic ganglia.
Collapse
Affiliation(s)
- Toshiko Tsumori
- Department of Anatomy and Morphological Neuroscience, Shimane University School of Medicine, Izumo 693-8501, Japan
| | | | | | | | | |
Collapse
|
39
|
Richardson J, Cruz MT, Majumdar U, Lewin A, Kingsbury KA, Dezfuli G, Vicini S, Verbalis JG, Dretchen KL, Gillis RA, Sahibzada N. Melanocortin signaling in the brainstem influences vagal outflow to the stomach. J Neurosci 2013; 33:13286-99. [PMID: 23946387 PMCID: PMC3742919 DOI: 10.1523/jneurosci.0780-13.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 06/11/2013] [Accepted: 06/13/2013] [Indexed: 01/23/2023] Open
Abstract
Activation of melanocortin 4 receptors (MC4-Rs) in brain nuclei associated with food intake profoundly influences consummatory behavior. Of these nuclei, the dorsal motor vagal nucleus (DMV), which has a dense concentration of MC4-Rs, is an important regulator of gastric tone and motility. Hence, the present study sought to examine the role of MC4-Rs in this nucleus on these activities. Using an in vivo approach, MC4-R agonists, melanotan-II (MT-II) or α-melanocyte stimulating hormone (α-MSH), were unilaterally microinjected into the DMV of rats, and their effects were noted on gastric activity. MT-II decreased phasic contractions, whereas α-MSH increased their amplitude. Both effects were blocked by the MC4-R antagonist SHU9119 or by ipsilateral vagotomy. Microinjection of the agonists (MT-II and α-MSH) into the overlying nucleus of the solitary tract (NTS), an important component of "vago-vagal" gastric circuitry, decreased phasic contractions. In addition, α-MSH reduced gastric tone and mean arterial blood pressure. To study the underlying mechanisms of the effect of MC4-R stimulation on gastric activity, electrophysiological recordings were made from labeled DMV antrum neurons in rat pups and MC4-R(-/-) mice. Bath application of MT-II or α-MSH significantly reduced spontaneous action potentials (but not in MC4-R(-/-) mice). However, in low-calcium ACSF, MT-II decreased neuronal firing, whereas α-MSH increased it. These effects mirror those of our in vivo DMV studies. Altogether, our novel findings show that activation of MC4-Rs in the brainstem, particularly in the medial NTS by the endogenous peptide α-MSH, modulates gastric activity, which may have physiological relevance for food intake and gastric function.
Collapse
Affiliation(s)
| | - Maureen T. Cruz
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
| | | | | | | | - Ghazaul Dezfuli
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
| | | | | | | | | | | |
Collapse
|
40
|
Sohn JW, Elmquist JK, Williams KW. Neuronal circuits that regulate feeding behavior and metabolism. Trends Neurosci 2013; 36:504-12. [PMID: 23790727 DOI: 10.1016/j.tins.2013.05.003] [Citation(s) in RCA: 187] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 05/06/2013] [Accepted: 05/20/2013] [Indexed: 12/24/2022]
Abstract
Neurons within the central nervous system receive humoral and central (neurotransmitter or neuropeptide) signals that ultimately regulate ingestive behavior and metabolism. Recent advances in mouse genetics combined with neuroanatomical and electrophysiological techniques have contributed to a better understanding of these central mechanisms. This review integrates recently defined cellular mechanisms and neural circuits relevant to the regulation of feeding behavior, energy expenditure, and glucose homeostasis by metabolic signals.
Collapse
Affiliation(s)
- Jong-Woo Sohn
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | | | | |
Collapse
|
41
|
Double deletion of melanocortin 4 receptors and SAPAP3 corrects compulsive behavior and obesity in mice. Proc Natl Acad Sci U S A 2013; 110:10759-64. [PMID: 23754400 DOI: 10.1073/pnas.1308195110] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Compulsive behavior is a debilitating clinical feature of many forms of neuropsychiatric disease, including Tourette syndrome, obsessive-compulsive spectrum disorders, eating disorders, and autism. Although several studies link striatal dysfunction to compulsivity, the pathophysiology remains poorly understood. Here, we show that both constitutive and induced genetic deletion of the gene encoding the melanocortin 4 receptor (MC4R), as well as pharmacologic inhibition of MC4R signaling, normalize compulsive grooming and striatal electrophysiologic impairments in synapse-associated protein 90/postsynaptic density protein 95-associated protein 3 (SAPAP3)-null mice, a model of human obsessive-compulsive disorder. Unexpectedly, genetic deletion of SAPAP3 restores normal weight and metabolic features of MC4R-null mice, a model of human obesity. Our findings offer insights into the pathophysiology and treatment of both compulsive behavior and eating disorders.
Collapse
|
42
|
Gautron L, Lee CE, Lee S, Elmquist JK. Melanocortin-4 receptor expression in different classes of spinal and vagal primary afferent neurons in the mouse. J Comp Neurol 2013; 520:3933-48. [PMID: 22592759 DOI: 10.1002/cne.23137] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Melanocortin-4 receptor (MC4R) ligands are known to modulate nociception, but the site of action of MC4R signaling on nociception remains to be elucidated. The current study investigated MC4R expression in dorsal root ganglia (DRG) of the MC4R-GFP reporter mouse. Because MC4R is known to be expressed in vagal afferent neurons in the nodose ganglion (NG), we also systematically compared MC4R-expressing vagal and spinal afferent neurons. Abundant green fluorescent protein (GFP) immunoreactivity was found in about 45% of DRG neuronal profiles (at the mid-thoracic level), the majority being small-sized profiles. Immunohistochemistry combined with in situ hybridization confirmed that GFP was genuinely produced in MC4R-expressing neurons in the DRG. While a large number of GFP profiles in the DRG coexpressed Nav1.8 mRNA (84%) and bound isolectin B4 (72%), relatively few GFP profiles were positive for NF200 (16%) or CGRP (13%), suggesting preferential MC4R expression in C-fiber nonpeptidergic neurons. By contrast, GFP in the NG frequently colocalized with Nav1.8 mRNA (64%) and NF200 (29%), but only to a moderate extent with isolectin B4 (16%). Lastly, very few GFP profiles in the NG expressed CGRP (5%) or CART (4%). Together, our findings demonstrate variegated MC4R expression in different classes of vagal and spinal primary afferent neurons, and underscore the role of the melanocortin system in modulating nociceptive and nonnociceptive peripheral sensory modalities.
Collapse
Affiliation(s)
- Laurent Gautron
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9077, USA.
| | | | | | | |
Collapse
|
43
|
Cui H, Sohn JW, Gautron L, Funahashi H, Williams KW, Elmquist JK, Lutter M. Neuroanatomy of melanocortin-4 receptor pathway in the lateral hypothalamic area. J Comp Neurol 2013; 520:4168-83. [PMID: 22605619 DOI: 10.1002/cne.23145] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The central melanocortin system regulates body energy homeostasis including the melanocortin-4 receptor (MC4R). The lateral hypothalamic area (LHA) receives dense melanocortinergic inputs from the arcuate nucleus of the hypothalamus and regulates multiple processes including food intake, reward behaviors, and autonomic function. By using a mouse line in which green fluorescent protein (GFP) is expressed under control of the MC4R gene promoter, we systemically investigated MC4R signaling in the LHA by combining double immunohistochemistry, electrophysiology, and retrograde tracing techniques. We found that LHA MC4R-GFP neurons coexpress neurotensin as well as the leptin receptor but do not coexpress other peptide neurotransmitters found in the LHA including orexin, melanin-concentrating hormone, and nesfatin-1. Furthermore, electrophysiological recording demonstrated that leptin, but not the MC4R agonist melanotan II, hyperpolarizes the majority of LHA MC4R-GFP neurons in an ATP- sensitive potassium channel-dependent manner. Retrograde tracing revealed that LHA MC4R-GFP neurons do not project to the ventral tegmental area, dorsal raphe nucleus, nucleus accumbens, and spinal cord, and only limited number of neurons project to the nucleus of the solitary tract and parabrachial nucleus. Our findings provide new insights into MC4R signaling in the LHA and its potential implications in homeostatic regulation of body energy balance.
Collapse
Affiliation(s)
- Huxing Cui
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-9127, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Sohn JW, Harris LE, Berglund ED, Liu T, Vong L, Lowell BB, Balthasar N, Williams KW, Elmquist JK. Melanocortin 4 receptors reciprocally regulate sympathetic and parasympathetic preganglionic neurons. Cell 2013; 152:612-9. [PMID: 23374353 DOI: 10.1016/j.cell.2012.12.022] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 11/20/2012] [Accepted: 12/18/2012] [Indexed: 10/27/2022]
Abstract
Melanocortin 4 receptors (MC4Rs) in the central nervous system are key regulators of energy and glucose homeostasis. Notably, obese patients with MC4R mutations are hyperinsulinemic and resistant to obesity-induced hypertension. Although these effects are probably dependent upon the activity of the autonomic nervous system, the cellular effects of MC4Rs on parasympathetic and sympathetic neurons remain undefined. Here, we show that MC4R agonists inhibit parasympathetic preganglionic neurons in the brainstem. In contrast, MC4R agonists activate sympathetic preganglionic neurons in the spinal cord. Deletion of MC4Rs in cholinergic neurons resulted in elevated levels of insulin. Furthermore, re-expression of MC4Rs specifically in cholinergic neurons (including sympathetic preganglionic neurons) restores obesity-associated hypertension in MC4R null mice. These findings provide a cellular correlate of the autonomic side effects associated with MC4R agonists and demonstrate a role for MC4Rs expressed in cholinergic neurons in the regulation of insulin levels and in the development of obesity-induced hypertension.
Collapse
Affiliation(s)
- Jong-Woo Sohn
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ross AP, Ben-Zacharia A, Harris C, Smrtka J. Multiple sclerosis, relapses, and the mechanism of action of adrenocorticotropic hormone. Front Neurol 2013; 4:21. [PMID: 23482896 PMCID: PMC3591751 DOI: 10.3389/fneur.2013.00021] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 02/13/2013] [Indexed: 12/27/2022] Open
Abstract
Relapses in multiple sclerosis (MS) are disruptive and frequently disabling for patients, and their treatment is often a challenge to clinicians. Despite progress in the understanding of the pathophysiology of MS and development of new treatments for long-term management of MS, options for treating relapses have not changed substantially over the past few decades. Corticosteroids, a component of the hypothalamic-pituitary-adrenal axis that modulate immune responses and reduce inflammation, are currently the mainstay of relapse treatment. Adrenocorticotropic hormone (ACTH) gel is another treatment option. Although it has long been assumed that the efficacy of ACTH in treating relapses depends on the peptide’s ability to increase endogenous corticosteroid production, evidence from research on the melanocortin system suggests that steroidogenesis may only partly account for ACTH influences. Indeed, the melanocortin peptides [ACTH and α-, β-, γ-melanocyte-stimulating hormones (MSH)] and their receptors (Melanocortin receptors, MCRs) exert multiple actions, including modulation of inflammatory and immune mediator production. MCRs are widely distributed within the central nervous system and in peripheral tissues including immune cells (e.g., macrophages). This suggests that the mechanism of action of ACTH includes not only steroid-mediated indirect effects, but also direct anti-inflammatory and immune-modulating actions via the melanocortin system. An increased understanding of the role of the melanocortin system, particularly ACTH, in the immune and inflammatory processes underlying relapses may help to improve relapse management.
Collapse
Affiliation(s)
- Amy Perrin Ross
- Department of Neurosciences, Loyola University Chicago Chicago, IL, USA
| | | | | | | |
Collapse
|
46
|
Zechner JF, Mirshahi UL, Satapati S, Berglund ED, Rossi J, Scott MM, Still CD, Gerhard GS, Burgess SC, Mirshahi T, Aguirre V. Weight-independent effects of roux-en-Y gastric bypass on glucose homeostasis via melanocortin-4 receptors in mice and humans. Gastroenterology 2013; 144:580-590.e7. [PMID: 23159449 PMCID: PMC3835150 DOI: 10.1053/j.gastro.2012.11.022] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 11/08/2012] [Accepted: 11/11/2012] [Indexed: 01/06/2023]
Abstract
BACKGROUND & AIMS Roux-en-Y gastric bypass (RYGB) improves glucose homeostasis independently of changes in body weight by unknown mechanisms. Melanocortin-4 receptors (MC4R) have weight-independent effects on glucose homeostasis, via autonomic neurons, and also might contribute to weight loss after RYGB. We investigated whether MC4Rs mediate effects of RYGB, such as its weight-independent effects on glucose homeostasis, in mice and humans. METHODS We studied C57BL/6 mice with diet-induced obesity, MC4R-deficient mice, and mice that re-express MC4R specifically in autonomic neurons after RYGB or sham surgeries. We also sequenced the MC4R locus in patients undergoing RYGB to investigate diabetes resolution in carriers of rare MC4R variants. RESULTS MC4Rs in autonomic brainstem neurons (including the parasympathetic dorsal motor vagus) mediated improved glucose homeostasis independent of changes in body weight. In contrast, MC4Rs in cholinergic preganglionic motor neurons (sympathetic and parasympathetic) mediated RYGB-induced increased energy expenditure and weight loss. Increased energy expenditure after RYGB is the predominant mechanism of weight loss and confers resistance to weight gain from a high-fat diet, the effects of which are MC4R-dependent. MC4R-dependent effects of RYGB still occurred in mice with Mc4r haplosufficiency, and early stage diabetes resolved at a similar rate in patients with rare variants of MC4R and noncarriers. However, carriers of MC4R (I251L), a rare variant associated with increased weight loss after RYGB and increased basal activity in vitro, were more likely to have early and weight-independent resolution of diabetes than noncarriers, indicating a role for MC4Rs in the effects of RYGB. CONCLUSIONS MC4Rs in autonomic neurons mediate beneficial effects of RYGB, including weight-independent improved glucose homeostasis, in mice and humans.
Collapse
Affiliation(s)
- Juliet F. Zechner
- UT Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Hypothalamic Research, Department of Medicine
| | - Uyenlinh L. Mirshahi
- Weis Center for Research and Geisinger Obesity Research Institute, Geisinger Clinic, Danville, PA 17822, USA
| | - Santhosh Satapati
- UT Southwestern Medical Center, Dallas, TX 75390, USA
- Advanced Imaging Center, Department of Pharmacology
| | - Eric D. Berglund
- UT Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Hypothalamic Research, Department of Medicine
| | - Jari Rossi
- Institute of Biomedicine, Anatomy, University of Helsinki, Helsinki, Finland
| | - Michael M. Scott
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22903, USA
| | - Christopher D. Still
- Weis Center for Research and Geisinger Obesity Research Institute, Geisinger Clinic, Danville, PA 17822, USA
| | - Glenn S. Gerhard
- Weis Center for Research and Geisinger Obesity Research Institute, Geisinger Clinic, Danville, PA 17822, USA
| | - Shawn C. Burgess
- UT Southwestern Medical Center, Dallas, TX 75390, USA
- Advanced Imaging Center, Department of Pharmacology
| | - Tooraj Mirshahi
- Weis Center for Research and Geisinger Obesity Research Institute, Geisinger Clinic, Danville, PA 17822, USA
| | - Vincent Aguirre
- UT Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Hypothalamic Research, Department of Medicine
| |
Collapse
|
47
|
Rinne P, Harjunpää J, Mäkelä S, Savontaus E. Genetic and pharmacological mouse models of chronic melanocortin activation show enhanced baroreflex control of heart rate. ACTA ACUST UNITED AC 2013; 182:19-27. [DOI: 10.1016/j.regpep.2012.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 09/05/2012] [Accepted: 12/17/2012] [Indexed: 10/27/2022]
|
48
|
Andresen MC, Fawley JA, Hofmann ME. Peptide and lipid modulation of glutamatergic afferent synaptic transmission in the solitary tract nucleus. Front Neurosci 2013; 6:191. [PMID: 23335875 PMCID: PMC3541483 DOI: 10.3389/fnins.2012.00191] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 12/17/2012] [Indexed: 12/21/2022] Open
Abstract
The brainstem nucleus of the solitary tract (NTS) holds the first central neurons in major homeostatic reflex pathways. These homeostatic reflexes regulate and coordinate multiple organ systems from gastrointestinal to cardiopulmonary functions. The core of many of these pathways arise from cranial visceral afferent neurons that enter the brain as the solitary tract (ST) with more than two-thirds arising from the gastrointestinal system. About one quarter of ST afferents have myelinated axons but the majority are classed as unmyelinated C-fibers. All ST afferents release the fast neurotransmitter glutamate with remarkably similar, high-probability release characteristics. Second order NTS neurons receive surprisingly limited primary afferent information with one or two individual inputs converging on single second order NTS neurons. A- and C-fiber afferents never mix at NTS second order neurons. Many transmitters modify the basic glutamatergic excitatory postsynaptic current often by reducing glutamate release or interrupting terminal depolarization. Thus, a distinguishing feature of ST transmission is presynaptic expression of G-protein coupled receptors for peptides common to peripheral or forebrain (e.g., hypothalamus) neuron sources. Presynaptic receptors for angiotensin (AT1), vasopressin (V1a), oxytocin, opioid (MOR), ghrelin (GHSR1), and cholecystokinin differentially control glutamate release on particular subsets of neurons with most other ST afferents unaffected. Lastly, lipid-like signals are transduced by two key ST presynaptic receptors, the transient receptor potential vanilloid type 1 and the cannabinoid receptor that oppositely control glutamate release. Increasing evidence suggests that peripheral nervous signaling mechanisms are repurposed at central terminals to control excitation and are major sites of signal integration of peripheral and central inputs particularly from the hypothalamus.
Collapse
Affiliation(s)
- Michael C Andresen
- Department of Physiology and Pharmacology, Oregon Health and Science University Portland, OR, USA
| | | | | |
Collapse
|
49
|
Begriche K, Girardet C, McDonald P, Butler AA. Melanocortin-3 receptors and metabolic homeostasis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 114:109-46. [PMID: 23317784 DOI: 10.1016/b978-0-12-386933-3.00004-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Attenuated activity of the central nervous melanocortin system causes obesity and insulin resistance. Obese rodents treated with melanocortins exhibit improvements in obesity and metabolic homeostasis that are not mutually dependent, suggesting metabolic actions that are independent of weight changes. These responses are generally thought to involve G-protein-coupled receptors expressed in the brain. Melanocortin-4 receptors (MC4Rs) regulate satiety and autonomic nervous system and thyroid function. MC3Rs are expressed in hypothalamic and limbic regions involved in controlling ingestive behaviors and autonomic function. Mc3r-/- mice exhibit increased adiposity and an accelerated diet-induced obesity. While this phenotype is not dependent on hyperphagia, data on the regulation of food intake by MC3Rs are inconsistent. Recent investigations by our laboratory suggest a unique combination of behavioral and metabolic disorders in Mc3r-/- mice. MC3Rs are critical for the expression of the anticipatory response and metabolic homeostasis when food intake occurs outside the normal voluntary rhythms driven by photoperiod. Using a Cre-Lox strategy, we can now investigate MC3Rs expressed in different brain regions and organ systems in the periphery. While focusing on the functions of neural MC3Rs, early results suggest an additional layer of complexity with central and peripheral MC3Rs involved in the defense of body weight.
Collapse
Affiliation(s)
- Karima Begriche
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, Florida, USA
| | | | | | | |
Collapse
|
50
|
Granell S, Serra-Juhé C, Martos-Moreno GÁ, Díaz F, Pérez-Jurado LA, Baldini G, Argente J. A novel melanocortin-4 receptor mutation MC4R-P272L associated with severe obesity has increased propensity to be ubiquitinated in the ER in the face of correct folding. PLoS One 2012; 7:e50894. [PMID: 23251400 PMCID: PMC3520997 DOI: 10.1371/journal.pone.0050894] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 10/26/2012] [Indexed: 11/28/2022] Open
Abstract
Heterozygous mutations in the melanocortin-4 receptor (MC4R) gene represent the most frequent cause of monogenic obesity in humans. MC4R mutation analysis in a cohort of 77 children with morbid obesity identified previously unreported heterozygous mutations (P272L, N74I) in two patients inherited from their obese mothers. A rare polymorphism (I251L, allelic frequency: 1/100) reported to protect against obesity was found in another obese patient. When expressed in neuronal cells, the cell surface abundance of wild-type MC4R and of the N74I and I251L variants and the cAMP generated by these receptors in response to exposure to the agonist, α-MSH, were not different. Conversely, MC4R P272L was retained in the endoplasmic reticulum and had reduced cell surface expression and signaling (by ≈3-fold). The chemical chaperone PBA, which promotes protein folding of wild-type MC4R, had minimal effects on the distribution and signaling of the P272L variant. In contrast, incubation with UBE-41, a specific inhibitor of ubiquitin activating enzyme E1, inhibited ubiquitination of MC4R P272L and increased its cell surface expression and signaling to similar levels as wild-type MC4R. UBE41 had much less profound effects on MC4R I316S, another obesity-linked MC4R variant trapped in the ER. These data suggest that P272L is retained in the ER by a propensity to be ubiquitinated in the face of correct folding, which is only minimally shared by MC4R I316S. Thus, studies that combine clinical screening of obese patients and investigation of the functional defects of the obesity-linked MC4R variants can identify specific ways to correct these defects and are the first steps towards personalized medicine.
Collapse
Affiliation(s)
- Susana Granell
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Clara Serra-Juhé
- Unitat de Genètica, Universitat Pompeu Fabra, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Gabriel Á. Martos-Moreno
- Department of Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
| | - Francisca Díaz
- Department of Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
| | - Luis A. Pérez-Jurado
- Unitat de Genètica, Universitat Pompeu Fabra, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Giulia Baldini
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- * E-mail: (GB); (JA)
| | - Jesús Argente
- Department of Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid and Centro de Investigación Biomédica en Red de fisiopatología de la obesidad y nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- * E-mail: (GB); (JA)
| |
Collapse
|