1
|
Pirník Z, Szadvári I, Borbélyová V, Tomova A. Altered sex differences related to food intake, hedonic preference, and FosB/deltaFosB expression within central neural circuit involved in homeostatic and hedonic food intake regulation in Shank3B mouse model of autism spectrum disorder. Neurochem Int 2024:105895. [PMID: 39461669 DOI: 10.1016/j.neuint.2024.105895] [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: 05/27/2024] [Revised: 10/09/2024] [Accepted: 10/24/2024] [Indexed: 10/29/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder accompanied by narrow interests, difficulties in communication and social interaction, and repetitive behavior. In addition, ASD is frequently associated with eating and feeding problems. Although the symptoms of ASD are more likely to be observed in boys, the prevalence of eating disorders is more common in females. The ingestive behavior is regulated by the integrative system of the brain, which involves both homeostatic and hedonic neural circuits. Sex differences in the physiology of food intake depend on sex hormones regulating the expression of the ASD-associated Shank genes. Shank3 mutation leads to ASD-like traits and Shank3B -/- mice have been established as an animal model to study the neurobiology of ASD. Therefore, the long-lasting neuronal activity in the central neural circuit related to the homeostatic and hedonic regulation of food intake was evaluated in both sexes of Shank3B mice, followed by the evaluation of the food intake and preference. In the Shank3B +/+ genotype, well-preserved relationships in the tonic activity within the homeostatic neural network together with the relationships between ingestion and hedonic preference were observed in males but were reduced in females. These interrelations were partially or completely lost in the mice with the Shank3B -/- genotype. A decreased hedonic preference for the sweet taste but increased total food intake was found in the Shank3B -/- mice. In the Shank3B -/- group, there were altered sex differences related to the amount of tonic cell activity in the hedonic and homeostatic neural networks, together with altered sex differences in sweet and sweet-fat solution intake. Furthermore, the Shank3B -/- females exhibited an increased intake and preference for cheese compared to the Shank3B +/+ ones. The obtained data indicate altered functional crosstalk between the central homeostatic and hedonic neural circuits involved in the regulation of food intake in ASD.
Collapse
Affiliation(s)
- Zdenko Pirník
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia; Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia; Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Ivan Szadvári
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Veronika Borbélyová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Aleksandra Tomova
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
2
|
Neyens DM, Brenner L, Calkins R, Winzenried ET, Ritter RC, Appleyard SM. CCK-sensitive C fibers activate NTS leptin receptor-expressing neurons via NMDA receptors. Am J Physiol Regul Integr Comp Physiol 2024; 326:R383-R400. [PMID: 38105761 PMCID: PMC11381032 DOI: 10.1152/ajpregu.00238.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
The hormone leptin reduces food intake through actions in the peripheral and central nervous systems, including in the hindbrain nucleus of the solitary tract (NTS). The NTS receives viscerosensory information via vagal afferents, including information from the gastrointestinal tract, which is then relayed to other central nervous system (CNS) sites critical for control of food intake. Leptin receptors (lepRs) are expressed by a subpopulation of NTS neurons, and knockdown of these receptors increases both food intake and body weight. Recently, we demonstrated that leptin increases vagal activation of lepR-expressing neurons via increased NMDA receptor (NMDAR) currents, thereby potentiating vagally evoked firing. Furthermore, chemogenetic activation of these neurons was recently shown to inhibit food intake. However, the vagal inputs these neurons receive had not been characterized. Here we performed whole cell recordings in brain slices taken from lepRCre × floxedTdTomato mice and found that lepR neurons of the NTS are directly activated by monosynaptic inputs from C-type afferents sensitive to the transient receptor potential vanilloid type 1 (TRPV1) agonist capsaicin. CCK administered onto NTS slices stimulated spontaneous glutamate release onto lepR neurons and induced action potential firing, an effect mediated by CCKR1. Interestingly, NMDAR activation contributed to the current carried by spontaneous excitatory postsynaptic currents (EPSCs) and enhanced CCK-induced firing. Peripheral CCK also increased c-fos expression in these neurons, suggesting they are activated by CCK-sensitive vagal afferents in vivo. Our results indicate that the majority of NTS lepR neurons receive direct inputs from CCK-sensitive C vagal-type afferents, with both peripheral and central CCK capable of activating these neurons and NMDARs able to potentiate these effects.
Collapse
Affiliation(s)
- Drew M Neyens
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| | - Lynne Brenner
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| | - Rowan Calkins
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| | - Eric T Winzenried
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| | - Robert C Ritter
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| | - Suzanne M Appleyard
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| |
Collapse
|
3
|
Bodas DS, Maduskar A, Kaniganti T, Wakhloo D, Balasubramanian A, Subhedar N, Ghose A. Convergent Energy State-Dependent Antagonistic Signaling by Cocaine- and Amphetamine-Regulated Transcript (CART) and Neuropeptide Y (NPY) Modulates the Plasticity of Forebrain Neurons to Regulate Feeding in Zebrafish. J Neurosci 2023; 43:1089-1110. [PMID: 36599680 PMCID: PMC9962846 DOI: 10.1523/jneurosci.2426-21.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 11/28/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023] Open
Abstract
Dynamic reconfiguration of circuit function subserves the flexibility of innate behaviors tuned to physiological states. Internal energy stores adaptively regulate feeding-associated behaviors and integrate opposing hunger and satiety signals at the level of neural circuits. Across vertebrate lineages, the neuropeptides cocaine- and amphetamine-regulated transcript (CART) and neuropeptide Y (NPY) have potent anorexic and orexic functions, respectively, and show energy-state-dependent expression in interoceptive neurons. However, how the antagonistic activities of these peptides modulate circuit plasticity remains unclear. Using behavioral, neuroanatomical, and activity analysis in adult zebrafish of both sexes, along with pharmacological interventions, we show that CART and NPY activities converge on a population of neurons in the dorsomedial telencephalon (Dm). Although CART facilitates glutamatergic neurotransmission at the Dm, NPY dampens the response to glutamate. In energy-rich states, CART enhances NMDA receptor (NMDAR) function by protein kinase A/protein kinase C (PKA/PKC)-mediated phosphorylation of the NR1 subunit of the NMDAR complex. Conversely, starvation triggers NPY-mediated reduction in phosphorylated NR1 via calcineurin activation and inhibition of cAMP production leading to reduced responsiveness to glutamate. Our data identify convergent integration of CART and NPY inputs by the Dm neurons to generate nutritional state-dependent circuit plasticity that is correlated with the behavioral switch induced by the opposing actions of satiety and hunger signals.SIGNIFICANCE STATEMENT Internal energy needs reconfigure neuronal circuits to adaptively regulate feeding behavior. Energy-state-dependent neuropeptide release can signal energy status to feeding-associated circuits and modulate circuit function. CART and NPY are major anorexic and orexic factors, respectively, but the intracellular signaling pathways used by these peptides to alter circuit function remain uncharacterized. We show that CART and NPY-expressing neurons from energy-state interoceptive areas project to a novel telencephalic region, Dm, in adult zebrafish. CART increases the excitability of Dm neurons, whereas NPY opposes CART activity. Antagonistic signaling by CART and NPY converge onto NMDA-receptor function to modulate glutamatergic neurotransmission. Thus, opposing activities of anorexic CART and orexic NPY reconfigure circuit function to generate flexibility in feeding behavior.
Collapse
Affiliation(s)
- Devika S Bodas
- Indian Institute of Science Education and Research, Pune, Pune 411008, India
| | - Aditi Maduskar
- Indian Institute of Science Education and Research, Pune, Pune 411008, India
| | - Tarun Kaniganti
- Indian Institute of Science Education and Research, Pune, Pune 411008, India
| | - Debia Wakhloo
- Indian Institute of Science Education and Research, Pune, Pune 411008, India
| | | | - Nishikant Subhedar
- Indian Institute of Science Education and Research, Pune, Pune 411008, India
| | - Aurnab Ghose
- Indian Institute of Science Education and Research, Pune, Pune 411008, India
| |
Collapse
|
4
|
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: 59] [Impact Index Per Article: 29.5] [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
|
5
|
Glial Modulation of Energy Balance: The Dorsal Vagal Complex Is No Exception. Int J Mol Sci 2022; 23:ijms23020960. [PMID: 35055143 PMCID: PMC8779587 DOI: 10.3390/ijms23020960] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 02/04/2023] Open
Abstract
The avoidance of being overweight or obese is a daily challenge for a growing number of people. The growing proportion of people suffering from a nutritional imbalance in many parts of the world exemplifies this challenge and emphasizes the need for a better understanding of the mechanisms that regulate nutritional balance. Until recently, research on the central regulation of food intake primarily focused on neuronal signaling, with little attention paid to the role of glial cells. Over the last few decades, our understanding of glial cells has changed dramatically. These cells are increasingly regarded as important neuronal partners, contributing not just to cerebral homeostasis, but also to cerebral signaling. Our understanding of the central regulation of energy balance is part of this (r)evolution. Evidence is accumulating that glial cells play a dynamic role in the modulation of energy balance. In the present review, we summarize recent data indicating that the multifaceted glial compartment of the brainstem dorsal vagal complex (DVC) should be considered in research aimed at identifying feeding-related processes operating at this level.
Collapse
|
6
|
Targeting the T-type calcium channel Cav3.2 in GABAergic arcuate nucleus neurons to treat obesity. Mol Metab 2021; 54:101391. [PMID: 34767997 PMCID: PMC8640109 DOI: 10.1016/j.molmet.2021.101391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/20/2021] [Accepted: 11/02/2021] [Indexed: 01/16/2023] Open
Abstract
OBJECTIVE Cav3.2, a T-type low voltage-activated calcium channel widely expressed throughout the central nervous system, plays a vital role in neuronal excitability and various physiological functions. However, the effects of Cav3.2 on energy homeostasis remain unclear. Here, we examined the role of Cav3.2 expressed by hypothalamic GABAergic neurons in the regulation of food intake and body weight in mice and explored the underlying mechanisms. METHODS Male congenital Cana1h (the gene coding for Cav3.2) global knockout (Cav3.2KO) mice and their wild type (WT) littermates were first used for metabolic phenotyping studies. By using the CRISPR-Cas9 technique, Cav3.2 was selectively deleted from GABAergic neurons in the arcuate nucleus of the hypothalamus (ARH) by specifically overexpressing Cas9 protein and Cav3.2-targeting sgRNAs in ARH Vgat (VgatARH) neurons. These male mutants (Cav3.2KO-VgatARH) were used to determine whether Cav3.2 expressed by VgatARH neurons is required for the proper regulation of energy balance. Subsequently, we used an electrophysiological patch-clamp recording in ex vivo brain slices to explore the impact of Cav3.2KO on the cellular excitability of VgatARH neurons. RESULTS Male Cav3.2KO mice had significantly lower food intake than their WT littermate controls when fed with either a normal chow diet (NCD) or a high-fat diet (HFD). This hypophagia phenotype was associated with increased energy expenditure and decreased fat mass, lean mass, and total body weight. Selective deletion of Cav3.2 in VgatARH neurons resulted in similar feeding inhibition and lean phenotype without changing energy expenditure. These data provides an intrinsic mechanism to support the previous finding on ARH non-AgRP GABA neurons in regulating diet-induced obesity. Lastly, we found that naringenin extract, a predominant flavanone found in various fruits and herbs and known to act on Cav3.2, decreased the firing activity of VgatARH neurons and reduced food intake and body weight. These naringenin-induced inhibitions were fully blocked in Cav3.2KO-VgatARH mice. CONCLUSION Our results identified Cav3.2 expressed by VgatARH neurons as an essential intrinsic modulator for food intake and energy homeostasis, which is a potential therapeutic target in the treatment of obesity.
Collapse
|
7
|
Kochetova OV, Avzaletdinova DS, Shangareeva ZA, Akhmadishina LZ, Korytina GF, Victorov VV, Victorova TV. Association of Polymorphisms in Neurotransmitter Genes and the TMEM18 Gene with Eating Behavior in Obese Patients. RUSS J GENET+ 2021. [DOI: 10.1134/s1022795421050045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
8
|
Schalla MA, Taché Y, Stengel A. Neuroendocrine Peptides of the Gut and Their Role in the Regulation of Food Intake. Compr Physiol 2021; 11:1679-1730. [PMID: 33792904 DOI: 10.1002/cphy.c200007] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The regulation of food intake encompasses complex interplays between the gut and the brain. Among them, the gastrointestinal tract releases different peptides that communicate the metabolic state to specific nuclei in the hindbrain and the hypothalamus. The present overview gives emphasis on seven peptides that are produced by and secreted from specialized enteroendocrine cells along the gastrointestinal tract in relation with the nutritional status. These established modulators of feeding are ghrelin and nesfatin-1 secreted from gastric X/A-like cells, cholecystokinin (CCK) secreted from duodenal I-cells, glucagon-like peptide 1 (GLP-1), oxyntomodulin, and peptide YY (PYY) secreted from intestinal L-cells and uroguanylin (UGN) released from enterochromaffin (EC) cells. © 2021 American Physiological Society. Compr Physiol 11:1679-1730, 2021.
Collapse
Affiliation(s)
- Martha A Schalla
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Yvette Taché
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, David Geffen School of Medicine, UCLA, Los Angeles, California, USA.,VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Andreas Stengel
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
9
|
Browning KN, Carson KE. Central Neurocircuits Regulating Food Intake in Response to Gut Inputs-Preclinical Evidence. Nutrients 2021; 13:nu13030908. [PMID: 33799575 PMCID: PMC7998662 DOI: 10.3390/nu13030908] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/02/2021] [Accepted: 03/07/2021] [Indexed: 02/07/2023] Open
Abstract
The regulation of energy balance requires the complex integration of homeostatic and hedonic pathways, but sensory inputs from the gastrointestinal (GI) tract are increasingly recognized as playing critical roles. The stomach and small intestine relay sensory information to the central nervous system (CNS) via the sensory afferent vagus nerve. This vast volume of complex sensory information is received by neurons of the nucleus of the tractus solitarius (NTS) and is integrated with responses to circulating factors as well as descending inputs from the brainstem, midbrain, and forebrain nuclei involved in autonomic regulation. The integrated signal is relayed to the adjacent dorsal motor nucleus of the vagus (DMV), which supplies the motor output response via the efferent vagus nerve to regulate and modulate gastric motility, tone, secretion, and emptying, as well as intestinal motility and transit; the precise coordination of these responses is essential for the control of meal size, meal termination, and nutrient absorption. The interconnectivity of the NTS implies that many other CNS areas are capable of modulating vagal efferent output, emphasized by the many CNS disorders associated with dysregulated GI functions including feeding. This review will summarize the role of major CNS centers to gut-related inputs in the regulation of gastric function with specific reference to the regulation of food intake.
Collapse
|
10
|
Leptin Sensitizes NTS Neurons to Vagal Input by Increasing Postsynaptic NMDA Receptor Currents. J Neurosci 2020; 40:7054-7064. [PMID: 32817248 DOI: 10.1523/jneurosci.1865-19.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/05/2019] [Accepted: 07/27/2020] [Indexed: 11/21/2022] Open
Abstract
Leptin signaling within the nucleus of the solitary tract (NTS) contributes to the control of food intake, and injections of leptin into the NTS reduce meal size and increase the efficacy of vagus-mediated satiation signals. Leptin receptors (LepRs) are expressed by vagal afferents as well as by a population of NTS neurons. However, the electrophysiological properties of LepR-expressing NTS neurons have not been well characterized, and it is unclear how leptin might act on these neurons to reduce food intake. To address this question, we recorded from LepR-expressing neurons in horizontal brain slices containing the NTS from male and female LepR-Cre X Rosa-tdTomato mice. We found that the vast majority of NTS LepR neurons received monosynaptic innervation from vagal afferent fibers and LepR neurons exhibited large synaptic NMDA receptor (NMDAR)-mediated currents compared with non-LepR neurons. During high-frequency stimulation of vagal afferents, leptin increased the size of NMDAR-mediated currents, but not AMPAR-mediated currents. Leptin also increased the size of evoked EPSPs and the ability of low-intensity solitary tract stimulation to evoke action potentials in LepR neurons. These effects of leptin were blocked by bath applying a competitive NMDAR antagonist (DCPP-ene) or by an NMDAR channel blocker applied through the recording pipette (MK-801). Last, feeding studies using male rats demonstrate that intra-NTS injections of DCPP-ene attenuate reduction of overnight food intake following intra-NTS leptin injection. Our results suggest that leptin acts in the NTS to reduce food intake by increasing NMDAR-mediated currents, thus enhancing NTS sensitivity to vagal inputs.SIGNIFICANCE STATEMENT Leptin is a hormone that critically impacts food intake and energy homeostasis. The nucleus of the solitary tract (NTS) is activated by vagal afferents from the gastrointestinal tract, which promotes termination of a meal. Injection of leptin into the NTS inhibits food intake, while knockdown of leptin receptors (LepRs) in NTS neurons increases food intake. However, little was known about how leptin acts in the NTS neurons to inhibit food intake. We found that leptin increases the sensitivity of LepR-expressing neurons to vagal inputs by increasing NMDA receptor-mediated synaptic currents and that NTS NMDAR activation contributes to leptin-induced reduction of food intake. These findings suggest a novel mechanism by which leptin, acting in the NTS, could potentiate gastrointestinal satiation signals.
Collapse
|
11
|
Wald HS, Grill HJ. Individual Differences in Behavioral Responses to Palatable Food or to Cholecystokinin Predict Subsequent Diet-Induced Obesity. Obesity (Silver Spring) 2019; 27:943-949. [PMID: 30998842 PMCID: PMC6533155 DOI: 10.1002/oby.22459] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/13/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study investigated whether individual differences in behavioral responses to palatable food and to the satiation signal cholecystokinin (CCK) in outbred chow-maintained Sprague-Dawley rats enabled prediction of individual differences in weight gained after subsequent high-fat/high-sugar diet (HFHSD) maintenance. METHODS Meal size, meal number, and early dark cycle intake during initial HFHSD exposure were measured, as were early dark cycle sucrose solution and chow intake, chow meal size and meal number, the intake-suppressive effects of 0.5-µg/kg CCK injection, and CCK-induced c-Fos activation in the nucleus tractus solitarius. Subsequently, rats were maintained on an HFHSD for 5 weeks, and weight gain was determined. RESULTS Rats that took larger and less frequent meals on the first day of HFHSD exposure, whose early dark cycle intake (HFHSD and sucrose) was larger during initial HFHSD exposure, gained more weight after HFHSD maintenance. Rats with lesser sucrose intake suppression in response to CCK gained more weight after HFHSD maintenance and displayed reduced CCK-induced c-Fos activation in the nucleus tractus solitarius. CONCLUSIONS Together, these data identify individual differences in behavioral responses to palatable food and to CCK as novel predictors of diet-induced obesity.
Collapse
Affiliation(s)
- Hallie S Wald
- Graduate Group in Psychology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Harvey J Grill
- Graduate Group in Psychology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Graduate Group in Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
Huston NJ, Brenner LA, Taylor ZC, Ritter RC. NPY2 receptor activation in the dorsal vagal complex increases food intake and attenuates CCK-induced satiation in male rats. Am J Physiol Regul Integr Comp Physiol 2019; 316:R406-R416. [PMID: 30726118 DOI: 10.1152/ajpregu.00011.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Neuropeptide Y (NPY), peptide YY (PYY), and their cognate receptors (YR) are expressed by subpopulations of central and peripheral nervous system neurons. Intracerebroventricular injections of NPY or PYY increase food intake, and intrahypothalamic NPY1 or NPY5 receptor agonist injections also increase food intake. In contrast, injection of PYY in the periphery reduces food intake, apparently by activating peripheral Y2R. The dorsal vagal complex (DVC) of the hindbrain is the site where vagal afferents relay gut satiation signals to the brain. While contributions of the DVC are increasingly investigated, a role for DVC YR in control of food intake has not been examined systematically. We used in situ hybridization to confirm expression of Y1R and Y2R, but not Y5R, in the DVC and vagal afferent neurons. We found that nanoinjections of a Y2R agonist, PYY-(3-36), into the DVC significantly increased food intake over a 4-h period in satiated male rats. PYY-(3-36)-evoked food intake was prevented by injection of a selective Y2R antagonist. Injection of a Y1R/Y5R-preferring agonist into the DVC failed to increase food intake at doses reported to increase food intake following hypothalamic injection. Finally, injection of PYY-(3-36) into the DVC prevented reduction of 30-min food intake following intraperitoneal injection of cholecystokinin (CCK). Our results indicate that activation of DVC Y2R, unlike hypothalamic or peripheral Y2R, increases food intake. Furthermore, in the context of available electrophysiological observations, our results are consistent with the hypothesis that DVC Y2R control food intake by dampening vagally mediated satiation signals in the DVC.
Collapse
Affiliation(s)
- Nathaneal J Huston
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington
| | - Lynne A Brenner
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington
| | - Zachary C Taylor
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington
| | - Robert C Ritter
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington
| |
Collapse
|
13
|
Deng SN, Yan YH, Zhu TL, Ma BK, Fan HR, Liu YM, Li WG, Li F. Long-Term NMDAR Antagonism Correlates Weight Loss With Less Eating. Front Psychiatry 2019; 10:15. [PMID: 30800078 PMCID: PMC6375831 DOI: 10.3389/fpsyt.2019.00015] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/10/2019] [Indexed: 12/12/2022] Open
Abstract
Memantine hydrochloride is an uncompetitive N-methyl-D-aspartate (NMDA) antagonist for treatment of moderate-to-severe Alzheimer's disease. Several studies have shown that memantine can significantly correct the binge-like eating behavior in human and animal models. People with overeating behavior are more likely to be obese. Therefore, we suppose that memantine would be a good candidate for the treatment of obesity. In this study, memantine was shown to increase weight loss in obese mice induced by high fat diet. Memantine was shown to decrease food intake without inducing abdominal discomfort and anxiety, suggesting that this compound would be a good candidate drug for obesity control.
Collapse
Affiliation(s)
- Shi-Ning Deng
- Developmental and Behavioral Pediatric Department and Child Primary Care Department, Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Shanghai Institute for Pediatric Research, Xinhua Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Hua Yan
- Key Laboratory of Brain Functional Genomics, Ministry of Education, East China Normal University, Shanghai, China
| | - Tai-Lin Zhu
- Key Laboratory of Brain Functional Genomics, Ministry of Education, East China Normal University, Shanghai, China
| | - Bing-Ke Ma
- Key Laboratory of Brain Functional Genomics, Ministry of Education, East China Normal University, Shanghai, China
| | - Hui-Ran Fan
- Developmental and Behavioral Pediatric Department and Child Primary Care Department, Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Shanghai Institute for Pediatric Research, Xinhua Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan-Mei Liu
- Developmental and Behavioral Pediatric Department and Child Primary Care Department, Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Shanghai Institute for Pediatric Research, Xinhua Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Guang Li
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Li
- Developmental and Behavioral Pediatric Department and Child Primary Care Department, Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Shanghai Institute for Pediatric Research, Xinhua Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Dafalla AI, Mhalhal TR, Hiscocks K, Heath J, Sayegh AI. Non-sulfated cholecystokinin-8 increases enteric and hindbrain Fos-like immunoreactivity in male Sprague Dawley rats. Brain Res 2018; 1708:200-206. [PMID: 30571983 DOI: 10.1016/j.brainres.2018.12.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/13/2022]
Abstract
Recently, we reported that non-sulfated cholecystokinin-8 (NS CCK-8) reduces food intake by cholecystokinin-B receptors (CCK-BR). To examine a possible site of action for this peptide, and based on the fact that both NS CCK-8 and CCK-BR are found centrally and peripherally, in the current study we hypothesized that NS CCK-8 increases Fos-like immunoreactivity (Fos-LI, a neuronal activation marker) in the dorsal vagal complex (DVC) of the hindbrain and the myenteric and submucosal plexuses of the small intestine. We found that intraperitoneal NS CCK-8 (0.5 nmol/kg) increases Fos-LI in the DVC, the myenteric and the submucosal plexuses of the duodenum and the myenteric plexus of the jejunum. The findings suggest, but does not prove, a potential role for the DVC and the enteric neurons in the feeding responses evoked by NS CCK-8.
Collapse
Affiliation(s)
- Amged I Dafalla
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee AL36088, United States
| | - Thaer R Mhalhal
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee AL36088, United States
| | - Kenneth Hiscocks
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee AL36088, United States
| | - John Heath
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee AL36088, United States
| | - Ayman I Sayegh
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee AL36088, United States.
| |
Collapse
|
15
|
Minaya DM, Larson RW, Podlasz P, Czaja K. Glutamate-dependent regulation of food intake is altered with age through changes in NMDA receptor phenotypes on vagal afferent neurons. Physiol Behav 2018; 189:26-31. [PMID: 29476874 DOI: 10.1016/j.physbeh.2018.02.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/19/2018] [Accepted: 02/20/2018] [Indexed: 11/25/2022]
Abstract
Compared to younger individuals, older human subjects have significantly lower food intakes and an increased satiety response. N-methyl-d-aspartate (NMDA) receptors expressed by vagal afferent neurons originating from nodose ganglia (NG) are involved in modulating the satiety response. The present study investigated how NMDA receptor subunit phenotypes in NG neurons change with age and how these age-related alterations in food intake are modulated by presynaptic NMDA receptors in the NG of male Sprague Dawley rats (six week-old and sixty week-old). Food intake was measured at 30-, 60-, and 120-min following intraperitoneal administration of cholecystokinin (CCK) or the non-competitive NMDA receptor antagonist MK-801. Immunofluorescence was used to determine NMDA receptor subunit expression (NR1, NR2B, NR2C, and NR2D) in the NG. The results showed that, CCK reduced food intake at 30-, 60-, and 120-min post injection in both young and the middle-age animals, with no statistical difference between the groups at 30- and 60-min. In contrast, MK-801 produced an increase in food intake that was significantly higher in middle-age rats compared to young animals at all time points studied. NR1 subunit was expressed by almost all NG neurons in both age groups. In young rats, NR2B, NR2C, and NR2D subunits were expressed in 56.1%, 49.3%, and 13.9% of NG neurons, respectively. In contrast, only 30.3% of the neuronal population in middle-aged rats expressed NR2B subunit immunoreactivity, NR2C was present in 34.1%, and only 10.6% of total neurons expressed the NR2D subunit. In conclusion, glutamate-dependent regulation of food intake is altered with age and one of the potential mechanisms through which this age-related changes in intake occur is changes in NMDA receptor phenotypes on vagal afferent neurons located in NG.
Collapse
Affiliation(s)
- Dulce M Minaya
- Department of Veterinary Biosciences and Diagnostic Imaging, The University of Georgia, Athens 30602, GA, United States
| | - Rachel Wanty Larson
- Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman 99164-6520, WA, United States
| | - Piotr Podlasz
- Department of Pathophysiology, Forensic Veterinary and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, 10-719 Olsztyn, Poland
| | - Krzysztof Czaja
- Department of Veterinary Biosciences and Diagnostic Imaging, The University of Georgia, Athens 30602, GA, United States.
| |
Collapse
|
16
|
Kapica M, Puzio I, Kato I, Kuwahara A, Zabielski R, Antushevich H. Exogenous obestatin affects pancreatic enzyme secretion in rat through two opposite mechanisms, direct inhibition and vagally-mediated stimulation. JOURNAL OF ANIMAL AND FEED SCIENCES 2018. [DOI: 10.22358/jafs/89734/2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
17
|
A High-fat, High-sugar 'Western' Diet Alters Dorsal Striatal Glutamate, Opioid, and Dopamine Transmission in Mice. Neuroscience 2017; 372:1-15. [PMID: 29289718 DOI: 10.1016/j.neuroscience.2017.12.036] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/28/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022]
Abstract
Understanding neuroadaptations involved in obesity is critical for developing new approaches to treatment. Diet-induced neuroadaptations within the dorsal striatum have the capacity to drive excessive food seeking and consumption. Five-week-old C57BL/6J mice consumed a high-fat, high-sugar 'western diet' (WD) or a control 'standard diet' (SD) for 16 weeks. Weight gain, glucose tolerance, and insulin tolerance were measured to confirm an obese-like state. Following these 16 weeks, electrophysiological recordings were made from medium spiny neurons (MSNs) in the medial (DMS) and lateral (DLS) portions of dorsal striatum to evaluate diet effects on neuronal excitability and synaptic plasticity. In addition, fast-scan cyclic voltammetry evaluated dopamine transmission in these areas. WD mice gained significantly more weight and consumed more calories than SD mice and demonstrated impaired glucose tolerance. Electrophysiology data revealed that MSNs from WD mice demonstrated increased AMPA-to-NMDA receptor current ratio and prolonged spontaneous glutamate-mediated currents, specifically in the DLS. Evoked dopamine release was also significantly greater and reuptake slower in both subregions of WD striatum. Finally, dorsal striatal MSNs from WD mice were significantly less likely to demonstrate mu-opioid receptor-mediated synaptic plasticity. Neuronal excitability and GABAergic transmission were unaffected by diet in either striatal subregion. Our results demonstrate that a high-fat, high-sugar diet alters facets of glutamate, dopamine, and opioid signaling within the dorsal striatum, with some subregion specificity. These alterations within a brain area known to play a role in food motivation/consumption and habitual behavior are highly relevant for the clinical condition of obesity and its treatment.
Collapse
|
18
|
Ong ZY, Bongiorno DM, Hernando MA, Grill HJ. Effects of Endogenous Oxytocin Receptor Signaling in Nucleus Tractus Solitarius on Satiation-Mediated Feeding and Thermogenic Control in Male Rats. Endocrinology 2017; 158:2826-2836. [PMID: 28575174 PMCID: PMC5659667 DOI: 10.1210/en.2017-00200] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/24/2017] [Indexed: 12/15/2022]
Abstract
Central oxytocin receptor (OT-R) signaling reduces food intake and increases energy expenditure, but the central sites and mechanisms mediating these effects are unresolved. We showed previously that pharmacological activation of OT-R in hindbrain/nucleus tractus solitarius (NTS) amplifies the intake-inhibitory effects of gastrointestinal (GI) satiation signals. Unexplored were the energetic effects of hindbrain OT-R agonism and the physiological relevance of NTS OT-R signaling on food intake and energy expenditure control. Using a virally mediated OT-R knockdown (KD) strategy and a range of behavioral paradigms, this study examined the role of endogenous NTS OT-R signaling on satiation-mediated food intake inhibition and thermogenic control. Results showed that, compared with controls, NTS OT-R KD rats consumed larger meals, were less responsive to the intake-inhibitory effects of a self-ingested preload, and consumed more chow following a 24-hour fast. These data indicate that NTS OT-R signaling is necessary for normal satiation control. Whereas both control and NTS OT-R KD rats increased core temperature following high-fat diet maintenance (relative to chow maintenance), the percent increase in core temperature was greater in control compared with NTS OT-R KD rats during the light cycle. Hindbrain oxytocin agonist delivery increased core temperature in both control and NTS OT-R KD rats and the percent increase relative to vehicle treatment was not significantly different between groups. Together, data reveal a critical role for endogenous NTS OT-R signaling in mediating the intake-inhibitory effects of endogenous GI satiation signals and in diet-induced thermogenesis.
Collapse
Affiliation(s)
- Zhi Yi Ong
- Department of Psychology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Diana M. Bongiorno
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Mary Ann Hernando
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Harvey J. Grill
- Department of Psychology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
19
|
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: 3.0] [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
|
20
|
Ishimaru Y, Kozuka C, Nakajima K, Sasaki T. Expanding frontiers in weight-control research explored by young investigators. J Physiol Sci 2017; 67:83-95. [PMID: 27730500 PMCID: PMC5138253 DOI: 10.1007/s12576-016-0495-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 09/30/2016] [Indexed: 01/30/2023]
Abstract
At the 93rd annual meeting of the Physiological Society of Japan, a symposium entitled "Expanding frontiers in weight-control research explored by young investigators" was organized. The latest research on weight control was presented by young up-and-coming investigators. The symposium consisted of the following presentations: Gastrointestinal brush cells, immunity, and energy homeostasis; Impact of a brown rice-derived bioactive product on feeding regulation and fuel metabolism; A novel G protein-coupled receptor-regulated neuronal signaling pathway triggers sustained orexigenic effects; and NMDA receptor co-agonist D-serine regulates food preference. These four talks presented at the symposium were summarized as a series of short reviews in this review.
Collapse
Affiliation(s)
- Yoshiro Ishimaru
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan.
| | - Chisayo Kozuka
- Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan.
| | - Kenichiro Nakajima
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan.
| | - Tsutomu Sasaki
- Laboratory for Metabolic Signaling. Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan.
| |
Collapse
|
21
|
Control of Appetite and Food Preference by NMDA Receptor and Its Co-Agonist d-Serine. Int J Mol Sci 2016; 17:ijms17071081. [PMID: 27399680 PMCID: PMC4964457 DOI: 10.3390/ijms17071081] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 06/21/2016] [Accepted: 06/30/2016] [Indexed: 11/30/2022] Open
Abstract
Obesity causes a significant negative impact on health of human beings world-wide. The main reason for weight gain, which eventually leads to obesity, is excessive ingestion of energy above the body’s homeostatic needs. Therefore, the elucidation of detailed mechanisms for appetite control is necessary to prevent and treat obesity. N-methyl-d-aspartate (NMDA) receptor is a post-synaptic glutamate receptor and is important for excitatory neurotransmission. It is expressed throughout the nervous system, and is important for long-term potentiation. It requires both ligand (glutamate) and co-agonist (d-serine or glycine) for efficient opening of the channel to allow calcium influx. d-serine is contained in fermented foods and marine invertebrates, and brain d-serine level is maintained by synthesis in vivo and supply from food and gut microbiota. Although the NMDA receptor has been reported to take part in the central regulation of appetite, the role of d-serine had not been addressed. We recently reported that exogenous d-serine administration can suppress appetite and alter food preference. In this review, we will discuss how NMDA receptor and its co-agonist d-seine participate in the control of appetite and food preference, and elaborate on how this system could possibly be manipulated to suppress obesity.
Collapse
|
22
|
Duca FA, Bauer PV, Hamr SC, Lam TKT. Glucoregulatory Relevance of Small Intestinal Nutrient Sensing in Physiology, Bariatric Surgery, and Pharmacology. Cell Metab 2015. [PMID: 26212718 DOI: 10.1016/j.cmet.2015.07.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Emerging evidence suggests the gastrointestinal tract plays an important glucoregulatory role. In this perspective, we first review how the intestine senses ingested nutrients, initiating crucial negative feedback mechanisms through a gut-brain neuronal axis to regulate glycemia, mainly via reduction in hepatic glucose production. We then highlight how intestinal energy sensory mechanisms are responsible for the glucose-lowering effects of bariatric surgery, specifically duodenal-jejunal bypass, and the antidiabetic agents metformin and resveratrol. A better understanding of these pathways lays the groundwork for intestinally targeted drug therapy for the treatment of diabetes.
Collapse
Affiliation(s)
- Frank A Duca
- Toronto General Research Institute and Department of Medicine, UHN, Toronto, ON M5G 1L7, Canada
| | - Paige V Bauer
- Toronto General Research Institute and Department of Medicine, UHN, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sophie C Hamr
- Toronto General Research Institute and Department of Medicine, UHN, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tony K T Lam
- Toronto General Research Institute and Department of Medicine, UHN, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada.
| |
Collapse
|
23
|
Sasaki T, Kinoshita Y, Matsui S, Kakuta S, Yokota-Hashimoto H, Kinoshita K, Iwasaki Y, Kinoshita T, Yada T, Amano N, Kitamura T. N-methyl-d-aspartate receptor coagonist d-serine suppresses intake of high-preference food. Am J Physiol Regul Integr Comp Physiol 2015; 309:R561-75. [PMID: 26157056 DOI: 10.1152/ajpregu.00083.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 07/07/2015] [Indexed: 12/20/2022]
Abstract
d-Serine is abundant in the forebrain and physiologically important for modulating excitatory glutamatergic neurotransmission as a coagonist of synaptic N-methyl-d-aspartate (NMDA) receptor. NMDA signaling has been implicated in the control of food intake. However, the role of d-serine on appetite regulation is unknown. To clarify the effects of d-serine on appetite, we investigated the effect of oral d-serine ingestion on food intake in three different feeding paradigms (one-food access, two-food choice, and refeeding after 24-h fasting) using three different strains of male mice (C57Bl/6J, BKS, and ICR). The effect of d-serine was also tested in leptin signaling-deficient db/db mice and sensory-deafferented (capsaicin-treated) mice. The expression of orexigenic neuropeptides [neuropeptide Y (Npy) and agouti-related protein (Agrp)] in the hypothalamus was compared in fast/refed experiments. Conditioned taste aversion for high-fat diet (HFD) was tested in the d-serine-treated mice. Under the one-food-access paradigm, some of the d-serine-treated mice showed starvation, but not when fed normal chow. HFD feeding with d-serine ingestion did not cause aversion. Under the two-food-choice paradigm, d-serine suppressed the intake of high-preference food but not normal chow. d-Serine also effectively suppressed HFD intake but not normal chow in db/db mice and sensory-deafferented mice. In addition, d-serine suppressed normal chow intake after 24-h fasting despite higher orexigenic gene expression in the hypothalamus. d-Serine failed to suppress HFD intake in the presence of L-701,324, the selective and full antagonist at the glycine-binding site of the NMDA receptor. Therefore, d-serine suppresses the intake of high-preference food through coagonism toward NMDA receptors.
Collapse
Affiliation(s)
- Tsutomu Sasaki
- Laboratory of Metabolic Signal, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan;
| | - Yoshihiro Kinoshita
- Department of Psychiatry, School of Medicine, Shinshu University, Matsumoto, Nagano, Japan
| | - Sho Matsui
- Laboratory of Metabolic Signal, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | - Shigeru Kakuta
- Research Center for Human and Environmental Sciences, Shinshu University, Matsumoto, Nagano, Japan
| | - Hiromi Yokota-Hashimoto
- Laboratory of Metabolic Signal, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | - Kuni Kinoshita
- Department of Psychiatry, School of Medicine, Shinshu University, Matsumoto, Nagano, Japan
| | - Yusaku Iwasaki
- Division of Integrated Physiology, Department of Physiology, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan; and
| | - Toshio Kinoshita
- Department of Analytical Chemistry, School of Pharmacy, Kitasato University, Tokyo, Tokyo, Japan
| | - Toshihiko Yada
- Division of Integrated Physiology, Department of Physiology, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan; and
| | - Naoji Amano
- Department of Psychiatry, School of Medicine, Shinshu University, Matsumoto, Nagano, Japan
| | - Tadahiro Kitamura
- Laboratory of Metabolic Signal, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| |
Collapse
|
24
|
Sleeve gastrectomy and Roux-en-Y gastric bypass alter the gut-brain communication. Neural Plast 2015; 2015:601985. [PMID: 25722893 PMCID: PMC4333325 DOI: 10.1155/2015/601985] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 10/15/2014] [Accepted: 10/17/2014] [Indexed: 12/15/2022] Open
Abstract
This study investigated the anatomical integrity of vagal innervation of the gastrointestinal tract following vertical sleeve gastrectomy (VSG) and Roux-en-Y gastric bypass (RYGB) operations. The retrograde tracer fast blue (FB) was injected into the stomach to label vagal neurons originating from nodose ganglion (NG) and dorsal motor nucleus of the vagus (DMV). Microglia activation was determined by quantifying changes in the fluorescent staining of hindbrain sections against an ionizing calcium adapter binding molecule 1 (Iba1). Reorganization of vagal afferents in the hindbrain was studied by fluorescent staining against isolectin 4 (IB4). The density of Iba1- and IB4-immunoreactivity was analyzed using Nikon Elements software. There was no difference in the number of FB-labeled neurons located in NG and DMV between VSG and VSG-sham rats. RYGB, but not RYGB-sham rats, showed a dramatic reduction in number of FB-labeled neurons located in the NG and DMV. VSG increased, while the RYGB operation decreased, the density of vagal afferents in the nucleus tractus solitarius (NTS). The RYGB operation, but not the VSG procedure, significantly activated microglia in the NTS and DMV. Results of this study show that the RYGB, but not the VSG procedure, triggers microglia activation in vagal structures and remodels gut-brain communication.
Collapse
|
25
|
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: 2.0] [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
|
26
|
Zhao H, Peters JH, Zhu M, Page SJ, Ritter RC, Appleyard SM. Frequency-dependent facilitation of synaptic throughput via postsynaptic NMDA receptors in the nucleus of the solitary tract. J Physiol 2014; 593:111-25. [PMID: 25281729 DOI: 10.1113/jphysiol.2013.258103] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 09/15/2014] [Indexed: 12/28/2022] Open
Abstract
Hindbrain NMDA receptors play important roles in reflexive and behavioural responses to vagal activation. NMDA receptors have also been shown to contribute to the synaptic responses of neurons in the nucleus of the solitary tract (NTS), but their exact role remains unclear. In this study we used whole cell patch-clamping techniques in rat horizontal brain slice to investigate the role of NMDA receptors in the fidelity of transmission across solitary tract afferent-NTS neuron synapses. Results show that NMDA receptors contribute up to 70% of the charge transferred across the synapse at high (>5 Hz) firing rates, but have little contribution at lower firing frequencies. Results also show that NMDA receptors critically contribute to the fidelity of transmission across these synapses during high frequency (>5 Hz) afferent discharge rates. This novel role of NMDA receptors may explain in part how primary visceral afferents, including vagal afferents, can maintain fidelity of transmission across a broad range of firing frequencies. Neurons within the nucleus of the solitary tract (NTS) receive vagal afferent innervations that initiate gastrointestinal and cardiovascular reflexes. Glutamate is the fast excitatory neurotransmitter released in the NTS by vagal afferents, which arrive there via the solitary tract (ST). ST stimulation elicits excitatory postsynaptic currents (EPSCs) in NTS neurons mediated by both AMPA- and NMDA-type glutamate receptors (-Rs). Vagal afferents exhibit a high probability of vesicle release and exhibit robust frequency-dependent depression due to presynaptic vesicle depletion. Nonetheless, synaptic throughput is maintained even at high frequencies of afferent activation. Here we test the hypothesis that postsynaptic NMDA-Rs are essential in maintaining throughput across ST-NTS synapses. Using patch clamp electrophysiology in horizontal brainstem slices, we found that NMDA-Rs, including NR2B subtypes, carry up to 70% of the charge transferred across the synapse during high frequency stimulations (>5 Hz). In contrast, their relative contribution to the ST-EPSC is much less during low (<2 Hz) frequency stimulations. Afferent-driven activation of NMDA-Rs produces a sustained depolarization during high, but not low, frequencies of stimulation as a result of relatively slow decay kinetics. Hence, NMDA-Rs are critical for maintaining action potential generation at high firing rates. These results demonstrate a novel role for NMDA-Rs enabling a high probability of release synapse to maintain the fidelity of synaptic transmission during high frequency firing when glutamate release and AMPA-R responses are reduced. They also suggest why NMDA-Rs are critical for responses that may depend on high rates of afferent discharge.
Collapse
Affiliation(s)
- Huan Zhao
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, 99164, USA
| | | | | | | | | | | |
Collapse
|
27
|
de Lartigue G. Putative roles of neuropeptides in vagal afferent signaling. Physiol Behav 2014; 136:155-69. [PMID: 24650553 DOI: 10.1016/j.physbeh.2014.03.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/23/2014] [Accepted: 03/10/2014] [Indexed: 02/06/2023]
Abstract
The vagus nerve is a major pathway by which information is communicated between the brain and peripheral organs. Sensory neurons of the vagus are located in the nodose ganglia. These vagal afferent neurons innervate the heart, the lung and the gastrointestinal tract, and convey information about peripheral signals to the brain important in the control of cardiovascular tone, respiratory tone, and satiation, respectively. Glutamate is thought to be the primary neurotransmitter involved in conveying all of this information to the brain. It remains unclear how a single neurotransmitter can regulate such an extensive list of physiological functions from a wide range of visceral sites. Many neurotransmitters have been identified in vagal afferent neurons and have been suggested to modulate the physiological functions of glutamate. Specifically, the anorectic peptide transmitters, cocaine and amphetamine regulated transcript (CART) and the orexigenic peptide transmitters, melanin concentrating hormone (MCH) are differentially regulated in vagal afferent neurons and have opposing effects on food intake. Using these two peptides as a model, this review will discuss the potential role of peptide transmitters in providing a more precise and refined modulatory control of the broad physiological functions of glutamate, especially in relation to the control of feeding.
Collapse
Affiliation(s)
- Guillaume de Lartigue
- Dept Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, Davis, CA 95616, USA.
| |
Collapse
|
28
|
Campos CA, Shiina H, Silvas M, Page S, Ritter RC. Vagal afferent NMDA receptors modulate CCK-induced reduction of food intake through synapsin I phosphorylation in adult male rats. Endocrinology 2013; 154:2613-25. [PMID: 23715865 PMCID: PMC3713210 DOI: 10.1210/en.2013-1062] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Vagal afferent nerve fibers transmit gastrointestinal satiation signals to the brain via synapses in the nucleus of the solitary tract (NTS). Despite their pivotal role in energy homeostasis, little is known about the cellular mechanisms enabling fleeting synaptic events at vagal sensory endings to sustain behavioral changes lasting minutes to hours. Previous reports suggest that the reduction of food intake by the satiation peptide, cholecystokinin (CCK), requires activation of N-methyl-D-aspartate-type glutamate receptors (NMDAR) in the NTS, with subsequent phosphorylation of ERK1/2 (pERK1/2) in NTS vagal afferent terminals. The synaptic vesicle protein synapsin I is phosphorylated by pERK1/2 at serines 62 and 67. This pERK1/2-catalyzed phosphorylation increases synaptic strength by increasing the readily releasable pool of the neurotransmitter. Conversely, dephosphorylation of serines 62 and 67 by calcineurin reduces the size of the readily releasable transmitter pool. Hence, the balance of synapsin I phosphorylation and dephosphorylation can modulate synaptic strength. We postulated that CCK-evoked activation of vagal afferent NMDARs results in pERK1/2-catalyzed phosphorylation of synapsin I in vagal afferent terminals, leading to the suppression of food intake. We found that CCK injection increased the phosphorylation of synapsin I in the NTS and that this increase is abolished after surgical or chemical ablation of vagal afferent fibers. Furthermore, fourth ventricle injection of an NMDAR antagonist or the mitogen-activated ERK kinase inhibitor blocked CCK-induced synapsin I phosphorylation, indicating that synapsin phosphorylation in vagal afferent terminals depends on NMDAR activation and ERK1/2 phosphorylation. Finally, hindbrain inhibition of calcineurin enhanced and prolonged synapsin I phosphorylation and potentiated reduction of food intake by CCK. Our findings are consistent with a mechanism in which NMDAR-dependent phosphorylation of ERK1/2 modulates satiation signals via synapsin I phosphorylation in vagal afferent endings.
Collapse
Affiliation(s)
- Carlos A Campos
- Department of Integrative Physiology and Neuroscience, Washington State University, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA.
| | | | | | | | | |
Collapse
|
29
|
Schwartz GJ, Zeltser LM. Functional organization of neuronal and humoral signals regulating feeding behavior. Annu Rev Nutr 2013; 33:1-21. [PMID: 23642202 DOI: 10.1146/annurev-nutr-071812-161125] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Energy homeostasis--ensuring that energy availability matches energy requirements--is essential for survival. One way that energy balance is achieved is through coordinated action of neural and neuroendocrine feeding circuits, which promote energy intake when energy supply is limited. Feeding behavior engages multiple somatic and visceral tissues distributed throughout the body--contraction of skeletal and smooth muscles in the head and along the upper digestive tract required to consume and digest food, as well as stimulation of endocrine and exocrine secretions from a wide range of organs. Accordingly, neurons that contribute to feeding behaviors are localized to central, peripheral, and enteric nervous systems. To promote energy balance, feeding circuits must be able to identify and respond to energy requirements, as well as the amount of energy available from internal and external sources, and then direct appropriate coordinated responses throughout the body.
Collapse
Affiliation(s)
- Gary J Schwartz
- Department of Medicine, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY 10461, USA
| | | |
Collapse
|
30
|
Blouet C, Schwartz GJ. Duodenal lipid sensing activates vagal afferents to regulate non-shivering brown fat thermogenesis in rats. PLoS One 2012; 7:e51898. [PMID: 23251649 PMCID: PMC3522613 DOI: 10.1371/journal.pone.0051898] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 11/08/2012] [Indexed: 11/18/2022] Open
Abstract
Previous evidence indicates that duodenal lipid sensing engages gut-brain neurocircuits to determine food intake and hepatic glucose production, but a potential role for gut-brain communication in the control of energy expenditure remains to be determined. Here, we tested the hypothesis that duodenal lipid sensing activates a gut-brain-brown adipose tissue neuraxis to regulate thermogenesis. We demonstrate that direct administration of lipids into the duodenum increases brown fat temperature. Co-infusion of the local anesthetic tetracaine with duodenal lipids abolished the lipid-induced increase in brown fat temperature. Systemic administration of the CCKA receptor antagonist devazepide blocked the ability of duodenal lipids to increase brown fat thermogenesis. Parenchymal administration of the N-methyl-d-aspartate receptor blocker MK-801 directly into the caudomedial nucleus of the solitary tract also abolished duodenal lipid-induced activation of brown fat thermogenesis. These findings establish that duodenal lipid sensing activates a gut-brain-brown fat axis to determine brown fat temperature, and thereby reveal a previously unappreciated pathway that regulates thermogenesis.
Collapse
Affiliation(s)
- Clémence Blouet
- Departments of Medicine and Neuroscience, Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Gary J. Schwartz
- Departments of Medicine and Neuroscience, Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
31
|
Iwasaki Y, Yada T. Vagal afferents sense meal-associated gastrointestinal and pancreatic hormones: mechanism and physiological role. Neuropeptides 2012; 46:291-7. [PMID: 23020951 DOI: 10.1016/j.npep.2012.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 08/17/2012] [Accepted: 08/17/2012] [Indexed: 01/08/2023]
Abstract
Some gastrointestinal and pancreatic hormones are potently secreted by meal intake and reduce food intake, therefore these hormones play a role in the meal-evoked satiety peptides. Previous reports have demonstrated that peripheral administration of these gastrointestinal or pancreatic hormones decrease feeding and the anorectic effects are abolished by lesions of vagal afferent nerves using surgical or chemical protocols, indicative of the involvement of the vagal afferents. Vagal afferent nerves link between several peripheral organs and the nucleus tractus solitarius of the brainstem. The present review focuses on cholecystokinin, peptide YY(3-36), pancreatic polypeptide, and nesfatin-1 released from endocrine cells of the gut and pancreas. These hormonal peptides directly act on and increase cytosolic Ca(2+) in vagal afferent nodose ganglion neurons and finally suppress food intake via vagal afferents. Therefore, peripheral terminals of vagal afferents could sense gastrointestinal and pancreatic hormones and regulate food intake. Here, we review how the vagal afferent neurons sense a variety of gastrointestinal and pancreatic hormones and discuss its physiological significance in regulation of feeding.
Collapse
Affiliation(s)
- Yusaku Iwasaki
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, 3311-1, Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | | |
Collapse
|
32
|
The role of NMDA receptors in human eating behavior: evidence from a case of anti-NMDA receptor encephalitis. Cogn Behav Neurol 2012; 25:93-7. [PMID: 22596107 DOI: 10.1097/wnn.0b013e31825921a6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Research in animal models has implicated N-methyl-D-aspartate (NMDA) receptors (NMDARs) in the control of food intake. Until now, these findings have been not replicated in humans. Here we describe a 22-year-old woman with anti-NMDAR encephalitis and no prior neurological or psychiatric history. Her clinical course was marked by successive eating disorders: anorexia followed by hyperphagia. We propose that, much as they do in other animals, NMDARs in humans interact with the neuroendocrine, homeostatic, and reward systems controlling food intake in the central and peripheral nervous system structures related to feeding and satiety.
Collapse
|
33
|
Singru PS, Wittmann G, Farkas E, Zséli G, Fekete C, Lechan RM. Refeeding-activated glutamatergic neurons in the hypothalamic paraventricular nucleus (PVN) mediate effects of melanocortin signaling in the nucleus tractus solitarius (NTS). Endocrinology 2012; 153:3804-14. [PMID: 22700769 PMCID: PMC3404351 DOI: 10.1210/en.2012-1235] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
We previously demonstrated that refeeding after a prolonged fast activates a subset of neurons in the ventral parvocellular subdivision of the paraventricular nucleus (PVNv) as a result of increased melanocortin signaling. To determine whether these neurons contribute to satiety by projecting to the nucleus tractus solitarius (NTS), the retrogradely transported marker substance, cholera toxin-β (CTB), was injected into the dorsal vagal complex of rats that were subsequently fasted and refed for 2 h. By double-labeling immunohistochemistry, CTB accumulation was found in the cytoplasm of the majority of refeeding-activated c-Fos neurons in the ventral parvocellular subdivision of the hypothalamic paraventricular nucleus (PVNv). In addition, a large number of refeeding-activated c-Fos-expressing neurons were observed in the lateral parvocellular subdivision (PVNl) that also contained CTB and were innervated by axon terminals of proopiomelanocortin neurons. To visualize the location of neuronal activation within the NTS by melanocortin-activated PVN neurons, α-MSH was focally injected into the PVN, resulting in an increased number of c-Fos-containing neurons in the PVN and in the NTS, primarily in the medial and commissural parts. All refeeding-activated neurons in the PVNv and PVNl expressed the mRNA of the glutamatergic marker, type 2 vesicular glutamate transporter (VGLUT2), indicating their glutamatergic phenotype, but only rare neurons contained oxytocin. These data suggest that melanocortin-activated neurons in the PVNv and PVNl may contribute to refeeding-induced satiety through effects on the NTS and may alter the sensitivity of NTS neurons to vagal satiety inputs via glutamate excitation.
Collapse
Affiliation(s)
- Praful S Singru
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Tufts Medical Center, no. 268, 800 Washington Street, Boston, Massachusetts 02111, USA
| | | | | | | | | | | |
Collapse
|
34
|
Campos CA, Wright JS, Czaja K, Ritter RC. CCK-induced reduction of food intake and hindbrain MAPK signaling are mediated by NMDA receptor activation. Endocrinology 2012; 153:2633-46. [PMID: 22508518 PMCID: PMC3359610 DOI: 10.1210/en.2012-1025] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 03/23/2012] [Indexed: 01/28/2023]
Abstract
The dorsal vagal complex of the hindbrain, including the nucleus of the solitary tract (NTS), receives neural and humoral afferents that contribute to the process of satiation. The gut peptide, cholecystokinin (CCK), promotes satiation by activating gastrointestinal vagal afferents that synapse in the NTS. Previously, we demonstrated that hindbrain administration of N-methyl-D-aspartate (NMDA)-type glutamate receptor antagonists attenuate reduction of food intake after ip CCK-8 injection, indicating that these receptors play a necessary role in control of food intake by CCK. However, the signaling pathways through which hindbrain NMDA receptors contribute to CCK-induced reduction of food intake have not been investigated. Here we report CCK increases phospho-ERK1/2 in NTS neurons and in identified vagal afferent endings in the NTS. CCK-evoked phospho-ERK1/2 in the NTS was attenuated in rats pretreated with capsaicin and was abolished by systemic injection of a CCK1 receptor antagonist, indicating that phosphorylation of ERK1/2 occurs in and is mediated by gastrointestinal vagal afferents. Fourth ventricle injection of a competitive NMDA receptor antagonist, prevented CCK-induced phosphorylation of ERK1/2 in hindbrain neurons and in vagal afferent endings, as did direct inhibition of MAPK kinase. Finally, fourth ventricle administration of either a MAPK kinase inhibitor or NMDA receptor antagonist prevented the reduction of food intake by CCK. We conclude that activation of NMDA receptors in the hindbrain is necessary for CCK-induced ERK1/2 phosphorylation in the NTS and consequent reduction of food intake.
Collapse
Affiliation(s)
- Carlos A Campos
- Department of Veterinary and Comparative Anatomy, Pharmacology, and Physiology and Programs in Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, Washington 99164-6520, USA
| | | | | | | |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Cholecystokinin (CCK) controls nutrient delivery to the small intestine by inhibiting food intake and gastric emptying. This review deals with recent work shedding new light on how and when. RECENT FINDINGS Intestinal I-cells release CCK in response to dietary lipid and protein through mechanisms involving the G-protein-coupled receptors GPR40 and calcium-sensing receptor. Vagal afferent neurons are a primary target of CCK and are now recognized as an important site of integration of peripheral signals regulating ingestion. In addition to regulating vagal afferent nerve discharge, CCK also controls the expression of receptors and peptide neurotransmitters by these neurons; these actions are potentiated by leptin and inhibited by ghrelin. The responses of vagal afferent neurons to CCK are attenuated in obesity. Studies of human central nervous system responses using functional magnetic resonance imaging indicate activation of brainstem, hypothalamus and motor cortex by ingested fatty acid that is inhibited by a CCK-1 receptor antagonist. CCK may also play a role in adaptive responses in pancreatic islets by maintaining β-cell mass and acting as an incretin in certain circumstances. SUMMARY CCK mediates inhibition of food intake in response to ingested lipid and protein; resistance to CCK occurs in obesity and may contribute to altered mechanisms regulating food intake.
Collapse
Affiliation(s)
- Graham J Dockray
- Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| |
Collapse
|