1
|
Bachor TP, Hwang E, Yulyaningsih E, Attal K, Mifsud F, Pham V, Vagena E, Huarcaya R, Valdearcos M, Vaisse C, Williams KW, Emmerson PJ, Xu AW. Identification of AgRP cells in the murine hindbrain that drive feeding. Mol Metab 2024; 80:101886. [PMID: 38246589 PMCID: PMC10844855 DOI: 10.1016/j.molmet.2024.101886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
OBJECTIVE The central melanocortin system is essential for the regulation of food intake and body weight. Agouti-related protein (AgRP) is the sole orexigenic component of the central melanocortin system and is conserved across mammalian species. AgRP is currently known to be expressed exclusively in the mediobasal hypothalamus, and hypothalamic AgRP-expressing neurons are essential for feeding. Here we characterized a previously unknown population of AgRP cells in the mouse hindbrain. METHODS Expression of AgRP in the hindbrain was investigated using gene expression analysis, single-cell RNA sequencing, immunofluorescent analysis and multiple transgenic mice with reporter expressions. Activation of AgRP neurons was achieved by Designer Receptors Exclusively Activated by Designer Drugs (DREADD) and by transcranial focal photo-stimulation using a step-function opsin with ultra-high light sensitivity (SOUL). RESULTS AgRP expressing cells were present in the area postrema (AP) and the adjacent subpostrema area (SubP) and commissural nucleus of the solitary tract (cNTS) of the mouse hindbrain (termed AgRPHind herein). AgRPHind cells consisted of locally projecting neurons as well as tanycyte-like cells. Food deprivation stimulated hindbrain Agrp expression as well as neuronal activity of subsets of AgRPHind cells. In adult mice that lacked hypothalamic AgRP neurons, chemogenetic activation of AgRP neurons resulted in hyperphagia and weight gain. In addition, transcranial focal photo-stimulation of hindbrain AgRP cells increased food intake in adult mice with or without hypothalamic AgRP neurons. CONCLUSIONS Our study indicates that the central melanocortin system in the hindbrain possesses an orexigenic component, and that AgRPHind neurons stimulate feeding independently of hypothalamic AgRP neurons.
Collapse
Affiliation(s)
- Tomas P Bachor
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Eunsang Hwang
- Center for Hypothalamic Research, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Ernie Yulyaningsih
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Kush Attal
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Francois Mifsud
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Viana Pham
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Eirini Vagena
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Renzo Huarcaya
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Martin Valdearcos
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Christian Vaisse
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA
| | - Kevin W Williams
- Center for Hypothalamic Research, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Paul J Emmerson
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly & Company, Indianapolis, IN, USA
| | - Allison W Xu
- Diabetes Center and Department of Anatomy, University of California, San Francisco, California, USA.
| |
Collapse
|
2
|
Metzger PJ, Zhang A, Carlson BA, Sun H, Cui Z, Li Y, Jahnke MT, Layton DR, Gupta MB, Liu N, Kostenis E, Gavrilova O, Chen M, Weinstein LS. A human obesity-associated MC4R mutation with defective Gq/11α signaling leads to hyperphagia in mice. J Clin Invest 2024; 134:e165418. [PMID: 38175730 PMCID: PMC10869179 DOI: 10.1172/jci165418] [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: 09/14/2022] [Accepted: 12/21/2023] [Indexed: 01/06/2024] Open
Abstract
Melanocortin 4 receptor (MC4R) mutations are the most common cause of human monogenic obesity and are associated with hyperphagia and increased linear growth. While MC4R is known to activate Gsα/cAMP signaling, a substantial proportion of obesity-associated MC4R mutations do not affect MC4R/Gsα signaling. To further explore the role of specific MC4R signaling pathways in the regulation of energy balance, we examined the signaling properties of one such mutant, MC4R (F51L), as well as the metabolic consequences of MC4RF51L mutation in mice. The MC4RF51L mutation produced a specific defect in MC4R/Gq/11α signaling and led to obesity, hyperphagia, and increased linear growth in mice. The ability of a melanocortin agonist to acutely inhibit food intake when delivered to the paraventricular nucleus (PVN) was lost in MC4RF51L mice, as well as in WT mice in which a specific Gq/11α inhibitor was delivered to the PVN; this provided evidence that a Gsα-independent signaling pathway, namely Gq/11α, significantly contributes to the actions of MC4R on food intake and linear growth. These results suggest that a biased MC4R agonist that primarily activates Gq/11α may be a potential agent to treat obesity with limited untoward cardiovascular and other side effects.
Collapse
Affiliation(s)
| | | | | | - Hui Sun
- Metabolic Diseases Branch and
| | - Zhenzhong Cui
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | | | | | | | | | - Naili Liu
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Evi Kostenis
- Molecular, Cellular, and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Oksana Gavrilova
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | | | | |
Collapse
|
3
|
Busebee B, Ghusn W, Cifuentes L, Acosta A. Obesity: A Review of Pathophysiology and Classification. Mayo Clin Proc 2023; 98:1842-1857. [PMID: 37831039 PMCID: PMC10843116 DOI: 10.1016/j.mayocp.2023.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 10/14/2023]
Abstract
Obesity is a chronic, multifactorial, and morbid disease. In the United States, 69% of adults are overweight or have obesity, and the global prevalence of obesity is increasing. Obesity is influenced by genetic, neurologic, metabolic, enteric, and behavioral processes. It remains a key modifiable risk factor for many comorbid diseases, including cardiovascular disease, diabetes mellitus, and cancer. Whereas there are recent and significant advances in obesity therapy, including diets, lifestyle modifications, pharmacotherapies, endoscopic procedures, and bariatric surgeries, there is an immense need for a better understanding of the heterogeneity in the pathophysiologic process of obesity and outcomes. Here we review salient pathophysiologic mechanisms underlying the development and morbidity of obesity as well as pathophysiologically based classification systems that inform current obesity management and may inform improved and individualized management in the future.
Collapse
Affiliation(s)
| | - Wissam Ghusn
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Lizeth Cifuentes
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
4
|
Ruggiero-Ruff RE, Villa PA, Hijleh SA, Avalos B, DiPatrizio NV, Haga-Yamanaka S, Coss D. Increased body weight in mice with fragile X messenger ribonucleoprotein 1 (Fmr1) gene mutation is associated with hypothalamic dysfunction. Sci Rep 2023; 13:12666. [PMID: 37542065 PMCID: PMC10403586 DOI: 10.1038/s41598-023-39643-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/28/2023] [Indexed: 08/06/2023] Open
Abstract
Mutations in the Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene are linked to Fragile X Syndrome, the most common monogenic cause of intellectual disability and autism. People affected with mutations in FMR1 have higher incidence of obesity, but the mechanisms are largely unknown. In the current study, we determined that male Fmr1 knockout mice (KO, Fmr1-/y), but not female Fmr1-/-, exhibit increased weight when compared to wild-type controls, similarly to humans with FMR1 mutations. No differences in food or water intake were found between groups; however, male Fmr1-/y display lower locomotor activity, especially during their active phase. Moreover, Fmr1-/y have olfactory dysfunction determined by buried food test, although they exhibit increased compulsive behavior, determined by marble burying test. Since olfactory brain regions communicate with hypothalamic regions that regulate food intake, including POMC neurons that also regulate locomotion, we examined POMC neuron innervation and numbers in Fmr1-/y mice. POMC neurons express Fmrp, and POMC neurons in Fmr1-/y have higher inhibitory GABAergic synaptic inputs. Consistent with increased inhibitory innervation, POMC neurons in the Fmr1-/y mice exhibit lower activity, based on cFOS expression. Notably, Fmr1-/y mice have fewer POMC neurons than controls, specifically in the rostral arcuate nucleus, which could contribute to decreased locomotion and increased body weight. These results suggest a role for Fmr1 in the regulation of POMC neuron function and the etiology of Fmr1-linked obesity.
Collapse
Affiliation(s)
- Rebecca E Ruggiero-Ruff
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Pedro A Villa
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Sarah Abu Hijleh
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Bryant Avalos
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Nicholas V DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Sachiko Haga-Yamanaka
- Department of Molecular, Cell, and Systems Biology, College of Natural and Agricultural Sciences, University of California, Riverside, Riverside, USA
| | - Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA.
| |
Collapse
|
5
|
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: 6] [Impact Index Per Article: 3.0] [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
|
6
|
Holt MK. The ins and outs of the caudal nucleus of the solitary tract: An overview of cellular populations and anatomical connections. J Neuroendocrinol 2022; 34:e13132. [PMID: 35509189 PMCID: PMC9286632 DOI: 10.1111/jne.13132] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/25/2022] [Accepted: 03/22/2022] [Indexed: 11/29/2022]
Abstract
The body and brain are in constant two-way communication. Driving this communication is a region in the lower brainstem: the dorsal vagal complex. Within the dorsal vagal complex, the caudal nucleus of the solitary tract (cNTS) is a major first stop for incoming information from the body to the brain carried by the vagus nerve. The anatomy of this region makes it ideally positioned to respond to signals of change in both emotional and bodily states. In turn, the cNTS controls the activity of regions throughout the brain that are involved in the control of both behaviour and physiology. This review is intended to help anyone with an interest in the cNTS. First, I provide an overview of the architecture of the cNTS and outline the wide range of neurotransmitters expressed in subsets of neurons in the cNTS. Next, in detail, I discuss the known inputs and outputs of the cNTS and briefly highlight what is known regarding the neurochemical makeup and function of those connections. Then, I discuss one group of cNTS neurons: glucagon-like peptide-1 (GLP-1)-expressing neurons. GLP-1 neurons serve as a good example of a group of cNTS neurons, which receive input from varied sources and have the ability to modulate both behaviour and physiology. Finally, I consider what we might learn about other cNTS neurons from our study of GLP-1 neurons and why it is important to remember that the manipulation of molecularly defined subsets of cNTS neurons is likely to affect physiology and behaviours beyond those monitored in individual experiments.
Collapse
Affiliation(s)
- Marie K. Holt
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| |
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: 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
|
8
|
Khodarahmi M, Niknam M, Farhangi MA. Personalized gene-diet study of rs2239670 gene variants and dietary patterns among obese adults. Clin Nutr ESPEN 2022; 47:358-366. [DOI: 10.1016/j.clnesp.2021.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 10/31/2021] [Accepted: 11/08/2021] [Indexed: 12/29/2022]
|
9
|
Metz MJ, Daimon CM, King CM, Rau AR, Hentges ST. Individual arcuate nucleus proopiomelanocortin neurons project to select target sites. Am J Physiol Regul Integr Comp Physiol 2021; 321:R982-R989. [PMID: 34755553 PMCID: PMC8714814 DOI: 10.1152/ajpregu.00169.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 11/22/2022]
Abstract
Proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARH) are a diverse group of neurons that project widely to different brain regions. It is unknown how this small population of neurons organizes its efferent projections. In this study, we hypothesized that individual ARH POMC neurons exclusively innervate select target regions. To investigate this hypothesis, we first verified that only a fraction of ARH POMC neurons innervate the lateral hypothalamus (LH), the paraventricular nucleus of the hypothalamus (PVN), the periaqueductal gray (PAG), or the ventral tegmental area (VTA) using the retrograde tracer cholera toxin B (CTB). Next, two versions of CTB conjugated to distinct fluorophores were injected bilaterally into two of the regions such that PVN and VTA, PAG and VTA, or LH and PVN received tracers simultaneously. These pairs of target sites were chosen based on function and location. Few individual ARH POMC neurons projected to two brain regions at once, suggesting that there are ARH POMC neuron subpopulations organized by their efferent projections. We also investigated whether increasing the activity of POMC neurons could increase the number of ARH POMC neurons labeled with CTB, implying an increase in new synaptic connections to downstream regions. However, chemogenetic enhancement of POMC neuron activity did not increase retrograde tracing of CTB back to ARH POMC neurons from either the LH, PVN, or VTA. Overall, subpopulations of ARH POMC neurons with distinct efferent projections may serve as a way for the POMC population to organize its many functions.
Collapse
Affiliation(s)
- Marissa J Metz
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Caitlin M Daimon
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Connie M King
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Andrew R Rau
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Shane T Hentges
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
10
|
Lhomme T, Clasadonte J, Imbernon M, Fernandois D, Sauve F, Caron E, Lima N, Heras V, Martinez-Corral I, Müller-Fielitz H, Rasika S, Schwaninger M, Nogueiras R, Prevot V. Tanycytic networks mediate energy balance by feeding lactate to glucose-insensitive POMC neurons. J Clin Invest 2021; 131:e140521. [PMID: 34324439 DOI: 10.1172/jci140521] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
Hypothalamic glucose sensing enables an organism to match energy expenditure and food intake to circulating levels of glucose, the main energy source of the brain. Here, we established that tanycytes of the hypothalamic arcuate nucleus, specialized glia that line the wall of the third ventricle, convert brain glucose supplies into lactate that they transmit through monocarboxylate transporters to arcuate proopiomelanocortin neurons, which integrate this signal to drive their activity and to adapt the metabolic response to meet physiological demands. Furthermore, this transmission required the formation of extensive Connexin-43 gap-junction-mediated metabolic networks by arcuate tanycytes. Selectively suppressing either tanycytic monocarboxylate transporters or gap junctions resulted in altered feeding behavior and energy metabolism. Tanycytic intercellular communication and lactate production are thus integral to the mechanism by which hypothalamic neurons that regulate energy and glucose homeostasis efficiently perceive alterations in systemic glucose levels as a function of the physiological state of the organism.
Collapse
Affiliation(s)
- Tori Lhomme
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Jerome Clasadonte
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Monica Imbernon
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Daniela Fernandois
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Florent Sauve
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Emilie Caron
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Natalia Lima
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Violeta Heras
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ines Martinez-Corral
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Helge Müller-Fielitz
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - S Rasika
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Ruben Nogueiras
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| |
Collapse
|
11
|
Sarver DC, Stewart AN, Rodriguez S, Little HC, Aja S, Wong GW. Loss of CTRP4 alters adiposity and food intake behaviors in obese mice. Am J Physiol Endocrinol Metab 2020; 319:E1084-E1100. [PMID: 33017221 PMCID: PMC7792665 DOI: 10.1152/ajpendo.00448.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Central and peripheral mechanisms are both required for proper control of energy homeostasis. Among circulating plasma proteins, C1q/TNF-related proteins (CTRPs) have recently emerged as important regulators of sugar and fat metabolism. CTRP4, expressed in brain and adipose tissue, is unique among the family members in having two tandem globular C1q domains. We previously showed that central administration of recombinant CTRP4 suppresses food intake, suggesting a central nervous system role in regulating ingestive physiology. Whether this effect is pharmacological or physiological remains unclear. We used a loss-of-function knockout (KO) mouse model to clarify the physiological role of CTRP4. Under basal conditions, CTRP4 deficiency increased serum cholesterol levels and impaired glucose tolerance in male but not female mice fed a control low-fat diet. When challenged with a high-fat diet, male and female KO mice responded differently to weight gain and had different food intake patterns. On an obesogenic diet, male KO mice had similar weight gain as wild-type littermates. When fed ad libitum, KO male mice had greater meal number, shorter intermeal interval, and reduced satiety ratio. Female KO mice, in contrast, had lower body weight and adiposity. In the refeeding period following food deprivation, female KO mice had significantly higher food intake due to longer meal duration and reduced satiety ratio. Collectively, our data provide genetic evidence for a sex-dependent physiological role of CTRP4 in modulating food intake patterns and systemic energy metabolism.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ashley N Stewart
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Susana Rodriguez
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hannah C Little
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
12
|
Qin L, Tiwari AK, Zai CC, Freeman N, Zhai D, Liu F, Stachelscheid H, Mergenthaler P, Kennedy JL, Müller DJ. Regulation of melanocortin-4-receptor (MC4R) expression by SNP rs17066842 is dependent on glucose concentration. Eur Neuropsychopharmacol 2020; 37:39-48. [PMID: 32684494 DOI: 10.1016/j.euroneuro.2020.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 05/18/2020] [Accepted: 05/24/2020] [Indexed: 11/17/2022]
Abstract
Melanocortin-4-receptor (MC4R) gene codes for a G-protein-coupled receptor that is highly expressed in the hypothalamus and involved in the regulation of appetite. Single-nucleotide polymorphisms (SNPs) in the MC4R gene region have been associated with obesity, type 2-diabetes (T2D) and with antipsychotic-induced weight gain. Of these, rs17066842 (G>A) in the MC4R promoter region is the top variant associated with obesity and diabetes. In this study, we investigated the effect of rs17066842 on MC4R expression at various glucose concentrations using reporter gene expression in the SH-SY5Y cell line and regulation of MC4R expression in human cerebral organoids. We observed that higher glucose concentrations significantly reduced MC4R mRNA expression in SH-SY5Y cells. In addition, at high glucose concentrations, the luciferase reporter plasmid containing the MC4R promoter insert with the G-allele of rs170066842 showed significantly reduced activity compared to the A-allele carrying plasmid. The immediate early gene product, early growth-response 1 (EGR-1), was identified to bind to the sequence containing the G-allele at rs17066842 but not to the A-allele-containing sequence. Interestingly, in human induced pluripotent stem cell (hiPSC)-derived cerebral organoids, we observed increased MC4R expression in response to high glucose exposure. These opposite observations might suggest that glucose regulation is complex and may be cell-specific. This study provides evidence that rs17066842 regulates MC4R gene expression through binding of EGR-1 and that this process is influenced by glucose concentration.
Collapse
Affiliation(s)
- Li Qin
- Pharmacogenetics Research Clinic, Molecular Brain Science, Centre for Addiction and Mental Health, Toronto M5T 1R8, ON, Canada
| | - Arun K Tiwari
- Pharmacogenetics Research Clinic, Molecular Brain Science, Centre for Addiction and Mental Health, Toronto M5T 1R8, ON, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario M5T 1R8, Canada
| | - Clement C Zai
- Pharmacogenetics Research Clinic, Molecular Brain Science, Centre for Addiction and Mental Health, Toronto M5T 1R8, ON, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario M5T 1R8, Canada
| | - Natalie Freeman
- Pharmacogenetics Research Clinic, Molecular Brain Science, Centre for Addiction and Mental Health, Toronto M5T 1R8, ON, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada
| | - Dongxu Zhai
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario M5T 1R8, Canada
| | - Fang Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario M5T 1R8, Canada
| | - Harald Stachelscheid
- Berlin Institute of Health, Stem Cell Core Facility, 13353 Berlin, Germany; Charité - Universitätsmedizin Berlin, Charité-BIH Centrum Therapy and Research 13353 Berlin, Germany
| | - Philipp Mergenthaler
- Charité-Universitätsmedizin Berlin, Department of Experimental Neurology, Department of Neurology, Center for Stroke Research Berlin, 10117 Berlin, Germany; Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - James L Kennedy
- Pharmacogenetics Research Clinic, Molecular Brain Science, Centre for Addiction and Mental Health, Toronto M5T 1R8, ON, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario M5T 1R8, Canada
| | - Daniel J Müller
- Pharmacogenetics Research Clinic, Molecular Brain Science, Centre for Addiction and Mental Health, Toronto M5T 1R8, ON, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario M5T 1R8, Canada.
| |
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: 0.5] [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: 0.5] [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
|
Wang L, De Solis AJ, Goffer Y, Birkenbach KE, Engle SE, Tanis R, Levenson JM, Li X, Rausch R, Purohit M, Lee JY, Tan J, De Rosa MC, Doege CA, Aaron HL, Martins GJ, Brüning JC, Egli D, Costa R, Berbari N, Leibel RL, Stratigopoulos G. Ciliary gene RPGRIP1L is required for hypothalamic arcuate neuron development. JCI Insight 2019; 4:e123337. [PMID: 30728336 PMCID: PMC6413800 DOI: 10.1172/jci.insight.123337] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 01/03/2019] [Indexed: 12/16/2022] Open
Abstract
Intronic polymorphisms in the α-ketoglutarate-dependent dioxygenase gene (FTO) that are highly associated with increased body weight have been implicated in the transcriptional control of a nearby ciliary gene, retinitis pigmentosa GTPase regulator-interacting protein-1 like (RPGRIP1L). Previous studies have shown that congenital Rpgrip1l hypomorphism in murine proopiomelanocortin (Pomc) neurons causes obesity by increasing food intake. Here, we show by congenital and adult-onset Rpgrip1l deletion in Pomc-expressing neurons that the hyperphagia and obesity are likely due to neurodevelopmental effects that are characterized by a reduction in the Pomc/Neuropeptide Y (Npy) neuronal number ratio and marked increases in arcuate hypothalamic-paraventricular hypothalamic (ARH-PVH) axonal projections. Biallelic RPGRIP1L mutations result in fewer cilia-positive human induced pluripotent stem cell-derived (iPSC-derived) neurons and blunted responses to Sonic Hedgehog (SHH). Isogenic human ARH-like embryonic stem cell-derived (ESc-derived) neurons homozygous for the obesity-risk alleles at rs8050136 or rs1421085 have decreased RPGRIP1L expression and have lower numbers of POMC neurons. RPGRIP1L overexpression increases POMC cell number. These findings suggest that apparently functional intronic polymorphisms affect hypothalamic RPGRIP1L expression and impact development of POMC neurons and their derivatives, leading to hyperphagia and increased adiposity.
Collapse
Affiliation(s)
- Liheng Wang
- Naomi Berrie Diabetes Center and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Alain J. De Solis
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Yossef Goffer
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Kathryn E. Birkenbach
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Staci E. Engle
- Department of Biology, Indiana University-Purdue University, Indianapolis, Indiana, USA
| | - Ross Tanis
- University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Jacob M. Levenson
- University of Central Florida College of Medicine, Orlando, Florida, USA
| | - Xueting Li
- Institute of Human Nutrition graduate program, Columbia University, New York, New York, USA
| | - Richard Rausch
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Manika Purohit
- Zuckerman Institute, Columbia University, New York, New York, USA
| | - Jen-Yi Lee
- Cancer Research Laboratory Molecular Imaging Center, University of California, Berkeley, 94720, USA
| | - Jerica Tan
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Maria Caterina De Rosa
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Claudia A. Doege
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Holly L. Aaron
- Cancer Research Laboratory Molecular Imaging Center, University of California, Berkeley, 94720, USA
| | | | - Jens C. Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- National Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Dieter Egli
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Rui Costa
- Zuckerman Institute, Columbia University, New York, New York, USA
| | - Nicolas Berbari
- Department of Biology, Indiana University-Purdue University, Indianapolis, Indiana, USA
| | - Rudolph L. Leibel
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - George Stratigopoulos
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| |
Collapse
|
16
|
Abstract
This review evaluates published studies regarding alpha-melanocyte stimulating hormone (α-MSH) in ghrelin-elicited feeding and gut motility. We have sought to integrate all available evidences to provide a complete review on the properties of melanocortin receptors (MCR) and the potential clinical treatment of α-MSH after ghrelin-elicited feeding and gut motility. The available studies were grouped into four categories: food intake, gastric emptying, small intestinal transit, and colonic transit. As we describe, the literature provides evidence of the ability of ghrelin to increase food intake, gastric emptying, small intestinal transit, and colonic transit. α-MSH, which displays high affinity for the MC3 and MC4 receptors, can competitively activate MCRs with agouti-related protein stimulated by ghrelin, and partly attenuates the effect of acyl ghrelin on food intake. Central ghrelin-induced acceleration of gastric emptying is not mediated by MCRs, but the acceleration of the small intestinal transit is at least partly mediated via MCRs in the brain. Similar to fecal pellets and total fecal weight, distal colonic motility and secretion are partly mediated by MCRs in the brain. The interplay between acyl ghrelin and MCRs may provide a new therapeutic avenue to ameliorate anorexia and constipation.
Collapse
Affiliation(s)
- Hsien-Hao Huang
- Department of Emergency Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Institute of Emergency and Critical Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan, ROC
| | - Chih-Yen Chen
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan, ROC
- Taiwan Association for the Study of Small Intestinal Diseases, Guishan, Taiwan, ROC
- Chinese Taipei Society for the Study of Obesity, Taipei, Taiwan, ROC
| |
Collapse
|
17
|
Davis XS, Grill H. The hindbrain is a site of energy balance action for prolactin-releasing peptide: feeding and thermic effects from GPR10 stimulation of the nucleus tractus solitarius/area postrema. Psychopharmacology (Berl) 2018; 235:2287-2301. [PMID: 29796829 PMCID: PMC8019516 DOI: 10.1007/s00213-018-4925-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/07/2018] [Indexed: 01/24/2023]
Abstract
PURPOSE Prolactin-releasing peptide (PrRP) is a neuropeptide that suppresses food intake and increases body temperature when delivered to the forebrain ventricularly or parenchymally. However, PrRP's receptor GPR10 is widely distributed throughout the brain with particularly high levels found in the dorsomedial hindbrain. Thus, we hypothesized that hindbrain-directed PrRP administration would affect energy balance and motivated feeding behavior. METHODS To address this hypothesis, a range of behavioral and physiologic variables were measured in Sprague-Dawley rats that received PrRP delivered to the fourth ventricle (4V) or the nucleus of the solitary tract (NTS) at the level of the area postrema (AP). RESULTS 4V PrRP delivery decreased chow intake and body weight, in part, through decreasing meal size in ad libitum maintained rats tested at dark onset. PrRP inhibited feeding when delivered to the nucleus tractus solitarius (NTS), but not to more ventral hindbrain structures. In addition, 4V as well as direct NTS administration of PrRP increased core temperature. By contrast, 4V PrRP did not reduce ad libitum intake of highly palatable food or the motivation to work for or seek palatable foods. CONCLUSIONS The dorsomedial hindbrain and NTS/AP, in particular, are sites of action in PrRP/GPR10-mediated control of chow intake, core temperature, and body weight.
Collapse
Affiliation(s)
- X. S. Davis
- Department of Psychology, University of Pennsylvania, 433 S. University Avenue, Rm. 327, Philadelphia, PA 19104, USA
| | - H.J. Grill
- Department of Psychology, University of Pennsylvania, 433 S. University Avenue, Rm. 327, Philadelphia, PA 19104, USA
| |
Collapse
|
18
|
Wang C, He Y, Xu P, Yang Y, Saito K, Xia Y, Yan X, Hinton A, Yan C, Ding H, Yu L, Shu G, Gupta R, Wu Q, Tong Q, Lagor WR, Flores ER, Xu Y. TAp63 contributes to sexual dimorphism in POMC neuron functions and energy homeostasis. Nat Commun 2018; 9:1544. [PMID: 29670083 PMCID: PMC5906443 DOI: 10.1038/s41467-018-03796-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 03/13/2018] [Indexed: 01/09/2023] Open
Abstract
Sexual dimorphism exists in energy balance, but the underlying mechanisms remain unclear. Here we show that the female mice have more pro-opiomelanocortin (POMC) neurons in the arcuate nucleus of hypothalamus than males, and female POMC neurons display higher neural activities, compared to male counterparts. Strikingly, deletion of the transcription factor, TAp63, in POMC neurons confers "male-like" diet-induced obesity (DIO) in female mice associated with decreased POMC neural activities; but the same deletion does not affect male mice. Our results indicate that TAp63 in female POMC neurons contributes to the enhanced POMC neuron functions and resistance to obesity in females. Thus, TAp63 in POMC neurons is one key molecular driver for the sexual dimorphism in energy homeostasis.
Collapse
Affiliation(s)
- Chunmei Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yanlin He
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Pingwen Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yongjie Yang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kenji Saito
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yan Xia
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xiaofeng Yan
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Antentor Hinton
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chunling Yan
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Hongfang Ding
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Likai Yu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Gang Shu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Rajat Gupta
- Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Qi Wu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - William R Lagor
- Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Elsa R Flores
- Department of Molecular Oncology, Cancer Biology and Evolution Program, Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
19
|
Dezfuli G, Gillis RA, Tatge JE, Duncan KR, Dretchen KL, Jackson PG, Verbalis JG, Sahibzada N. Subdiaphragmatic Vagotomy With Pyloroplasty Ameliorates the Obesity Caused by Genetic Deletion of the Melanocortin 4 Receptor in the Mouse. Front Neurosci 2018; 12:104. [PMID: 29545738 PMCID: PMC5838008 DOI: 10.3389/fnins.2018.00104] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 02/12/2018] [Indexed: 12/22/2022] Open
Abstract
Background/Objectives: We tested the hypothesis that abolishing vagal nerve activity will reverse the obesity phenotype of melanocortin 4 receptor knockout mice (Mc4r−/−). Subjects/Methods: In two separate studies, we examined the efficacy of bilateral subdiaphragmatic vagotomy (SDV) with pyloroplasty in the prevention and treatment of obesity in Mc4r−/− mice. Results: In the first study, SDV prevented >20% increase in body weight (BW) associated with this genotype. This was correlated with a transient reduction in overall food intake (FI) in the preventative arm of the study. Initially, SDV mice had reduced weekly FI; however, FI normalized to that of controls and baseline FI within the 8-week study period. In the second study, the severe obesity that is characteristic of the adult Mc4r−/− genotype was significantly improved by SDV with a magnitude of 30% loss in excess BW over a 4-week period. Consistent with the first preventative study, within the treatment arm, SDV mice also demonstrated a transient reduction in FI relative to control and baseline levels that normalized over subsequent weeks. In addition to the accompanying loss in weight, mice subjected to SDV showed a decrease in respiratory exchange ratio (RER), and an increase in locomotor activity (LA). Analysis of the white fat-pad deposits of these mice showed that they were significantly less than the control groups. Conclusions: Altogether, our data demonstrates that SDV both prevents gain in BW and causes weight loss in severely obese Mc4r−/− mice. Moreover, it suggests that an important aspect of weight reduction for this type of monogenic obesity involves loss of signaling in vagal motor neurons.
Collapse
Affiliation(s)
- Ghazaul Dezfuli
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Richard A Gillis
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Jaclyn E Tatge
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Kimbell R Duncan
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Kenneth L Dretchen
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Patrick G Jackson
- Department of Surgery, Georgetown University Medical Center, Washington, DC, United States
| | - Joseph G Verbalis
- Department of Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - Niaz Sahibzada
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
20
|
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: 2.5] [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]
|
21
|
Al-Najim W, le Roux CW, Docherty NG. Integrated insights into the role of alpha-melanocyte stimulatory hormone in the control of food intake and glycaemia. Peptides 2018; 100:243-248. [PMID: 29412826 DOI: 10.1016/j.peptides.2017.12.003] [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: 10/01/2017] [Revised: 12/01/2017] [Accepted: 12/04/2017] [Indexed: 10/18/2022]
Abstract
Identifying peptide hormones with multipotent actions on both weight and glycaemia can have a significant impact on therapeutic options in the treatment of obesity and diabetes. This has been exemplified by recent advances involving pharmacological exploitation of glucagon-like peptide 1 biology. Herein, we summarise evidence supporting the potential candidacy in this light of alpha-melanocyte stimulatory hormone, an endogenous peptide hormone and a breakdown product of the neuropeptide pro-opiomelanocortin. We reference its well described central actions in the control of food intake and moreover highlight new data pointing to an important role for this peptide hormone in the periphery, in relation to glycaemic control.
Collapse
Affiliation(s)
- Werd Al-Najim
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Ireland; Investigative Science, Imperial College London, UK
| | - Carel W le Roux
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Ireland; Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden; Investigative Science, Imperial College London, UK
| | - Neil G Docherty
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Ireland; Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden.
| |
Collapse
|
22
|
α-MSH Influences the Excitability of Feeding-Related Neurons in the Hypothalamus and Dorsal Vagal Complex of Rats. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2034691. [PMID: 29318141 PMCID: PMC5727559 DOI: 10.1155/2017/2034691] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 10/07/2017] [Accepted: 10/22/2017] [Indexed: 11/23/2022]
Abstract
Alpha-melanocyte-stimulating hormone (α-MSH) is processed from proopiomelanocortin (POMC) and acts on the melanocortin receptors, MC3 and MC4. α-MSH plays a key role in energy homeostasis. In the present study, to shed light on the mechanisms by which α-MSH exerts its anorectic effects, extracellular neuronal activity was recorded in the hypothalamus and the dorsal vagal complex (DVC) of anesthetized rats. We examined the impact of α-MSH on glucose-sensing neurons and gastric distension (GD) sensitive neurons. In the lateral hypothalamus (LHA), α-MSH inhibited 75.0% of the glucose-inhibited (GI) neurons. In the ventromedial nucleus (VMN), most glucose-sensitive neurons were glucose-excited (GE) neurons, which were mainly activated by α-MSH. In the paraventricular nucleus (PVN), α-MSH suppressed the majority of GI neurons and excited most GE neurons. In the DVC, among the 20 GI neurons examined for a response to α-MSH, 1 was activated, 16 were depressed, and 3 failed to respond. Nineteen of 24 GE neurons were activated by α-MSH administration. Additionally, among the 42 DVC neurons examined for responses to GD, 23 were excited (GD-EXC) and 19 were inhibited (GD-INH). Fifteen of 20 GD-EXC neurons were excited, whereas 11 out of 14 GD-INH neurons were suppressed by α-MSH. All these responses were abolished by pretreatment with the MC3/4R antagonist, SHU9119. In conclusion, the activity of glucose-sensitive neurons and GD-sensitive neurons in the hypothalamus and DVC can be modulated by α-MSH.
Collapse
|
23
|
Lau J, Farzi A, Qi Y, Heilbronn R, Mietzsch M, Shi YC, Herzog H. CART neurons in the arcuate nucleus and lateral hypothalamic area exert differential controls on energy homeostasis. Mol Metab 2017; 7:102-118. [PMID: 29146410 PMCID: PMC5784325 DOI: 10.1016/j.molmet.2017.10.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 10/27/2017] [Accepted: 10/30/2017] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE The cocaine- and amphetamine-regulated transcript (CART) codes for a pivotal neuropeptide important in the control of appetite and energy homeostasis. However, limited understanding exists for the defined effector sites underlying CART function, as discrepant effects of central CART administration have been reported. METHODS By combining Cart-cre knock-in mice with a Cart adeno-associated viral vector designed using the flip-excision switch (AAV-FLEX) technology, specific reintroduction or overexpression of CART selectively in CART neurons in the arcuate nucleus (Arc) and lateral hypothalamic area (LHA), respectively, was achieved. The effects on energy homeostasis control were investigated. RESULTS Here we show that CART neuron-specific reintroduction of CART into the Arc and LHA leads to distinct effects on energy homeostasis control. Specifically, CART reintroduction into the Arc of otherwise CART-deficient Cartcre/cre mice markedly decreased fat mass and body weight, whereas CART reintroduction into the LHA caused significant fat mass gain and lean mass loss, but overall unaltered body weight. The reduced adiposity in ArcCART;Cartcre/cre mice was associated with an increase in both energy expenditure and physical activity, along with significantly decreased Npy mRNA levels in the Arc but with no change in food consumption. Distinctively, the elevated fat mass in LHACART;Cartcre/cre mice was accompanied by diminished insulin responsiveness and glucose tolerance, greater spontaneous food intake, and reduced energy expenditure, which is consistent with the observed decrease of brown adipose tissue temperature. This is also in line with significantly reduced tyrosine hydroxylase (Th) and notably increased corticotropin-releasing hormone (Crh) mRNA expressions in the paraventricular nucleus (PVN). CONCLUSIONS Taken together, these results identify catabolic and anabolic effects of CART in the Arc and LHA, respectively, demonstrating for the first time the distinct and region-specific functions of CART in controlling feeding and energy homeostasis.
Collapse
Affiliation(s)
- Jackie Lau
- Neuroscience Division, Garvan Institute of Medical Research, St. Vincent's Hospital, Darlinghurst, NSW 2010, Sydney, Australia
| | - Aitak Farzi
- Neuroscience Division, Garvan Institute of Medical Research, St. Vincent's Hospital, Darlinghurst, NSW 2010, Sydney, Australia
| | - Yue Qi
- Neuroscience Division, Garvan Institute of Medical Research, St. Vincent's Hospital, Darlinghurst, NSW 2010, Sydney, Australia
| | - Regine Heilbronn
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, Campus Benjamin Franklin, Germany
| | - Mario Mietzsch
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, Campus Benjamin Franklin, Germany
| | - Yan-Chuan Shi
- Neuroscience Division, Garvan Institute of Medical Research, St. Vincent's Hospital, Darlinghurst, NSW 2010, Sydney, Australia.
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research, St. Vincent's Hospital, Darlinghurst, NSW 2010, Sydney, Australia.
| |
Collapse
|
24
|
Inhibition of central de novo ceramide synthesis restores insulin signaling in hypothalamus and enhances β-cell function of obese Zucker rats. Mol Metab 2017; 8:23-36. [PMID: 29233519 PMCID: PMC5985020 DOI: 10.1016/j.molmet.2017.10.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/17/2017] [Accepted: 10/27/2017] [Indexed: 12/29/2022] Open
Abstract
Objectives Hypothalamic lipotoxicity has been shown to induce central insulin resistance and dysregulation of glucose homeostasis; nevertheless, elucidation of the regulatory mechanisms remains incomplete. Here, we aimed to determine the role of de novo ceramide synthesis in hypothalamus on the onset of central insulin resistance and the dysregulation of glucose homeostasis induced by obesity. Methods Hypothalamic GT1-7 neuronal cells were treated with palmitate. De novo ceramide synthesis was inhibited either by pharmacological (myriocin) or molecular (si-Serine Palmitoyl Transferase 2, siSPT2) approaches. Obese Zucker rats (OZR) were intracerebroventricularly infused with myriocin to inhibit de novo ceramide synthesis. Insulin resistance was determined by quantification of Akt phosphorylation. Ceramide levels were quantified either by a radioactive kinase assay or by mass spectrometry analysis. Glucose homeostasis were evaluated in myriocin-treated OZR. Basal and glucose-stimulated parasympathetic tonus was recorded in OZR. Insulin secretion from islets and β-cell mass was also determined. Results We show that palmitate impaired insulin signaling and increased ceramide levels in hypothalamic neuronal GT1-7 cells. In addition, the use of deuterated palmitic acid demonstrated that palmitate activated several enzymes of the de novo ceramide synthesis pathway in hypothalamic cells. Importantly, myriocin and siSPT2 treatment restored insulin signaling in palmitate-treated GT1-7 cells. Protein kinase C (PKC) inhibitor or a dominant-negative PKCζ also counteracted palmitate-induced insulin resistance. Interestingly, attenuating the increase in levels of hypothalamic ceramides with intracerebroventricular infusion of myriocin in OZR improved their hypothalamic insulin-sensitivity. Importantly, central myriocin treatment partially restored glucose tolerance in OZR. This latter effect is related to the restoration of glucose-stimulated insulin secretion and an increase in β-cell mass of OZR. Electrophysiological recordings also showed an improvement of glucose-stimulated parasympathetic nerve activity in OZR centrally treated with myriocin. Conclusion Our results highlight a key role of hypothalamic de novo ceramide synthesis in central insulin resistance installation and glucose homeostasis dysregulation associated with obesity. de novo ceramide synthesis induces hypothalamic insulin resistance through PKCζ. Hypothalamic ceramides induce glucose homeostasis dysregulation seen with obesity. Hypothalamic ceramides mediate inhibition of insulin secretion induced by obesity. Hypothalamic ceramides decreases β cell mass in obese rats. Hypothalamic ceramides decreases parasympathetic tonus.
Collapse
|
25
|
Huang HH, Chen LY, Doong ML, Chang SC, Chen CY. α-melanocyte stimulating hormone modulates the central acyl ghrelin-induced stimulation of feeding, gastrointestinal motility, and colonic secretion. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:2377-2386. [PMID: 28860709 PMCID: PMC5566386 DOI: 10.2147/dddt.s143749] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background Acyl ghrelin-induced intake depends on hypothalamic neuropeptide Y and agouti-related protein (AgRP) neurotransmitters. Intracerebroventricular (ICV) injection of AgRP increases feeding through competitive antagonism at melanocortin receptors. ICV administration of α-melanocyte stimulating hormone (α-MSH), a natural antagonist of AgRP, may modulate the acyl ghrelin-induced orexigenic effect. Objective This study aimed to investigate the modulating effect of α-MSH on the central acyl ghrelin-induced food intake, gastrointestinal motility, and colonic secretion in rats. Methods and procedures We examined the effects of α-MSH and acyl ghrelin on food intake, gastric emptying, small intestinal transit, colonic motility, and secretion in conscious rats with a chronic implant of ICV catheters. Results ICV injection of O-n-octanoylated ghrelin (0.1 nmol/rat) significantly increased the cumulative food intake up to 8 h (P<0.01), enhanced non-nutrient semi-liquid gastric emptying (P<0.001), increased the geometric center and running percentage of small intestinal transit (P<0.001), accelerated colonic transit time (P<0.05), and increased fecal pellet output (P<0.01) and total fecal weight (P<0.01). Pretreatment with ICV injection of α-MSH (1.0 and 2.0 nmol/rat) attenuated the acyl ghrelin-induced hyperphagic effect, fecal pellet output, and total fecal weight, while higher dose of α-MSH (2.0 nmol/rat) attenuated the increase in the geometric center of small intestinal transit (P<0.01). However, neither dose of α-MSH altered acyl ghrelin-stimulated gastroprokinetic effect, increase in the running percentage of small intestinal transit, nor accelerated colonic transit time. Conclusion α-MSH is involved in central acyl ghrelin-elicited feeding, small intestinal transit, fecal pellet output, and fecal weight. α-MSH does not affect central acyl ghrelin-induced acceleration of gastric emptying and colonic transit time in rats.
Collapse
Affiliation(s)
- Hsien-Hao Huang
- Institute of Clinical Medicine, National Yang-Ming University of Medicine.,Department of Emergency Medicine, Taipei Veterans General Hospital
| | - Liang-Yu Chen
- Aging and Health Research Center, National Yang-Ming University.,Center for Geriatrics and Gerontology, Taipei Veterans General Hospital
| | - Ming-Luen Doong
- Institute of Physiology, National Yang-Ming University School of Medicine
| | - Shi-Chuan Chang
- Institute of Emergency and Critical Medicine, National Yang-Ming University School of Medicine.,Department of Chest Medicine, Taipei Veterans General Hospital
| | - Chih-Yen Chen
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital.,Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei.,Taiwan Association for the Study of Small Intestinal Diseases, Guishan, Taiwan
| |
Collapse
|
26
|
Modulatory function of NMDA glutamate receptor on MC3/MC4 receptors agonist-induced hypophagia in neonatal meat-type chicken. Vet Res Commun 2017; 41:241-248. [DOI: 10.1007/s11259-017-9693-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/24/2017] [Indexed: 10/19/2022]
|
27
|
Sun J, Gao Y, Yao T, Huang Y, He Z, Kong X, Yu KJ, Wang RT, Guo H, Yan J, Chang Y, Chen H, Scherer PE, Liu T, Williams KW. Adiponectin potentiates the acute effects of leptin in arcuate Pomc neurons. Mol Metab 2016; 5:882-891. [PMID: 27689001 PMCID: PMC5034606 DOI: 10.1016/j.molmet.2016.08.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/08/2016] [Accepted: 08/10/2016] [Indexed: 01/06/2023] Open
Abstract
Objective Adiponectin receptors (AdipoRs) are located on neurons of the hypothalamus involved in metabolic regulation – including arcuate proopiomelanocortin (Pomc) and Neuropeptide Y/Agouti-related peptide (NPY/AgRP) neurons. AdipoRs play a critical role in regulating glucose and fatty acid metabolism by initiating several signaling cascades overlapping with Leptin receptors (LepRs). However, the mechanism by which adiponectin regulates cellular activity in the brain remains undefined. Methods In order to resolve this issue, we utilized neuron-specific transgenic mouse models to identify Pomc and NPY/AgRP neurons which express LepRs for patch-clamp electrophysiology experiments. Results We found that leptin and adiponectin synergistically activated melanocortin neurons in the arcuate nucleus. Conversely, NPY/AgRP neurons were inhibited in response to adiponectin. The adiponectin-induced depolarization of arcuate Pomc neurons occurred via activation of Phosphoinositide-3-kinase (PI3K) signaling, independent of 5′ AMP-activated protein kinase (AMPK) activity. Adiponectin also activated melanocortin neurons at various physiological glucose levels. Conclusions Our results demonstrate a requirement for PI3K signaling in the acute adiponectin-induced effects on the cellular activity of arcuate melanocortin neurons. Moreover, these data provide evidence for PI3K as a substrate for both leptin and adiponectin to regulate energy balance and glucose metabolism via melanocortin activity. Adiponectin activates arcuate Pomc neurons. Adiponectin-induced activation of Pomc neurons requires PI3K (independent of AMPK). Adiponectin inhibits adjacent NPY/AgRP neurons (disinhibiting arcuate Pomc neurons). Leptin potentiates the effects of adiponectin arcuate Pomc neurons.
Collapse
Affiliation(s)
- Jia Sun
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Division of Hypothalamic Research, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Yong Gao
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Division of Hypothalamic Research, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Ting Yao
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China; Division of Hypothalamic Research, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Yiru Huang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Division of Hypothalamic Research, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Zhenyan He
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Division of Hypothalamic Research, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Xingxing Kong
- Division of Endocrinology, Beth Israel Deaconess Medical Center and Harvard Medical School, Harvard University, Boston, MA, 02115, USA
| | - Kai-Jiang Yu
- Department of Intensive Care Unit, The Third Affiliated Hospital, Harbin Medical University, No. 150 Haping St, Nangang District, Harbin, 150081, China
| | - Rui-Tao Wang
- Department of Intensive Care Unit, The Third Affiliated Hospital, Harbin Medical University, No. 150 Haping St, Nangang District, Harbin, 150081, China
| | - Hongbo Guo
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jianqun Yan
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| | - Yongsheng Chang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong Chen
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Tiemin Liu
- Department of Intensive Care Unit, The Third Affiliated Hospital, Harbin Medical University, No. 150 Haping St, Nangang District, Harbin, 150081, China; Division of Hypothalamic Research, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Kevin W Williams
- Division of Hypothalamic Research, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA.
| |
Collapse
|
28
|
Melanocortin 3 Receptor Signaling in Midbrain Dopamine Neurons Increases the Motivation for Food Reward. Neuropsychopharmacology 2016; 41:2241-51. [PMID: 26852738 PMCID: PMC4946052 DOI: 10.1038/npp.2016.19] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/31/2016] [Accepted: 02/01/2016] [Indexed: 01/21/2023]
Abstract
The central melanocortin (MC) system mediates its effects on food intake via MC3 (MC3R) and MC4 receptors (MC4R). Although the role of MC4R in meal size determination, satiation, food preference, and motivation is well established, the involvement of MC3R in the modulation of food intake has been less explored. Here, we investigated the role of MC3R on the incentive motivation for food, which is a crucial component of feeding behavior. Dopaminergic neurons within the ventral tegmental area (VTA) have a crucial role in the motivation for food. We here report that MC3Rs are expressed on VTA dopaminergic neurons and that pro-opiomelanocortinergic (POMC) neurons in the arcuate nucleus of the hypothalamus (Arc) innervate these VTA dopaminergic neurons. Our findings show that intracerebroventricular or intra-VTA infusion of the selective MC3R agonist γMSH increases responding for sucrose under a progressive ratio schedule of reinforcement, but not free sucrose consumption in rats. Furthermore, ex vivo electrophysiological recordings show increased VTA dopaminergic neuronal activity upon γMSH application. Consistent with a dopamine-mediated effect of γMSH, the increased motivation for sucrose after intra-VTA infusion of γMSH was blocked by pretreatment with the dopamine receptor antagonist α-flupenthixol. Taken together, we demonstrate an Arc POMC projection onto VTA dopaminergic neurons that modulates motivation for palatable food via activation of MC3R signaling.
Collapse
|
29
|
Stijnen P, Ramos-Molina B, O'Rahilly S, Creemers JWM. PCSK1 Mutations and Human Endocrinopathies: From Obesity to Gastrointestinal Disorders. Endocr Rev 2016; 37:347-71. [PMID: 27187081 DOI: 10.1210/er.2015-1117] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prohormone convertase 1/3, encoded by the PCSK1 gene, is a serine endoprotease that is involved in the processing of a variety of proneuropeptides and prohormones. Humans who are homozygous or compound heterozygous for loss-of-function mutations in PCSK1 exhibit a variable and pleiotropic syndrome consisting of some or all of the following: obesity, malabsorptive diarrhea, hypogonadotropic hypogonadism, altered thyroid and adrenal function, and impaired regulation of plasma glucose levels in association with elevated circulating proinsulin-to-insulin ratio. Recently, more common variants in the PCSK1 gene have been found to be associated with alterations in body mass index, increased circulating proinsulin levels, and defects in glucose homeostasis. This review provides an overview of the endocrinopathies and other disorders observed in prohormone convertase 1/3-deficient patients, discusses the possible biochemical basis for these manifestations of the disease, and proposes a model whereby certain missense mutations in PCSK1 may result in proteins with a dominant negative action.
Collapse
Affiliation(s)
- Pieter Stijnen
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Bruno Ramos-Molina
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Stephen O'Rahilly
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - John W M Creemers
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
30
|
Üner A, Gonçalves GH, Li W, Porceban M, Caron N, Schönke M, Delpire E, Sakimura K, Bjørbæk C. The role of GluN2A and GluN2B NMDA receptor subunits in AgRP and POMC neurons on body weight and glucose homeostasis. Mol Metab 2015; 4:678-91. [PMID: 26500840 PMCID: PMC4588453 DOI: 10.1016/j.molmet.2015.06.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 06/15/2015] [Accepted: 06/19/2015] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE Hypothalamic agouti-related peptide (AgRP) and pro-opiomelanocortin (POMC) expressing neurons play critical roles in control of energy balance. Glutamatergic input via n-methyl-d-aspartate receptors (NMDARs) is pivotal for regulation of neuronal activity and is required in AgRP neurons for normal body weight homeostasis. NMDARs typically consist of the obligatory GluN1 subunit and different GluN2 subunits, the latter exerting crucial differential effects on channel activity and neuronal function. Currently, the role of specific GluN2 subunits in AgRP and POMC neurons on whole body energy and glucose balance is unknown. METHODS We used the cre-lox system to genetically delete GluN2A or GluN2B only from AgRP or POMC neurons in mice. Mice were then subjected to metabolic analyses and assessment of AgRP and POMC neuronal function through morphological studies. RESULTS We show that loss of GluN2B from AgRP neurons reduces body weight, fat mass, and food intake, whereas GluN2B in POMC neurons is not required for normal energy balance control. GluN2A subunits in either AgRP or POMC neurons are not required for regulation of body weight. Deletion of GluN2B reduces the number of AgRP neurons and decreases their dendritic length. In addition, loss of GluN2B in AgRP neurons of the morbidly obese and severely diabetic leptin-deficient Lep (ob/ob) mice does not affect body weight and food intake but, remarkably, leads to full correction of hyperglycemia. Lep (ob/ob) mice lacking GluN2B in AgRP neurons are also more sensitive to leptin's anti-obesity actions. CONCLUSIONS GluN2B-containing NMDA receptors in AgRP neurons play a critical role in central control of body weight homeostasis and blood glucose balance via mechanisms that likely involve regulation of AgRP neuronal survival and structure, and modulation of hypothalamic leptin action.
Collapse
Key Words
- AAC, area above the curve
- AMPARs, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors
- ANCOVA, analysis of covariance
- ANOVA, analysis of variance
- AUC, area under the curve
- AgRP
- AgRP, agouti-related peptide
- CNS, central nervous system
- DAB, 3,3′-diaminobenzidine
- DIO, diet-induced obesity
- DREADD, Designer Receptor Exclusively Activated by Dedigner Drugs
- EPSCs, excitatory post-synaptic synaptic currents
- GABA, gamma-aminobutyric acid
- GTT, glucose tolerance test
- GluN2B
- Glycemia
- HFD, high-fat diet
- HSD, honestly significant difference
- ITT, insulin tolerance test
- KO, knockout
- LTD, long-term depression
- LTP, long-term potentiation
- Lepob/ob mice, obese leptin-deficient mice
- Leptin
- Metabolism
- NMDAR
- NMDARs, N-methyl-d-aspartate receptors
- PBS, phosphate-buffered saline
- POMC, pro-opiomelanocortin
- PVN, paraventricular nucleus
- RT, room temperature
- hrGFP, humanized renilla GFP
Collapse
Affiliation(s)
- Aykut Üner
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Gabriel H.M. Gonçalves
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Wenjing Li
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Matheus Porceban
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Nicole Caron
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Milena Schönke
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical School, Nashville, TN 37232, USA
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Christian Bjørbæk
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
31
|
Iemolo A, Ferragud A, Cottone P, Sabino V. Pituitary Adenylate Cyclase-Activating Peptide in the Central Amygdala Causes Anorexia and Body Weight Loss via the Melanocortin and the TrkB Systems. Neuropsychopharmacology 2015; 40:1846-55. [PMID: 25649277 PMCID: PMC4839508 DOI: 10.1038/npp.2015.34] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/10/2015] [Accepted: 01/11/2015] [Indexed: 02/03/2023]
Abstract
Growing evidence suggests that the pituitary adenylate cyclase-activating polypeptide (PACAP)/PAC1 receptor system represents one of the main regulators of the behavioral, endocrine, and autonomic responses to stress. Although induction of anorexia is a well-documented effect of PACAP, the central sites underlying this phenomenon are poorly understood. The present studies addressed this question by examining the neuroanatomical, behavioral, and pharmacological mechanisms mediating the anorexia produced by PACAP in the central nucleus of the amygdala (CeA), a limbic structure implicated in the emotional components of ingestive behavior. Male rats were microinfused with PACAP (0-1 μg per rat) into the CeA and home-cage food intake, body weight change, microstructural analysis of food intake, and locomotor activity were assessed. Intra-CeA (but not intra-basolateral amygdala) PACAP dose-dependently induced anorexia and body weight loss without affecting locomotor activity. PACAP-treated rats ate smaller meals of normal duration, revealing that PACAP slowed feeding within meals by decreasing the regularity and maintenance of feeding from pellet-to-pellet; postprandial satiety was unaffected. Intra-CeA PACAP-induced anorexia was blocked by coinfusion of either the melanocortin receptor 3/4 antagonist SHU 9119 or the tyrosine kinase B (TrKB) inhibitor k-252a, but not the CRF receptor antagonist D-Phe-CRF(12-41). These results indicate that the CeA is one of the brain areas through which the PACAP system promotes anorexia and that PACAP preferentially lessens the maintenance of feeding in rats, effects opposite to those of palatable food. We also demonstrate that PACAP in the CeA exerts its anorectic effects via local melanocortin and the TrKB systems, and independently from CRF.
Collapse
Affiliation(s)
- Attilio Iemolo
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA,Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Antonio Ferragud
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA,Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA,Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA,Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118 USA, Tel: +1 617 638 4327, Fax: +1 617 638 5668, E-mail: or
| | - Valentina Sabino
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA,Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA,Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118 USA, Tel: +1 617 638 4327, Fax: +1 617 638 5668, E-mail: or
| |
Collapse
|
32
|
Cheng ML, Wang HC, Hsu KC, Hwang JS. Anti-inflammatory peptides from enzymatic hydrolysates of tuna cooking juice. FOOD AGR IMMUNOL 2015. [DOI: 10.1080/09540105.2015.1036352] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- Mei-Li Cheng
- Department of Nursing, National Taichung University of Science and Technology, 129 Sec. 3, Sanmin Rd., Taichung 40401, Taiwan, Republic of China
| | - Hsuan-Chi Wang
- Department of Nursing, National Taichung University of Science and Technology, 129 Sec. 3, Sanmin Rd., Taichung 40401, Taiwan, Republic of China
| | - Kuo-Chiang Hsu
- School of Nutrition and Institute of Nutrition, China Medical University, No.110, Sec. 1, Chien-Kuo N. Rd, Taichung, Taiwan, Republic of China
| | - Jyh-Sheng Hwang
- Department of Food Science and Technology, Hungkuang University, No.1018, Sec. 6, Taiwan Boulevard, Shalu District, Taichung City 43302, Taiwan, Republic of China
| |
Collapse
|
33
|
Lam DD, Attard CA, Mercer AJ, Myers MG, Rubinstein M, Low MJ. Conditional expression of Pomc in the Lepr-positive subpopulation of POMC neurons is sufficient for normal energy homeostasis and metabolism. Endocrinology 2015; 156:1292-302. [PMID: 25594696 PMCID: PMC4399319 DOI: 10.1210/en.2014-1373] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Peptides derived from the proopiomelanocortin (POMC) precursor are critical for the normal regulation of many physiological parameters, and POMC deficiency results in severe obesity and metabolic dysfunction. Conversely, augmentation of central nervous system melanocortin function is a promising therapeutic avenue for obesity and diabetes but is confounded by detrimental cardiovascular effects including hypertension. Because the hypothalamic population of POMC-expressing neurons is neurochemically and neuroanatomically heterogeneous, there is interest in the possible dissociation of functionally distinct POMC neuron subpopulations. We used a Cre recombinase-dependent and hypothalamus-specific reactivatable PomcNEO allele to restrict Pomc expression to hypothalamic neurons expressing leptin receptor (Lepr) in mice. In contrast to mice with total hypothalamic Pomc deficiency, which are severely obese, mice with Lepr-restricted Pomc expression displayed fully normal body weight, food consumption, glucose homeostasis, and locomotor activity. Thus, Lepr+ POMC neurons, which constitute approximately two-thirds of the total POMC neuron population, are sufficient for normal regulation of these parameters. This functional dissociation approach represents a promising avenue for isolating therapeutically relevant POMC neuron subpopulations.
Collapse
Affiliation(s)
- Daniel D Lam
- Department of Molecular and Integrative Physiology (D.D.L., C.A.A., A.J.M., M.R., M.J.L.), and Department of Internal Medicine (M.G.M.), Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, Michigan 48105; and Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (M.R.), Consejo Nacional de Investigaciones Científicas y Técnicas and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, 1428 Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
34
|
Wang D, He X, Zhao Z, Feng Q, Lin R, Sun Y, Ding T, Xu F, Luo M, Zhan C. Whole-brain mapping of the direct inputs and axonal projections of POMC and AgRP neurons. Front Neuroanat 2015; 9:40. [PMID: 25870542 PMCID: PMC4375998 DOI: 10.3389/fnana.2015.00040] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/12/2015] [Indexed: 01/21/2023] Open
Abstract
Pro-opiomelanocortin (POMC) neurons in the arcuate nucleus (ARC) of the hypothalamus and nucleus tractus solitarius (NTS) of the brainstem play important roles in suppressing food intake and maintaining energy homeostasis. Previous tract-tracing studies have revealed the axonal connection patterns of these two brain areas, but the intermingling of POMC neurons with other neuron types has made it challenging to precisely identify the inputs and outputs of POMC neurons. In this study, we used the modified rabies virus to map the brain areas that provide direct inputs to the POMC neurons in the ARC and NTS as well as the inputs to the ARC AgRP neurons for comparison. ARC POMC neurons receive inputs from dozens of discrete structures throughout the forebrain and brainstem. The brain areas containing the presynaptic partners of ARC POMC neurons largely overlap with those of ARC AgRP neurons, although POMC neurons receive relatively broader, denser inputs. Furthermore, POMC neurons in the NTS receive direct inputs predominantly from the brainstem and show very different innervation patterns for POMC neurons in the ARC. By selectively expressing fluorescent markers in the ARC and NTS POMC neurons, we found that almost all of their major presynaptic partners are innervated by POMC neurons in the two areas, suggesting that there are strong reciprocal projections among the major POMC neural pathways. By comprehensively chartering the whole-brain connections of the central melanocortin system in a cell-type-specific manner, this study lays the foundation for dissecting the roles and underlying circuit mechanisms of specific neural pathways in regulating energy homeostasis.
Collapse
Affiliation(s)
- Daqing Wang
- School of Life Sciences, Tsinghua University Beijing China ; National Institute of Biological Sciences Beijing, China
| | - Xiaobing He
- Key Laboratory of Magnetic Resonance in Biological Systems and State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences Wuhan, China ; University of Chinese Academy of Sciences Beijing, China
| | - Zhe Zhao
- National Institute of Biological Sciences Beijing, China
| | - Qiru Feng
- National Institute of Biological Sciences Beijing, China
| | - Rui Lin
- National Institute of Biological Sciences Beijing, China
| | - Yue Sun
- National Institute of Biological Sciences Beijing, China
| | - Ting Ding
- Key Laboratory of Magnetic Resonance in Biological Systems and State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences Wuhan, China
| | - Fuqiang Xu
- Key Laboratory of Magnetic Resonance in Biological Systems and State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences Wuhan, China ; University of Chinese Academy of Sciences Beijing, China ; Wuhan National Laboratory for Optoelectronics Wuhan, China
| | - Minmin Luo
- School of Life Sciences, Tsinghua University Beijing China ; National Institute of Biological Sciences Beijing, China
| | - Cheng Zhan
- National Institute of Biological Sciences Beijing, China
| |
Collapse
|
35
|
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: 2.0] [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
|
36
|
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
|
37
|
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: 2.0] [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
|
38
|
Lau J, Herzog H. CART in the regulation of appetite and energy homeostasis. Front Neurosci 2014; 8:313. [PMID: 25352770 PMCID: PMC4195273 DOI: 10.3389/fnins.2014.00313] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/17/2014] [Indexed: 12/20/2022] Open
Abstract
The cocaine- and amphetamine-regulated transcript (CART) has been the subject of significant interest for over a decade. Work to decipher the detailed mechanism of CART function has been hampered by the lack of specific pharmacological tools like antagonists and the absence of a specific CART receptor(s). However, extensive research has been devoted to elucidate the role of the CART peptide and it is now evident that CART is a key neurotransmitter and hormone involved in the regulation of diverse biological processes, including food intake, maintenance of body weight, reward and addiction, stress response, psychostimulant effects and endocrine functions (Rogge et al., 2008; Subhedar et al., 2014). In this review, we focus on knowledge gained on CART's role in controlling appetite and energy homeostasis, and also address certain species differences between rodents and humans.
Collapse
Affiliation(s)
- Jackie Lau
- Neuroscience Division, Garvan Institute of Medical Research Sydney, NSW, Australia
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research Sydney, NSW, Australia
| |
Collapse
|
39
|
Lira LA, Almeida LC, Silva AA, Cavalcante TC, Melo DD, Souza JA, Campina RC, Souza SL. Perinatal undernutrition increases meal size and neuronal activation of the nucleus of the solitary tract in response to feeding stimulation in adult rats. Int J Dev Neurosci 2014; 38:23-9. [DOI: 10.1016/j.ijdevneu.2014.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Revised: 07/27/2014] [Accepted: 07/27/2014] [Indexed: 10/24/2022] Open
Affiliation(s)
- Lívia A. Lira
- Postgraduate Neuropsychiatry and Behavioral SciencesUniversidade Federal Pernambuco – UFPERecifePEBrazil
| | - Larissa C.A. Almeida
- Postgraduate Neuropsychiatry and Behavioral SciencesUniversidade Federal Pernambuco – UFPERecifePEBrazil
| | - Amanda A.M. Silva
- Postgraduate Neuropsychiatry and Behavioral SciencesUniversidade Federal Pernambuco – UFPERecifePEBrazil
| | | | - Diogo D.C.B. Melo
- Postgraduate Neuropsychiatry and Behavioral SciencesUniversidade Federal Pernambuco – UFPERecifePEBrazil
| | - Julliet A. Souza
- Department of NutritionUniversidade Federal de Pernambuco – UFPERecifePEBrazil
| | - Renata C.F. Campina
- Department of AnatomyUniversidade Federal de Pernambuco – UFPERecifePEBrazil
- Postgraduate Neuropsychiatry and Behavioral SciencesUniversidade Federal Pernambuco – UFPERecifePEBrazil
| | - Sandra L. Souza
- Department of AnatomyUniversidade Federal de Pernambuco – UFPERecifePEBrazil
- Postgraduate Neuropsychiatry and Behavioral SciencesUniversidade Federal Pernambuco – UFPERecifePEBrazil
- Department of NutritionUniversidade Federal de Pernambuco – UFPERecifePEBrazil
| |
Collapse
|
40
|
Guan X. The CNS glucagon-like peptide-2 receptor in the control of energy balance and glucose homeostasis. Am J Physiol Regul Integr Comp Physiol 2014; 307:R585-96. [PMID: 24990862 DOI: 10.1152/ajpregu.00096.2014] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The gut-brain axis plays a key role in the control of energy balance and glucose homeostasis. In response to luminal stimulation of macronutrients and microbiota-derived metabolites (secondary bile acids and short chain fatty acids), glucagon-like peptides (GLP-1 and -2) are cosecreted from endocrine L cells in the gut and coreleased from preproglucagonergic neurons in the brain stem. Glucagon-like peptides are proposed as key mediators for bariatric surgery-improved glycemic control and energy balance. Little is known about the GLP-2 receptor (Glp2r)-mediated physiological roles in the control of food intake and glucose homeostasis, yet Glp1r has been studied extensively. This review will highlight the physiological relevance of the central nervous system (CNS) Glp2r in the control of energy balance and glucose homeostasis and focuses on cellular mechanisms underlying the CNS Glp2r-mediated neural circuitry and intracellular PI3K signaling pathway. New evidence (obtained from Glp2r tissue-specific KO mice) indicates that the Glp2r in POMC neurons is essential for suppressing feeding behavior, gastrointestinal motility, and hepatic glucose production. Mice with Glp2r deletion selectively in POMC neurons exhibit hyperphagic behavior, accelerated gastric emptying, glucose intolerance, and hepatic insulin resistance. GLP-2 differentially modulates postsynaptic membrane excitability of hypothalamic POMC neurons in Glp2r- and PI3K-dependent manners. GLP-2 activates the PI3K-Akt-FoxO1 signaling pathway in POMC neurons by Glp2r-p85α interaction. Intracerebroventricular GLP-2 augments glucose tolerance, suppresses glucose production, and enhances insulin sensitivity, which require PI3K (p110α) activation in POMC neurons. Thus, the CNS Glp2r plays a physiological role in the control of food intake and glucose homeostasis. This review will also discuss key questions for future studies.
Collapse
Affiliation(s)
- Xinfu Guan
- U.S. Department of Agriculture/Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics; and Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
41
|
Eerola K, Rinne P, Penttinen AM, Vähätalo L, Savontaus M, Savontaus E. α-MSH overexpression in the nucleus tractus solitarius decreases fat mass and elevates heart rate. J Endocrinol 2014; 222:123-36. [PMID: 24829220 DOI: 10.1530/joe-14-0064] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The POMC pathway is involved in the regulation of energy and cardiovascular homeostasis in the hypothalamus and the brain stem. Although the acute effects of POMC-derived peptides in different brain locations have been elucidated, the chronic site-specific effects of distinct peptides remain to be studied. To this end, we used a lentiviral gene delivery vector to study the long-term effects of α-MSH in the nucleus tractus solitarius (NTS) of the brain stem. The α-MSH vector (LVi-α-MSH-EGFP) based on the N-terminal POMC sequence and a control vector (LVi-EGFP) were delivered into the NTS of C57BL/6N male mice fed on a western diet. Effects on body weight and composition, feeding, glucose metabolism, and hemodynamics by telemetric analyses were studied during the 12-week follow-up. The LVi-α-MSH-EGFP-treated mice had a significantly smaller gain in the fat mass compared with LVi-EGFP-injected mice. There was a small initial decrease in food intake and no differences in the physical activity. Glucose metabolism was not changed compared with the control. LVi-α-MSH-EGFP increased the heart rate (HR), which was attenuated by adrenergic blockade suggesting an increased sympathetic activity. Reduced response to muscarinic blockade suggested a decreased parasympathetic activity. Fitting with sympathetic activation, LVi-α-MSH-EGFP treatment reduced urine secretion. Thus, the results demonstrate that long-term α-MSH overexpression in the NTS attenuates diet-induced obesity. Modulation of autonomic nervous system tone increased the HR and most probably contributed to an anti-obesity effect. The results underline the key role of NTS in the α-MSH-induced long-term effects on adiposity and in regulation of sympathetic and parasympathetic activities.
Collapse
Affiliation(s)
- K Eerola
- Department of PharmacologyDrug Development and Therapeutics and Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520 Turku, FinlandTurku Centre for BiotechnologyUniversity of Turku, Turku, FinlandDrug Research Doctoral ProgramUniversity of Turku, Turku, FinlandHeart CenterTurku University Hospital and University of Turku, Turku, FinlandUnit of Clinical PharmacologyTurku University Hospital, Turku, FinlandDepartment of PharmacologyDrug Development and Therapeutics and Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520 Turku, FinlandTurku Centre for BiotechnologyUniversity of Turku, Turku, FinlandDrug Research Doctoral ProgramUniversity of Turku, Turku, FinlandHeart CenterTurku University Hospital and University of Turku, Turku, FinlandUnit of Clinical PharmacologyTurku University Hospital, Turku, FinlandDepartment of PharmacologyDrug Development and Therapeutics and Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520 Turku, FinlandTurku Centre for BiotechnologyUniversity of Turku, Turku, FinlandDrug Research Doctoral ProgramUniversity of Turku, Turku, FinlandHeart CenterTurku University Hospital and University of Turku, Turku, FinlandUnit of Clinical PharmacologyTurku University Hospital, Turku, Finland
| | - P Rinne
- Department of PharmacologyDrug Development and Therapeutics and Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520 Turku, FinlandTurku Centre for BiotechnologyUniversity of Turku, Turku, FinlandDrug Research Doctoral ProgramUniversity of Turku, Turku, FinlandHeart CenterTurku University Hospital and University of Turku, Turku, FinlandUnit of Clinical PharmacologyTurku University Hospital, Turku, Finland
| | - A M Penttinen
- Department of PharmacologyDrug Development and Therapeutics and Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520 Turku, FinlandTurku Centre for BiotechnologyUniversity of Turku, Turku, FinlandDrug Research Doctoral ProgramUniversity of Turku, Turku, FinlandHeart CenterTurku University Hospital and University of Turku, Turku, FinlandUnit of Clinical PharmacologyTurku University Hospital, Turku, Finland
| | - L Vähätalo
- Department of PharmacologyDrug Development and Therapeutics and Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520 Turku, FinlandTurku Centre for BiotechnologyUniversity of Turku, Turku, FinlandDrug Research Doctoral ProgramUniversity of Turku, Turku, FinlandHeart CenterTurku University Hospital and University of Turku, Turku, FinlandUnit of Clinical PharmacologyTurku University Hospital, Turku, FinlandDepartment of PharmacologyDrug Development and Therapeutics and Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520 Turku, FinlandTurku Centre for BiotechnologyUniversity of Turku, Turku, FinlandDrug Research Doctoral ProgramUniversity of Turku, Turku, FinlandHeart CenterTurku University Hospital and University of Turku, Turku, FinlandUnit of Clinical PharmacologyTurku University Hospital, Turku, Finland
| | - M Savontaus
- Department of PharmacologyDrug Development and Therapeutics and Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520 Turku, FinlandTurku Centre for BiotechnologyUniversity of Turku, Turku, FinlandDrug Research Doctoral ProgramUniversity of Turku, Turku, FinlandHeart CenterTurku University Hospital and University of Turku, Turku, FinlandUnit of Clinical PharmacologyTurku University Hospital, Turku, FinlandDepartment of PharmacologyDrug Development and Therapeutics and Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520 Turku, FinlandTurku Centre for BiotechnologyUniversity of Turku, Turku, FinlandDrug Research Doctoral ProgramUniversity of Turku, Turku, FinlandHeart CenterTurku University Hospital and University of Turku, Turku, FinlandUnit of Clinical PharmacologyTurku University Hospital, Turku, Finland
| | - E Savontaus
- Department of PharmacologyDrug Development and Therapeutics and Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520 Turku, FinlandTurku Centre for BiotechnologyUniversity of Turku, Turku, FinlandDrug Research Doctoral ProgramUniversity of Turku, Turku, FinlandHeart CenterTurku University Hospital and University of Turku, Turku, FinlandUnit of Clinical PharmacologyTurku University Hospital, Turku, FinlandDepartment of PharmacologyDrug Development and Therapeutics and Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520 Turku, FinlandTurku Centre for BiotechnologyUniversity of Turku, Turku, FinlandDrug Research Doctoral ProgramUniversity of Turku, Turku, FinlandHeart CenterTurku University Hospital and University of Turku, Turku, FinlandUnit of Clinical PharmacologyTurku University Hospital, Turku, Finland
| |
Collapse
|
42
|
de Carvalho Borges B, Rorato R, Uchoa ET, Marangon P, da Silva GSF, de Paula FJ, Branco LGS, Antunes-Rodrigues J, Elias LLK. High-fat diet induces site-specific unresponsiveness to LPS-stimulated STAT3 activation in the hypothalamus. Am J Physiol Regul Integr Comp Physiol 2014; 306:R34-44. [DOI: 10.1152/ajpregu.00147.2013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Hypophagia induced by inflammation is associated with Janus kinase (JAK)-2/signal transducer and activator of transcription (STAT) 3 signaling pathway, and leptin-mediated hypophagia is also mediated by JAK2-STAT3 pathway. We have previously reported that lipopolysaccharide (LPS) did not reduce food intake in leptin-resistant high-fat diet (HFD) rats but maintained body weight loss. We investigated whether changes in p-STAT3 expression in the hypothalamus and brain stem could account for the desensitization of hypophagia in HFD animals after a low LPS dose (100 μg/kg). Wistar rats fed standard diet (3.95 kcal/g) or HFD (6.3 kcal/g) for 8 wk were assigned into control diet-saline, control diet-LPS, HFD-saline, and HFD-LPS groups. LPS reduced feeding in the control diet but not HFD. This group showed no p-STAT3 expression in the paraventricular nucleus (PVN) and ventromedial hypothalamic nucleus (VMH), but sustained, though lower than control, p-STAT3 in the nucleus of the solitary tract (NTS) and raphe pallidus (RPa). LPS decreased body weight in HFD rats and increased Fos expression in the NTS. LPS increased body temperature, oxygen consumption, and energy expenditure in both control diet and HFD rats, and this response was more pronounced in HFD-LPS group. Brown adipose tissue (BAT) thermogenesis and increased energy expenditure seem to contribute to body weight loss in HFD-LPS. This response might be related with increased brain stem activation. In conclusion, LPS activates STAT3-mediated pathway in the hypothalamus and brain stem, leading to hypophagia, however, LPS effects on food intake, but not body weight loss, are abolished by leptin resistance induced by HFD. The preserved STAT3 phosphorylation in the brain stem suggests that unresponsiveness to LPS on STAT3 activation under HFD might be selective to the hypothalamus.
Collapse
Affiliation(s)
| | | | | | | | - Glauber S. F. da Silva
- Department of Physiology and
- Department of Morphology, Physiology and Basic Pathology, Dental School of Ribeirao Preto, University of Sao Paulo, Brazil
| | - Francisco José de Paula
- Department of Internal Medicine, School of Medicine of Ribeirao Preto, University of Sao Paulo, Brazil; and
| | - Luiz G. S. Branco
- Department of Morphology, Physiology and Basic Pathology, Dental School of Ribeirao Preto, University of Sao Paulo, Brazil
| | | | | |
Collapse
|
43
|
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
|
44
|
Abstract
Autophagy is a catabolic process involving the rearrangement of subcellular membranes to sequester cytoplasm and organelles for delivery to lysosomes, where the sequestered material is degraded and recycled. Autophagy is important for maintenance of intracellular energy homeostasis and the quality control of organelles such as the endoplasmic reticulum (ER) and mitochondria, which suggests that dysregulated autophagy might play a role in the pathogenesis of metabolic disorders and diabetes. In an attempt to elucidate the role of autophagy in metabolic disorders, diverse in vivo and in vitro models have been employed. Site-specific autophagy knockout models that are autophagy-deficient specifically in pancreatic β-cells, skeletal muscle, adipose tissues or liver have been produced. These models have generated valuable information regarding the role of autophagy in body metabolism. The role of autophagy in the hypothalamus, which controls whole body energy balance, appetite and energy expenditure, has also been investigated. Thus, mice with autophagy deficiency in the hypothalamus have shown diverse phenotypes (lean vs. obese) depending on the site of autophagy deficiency or the method of autophagy abrogation.
Collapse
Affiliation(s)
- Min-Seon Kim
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 138-736, Korea
| | | | | |
Collapse
|
45
|
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
|
46
|
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: 1.7] [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
|
47
|
Mul JD, Spruijt BM, Brakkee JH, Adan RAH. Melanocortin MC(4) receptor-mediated feeding and grooming in rodents. Eur J Pharmacol 2013; 719:192-201. [PMID: 23872405 DOI: 10.1016/j.ejphar.2013.04.060] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 03/27/2013] [Accepted: 04/03/2013] [Indexed: 02/04/2023]
Abstract
Decades ago it was recognized that the pharmacological profile of melanocortin ligands that stimulated grooming behavior in rats was strikingly similar to that of Xenopus laevis melanophore pigment dispersion. After cloning of the melanocortin MC1 receptor, expressed in melanocytes, and the melanocortin MC4 receptor, expressed mainly in brain, the pharmacological profiles of these receptors appeared to be very similar and it was demonstrated that these receptors mediate melanocortin-induced pigmentation and grooming respectively. Grooming is a low priority behavior that is concerned with care of body surface. Activation of central melanocortin MC4 receptors is also associated with meal termination, and continued postprandial stimulation of melanocortin MC4 receptors may stimulate natural postprandial grooming behavior as part of the behavioral satiety sequence. Indeed, melanocortins fail to suppress food intake or induce grooming behavior in melanocortin MC4 receptor-deficient rats. This review will focus on how melanocortins affect grooming behavior through the melanocortin MC4 receptor, and how melanocortin MC4 receptors mediate feeding behavior. This review also illustrates how melanocortins were the most likely candidates to mediate grooming and feeding based on the natural behaviors they induced.
Collapse
Affiliation(s)
- Joram D Mul
- Metabolic Diseases Institute, University of Cincinnati, 2170 East Galbraith Road, 45237 Cincinnati, Ohio, USA.
| | - Berry M Spruijt
- Department of Biology, Faculty of Beta Sciences, University of Utrecht, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Jan H Brakkee
- Rudolf Magnus Institute of Neuroscience, Department of Neuroscience and Pharmacology, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | - Roger A H Adan
- Rudolf Magnus Institute of Neuroscience, Department of Neuroscience and Pharmacology, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
48
|
Acute and long-term suppression of feeding behavior by POMC neurons in the brainstem and hypothalamus, respectively. J Neurosci 2013; 33:3624-32. [PMID: 23426689 DOI: 10.1523/jneurosci.2742-12.2013] [Citation(s) in RCA: 312] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
POMC-derived melanocortins inhibit food intake. In the adult rodent brain, POMC-expressing neurons are located in the arcuate nucleus (ARC) and the nucleus tractus solitarius (NTS), but it remains unclear how POMC neurons in these two brain nuclei regulate feeding behavior and metabolism differentially. Using pharmacogenetic methods to activate or deplete neuron groups in separate brain areas, in the present study, we show that POMC neurons in the ARC and NTS suppress feeding behavior at different time scales. Neurons were activated using the DREADD (designer receptors exclusively activated by designer drugs) method. The evolved human M3-muscarinic receptor was expressed in a selective population of POMC neurons by stereotaxic infusion of Cre-recombinase-dependent, adeno-associated virus vectors into the ARC or NTS of POMC-Cre mice. After injection of the human M3-muscarinic receptor ligand clozapine-N-oxide (1 mg/kg, i.p.), acute activation of NTS POMC neurons produced an immediate inhibition of feeding behavior. In contrast, chronic stimulation was required for ARC POMC neurons to suppress food intake. Using adeno-associated virus delivery of the diphtheria toxin receptor gene, we found that diphtheria toxin-induced ablation of POMC neurons in the ARC but not the NTS, increased food intake, reduced energy expenditure, and ultimately resulted in obesity and metabolic and endocrine disorders. Our results reveal different behavioral functions of POMC neurons in the ARC and NTS, suggesting that POMC neurons regulate feeding and energy homeostasis by integrating long-term adiposity signals from the hypothalamus and short-term satiety signals from the brainstem.
Collapse
|
49
|
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]
|
50
|
Roseberry AG. Altered feeding and body weight following melanocortin administration to the ventral tegmental area in adult rats. Psychopharmacology (Berl) 2013; 226:25-34. [PMID: 23010797 DOI: 10.1007/s00213-012-2879-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 09/07/2012] [Indexed: 12/01/2022]
Abstract
RATIONALE The melanocortin system is an important component of the brain circuitry controlling feeding and body weight, and most of the effects of melanocortins are attributed to their actions in hypothalamic and brainstem nuclei. The mesolimbic dopamine system is another component of the central circuitry controlling feeding, and there is evidence that melanocortins can act on mesolimbic dopamine pathways. It is unknown, however, whether melanocortins can act on the mesolimbic dopamine system to regulate feeding. OBJECTIVE These studies tested whether injection of melanocortin receptor agonists and antagonists directly into the ventral tegmental area (VTA) of adult rats affects feeding and body weight. METHODS Varying doses of the melanocortin receptor agonist, MTII, or the melanocortin receptor antagonist, SHU9119, were injected directly into the VTA, and food intake was measured at specific intervals. In addition, melanocortin receptors in the VTA were chronically blocked through repeated daily injections of SHU9119 into the VTA, and the resulting effects on food intake and body weight were determined. RESULTS Injection of MTII into the VTA dose-dependently inhibited feeding for up to 24 h, while injection of SHU9119 into the VTA dose-dependently stimulated feeding for up to 24 h. In addition, chronic blockade of melanocortin receptors in the VTA increased feeding, body weight, and caloric efficiency. CONCLUSIONS These studies demonstrate that melanocortins can control feeding and body weight by acting in the VTA and suggest that endogenous melanocortins control feeding in part through actions on the mesolimbic dopamine system in vivo.
Collapse
Affiliation(s)
- Aaron G Roseberry
- Department of Biology, Georgia State University, 24 Peachtree Center Ave NE, Suite 430SA, Atlanta, GA 30303, USA.
| |
Collapse
|