1
|
Fang LZ, Hirasawa M. Refining the role of the paraventricular hypothalamus in feeding behavior. Proc Natl Acad Sci U S A 2025; 122:e2506038122. [PMID: 40324098 DOI: 10.1073/pnas.2506038122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025] Open
Affiliation(s)
- Lisa Z Fang
- Department of Anesthesiology, Washington University in St Louis, St Louis, MO 63110
| | - Michiru Hirasawa
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| |
Collapse
|
2
|
Kongstorp M, Karnani MM, McCutcheon JE. Does the lateral hypothalamus govern the transition between appetitive and consummatory feeding? Neuropharmacology 2025; 275:110438. [PMID: 40194590 DOI: 10.1016/j.neuropharm.2025.110438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 04/09/2025]
Abstract
Feeding is a cyclic behaviour that includes appetitive, consummatory and termination phases. Identifying the neural circuits controlling these phases and triggering specific transitions between phases would be a key advance in understanding feeding behaviour. The lateral hypothalamus (LH) has long been recognized for its central role in feeding. We review evidence suggesting that the LH acts as a regulator of the appetitive-consummatory transition using a switchboard-like circuit architecture. Within the LH, several neuronal subpopulations can be defined based on molecular markers, and - although these subpopulations are functionally diverse - they contribute to appetitive and consummatory behaviours to varying extents. We summarise the current evidence on whether these subpopulations have functional identities and speculate on the role of the LH as a controller of behavioural transitions.
Collapse
Affiliation(s)
- Mette Kongstorp
- Department of Psychology, UiT The Arctic University of Norway, Huginbakken 32, 9037, Tromsø, Norway
| | - Mahesh M Karnani
- Centre for Discovery Brain Sciences, University of Edinburgh, 1 George Square, Edinburgh, EH8 9JZ, UK
| | - James E McCutcheon
- Department of Psychology, UiT The Arctic University of Norway, Huginbakken 32, 9037, Tromsø, Norway.
| |
Collapse
|
3
|
Iremonger KJ, Power EM. The paraventricular nucleus of the hypothalamus: a key node in the control of behavioural states. J Physiol 2025; 603:2231-2243. [PMID: 40119815 PMCID: PMC12013795 DOI: 10.1113/jp288366] [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: 01/07/2025] [Accepted: 03/05/2025] [Indexed: 03/24/2025] Open
Abstract
The paraventricular nucleus (PVN) of the hypothalamus contains diverse populations of neuropeptide-producing neurons. These include neurons that synthesise oxytocin, vasopressin, corticotropin-releasing hormone, thyrotropin-releasing hormone and somatostatin. While it is well established that these neurons control the secretion of neuroendocrine hormones, there is growing evidence that they also control the expression of important homeostatic behaviours. Here we review recent data showing a critical role of PVN neurons in controlling arousal, social behaviour, defensive behaviour and pain. Collectively, this suggests that the PVN is a key node in a wider neural network controlling behavioural states.
Collapse
Affiliation(s)
- Karl J. Iremonger
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| | - Emmet M. Power
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| |
Collapse
|
4
|
Haddad HK, Mercado-Reyes JI, Mustafá ER, D’Souza SP, Chung CS, Nestor RRM, Olinski LE, Martinez Damonte V, Saskin J, Vemaraju S, Raingo J, Kauer JA, Lang RA, Oancea E. Hypothalamic opsin 3 suppresses MC4R signaling and potentiates Kir7.1 to promote food consumption. Proc Natl Acad Sci U S A 2025; 122:e2403891122. [PMID: 39951488 PMCID: PMC11874419 DOI: 10.1073/pnas.2403891122] [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: 02/23/2024] [Accepted: 12/02/2024] [Indexed: 02/16/2025] Open
Abstract
Mammalian opsin 3 (OPN3) is a member of the opsin family of G-protein-coupled receptors with ambiguous light sensitivity. OPN3 was first identified in the brain (and named encephalopsin) and subsequently found to be expressed in other tissues. In adipocytes, OPN3 is necessary for light responses that modulate lipolysis and glucose uptake, while OPN3 in human skin melanocytes regulates pigmentation in a light-independent manner. Despite its initial discovery in the brain, OPN3 functional mechanisms in the brain remain elusive. Here, we investigated the molecular mechanism of OPN3 function in the paraventricular nucleus (PVN) of the hypothalamus. We show that Opn3 is coexpressed with the melanocortin 4 receptor (Mc4r) in a population of PVN neurons, where it negatively regulates MC4R-mediated cAMP signaling in a specific and Gαi/o-dependent manner. Under baseline conditions, OPN3 via Gαi/o potentiates the activity of the inward rectifying Kir7.1 channel, previously shown to be closed in response to agonist-mediated activation of MC4R in a Gαs-independent manner. In mice, we found that Opn3 in Mc4r-expressing neurons regulates food consumption. Our results reveal the first mechanistic insight into OPN3 function in the hypothalamus, uncovering a unique mechanism by which OPN3 functions to potentiate Kir7.1 activity and negatively regulate MC4R-mediated cAMP signaling, thereby promoting food intake.
Collapse
Affiliation(s)
- Hala K. Haddad
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Jonathan I. Mercado-Reyes
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - E. Román Mustafá
- Electrophysiology Lab, Instituto Multidisciplinario de Biología Celular, La Plata, Buenos Aires1900, Argentina
| | - Shane P. D’Souza
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - C. Sean Chung
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Ramses R. M. Nestor
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Lauren E. Olinski
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Valentina Martinez Damonte
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
| | - Joshua Saskin
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Shruti Vemaraju
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Jesica Raingo
- Electrophysiology Lab, Instituto Multidisciplinario de Biología Celular, La Plata, Buenos Aires1900, Argentina
| | - Julie A. Kauer
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
| | - Richard A. Lang
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Department of Ophthalmology, University of Cincinnati, Cincinnati, OH45229
| | - Elena Oancea
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| |
Collapse
|
5
|
Qiu T, Fu O. GPCRs in hypothalamic neurons and their roles in controlling food intake and metabolism. Front Mol Neurosci 2025; 18:1536577. [PMID: 39974186 PMCID: PMC11835808 DOI: 10.3389/fnmol.2025.1536577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/24/2025] [Indexed: 02/21/2025] Open
Abstract
G-protein coupled receptor (GPCR) subtypes within the hypothalamus play a pivotal role in maintaining body homeostasis, particularly in the regulation of food intake and energy metabolism. This review provides an overview of classical loss and gain-of-function studies on GPCRs related to feeding and metabolism, with a focus on emerging cell-type-specific investigations. These studies reveal that diverse GPCR-expressing neuronal populations are intricately linked to feeding and energy balance. We also discuss recent findings that highlight the interaction of distinct peptide-GPCR systems in modulating complex feeding behaviors.
Collapse
Affiliation(s)
- Tian Qiu
- School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
- Laboratory of Food Perception Science, Science Center for Future Foods, Jiangnan University, Wuxi, Jiangsu, China
| | - Ou Fu
- School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
- Laboratory of Food Perception Science, Science Center for Future Foods, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
6
|
Duran M, Willis JR, Dalvi N, Fokakis Z, Virkus SA, Hardaway JA. Integration of Glucagon-Like Peptide 1 Receptor Actions Through the Central Amygdala. Endocrinology 2025; 166:bqaf019. [PMID: 39888375 PMCID: PMC11850305 DOI: 10.1210/endocr/bqaf019] [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: 10/16/2024] [Revised: 12/31/2024] [Accepted: 01/25/2025] [Indexed: 02/01/2025]
Abstract
Understanding the detailed mechanism of action of glucagon-like peptide 1 receptor (GLP-1R) agonists on distinct topographic and genetically defined brain circuits is critical for improving the efficacy and mitigating adverse side effects of these compounds. In this mini-review, we propose that the central nucleus of the amygdala (CeA) is a critical mediator of GLP-1R agonist-driven hypophagia. Here, we review the extant literature demonstrating CeA activation via GLP-1R agonists across multiple species and through multiple routes of administration. The precise role of GLP-1Rs within the CeA is unclear but the site-specific GLP-1Rs may mediate distinct behavioral and physiological hallmarks of GLP-1R agonists on food intake. Thus, we propose important novel directions and methods to test the role of the CeA in mediating GLP-1R actions.
Collapse
Affiliation(s)
- Miguel Duran
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer R Willis
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nilay Dalvi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Zoe Fokakis
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sonja A Virkus
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - J Andrew Hardaway
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
7
|
Gruber T, Lechner F, Krieger JP, García-Cáceres C. Neuroendocrine gut-brain signaling in obesity. Trends Endocrinol Metab 2025; 36:42-54. [PMID: 38821753 DOI: 10.1016/j.tem.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 06/02/2024]
Abstract
The past decades have witnessed the rise and fall of several, largely unsuccessful, therapeutic attempts to bring the escalating obesity pandemic to a halt. Looking back to look ahead, the field has now put its highest hopes in translating insights from how the gastrointestinal (GI) tract communicates with the brain to calibrate behavior, physiology, and metabolism. A major focus of this review is to summarize the latest advances in comprehending the neuroendocrine aspects of this so-called 'gut-brain axis' and to explore novel concepts, cutting-edge technologies, and recent paradigm-shifting experiments. These exciting insights continue to refine our understanding of gut-brain crosstalk and are poised to promote the development of additional therapeutic avenues at the dawn of a new era of antiobesity therapeutics.
Collapse
Affiliation(s)
- Tim Gruber
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49506, USA; Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49506, USA; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.
| | - Franziska Lechner
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany
| | - Jean-Philippe Krieger
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland; Institute of Neuroscience and Physiology, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Cristina García-Cáceres
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
| |
Collapse
|
8
|
Zhang SX, Kim A, Madara JC, Zhu PK, Christenson LF, Lutas A, Kalugin PN, Sunkavalli PS, Jin Y, Pal A, Tian L, Lowell BB, Andermann ML. Stochastic neuropeptide signals compete to calibrate the rate of satiation. Nature 2025; 637:137-144. [PMID: 39506113 PMCID: PMC11981016 DOI: 10.1038/s41586-024-08164-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/07/2024] [Indexed: 11/08/2024]
Abstract
Neuropeptides have important roles in neural plasticity, spiking and behaviour1. Yet, many fundamental questions remain regarding their spatiotemporal transmission, integration and functions in the awake brain. Here we examined how MC4R-expressing neurons in the paraventricular nucleus of the hypothalamus (PVHMC4R) integrate neuropeptide signals to modulate feeding-related fast synaptic transmission and titrate the transition to satiety2-6. We show that hunger-promoting AgRP axons release the neuropeptide NPY to decrease the second messenger cAMP in PVHMC4R neurons, while satiety-promoting POMC axons release the neuropeptide αMSH to increase cAMP. Each release event is all-or-none, stochastic and can impact multiple neurons within an approximately 100-µm-diameter region. After release, NPY and αMSH peptides compete to control cAMP-the amplitude and persistence of NPY signalling is blunted by high αMSH in the fed state, while αMSH signalling is blunted by high NPY in the fasted state. Feeding resolves this competition by simultaneously elevating αMSH release and suppressing NPY release7,8, thereby sustaining elevated cAMP in PVHMC4R neurons throughout a meal. In turn, elevated cAMP facilitates potentiation of feeding-related excitatory inputs with each bite to gradually promote satiation across many minutes. Our findings highlight biochemical modes of peptide signal integration and information accumulation to guide behavioural state transitions.
Collapse
Affiliation(s)
- Stephen X Zhang
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Angela Kim
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Joseph C Madara
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Paula K Zhu
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lauren F Christenson
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Andrew Lutas
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter N Kalugin
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Praneel S Sunkavalli
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yihan Jin
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Neuroscience Graduate Group, Center for Neuroscience, University of California, Davis, Sacramento, CA, USA
- Max Planck Florida Institute for Neuroscience, One Max Planck Way, Jupiter, FL, USA
| | - Akash Pal
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Max Planck Florida Institute for Neuroscience, One Max Planck Way, Jupiter, FL, USA
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Max Planck Florida Institute for Neuroscience, One Max Planck Way, Jupiter, FL, USA
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mark L Andermann
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
9
|
Mahishi D, Agrawal N, Jiang W, Yapici N. From Mammals to Insects: Exploring the Genetic and Neural Basis of Eating Behavior. Annu Rev Genet 2024; 58:455-485. [PMID: 39585905 DOI: 10.1146/annurev-genet-111523-102202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Obesity and anorexia are life-threatening diseases that are still poorly understood at the genetic and neuronal levels. Patients suffering from these conditions experience disrupted regulation of food consumption, leading to extreme weight gain or loss and, in severe situations, death from metabolic dysfunction. Despite the development of various behavioral and pharmacological interventions, current treatments often yield limited and short-lived success. To address this, a deeper understanding of the genetic and neural mechanisms underlying food perception and appetite regulation is essential for identifying new drug targets and developing more effective treatment methods. This review summarizes the progress of past research in understanding the genetic and neural mechanisms controlling food consumption and appetite regulation, focusing on two key model organisms: the fruit fly Drosophila melanogaster and the mouse Mus musculus. These studies investigate how the brain senses energy and nutrient deficiency, how sensory signals trigger appetitive behaviors, and how food intake is regulated through interconnected neural circuits in the brain.
Collapse
Affiliation(s)
- Deepthi Mahishi
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| | - Naman Agrawal
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| | - Wenshuai Jiang
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| | - Nilay Yapici
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| |
Collapse
|
10
|
Altafi M, Chen C, Korotkova T, Ponomarenko A. Sequential Activation of Lateral Hypothalamic Neuronal Populations during Feeding and Their Assembly by Gamma Oscillations. J Neurosci 2024; 44:e0518242024. [PMID: 39256049 PMCID: PMC11502232 DOI: 10.1523/jneurosci.0518-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 07/18/2024] [Accepted: 08/10/2024] [Indexed: 09/12/2024] Open
Abstract
Neural circuits supporting innate behaviors, such as feeding, exploration, and social interaction, intermingle in the lateral hypothalamus (LH). Although previous studies have shown that individual LH neurons change their firing relative to the baseline during one or more behaviors, the firing rate dynamics of LH populations within behavioral episodes and the coordination of behavior-related LH populations remain largely unknown. Here, using unsupervised graph-based clustering of LH neurons firing rate dynamics in freely behaving male mice, we identified distinct populations of cells whose activity corresponds to feeding, specific times during feeding bouts, or other innate behaviors-social interaction and novel object exploration. Feeding-related cells fired together with a higher probability during slow and fast gamma oscillations (30-60 and 60-90 Hz) than during nonrhythmic epochs. In contrast, the cofiring of neurons signaling other behaviors than feeding was overall similar between slow gamma and nonrhythmic epochs but increased during fast gamma oscillations. These results reveal a neural organization of ethological hierarchies in the LH and point to behavior-specific motivational systems, the dysfunction of which may contribute to mental disorders.
Collapse
Affiliation(s)
- Mahsa Altafi
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen 91054, Germany
| | - Changwan Chen
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne/University Clinic Cologne, Cologne 50931, Germany
- Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Tatiana Korotkova
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne/University Clinic Cologne, Cologne 50931, Germany
- Max Planck Institute for Metabolism Research, Cologne 50931, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), Cologne 50931, Germany
| | - Alexey Ponomarenko
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen 91054, Germany
| |
Collapse
|
11
|
Xing M, Li Y, Zhang Y, Zhou J, Ma D, Zhang M, Tang M, Ouyang T, Zhang F, Shi X, Sun J, Chen Z, Zhang WJ, Zhang S, Xie X. Paraventricular hypothalamic RUVBL2 neurons suppress appetite by enhancing excitatory synaptic transmission in distinct neurocircuits. Nat Commun 2024; 15:8939. [PMID: 39414808 PMCID: PMC11484884 DOI: 10.1038/s41467-024-53258-6] [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: 03/08/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
The paraventricular hypothalamus (PVH) is crucial for food intake control, yet the presynaptic mechanisms underlying PVH neurons remain unclear. Here, we show that RUVBL2 in the PVH is significantly reduced during energy deficit, and knockout (KO) of PVH RUVBL2 results in hyperphagic obesity in mice. RUVBL2-expressing neurons in the PVH (PVHRUVBL2) exert the anorexigenic effect by projecting to the arcuate hypothalamus, the dorsomedial hypothalamus, and the parabrachial complex. We further demonstrate that PVHRUVBL2 neurons form the synaptic connections with POMC and AgRP neurons in the ARC. PVH RUVBL2 KO impairs the excitatory synaptic transmission by reducing presynaptic boutons and synaptic vesicles near active zone. Finally, RUVBL2 overexpression in the PVH suppresses food intake and protects against diet induced obesity. Together, this study demonstrates an essential role for PVH RUVBL2 in food intake control, and suggests that modulation of synaptic plasticity could be an effective way to curb appetite and obesity.
Collapse
Affiliation(s)
- Mingming Xing
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Yang Li
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuqi Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Juemou Zhou
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Danting Ma
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Mengqi Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Minglei Tang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Ting Ouyang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Fumiao Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Xiaofeng Shi
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Jianyuan Sun
- University of Chinese Academy of Sciences, Beijing, 100049, China
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Weiping J Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Shuli Zhang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xiangyang Xie
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China.
| |
Collapse
|
12
|
Liu Y, Lin D, Najam SS, Huang S, Song M, Sirakawin C, Zhao C, Jiang H, Konopka W, Herzig S, Vinnikov IA. Functional redundancy between glucocorticoid and mineralocorticoid receptors in mature corticotropin-releasing hormone neurons protects from obesity. Obesity (Silver Spring) 2024; 32:1885-1896. [PMID: 39315404 DOI: 10.1002/oby.24116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/13/2024] [Accepted: 06/23/2024] [Indexed: 09/25/2024]
Abstract
OBJECTIVE Here, we aimed to investigate the role of glucocorticoid and mineralocorticoid receptors (GRs and MRs, respectively) in the regulation of energy homeostasis. METHODS We used three mouse models with simultaneous deletion of GRs and MRs in either forebrain neurons, the paraventricular nucleus, or corticotropin-releasing hormone (CRH) neurons and compared them with wild-type controls or isolated knockout groups. In addition to body weight, food intake, energy expenditure, insulin sensitivity, fat/lean mass distribution, and plasma corticosterone levels, we also performed transcriptomic analysis of CRH neurons and assessed their response to melanocortinergic stimulation. RESULTS Similar to global double-knockout models, deletion of GRs and MRs specifically in mature CRH neurons resulted in obesity. Importantly, the latter was accompanied by insulin resistance, but not increased plasma corticosterone levels. Transcriptomic analysis of these neurons revealed upregulation of several genes involved in postsynaptic signal transduction, including the Ptk2b gene, which encodes proline-rich tyrosine kinase 2. Knockout of both nuclear receptors leads to upregulation of Ptk2b in CRH neurons, which results in their diminished responsiveness to melanocortinergic stimulation. CONCLUSIONS Our data demonstrate the functional redundancy of GRs and MRs in CRH neurons to maintain energy homeostasis and prevent obesity. Simultaneous targeting of both receptors might represent an unprecedented approach to counteract obesity.
Collapse
Affiliation(s)
- Yu Liu
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Dongfa Lin
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun, China
| | - Syeda Sadia Najam
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Shangyuan Huang
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Muyi Song
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Chaweewan Sirakawin
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Catherine Zhao
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Haixia Jiang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Witold Konopka
- Laboratory of Neuroplasticity and Metabolism, Department of Life Sciences and Biotechnology, Łukasiewicz PORT Polish Center for Technology Development, Wrocław, Poland
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Munich Germany; Joint Heidelberg-IDC Translational Diabetes Unit, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany; Molecular Metabolic Control, Technical University Munich, Munich, Germany; German Center for Diabetes Research, Munich, Germany
| | - Ilya A Vinnikov
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
13
|
Chesters RA, Zhu J, Coull BM, Baidoe-Ansah D, Baumer L, Palm L, Klinghammer N, Chen S, Hahm A, Yagoub S, Cantacorps L, Bernardi D, Ritter K, Lippert RN. Fasting-induced activity changes in MC3R neurons of the paraventricular nucleus of the thalamus. Life Sci Alliance 2024; 7:e202402754. [PMID: 39107065 PMCID: PMC11303869 DOI: 10.26508/lsa.202402754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/09/2024] Open
Abstract
The brain controls energy homeostasis by regulating food intake through signaling within the melanocortin system. Whilst we understand the role of the hypothalamus within this system, how extra-hypothalamic brain regions are involved in controlling energy balance remains unclear. Here we show that the melanocortin 3 receptor (MC3R) is expressed in the paraventricular nucleus of the thalamus (PVT). We tested whether fasting would change the activity of MC3R neurons in this region by assessing the levels of c-Fos and pCREB as neuronal activity markers. We determined that overnight fasting causes a significant reduction in pCREB levels within PVT-MC3R neurons. We then questioned whether perturbation of MC3R signaling, during fasting, would result in altered refeeding. Using chemogenetic approaches, we show that modulation of MC3R activity, during the fasting period, does not impact body weight regain or total food intake in the refeeding period. However, we did observe significant differences in the pattern of feeding-related behavior. These findings suggest that the PVT is a region where MC3R neurons respond to energy deprivation and modulate refeeding behavior.
Collapse
Affiliation(s)
- Robert A Chesters
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Jiajie Zhu
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin, Berlin, Germany
| | - Bethany M Coull
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin, Berlin, Germany
| | - David Baidoe-Ansah
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin, Berlin, Germany
| | - Lea Baumer
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Lydia Palm
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Niklas Klinghammer
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Seve Chen
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Anneke Hahm
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Selma Yagoub
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Lídia Cantacorps
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Daniel Bernardi
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition Nuthetal, Germany
| | - Katrin Ritter
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition Nuthetal, Germany
| | - Rachel N Lippert
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
14
|
Sandoval-Caballero C, Jara J, Luarte L, Jiménez Y, Teske JA, Perez-Leighton C. Control of motivation for sucrose in the paraventricular hypothalamic nucleus by dynorphin peptides and the kappa opioid receptor. Appetite 2024; 200:107504. [PMID: 38768926 DOI: 10.1016/j.appet.2024.107504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/14/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
The dynorphin peptides are the endogenous ligands for the kappa opioid receptor (KOR) and regulate food intake. Administration of dynorphin-A1-13 (DYN) in the paraventricular hypothalamic nucleus (PVN) increases palatable food intake, and this effect is blocked by co-administration of the orexin-A neuropeptide, which is co-released with DYN in PVN from neurons located in the lateral hypothalamus. While PVN administration of DYN increases palatable food intake, whether it increases food-seeking behaviors has yet to be examined. We tested the effects of DYN and norBNI (a KOR antagonist) on the seeking and consumption of sucrose using a progressive ratio (PR) and demand curve (DC) tasks. In PVN, DYN did not alter the sucrose breaking point (BP) in the PR task nor the elasticity or intensity of demand for sucrose in the DC task. Still, DYN reduced the delay in obtaining sucrose and increased licks during sucrose intake in the PR task, irrespective of the co-administration of orexin-A. In PVN, norBNI increased the delay in obtaining sucrose and reduced licks during sucrose intake in the PR task while increasing elasticity without altering intensity of demand in the DC task. However, subcutaneous norBNI reduced the BP for sucrose and increased the delay in obtaining sucrose in the PR task while reducing the elasticity of demand. Together, these data show different effects of systemic and PVN blockade of KOR on food-seeking, consummatory behaviors, and incentive motivation for sucrose and suggest that KOR activity in PVN is necessary but not sufficient to drive seeking behaviors for palatable food.
Collapse
Affiliation(s)
- C Sandoval-Caballero
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - J Jara
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - L Luarte
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Y Jiménez
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - J A Teske
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, Arizona, USA
| | - C Perez-Leighton
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
15
|
Liu H, Bean JC, Li Y, Yu M, Ginnard OZ, Conde KM, Wang M, Fang X, Liu H, Tu L, Yin N, Han J, Yang Y, Tong Q, Arenkiel BR, Wang C, He Y, Xu Y. Distinct basal forebrain-originated neural circuits promote homoeostatic feeding and suppress hedonic feeding in male mice. Nat Metab 2024; 6:1775-1790. [PMID: 39112722 PMCID: PMC11881791 DOI: 10.1038/s42255-024-01099-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/08/2024] [Indexed: 09/26/2024]
Abstract
Feeding behaviour is influenced by two primary factors: homoeostatic needs driven by hunger and hedonic desires for pleasure even in the absence of hunger. While efficient homoeostatic feeding is vital for survival, excessive hedonic feeding can lead to adverse consequences such as obesity and metabolic dysregulations. However, the neurobiological mechanisms that orchestrate homoeostatic versus hedonic food consumption remain largely unknown. Here we show that GABAergic proenkephalin (Penk) neurons in the diagonal band of Broca (DBB) of male mice respond to food presentation. We further demonstrate that a subset of DBBPenk neurons that project to the paraventricular nucleus of the hypothalamus are preferentially activated upon food presentation during fasting periods and transmit a positive valence to facilitate feeding. On the other hand, a separate subset of DBBPenk neurons that project to the lateral hypothalamus are preferentially activated when detecting a high-fat high-sugar (HFHS) diet and transmit a negative valence to inhibit food consumption. Notably, when given free choice of chow and HFHS diets, mice with the whole DBBPenk population ablated exhibit reduced consumption of chow but increased intake of the HFHS diet, resulting in accelerated development of obesity and metabolic disturbances. Together, we identify a molecularly defined neural population in male mice that is crucial for the maintenance of energy balance by facilitating homoeostatic feeding while suppressing hedonic overeating.
Collapse
Affiliation(s)
- Hailan Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan C Bean
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yongxiang Li
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Meng Yu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Olivia Z Ginnard
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Kristine M Conde
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Mengjie Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Xing Fang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Hesong Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Longlong Tu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Na Yin
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Junying Han
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yongjie Yang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Chunmei Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yang He
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
16
|
Cai H, Schnapp WI, Mann S, Miscevic M, Shcmit MB, Conteras M, Fang C. Neural circuits regulation of satiation. Appetite 2024; 200:107512. [PMID: 38801994 PMCID: PMC11227400 DOI: 10.1016/j.appet.2024.107512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
Terminating a meal after achieving satiation is a critical step in maintaining a healthy energy balance. Despite the extensive collection of information over the last few decades regarding the neural mechanisms controlling overall eating, the mechanism underlying different temporal phases of eating behaviors, especially satiation, remains incompletely understood and is typically embedded in studies that measure the total amount of food intake. In this review, we summarize the neural circuits that detect and integrate satiation signals to suppress appetite, from interoceptive sensory inputs to the final motor outputs. Due to the well-established role of cholecystokinin (CCK) in regulating the satiation, we focus on the neural circuits that are involved in regulating the satiation effect caused by CCK. We also discuss several general principles of how these neural circuits control satiation, as well as the limitations of our current understanding of the circuits function. With the application of new techniques involving sophisticated cell-type-specific manipulation and mapping, as well as real-time recordings, it is now possible to gain a better understanding of the mechanisms specifically underlying satiation.
Collapse
Affiliation(s)
- Haijiang Cai
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA; Bio 5 Institute and Department of Neurology, University of Arizona, Tucson, AZ, 85721, USA.
| | - Wesley I Schnapp
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA; Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
| | - Shivani Mann
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
| | - Masa Miscevic
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA; Graduate Interdisciplinary Program in Physiological Sciences, University of Arizona, Tucson, AZ, 85721, USA
| | - Matthew B Shcmit
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA; Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
| | - Marco Conteras
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
| | - Caohui Fang
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
17
|
Yagoub S, Chesters RA, Ott J, Zhu J, Cantacorps L, Ritter K, Lippert RN. Acute elevated dietary fat alone is not sufficient to decrease AgRP projections in the paraventricular nucleus of the hypothalamus in mice. Sci Rep 2024; 14:20043. [PMID: 39209992 PMCID: PMC11362280 DOI: 10.1038/s41598-024-70870-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Within the brain, the connections between neurons are constantly changing in response to environmental stimuli. A prime environmental regulator of neuronal activity is diet, and previous work has highlighted changes in hypothalamic connections in response to diets high in dietary fat and elevated sucrose. We sought to determine if the change in hypothalamic neuronal connections was driven primarily by an elevation in dietary fat alone. Analysis was performed in both male and female animals. We measured Agouti-related peptide (AgRP) neuropeptide and Synaptophysin markers in the paraventricular nucleus of the hypothalamus (PVH) in response to an acute 48 h high fat diet challenge. Using two image analysis methods described in previous studies, an effect of a high fat diet on AgRP neuronal projections in the PVH of male or female mice was not identified. These results suggest that it may not be dietary fat alone that is responsible for the previously published alterations in hypothalamic connections. Future work should focus on deciphering the role of individual macronutrients on neuroanatomical and functional changes.
Collapse
Affiliation(s)
- Selma Yagoub
- German Institute for Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- University of Potsdam, Potsdam, Germany
| | - Robert A Chesters
- German Institute for Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jonathan Ott
- German Institute for Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- University of Potsdam, Potsdam, Germany
| | - Jiajie Zhu
- German Institute for Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- University of Potsdam, Potsdam, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lídia Cantacorps
- German Institute for Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Katrin Ritter
- German Institute for Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Rachel N Lippert
- German Institute for Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
18
|
Laule C, Sayar-Atasoy N, Aklan I, Kim H, Ates T, Davis D, Atasoy D. Stress integration by an ascending adrenergic-melanocortin circuit. Neuropsychopharmacology 2024; 49:1361-1372. [PMID: 38326456 PMCID: PMC11251172 DOI: 10.1038/s41386-024-01810-9] [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: 08/22/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/09/2024]
Abstract
Stress is thought to be an important contributing factor for eating disorders; however, neural substrates underlying the complex relationship between stress and appetite are not fully understood. Using in vivo recordings from awake behaving mice, we show that various acute stressors activate catecholaminergic nucleus tractus solitarius (NTSTH) projections in the paraventricular hypothalamus (PVH). Remarkably, the resulting adrenergic tone inhibits MC4R-expressing neurons (PVHMC4R), which are known for their role in feeding suppression. We found that PVHMC4R silencing encodes negative valence in sated mice and is required for avoidance induced by visceral malaise. Collectively, these findings establish PVHMC4R neurons as an effector of stress-activated brainstem adrenergic input in addition to the well-established hypothalamic-pituitary-adrenal axis. Convergent modulation of stress and feeding by PVHMC4R neurons implicates NTSTH → PVHMC4R input in stress-associated appetite disorders.
Collapse
Affiliation(s)
- Connor Laule
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nilufer Sayar-Atasoy
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Iltan Aklan
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Hyojin Kim
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Tayfun Ates
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Debbie Davis
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
19
|
Li Y, Kim M, Jiang L, Baron L, Faulkner LD, Olson DP, Li X, Gannot N, Li P, Rui L. SH2B1 Defends Against Energy Imbalance, Obesity, and Metabolic Disease via a Paraventricular Hypothalamus→Dorsal Raphe Nucleus Neurocircuit. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400437. [PMID: 38885417 PMCID: PMC11336965 DOI: 10.1002/advs.202400437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/01/2024] [Indexed: 06/20/2024]
Abstract
SH2B1 mutations are associated with obesity, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD) in humans. Global deletion of Sh2b1 results in severe obesity, type 2 diabetes, and MASLD in mice. Neuron-specific restoration of SH2B1 rescues the obesity phenotype of Sh2b1-null mice, indicating that the brain is a main SH2B1 target. However, SH2B1 neurocircuits remain elusive. SH2B1-expressing neurons in the paraventricular hypothalamus (PVHSH2B1) and a PVHSH2B1→dorsal raphe nucleus (DRN) neurocircuit are identified here. PVHSH2B1 axons monosynaptically innervate DRN neurons. Optogenetic stimulation of PVHSH2B1 axonal fibers in the DRN suppresses food intake. Chronic inhibition of PVHSH2B1 neurons causes obesity. In male and female mice, either embryonic-onset or adult-onset deletion of Sh2b1 in PVH neurons causes energy imbalance, obesity, insulin resistance, glucose intolerance, and MASLD. Ablation of Sh2b1 in the DRN-projecting PVHSH2B1 subpopulation also causes energy imbalance, obesity, and metabolic disorders. Conversely, SH2B1 overexpression in either total or DRN-projecting PVHSH2B1 neurons protects against diet-induced obesity. SH2B1 binds to TrkB and enhances brain-derived neurotrophic factor (BDNF) signaling. Ablation of Sh2b1 in PVHSH2B1 neurons induces BDNF resistance in the PVH, contributing to obesity. In conclusion, these results unveil a previously unrecognized PVHSH2B1→DRN neurocircuit through which SH2B1 defends against obesity by enhancing BDNF/TrkB signaling.
Collapse
Affiliation(s)
- Yuan Li
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Min‐Hyun Kim
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- College of Health SolutionsArizona State UniversityPhoenixAZ85004USA
| | - Lin Jiang
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Lorelei Baron
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Latrice D. Faulkner
- Department of PediatricsUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - David P. Olson
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Department of PediatricsUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Elizabeth Weiser Caswell Diabetes InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Xingyu Li
- Life Sciences InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Noam Gannot
- Life Sciences InstituteUniversity of MichiganAnn ArborMI48109USA
- Department of Biologic and Materials SciencesSchool of DentistryUniversity of MichiganAnn ArborMI48109USA
| | - Peng Li
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Life Sciences InstituteUniversity of MichiganAnn ArborMI48109USA
- Department of Biologic and Materials SciencesSchool of DentistryUniversity of MichiganAnn ArborMI48109USA
| | - Liangyou Rui
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Elizabeth Weiser Caswell Diabetes InstituteUniversity of MichiganAnn ArborMI48109USA
- Division of Gastroenterology and HepatologyDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| |
Collapse
|
20
|
Lu Y, Wang L, Luo F, Savani R, Rossi MA, Pang ZP. Dorsolateral septum GLP-1R neurons regulate feeding via lateral hypothalamic projections. Mol Metab 2024; 85:101960. [PMID: 38763494 PMCID: PMC11153235 DOI: 10.1016/j.molmet.2024.101960] [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: 03/25/2024] [Revised: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024] Open
Abstract
OBJECTIVE Although glucagon-like peptide 1 (GLP-1) is known to regulate feeding, the central mechanisms contributing to this function remain enigmatic. Here, we aim to test the role of neurons expressing GLP-1 receptors (GLP-1R) in the dorsolateral septum (dLS; dLSGLP-1R) that project to the lateral hypothalamic area (LHA) on food intake and determine the relationship with feeding regulation. METHODS Using chemogenetic manipulations, we assessed how activation or inhibition of dLSGLP-1R neurons affected food intake in Glp1r-ires-Cre mice. Then, we used channelrhodopsin-assisted circuit mapping, chemogenetics, and electrophysiological recordings to identify and assess the role of the pathway from dLSGLP-1R →LHA projections in regulating food intake. RESULTS Chemogenetic inhibition of dLSGLP-1R neurons increases food intake. LHA is a major downstream target of dLSGLP-1R neurons. The dLSGLP-1R→LHA projections are GABAergic, and chemogenetic inhibition of this pathway also promotes food intake. While chemogenetic activation of dLSGLP-1R→LHA projections modestly decreases food intake, optogenetic stimulation of the dLSGLP-1R→LHA projection terminals in the LHA rapidly suppresses feeding behavior. Finally, we demonstrate that the GLP-1R agonist, Exendin 4 enhances dLSGLP-1R →LHA GABA release. CONCLUSIONS Together, these results demonstrate that dLS-GLP-1R neurons and the inhibitory pathway to LHA can regulate feeding behavior, which might serve as a potential therapeutic target for the treatment of eating disorders or obesity.
Collapse
Affiliation(s)
- Yi Lu
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Le Wang
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Fang Luo
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Rohan Savani
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Mark A Rossi
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; Brain Health Institute, Rutgers University, New Brunswick, NJ 08901, USA; Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
| | - Zhiping P Pang
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
| |
Collapse
|
21
|
Zhu M, Jun S, Nie X, Chen J, Hao Y, Yu H, Zhang X, Sun L, Liu Y, Yuan X, Yuan F, Wang S. Mapping of afferent and efferent connections of phenylethanolamine N-methyltransferase-expressing neurons in the nucleus tractus solitarii. CNS Neurosci Ther 2024; 30:e14808. [PMID: 38887205 PMCID: PMC11183208 DOI: 10.1111/cns.14808] [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: 02/14/2024] [Revised: 05/18/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVE Phenylethanolamine N-methyltransferase (PNMT)-expressing neurons in the nucleus tractus solitarii (NTS) contribute to the regulation of autonomic functions. However, the neural circuits linking these neurons to other brain regions remain unclear. This study aims to investigate the connectivity mechanisms of the PNMT-expressing neurons in the NTS (NTSPNMT neurons). METHODS The methodologies employed in this study included a modified rabies virus-based retrograde neural tracing technique, conventional viral anterograde tracing, and immunohistochemical staining procedures. RESULTS A total of 43 upstream nuclei projecting to NTSPNMT neurons were identified, spanning several key brain regions including the medulla oblongata, pons, midbrain, cerebellum, diencephalon, and telencephalon. Notably, dense projections to the NTSPNMT neurons were observed from the central amygdaloid nucleus, paraventricular nucleus of the hypothalamus, area postrema, and the gigantocellular reticular nucleus. In contrast, the ventrolateral medulla, lateral parabrachial nucleus, and lateral hypothalamic area were identified as the primary destinations for axon terminals originating from NTSPNMT neurons. Additionally, reciprocal projections were evident among 21 nuclei, primarily situated within the medulla oblongata. CONCLUSION Our research findings demonstrate that NTSPNMT neurons form extensive connections with numerous nuclei, emphasizing their essential role in the homeostatic regulation of vital autonomic functions.
Collapse
Affiliation(s)
- Mengchu Zhu
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
- Department of Laboratory DiagnosticsHebei Medical UniversityShijiazhuangHebeiChina
| | - Shirui Jun
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Xiaojun Nie
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Jinting Chen
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Yinchao Hao
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Hongxiao Yu
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Xiang Zhang
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Lu Sun
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Yuelin Liu
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Xiangshan Yuan
- Department of Anatomy and Histoembryology, School of Basic Medical SciencesFudan UniversityShanghaiChina
- Department of NeurologyJinshan Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Fang Yuan
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
- Hebei Key Laboratory of NeurophysiologyShijiazhuangHebei ProvinceChina
| | - Sheng Wang
- Department of NeurobiologyHebei Medical UniversityShijiazhuangHebeiChina
- Hebei Key Laboratory of NeurophysiologyShijiazhuangHebei ProvinceChina
| |
Collapse
|
22
|
Perez-Leighton C, Kerr B, Scherer PE, Baudrand R, Cortés V. The interplay between leptin, glucocorticoids, and GLP1 regulates food intake and feeding behaviour. Biol Rev Camb Philos Soc 2024; 99:653-674. [PMID: 38072002 DOI: 10.1111/brv.13039] [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: 03/29/2023] [Revised: 11/21/2023] [Accepted: 12/01/2023] [Indexed: 05/09/2024]
Abstract
Nutritional, endocrine, and neurological signals converge in multiple brain centres to control feeding behaviour and food intake as part of the allostatic regulation of energy balance. Among the several neuroendocrine systems involved, the leptin, glucocorticoid, and glucagon-like peptide 1 (GLP1) systems have been extensively researched. Leptin is at the top hierarchical level since its complete absence is sufficient to trigger severe hyperphagia. Glucocorticoids are key regulators of the energy balance adaptation to stress and their sustained excess leads to excessive adiposity and metabolic perturbations. GLP1 participates in metabolic adaptation to food intake, regulating insulin secretion and satiety by parallel central and peripheral signalling systems. Herein, we review the brain and peripheral targets of these three hormone systems that integrate to regulate food intake, feeding behaviour, and metabolic homeostasis. We examine the functional relationships between leptin, glucocorticoids, and GLP1 at the central and peripheral levels, including the cross-regulation of their circulating levels and their cooperative or antagonistic actions at different brain centres. The pathophysiological roles of these neuroendocrine systems in dysregulated intake are explored in the two extremes of body adiposity - obesity and lipodystrophy - and eating behaviour disorders.
Collapse
Affiliation(s)
- Claudio Perez-Leighton
- Departmento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
| | - Bredford Kerr
- Centro de Biología Celular y Biomedicina-CEBICEM, Facultad de Medicina y Ciencia, Universidad San Sebastián, Carmen Sylva 2444, Providencia, Santiago, Chile
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - René Baudrand
- Departmento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
- Centro Translacional de Endocrinología (CETREN), Facultad de Medicina, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
| | - Víctor Cortés
- Departmento de Nutrición, Diabetes y Metabolismo, Facultad de Medicina, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
| |
Collapse
|
23
|
Liskiewicz A, Müller TD. Regulation of energy metabolism through central GIPR signaling. Peptides 2024; 176:171198. [PMID: 38527521 DOI: 10.1016/j.peptides.2024.171198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/13/2024] [Accepted: 03/22/2024] [Indexed: 03/27/2024]
Abstract
In recent years, significant progress has been made to pharmacologically combat the obesity pandemic, particularly with regard to biochemically tailored drugs that simultaneously target the receptors for glucagon-like peptide-1 (GLP-1) and the glucose-dependent insulinotropic polypeptide (GIP). But while the pharmacological benefits of GLP-1 receptor (GLP-1R) agonism are widely acknowledged, the role of the GIP system in regulating systems metabolism remains controversial. When given in adjunct to GLP-1R agonism, both agonism and antagonism of the GIP receptor (GIPR) improves metabolic outcome in preclinical and clinical studies, and despite persistent concerns about its potential obesogenic nature, there is accumulating evidence indicating that GIP has beneficial metabolic effects via central GIPR agonism. Nonetheless, despite growing recognition of the GIP system as a valuable pharmacological target, there remains great uncertainty as to where and how GIP acts in the brain to regulate metabolism, and how GIPR agonism may differ from GIPR antagonism in control of energy metabolism. In this review we highlight current knowledge on the central action of GIP, and discuss open questions related to its multifaceted biology in the brain and the periphery.
Collapse
Affiliation(s)
- Arkadiusz Liskiewicz
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany.
| |
Collapse
|
24
|
Xu XY, Wang JX, Chen JL, Dai M, Wang YM, Chen Q, Li YH, Zhu GQ, Chen AD. GLP-1 in the Hypothalamic Paraventricular Nucleus Promotes Sympathetic Activation and Hypertension. J Neurosci 2024; 44:e2032232024. [PMID: 38565292 PMCID: PMC11112640 DOI: 10.1523/jneurosci.2032-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/04/2024] Open
Abstract
Glucagon-like peptide-1 (GLP-1) and its analogs are widely used for diabetes treatment. The paraventricular nucleus (PVN) is crucial for regulating cardiovascular activity. This study aims to determine the roles of GLP-1 and its receptors (GLP-1R) in the PVN in regulating sympathetic outflow and blood pressure. Experiments were carried out in male normotensive rats and spontaneously hypertensive rats (SHR). Renal sympathetic nerve activity (RSNA) and mean arterial pressure (MAP) were recorded. GLP-1 and GLP-1R expressions were present in the PVN. PVN microinjection of GLP-1R agonist recombinant human GLP-1 (rhGLP-1) or EX-4 increased RSNA and MAP, which were prevented by GLP-1R antagonist exendin 9-39 (EX9-39) or GLP-1R antagonist 1, superoxide scavenger tempol, antioxidant N-acetylcysteine, NADPH oxidase (NOX) inhibitor apocynin, adenylyl cyclase (AC) inhibitor SQ22536 or protein kinase A (PKA) inhibitor H89. PVN microinjection of rhGLP-1 increased superoxide production, NADPH oxidase activity, cAMP level, AC, and PKA activity, which were prevented by SQ22536 or H89. GLP-1 and GLP-1R were upregulated in the PVN of SHR. PVN microinjection of GLP-1 agonist increased RSNA and MAP in both WKY and SHR, but GLP-1 antagonists caused greater effects in reducing RSNA and MAP in SHR than in WKY. The increased superoxide production and NADPH oxidase activity in the PVN of SHR were augmented by GLP-1R agonists but attenuated by GLP-1R antagonists. These results indicate that activation of GLP-1R in the PVN increased sympathetic outflow and blood pressure via cAMP-PKA-mediated NADPH oxidase activation and subsequent superoxide production. GLP-1 and GLP-1R upregulation in the PVN partially contributes to sympathetic overactivity and hypertension.
Collapse
Affiliation(s)
- Xiao-Yu Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Jing-Xiao Wang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Jun-Liu Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Min Dai
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Yi-Ming Wang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Qi Chen
- Department of Pathophysiology, Nanjing Medical University, Nanjing 211166, China
| | - Yue-Hua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing 211166, China
| | - Guo-Qing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Ai-Dong Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
25
|
Lu Y, Wang L, Luo F, Savani R, Rossi MA, Pang ZP. Dorsolateral septum GLP-1R neurons regulate feeding via lateral hypothalamic projections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586855. [PMID: 38585874 PMCID: PMC10996601 DOI: 10.1101/2024.03.26.586855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Objective Although glucagon-like peptide 1 (GLP-1) is known to regulate feeding, the central mechanisms contributing to this function remain enigmatic. Here, we aim to test the role of neurons expressing GLP-1 receptors (GLP-1R) in the dorsolateral septum (dLS; dLS GLP-1R ) and their downstream projections on food intake and determine the relationship with feeding regulation. Methods Using chemogenetic manipulations, we assessed how activation or inhibition of dLS GLP-1R neurons affected food intake in Glp1r-ires-Cre mice. Then, we used channelrhodopsin-assisted circuit mapping, chemogenetics, and electrophysiological recordings to identify and assess the role of the pathway from dLS GLP-1R neurons to the lateral hypothalamic area (LHA) in regulating food intake. Results Chemogenetic inhibition of dLS GLP-1R neurons increases food intake. LHA is a major downstream target of dLS GLP-1R neurons. The dLS GLP-1R →LHA projections are GABAergic, and chemogenetic inhibition of this pathway also promotes food intake. While chemogenetic activation of dLS GLP-1R →LHA projections modestly decreases food intake, optogenetic stimulation of the dLS GLP-1R →LHA projection terminals in the LHA rapidly suppressed feeding behavior. Finally, we demonstrate that the GLP-1R agonist, Exendin 4 enhances dLS GLP-1R →LHA GABA release. Conclusions Together, these results demonstrate that dLS-GLP-1R neurons and the inhibitory pathway to LHA can regulate feeding behavior, which might serve as a potential therapeutic target for the treatment of eating disorders or obesity. Highlights Chemogenetic inhibition of dLS GLP-1R neurons boosts food intake in mice dLS GLP-1R neuron activation does not alter feeding, likely by collateral inhibition dLS GLP-1R neurons project to LHA and release GABA Activation of dLS GLP-1R →LHA axonal terminals suppresses food intake GLP-1R agonism enhances dLS GLP-1R →LHA GABA release via a presynaptic mechanism.
Collapse
|
26
|
Wang L, Savani R, Bernabucci M, Lu Y, Singh I, Xu W, El Ouaamari A, Wheeler MB, Grill HJ, Rossi MA, Pang ZP. State-dependent central synaptic regulation by GLP-1 is essential for energy homeostasis. RESEARCH SQUARE 2024:rs.3.rs-3929981. [PMID: 38559032 PMCID: PMC10980146 DOI: 10.21203/rs.3.rs-3929981/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Central nervous system (CNS) control of metabolism plays a pivotal role in maintaining energy homeostasis. Glucagon-like peptide-1 (GLP-1, encoded by Gcg), secreted by a distinct population of neurons located within the nucleus tractus solitarius (NTS), suppresses feeding through projections to multiple brain targets1-3. Although GLP-1 analogs are proven clinically effective in treating type 2 diabetes and obesity4, the mechanisms of GLP-1 action within the brain remain unclear. Here, we investigate the involvement of GLP-1 receptor (GLP-1R) mediated signaling in a descending circuit formed by GLP-1R neurons in the paraventricular hypothalamic nucleus (PVNGLP-1R) that project to dorsal vagal complex (DVC) neurons of the brain stem in mice. PVNGLP- 1R→DVC synapses release glutamate that is augmented by GLP-1 via a presynaptic mechanism. Chemogenetic activation of PVNGLP-1R→DVC neurons suppresses feeding. The PVNGLP-1R→DVC synaptic transmission is dynamically regulated by energy states. In a state of energy deficit, synaptic strength is weaker but is more profoundly augmented by GLP-1R signaling compared to an energy-replete state. In an obese state, the dynamic synaptic strength changes in the PVNGLP-1R→DVC descending circuit are disrupted. Blocking PVNGLP-1R→DVC synaptic release or ablation of GLP-1R in the presynaptic compartment increases food intake and causes obesity, elevated blood glucose, and impaired insulin sensitivity. These findings suggest that the state-dependent synaptic plasticity in this PVNGLP-1R→DVC descending circuit mediated by GLP-1R signaling is an essential regulator of energy homeostasis.
Collapse
Affiliation(s)
- Le Wang
- The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Rohan Savani
- The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Matteo Bernabucci
- The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Yi Lu
- The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Ishnoor Singh
- The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Department of Physiology, University of Toronto, Ontario, Canada
| | - Wei Xu
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
| | | | | | - Harvey J. Grill
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark A. Rossi
- The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Brain Health Institute, Rutgers University, New Brunswick, NJ, USA
| | - Zhiping P. Pang
- The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
27
|
De Solis AJ, Del Río-Martín A, Radermacher J, Chen W, Steuernagel L, Bauder CA, Eggersmann FR, Morgan DA, Cremer AL, Sué M, Germer M, Kukat C, Vollmar S, Backes H, Rahmouni K, Kloppenburg P, Brüning JC. Reciprocal activity of AgRP and POMC neurons governs coordinated control of feeding and metabolism. Nat Metab 2024; 6:473-493. [PMID: 38378998 DOI: 10.1038/s42255-024-00987-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/16/2024] [Indexed: 02/22/2024]
Abstract
Agouti-related peptide (AgRP)-expressing and proopiomelanocortin (POMC)-expressing neurons reciprocally regulate food intake. Here, we combine non-interacting recombinases to simultaneously express functionally opposing chemogenetic receptors in AgRP and POMC neurons for comparing metabolic responses in male and female mice with simultaneous activation of AgRP and inhibition of POMC neurons with isolated activation of AgRP neurons or isolated inhibition of POMC neurons. We show that food intake is regulated by the additive effect of AgRP neuron activation and POMC neuron inhibition, while systemic insulin sensitivity and gluconeogenesis are differentially modulated by isolated-versus-simultaneous regulation of AgRP and POMC neurons. We identify a neurocircuit engaging Npy1R-expressing neurons in the paraventricular nucleus of the hypothalamus, where activated AgRP neurons and inhibited POMC neurons cooperate to promote food consumption and activate Th+ neurons in the nucleus tractus solitarii. Collectively, these results unveil how food intake is precisely regulated by the simultaneous bidirectional interplay between AgRP and POMC neurocircuits.
Collapse
Affiliation(s)
- Alain J De Solis
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, 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
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Almudena Del Río-Martín
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, 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
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Jan Radermacher
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute for Zoology, Biocenter, University of Cologne, Cologne, Germany
| | - Weiyi Chen
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, 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
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, 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
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Corinna A Bauder
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, 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
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Fynn R Eggersmann
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute for Zoology, Biocenter, University of Cologne, Cologne, Germany
| | - Donald A Morgan
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Anna-Lena Cremer
- Multimodal Imaging of Brain Metabolism Group, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Michael Sué
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Maximilian Germer
- FACS & Imaging Core Facility, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Christian Kukat
- FACS & Imaging Core Facility, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Stefan Vollmar
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Heiko Backes
- Multimodal Imaging of Brain Metabolism Group, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Peter Kloppenburg
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute for Zoology, Biocenter, University of Cologne, Cologne, Germany
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, 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.
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany.
- National Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
28
|
Rui L. Simultaneous targeting of POMC and AgRP neurons. Nat Metab 2024; 6:382-383. [PMID: 38378997 DOI: 10.1038/s42255-023-00951-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Affiliation(s)
- Liangyou Rui
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
29
|
Li SY, Cao JJ, Tan K, Fan L, Wang YQ, Shen ZX, Li SS, Wu C, Zhou H, Xu HT. CRH neurons in the lateral hypothalamic area regulate feeding behavior of mice. Curr Biol 2023; 33:4827-4843.e7. [PMID: 37848038 DOI: 10.1016/j.cub.2023.09.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/15/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023]
Abstract
Food cues serve as pivotal triggers for eliciting physiological responses that subsequently influence food consumption. The magnitude of response induced by these cues stands as a critical determinant in the context of obesity risk. Nonetheless, the underlying neural mechanism that underpins how cues associated with edible food potentiate feeding behaviors remains uncertain. In this study, we revealed that corticotropin-releasing hormone (CRH)-expressing neurons in the lateral hypothalamic area played a crucial role in promoting consummatory behaviors in mice, shedding light on this intricate process. By employing an array of diverse assays, we initially established the activation of these neurons during feeding. Manipulations using optogenetic and chemogenetic assays revealed that their activation amplified appetite and promoted feeding behaviors, whereas inhibition decreased them. Additionally, our investigation identified downstream targets, including the ventral tegmental area, and underscored the pivotal involvement of the CRH neuropeptide itself in orchestrating this regulatory network. This research casts a clarifying light on the neural mechanism underlying the augmentation of appetite and the facilitation of feeding behaviors in response to food cues. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Song-Yun Li
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Lingang Laboratory, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, 555 Qiangye Road, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing-Juan Cao
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Lingang Laboratory, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, 555 Qiangye Road, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kang Tan
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Lingang Laboratory, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, 555 Qiangye Road, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liu Fan
- Lingang Laboratory, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, 555 Qiangye Road, Shanghai 201210, China
| | - Ya-Qian Wang
- Lingang Laboratory, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, 555 Qiangye Road, Shanghai 201210, China
| | - Zi-Xuan Shen
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Lingang Laboratory, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, 555 Qiangye Road, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai-Shuai Li
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chao Wu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hui Zhou
- Lingang Laboratory, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, 555 Qiangye Road, Shanghai 201210, China
| | - Hua-Tai Xu
- Lingang Laboratory, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, 555 Qiangye Road, Shanghai 201210, China.
| |
Collapse
|
30
|
Herb BR, Glover HJ, Bhaduri A, Colantuoni C, Bale TL, Siletti K, Hodge R, Lein E, Kriegstein AR, Doege CA, Ament SA. Single-cell genomics reveals region-specific developmental trajectories underlying neuronal diversity in the human hypothalamus. SCIENCE ADVANCES 2023; 9:eadf6251. [PMID: 37939194 PMCID: PMC10631741 DOI: 10.1126/sciadv.adf6251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 10/24/2023] [Indexed: 11/10/2023]
Abstract
The development and diversity of neuronal subtypes in the human hypothalamus has been insufficiently characterized. To address this, we integrated transcriptomic data from 241,096 cells (126,840 newly generated) in the prenatal and adult human hypothalamus to reveal a temporal trajectory from proliferative stem cell populations to mature hypothalamic cell types. Iterative clustering of the adult neurons identified 108 robust transcriptionally distinct neuronal subtypes representing 10 hypothalamic nuclei. Pseudotime trajectories provided insights into the genes driving formation of these nuclei. Comparisons to single-cell transcriptomic data from the mouse hypothalamus suggested extensive conservation of neuronal subtypes despite certain differences in species-enriched gene expression. The uniqueness of hypothalamic neuronal lineages was examined developmentally by comparing excitatory lineages present in cortex and inhibitory lineages in ganglionic eminence, revealing both distinct and shared drivers of neuronal maturation across the human forebrain. These results provide a comprehensive transcriptomic view of human hypothalamus development through gestation and adulthood at cellular resolution.
Collapse
Affiliation(s)
- Brian R. Herb
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
- UM-MIND, University of Maryland School of Medicine, Baltimore, MD, USA
- Kahlert Institute for Addiction Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hannah J. Glover
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Aparna Bhaduri
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Carlo Colantuoni
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tracy L. Bale
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kimberly Siletti
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rebecca Hodge
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Ed Lein
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Arnold R. Kriegstein
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Claudia A. Doege
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Seth A. Ament
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- UM-MIND, University of Maryland School of Medicine, Baltimore, MD, USA
- Kahlert Institute for Addiction Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
31
|
Gruber T, Lechner F, Murat C, Contreras RE, Sanchez-Quant E, Miok V, Makris K, Le Thuc O, González-García I, García-Clave E, Althammer F, Krabichler Q, DeCamp LM, Jones RG, Lutter D, Williams RH, Pfluger PT, Müller TD, Woods SC, Pospisilik JA, Martinez-Jimenez CP, Tschöp MH, Grinevich V, García-Cáceres C. High-calorie diets uncouple hypothalamic oxytocin neurons from a gut-to-brain satiation pathway via κ-opioid signaling. Cell Rep 2023; 42:113305. [PMID: 37864798 PMCID: PMC10636643 DOI: 10.1016/j.celrep.2023.113305] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/21/2023] [Accepted: 10/04/2023] [Indexed: 10/23/2023] Open
Abstract
Oxytocin-expressing paraventricular hypothalamic neurons (PVNOT neurons) integrate afferent signals from the gut, including cholecystokinin (CCK), to adjust whole-body energy homeostasis. However, the molecular underpinnings by which PVNOT neurons orchestrate gut-to-brain feeding control remain unclear. Here, we show that mice undergoing selective ablation of PVNOT neurons fail to reduce food intake in response to CCK and develop hyperphagic obesity on a chow diet. Notably, exposing wild-type mice to a high-fat/high-sugar (HFHS) diet recapitulates this insensitivity toward CCK, which is linked to diet-induced transcriptional and electrophysiological aberrations specifically in PVNOT neurons. Restoring OT pathways in diet-induced obese (DIO) mice via chemogenetics or polypharmacology sufficiently re-establishes CCK's anorexigenic effects. Last, by single-cell profiling, we identify a specialized PVNOT neuronal subpopulation with increased κ-opioid signaling under an HFHS diet, which restrains their CCK-evoked activation. In sum, we document a (patho)mechanism by which PVNOT signaling uncouples a gut-brain satiation pathway under obesogenic conditions.
Collapse
Affiliation(s)
- Tim Gruber
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49506, USA; Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49506, USA.
| | - Franziska Lechner
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Cahuê Murat
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Raian E Contreras
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Eva Sanchez-Quant
- Helmholtz Pioneer Campus (HPC), Helmholtz Zentrum München, Neuherberg, Germany
| | - Viktorian Miok
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Konstantinos Makris
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Computational Discovery Research, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Ophélia Le Thuc
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Ismael González-García
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Elena García-Clave
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | | | - Quirin Krabichler
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Lisa M DeCamp
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49506, USA
| | - Russell G Jones
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49506, USA
| | - Dominik Lutter
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Computational Discovery Research, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Rhiannan H Williams
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute for Neurogenomics, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Paul T Pfluger
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Research Unit NeuroBiology of Diabetes, Helmholtz Zentrum München, 85764 Neuherberg, Germany; Neurobiology of Diabetes, TUM School of Medicine, Technical University Munich, 80333 Munich, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Department of Pharmacology and Experimental Therapy, Institute for Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls Hospitals and Clinics, Tübingen, Germany
| | - Stephen C Woods
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - John Andrew Pospisilik
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49506, USA; Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49506, USA
| | - Celia P Martinez-Jimenez
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49506, USA; TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität, Munich, Germany
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA.
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
| |
Collapse
|
32
|
Sweeney P, Gimenez LE, Hernandez CC, Cone RD. Targeting the central melanocortin system for the treatment of metabolic disorders. Nat Rev Endocrinol 2023; 19:507-519. [PMID: 37365323 DOI: 10.1038/s41574-023-00855-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/18/2023] [Indexed: 06/28/2023]
Abstract
A large body of preclinical and clinical data shows that the central melanocortin system is a promising therapeutic target for treating various metabolic disorders such as obesity and cachexia, as well as anorexia nervosa. Setmelanotide, which functions by engaging the central melanocortin circuitry, was approved by the FDA in 2020 for use in certain forms of syndromic obesity. Furthermore, the FDA approvals in 2019 of two peptide drugs targeting melanocortin receptors for the treatment of generalized hypoactive sexual desire disorder (bremelanotide) and erythropoietic protoporphyria-associated phototoxicity (afamelanotide) demonstrate the safety of this class of peptides. These approvals have also renewed excitement in the development of therapeutics targeting the melanocortin system. Here, we review the anatomy and function of the melanocortin system, discuss progress and challenges in developing melanocortin receptor-based therapeutics, and outline potential metabolic and behavioural disorders that could be addressed using pharmacological agents targeting these receptors.
Collapse
Affiliation(s)
- Patrick Sweeney
- School of Molecular and Cellular Biology, College of Liberal Arts and Sciences, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Luis E Gimenez
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | | | - Roger D Cone
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular and Integrative Physiology, School of Medicine, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular, Cellular, and Developmental Biology, College of Literature Science and the Arts, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
33
|
Santiago-Marrero I, Liu F, Wang H, Arzola EP, Xiong WC, Mei L. Energy Expenditure Homeostasis Requires ErbB4, an Obesity Risk Gene, in the Paraventricular Nucleus. eNeuro 2023; 10:ENEURO.0139-23.2023. [PMID: 37669858 PMCID: PMC10521346 DOI: 10.1523/eneuro.0139-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/07/2023] Open
Abstract
Obesity affects more than a third adult population in the United States; the prevalence is even higher in patients with major depression disorders. GWAS studies identify the receptor tyrosine kinase ErbB4 as a risk gene for obesity and for major depression disorders. We found that ErbB4 was enriched in the paraventricular nucleus of the hypothalamus (PVH). To investigate its role in metabolism, we deleted ErbB4 by injecting a Cre-expressing virus into the PVH of ErbB4-floxed male mice and found that PVH ErbB4 deletion increased weight gain without altering food intake. ErbB4 PVH deletion also reduced nighttime activity and decreased intrascapular brown adipose tissue (iBAT) thermogenesis. Analysis of covariance (ANCOVA) revealed that ErbB4 PVH deletion reduced O2 consumption, CO2 production and heat generation in a manner independent of body weight. Immunostaining experiments show that ErbB4+ neurons in the PVH were positive for oxytocin (OXT); ErbB4 PVH deletion reduces serum levels of OXT. We characterized mice where ErbB4 was specifically mutated in OXT+ neurons and found reduction in energy expenditure, phenotypes similar to PVH ErbB4 deletion. Taken together, our data indicate that ErbB4 in the PVH regulates metabolism likely through regulation of OXT expressing neurons, reveal a novel function of ErbB4 and provide insight into pathophysiological mechanisms of depression-associated obesity.
Collapse
Affiliation(s)
- Ivan Santiago-Marrero
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Fang Liu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Hongsheng Wang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Emily P Arzola
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912
- Chinese Institutes for Medical Research, Beijing 100005, China
- Capital Medical University, Beijing 100054, China
| |
Collapse
|
34
|
Jovanovic P, Pool AH, Morones N, Wang Y, Novinbakht E, Keshishian N, Jang K, Oka Y, Riera CE. A sex-specific thermogenic neurocircuit induced by predator smell recruiting cholecystokinin neurons in the dorsomedial hypothalamus. Nat Commun 2023; 14:4937. [PMID: 37582805 PMCID: PMC10427624 DOI: 10.1038/s41467-023-40484-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 07/31/2023] [Indexed: 08/17/2023] Open
Abstract
Olfactory cues are vital for prey animals like rodents to perceive and evade predators. Stress-induced hyperthermia, via brown adipose tissue (BAT) thermogenesis, boosts physical performance and facilitates escape. However, many aspects of this response, including thermogenic control and sex-specific effects, remain enigmatic. Our study unveils that the predator odor trimethylthiazoline (TMT) elicits BAT thermogenesis, suppresses feeding, and drives glucocorticoid release in female mice. Chemogenetic stimulation of olfactory bulb (OB) mitral cells recapitulates the thermogenic output of this response and associated stress hormone corticosterone release in female mice. Neuronal projections from OB to medial amygdala (MeA) and dorsomedial hypothalamus (DMH) exhibit female-specific cFos activity toward odors. Cell sorting and single-cell RNA-sequencing of DMH identify cholecystokinin (CCK)-expressing neurons as recipients of predator odor cues. Chemogenetic manipulation and neuronal silencing of DMHCCK neurons further implicate these neurons in the propagation of predator odor-associated thermogenesis and food intake suppression, highlighting their role in female stress-induced hyperthermia.
Collapse
Affiliation(s)
- Predrag Jovanovic
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Allan-Hermann Pool
- Department of Neuroscience, Department of Anesthesiology and Pain Management, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nancy Morones
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Yidan Wang
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Edward Novinbakht
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Nareg Keshishian
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Kaitlyn Jang
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Yuki Oka
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Celine E Riera
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA.
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA.
- Department of Neurology, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA.
| |
Collapse
|
35
|
Zhang SX, Kim A, Madara JC, Zhu PK, Christenson LF, Lutas A, Kalugin PN, Jin Y, Pal A, Tian L, Lowell BB, Andermann ML. Competition between stochastic neuropeptide signals calibrates the rate of satiation. RESEARCH SQUARE 2023:rs.3.rs-3185572. [PMID: 37546985 PMCID: PMC10402269 DOI: 10.21203/rs.3.rs-3185572/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
We investigated how transmission of hunger- and satiety-promoting neuropeptides, NPY and αMSH, is integrated at the level of intracellular signaling to control feeding. Receptors for these peptides use the second messenger cAMP. How cAMP integrates opposing peptide signals to regulate energy balance, and the in vivo spatiotemporal dynamics of endogenous peptidergic signaling, remain largely unknown. We show that AgRP axon stimulation in the paraventricular hypothalamus evokes probabilistic NPY release that triggers stochastic cAMP decrements in downstream MC4R-expressing neurons (PVHMC4R). Meanwhile, POMC axon stimulation triggers stochastic, αMSH-dependent cAMP increments. Release of either peptide impacts a ~100 μm diameter region, and when these peptide signals overlap, they compete to control cAMP. The competition is reflected by hunger-state-dependent differences in the amplitude and persistence of cAMP transients: hunger peptides are more efficacious in the fasted state, satiety peptides in the fed state. Feeding resolves the competition by simultaneously elevating αMSH release and suppressing NPY release, thereby sustaining elevated cAMP in PVHMC4R neurons. In turn, cAMP potentiates feeding-related excitatory inputs and promotes satiation across minutes. Our findings highlight how biochemical integration of opposing, quantal peptide signals during energy intake orchestrates a gradual transition between stable states of hunger and satiety.
Collapse
Affiliation(s)
- Stephen X Zhang
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Co-corresponding authors
| | - Angela Kim
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Joseph C Madara
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Paula K Zhu
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Lauren F Christenson
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Andrew Lutas
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Present address: Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter N Kalugin
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Program in Neuroscience, Harvard University, Cambridge, MA 02138, USA
| | - Yihan Jin
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Akash Pal
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Mark L Andermann
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Program in Neuroscience, Harvard University, Cambridge, MA 02138, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
- Co-corresponding authors
| |
Collapse
|
36
|
Zhang SX, Kim A, Madara JC, Zhu PK, Christenson LF, Lutas A, Kalugin PN, Jin Y, Pal A, Tian L, Lowell BB, Andermann ML. Competition between stochastic neuropeptide signals calibrates the rate of satiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.11.548551. [PMID: 37503012 PMCID: PMC10369917 DOI: 10.1101/2023.07.11.548551] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
We investigated how transmission of hunger- and satiety-promoting neuropeptides, NPY and αMSH, is integrated at the level of intracellular signaling to control feeding. Receptors for these peptides use the second messenger cAMP, but the messenger's spatiotemporal dynamics and role in energy balance are controversial. We show that AgRP axon stimulation in the paraventricular hypothalamus evokes probabilistic and spatially restricted NPY release that triggers stochastic cAMP decrements in downstream MC4R-expressing neurons (PVH MC4R ). Meanwhile, POMC axon stimulation triggers stochastic, αMSH-dependent cAMP increments. NPY and αMSH competitively control cAMP, as reflected by hunger-state-dependent differences in the amplitude and persistence of cAMP transients evoked by each peptide. During feeding bouts, elevated αMSH release and suppressed NPY release cooperatively sustain elevated cAMP in PVH MC4R neurons, thereby potentiating feeding-related excitatory inputs and promoting satiation across minutes. Our findings highlight how state-dependent integration of opposing, quantal peptidergic events by a common biochemical target calibrates energy intake.
Collapse
|
37
|
Haspula D, Cui Z. Neurochemical Basis of Inter-Organ Crosstalk in Health and Obesity: Focus on the Hypothalamus and the Brainstem. Cells 2023; 12:1801. [PMID: 37443835 PMCID: PMC10341274 DOI: 10.3390/cells12131801] [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: 05/15/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Precise neural regulation is required for maintenance of energy homeostasis. Essential to this are the hypothalamic and brainstem nuclei which are located adjacent and supra-adjacent to the circumventricular organs. They comprise multiple distinct neuronal populations which receive inputs not only from other brain regions, but also from circulating signals such as hormones, nutrients, metabolites and postprandial signals. Hence, they are ideally placed to exert a multi-tier control over metabolism. The neuronal sub-populations present in these key metabolically relevant nuclei regulate various facets of energy balance which includes appetite/satiety control, substrate utilization by peripheral organs and glucose homeostasis. In situations of heightened energy demand or excess, they maintain energy homeostasis by restoring the balance between energy intake and expenditure. While research on the metabolic role of the central nervous system has progressed rapidly, the neural circuitry and molecular mechanisms involved in regulating distinct metabolic functions have only gained traction in the last few decades. The focus of this review is to provide an updated summary of the mechanisms by which the various neuronal subpopulations, mainly located in the hypothalamus and the brainstem, regulate key metabolic functions.
Collapse
Affiliation(s)
- Dhanush Haspula
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Zhenzhong Cui
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA;
| |
Collapse
|
38
|
Correa-da-Silva F, Kalsbeek MJ, Gadella FS, Oppersma J, Jiang W, Wolff SEC, Korpel NL, Swaab DF, Fliers E, Kalsbeek A, Yi CX. Reduction of oxytocin-containing neurons and enhanced glymphatic activity in the hypothalamic paraventricular nucleus of patients with type 2 diabetes mellitus. Acta Neuropathol Commun 2023; 11:107. [PMID: 37400893 DOI: 10.1186/s40478-023-01606-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/15/2023] [Indexed: 07/05/2023] Open
Abstract
Evidence from animal experiments has shown that the hypothalamic paraventricular nucleus (PVN) plays a key role in regulating body weight and blood glucose levels. However, it is unclear whether neuron populations in the human PVN are involved in the development of type 2 diabetes mellitus (T2DM). To address this, we investigated the neuronal and glial populations in the PVN of 26 T2DM patients and 20 matched controls. Our findings revealed a significant reduction in oxytocin (Oxt) neuron density in the PVN of T2DM patients compared to controls, while other neuronal populations remained unchanged. This suggests that Oxt neurons may play a specific role in the pathophysiology of T2DM. Interestingly, the reduction in Oxt neurons was accompanied by a decreased melanocortinergic input in to the PVN as reflected by a reduction in alpha-MSH immunoreactivity. We also analysed two glial cell populations, as they are important for maintaining a healthy neural microenvironment. We found that microglial density, phagocytic capacity, and their proximity to neurons were not altered in T2DM patients, indicating that the loss of Oxt neurons is independent of changes in microglial immunity. However, we did observe a reduction in the number of astrocytes, which are crucial for providing trophic support to local neurons. Moreover, a specific subpopulation of astrocytes characterized by aquaporin 4 expression was overrepresented in T2DM patients. Since this subset of astrocytes is linked to the glymphatic system, their overrepresentation might point to alterations in the hypothalamic waste clearance system in T2DM. Our study shows selective loss of Oxt neurons in the PVN of T2DM individuals in association with astrocytic reduction and gliovascular remodelling. Therefore, hypothalamic Oxt neurons may represent a potential target for T2DM treatment modalities.
Collapse
Affiliation(s)
- Felipe Correa-da-Silva
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Laboratory of Endocrinology, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Martin J Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Laboratory of Endocrinology, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Femke S Gadella
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Laboratory of Endocrinology, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jorn Oppersma
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Laboratory of Endocrinology, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Wei Jiang
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Laboratory of Endocrinology, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Samantha E C Wolff
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Laboratory of Endocrinology, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Nikita L Korpel
- Laboratory of Endocrinology, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Dick F Swaab
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Laboratory of Endocrinology, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Laboratory of Endocrinology, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands.
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
39
|
Hwang E, Portillo B, Grose K, Fujikawa T, Williams KW. Exercise-induced hypothalamic neuroplasticity: Implications for energy and glucose metabolism. Mol Metab 2023; 73:101745. [PMID: 37268247 PMCID: PMC10326746 DOI: 10.1016/j.molmet.2023.101745] [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: 11/11/2022] [Revised: 05/15/2023] [Accepted: 05/29/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Neuroplasticity refers to the brain's ability to undergo functional and structural changes in response to diverse challenges. Converging evidence supports the notion that exercise serves as a metabolic challenge, triggering the release of multiple factors both in the periphery and within the brain. These factors actively contribute to plasticity in the brain, and in turn, regulate energy and glucose metabolism. SCOPE OF REVIEW The primary focus of this review is to explore the impact of exercise-induced plasticity in the brain on metabolic homeostasis, with an emphasis on the role of the hypothalamus in this process. Additionally, the review provides an overview of various factors induced by exercise that contribute to energy balance and glucose metabolism. Notably, these factors exert their effects, at least in part, through actions within the hypothalamus and more broadly in the central nervous system. MAJOR CONCLUSIONS Exercise elicits both transient and sustained changes in metabolism, accompanied by changes in neural activity within specific brain regions. Importantly, the contribution of exercise-induced plasticity and the underlying mechanisms by which neuroplasticity influences the effects of exercise are not well understood. Recent work has begun to overcome this gap in knowledge by examining the complex interactions of exercise-induced factors which alter neural circuit properties to influence metabolism.
Collapse
Affiliation(s)
- Eunsang Hwang
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Bryan Portillo
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Kyle Grose
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Teppei Fujikawa
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Kevin W Williams
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA.
| |
Collapse
|
40
|
Mattar P, Jaque C, Teske JA, Morselli E, Kerr B, Cortés V, Baudrand R, Perez-Leighton CE. Impact of short and long exposure to cafeteria diet on food intake and white adipose tissue lipolysis mediated by glucagon-like peptide 1 receptor. Front Endocrinol (Lausanne) 2023; 14:1164047. [PMID: 37293487 PMCID: PMC10244886 DOI: 10.3389/fendo.2023.1164047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/05/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction The modern food environment facilitates excessive calorie intake, a major driver of obesity. Glucagon-like peptide 1 (GLP1) is a neuroendocrine peptide that has been the basis for developing new pharmacotherapies against obesity. The GLP1 receptor (GLP1R) is expressed in central and peripheral tissues, and activation of GLP1R reduces food intake, increases the expression of thermogenic proteins in brown adipose tissue (BAT), and enhances lipolysis in white adipose tissue (WAT). Obesity decreases the efficiency of GLP1R agonists in reducing food intake and body weight. Still, whether palatable food intake before or during the early development of obesity reduces the effects of GLP1R agonists on food intake and adipose tissue metabolism remains undetermined. Further, whether GLP1R expressed in WAT contributes to these effects is unclear. Methods Food intake, expression of thermogenic BAT proteins, and WAT lipolysis were measured after central or peripheral administration of Exendin-4 (EX4), a GLP1R agonist, to mice under intermittent-short exposure to CAF diet (3 h/d for 8 days) or a longer-continuous exposure to CAF diet (24 h/d for 15 days). Ex-vivo lipolysis was measured after EX4 exposure to WAT samples from mice fed CAF or control diet for 12 weeks. . Results During intermittent-short exposure to CAF diet (3 h/d for 8 days), third ventricle injection (ICV) and intra-peritoneal administration of EX4 reduced palatable food intake. Yet, during a longer-continuous exposure to CAF diet (24 h/d for 15 days), only ICV EX4 administration reduced food intake and body weight. However, this exposure to CAF diet blocked the increase in uncoupling protein 1 (UCP1) caused by ICV EX4 administration in mice fed control diet. Finally, GLP1R expression in WAT was minimal, and EX4 failed to increase lipolysis ex-vivo in WAT tissue samples from mice fed CAF or control diet for 12 weeks. . Discussion Exposure to a CAF diet during the early stages of obesity reduces the effects of peripheral and central GLP1R agonists, and WAT does not express a functional GLP1 receptor. These data support that exposure to the obesogenic food environment, without the development or manifestation of obesity, can alter the response to GLP1R agonists. .
Collapse
Affiliation(s)
- Pamela Mattar
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian Jaque
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jennifer A. Teske
- Department of Physiology, School of Nutritional Sciences and Wellness, Graduate Interdisciplinary Programs in Physiological Sciences and Neuroscience, University of Arizona, Tucson, AZ, United States
- Department of Food Science and Nutrition at the University of Minnesota, Saint Paul, MN, United States
| | - Eugenia Morselli
- Department of Basic Sciences, Faculty of Medicine and Sciences, Universidad San Sebastián, Santiago, Chile
| | - Bredford Kerr
- Centro de Biología Celular y Biomedicina-CEBICEM, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Víctor Cortés
- Department of Nutrition, Diabetes, and Metabolism, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rene Baudrand
- Department of Endocrinology, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Centro Traslacional de Endocrinologia UC CETREN, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | | |
Collapse
|
41
|
Liu H, Li X, Li P, Hai R, Li J, Fan Q, Wang X, Chen Y, Cao X, Zhang X, Gao R, Wang K, Du C. Glutamatergic melanocortin-4 receptor neurons regulate body weight. FASEB J 2023; 37:e22920. [PMID: 37078546 DOI: 10.1096/fj.202201786r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/22/2023] [Accepted: 03/30/2023] [Indexed: 04/21/2023]
Abstract
The locus coeruleus (LC), enriched in vesicular glutamate transporter 2 (VGlut2) neurons, is a potential homeostasis-regulating hub. However, the identity of melanocortin-4 receptor (MC4R) neurons in the paraventricular nucleus (PVN) of the hypothalamus, PVNVGlut2::MC4R and LCVGlut2::MC4R regulation of body weight, and axonal projections of LCVGlut2 neurons remain unclear. Conditional knockout of MC4R in chimeric mice was used to confirm the effects of VGlut2. Interscapular brown adipose tissue was injected with pseudorabies virus to study the central nervous system projections. We mapped the LCVGlut2 circuitry. Based on the Cre-LoxP recombination system, specific knockdown of MC4R in VGlut2 neurons resulted in weight gain in chimeric mice. Adeno-associated virus-mediated knockdown of MC4R expression in the PVN and LC had potential superimposed effects on weight gain, demonstrating the importance of VGlut2 neurons. Unlike these wide-ranging efferent projections, the PVN, hypothalamic arcuate nucleus, supraoptic nucleus of the lateral olfactory tegmental nuclei, and nucleus tractus solitarius send excitatory projections to LCVGlut2 neurons. The PVN → LC glutamatergic MC4R long-term neural circuit positively affected weight management and could help treat obesity.
Collapse
Affiliation(s)
- Haodong Liu
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Xiaojing Li
- College of Horticulture and Plant Protection, Inner Mongolia Agricultural University, Hohhot, China
| | - Penghui Li
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Rihan Hai
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
- Vocational and Technical College, Inner Mongolia Agricultural University, Baotou, China
| | - Jiacheng Li
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Qi Fan
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Xing Wang
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Yujie Chen
- Vocational and Technical College, Inner Mongolia Agricultural University, Baotou, China
| | - Xiaojuan Cao
- Vocational and Technical College, Inner Mongolia Agricultural University, Baotou, China
| | - Xiaoyu Zhang
- Vocational and Technical College, Inner Mongolia Agricultural University, Baotou, China
| | - Ruifeng Gao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Kun Wang
- Institute of Cereal and Oil Crops, Hebei Academy of Agricultural and Forestry Sciences, Shijiazhuang, China
- Key Laboratory of Crop Cultivation Physiology and Green Production in Hebei Province, Shijiazhuang, China
| | - Chenguang Du
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
- Vocational and Technical College, Inner Mongolia Agricultural University, Baotou, China
| |
Collapse
|
42
|
Semple EA, Harberson MT, Xu B, Rashleigh R, Cartwright TL, Braun JJ, Custer AC, Liu C, Hill JW. Melanocortin 4 receptor signaling in Sim1 neurons permits sexual receptivity in female mice. Front Endocrinol (Lausanne) 2023; 14:983670. [PMID: 37033219 PMCID: PMC10080118 DOI: 10.3389/fendo.2023.983670] [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: 07/01/2022] [Accepted: 03/14/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Female sexual dysfunction affects approximately 40% of women in the United States, yet few therapeutic options exist for these patients. The melanocortin system is a new treatment target for hypoactive sexual desire disorder (HSDD), but the neuronal pathways involved are unclear. Methods In this study, the sexual behavior of female MC4R knockout mice lacking melanocortin 4 receptors (MC4Rs) was examined. The mice were then bred to express MC4Rs exclusively on Sim1 neurons (tbMC4RSim1 mice) or on oxytocin neurons (tbMC4ROxt mice) to examine the effect on sexual responsiveness. Results MC4R knockout mice were found to approach males less and have reduced receptivity to copulation, as indicated by a low lordosis quotient. These changes were independent of body weight. Lordosis behavior was normalized in tbMC4RSim1 mice and improved in tbMC4ROxt mice. In contrast, approach behavior was unchanged in tbMC4RSim1 mice but greatly increased in tbMC4ROxt animals. The changes were independent of melanocortin-driven metabolic effects. Discussion These results implicate MC4R signaling in Oxt neurons in appetitive behaviors and MC4R signaling in Sim1 neurons in female sexual receptivity, while suggesting melanocortin-driven sexual function does not rely on metabolic neural circuits.
Collapse
Affiliation(s)
- Erin A. Semple
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Mitchell T. Harberson
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Baijie Xu
- Center for Hypothalamic Research, University of Texas Southwestern, Dallas, TX, United States
| | - Rebecca Rashleigh
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Tori L. Cartwright
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Jessica J. Braun
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Amy C. Custer
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Chen Liu
- Center for Hypothalamic Research, University of Texas Southwestern, Dallas, TX, United States
| | - Jennifer W. Hill
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH, United States
| |
Collapse
|
43
|
Elevated Glucagon-like Peptide-1 Receptor Level in the Paraventricular Hypothalamic Nucleus of Type 2 Diabetes Mellitus Patients. Int J Mol Sci 2022; 23:ijms232415945. [PMID: 36555587 PMCID: PMC9781792 DOI: 10.3390/ijms232415945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) agonists have been approved for the treatment of type 2 diabetes mellitus (T2DM); however, the brain actions of these drugs are not properly established. We used post mortem microdissected human hypothalamic samples for RT-qPCR and Western blotting. For in situ hybridization histochemistry and immunolabelling, parallel cryosections were prepared from the hypothalamus. We developed in situ hybridization probes for human GLP-1R and oxytocin. In addition, GLP-1 and oxytocin were visualized by immunohistochemistry. Radioactive in situ hybridization histochemistry revealed abundant GLP-1R labelling in the human paraventricular hypothalamic nucleus (PVN), particularly in its magnocellular subdivision (PVNmc). Quantitative analysis of the mRNA signal demonstrated increased GLP-1R expression in the PVNmc in post mortem hypothalamic samples from T2DM subjects as compared to controls, while there was no difference in the expression level of GLP-1R in the other subdivisions of the PVN, the hypothalamic dorsomedial and infundibular nuclei. Our results in the PVN were confirmed by RT-qPCR. Furthermore, we demonstrated by Western blot technique that the GLP-1R protein level was also elevated in the PVN of T2DM patients. GLP-1 fibre terminals were also observed in the PVNmc closely apposing oxytocin neurons using immunohistochemistry. The data suggest that GLP-1 activates GLP-1Rs in the PVNmc and that GLP-1R is elevated in T2DM patients, which may be related to the dysregulation of feeding behaviour and glucose homeostasis in T2DM.
Collapse
|
44
|
Deng Y, Dickey JE, Saito K, Deng G, Singh U, Jiang J, Toth BA, Zhu Z, Zingman LV, Resch JM, Grobe JL, Cui H. Elucidating the role of Rgs2 expression in the PVN for metabolic homeostasis in mice. Mol Metab 2022; 66:101622. [PMID: 36307046 PMCID: PMC9638802 DOI: 10.1016/j.molmet.2022.101622] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/09/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE RGS2 is a GTPase activating protein that modulates GPCR-Gα signaling and mice lacking RGS2 globally exhibit metabolic alterations. While RGS2 is known to be broadly expressed throughout the body including the brain, the relative contribution of brain RGS2 to metabolic homeostasis remains unknown. The purpose of this study was to characterize RGS2 expression in the paraventricular nucleus of hypothalamus (PVN) and test its role in metabolic homeostasis. METHODS We used a combination of RNAscope in situ hybridization (ISH), immunohistochemistry, and bioinformatic analyses to characterize the pattern of Rgs2 expression in the PVN. We then created mice lacking Rgs2 either prenatally or postnatally in the PVN and evaluated their metabolic consequences. RESULTS RNAscope ISH analysis revealed a broad but regionally enriched Rgs2 mRNA expression throughout the mouse brain, with the highest expression being observed in the PVN along with several other brain regions, such as the arcuate nucleus of hypothalamus and the dorsal raphe nucleus. Within the PVN, we found that Rgs2 is specifically enriched in CRH+ endocrine neurons and is further increased by calorie restriction. Functionally, although Sim1-Cre-mediated prenatal deletion of Rgs2 in PVN neurons had no major effects on metabolic homeostasis, AAV-mediated adult deletion of Rgs2 in the PVN led to significantly increased food intake, body weight (both fat and fat-free masses), body length, and blood glucose levels in both male and female mice. Strikingly, we found that prolonged postnatal loss of Rgs2 leads to neuronal cell death in the PVN, while rapid body weight gain in the early phase of viral-mediated PVN Rgs2 deletion is independent of PVN neuronal loss. CONCLUSIONS Our results provide the first evidence to show that PVN Rgs2 expression is not only sensitive to metabolic challenge but also critically required for PVN endocrine neurons to function and maintain metabolic homeostasis.
Collapse
Affiliation(s)
- Yue Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jacob E Dickey
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Kenji Saito
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Guorui Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Uday Singh
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jingwei Jiang
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Brandon A Toth
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Zhiyong Zhu
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Leonid V Zingman
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jon M Resch
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States; F.O.E. Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Huxing Cui
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States; F.O.E. Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, United States.
| |
Collapse
|
45
|
Wang M, Wang X, Jiang B, Zhai Y, Zheng J, Yang L, Tai X, Li Y, Fu S, Xu J, Lei X, Kuang Z, Zhang C, Bai X, Li M, Zan T, Qu S, Li Q, Zhang C. Identification of MRAP protein family as broad-spectrum GPCR modulators. Clin Transl Med 2022; 12:e1091. [PMID: 36314066 PMCID: PMC9619224 DOI: 10.1002/ctm2.1091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The melanocortin receptor accessory proteins (MRAP1 and MRAP2) are well-known endocrine regulators for the trafficking and signalling of all five melanocortin receptors (MC1R-MC5R). The observation of MRAP2 on regulating several non-melanocortin G protein-coupled receptors (GPCRs) has been sporadically reported, whereas other endogenous GPCR partners of the MRAP protein family are largely unknown. METHODS Here, we performed single-cell transcriptome analysis and drew a fine GPCR blueprint and MRAPs-associated network of two major endocrine organs, the hypothalamus and adrenal gland at single-cell resolution. We also integrated multiple bulk RNA-seq profiles and single-cell datasets of human and mouse tissues, and narrowed down a list of 48 GPCRs with strong endogenous co-expression correlation with MRAPs. RESULTS 36 and 46 metabolic-related GPCRs were consequently identified as novel interacting partners of MRAP1 or MRAP2, respectively. MRAPs exhibited protein-protein interactions and varying pharmacological properties on the surface translocation, constitutive activities and ligand-stimulated downstream signalling of these GPCRs. Knockdown of MRAP2 expression by hypothalamic administration of adeno-associated virus (AAV) packed shRNA stimulated body weight gain in mouse model. Co-injection of corticotropinreleasing factor (CRF), the agonist of corticotropin releasing hormone receptor 1 (CRHR1), suppressed feeding behaviour in a MRAP2-dependent manner. CONCLUSIONS Collectively, our study has comprehensively elucidated the complex GPCR networks in two major endocrine organs and redefined the MRAP protein family as broad-spectrum GPCR modulators. MRAP proteins not only serve as a vital endocrine pivot on the regulation of global GPCR activities in vivo that could explain the composite physiological phenotypes of the MRAP2 null murine model but also provide us with new insights of the phenotyping investigation of GPCR-MRAP functional complexes.
Collapse
Affiliation(s)
- Meng Wang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaozhu Wang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bopei Jiang
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yue Zhai
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Jihong Zheng
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Liu Yang
- Department of Endocrinology and MetabolismNational Metabolic Management CenterShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
| | - Xiaolu Tai
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yunpeng Li
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Shaliu Fu
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Jing Xu
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Xiaowei Lei
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Zhe Kuang
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Cong Zhang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xuanxuan Bai
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Mingyu Li
- Fujian Provincial Key Laboratory of Innovative Drug Target ResearchSchool of Pharmaceutical SciencesXiamen UniversityXiamenChina
| | - Tao Zan
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shen Qu
- Department of Endocrinology and MetabolismNational Metabolic Management CenterShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
| | - Qingfeng Li
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chao Zhang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
46
|
Wang X, Cui X, Li Y, Li F, Li Y, Dai J, Hu H, Wang X, Sun J, Yang Y, Zhang S. MC4R Deficiency Causes Dysregulation of Postsynaptic Excitatory Synaptic Transmission as a Crucial Culprit for Obesity. Diabetes 2022; 71:2331-2343. [PMID: 35926095 DOI: 10.2337/db22-0162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 07/30/2022] [Indexed: 11/13/2022]
Abstract
Melanocortin 4 receptor (MC4R) in the paraventricular nucleus of the hypothalamus (PVH) shows bidirectional characterization in modulating food intake and energy homeostasis. We demonstrate that MC4R knockdown (KD) in the PVH can attenuate AMPA receptor (AMPAR)-mediated postsynaptic responses by altering the phosphorylation of AMPAR GluA1 subunit through the protein kinase A (PKA)-dependent signaling cascade and simultaneously lead to rapid body weight gain. Furthermore, PKA KD in the PVH engendered similar electrophysiological and behavioral phenotypes as in MC4R KD mice. Importantly, we observed that the reduction of AMPAR GluA1 expression not only led to attenuated synaptic responses but also caused body weight gain, suggesting that the aberration of synaptic responses may be one of the crucial pathogeny of obesity. Our study provides the synaptic and molecular explanations of how body weight is regulated by MC4R in the PVH.
Collapse
Affiliation(s)
- Xiaohui Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Cui
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yang Li
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yue Li
- Guangwai Community Health Service Center of Xicheng District, Beijing, China
| | - Jinye Dai
- Howard Hughes Medical Institute and Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA
| | - Han Hu
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xuefeng Wang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jianyuan Sun
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Beijing, China
| | - Yan Yang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuli Zhang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
47
|
Herrera Moro Chao D, Kirchner MK, Pham C, Foppen E, Denis RGP, Castel J, Morel C, Montalban E, Hassouna R, Bui LC, Renault J, Mouffle C, García-Cáceres C, Tschöp MH, Li D, Martin C, Stern JE, Luquet SH. Hypothalamic astrocytes control systemic glucose metabolism and energy balance. Cell Metab 2022; 34:1532-1547.e6. [PMID: 36198294 PMCID: PMC9615252 DOI: 10.1016/j.cmet.2022.09.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/23/2022] [Accepted: 09/02/2022] [Indexed: 01/29/2023]
Abstract
The hypothalamus is key in the control of energy balance. However, strategies targeting hypothalamic neurons have failed to provide viable options to treat most metabolic diseases. Conversely, the role of astrocytes in systemic metabolic control has remained largely unexplored. Here, we show that obesity promotes anatomically restricted remodeling of hypothalamic astrocyte activity. In the paraventricular nucleus (PVN) of the hypothalamus, chemogenetic manipulation of astrocytes results in bidirectional control of neighboring neuron activity, autonomic outflow, glucose metabolism, and energy balance. This process recruits a mechanism involving the astrocytic control of ambient glutamate levels, which becomes defective in obesity. Positive or negative chemogenetic manipulation of PVN astrocyte Ca2+ signals, respectively, worsens or improves metabolic status of diet-induced obese mice. Collectively, these findings highlight a yet unappreciated role for astrocytes in the direct control of systemic metabolism and suggest potential targets for anti-obesity strategy.
Collapse
Affiliation(s)
| | - Matthew K Kirchner
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA; Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA 30302, USA
| | - Cuong Pham
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, Sorbonne Universite, Paris 75005, France
| | - Ewout Foppen
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Raphael G P Denis
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France; Institut Cochin, Université Paris Cité, INSERM U1016, CNRS UMR 8104, 75014 Paris, France
| | - Julien Castel
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Chloe Morel
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Enrica Montalban
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Rim Hassouna
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Linh-Chi Bui
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Justine Renault
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Christine Mouffle
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, Sorbonne Universite, Paris 75005, France
| | - Cristina García-Cáceres
- Helmholtz Diabetes Center (HDC) & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg, 85764, Germany; Division of Metabolic Diseases, Technische Universität München, Munich, 80333, Germany; Medizinische Klinik and Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Matthias H Tschöp
- Helmholtz Diabetes Center (HDC) & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg, 85764, Germany; Division of Metabolic Diseases, Technische Universität München, Munich, 80333, Germany
| | - Dongdong Li
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, Sorbonne Universite, Paris 75005, France
| | - Claire Martin
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Javier E Stern
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA; Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA 30302, USA
| | - Serge H Luquet
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France.
| |
Collapse
|
48
|
Steuernagel L, Lam BYH, Klemm P, Dowsett GKC, Bauder CA, Tadross JA, Hitschfeld TS, Del Rio Martin A, Chen W, de Solis AJ, Fenselau H, Davidsen P, Cimino I, Kohnke SN, Rimmington D, Coll AP, Beyer A, Yeo GSH, Brüning JC. HypoMap-a unified single-cell gene expression atlas of the murine hypothalamus. Nat Metab 2022; 4:1402-1419. [PMID: 36266547 PMCID: PMC9584816 DOI: 10.1038/s42255-022-00657-y] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 09/07/2022] [Indexed: 01/20/2023]
Abstract
The hypothalamus plays a key role in coordinating fundamental body functions. Despite recent progress in single-cell technologies, a unified catalog and molecular characterization of the heterogeneous cell types and, specifically, neuronal subtypes in this brain region are still lacking. Here, we present an integrated reference atlas, 'HypoMap,' of the murine hypothalamus, consisting of 384,925 cells, with the ability to incorporate new additional experiments. We validate HypoMap by comparing data collected from Smart-Seq+Fluidigm C1 and bulk RNA sequencing of selected neuronal cell types with different degrees of cellular heterogeneity. Finally, via HypoMap, we identify classes of neurons expressing glucagon-like peptide-1 receptor (Glp1r) and prepronociceptin (Pnoc), and validate them using single-molecule in situ hybridization. Collectively, HypoMap provides a unified framework for the systematic functional annotation of murine hypothalamic cell types, and it can serve as an important platform to unravel the functional organization of hypothalamic neurocircuits and to identify druggable targets for treating metabolic disorders.
Collapse
Affiliation(s)
- Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Brian Y H Lam
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Paul Klemm
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Georgina K C Dowsett
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Corinna A Bauder
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - John A Tadross
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
- Cambridge Genomics Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Tamara Sotelo Hitschfeld
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Almudena Del Rio Martin
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Weiyi Chen
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Alain J de Solis
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Henning Fenselau
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
| | | | - Irene Cimino
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Sara N Kohnke
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Debra Rimmington
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Anthony P Coll
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Andreas Beyer
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany.
| | - Giles S H Yeo
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK.
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany.
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany.
- National Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
49
|
Role of Ion Channels in the Chemotransduction and Mechanotransduction in Digestive Function and Feeding Behavior. Int J Mol Sci 2022; 23:ijms23169358. [PMID: 36012643 PMCID: PMC9409042 DOI: 10.3390/ijms23169358] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Abstract
The gastrointestinal tract constantly communicates with the environment, receiving and processing a wide range of information. The contents of the gastrointestinal tract and the gastrointestinal tract generate mechanical and chemical signals, which are essential for regulating digestive function and feeding behavior. There are many receptors here that sense intestinal contents, including nutrients, microbes, hormones, and small molecule compounds. In signal transduction, ion channels are indispensable as an essential component that can generate intracellular ionic changes or electrical signals. Ion channels generate electrical activity in numerous neurons and, more importantly, alter the action of non-neurons simply and effectively, and also affect satiety, molecular secretion, intestinal secretion, and motility through mechanisms of peripheral sensation, signaling, and altered cellular function. In this review, we focus on the identity of ion channels in chemosensing and mechanosensing in the gastrointestinal tract.
Collapse
|
50
|
Alcantara IC, Tapia APM, Aponte Y, Krashes MJ. Acts of appetite: neural circuits governing the appetitive, consummatory, and terminating phases of feeding. Nat Metab 2022; 4:836-847. [PMID: 35879462 PMCID: PMC10852214 DOI: 10.1038/s42255-022-00611-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 06/16/2022] [Indexed: 12/11/2022]
Abstract
The overconsumption of highly caloric and palatable foods has caused a surge in obesity rates in the past half century, thereby posing a healthcare challenge due to the array of comorbidities linked to heightened body fat accrual. Developing treatments to manage body weight requires a grasp of the neurobiological basis of appetite. In this Review, we discuss advances in neuroscience that have identified brain regions and neural circuits that coordinate distinct phases of eating: food procurement, food consumption, and meal termination. While pioneering work identified several hypothalamic nuclei to be involved in feeding, more recent studies have explored how neuronal populations beyond the hypothalamus, such as the mesolimbic pathway and nodes in the hindbrain, interconnect to modulate appetite. We also examine how long-term exposure to a calorically dense diet rewires feeding circuits and alters the response of motivational systems to food. Understanding how the nervous system regulates eating behaviour will bolster the development of medical strategies that will help individuals to maintain a healthy body weight.
Collapse
Affiliation(s)
- Ivan C Alcantara
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
- Department of Neuroscience, Brown University, Providence, RI, USA
| | | | - Yeka Aponte
- National Institute on Drug Abuse (NIDA), National Institutes of Health, Baltimore, MD, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Michael J Krashes
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA.
- National Institute on Drug Abuse (NIDA), National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|