1
|
Ji RL, Tao YX. Biased signaling in drug discovery and precision medicine. Pharmacol Ther 2025; 268:108804. [PMID: 39904401 DOI: 10.1016/j.pharmthera.2025.108804] [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: 10/28/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
Receptors are crucial for converting chemical and environmental signals into cellular responses, making them prime targets in drug discovery, with about 70% of drugs targeting these receptors. Biased signaling, or functional selectivity, has revolutionized drug development by enabling precise modulation of receptor signaling pathways. This concept is more firmly established in G protein-coupled receptor and has now been applied to other receptor types, including ion channels, receptor tyrosine kinases, and nuclear receptors. Advances in structural biology have further refined our understanding of biased signaling. This targeted approach enhances therapeutic efficacy and potentially reduces side effects. Numerous biased drugs have been developed and approved as therapeutics to treat various diseases, demonstrating their significant therapeutic potential. This review provides a comprehensive overview of biased signaling in drug discovery and disease treatment, highlighting recent advancements and exploring the therapeutic potential of these innovative modulators across various diseases.
Collapse
Affiliation(s)
- Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
2
|
Leon-Mercado L, Menendez-Montes I, Tao J, Chen B, Olson DP, Mackaaij C, Cleypool CGJ, Gautron L. Hypoxia inducible factor-dependent upregulation of Agrp in glomus type I cells of the carotid body. Mol Metab 2025; 92:102095. [PMID: 39793758 PMCID: PMC11786784 DOI: 10.1016/j.molmet.2025.102095] [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: 09/14/2024] [Revised: 12/11/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Agouti-related peptide (AgRP) is a well-established potent orexigenic peptide primarily expressed in hypothalamic neurons. Nevertheless, the expression and functional significance of extrahypothalamic AgRP remain poorly understood. In this study, utilizing histological and molecular biology techniques, we have identified a significant expression of Agrp mRNA and AgRP peptide production in glomus type I cells within the mouse carotid body (CB). Furthermore, we have uncovered evidence supporting the expression of the AgRP receptor melanocortin receptor 3 (Mc3r) in adjacent sympathetic neurons, suggesting a potential local paracrine role for AgRP within the CB. Importantly, AgRP immunoreactivity was also identified in glomus type I cells of the human CB. Given the unexpected abundance of AgRP in glomus type I cells, a chemoreceptor cell specialized in oxygen sensing, we proceeded to investigate whether Agrp expression in the CB is regulated by hypoxemia and associated oxygen-sensing molecular mechanisms. In vitro luciferase assays reveal that hypoxia stimulates the human and mouse Agrp promoters in a Hypoxia Inducible Factor (HIF1/2)-dependent manner. Our in vivo experiments further demonstrate that exposure to environmental hypoxia (10%) robustly induces Agrp expression in type I glomus cells of mice. Furthermore, these findings collectively highlight the hitherto unknown source of AgRP in murine and human type I glomus cells and underscore the direct control of Agrp transcription by HIF signaling.
Collapse
Affiliation(s)
- Luis Leon-Mercado
- Center for Hypothalamic Research and Department of Internal medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ivan Menendez-Montes
- Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jonathan Tao
- Center for Hypothalamic Research and Department of Internal medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bandy Chen
- Center for Hypothalamic Research and Department of Internal medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - David P Olson
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - C Mackaaij
- Division of Surgical Specialties, Department of Anatomy, University Medical Center, Utrecht, the Netherlands
| | - C G J Cleypool
- Division of Surgical Specialties, Department of Anatomy, University Medical Center, Utrecht, the Netherlands
| | - Laurent Gautron
- Center for Hypothalamic Research and Department of Internal medicine, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
3
|
Minakhina S, Kim SY, Wondisford FE. Regulation of hypothalamic reactive oxygen species and feeding behavior by phosphorylation of the beta 2 thyroid hormone receptor isoform. Sci Rep 2024; 14:7200. [PMID: 38531895 PMCID: PMC10965981 DOI: 10.1038/s41598-024-57364-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 03/18/2024] [Indexed: 03/28/2024] Open
Abstract
Unlike other thyroid hormone receptors (THRs), the beta 2 isoform (THRB2) has a restricted expression pattern and is uniquely and abundantly phosphorylated at a conserved serine residue S101 (S102 in humans). Using tagged and or phosphorylation-defective (S101A) THRB2 mutant mice, we show that THRB2 is present in a large subset of POMC neurons and mitigates ROS accumulation during ROS-triggering events, such as fasting/refeeding or high fat diet (HFD). Excessive ROS accumulation in mutant POMC neurons was accompanied by a skewed production of orexigenic/anorexigenic hormones, resulting in elevated food intake. The prolonged exposure to pathogenic hypothalamic ROS levels during HFD feeding lead to a significant loss of POMC neurons in mutant versus wild-type (WT) mice. In cultured cells, the presence of WT THRB2 isoform, but not other THRs, or THRB2S101A, reduced ROS accumulation upon exogenous induction of oxidative stress by tert-butyl hydroperoxide. The protective function of phospho-THRB2 (pTHRB2) did not require thyroid hormone (TH), suggesting a TH-independent role of the THRB2 isoform, and phospho-S101 in particular, in regulating oxidative stress. We propose that pTHRB2 has a fundamental role in neuronal protection against ROS cellular damage, and mitigates hypothalamic pathological changes found in diet-induced obesity.
Collapse
Affiliation(s)
- Svetlana Minakhina
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
- Mount Sinai School of Medicine, New York, NY, USA.
| | - Sun Young Kim
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Fredric E Wondisford
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
- University of Arizona College of Medicine, Phoenix, AZ, USA.
| |
Collapse
|
4
|
Aitken CM, Jaramillo JCM, Davis W, Brennan‐Xie L, McDougall SJ, Lawrence AJ, Ryan PJ. Feeding signals inhibit fluid-satiation signals in the mouse lateral parabrachial nucleus to increase intake of highly palatable, caloric solutions. J Neurochem 2023; 167:648-667. [PMID: 37855271 PMCID: PMC10952698 DOI: 10.1111/jnc.15991] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/20/2023]
Abstract
Chemogenetic activation of oxytocin receptor-expressing neurons in the parabrachial nucleus (OxtrPBN neurons) acts as a satiation signal for water. In this research, we investigated the effect of activating OxtrPBN neurons on satiation for different types of fluids. Chemogenetic activation of OxtrPBN neurons in male and female transgenic OxtrCre mice robustly suppressed the rapid, initial (15-min) intake of several solutions after dehydration: water, sucrose, ethanol and saccharin, but only slightly decreased intake of Ensure®, a highly caloric solution (1 kcal/mL; containing 3.72 g protein, 3.27 g fat, 13.42 g carbohydrates, and 1.01 g dietary fibre per 100 mL). OxtrPBN neuron activation also suppressed cumulative, longer-term (2-h) intake of lower caloric, less palatable solutions, but not highly caloric, palatable solutions. These results suggest that OxtrPBN neurons predominantly control initial fluid-satiation responses after rehydration, but not longer-term intake of highly caloric, palatable solutions. The suppression of fluid intake was not because of anxiogenesis, but because OxtrPBN neuron activation decreased anxiety-like behaviour. To investigate the role of different PBN subdivisions on the intake of different solutions, we examined FOS as a proxy marker of PBN neuron activation. Different PBN subdivisions were activated by different solutions: the dorsolateral PBN similarly by all fluids; the external lateral PBN by caloric but not non-caloric solutions; and the central lateral PBN primarily by highly palatable solutions, suggesting PBN subdivisions regulate different aspects of fluid intake. To explore the possible mechanisms underlying the minimal suppression of Ensure® after OxtrPBN neuron activation, we demonstrated in in vitro slice recordings that the feeding-associated agouti-related peptide (AgRP) inhibited OxtrPBN neuron firing in a concentration-related manner, suggesting possible inhibition by feeding-related neurocircuitry of fluid satiation neurocircuitry. Overall, this research suggests that although palatable beverages like sucrose- and ethanol-containing beverages activate fluid satiation signals encoded by OxtrPBN neurons, these neurons can be inhibited by hunger-related signals (agouti-related peptide, AgRP), which may explain why these fluids are often consumed in excess of what is required for fluid satiation.
Collapse
Affiliation(s)
- Connor M. Aitken
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Janine C. M. Jaramillo
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Warren Davis
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Liam Brennan‐Xie
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Stuart J. McDougall
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Andrew J. Lawrence
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Philip J. Ryan
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
5
|
Kupcova I, Danisovic L, Grgac I, Harsanyi S. Anxiety and Depression: What Do We Know of Neuropeptides? Behav Sci (Basel) 2022; 12:262. [PMID: 36004833 PMCID: PMC9405013 DOI: 10.3390/bs12080262] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/11/2022] [Accepted: 07/27/2022] [Indexed: 12/04/2022] Open
Abstract
In modern society, there has been a rising trend of depression and anxiety. This trend heavily impacts the population's mental health and thus contributes significantly to morbidity and, in the worst case, to suicides. Modern medicine, with many antidepressants and anxiolytics at hand, is still unable to achieve remission in many patients. The pathophysiology of depression and anxiety is still only marginally understood, which encouraged researchers to focus on neuropeptides, as they are a vast group of signaling molecules in the nervous system. Neuropeptides are involved in the regulation of many physiological functions. Some act as neuromodulators and are often co-released with neurotransmitters that allow for reciprocal communication between the brain and the body. Most studied in the past were the antidepressant and anxiolytic effects of oxytocin, vasopressin or neuropeptide Y and S, or Substance P. However, in recent years, more and more novel neuropeptides have been added to the list, with implications for the research and development of new targets, diagnostic elements, and even therapies to treat anxiety and depressive disorders. In this review, we take a close look at all currently studied neuropeptides, their related pathways, their roles in stress adaptation, and the etiology of anxiety and depression in humans and animal models. We will focus on the latest research and information regarding these associated neuropeptides and thus picture their potential uses in the future.
Collapse
Affiliation(s)
- Ida Kupcova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (I.K.); (L.D.)
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (I.K.); (L.D.)
| | - Ivan Grgac
- Institute of Anatomy, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia;
| | - Stefan Harsanyi
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (I.K.); (L.D.)
| |
Collapse
|
6
|
Liu Z, Hruby VJ. MC4R biased signalling and the conformational basis of biological function selections. J Cell Mol Med 2022; 26:4125-4136. [PMID: 35818295 PMCID: PMC9344818 DOI: 10.1111/jcmm.17441] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 05/26/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022] Open
Abstract
The MC4R, a GPCR, has long been a major target for obesity treatment. As the most well‐studied melanocortin receptor subtype, the evolutionary knowledge pushes the drug development and structure–activity relationship (SAR) moving forward. The past decades have witnessed the evolution of scientists' view on GPCRs gradually from the control of a single canonical signalling pathway via a bilateral ‘active‐inactive’ model to a multi‐state alternative model where the ligands' binding affects the selection of the downstream signalling. This evolution brings the concept of biased signalling and the beginning of the next generation of peptide drug development, with the aim of turning from receptor subtype specificity to signalling pathway selectivity. The determination of the value structures of the MC4R revealed insights into the working mechanism of MC4R activation upon binding of agonists. However, new challenge has risen as we seek to unravel the mystery of MC4R signalling selection. Thus, more biased agonists and ligands with representative biological functions are needed to solve the rest of the puzzle.
Collapse
Affiliation(s)
- Zekun Liu
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona, USA
| | - Victor J Hruby
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
7
|
Tu L, Fukuda M, Tong Q, Xu Y. The ventromedial hypothalamic nucleus: watchdog of whole-body glucose homeostasis. Cell Biosci 2022; 12:71. [PMID: 35619170 PMCID: PMC9134642 DOI: 10.1186/s13578-022-00799-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/25/2022] [Indexed: 02/06/2023] Open
Abstract
The brain, particularly the ventromedial hypothalamic nucleus (VMH), has been long known for its involvement in glucose sensing and whole-body glucose homeostasis. However, it is still not fully understood how the brain detects and responds to the changes in the circulating glucose levels, as well as brain-body coordinated control of glucose homeostasis. In this review, we address the growing evidence implicating the brain in glucose homeostasis, especially in the contexts of hypoglycemia and diabetes. In addition to neurons, we emphasize the potential roles played by non-neuronal cells, as well as extracellular matrix in the hypothalamus in whole-body glucose homeostasis. Further, we review the ionic mechanisms by which glucose-sensing neurons sense fluctuations of ambient glucose levels. We also introduce the significant implications of heterogeneous neurons in the VMH upon glucose sensing and whole-body glucose homeostasis, in which sex difference is also addressed. Meanwhile, research gaps have also been identified, which necessities further mechanistic studies in future.
Collapse
Affiliation(s)
- Longlong Tu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street #8066, Houston, TX, 77030, USA
| | - Makoto Fukuda
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street #8066, Houston, TX, 77030, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street #8066, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
8
|
Ameroso D, Meng A, Chen S, Felsted J, Dulla CG, Rios M. Astrocytic BDNF signaling within the ventromedial hypothalamus regulates energy homeostasis. Nat Metab 2022; 4:627-643. [PMID: 35501599 PMCID: PMC9177635 DOI: 10.1038/s42255-022-00566-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/28/2022] [Indexed: 11/12/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is essential for maintaining energy and glucose balance within the central nervous system. Because the study of its metabolic actions has been limited to effects in neuronal cells, its role in other cell types within the brain remains poorly understood. Here we show that astrocytic BDNF signaling within the ventromedial hypothalamus (VMH) modulates neuronal activity in response to changes in energy status. This occurs via the truncated TrkB.T1 receptor. Accordingly, either fasting or central BDNF depletion enhances astrocytic synaptic glutamate clearance, thereby decreasing neuronal activity in mice. Notably, selective depletion of TrkB.T1 in VMH astrocytes blunts the effects of energy status on excitatory transmission, as well as on responses to leptin, glucose and lipids. These effects are driven by increased astrocytic invasion of excitatory synapses, enhanced glutamate reuptake and decreased neuronal activity. We thus identify BDNF/TrkB.T1 signaling in VMH astrocytes as an essential mechanism that participates in energy and glucose homeostasis.
Collapse
Affiliation(s)
- Dominique Ameroso
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Alice Meng
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Stella Chen
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Jennifer Felsted
- Graduate Program in Biochemical and Molecular Nutrition, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Chris G Dulla
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Maribel Rios
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
9
|
Abstract
The role of central estrogen in cognitive, metabolic, and reproductive health has long fascinated the lay public and scientists alike. In the last two decades, insight into estrogen signaling in the brain and its impact on female physiology is beginning to catch up with the vast information already established for its actions on peripheral tissues. Using newer methods to manipulate estrogen signaling in hormone-sensitive brain regions, neuroscientists are now identifying the molecular pathways and neuronal subtypes required for controlling sex-dependent energy allocation. However, the immense cellular complexity of these hormone-sensitive brain regions makes it clear that more research is needed to fully appreciate how estrogen modulates neural circuits to regulate physiological and behavioral end points. Such insight is essential for understanding how natural or drug-induced hormone fluctuations across lifespan affect women's health.
Collapse
Affiliation(s)
- Holly A Ingraham
- Department of Cellular and Molecular Pharmacology, School of Medicine, Mission Bay, University of California, San Francisco, California, USA;
| | - Candice B Herber
- Department of Cellular and Molecular Pharmacology, School of Medicine, Mission Bay, University of California, San Francisco, California, USA;
| | - William C Krause
- Department of Cellular and Molecular Pharmacology, School of Medicine, Mission Bay, University of California, San Francisco, California, USA;
| |
Collapse
|
10
|
Fortin SM, Chen J, Grill HJ, Hayes MR. The Mesencephalic Trigeminal Nucleus Controls Food Intake and Body Weight via Hindbrain POMC Projections. Nutrients 2021; 13:nu13051642. [PMID: 34068091 PMCID: PMC8152732 DOI: 10.3390/nu13051642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/30/2021] [Accepted: 05/10/2021] [Indexed: 12/20/2022] Open
Abstract
The mesencephalic trigeminal nucleus (Mes5) processes oral sensory–motor information, but its role in the control of energy balance remains unexplored. Here, using fluorescent in situ hybridization, we show that the Mes5 expresses the melanocortin-4 receptor. Consistent with MC4R activation in other areas of the brain, we found that Mes5 microinjection of the MC4R agonist melanotan-II (MTII) suppresses food intake and body weight in the mouse. Furthermore, NTS POMC-projecting neurons to the Mes5 can be chemogenetically activated to drive a suppression in food intake. Taken together, these findings highlight the Mes5 as a novel target of melanocortinergic control of food intake and body weight regulation, although elucidating the endogenous role of this circuit requires future study. While we observed the sufficiency of Mes5 MC4Rs for food intake and body weight suppression, these receptors do not appear to be necessary for food intake or body weight control. Collectively, the data presented here support the functional relevance of the NTS POMC to Mes5 projection pathway as a novel circuit that can be targeted to modulate food intake and body weight.
Collapse
Affiliation(s)
- Samantha M. Fortin
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.M.F.); (J.C.)
| | - Jack Chen
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.M.F.); (J.C.)
| | - Harvey J. Grill
- Department of Psychology, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Matthew R. Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.M.F.); (J.C.)
- Correspondence:
| |
Collapse
|
11
|
Hypothalamic Pomc Neurons Innervate the Spinal Cord and Modulate the Excitability of Premotor Circuits. Curr Biol 2020; 30:4579-4593.e7. [PMID: 32976803 DOI: 10.1016/j.cub.2020.08.103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 07/30/2020] [Accepted: 08/27/2020] [Indexed: 12/20/2022]
Abstract
Locomotion requires energy, yet animals need to increase locomotion in order to find and consume food in energy-deprived states. While such energy homeostatic coordination suggests brain origin, whether the central melanocortin 4 receptor (Mc4r) system directly modulates locomotion through motor circuits is unknown. Here, we report that hypothalamic Pomc neurons in zebrafish and mice have long-range projections into spinal cord regions harboring Mc4r-expressing V2a interneurons, crucial components of the premotor networks. Furthermore, in zebrafish, Mc4r activation decreases the excitability of spinal V2a neurons as well as swimming and foraging, while systemic or V2a neuron-specific blockage of Mc4r promotes locomotion. In contrast, in mice, electrophysiological recordings revealed that two-thirds of V2a neurons in lamina X are excited by the Mc4r agonist α-MSH, and acute inhibition of Mc4r signaling reduces locomotor activity. In addition, we found other Mc4r neurons in spinal lamina X that are inhibited by α-MSH, which is in line with previous studies in rodents where Mc4r agonists reduced locomotor activity. Collectively, our studies identify spinal V2a interneurons as evolutionary conserved second-order neurons of the central Mc4r system, providing a direct anatomical and functional link between energy homeostasis and locomotor control systems. The net effects of this modulatory system on locomotor activity can vary between different vertebrate species and, possibly, even within one species. We discuss the biological sense of this phenomenon in light of the ambiguity of locomotion on energy balance and the different living conditions of the different species.
Collapse
|
12
|
The Role of Ventromedial Hypothalamus Receptors in the Central Regulation of Food Intake. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-020-10120-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
13
|
Shi Z, Zhao D, Cassaglia PA, Brooks VL. Sites and sources of sympathoexcitation in obese male rats: role of brain insulin. Am J Physiol Regul Integr Comp Physiol 2020; 318:R634-R648. [PMID: 31967846 PMCID: PMC7099464 DOI: 10.1152/ajpregu.00317.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 12/21/2022]
Abstract
In males, obesity increases sympathetic nerve activity (SNA), but the mechanisms are unclear. Here, we investigate insulin, via an action in the arcuate nucleus (ArcN), and downstream neuropathways, including melanocortin receptor 3/4 (MC3/4R) in the hypothalamic paraventricular nucleus (PVN) and dorsal medial hypothalamus (DMH). We studied conscious and α-chloralose-anesthetized Sprague-Dawley rats fed a high-fat diet, which causes obesity prone (OP) rats to accrue excess fat and obesity-resistant (OR) rats to maintain fat content, similar to rats fed a standard control (CON) diet. Nonspecific blockade of the ArcN with muscimol and specific blockade of ArcN insulin receptors (InsR) decreased lumbar SNA (LSNA), heart rate (HR), and mean arterial pressure (MAP) in OP, but not OR or CON, rats, indicating that insulin supports LSNA in obese males. In conscious rats, intracerebroventricular infusion of insulin increased MAP only in OP rats and also improved HR baroreflex function from subnormal to supranormal. The brain sensitization to insulin may elucidate how insulin can drive central SNA pathways when transport of insulin across the blood-brain barrier may be impaired. Blockade of PVN, but not DMH, MC3/4R with SHU9119 decreased LSNA, HR, and, MAP in OP, but not OR or CON, rats. Interestingly, nanoinjection of the MC3/4R agonist melanotan II (MTII) into the PVN increased LSNA only in OP rats, similar to PVN MTII-induced increases in LSNA in CON rats after blockade of sympathoinhibitory neuropeptide Y Y1 receptors. ArcN InsR expression was not increased in OP rats. Collectively, these data indicate that obesity increases SNA, in part via increased InsR signaling and downstream PVN MC3/4R.
Collapse
Affiliation(s)
- Zhigang Shi
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon
| | - Ding Zhao
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon
- School of Pharmacy, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Priscila A Cassaglia
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon
| | - Virginia L Brooks
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
14
|
Hurley MM, Anderson EM, Chen C, Maunze B, Hess EM, Block ME, Patel N, Cooper Z, McCoy R, Dabra T, Conley W, Reilly MJ, Hearing M, Choi S. Acute Blockade of PACAP-Dependent Activity in the Ventromedial Nucleus of the Hypothalamus Disrupts Leptin-Induced Behavioral and Molecular Changes in Rats. Neuroendocrinology 2019; 110:271-281. [PMID: 31167202 PMCID: PMC6895395 DOI: 10.1159/000501337] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/05/2019] [Indexed: 12/18/2022]
Abstract
Leptin signaling pathways, stemming primarily from the hypothalamus, are necessary for maintaining normal energy homeostasis and body weight. In both rodents and humans, dysregulation of leptin signaling leads to morbid obesity and diabetes. Since leptin resistance is considered a primary factor underlying obesity, understanding the regulation of leptin signaling could lead to therapeutic tools and provide insights into the causality of obesity. While leptin actions in some hypothalamic regions such as the arcuate nuclei have been characterized, less is known about leptin activity in the hypothalamic ventromedial nuclei (VMN). Recently, pituitary adenylate cyclase-activating polypeptide (PACAP) has been shown to reduce feeding behavior and alter metabolism when administered into the VMN in a pattern similar to that of leptin. In the current study, we examined whether leptin and PACAP actions in the VMN share overlapping pathways in the regulation of energy balance. Interestingly, PACAP administration into the VMN increased STAT3 phosphorylation and SOCS3 mRNA expression, both of which are hallmarks of leptin receptor activation. In addition, BDNF mRNA expression in the VMN was increased by both leptin and PACAP administration. Moreover, antagonizing PACAP receptors fully reversed the behavioral and cellular effects of leptin injections into the VMN. Electrophysiological studies further illustrated that leptin-induced effects on VMN neurons were blocked by antagonizing PACAP receptors. We conclude that leptin dependency on PACAP signaling in the VMN suggests a potential common signaling cascade, allowing a tonically and systemically secreted neuropeptide to be more precisely regulated by central neuropeptides.
Collapse
Affiliation(s)
- Matthew M Hurley
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Eden M Anderson
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Christopher Chen
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Brian Maunze
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Evan M Hess
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Megan E Block
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Neerali Patel
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Zane Cooper
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Riley McCoy
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Tanya Dabra
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - William Conley
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Michael J Reilly
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Matthew Hearing
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - SuJean Choi
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA,
| |
Collapse
|
15
|
van den Pol AN, Acuna C, Davis JN, Huang H, Zhang X. Defining the caudal hypothalamic arcuate nucleus with a focus on anorexic excitatory neurons. J Physiol 2019; 597:1605-1625. [PMID: 30618146 PMCID: PMC6418765 DOI: 10.1113/jp277152] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/03/2019] [Indexed: 01/11/2023] Open
Abstract
KEY POINTS Excitatory glutamate neurons are sparse in the rostral hypothalamic arcuate nucleus (ARC), the subregion that has received the most attention in the past. In striking contrast, excitatory neurons are far more common (by a factor of 10) in the caudal ARC, an area which has received relatively little attention. These glutamate cells may play a negative role in energy balance and food intake. They can show an increase in phosphorylated Stat-3 in the presence of leptin, are electrically excited by the anorectic neuromodulator cholecystokinin, and inhibited by orexigenic neuromodulators neuropeptide Y, met-enkephalin, dynorphin and the catecholamine dopamine. The neurons project local axonal connections that excite other ARC neurons including proopiomelanocortin neurons that can play an important role in obesity. These data are consistent with models suggesting that the ARC glutamatergic neurons may play both a rapid and a slower role in acting as anorectic neurons in CNS control of food intake and energy homeostasis. ABSTRACT Here we interrogate a unique class of excitatory neurons in the hypothalamic arcuate nucleus (ARC) that utilizes glutamate as a fast neurotransmitter using mice expressing GFP under control of the vesicular glutamate transporter 2 (vGluT2) promoter. These neurons show a unique distribution, synaptic characterization, cellular physiology and response to neuropeptides involved in energy homeostasis. Although apparently not previously appreciated, the caudal ARC showed a far greater density of vGluT2 cells than the rostral ARC, as seen in transgenic vGluT2-GFP mice and mRNA analysis. After food deprivation, leptin induced an increase in phosphorylated Stat-3 in vGluT2-positive neurons, indicating a response to hormonal cues of energy state. Based on whole-cell recording electrophysiology in brain slices, vGluT2 neurons were spontaneously active with a spike frequency around 2 Hz. vGluT2 cells were responsive to a number of neuropeptides related to energy homeostasis; they were excited by the anorectic peptide cholecystokinin, but inhibited by orexigenic neuropeptide Y, dynorphin and met-enkephalin, consistent with an anorexic role in energy homeostasis. Dopamine, associated with the hedonic aspect of enhancing food intake, inhibited vGluT2 neurons. Optogenetic excitation of vGluT2 cells evoked EPSCs in neighbouring neurons, indicating local synaptic excitation of other ARC neurons. Microdrop excitation of ARC glutamate cells in brain slices rapidly increased excitatory synaptic activity in anorexigenic proopiomelanocortin neurons. Together these data support the perspective that vGluT2 cells may be more prevalent in the ARC than previously appreciated, and play predominantly an anorectic role in energy metabolism.
Collapse
Affiliation(s)
| | - Claudio Acuna
- Department of NeurosurgeryYale University School of MedicineNew HavenCT06520USA
| | - John N. Davis
- Department of NeurosurgeryYale University School of MedicineNew HavenCT06520USA
| | - Hao Huang
- Department of NeurosurgeryYale University School of MedicineNew HavenCT06520USA
| | - Xiaobing Zhang
- Department of NeurosurgeryYale University School of MedicineNew HavenCT06520USA
| |
Collapse
|
16
|
Girardet C, Marks DL, Butler AA. Melanocortin-3 Receptors Expressed on Agouti-Related Peptide Neurons Inhibit Feeding Behavior in Female Mice. Obesity (Silver Spring) 2018; 26:1849-1855. [PMID: 30426710 PMCID: PMC7294842 DOI: 10.1002/oby.22306] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Activation of hypothalamic agouti-related peptide expressing (AgRP)+ve neurons during energy deficit is a negative valence signal, rapidly activating food-seeking behaviors. This study examined the roles of melanocortin-3 receptors (MC3Rs) coexpressed in a subpopulation of AgRP+ve neurons. METHODS AgRP-MC3R mice expressing MC3Rs selectively in AgRP+ve neurons were generated by crossing AgRP-IRES-Cre mice with LoxTBMc3r mice containing a "loxP-STOP-loxP" sequence in the 5' untranslated region. Body weight, body composition, and feeding behavior were assessed during ad libitum and time-restricted feeding conditions. RESULTS In females, food intake of AgRP-IRES-Cre+ve (n = 7) or AgRP-IRES-Cre-ve (n = 9) mice was not significantly different; these mice were therefore pooled to form the "control" group. Female AgRP-MC3R mice exhibited lower food intake (25.4 ± 2.4 kJ/12 h; n = 6) compared with controls (35.3 ± 1.8 kJ/12 h; n = 16) and LoxTBMc3r mice (32.1 ± 2.1 kJ/12 h; n = 9) in the active phase during the dark period. Food intake during the rest phase (lights on) when mice consume less food (9-10 kJ) was normal between genotypes. Body weight and composition of AgRP-MC3R and LoxTBMc3r mice were similar, suggesting compensatory mechanisms for reduced calorie intake. Remarkably, AgRP-MC3R mice continued to consume less food during refeeding after fasting and time-restricted feeding. CONCLUSIONS MC3Rs expressed on AgRP+ve neurons appear to exert a strong inhibitory signal on hypothalamic networks governing feeding behavior.
Collapse
Affiliation(s)
- Clemence Girardet
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Daniel L. Marks
- Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Health & Science University, Mail Code L481 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Andrew A. Butler
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| |
Collapse
|
17
|
Ghamari-Langroudi M, Cakir I, Lippert RN, Sweeney P, Litt MJ, Ellacott KLJ, Cone RD. Regulation of energy rheostasis by the melanocortin-3 receptor. SCIENCE ADVANCES 2018; 4:eaat0866. [PMID: 30140740 PMCID: PMC6105298 DOI: 10.1126/sciadv.aat0866] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 07/18/2018] [Indexed: 05/19/2023]
Abstract
Like most homeostatic systems, adiposity in mammals is defended between upper and lower boundary conditions. While leptin and melanocortin-4 receptor (MC4R) signaling are required for defending energy set point, mechanisms controlling upper and lower homeostatic boundaries are less well understood. In contrast to the MC4R, deletion of the MC3R does not produce measurable hyperphagia or hypometabolism under normal conditions. However, we demonstrate that MC3R is required bidirectionally for controlling responses to external homeostatic challenges, such as caloric restriction or calorie-rich diet. MC3R is also required for regulated excursion from set point, or rheostasis, during pregnancy. Further, we demonstrate a molecular mechanism: MC3R provides regulatory inputs to melanocortin signaling, acting presynaptically on agouti-related protein neurons to regulate γ-aminobutyric acid release onto anorexigenic MC4R neurons, exerting boundary control on the activity of MC4R neurons. Thus, the MC3R is a critical regulator of boundary controls on melanocortin signaling, providing rheostatic control on energy storage.
Collapse
Affiliation(s)
- Masoud Ghamari-Langroudi
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 702 Light Hall, Nashville, TN 37232, USA
- Corresponding author. (M.G.-L.); (R.D.C.)
| | - Isin Cakir
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 702 Light Hall, Nashville, TN 37232, USA
- Life Sciences Institute and Department of Molecular and Integrative Physiology, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI 48109–2216, USA
| | - Rachel N. Lippert
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 702 Light Hall, Nashville, TN 37232, USA
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Patrick Sweeney
- Life Sciences Institute and Department of Molecular and Integrative Physiology, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI 48109–2216, USA
| | - Michael J. Litt
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 702 Light Hall, Nashville, TN 37232, USA
| | - Kate L. J. Ellacott
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 702 Light Hall, Nashville, TN 37232, USA
- University of Exeter Medical School, Exeter, UK
| | - Roger D. Cone
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 702 Light Hall, Nashville, TN 37232, USA
- Life Sciences Institute and Department of Molecular and Integrative Physiology, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI 48109–2216, USA
- Corresponding author. (M.G.-L.); (R.D.C.)
| |
Collapse
|
18
|
Optogenetic Study of Anterior BNST and Basomedial Amygdala Projections to the Ventromedial Hypothalamus. eNeuro 2018; 5:eN-CFN-0204-18. [PMID: 29971248 PMCID: PMC6027956 DOI: 10.1523/eneuro.0204-18.2018] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 05/26/2018] [Indexed: 12/30/2022] Open
Abstract
The basomedial amygdala (BM) influences the ventromedial nucleus of the hypothalamus (VMH) through direct glutamatergic projections as well as indirectly, through the anterior part of the bed nucleus of the stria terminalis (BNSTa). However, BM and BNSTa axons end in a segregated fashion in VMH. BM projects to the core of VMH, where VMH’s projection cells are located, whereas BNSTa projects to the shell of VMH, where GABAergic cells that inhibit core neurons are concentrated. However, the consequences of this dual regulation of VMH by BM and BNSTa are unknown. To study this question, we recorded the responses of VMH’s shell and core neurons to the optogenetic activation of BM or BNSTa inputs in transgenic mice that selectively express Cre-recombinase in glutamatergic or GABAergic neurons. Glutamatergic BM inputs fired most core neurons but elicited no response in GABAergic shell neurons. Following BM infusions of AAV-EF1α-DIO-hChR2-mCherry in Vgat-ires-Cre-Ai6 mice, no anterograde labeling was observed in the VMH, suggesting that GABAergic BM neurons do not project to the VMH. In contrast, BNSTa sent mostly GABAergic projections that inhibited both shell and core neurons. However, BNSTa-evoked IPSPs had a higher amplitude in shell neurons. Since we also found that activation of GABAergic shell neurons causes an inhibition of core neurons, these results suggest that depending on the firing rate of shell neurons, BNSTa inputs could elicit a net inhibition or disinhibition of core neurons. Thus, the dual regulation of VMH by BM and BNSTa imparts flexibility to this regulator of defensive and social behaviors.
Collapse
|
19
|
Functional Interrogation of the AgRP Neural Circuits in Control of Appetite, Body Weight, and Behaviors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1090:1-16. [PMID: 30390282 DOI: 10.1007/978-981-13-1286-1_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurons expressing agouti-related protein (AgRP), the so-called hunger neurons, protect mammals from starvation by promoting food-seeking behaviors (Trends Neurosci 36:504-512, 2013). Now an increasing amount of evidence show that these hunger-sensing neurons not only motivate animals to forage and ingest food but also help conserve energy by inhibiting innate processes that demand large amounts of energy such as growth, reproduction, and stress response. It has further been perceived that AgRP neurons transmit signals with negative valence to reward and cognitive centers so as to engage the motivational behavior toward seeking and obtaining foods (Physiol Behav 190:34-42, 2017). Recent advancement in genome editing and neurotechniques unleashed an escalated research of uniquely defined neuronal populations and neural circuits underlying the behavioral regulation of body weight and food responses (Nat Biotechnol 32:347-355, 2014; Proc Natl Acad Sci 113, 2016). In this chapter we will review literatures describing the functional organization of the AgRP circuit and its correlative signaling components that influence ingestive, foraging, motivational, and cognitive responses, a framework that reshaped our thinking toward the new hope and challenges in treatment of obesity and eating disorders.
Collapse
|
20
|
α-MSH Influences the Excitability of Feeding-Related Neurons in the Hypothalamus and Dorsal Vagal Complex of Rats. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2034691. [PMID: 29318141 PMCID: PMC5727559 DOI: 10.1155/2017/2034691] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 10/07/2017] [Accepted: 10/22/2017] [Indexed: 11/23/2022]
Abstract
Alpha-melanocyte-stimulating hormone (α-MSH) is processed from proopiomelanocortin (POMC) and acts on the melanocortin receptors, MC3 and MC4. α-MSH plays a key role in energy homeostasis. In the present study, to shed light on the mechanisms by which α-MSH exerts its anorectic effects, extracellular neuronal activity was recorded in the hypothalamus and the dorsal vagal complex (DVC) of anesthetized rats. We examined the impact of α-MSH on glucose-sensing neurons and gastric distension (GD) sensitive neurons. In the lateral hypothalamus (LHA), α-MSH inhibited 75.0% of the glucose-inhibited (GI) neurons. In the ventromedial nucleus (VMN), most glucose-sensitive neurons were glucose-excited (GE) neurons, which were mainly activated by α-MSH. In the paraventricular nucleus (PVN), α-MSH suppressed the majority of GI neurons and excited most GE neurons. In the DVC, among the 20 GI neurons examined for a response to α-MSH, 1 was activated, 16 were depressed, and 3 failed to respond. Nineteen of 24 GE neurons were activated by α-MSH administration. Additionally, among the 42 DVC neurons examined for responses to GD, 23 were excited (GD-EXC) and 19 were inhibited (GD-INH). Fifteen of 20 GD-EXC neurons were excited, whereas 11 out of 14 GD-INH neurons were suppressed by α-MSH. All these responses were abolished by pretreatment with the MC3/4R antagonist, SHU9119. In conclusion, the activity of glucose-sensitive neurons and GD-sensitive neurons in the hypothalamus and DVC can be modulated by α-MSH.
Collapse
|
21
|
Gupta R, Ma Y, Wang M, Whim MD. AgRP-Expressing Adrenal Chromaffin Cells Are Involved in the Sympathetic Response to Fasting. Endocrinology 2017; 158:2572-2584. [PMID: 28531318 PMCID: PMC5551550 DOI: 10.1210/en.2016-1268] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 05/16/2017] [Indexed: 01/23/2023]
Abstract
Fasting evokes a homeostatic response that maintains circulating levels of energy-rich metabolites and increases the drive to eat. Centrally, this reflex activates a small population of hypothalamic neurons that are characterized by the expression of AgRP, a neuropeptide with an extremely restricted distribution. Apart from the hypothalamus, the only other site with substantial expression is the adrenal gland, but there is disagreement about which cells synthesize AgRP. Using immunohistochemistry, flow cytometry, and reverse transcription-polymerase chain reaction, we show AgRP is present in the mouse adrenal medulla and is expressed by neuroendocrine chromaffin cells that also synthesize the catecholamines and neuropeptide Y. Short-term fasting led to an increase in adrenal AgRP expression. Because AgRP can act as an antagonist at MC3/4 receptors, we tested whether melanotan II, an MC3/4 receptor agonist, could regulate pre- and postsynaptic signaling within the adrenal medulla. Melanotan II decreased the paired-pulse ratio of evoked synaptic currents recorded in chromaffin cells; this effect was blocked by exogenous AgRP. In contrast, neither melanotan II nor AgRP altered the optogenetically evoked release of catecholamines from isolated chromaffin cells. These results are consistent with the idea that AgRP regulates the strength of the sympathetic input by modulation of presynaptic MC3/4 receptors located on preganglionic neurons. We conclude that a small population of neuroendocrine cells in the adrenal medulla, and the arcuate nucleus of the hypothalamus, express AgRP and neuropeptide Y and are functionally involved in the systemic response to fasting.
Collapse
Affiliation(s)
- Rajesh Gupta
- Department of Cell Biology & Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Yunbing Ma
- Department of Cell Biology & Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Manqi Wang
- Department of Cell Biology & Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Matthew D. Whim
- Department of Cell Biology & Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| |
Collapse
|
22
|
Functional Heterogeneity in the Bed Nucleus of the Stria Terminalis. J Neurosci 2017; 36:8038-49. [PMID: 27488624 DOI: 10.1523/jneurosci.0856-16.2016] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/05/2016] [Indexed: 11/21/2022] Open
Abstract
Early work stressed the differing involvement of the central amygdala (CeA) and bed nucleus of the stria terminalis (BNST) in the genesis of fear versus anxiety, respectively. In 2009, Walker, Miles, and Davis proposed a model of amygdala-BNST interactions to explain these functional differences. This model became extremely influential and now guides a new wave of studies on the role of BNST in humans. Here, we consider evidence for and against this model, in the process highlighting central principles of BNST organization. This analysis leads us to conclude that BNST's influence is not limited to the generation of anxiety-like responses to diffuse threats, but that it also shapes the impact of discrete threatening stimuli. It is likely that BNST-CeA interactions are involved in modulating responses to such threats. In addition, whereas current views emphasize the contributions of the anterolateral BNST region in anxiety, accumulating data indicate that the anteromedial and anteroventral regions also play a critical role. The presence of multiple functional subregions within the small volume of BNST raises significant technical obstacles for functional imaging studies in humans.
Collapse
|
23
|
Roltsch Hellard EA, Impastato RA, Gilpin NW. Intra-cerebral and intra-nasal melanocortin-4 receptor antagonist blocks withdrawal hyperalgesia in alcohol-dependent rats. Addict Biol 2017; 22:692-701. [PMID: 26804193 DOI: 10.1111/adb.12360] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/02/2015] [Accepted: 12/04/2015] [Indexed: 01/08/2023]
Abstract
Humans diagnosed with alcohol use disorder are more sensitive to painful stimuli during withdrawal, which suggests that excessive alcohol drinking worsens pain outcomes. Alcohol-dependent rats exhibit increases in nociceptive sensitivity during withdrawal. Data from animal models suggest that brain melanocortin-4 receptors (MC4Rs) mediate alcohol drinking and nociception. Here we tested: (1) the effect of alcohol dependence on thermal nociception in rats, and (2) the ability of acute alcohol and (3) MC4R antagonists to reverse hyperalgesia during withdrawal in alcohol-dependent rats. Rats were trained to self-administer operant alcohol and were tested for baseline thermal nociception. Half of the rats were made dependent on alcohol, then all rats were cannulated in the lateral ventricle. We tested the effects of acute alcohol drinking, acute fixed-dose alcohol, intra-ventricular agouti-related protein (endogenous MC4R antagonist), intra-ventricular HS014 (synthetic MC4R antagonist) and intra-nasal HS014 on hyperalgesia during withdrawal in alcohol-dependent rats, relative to non-dependent drinkers and alcohol-naïve controls. Alcohol-dependent rats exhibit thermal hyperalgesia that is abolished by alcohol drinking, bolus alcohol and intra-ventricular and intra-nasal MC4R antagonists. These manipulations did not affect thermal nociception in non-dependent drinkers and alcohol-naïve controls, suggesting that alcohol dependence produces neuroadaptations in brain MC4R systems. These results suggest that brain MC4R systems may be an effective therapeutic target for reducing nociception in the alcohol-dependent organism.
Collapse
Affiliation(s)
- Emily A. Roltsch Hellard
- Department of Physiology; Louisiana State University Health Sciences Center; New Orleans LA 70112 USA
- Currently at Department of Neuroscience and Experimental Therapeutics; Texas A&M Health Sciences Center; Bryan TX 77807 USA
| | - Renata A. Impastato
- Department of Physiology; Louisiana State University Health Sciences Center; New Orleans LA 70112 USA
| | - Nicholas W. Gilpin
- Department of Physiology; Louisiana State University Health Sciences Center; New Orleans LA 70112 USA
- Neuroscience Center of Excellence; Louisiana State University Health Sciences Center; New Orleans LA 70112 USA
| |
Collapse
|
24
|
Yang LK, Tao YX. Biased signaling at neural melanocortin receptors in regulation of energy homeostasis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2486-2495. [PMID: 28433713 DOI: 10.1016/j.bbadis.2017.04.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/09/2017] [Accepted: 04/10/2017] [Indexed: 12/15/2022]
Abstract
The global prevalence of obesity highlights the importance of understanding on regulation of energy homeostasis. The central melanocortin system is an important intersection connecting the neural pathways controlling satiety and energy expenditure to regulate energy homeostasis by sensing and integrating the signals of external stimuli. In this system, neural melanocortin receptors (MCRs), melanocortin-3 and -4 receptors (MC3R and MC4R), play crucial roles in the regulation of energy homeostasis. Recently, multiple intracellular signaling pathways and biased signaling at neural MCRs have been discovered, providing new insights into neural MCR signaling. This review attempts to summarize biased signaling including biased receptor mutants (both naturally occurring and lab-generated) and biased ligands at neural MCRs, and to provide a better understanding of obesity pathogenesis and new therapeutic implications for obesity treatment.
Collapse
Affiliation(s)
- Li-Kun Yang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
25
|
Origins and Functions of the Ventrolateral VMH: A Complex Neuronal Cluster Orchestrating Sex Differences in Metabolism and Behavior. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1043:199-213. [PMID: 29224096 DOI: 10.1007/978-3-319-70178-3_10] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The neuroendocrine brain or hypothalamus has emerged as one of the most highly sexually dimorphic brain regions in mammals, and specifically in rodents. It is not surprising that hypothalamic nuclei play a pivotal role in controlling sex-dependent physiology. This brain region functions as a chief executive officer or master regulator of homeostatic physiological systems to integrate both external and internal signals. In this review, we describe sex differences in energy homeostasis that arise in one area of the hypothalamus, the ventrolateral subregion of the ventromedial hypothalamus (VMHvl) with a focus on how male and female neurons function in metabolic and behavioral aspects. Because other chapters within this book provide details on signaling pathways in the VMH that contribute to sex differences in metabolism, our discussion will be limited to how the sexually dimorphic VMHvl develops and what key regulators are thought to control the many functional and physiological endpoints attributed to this region. In the last decade, several exciting new studies using state-of-the-art genetic and molecular tools are beginning to provide some understanding as to how specific neurons contribute to the coordinated physiological responses needed by male and females. New technology that combines intersectional spatial and genetic approaches is now allowing further refinement in how we describe, probe, and manipulate critical male and female neurocircuits involved in metabolism.
Collapse
|
26
|
Cheung CC, Krause WC, Edwards RH, Yang CF, Shah NM, Hnasko TS, Ingraham HA. Sex-dependent changes in metabolism and behavior, as well as reduced anxiety after eliminating ventromedial hypothalamus excitatory output. Mol Metab 2015; 4:857-66. [PMID: 26629409 PMCID: PMC4632173 DOI: 10.1016/j.molmet.2015.09.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 08/26/2015] [Accepted: 09/02/2015] [Indexed: 12/17/2022] Open
Abstract
Objectives The ventromedial hypothalamic nucleus (VMH) regulates energy homeostasis as well as social and emotional behaviors. Nearly all VMH neurons, including those in the sexually dimorphic ventrolateral VMH (VMHvl) subregion, release the excitatory neurotransmitter glutamate and use the vesicular glutamate transporter 2 (Vglut2). Here, we asked how glutamatergic signaling contributes to the collective metabolic and behavioral responses attributed to the VMH and VMHvl. Methods Using Sf1-Cre and a Vglut2 floxed allele, Vglut2 was knocked-out in SF-1 VMH neurons (Vglut2Sf1-Cre). Metabolic and neurobehavioral assays were carried out initially on Vglut2fl/fl and Vglut2Sf1-Cre mice in a mixed, and then in the C57BL/6 genetic background, which is prone to hyperglycemia and diet induced obesity (DIO). Results Several phenotypes observed in Vglut2Sf1-Cre mice were largely unexpected based on prior studies that have perturbed VMH development or VMH glutamate signaling. In our hands, Vglut2Sf1-Cre mice failed to exhibit the anticipated increase in body weight after high fat diet (HFD) or the impaired glucose homeostasis after fasting. Instead, there was a significant sex-dependent attenuation of DIO in Vglut2Sf1-Cre females. Vglut2Sf1-Cre males also display a sex-specific loss of conditioned-fear responses and aggression accompanied by more novelty-associated locomotion. Finally, unlike the higher anxiety noted in Sf1Nestin-Cre mice that lack a fully formed VMH, both male and female Vglut2Sf1-Cre mice were less anxious. Conclusions Loss of VMH glutamatergic signaling sharply decreased DIO in females, attenuated aggression and learned fear in males, and was anxiolytic in males and females. Collectively, our findings demonstrate that while glutamatergic output from the VMH appears largely dispensable for counter regulatory responses to hypoglycemia, it drives sex-dependent differences in metabolism and social behaviors and is essential for adaptive responses to anxiety-provoking stimuli in both sexes. Excitatory VMH output controls sex-dependent metabolic and behavioral phenotypes. Vglut2Sf1-Cre mice are not prone to diet-induced obesity or glucose misregulation. Loss of VMH glutamatergic signaling leads to negative energy state in females. Aggression and learned fear are lower in males lacking VMH excitatory output. VMH glutamatergic signaling drives normal anxiety responses in both sexes.
Collapse
Affiliation(s)
- Clement C Cheung
- Department of Cellular and Molecular Pharmacology, Mission Bay Campus, University of California, San Francisco 94143, United States
| | - William C Krause
- Department of Cellular and Molecular Pharmacology, Mission Bay Campus, University of California, San Francisco 94143, United States
| | - Robert H Edwards
- Department of Physiology and Neurology, Mission Bay Campus, University of California, San Francisco 94143, United States
| | - Cindy F Yang
- Department of Anatomy, Mission Bay Campus, University of California, San Francisco 94143, United States
| | - Nirao M Shah
- Department of Anatomy, Mission Bay Campus, University of California, San Francisco 94143, United States
| | - Thomas S Hnasko
- Department of Physiology and Neurology, Mission Bay Campus, University of California, San Francisco 94143, United States
| | - Holly A Ingraham
- Department of Cellular and Molecular Pharmacology, Mission Bay Campus, University of California, San Francisco 94143, United States
| |
Collapse
|
27
|
Rafael H. Omental transplantation for neuroendocrinological disorders. AMERICAN JOURNAL OF NEURODEGENERATIVE DISEASE 2015; 4:1-12. [PMID: 26389015 PMCID: PMC4568769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 08/28/2015] [Indexed: 06/05/2023]
Abstract
Neurosurgical evidences show that the aging process is initiated between 25 to 30 years of age, in the arcuate nucleus of the hypothalamus. Likewise, experimental and neurosurgical findings indicate that the progressive ischemia in the arcuate nucleus and adjacent nuclei are responsibles at the onset of obesity and, type 2 diabetes mellitus in adults, and essential arterial hypertension (EAH). On the contrary, an omental transplantation on the optic chiasma, carotid bifurcation and anterior perforated space can provoke rejuvenation, gradual loss of body weight, decrease or normalization of hyperglycemia and normalization of EAH; all of them, due to revascularization of the hypothalamic nuclei. Besides, our surgical method have best advantages than the bariatric surgery, against obesity and type 2 diabetes mellitus.
Collapse
|
28
|
Stocker SD, Gordon KW. Glutamate receptors in the hypothalamic paraventricular nucleus contribute to insulin-induced sympathoexcitation. J Neurophysiol 2014; 113:1302-9. [PMID: 25475355 DOI: 10.1152/jn.00764.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The sympathoexcitatory response to insulin is mediated by neurons in the arcuate nucleus (ARC) and hypothalamic paraventricular nucleus (PVH). Previous studies have reported that stimulation of ARC neurons increases sympathetic nerve activity (SNA) and arterial blood pressure (ABP) through glutamate receptor activation in the PVH. Therefore, the purpose of the present study was to determine whether glutamatergic neurotransmission in the PVH contributes to insulin-induced sympathoexcitation. Male Sprague-Dawley rats (275-400 g) were infused with isotonic saline or insulin (3.75 mU · kg(-1) · min(-1)) plus 50% dextrose to maintain euglycemia. Intravenous infusion of insulin significantly increased lumbar SNA without a significant change in mean ABP, renal SNA, heart rate, or blood glucose. Bilateral PVH injection of the excitatory amino acid antagonist kynurenic acid (KYN) lowered lumbar SNA and ABP of animals infused with insulin. Similarly, a cocktail of the NMDA antagonist DL-2-amino-5-phosphonopentanoic acid (AP5) and non-NMDA antagonist 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) reduced lumbar SNA and mean ABP during infusion of insulin. In a final experiment, bilateral PVH injection of AP5 only, but not CNQX, lowered lumbar SNA and mean ABP of animals infused with insulin. The peak changes in lumbar SNA and mean ABP of insulin-treated animals were not different between KYN, AP5 plus CNQX, or AP5 alone. These drug treatments did not alter any variable in animals infused with saline. Altogether, these findings suggest that glutamatergic NMDA neurotransmission in the PVH contributes to insulin-induced sympathoexcitation.
Collapse
Affiliation(s)
- Sean D Stocker
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania; and Department of Neural and Behavioral Neuroscience, Pennsylvania State College of Medicine, Hershey, Pennsylvania
| | - Kathryn W Gordon
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania; and
| |
Collapse
|
29
|
Agosti F, López Soto EJ, Cabral A, Castrogiovanni D, Schioth HB, Perelló M, Raingo J. Melanocortin 4 receptor activation inhibits presynaptic N-type calcium channels in amygdaloid complex neurons. Eur J Neurosci 2014; 40:2755-65. [PMID: 24943127 DOI: 10.1111/ejn.12650] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 04/30/2014] [Accepted: 05/05/2014] [Indexed: 12/22/2022]
Abstract
The melanocortin 4 receptor (MC4R) is a G protein-coupled receptor involved in food intake and energy expenditure regulation. MC4R activation modifies neuronal activity but the molecular mechanisms by which this regulation occurs remain unclear. Here, we tested the hypothesis that MC4R activation regulates the activity of voltage-gated calcium channels and, as a consequence, synaptic activity. We also tested whether the proposed effect occurs in the amygdala, a brain area known to mediate the anorexigenic actions of MC4R signaling. Using the patch-clamp technique, we found that the activation of MC4R with its agonist melanotan II specifically inhibited 34.5 ± 1.5% of N-type calcium currents in transiently transfected HEK293 cells. This inhibition was concentration-dependent, voltage-independent and occluded by the Gαs pathway inhibitor cholera toxin. Moreover, we found that melanotan II specifically inhibited 25.9 ± 2.0% of native N-type calcium currents and 55.4 ± 14.4% of evoked inhibitory postsynaptic currents in mouse cultured amygdala neurons. In vivo, we found that the MC4R agonist RO27-3225 increased the marker of cellular activity c-Fos in several components of the amygdala, whereas the N-type channel blocker ω conotoxin GVIA increased c-Fos expression exclusively in the central subdivision of the amygdala. Thus, MC4R specifically inhibited the presynaptic N-type channel subtype, and this inhibition may be important for the effects of melanocortin in the central subdivision of the amygdala.
Collapse
Affiliation(s)
- Francina Agosti
- Laboratory of Electrophysiology, Multidisciplinary Institute of Cell Biology (IMBICE), Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), La Plata, Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
30
|
Resch JM, Maunze B, Phillips KA, Choi S. Inhibition of food intake by PACAP in the hypothalamic ventromedial nuclei is mediated by NMDA receptors. Physiol Behav 2014; 133:230-5. [PMID: 24878316 DOI: 10.1016/j.physbeh.2014.05.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 05/02/2014] [Accepted: 05/07/2014] [Indexed: 11/17/2022]
Abstract
Central injections of pituitary adenylate cyclase-activating polypeptide (PACAP) into the ventromedial nuclei (VMN) of the hypothalamus produce hypophagia that is dependent upon the PAC1 receptor; however, the signaling downstream of this receptor in the VMN is unknown. Though PACAP signaling has many targets, this neuropeptide has been shown to influence glutamate signaling in several brain regions through mechanisms involving NMDA receptor potentiation via activation of the Src family of protein tyrosine kinases. With this in mind, we examined the Src-NMDA receptor signaling pathway as a target for PACAP signaling in the VMN that may mediate its effects on feeding behavior. Under nocturnal feeding conditions, NMDA receptor antagonism prior to PACAP administration into the VMN attenuated PACAP-mediated decreases in feeding suggesting that glutamatergic signaling via NMDA receptors is necessary for PACAP-induced hypophagia. Furthermore, PACAP administration into the VMN resulted in increased tyrosine phosphorylation of the GluN2B subunit of the NMDA receptor, and inhibition of Src kinase activity also blocked the effects of PACAP administration into the VMN on feeding behavior. These results indicate that PACAP neurotransmission in the VMN likely augments glutamate signaling by potentiating NMDA receptors activity through the tyrosine phosphorylation events mediated by the Src kinase family, and modulation of NMDA receptor activity by PACAP in the hypothalamus may be a primary mechanism for its regulation of food intake.
Collapse
Affiliation(s)
- Jon M Resch
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53201, USA
| | - Brian Maunze
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53201, USA
| | - Kailynn A Phillips
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53201, USA
| | - SuJean Choi
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53201, USA.
| |
Collapse
|
31
|
Cheung CC, Kurrasch DM, Liang JK, Ingraham HA. Genetic labeling of steroidogenic factor-1 (SF-1) neurons in mice reveals ventromedial nucleus of the hypothalamus (VMH) circuitry beginning at neurogenesis and development of a separate non-SF-1 neuronal cluster in the ventrolateral VMH. J Comp Neurol 2013; 521:1268-88. [PMID: 22987798 DOI: 10.1002/cne.23226] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 08/06/2012] [Accepted: 09/07/2012] [Indexed: 12/19/2022]
Abstract
The ventromedial nucleus of the hypothalamus (VMH) influences a wide variety of physiological responses. Here, using two distinct but complementary genetic tracing approaches in mice, we describe the development of VMH efferent projections, as marked by steroidogenic factor-1 (SF-1; NR5A1). SF-1 neurons were visualized by Tau-green fluorescent protein (GFP) expressed from the endogenous Sf-1 locus (Sf-1(TauGFP)) or by crossing the transgenic Sf1:Cre driver to a GFP reporter strain (Z/EG(Sf1:Cre)). Strikingly, VMH projections were visible early, at embryonic (E) 10.5, when few postmitotic SF1 neurons have been born, suggesting that formation of VMH circuitry begins at the onset of neurogenesis. At E14.5, comparison of these two reporter lines revealed that SF1-positive neurons in the ventrolateral VMH (VMH(vl)) persist in Z/EG(Sf1:Cre) embryos but are virtually absent in Sf-1(TauGFP). Therefore, although the entire VMH including the VMH(vl) shares a common lineage, the VMH(vl) further differentiates into a neuronal cluster devoid of SF-1. At birth, extensive VMH projections to broad regions of the brain were observed in both mouse reporter lines, matching well with those previously discovered by injection of axonal anterograde tracers in adult rats. In summary, our genetic tracing studies show that VMH efferent projections are highly conserved in rodents and are established far earlier than previously appreciated. Moreover, our results imply that neurons in the VMH(vl) adopt a distinct fate early in development, which might underlie the unique physiological functions associated with this VMH subregion.
Collapse
Affiliation(s)
- Clement C Cheung
- Department of Pediatrics, University of California, San Francisco, California 94143, USA.
| | | | | | | |
Collapse
|
32
|
Krashes MJ, Shah BP, Koda S, Lowell BB. Rapid versus delayed stimulation of feeding by the endogenously released AgRP neuron mediators GABA, NPY, and AgRP. Cell Metab 2013; 18:588-95. [PMID: 24093681 PMCID: PMC3822903 DOI: 10.1016/j.cmet.2013.09.009] [Citation(s) in RCA: 270] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 07/30/2013] [Accepted: 09/06/2013] [Indexed: 01/12/2023]
Abstract
Agouti-related peptide (AgRP) neurons of the hypothalamus release a fast transmitter (GABA) in addition to neuropeptides (neuropeptide Y [NPY] and Agouti-related peptide [AgRP]). This raises questions as to their respective functions. The acute activation of AgRP neurons robustly promotes food intake, while central injections of AgRP, NPY, or GABA agonist results in the marked escalation of food consumption with temporal variance. Given the orexigenic capability of all three of these neuroactive substances in conjunction with their coexpression in AgRP neurons, we looked to unravel their relative temporal role in driving food intake. After the acute stimulation of AgRP neurons with DREADD technology, we found that either GABA or NPY is required for the rapid stimulation of feeding, and the neuropeptide AgRP, through action on MC4 receptors, is sufficient to induce feeding over a delayed yet prolonged period. These studies help to elucidate the neurochemical mechanisms of AgRP neurons in controlling temporally distinct phases of eating.
Collapse
Affiliation(s)
- Michael J. Krashes
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Bhavik P. Shah
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Shuichi Koda
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Asubio Pharma Co., Ltd., Kobe 650-0047, Japan
| | - Bradford B. Lowell
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Correspondence: (B.B.L)
| |
Collapse
|
33
|
Toda C, Shiuchi T, Kageyama H, Okamoto S, Coutinho EA, Sato T, Okamatsu-Ogura Y, Yokota S, Takagi K, Tang L, Saito K, Shioda S, Minokoshi Y. Extracellular signal-regulated kinase in the ventromedial hypothalamus mediates leptin-induced glucose uptake in red-type skeletal muscle. Diabetes 2013; 62:2295-307. [PMID: 23530005 PMCID: PMC3712028 DOI: 10.2337/db12-1629] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Leptin is a key regulator of glucose metabolism in mammals, but the mechanisms of its action have remained elusive. We now show that signaling by extracellular signal-regulated kinase (ERK) and its upstream kinase MEK in the ventromedial hypothalamus (VMH) mediates the leptin-induced increase in glucose utilization as well as its insulin sensitivity in the whole body and in red-type skeletal muscle of mice through activation of the melanocortin receptor (MCR) in the VMH. In contrast, activation of signal transducer and activator of transcription 3 (STAT3), but not the MEK-ERK pathway, in the VMH by leptin enhances the insulin-induced suppression of endogenous glucose production in an MCR-independent manner, with this effect of leptin occurring only in the presence of an increased plasma concentration of insulin. Given that leptin requires 6 h to increase muscle glucose uptake, the transient activation of the MEK-ERK pathway in the VMH by leptin may play a role in the induction of synaptic plasticity in the VMH, resulting in the enhancement of MCR signaling in the nucleus and leading to an increase in insulin sensitivity in red-type muscle.
Collapse
Affiliation(s)
- Chitoku Toda
- Division of Endocrinology and Metabolism, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Tetsuya Shiuchi
- Department of Integrative Physiology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Haruaki Kageyama
- Department of Anatomy, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
- Faculty of Health Care, Kiryu University, Midori, Gunma, Japan
| | - Shiki Okamoto
- Division of Endocrinology and Metabolism, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Department of Physiological Sciences, Graduate University for Advanced Studies (Sokendai), Hayama, Kanagawa, Japan
| | - Eulalia A. Coutinho
- Division of Endocrinology and Metabolism, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Department of Physiological Sciences, Graduate University for Advanced Studies (Sokendai), Hayama, Kanagawa, Japan
| | - Tatsuya Sato
- Division of Endocrinology and Metabolism, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Department of Physiological Sciences, Graduate University for Advanced Studies (Sokendai), Hayama, Kanagawa, Japan
| | - Yuko Okamatsu-Ogura
- Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Shigefumi Yokota
- Division of Endocrinology and Metabolism, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Department of Physiological Sciences, Graduate University for Advanced Studies (Sokendai), Hayama, Kanagawa, Japan
| | - Kazuyo Takagi
- Division of Endocrinology and Metabolism, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Department of Physiological Sciences, Graduate University for Advanced Studies (Sokendai), Hayama, Kanagawa, Japan
| | - Lijun Tang
- Division of Endocrinology and Metabolism, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Department of Physiological Sciences, Graduate University for Advanced Studies (Sokendai), Hayama, Kanagawa, Japan
| | - Kumiko Saito
- Division of Endocrinology and Metabolism, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Seiji Shioda
- Department of Anatomy, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Yasuhiko Minokoshi
- Division of Endocrinology and Metabolism, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Department of Physiological Sciences, Graduate University for Advanced Studies (Sokendai), Hayama, Kanagawa, Japan
- Corresponding author: Yasuhiko Minokoshi,
| |
Collapse
|
34
|
Girardet C, Butler AA. Neural melanocortin receptors in obesity and related metabolic disorders. Biochim Biophys Acta Mol Basis Dis 2013; 1842:482-94. [PMID: 23680515 DOI: 10.1016/j.bbadis.2013.05.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 04/16/2013] [Accepted: 05/03/2013] [Indexed: 12/11/2022]
Abstract
Obesity is a global health issue, as it is associated with increased risk of developing chronic conditions associated with disorders of metabolism such as type 2 diabetes and cardiovascular disease. A better understanding of how excessive fat accumulation develops and causes diseases of the metabolic syndrome is urgently needed. The hypothalamic melanocortin system is an important point of convergence connecting signals of metabolic status with the neural circuitry that governs appetite and the autonomic and neuroendocrine system controling metabolism. This system has a critical role in the defense of body weight and maintenance of homeostasis. Two neural melanocortin receptors, melanocortin 3 and 4 receptors (MC3R and MC4R), play crucial roles in the regulation of energy balance. Mutations in the MC4R gene are the most common cause of monogenic obesity in humans, and a large literature indicates a role in regulating both energy intake through the control of satiety and energy expenditure. In contrast, MC3Rs have a more subtle role in energy homeostasis. Results from our lab indicate an important role for MC3Rs in synchronizing rhythms in foraging behavior with caloric cues and maintaining metabolic homeostasis during periods of nutrient scarcity. However, while deletion of the Mc3r gene in mice alters nutrient partitioning to favor accumulation of fat mass no obvious role for MC3R haploinsufficiency in human obesity has been reported. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.
Collapse
MESH Headings
- Animals
- Body Weight/genetics
- Cardiovascular Diseases/complications
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/pathology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Humans
- Metabolic Diseases/genetics
- Metabolic Diseases/metabolism
- Metabolic Diseases/pathology
- Mice
- Obesity/complications
- Obesity/genetics
- Obesity/metabolism
- Obesity/pathology
- Receptor, Melanocortin, Type 3/genetics
- Receptor, Melanocortin, Type 3/metabolism
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/metabolism
Collapse
Affiliation(s)
- Clemence Girardet
- Department of Metabolism and Aging, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Andrew A Butler
- Department of Metabolism and Aging, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA.
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW The obesity epidemic over the world has called to attention different ways to manage this development. As bariatric surgery today is the only manner by which rapid and sustained weight control can be achieved, new ways of treating obesity are under investigation. This review focuses on today's knowledge on satiety signaling as a means to combat obesity. RECENT FINDINGS The combined knowledge achieved from obesity surgery with gastric bypass and duodenal switch together with the pharmacological treatment of type 2 diabetes have given us some clues of how to manage obesity. The basis for our understanding is the present research focusing on the gut peptide hormones that are released in response to food intake, and the paucity of satiety signaling seems to prevail in obesity. This means that obese patients experience less activation of higher brain centers in association with a meal and therefore compensate with increased meal size or frequent food intake. SUMMARY Altered satiety signaling primarily emanating from the gastrointestinal tract seems to lead to the development of obesity and type 2 diabetes. Pharmacological tools that enhance the gut hormone signaling are in focus for the upcoming venues of treatment.
Collapse
|
36
|
Roa J. Role of GnRH Neurons and Their Neuronal Afferents as Key Integrators between Food Intake Regulatory Signals and the Control of Reproduction. Int J Endocrinol 2013; 2013:518046. [PMID: 24101924 PMCID: PMC3786537 DOI: 10.1155/2013/518046] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 08/07/2013] [Indexed: 11/17/2022] Open
Abstract
Reproductive function is regulated by a plethora of signals that integrate physiological and environmental information. Among others, metabolic factors are key components of this circuit since they inform about the propitious timing for reproduction depending on energy availability. This information is processed mainly at the hypothalamus that, in turn, modulates gonadotropin release from the pituitary and, thereby, gonadal activity. Metabolic hormones, such as leptin, insulin, and ghrelin, act as indicators of the energy status and convey this information to the reproductive axis regulating its activity. In this review, we will analyse the central mechanisms involved in the integration of this metabolic information and their contribution to the control of the reproductive function. Particular attention will be paid to summarize the participation of GnRH, Kiss1, NPY, and POMC neurons in this process and their possible interactions to contribute to the metabolic control of reproduction.
Collapse
Affiliation(s)
- Juan Roa
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Avenida Menéndez Pidal s/n, 14004 Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
- Instituto Maimónides de Investigaciones Biomédicas (IMIBIC)/Hospital Universitario Reina Sofia, Córdoba, Spain
- *Juan Roa:
| |
Collapse
|
37
|
Abstract
Neuropeptides are found in many mammalian CNS neurons where they play key roles in modulating neuronal activity. In contrast to amino acid transmitter release at the synapse, neuropeptide release is not restricted to the synaptic specialization, and after release, a neuropeptide may diffuse some distance to exert its action through a G protein-coupled receptor. Some neuropeptides such as hypocretin/orexin are synthesized only in single regions of the brain, and the neurons releasing these peptides probably have similar functional roles. Other peptides such as neuropeptide Y (NPY) are synthesized throughout the brain, and neurons that synthesize the peptide in one region have no anatomical or functional connection with NPY neurons in other brain regions. Here, I review converging data revealing a complex interaction between slow-acting neuromodulator peptides and fast-acting amino acid transmitters in the control of energy homeostasis, drug addiction, mood and motivation, sleep-wake states, and neuroendocrine regulation.
Collapse
|
38
|
Roa J, Herbison AE. Direct regulation of GnRH neuron excitability by arcuate nucleus POMC and NPY neuron neuropeptides in female mice. Endocrinology 2012; 153:5587-99. [PMID: 22948210 DOI: 10.1210/en.2012-1470] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Hypothalamic neuropeptide Y (NPY) and proopiomelanocortin (POMC) neurons act to sense and coordinate the brain's responses to metabolic cues. One neuronal network that is very sensitive to metabolic status is that controlling fertility. In this study, we investigated the impact of neuropeptides released by NPY and POMC neurons on the cellular excitability of GnRH neurons, the final output cells of the brain controlling fertility. The majority (∼70%) of GnRH neurons were activated by α-melanocyte-stimulating hormone, and this resulted from the direct postsynaptic activation of melanocortin receptor 3 and melanocortin receptor 4. A small population of GnRH neurons (∼15%) was excited by cocaine and amphetamine-regulated transcript or inhibited by β-endorphin. Agouti-related peptide, released by NPY neurons, was found to have variable inhibitory (∼10%) and stimulatory (∼25%) effects upon subpopulations of GnRH neurons. A variety of NPY and pancreatic polypeptide analogs was used to examine potential NPY interactions with GnRH neurons. Although porcine NPY (Y1/Y2/Y5 agonist) directly inhibited the firing of approximately 45% of GnRH neurons, [Leu(31),Pro(34)]-NPY (Y1/Y4/Y5 agonist) could excite (56%) or inhibit (19%). Experiments with further agonists indicated that Y1 receptors were responsible for suppressing GnRH neuron activity, whereas postsynaptic Y4 receptors were stimulatory. These results show that the activity of GnRH neurons is regulated in a complex manner by neuropeptides released by POMC and NPY neurons. This provides a direct route through which different metabolic cues can regulate fertility.
Collapse
Affiliation(s)
- Juan Roa
- Centre for Neuroendocrinology, Department of Physiology, University of Otago School of Medical Sciences, Dunedin, New Zealand
| | | |
Collapse
|
39
|
Variables controlling entry into and exit from the steady-state, one of two modes of feeding in Aplysia. PLoS One 2012; 7:e45241. [PMID: 23028872 PMCID: PMC3460933 DOI: 10.1371/journal.pone.0045241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 08/16/2012] [Indexed: 11/19/2022] Open
Abstract
Background Aplysia feeding is a model system for examining the neural mechanisms by which changes in motivational state control behavior. When food is intermittently present, Aplysia eat large meals controlled by a balance between food stimuli exciting feeding and gut stimuli inhibiting feeding. However, when food is continuously present animals are in a state in which feeding is relatively inhibited and animals eat little. We examined which stimuli provided by food and feeding initiate steady-state inhibition of feeding, and which stimuli maintain the inhibition. Results Multiple stimuli were found to control entry into the steady-state inhibition, and its maintenance. The major variable governing entry into the steady-state is fill of the gut with bulk provided by food, but this stimulus cannot alone cause entry into the steady-state. Food odor and nutritional stimuli such as increased hemolymph glucose and L-arginine concentrations also contribute to inhibition of feeding leading to entry into the steady-state. Although food odor can alone cause some inhibition of feeding, it does not amplify the effect of gut fill. By contrast, neither increased hemolymph glucose nor L-arginine alone inhibits feeding in hungry animals, but both amplify the inhibitory effects of food odor, and increased glucose also amplifies the effect of gut fill. The major variable maintaining the steady-state is the continued presence of food odor, which can alone maintain the steady-state for 48–72 hrs. Neither increased glucose nor L-arginine can alone preserve the steady-state, although they partially preserve it. Glucose and arginine partially extend the effect of food odor after 72 hrs. Conclusions These findings show that control of Aplysia feeding is more complex than was previously thought, in that multiple inhibitory factors interact in its control.
Collapse
|
40
|
Begriche K, Marston OJ, Rossi J, Burke LK, McDonald P, Heisler LK, Butler AA. Melanocortin-3 receptors are involved in adaptation to restricted feeding. GENES, BRAIN, AND BEHAVIOR 2012; 11:291-302. [PMID: 22353545 PMCID: PMC3319531 DOI: 10.1111/j.1601-183x.2012.00766.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 01/09/2012] [Accepted: 01/12/2012] [Indexed: 11/29/2022]
Abstract
The central nervous melanocortin system forms a neural network that maintains energy homeostasis. Actions involving neural melanocortin-3 receptors (MC3Rs) regulate the expression rhythms in ingestive behaviors and metabolism anticipating nutrient intake. Here, we characterized the response of Mc3r knockout (Mc3r(-/-)) and wild type (WT) mice to a restricted feeding (RF) schedule where food access was limited to a 4-h period mid light cycle using a mechanical barrier. Mc3r(-/-) mice adapted poorly to the food restriction schedule. Anticipatory activity and the initial bout of intense feeding activity associated with granting food access were attenuated in Mc3r(-/-) mice, resulting in increased weight loss relative to controls. To investigate whether activity in specific hypothalamic nuclei contribute to the Mc3r(-/-) phenotype observed, we assessed hypothalamic FOS-immunoreactivity (FOS-IR) associated with food restriction. Food access markedly increased FOS-IR in the dorsomedial hypothalamus (DMH), but not in the suprachiasmatic or ventromedial hypothalamic nuclei (SCN and VMN, respectively) compared to ad libitum fed mice. Mc3r(-/-) mice displayed a significant reduction in FOS-IR in the DMH during feeding. Analysis of MC3R signaling in vitro indicated dose-dependent stimulation of the extracellular signal-regulated kinase (ERK) pathway by the MC3R agonist d-Trp(8)-γMSH. Treatment of WT mice with d-Trp(8)-γMSH administered intracerebroventricularly increased the number of pERK neurons 1.7-fold in the DMH. These observations provide further support for the involvement of the MC3Rs in regulating adaptation to food restriction. Moreover, MC3Rs may modulate the activity of neurons in the DMH, a region previously linked to the expression of the anticipatory response to RF.
Collapse
Affiliation(s)
- K Begriche
- Department of Metabolism and Aging, The Scripps Research InstituteJupiter, FL, USA
| | - O J Marston
- Department of Pharmacology, University of CambridgeCambridge, United Kingdom
| | - J Rossi
- Department of Metabolism and Aging, The Scripps Research InstituteJupiter, FL, USA
| | - L K Burke
- Department of Pharmacology, University of CambridgeCambridge, United Kingdom
| | - P McDonald
- Department of Molecular Therapeutics and Translational Research Institute, The Scripps Research InstituteJupiter, FL, USA
| | - L K Heisler
- Department of Pharmacology, University of CambridgeCambridge, United Kingdom
| | - A A Butler
- Department of Metabolism and Aging, The Scripps Research InstituteJupiter, FL, USA
| |
Collapse
|
41
|
Biebermann H, Kühnen P, Kleinau G, Krude H. The neuroendocrine circuitry controlled by POMC, MSH, and AGRP. Handb Exp Pharmacol 2012:47-75. [PMID: 22249810 DOI: 10.1007/978-3-642-24716-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Obesity is one of the most challenging health problems worldwide. Over the past few decades, our knowledge concerning mechanisms of weight regulation has increased tremendously leading to the identification of the leptin-melanocortin pathway. The filling level of energy stores is signaled to the brain, and the information is integrated by hypothalamic nuclei, resulting in a well-orchestrated response to food intake and energy expenditure to ensure constant body weight. One of the key players in this system is proopiomelanocortin (POMC), a precursor of a variety of neuropeptides. POMC-derived alpha- and beta-MSH play an important role in energy homeostasis by activating melanocortin receptors expressed in the arcuate nucleus (MC3R) and in the nucleus paraventricularis (MC4R). Activation of these two G protein-coupled receptors is antagonized by agouti-related peptide (AgRP). Naturally occurring mutations in this system were identified in patients suffering from common obesity as well as in patients demonstrating a phenotype of severe early-onset obesity, adrenal insufficiency, red hair, and pale skin. Detailed understanding of the complex system of POMC-AgRP-MC3R-MC4R and their interaction with other hypothalamic as well as peripheral signals is a prerequisite to combat the obesity epidemic.
Collapse
Affiliation(s)
- Heike Biebermann
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | | | | | | |
Collapse
|
42
|
Cansell C, Denis RGP, Joly-Amado A, Castel J, Luquet S. Arcuate AgRP neurons and the regulation of energy balance. Front Endocrinol (Lausanne) 2012; 3:169. [PMID: 23293630 PMCID: PMC3530831 DOI: 10.3389/fendo.2012.00169] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 12/05/2012] [Indexed: 11/13/2022] Open
Abstract
The arcuate nucleus of the hypothalamus contains at least two populations of neurons that continuously monitor signals reflecting energy status and promote the appropriate behavioral and metabolic responses to changes in energy demand. Activation of neurons making pro-opiomelanocortin (POMC) decreases food intake and increases energy expenditure through activation of G protein-coupled melanocortin receptors via the release of α-melanocyte-stimulating hormone. Until recently, the prevailing idea was that the neighboring neurons [agouti-related protein (AgRP) neurons] co-expressing the orexigenic neuropeptides, AgRP, and neuropeptide Y increase feeding by opposing the anorexigenic actions of the POMC neurons. However, it has now been demonstrated that only AgRP neurons activation - not POMC neurons inhibition - is necessary and sufficient to promote feeding. Projections of AgRP-expressing axons innervate mesolimbic, midbrain, and pontine structures where they regulate feeding and feeding-independent functions such as reward or peripheral nutrient partitioning. AgRP neurons also make gamma aminobutyric acid , which is now thought to mediate many of critical functions of these neurons in a melanocortin-independent manner and on a timescale compatible with neuromodulation.
Collapse
Affiliation(s)
- Céline Cansell
- Unité de Biologie Fonctionnelle et Adaptative, CNRS-EAC 4413, Sorbonne Paris Cité, Université Paris Diderot-Paris 7Paris, France
| | - Raphaël G. P. Denis
- Unité de Biologie Fonctionnelle et Adaptative, CNRS-EAC 4413, Sorbonne Paris Cité, Université Paris Diderot-Paris 7Paris, France
| | - Aurélie Joly-Amado
- Unité de Biologie Fonctionnelle et Adaptative, CNRS-EAC 4413, Sorbonne Paris Cité, Université Paris Diderot-Paris 7Paris, France
| | - Julien Castel
- Unité de Biologie Fonctionnelle et Adaptative, CNRS-EAC 4413, Sorbonne Paris Cité, Université Paris Diderot-Paris 7Paris, France
- Centre National de la Recherche Scientifique EAC 4413Paris, France
| | - Serge Luquet
- Unité de Biologie Fonctionnelle et Adaptative, CNRS-EAC 4413, Sorbonne Paris Cité, Université Paris Diderot-Paris 7Paris, France
- Centre National de la Recherche Scientifique EAC 4413Paris, France
- *Correspondence: Serge Luquet, Unité de Biologie Fonctionnelle et Adaptative, CNRS-EAC 4413, Sorbonne Paris Cité, Université Paris Diderot-Paris 7, 4 rue Marie-Andrée Lagroua Weill-Hallé, Bâtiment Buffon, Case courrier 7126, 75205 Paris Cedex 13, France. e-mail:
| |
Collapse
|
43
|
Blum I, Lamont EW, Abizaid A. Competing clocks: Metabolic status moderates signals from the master circadian pacemaker. Neurosci Biobehav Rev 2012; 36:254-70. [DOI: 10.1016/j.neubiorev.2011.06.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 05/27/2011] [Accepted: 06/02/2011] [Indexed: 11/28/2022]
|
44
|
Jo YH. Endogenous BDNF regulates inhibitory synaptic transmission in the ventromedial nucleus of the hypothalamus. J Neurophysiol 2011; 107:42-9. [PMID: 21994261 DOI: 10.1152/jn.00353.2011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Output from steroidogenic factor-1 (SF-1) neurons in the ventromedial nucleus of the hypothalamus (VMH) is anorexigenic. SF-1 neurons express brain-derived neurotrophic factor (BDNF) that contributes to the regulation of food intake and body weight. Here I show that regulation of GABAergic inputs onto SF-1 neurons by endogenous BDNF determines the anorexigenic outcome from the VMH. Single-cell RT-PCR analysis reveals that one-third of SF-1 neurons express BDNF and that only a subset of BDNF-expressing SF-1 neurons coexpresses the melanocortin receptor type 4. Whole cell patch-clamp analysis of SF-1 neurons in the VMH shows that exogenous BDNF significantly increases the frequency of spontaneous GABAergic inhibitory postsynaptic currents (sIPSCs). This enhancement of GABA drive readily decreases the excitability of SF-1 neurons. However, treatment with BDNF has no significant effect on the frequency of TTX-independent GABAergic IPSCs. Moreover, TrkB receptors are not localized at the postsynaptic sites of GABAergic synapses on SF-1 neurons as there is no change in the amplitude of miniature IPSCs in the presence of BDNF. Dual patch-clamp recordings in mouse hypothalamic slices reveal that stimulation of one SF-1 neuron induces an increase in sIPSC frequency onto the neighboring SF-1 neuron. More importantly, this effect is blocked by a tyrosine kinase inhibitor. Hence, this increased GABA drive onto SF-1 neurons may, in part, explain the cellular mechanisms that mediate the anorexigenic effects of BDNF.
Collapse
Affiliation(s)
- Young-Hwan Jo
- Albert Einstein College of Medicine, Dept. of Medicine, Division of Endocrinology, 1300 Morris Park Ave., Bronx, NY 10461, USA.
| |
Collapse
|
45
|
Melanocortin system in cancer-related cachexia. Open Med (Wars) 2011. [DOI: 10.2478/s11536-011-0057-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AbstractThe melanocortin system plays a pivotal role in the regulation of appetite and energy balance. It was recognized to play an important role in the development of cancer-related cachexia, a debilitating condition characterized by progressive body wasting associated with anorexia, increased resting energy expediture and loss of fat as well as lean body mass that cannot be simply prevented or treated by adequate nutritional support.The recent advances in understanding of mechanisms underlying cancer-related cachexia led to consequent recognition of the melanocortin system as an important potential therapeutic target. Several molecules have been made available for animal experiments, including those with oral bioavailability, that act at various checkpoints of the melanocortin system and that might confer singificant benefits for the patients suffering from cancer-related cachexia. The application of melanocortin 4 receptor antagonists/agouti-related peptide agonists has been however restricted to animal models and more pharmacological data will be necessary to progress to clinical trials on humans. Still, pharmacological targeting of the melanocortin system seem to represent an elegant and promising way of treatment of cancer-related cachexia.
Collapse
|
46
|
Resch JM, Boisvert JP, Hourigan AE, Mueller CR, Yi SS, Choi S. Stimulation of the hypothalamic ventromedial nuclei by pituitary adenylate cyclase-activating polypeptide induces hypophagia and thermogenesis. Am J Physiol Regul Integr Comp Physiol 2011; 301:R1625-34. [PMID: 21957159 DOI: 10.1152/ajpregu.00334.2011] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Numerous studies have demonstrated that the hypothalamic ventromedial nuclei (VMN) regulate energy homeostasis by integrating and utilizing behavioral and metabolic mechanisms. The VMN heavily express pituitary adenylate cyclase-activating polypeptide (PACAP) type I receptors (PAC1R). Despite the receptor distribution, most PACAP experiments investigating affects on feeding have focused on intracerebroventricular administration or global knockout mice. To identify the specific contribution of PACAP signaling in the VMN, we injected PACAP directly into the VMN and measured feeding behavior and indices of energy expenditure. Following an acute injection of PACAP, nocturnal food intake was significantly reduced for 6 h after injections without evidence of malaise. In addition, PACAP-induced suppression of feeding also occurred following an overnight fast and could be blocked by a specific PAC1R antagonist. Metabolically, VMN-specific injections of PACAP significantly increased both core body temperature and spontaneous locomotor activity with a concurrent increase in brown adipose uncoupling protein 1 mRNA expression. To determine which signaling pathways were responsive to PACAP administration into the VMN, we measured mRNA expression of well-characterized hypothalamic neuropeptide regulators of feeding. One hour after PACAP administration, expression of pro-opiomelanocortin mRNA was significantly increased in the arcuate nuclei (ARC), with no changes in neuropeptide Y and agouti-related polypeptide mRNA levels. This suggests that PAC1R expressing VMN neurons projecting to pro-opiomelanocortin neurons contribute to hypophagia by involving melanocortin signaling. While the VMN also abundantly express PACAP protein, the present study demonstrates that PACAP input to the VMN can influence the control of energy homeostasis.
Collapse
Affiliation(s)
- Jon M Resch
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | | | | | | | | | | |
Collapse
|
47
|
Saba LM, Bennett B, Hoffman PL, Barcomb K, Ishii T, Kechris K, Tabakoff B. A systems genetic analysis of alcohol drinking by mice, rats and men: influence of brain GABAergic transmission. Neuropharmacology 2011; 60:1269-80. [PMID: 21185315 PMCID: PMC3079014 DOI: 10.1016/j.neuropharm.2010.12.019] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 12/01/2010] [Accepted: 12/15/2010] [Indexed: 11/19/2022]
Abstract
Genetic influences on the predisposition to complex behavioral or physiological traits can reflect genetic polymorphisms that lead to altered gene product function, and/or variations in gene expression levels. We have explored quantitative variations in an animal's alcohol consumption, using a genetical genomic/phenomic approach. In our studies, gene expression is correlated with amount of alcohol consumed, and genomic regions that regulate the alcohol consumption behavior and the quantitative levels of gene expression (behavioral and expression quantitative trait loci [QTL]) are determined and used as a filter to identify candidate genes predisposing the behavior. We determined QTLs for alcohol consumption using the LXS panel of recombinant inbred mice. We then identified genes that were: 1) differentially expressed between five high and five low alcohol-consuming lines or strains of mice; and 2) were physically located in, or had an expression QTL (eQTL) within the alcohol consumption QTLs. Comparison of mRNA and protein levels in brains of high and low alcohol consuming mice led us to a bioinformatic examination of potential regulation by microRNAs of an identified candidate transcript, Gnb1 (G protein beta subunit 1). We combined our current analysis with our earlier work identifying candidate genes for the alcohol consumption trait in mice, rats and humans. Our overall analysis leads us to postulate that the activity of the GABAergic system, and in particular GABA release and GABA receptor trafficking and signaling, which involves G protein function, contributes significantly to genetic variation in the predisposition to varying levels of alcohol consumption. This article is part of a Special Issue entitled 'Trends in neuropharmacology: in memory of Erminio Costa'.
Collapse
Affiliation(s)
- Laura M. Saba
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| | - Beth Bennett
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| | - Paula L. Hoffman
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| | - Kelsey Barcomb
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| | - Takao Ishii
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| | - Katerina Kechris
- Colorado School of Public Health, Campus Box B119, Aurora, CO 80045 USA,
| | - Boris Tabakoff
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| |
Collapse
|
48
|
Breit A, Büch TRH, Boekhoff I, Solinski HJ, Damm E, Gudermann T. Alternative G protein coupling and biased agonism: new insights into melanocortin-4 receptor signalling. Mol Cell Endocrinol 2011; 331:232-40. [PMID: 20674667 DOI: 10.1016/j.mce.2010.07.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Revised: 05/06/2010] [Accepted: 07/13/2010] [Indexed: 11/20/2022]
Abstract
The melanocortin-4 receptor (MC4R) is a prototypical G protein-coupled receptor (GPCR) that plays a considerable role in controlling appetite and energy homeostasis. Signalling initiated by MC4R is orchestrated by multiple agonists, inverse agonism and by interactions with accessory proteins. The exact molecular events translating MC4R signalling into its physiological role, however, are not fully understood. This review is an attempt to summarize new aspects of MC4R signalling in the context of its recently discovered alternative G protein coupling, and to give a perspective on how future research could improve our knowledge about the intertwining molecular mechanisms that are responsible for the regulation of energy homeostasis by the melanocortin system.
Collapse
Affiliation(s)
- Andreas Breit
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Goethestrasse 33, Ludwig-Maximilians-Universität München, 80336 München, Germany.
| | | | | | | | | | | |
Collapse
|
49
|
Myoung HJ, Kim G, Nam KW. Apigenin isolated from the seeds of Perilla frutescens britton var crispa (Benth.) inhibits food intake in C57BL/6J mice. Arch Pharm Res 2010; 33:1741-6. [DOI: 10.1007/s12272-010-1105-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 06/24/2010] [Accepted: 08/06/2010] [Indexed: 01/28/2023]
|
50
|
The nutritional induction of COUP-TFII gene expression in ventromedial hypothalamic neurons is mediated by the melanocortin pathway. PLoS One 2010; 5:e13464. [PMID: 20976162 PMCID: PMC2956692 DOI: 10.1371/journal.pone.0013464] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Accepted: 09/23/2010] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The nuclear receptor chicken ovalbumin upstream promoter transcription factor II (COUP-TFII) is an important coordinator of glucose homeostasis. We report, for the first time, a unique differential regulation of its expression by the nutritional status in the mouse hypothalamus compared to peripheral tissues. METHODOLOGY/PRINCIPAL FINDINGS Using hyperinsulinemic-euglycemic clamps and insulinopenic mice, we show that insulin upregulates its expression in the hypothalamus. Immunofluorescence studies demonstrate that COUP-TFII gene expression is restricted to a subpopulation of ventromedial hypothalamic neurons expressing the melanocortin receptor. In GT1-7 hypothalamic cells, the MC4-R agonist MTII leads to a dose dependant increase of COUP-TFII gene expression secondarily to a local increase in cAMP concentrations. Transfection experiments, using a COUP-TFII promoter containing a functional cAMP responsive element, suggest a direct transcriptional activation by cAMP. Finally, we show that the fed state or intracerebroventricular injections of MTII in mice induce an increased hypothalamic COUP-TFII expression associated with a decreased hepatic and pancreatic COUP-TFII expression. CONCLUSIONS/SIGNIFICANCE These observations strongly suggest that hypothalamic COUP-TFII gene expression could be a central integrator of insulin and melanocortin signaling pathway within the ventromedial hypothalamus. COUP-TFII could play a crucial role in brain integration of circulating signal of hunger and satiety involved in energy balance regulation.
Collapse
|