1
|
Yanik T, Durhan ST. Pro-Opiomelanocortin and Melanocortin Receptor 3 and 4 Mutations in Genetic Obesity. Biomolecules 2025; 15:209. [PMID: 40001512 PMCID: PMC11853658 DOI: 10.3390/biom15020209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Genetic obesity results from loss-of-function mutations, including those affecting the leptin-melanocortin system, which regulates body weight. Pro-opiomelanocortin (POMC)-derived neurohormones act as ligands for melanocortin receptors (MCRs), regulating the leptin-melanocortin pathway through protein-protein interactions. Loss-of-function mutations in the genes encoding POMC, MC3R, and MC4R can lead to the dysregulation of energy expenditure and feeding balance, early-onset obesity, and developmental dysregulation. Recent studies have identified new genetic regulatory mechanisms and potential biomarker regions for the POMC gene and MC4R secondary messenger pathway associated with obesity. Recent advances in crystal structure studies have enhanced our understanding of the protein interactions in this pathway. This narrative review focuses on recent developments in two key areas related to POMC regulation and the leptin-melanocortin pathway: (1) genetic variations in and functions of POMC, and (2) MC3R and MC4R variants that lead to genetic obesity in humans. Understanding these novel mutations in POMC and MC4R/MC3R, as well as their structural and intracellular mechanisms, may help identify strategies for the treatment and diagnosis of obesity, particularly childhood obesity.
Collapse
MESH Headings
- Humans
- Pro-Opiomelanocortin/genetics
- Pro-Opiomelanocortin/metabolism
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/metabolism
- Receptor, Melanocortin, Type 4/chemistry
- Obesity/genetics
- Obesity/metabolism
- Mutation
- Receptor, Melanocortin, Type 3/genetics
- Receptor, Melanocortin, Type 3/metabolism
- Receptor, Melanocortin, Type 3/chemistry
- Leptin/metabolism
- Leptin/genetics
- Animals
Collapse
Affiliation(s)
- Tulin Yanik
- Department of Biological Sciences, Middle East Technical University, Ankara 06800, Türkiye
| | - Seyda Tugce Durhan
- Department of Biochemistry, Middle East Technical University, Ankara 06800, Türkiye;
| |
Collapse
|
2
|
Mirasierra M, Fernández-Pérez A, Lizarbe B, Keiran N, Ruiz-Cañas L, Casarejos MJ, Cerdán S, Vendrell J, Fernández-Veledo S, Vallejo M. Alx3 deficiency disrupts energy homeostasis, alters body composition, and impairs hypothalamic regulation of food intake. Cell Mol Life Sci 2024; 81:343. [PMID: 39129011 PMCID: PMC11335267 DOI: 10.1007/s00018-024-05384-z] [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: 07/10/2023] [Revised: 07/03/2024] [Accepted: 07/27/2024] [Indexed: 08/13/2024]
Abstract
The coordination of food intake, energy storage, and expenditure involves complex interactions between hypothalamic neurons and peripheral tissues including pancreatic islets, adipocytes, muscle, and liver. Previous research shows that deficiency of the transcription factor Alx3 alters pancreatic islet-dependent glucose homeostasis. In this study we carried out a comprehensive assessment of metabolic alterations in Alx3 deficiency. We report that Alx3-deficient mice exhibit decreased food intake without changes in body weight, along with reduced energy expenditure and altered respiratory exchange ratio. Magnetic resonance imaging reveals increased adiposity and decreased muscle mass, which was associated with markers of motor and sympathetic denervation. By contrast, Alx3-deficient mice on a high-fat diet show attenuated weight gain and improved insulin sensitivity, compared to control mice. Gene expression analysis demonstrates altered lipogenic and lipolytic gene profiles. In wild type mice Alx3 is expressed in hypothalamic arcuate nucleus neurons, but not in major peripheral metabolic organs. Functional diffusion-weighted magnetic resonance imaging reveals selective hypothalamic responses to fasting in the arcuate nucleus of Alx3-deficient mice. Additionally, altered expression of proopiomelanocortin and melanocortin-3 receptor mRNA in the hypothalamus suggests impaired regulation of feeding behavior. This study highlights the crucial role for Alx3 in governing food intake, energy homeostasis, and metabolic nutrient partitioning, thereby influencing body mass composition.
Collapse
Affiliation(s)
- Mercedes Mirasierra
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Fernández-Pérez
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
- Centro para el Desarrollo Tecnológico e Industrial (CDTI), Madrid, Spain
| | - Blanca Lizarbe
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain
- Department of Biochemistry, Universidad Autónoma de Madrid, Madrid, Spain
| | - Noelia Keiran
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV) - Hospital Universitari de Tarragona Joan XXIII, Universitat Rovira i Virgili, Tarragona, Spain
| | - Laura Ruiz-Cañas
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain
- Chronic Diseases and Cancer Area 3, Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Madrid, Spain
| | - María José Casarejos
- Neuropharmacology Laboratory, Neurobiology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Madrid, Spain
| | - Sebastián Cerdán
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain
| | - Joan Vendrell
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV) - Hospital Universitari de Tarragona Joan XXIII, Universitat Rovira i Virgili, Tarragona, Spain
| | - Sonia Fernández-Veledo
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV) - Hospital Universitari de Tarragona Joan XXIII, Universitat Rovira i Virgili, Tarragona, Spain
| | - Mario Vallejo
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
3
|
Dietzsch AN, Al-Hasani H, Altschmied J, Bottermann K, Brendler J, Haendeler J, Horn S, Kaczmarek I, Körner A, Krause K, Landgraf K, Le Duc D, Lehmann L, Lehr S, Pick S, Ricken A, Schnorr R, Schulz A, Strnadová M, Velluva A, Zabri H, Schöneberg T, Thor D, Prömel S. Dysfunction of the adhesion G protein-coupled receptor latrophilin 1 (ADGRL1/LPHN1) increases the risk of obesity. Signal Transduct Target Ther 2024; 9:103. [PMID: 38664368 PMCID: PMC11045723 DOI: 10.1038/s41392-024-01810-7] [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: 08/21/2023] [Revised: 03/04/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Obesity is one of the diseases with severe health consequences and rapidly increasing worldwide prevalence. Understanding the complex network of food intake and energy balance regulation is an essential prerequisite for pharmacological intervention with obesity. G protein-coupled receptors (GPCRs) are among the main modulators of metabolism and energy balance. They, for instance, regulate appetite and satiety in certain hypothalamic neurons, as well as glucose and lipid metabolism and hormone secretion from adipocytes. Mutations in some GPCRs, such as the melanocortin receptor type 4 (MC4R), have been associated with early-onset obesity. Here, we identified the adhesion GPCR latrophilin 1 (ADGRL1/LPHN1) as a member of the regulating network governing food intake and the maintenance of energy balance. Deficiency of the highly conserved receptor in mice results in increased food consumption and severe obesity, accompanied by dysregulation of glucose homeostasis. Consistently, we identified a partially inactivating mutation in human ADGRL1/LPHN1 in a patient suffering from obesity. Therefore, we propose that LPHN1 dysfunction is a risk factor for obesity development.
Collapse
Affiliation(s)
- André Nguyen Dietzsch
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Hadi Al-Hasani
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Munich-Neuherberg, Germany
| | - Joachim Altschmied
- Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Cardiovascular Research Institute (CARID), Medical Faculty, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Katharina Bottermann
- Cardiovascular Research Institute (CARID), Medical Faculty, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Pharmacology, Medical Faculty, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jana Brendler
- Institute of Anatomy, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Judith Haendeler
- Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Cardiovascular Research Institute (CARID), Medical Faculty, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Susanne Horn
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Isabell Kaczmarek
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Antje Körner
- Center for Pediatric Research, Hospital for Children and Adolescents, Medical Faculty, Leipzig University, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Kerstin Krause
- Department of Endocrinology, Nephrology, Rheumatology, Leipzig University Medical Center, Leipzig, Germany
| | - Kathrin Landgraf
- Center for Pediatric Research, Hospital for Children and Adolescents, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Diana Le Duc
- Institute of Human Genetics, Leipzig University Medical Center, Leipzig, Germany
| | - Laura Lehmann
- Institute of Cell Biology, Department of Biology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Stefan Lehr
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Munich-Neuherberg, Germany
| | - Stephanie Pick
- Institute of Cell Biology, Department of Biology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Albert Ricken
- Institute of Anatomy, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Rene Schnorr
- Institute of Cell Biology, Department of Biology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Angela Schulz
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Martina Strnadová
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Akhil Velluva
- Institute of Human Genetics, Leipzig University Medical Center, Leipzig, Germany
| | - Heba Zabri
- Institute of Pharmacology, Medical Faculty, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
- School of Medicine, University of Global Health Equity, Kigali, Rwanda
| | - Doreen Thor
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany.
| | - Simone Prömel
- Institute of Cell Biology, Department of Biology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
4
|
Sandbaumhüter F, Nezhyva M, Andrén PE, Jansson ET. Label-Free Quantitative Thermal Proteome Profiling Reveals Target Transcription Factors with Activities Modulated by MC3R Signaling. Anal Chem 2023; 95:15400-15408. [PMID: 37804223 PMCID: PMC10585664 DOI: 10.1021/acs.analchem.3c03643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/20/2023] [Indexed: 10/09/2023]
Abstract
Thermal proteome profiling with label-free quantitation using ion-mobility-enhanced LC-MS offers versatile data sets, providing information on protein differential expression, thermal stability, and the activities of transcription factors. We developed a multidimensional data analysis workflow for label-free quantitative thermal proteome profiling (TPP) experiments that incorporates the aspects of gene set enrichment analysis, differential protein expression analysis, and inference of transcription factor activities from LC-MS data. We applied it to study the signaling processes downstream of melanocortin 3 receptor (MC3R) activation by endogenous agonists derived from the proopiomelanocortin prohormone: ACTH, α-MSH, and γ-MSH. The obtained information was used to map signaling pathways downstream of MC3R and to deduce transcription factors responsible for cellular response to ligand treatment. Using our workflow, we identified differentially expressed proteins and investigated their thermal stability. We found in total 298 proteins with altered thermal stability, resulting from MC3R activation. Out of these, several proteins were transcription factors, indicating them as being downstream target regulators that take part in the MC3R signaling cascade. We found transcription factors CCAR2, DDX21, HMGB2, SRSF7, and TET2 to have altered thermal stability. These apparent target transcription factors within the MC3R signaling cascade play important roles in immune responses. Additionally, we inferred the activities of the transcription factors identified in our data set. This was done with Bayesian statistics using the differential expression data we obtained with label-free quantitative LC-MS. The inferred transcription factor activities were validated in our bioinformatic pipeline by the phosphorylated peptide abundances that we observed, highlighting the importance of post-translational modifications in transcription factor regulation. Our multidimensional data analysis workflow allows for a comprehensive characterization of the signaling processes downstream of MC3R activation. It provides insights into protein differential expression, thermal stability, and activities of key transcription factors. All proteomic data generated in this study are publicly available at DOI: 10.6019/PXD039945.
Collapse
Affiliation(s)
| | - Mariya Nezhyva
- Department
of Pharmaceutical Biosciences, Uppsala University, 751 24 Uppsala, Sweden
| | - Per E. Andrén
- Department
of Pharmaceutical Biosciences, Uppsala University, 751 24 Uppsala, Sweden
- Science
for Life Laboratory, Spatial Mass Spectrometry, Uppsala University, 751 24 Uppsala, Sweden
| | - Erik T. Jansson
- Department
of Pharmaceutical Biosciences, Uppsala University, 751 24 Uppsala, Sweden
| |
Collapse
|
5
|
Romanova IV, Mikhailova EV, Mikhrina AL, Shpakov AO. Type 1 melanocortin receptors in pro-opiomelanocortin-, vasopressin-, and oxytocin-immunopositive neurons in different areas of mouse brain. Anat Rec (Hoboken) 2023; 306:2388-2399. [PMID: 35475324 DOI: 10.1002/ar.24934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/24/2022] [Accepted: 04/04/2022] [Indexed: 10/18/2022]
Abstract
Information on the localization of the Type 1 melanocortin receptors (MC1Rs) in different regions of the brain is very scarce. As a result, the role of MC1Rs in the functioning of brain neurons and in the central regulation of physiological functions has not been studied. This work aimed to study the expression and distribution of MС1Rs in different brain areas of female C57Bl/6J mice. Using real-time polymerase chain reaction, we demonstrated the Mс1R gene expression in the cerebral cortex, midbrain, hypothalamus, medulla oblongata, and hippocampus. Using an immunohistochemical approach, we showed the MС1R localization in neurons of the hypothalamic arcuate, paraventricular and supraoptic nuclei, nucleus tractus solitarius (NTS), dorsal hippocampus, substantia nigra, and cerebral cortex. Using double immunolabeling, the MC1Rs were visualized on the surface and in the bodies and outgrowths of pro-opiomelanocortin (POMC)-immunopositive neurons in the hypothalamic arcuate nucleus, NTS, hippocampal CA3 and CA1 regions, and cerebral cortex. Co-localization with POMC indicates that MC1R, like MC3R, is able to function as an autoreceptor. In the paraventricular and supraoptic nuclei, MC1Rs were visualized on the surface and in the cell bodies of vasopressin- and oxytocin-immunopositive neurons, indicating a relationship between hypothalamic MC1R signaling and vasopressin and oxytocin production. The data obtained indicate a wide distribution of MC1Rs in different areas of the mouse brain and their localization in POMC-, vasopressin- and oxytocin-immunopositive neurons, which may indicate the participation of MC1Rs in the control of many physiological processes in the central nervous system.
Collapse
Affiliation(s)
- Irina V Romanova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Elena V Mikhailova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Anastasiya L Mikhrina
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Alexander O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
6
|
Sweeney P, Gimenez LE, Hernandez CC, Cone RD. Targeting the central melanocortin system for the treatment of metabolic disorders. Nat Rev Endocrinol 2023; 19:507-519. [PMID: 37365323 DOI: 10.1038/s41574-023-00855-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/18/2023] [Indexed: 06/28/2023]
Abstract
A large body of preclinical and clinical data shows that the central melanocortin system is a promising therapeutic target for treating various metabolic disorders such as obesity and cachexia, as well as anorexia nervosa. Setmelanotide, which functions by engaging the central melanocortin circuitry, was approved by the FDA in 2020 for use in certain forms of syndromic obesity. Furthermore, the FDA approvals in 2019 of two peptide drugs targeting melanocortin receptors for the treatment of generalized hypoactive sexual desire disorder (bremelanotide) and erythropoietic protoporphyria-associated phototoxicity (afamelanotide) demonstrate the safety of this class of peptides. These approvals have also renewed excitement in the development of therapeutics targeting the melanocortin system. Here, we review the anatomy and function of the melanocortin system, discuss progress and challenges in developing melanocortin receptor-based therapeutics, and outline potential metabolic and behavioural disorders that could be addressed using pharmacological agents targeting these receptors.
Collapse
Affiliation(s)
- Patrick Sweeney
- School of Molecular and Cellular Biology, College of Liberal Arts and Sciences, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Luis E Gimenez
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | | | - Roger D Cone
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular and Integrative Physiology, School of Medicine, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular, Cellular, and Developmental Biology, College of Literature Science and the Arts, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
7
|
Abstract
Melanocortin 3 receptor (MC3R) is a G-protein coupled receptor which has been defined mostly as a regulator of the appetite/hunger balance mechanisms to date. In addition to its function regarding the weight gain and appetite control mechanisms of MC3R, recent studies have shown that MC3R controls growth, puberty, and circadian rhythms as well. Despite the drastic effects of MC3R deficiency in humans and other mammals, its cellular mechanisms are still under investigation. In this review paper, we aimed to point out the importance of MC3R regulations in three main areas: 1) its impact on weight and appetite control, 2) its role in the control of growth, puberty, and the circadian rhythm, and, 3) its protein-protein interactions and cellular mechanisms.
Collapse
Affiliation(s)
- Tulin Yanik
- Middle East Technical University, Department of Biological Sciences, Ankara, Turkey,* Address for Correspondence: Middle East Technical University, Department of Biological Sciences, Ankara, Turkey Phone: +90 312 210 64 65 E-mail:
| | - Seyda Tugce Durhan
- Middle East Technical University, Department of Biochemistry, Ankara, Turkey
| |
Collapse
|
8
|
Mo H, Yu H, Li Y, Ezeorba TPC, Zhang Z, Yao M, Yu J, Xiong D, Liu H, Wang L. Molecular cloning and functional characterization of melanocortin-3 receptor in grass carp (Ctenopharyngodon idella). FISH PHYSIOLOGY AND BIOCHEMISTRY 2023; 49:155-167. [PMID: 36547499 DOI: 10.1007/s10695-022-01164-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 12/13/2022] [Indexed: 06/17/2023]
Abstract
The melanocortin-3-receptor (MC3R) plays an important role in mammals' food intake and energy homeostasis. However, its physiological role in bony fishes, such as grass carp, has not been well understood. This study reports the molecular cloning, tissue distribution, and pharmacological characterization of grass carp melanocortin-3-receptor (ciMC3R). Phylogenetic and chromosomal synteny analyses indicated that ciMC3R was closest to cyprinid fishes in evolution. Quantitative PCR experiments revealed that the mRNA of ciMC3R was highly expressed in the brain of grass carp. The cytological function of ciMC3R was investigated by the co-transfection of pcDNA3.1-ciMC3R and the signal-pathway-specific luciferase into the HEK293T cells. Results revealed that the four agonists, α-MSH, β-MSH, ACTH, and NDP-MSH, potentiate the activation of ciMC3R and further increase the production of cAMP and upregulate the MAPK/ERK signaling, respectively. Our study will provide basic data for exploring the physiological functions of grass carp MC3R, especially in energy homeostasis and food intake.
Collapse
Affiliation(s)
- Haolin Mo
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China
| | - Huixia Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China
| | - Yang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China.
| | - Timothy P C Ezeorba
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Ihe Nsukka, Nsukka, 41001, Nigeria
| | - Zhihao Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China
| | - Mingxin Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China
| | - Jiajia Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China
| | - Dongmei Xiong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China
| | - Haixia Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China
| | - Lixin Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China.
| |
Collapse
|
9
|
Goit RK, Taylor AW, Yin Lo AC. The central melanocortin system as a treatment target for obesity and diabetes: A brief overview. Eur J Pharmacol 2022; 924:174956. [DOI: 10.1016/j.ejphar.2022.174956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 12/12/2022]
|
10
|
Sweeney P, Bedenbaugh MN, Maldonado J, Pan P, Fowler K, Williams SY, Gimenez LE, Ghamari-Langroudi M, Downing G, Gui Y, Hadley CK, Joy ST, Mapp AK, Simerly RB, Cone RD. The melanocortin-3 receptor is a pharmacological target for the regulation of anorexia. Sci Transl Med 2021; 13:13/590/eabd6434. [PMID: 33883274 DOI: 10.1126/scitranslmed.abd6434] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/19/2020] [Accepted: 03/25/2021] [Indexed: 12/14/2022]
Abstract
Ablation of hypothalamic AgRP (Agouti-related protein) neurons is known to lead to fatal anorexia, whereas their activation stimulates voracious feeding and suppresses other motivational states including fear and anxiety. Despite the critical role of AgRP neurons in bidirectionally controlling feeding, there are currently no therapeutics available specifically targeting this circuitry. The melanocortin-3 receptor (MC3R) is expressed in multiple brain regions and exhibits sexual dimorphism of expression in some of those regions in both mice and humans. MC3R deletion produced multiple forms of sexually dimorphic anorexia that resembled aspects of human anorexia nervosa. However, there was no sexual dimorphism in the expression of MC3R in AgRP neurons, 97% of which expressed MC3R. Chemogenetic manipulation of arcuate MC3R neurons and pharmacologic manipulation of MC3R each exerted potent bidirectional regulation over feeding behavior in male and female mice, whereas global ablation of MC3R-expressing cells produced fatal anorexia. Pharmacological effects of MC3R compounds on feeding were dependent on intact AgRP circuitry in the mice. Thus, the dominant effect of MC3R appears to be the regulation of the AgRP circuitry in both male and female mice, with sexually dimorphic sites playing specialized and subordinate roles in feeding behavior. Therefore, MC3R is a potential therapeutic target for disorders characterized by anorexia, as well as a potential target for weight loss therapeutics.
Collapse
Affiliation(s)
- Patrick Sweeney
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michelle N Bedenbaugh
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN 37240, USA
| | - Jose Maldonado
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN 37240, USA
| | - Pauline Pan
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Katelyn Fowler
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Luis E Gimenez
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Masoud Ghamari-Langroudi
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN 37240, USA
| | - Griffin Downing
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Molecular, Cellular, and Developmental Biology, School of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yijun Gui
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Molecular, Cellular, and Developmental Biology, School of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Colleen K Hadley
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Stephen T Joy
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anna K Mapp
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Chemistry, School of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Richard B Simerly
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN 37240, USA.
| | - Roger D Cone
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA. .,Department of Molecular, Cellular, and Developmental Biology, School of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Molecular and Integrative Physiology, School of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
11
|
Halter B, Ildari N, Cline MA, Gilbert ER. Ferulic acid, a phytochemical with transient anorexigenic effects in birds. Comp Biochem Physiol A Mol Integr Physiol 2021; 259:111015. [PMID: 34119636 DOI: 10.1016/j.cbpa.2021.111015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/07/2021] [Accepted: 06/07/2021] [Indexed: 11/18/2022]
Abstract
Ferulic acid (FA) is a phenolic acid found within the plant cell wall that has physiological benefits as an antioxidant. Although metabolic benefits of FA supplementation are described, lacking are reports of effects on appetite regulation. Thus, our objective was to determine if FA affects food or water intake, using chicks as a model. At 4 days post-hatch, broiler chicks were intraperitoneally injected with 0 (vehicle), 12.5, 25, or 50 mg/kg of FA. Chicks treated with 50 mg/kg of FA consumed 70% less food than controls at 30 min post-injection, and the effect dissipated thereafter. Water intake was not affected at any time. In a behavior analysis, FA-treated chicks defecated fewer times than vehicle-injected chicks, while other behaviors were not affected. There was an increase in c-Fos immunoreactivity within the hypothalamic arcuate nucleus (ARC) of FA-treated chicks, and no differences were detected in other nuclei. mRNA abundance was measured in the whole hypothalamus and the ARC. There was decreased hypothalamic galanin, ghrelin, melanocortin receptor 3, and pro-opiomelanocortin (POMC) mRNA in FA-treated chicks. Within the ARC, there was an increase in c-Fos mRNA and a decrease in POMC mRNA in response to FA. It is likely that the mechanism responsible for mediating FA's transient effects on food intake originates within the ARC, possibly involving POMC. A greater understanding of the short-term, mild appetite-suppressive effects of FA may have applications to treating eating disorders and modulating food intake in animal models of obesity.
Collapse
Affiliation(s)
- Bailey Halter
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| | | | - Mark A Cline
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA; School of Neuroscience, Virginia Tech, Blacksburg, VA, USA
| | - Elizabeth R Gilbert
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA; School of Neuroscience, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
12
|
Oviedo-Ojeda MF, Roque-Jiménez JA, Whalin M, Lee-Rangel HA, Relling AE. Effect of supplementation with different fatty acid profile to the dam in early gestation and to the offspring on the finishing diet on offspring growth and hypothalamus mRNA expression in sheep. J Anim Sci 2021; 99:6153448. [PMID: 33640974 DOI: 10.1093/jas/skab064] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/24/2021] [Indexed: 12/12/2022] Open
Abstract
Supplementation with omega-3 and omega-9 fatty acids (FA) during late gestation regulates offspring development; however, their effect in the first third of gestation is unknown in sheep. The objective of this experiment was to evaluate the effects of the maternal supplementation with an enriched source of monounsaturated FA (MUFA) or an enriched source of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) during the first third of gestation on productive performance on ewes and offspring, and hypothalamic neuropeptides on offspring. Seventy-nine post-weaning lambs, born of sheep supplemented in the first third of gestation with 1.61% Ca salts rich with MUFA or EPA+DHA (dam supplementation, DS), were distributed in a 2×2 factorial arrangement of treatments to finishing diets containing 1.48% of Ca salts of MUFA or EPA+DHA (lamb supplementation, LS). The finishing period of the offspring lasted for 56 d. During the finishing period dry matter intake (DMI, daily) and body weight (BW) were recorded. Plasma was collected for metabolites analysis. Twenty-four lambs were slaughtered, and hypothalamus was collected for mRNA expression of hormone receptors, neuropeptides, and lipid transport genes. The data were analyzed with a mixed model in SAS (9.4) using repeated measurements, when needed. There was a DS×LS interaction for BW (P = 0.10) where LS with EPA+DHA born from DS with MUFA were heavier than the other 3 treatments. Lambs born from DS with MUFA have a greater DMI (P < 0.01) than the offspring born from DS with EPA+DHA. Lambs born from MUFA supplemented dams had a greater (P ≤ 0.05) hypothalamus mRNA expression for cocaine and amphetamine regulated transcript, growth hormone receptor, metastasis suppressor 1, leptin receptor, pro-opiomelanocortin, and Neuropeptide Y. These results indicate that growth depends not on the type of FA during the finishing phase but the interaction of different sources of FA ad different stages. Also, supplementation with FA during early pregnancy changes productive performance and neuropeptides' mRNA expression of lambs independently of the finishing diet.
Collapse
Affiliation(s)
- Mario Francisco Oviedo-Ojeda
- Department of Animal Sciences, The Ohio State University, Ohio Agricultural Research and Development Center (OARDC), Wooster, OH 44691, USA.,Universidad Autónoma de San Luis Potosí, Facultad de Agronomía y Veterinaria, San Luis Potosí 78175, México
| | - José Alejandro Roque-Jiménez
- Department of Animal Sciences, The Ohio State University, Ohio Agricultural Research and Development Center (OARDC), Wooster, OH 44691, USA.,Universidad Autónoma de San Luis Potosí, Facultad de Agronomía y Veterinaria, San Luis Potosí 78175, México
| | - Megan Whalin
- Department of Animal Sciences, The Ohio State University, Ohio Agricultural Research and Development Center (OARDC), Wooster, OH 44691, USA
| | - Héctor Aarón Lee-Rangel
- Universidad Autónoma de San Luis Potosí, Facultad de Agronomía y Veterinaria, San Luis Potosí 78175, México
| | - Alejandro Enrique Relling
- Department of Animal Sciences, The Ohio State University, Ohio Agricultural Research and Development Center (OARDC), Wooster, OH 44691, USA
| |
Collapse
|
13
|
Micioni Di Bonaventura E, Botticelli L, Tomassoni D, Tayebati SK, Micioni Di Bonaventura MV, Cifani C. The Melanocortin System behind the Dysfunctional Eating Behaviors. Nutrients 2020; 12:E3502. [PMID: 33202557 PMCID: PMC7696960 DOI: 10.3390/nu12113502] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022] Open
Abstract
The dysfunction of melanocortin signaling has been associated with obesity, given the important role in the regulation of energy homeostasis, food intake, satiety and body weight. In the hypothalamus, the melanocortin-3 receptor (MC3R) and melanocortin-4 receptor (MC4R) contribute to the stability of these processes, but MC3R and MC4R are also localized in the mesolimbic dopamine system, the region that responds to the reinforcing properties of highly palatable food (HPF) and where these two receptors seem to affect food reward and motivation. Loss of function of the MC4R, resulting from genetic mutations, leads to overeating in humans, but to date, a clear understanding of the underlying mechanisms and behaviors that promote overconsumption of caloric foods remains unknown. Moreover, the MC4R demonstrated to be a crucial modulator of the stress response, factor that is known to be strictly related to binge eating behavior. In this review, we will explore the preclinical and clinical studies, and the controversies regarding the involvement of melanocortin system in altered eating patterns, especially binge eating behavior, food reward and motivation.
Collapse
Affiliation(s)
| | - Luca Botticelli
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| | - Daniele Tomassoni
- School of Bioscience and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy;
| | - Seyed Khosrow Tayebati
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| | | | - Carlo Cifani
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| |
Collapse
|
14
|
Azevedo EP, Tan B, Pomeranz LE, Ivan V, Fetcho R, Schneeberger M, Doerig KR, Liston C, Friedman JM, Stern SA. A limbic circuit selectively links active escape to food suppression. eLife 2020; 9:58894. [PMID: 32894221 PMCID: PMC7476759 DOI: 10.7554/elife.58894] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023] Open
Abstract
Stress has pleiotropic physiologic effects, but the neural circuits linking stress to these responses are not well understood. Here, we describe a novel population of lateral septum neurons expressing neurotensin (LSNts) in mice that are selectively tuned to specific types of stress. LSNts neurons increase their activity during active escape, responding to stress when flight is a viable option, but not when associated with freezing or immobility. Chemogenetic activation of LSNts neurons decreases food intake and body weight, without altering locomotion and anxiety. LSNts neurons co-express several molecules including Glp1r (glucagon-like peptide one receptor) and manipulations of Glp1r signaling in the LS recapitulates the behavioral effects of LSNts activation. Activation of LSNts terminals in the lateral hypothalamus (LH) also decreases food intake. These results show that LSNts neurons are selectively tuned to active escape stress and can reduce food consumption via effects on hypothalamic pathways.
Collapse
Affiliation(s)
- Estefania P Azevedo
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States
| | - Bowen Tan
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States
| | - Lisa E Pomeranz
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States
| | - Violet Ivan
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States
| | - Robert Fetcho
- Department of Psychiatry, Weill Cornell Medical College-New York Presbyterian Hospital, New York, United States
| | - Marc Schneeberger
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States
| | - Katherine R Doerig
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States
| | - Conor Liston
- Department of Psychiatry, Weill Cornell Medical College-New York Presbyterian Hospital, New York, United States
| | - Jeffrey M Friedman
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States.,Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Sarah A Stern
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States
| |
Collapse
|
15
|
Löw K, Roulin A, Kunz S. A proopiomelanocortin-derived peptide sequence enhances plasma stability of peptide drugs. FEBS Lett 2020; 594:2840-2866. [PMID: 32506501 DOI: 10.1002/1873-3468.13855] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/11/2020] [Accepted: 05/26/2020] [Indexed: 12/26/2022]
Abstract
Bioactive peptide drugs hold promise for therapeutic application due to their high potency and selectivity but display short plasma half-life. Examination of selected naturally occurring peptide hormones derived from proteolytic cleavage of the proopiomelanocortin (POMC) precursor lead to the identification of significant plasma-stabilizing properties of a 12-amino acid serine-rich orphan sequence NSSSSGSSGAGQ in human γ3-melanocyte-stimulating hormone (MSH) that is homologous to previously discovered NSn GGH (n = 4-24) sequences in owls. Notably, transfer of this sequence to des-acetyl-α-MSH and the therapeutically relevant peptide hormones neurotensin and glucagon-like peptide-1 likewise enhance their plasma stability without affecting receptor signaling. The stabilizing effect of the sequence module is independent of plasma components, suggesting a direct effect in cis. This natural sequence module may provide a possible strategy to enhance plasma stability, complementing existing methods of chemical modification.
Collapse
Affiliation(s)
- Karin Löw
- Institute of Microbiology, University Hospital Center and University of Lausanne, Switzerland.,Department of Ecology and Evolution, University of Lausanne, Switzerland
| | - Alexandre Roulin
- Department of Ecology and Evolution, University of Lausanne, Switzerland
| | - Stefan Kunz
- Institute of Microbiology, University Hospital Center and University of Lausanne, Switzerland
| |
Collapse
|
16
|
Kadiri JJ, Thapa K, Kaipio K, Cai M, Hruby VJ, Rinne P. Melanocortin 3 receptor activation with [D-Trp8]-γ-MSH suppresses inflammation in apolipoprotein E deficient mice. Eur J Pharmacol 2020; 880:173186. [PMID: 32416182 DOI: 10.1016/j.ejphar.2020.173186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/29/2020] [Accepted: 05/09/2020] [Indexed: 11/18/2022]
Abstract
The melanocortin MC1 and MC3 receptors elicit anti-inflammatory actions in leukocytes and activation of these receptors has been shown to alleviate arterial inflammation in experimental atherosclerosis. Thus, we aimed to investigate whether selective targeting of melanocortin MC3 receptor protects against atherosclerosis. Apolipoprotein E deficient (ApoE-/-) mice were fed high-fat diet for 12 weeks and randomly assigned to receive either vehicle (n = 11) or the selective melanocortin MC3 receptor agonist [D-Trp(8)]-gamma-melanocyte-stimulating hormone ([D-Trp8]-γ-MSH; 15 μg/day, n = 10) for the last 4 weeks. Lesion size as well as macrophage and collagen content in the aortic root plaques were determined. Furthermore, leukocyte counts in the blood and aorta and cytokine mRNA expression levels in the spleen, liver and aorta were quantified. No effect was observed in the body weight development or plasma cholesterol level between the two treatment groups. However, [D-Trp8]-γ-MSH treatment significantly reduced plasma levels of chemokine (C-C motif) ligands 2, 4 and 5. Likewise, cytokine and adhesion molecule expression levels were reduced in the spleen and liver of γ-MSH-treated mice, but not substantially in the aorta. In line with these findings, [D-Trp8]-γ-MSH treatment reduced leukocyte counts in the blood and aorta. Despite reduced inflammation, [D-Trp8]-γ-MSH did not change lesion size, macrophage content or collagen deposition of aortic root plaques. In conclusion, the findings indicate that selective activation of melanocortin MC3 receptor by [D-Trp8]-γ-MSH suppresses systemic and local inflammation and thereby also limits leukocyte accumulation in the aorta. However, the treatment was ineffective in reducing atherosclerotic plaque size.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/pharmacology
- Anti-Inflammatory Agents/therapeutic use
- Aorta/drug effects
- Aorta/immunology
- Aorta/pathology
- Cells, Cultured
- Cholesterol/blood
- Cytokines/blood
- Cytokines/genetics
- Diet, High-Fat
- Endothelial Cells
- Female
- Inflammation/immunology
- Leukocyte Count
- Liver/drug effects
- Liver/immunology
- Melanocyte-Stimulating Hormones/pharmacology
- Melanocyte-Stimulating Hormones/therapeutic use
- Mice, Knockout, ApoE
- Plaque, Atherosclerotic/drug therapy
- Plaque, Atherosclerotic/immunology
- Plaque, Atherosclerotic/pathology
- Receptor, Melanocortin, Type 3/agonists
- Receptor, Melanocortin, Type 3/immunology
- Spleen/drug effects
- Spleen/immunology
Collapse
Affiliation(s)
- James J Kadiri
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Finland; Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Keshav Thapa
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Finland; Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Katja Kaipio
- Department of Pathology, University of Turku, Turku, Finland
| | - Minying Cai
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | - Victor J Hruby
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | - Petteri Rinne
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Finland; Turku Center for Disease Modeling, University of Turku, Turku, Finland.
| |
Collapse
|
17
|
Molecular evolution of the proopiomelanocortin system in Barn owl species. PLoS One 2020; 15:e0231163. [PMID: 32369484 PMCID: PMC7199972 DOI: 10.1371/journal.pone.0231163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/17/2020] [Indexed: 01/20/2023] Open
Abstract
Examination of genetic polymorphisms in outbred wild-living species provides insights into the evolution of complex systems. In higher vertebrates, the proopiomelanocortin (POMC) precursor gives rise to α-, β-, and γ-melanocyte-stimulating hormones (MSH), which are involved in numerous physiological aspects. Genetic defects in POMC are linked to metabolic disorders in humans and animals. In the present study, we undertook an evolutionary genetic approach complemented with biochemistry to investigate the functional consequences of genetic polymorphisms in the POMC system of free-living outbred barn owl species (family Tytonidae) at the molecular level. Our phylogenetic studies revealed a striking correlation between a loss-of-function H9P mutation in the β-MSH receptor-binding motif and an extension of a poly-serine stretch in γ3-MSH to ≥7 residues that arose in the barn owl group 6–8 MYA ago. We found that extension of the poly-serine stretches in the γ-MSH locus affects POMC precursor processing, increasing γ3-MSH production at the expense of γ2-MSH and resulting in an overall reduction of γ-MSH signaling, which may be part of a negative feedback mechanism. Extension of the γ3-MSH poly-serine stretches ≥7 further markedly increases peptide hormone stability in plasma, which is conserved in humans, and is likely relevant to its endocrine function. In sum, our phylogenetic analysis of POMC in wild living owls uncovered a H9P β-MSH mutation subsequent to serine extension in γ3-MSH to 7 residues, which was then followed by further serine extension. The linked MSH mutations highlight the genetic plasticity enabled by the modular design of the POMC gene.
Collapse
|
18
|
Neuronal cAMP/PKA Signaling and Energy Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1090:31-48. [PMID: 30390284 DOI: 10.1007/978-981-13-1286-1_3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The brain plays a key role in the regulation of body weight and glucose metabolism. Peripheral signals including hormones, metabolites, and neural afferent signals are received and processed by the brain which in turn elicits proper behavioral and metabolic responses for maintaining energy and glucose homeostasis. The cAMP/protein kinase A (PKA) pathway acts downstream G-protein-coupled receptors (GPCR) to mediate the physiological effects of many hormones and neurotransmitters. Activated PKA phosphorylates various proteins including ion channels, enzymes, and transcription factors and regulates their activity. Recent studies have shown that neuronal cAMP/PKA activity in multiple brain regions are involved in the regulation of feeding, energy expenditure, and glucose homeostasis. In this chapter I summarize recent genetic and pharmacological studies concerning the regulation of body weight and glucose homeostasis by cAMP/PKA signaling in the brain.
Collapse
|
19
|
Zhang HJ, Xie HJ, Wang W, Wang ZQ, Tao YX. Pharmacology of the giant panda (Ailuropoda melanoleuca) melanocortin-3 receptor. Gen Comp Endocrinol 2019; 277:73-81. [PMID: 30391243 DOI: 10.1016/j.ygcen.2018.10.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/13/2018] [Accepted: 10/30/2018] [Indexed: 11/22/2022]
Abstract
The melanocortin-3 receptor (MC3R) is a member of the G protein-coupled receptor superfamily that plays a critical role in controlling energy balance and metabolism. Although pharmacological characterization of MC3R has been reported previously in several other species, there is no report on the MC3R from giant panda (Ailuropoda melanoleuca). This ancient species is known as a 'living fossil' and is among the most endangered animals in the world. Giant panda survive on a specialized diet of bamboo despite possessing a typical carnivorous digestive system. We report herein the molecular cloning and pharmacological characterization of amMC3R. Homology and phylogenetic analysis showed that amMC3R was highly homologous (>85%) to several other mammalian MC3Rs. Using human MC3R (hMC3R) as a control, the binding of five agonists, [Nle4, D-Phe7]-α-melanocyte stimulating hormone (NDP-MSH), α-, β-, γ-, and D-Trp8-γ-MSH, was investigated, as well as Gs-cAMP and pERK1/2 signaling. The results showed that amMC3R bound NDP- and D-Trp8-γ-MSH with the highest affinity, followed by α-, β-, and γ-MSH, with the same rank order as hMC3R. When stimulated with agonists, amMC3R displayed increased intracellular cAMP and activation of pERK1/2. These data suggest that the cloned amMC3R was a functional receptor. The availability of amMC3R and knowledge of its pharmacological functions will assist further investigation of its role in controlling energy balance and metabolism.
Collapse
Affiliation(s)
- Hai-Jie Zhang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China; Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Hua-Jie Xie
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China
| | - Wei Wang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Zhi-Qiang Wang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People's Republic of China.
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
20
|
Wang W, Guo DY, Lin YJ, Tao YX. Melanocortin Regulation of Inflammation. Front Endocrinol (Lausanne) 2019; 10:683. [PMID: 31649620 PMCID: PMC6794349 DOI: 10.3389/fendo.2019.00683] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/19/2019] [Indexed: 12/18/2022] Open
Abstract
Adrenocorticotropic hormone (ACTH), and α-, β-, and γ-melanocyte-stimulating hormones (α-, β-, γ-MSH), collectively known as melanocortins, together with their receptors (melanocortin receptors), are components of an ancient modulatory system. The clinical use of ACTH in the treatment of rheumatoid arthritis started in 1949, originally thought that the anti-inflammatory action was through hypothalamus-pituitary-adrenal axis and glucocorticoid-dependent. Subsequent decades have witnessed extensive attempts in unraveling the physiology and pharmacology of the melanocortin system. It is now known that ACTH, together with α-, β-, and γ-MSHs, also possess glucocorticoid-independent anti-inflammatory and immunomodulatory effects by activating the melanocortin receptors expressed in the brain or peripheral immune cells. This review will briefly introduce the melanocortin system and highlight the action of melanocortins in the regulation of immune functions from in vitro, in vivo, preclinical, and clinical studies. The potential therapeutic use of melanocortins are also summarized.
Collapse
Affiliation(s)
- Wei Wang
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
| | - Dong-Yu Guo
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
- *Correspondence: Dong-Yu Guo
| | - Yue-Jun Lin
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Ya-Xiong Tao
| |
Collapse
|
21
|
Eerola K, Virtanen S, Vähätalo L, Ailanen L, Cai M, Hruby V, Savontaus M, Savontaus E. Hypothalamic γ-melanocyte stimulating hormone gene delivery reduces fat mass in male mice. J Endocrinol 2018; 239:19–31. [PMID: 30307151 DOI: 10.1530/joe-18-0009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
γ-Melanocyte stimulating hormone (γ-MSH) is an endogenous agonist of the melanocortin 3-receptor (MC3R). Genetic disruption of MC3Rs increases adiposity and blunts responses to fasting, suggesting that increased MC3R signaling could be physiologically beneficial in the long term. Interestingly, several studies have concluded that activation of MC3Rs is orexigenic in the short term. Therefore, we aimed to examine the short- and long-term effects of γ-MSH in the hypothalamic arcuate nucleus (ARC) on energy homeostasis and hypothesized that the effect of MC3R agonism is dependent on the state of energy balance and nutrition. Lentiviral gene delivery was used to induce a continuous expression of γ-Msh only in the ARC of male C57Bl/6N mice. Parameters of body energy homeostasis were monitored as food was changed from chow (6 weeks) to Western diet (13 weeks) and back to chow (7 weeks). The γ-MSH treatment decreased the fat mass to lean mass ratio on chow, but the effect was attenuated on Western diet. After the switch back to chow, an enhanced loss in weight (−15% vs −6%) and fat mass (−37% vs −12%) and reduced cumulative food intake were observed in γ-MSH-treated animals. Fasting-induced feeding was increased on chow diet only; however, voluntary running wheel activity on Western diet was increased. The γ-MSH treatment also modulated the expression of key neuropeptides in the ARC favoring weight loss. We have shown that a chronic treatment intended to target ARC MC3Rs modulates energy balance in nutritional state-dependent manner. Enhancement of diet-induced weight loss could be beneficial in treatment of obesity.
Collapse
Affiliation(s)
- K Eerola
- Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | - S Virtanen
- Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - L Vähätalo
- Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - L Ailanen
- Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Drug Research Doctoral Program, University of Turku, Turku, Finland
| | - M Cai
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, USA
| | - V Hruby
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, USA
| | - M Savontaus
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - E Savontaus
- Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| |
Collapse
|
22
|
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
|
23
|
Gonçalves JPL, Palmer D, Meldal M. MC4R Agonists: Structural Overview on Antiobesity Therapeutics. Trends Pharmacol Sci 2018; 39:402-423. [PMID: 29478721 DOI: 10.1016/j.tips.2018.01.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/28/2018] [Accepted: 01/30/2018] [Indexed: 01/08/2023]
Abstract
The melanocortin-4 receptor (MC4R) regulates adipose tissue formation and energy homeostasis, and is believed to be a monogenic target for novel antiobesity therapeutics. Several research efforts targeting this receptor have identified potent and selective agonists. While viable agonists have been characterized in vitro, undesirable side effects frequently appeared during clinical trials. The most promising candidates have diverse structures, including linear peptides, cyclic peptides, and small molecules. Herein, we present a compilation of potent MC4R agonists and discuss the pivotal structural differences within those molecules that resulted in good selectivity for MC4R over other melanocortins. We provide insight on recent progress in the field and reflect on directions for development of new agonists.
Collapse
Affiliation(s)
- Juliana Pereira Lopes Gonçalves
- Center for Evolutionary Chemical Biology, Department of Chemistry, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark.
| | - Daniel Palmer
- Center for Evolutionary Chemical Biology, Department of Chemistry, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark
| | - Morten Meldal
- Center for Evolutionary Chemical Biology, Department of Chemistry, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark.
| |
Collapse
|
24
|
Neuropeptides, Inflammation, and Diabetic Wound Healing: Lessons from Experimental Models and Human Subjects. CONTEMPORARY DIABETES 2018. [DOI: 10.1007/978-3-319-89869-8_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
25
|
Thomas MA, Xue B. Mechanisms for AgRP neuron-mediated regulation of appetitive behaviors in rodents. Physiol Behav 2017; 190:34-42. [PMID: 29031550 DOI: 10.1016/j.physbeh.2017.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 08/29/2017] [Accepted: 10/06/2017] [Indexed: 12/19/2022]
Abstract
The obesity epidemic is a major health and economic burden facing both developed and developing countries worldwide. Interrogation of the central and peripheral mechanisms regulating ingestive behaviors have primarily focused on food intake, and in the process uncovered a detailed neuroanatomical framework controlling this behavior. However, these studies have largely ignored the behaviors that bring animals, including humans, in contact with food. It is therefore useful to dichotomize ingestive behaviors as appetitive (motivation to find and store food) and consummatory (consumption of food once found), and utilize an animal model that naturally displays these behaviors. Recent advances in genetics have facilitated the identification of several neuronal populations critical for regulating ingestive behaviors in mice, and novel functions of these neurons and neuropeptides in regulating appetitive behaviors in Siberian hamsters, a natural model of food foraging and food hoarding, have been identified. To this end, hypothalamic agouti-related protein/neuropeptide Y expressing neurons (AgRP neurons) have emerged as a critical regulator of ingestive behaviors. Recent studies by Dr. Timothy Bartness and others have identified several discrete mechanisms through which peripheral endocrine signals regulate AgRP neurons to control food foraging, food hoarding, and food intake. We review here recent advances in our understanding of the neuroendocrine control of ingestive behaviors in Siberian hamsters and other laboratory rodents, and identify novel mechanisms through which AgRP neurons mediate appetitive and consummatory behaviors.
Collapse
Affiliation(s)
- M Alex Thomas
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA; Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302, USA
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA; Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302, USA; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA.
| |
Collapse
|
26
|
Convergence between biological, behavioural and genetic determinants of obesity. Nat Rev Genet 2017; 18:731-748. [PMID: 28989171 DOI: 10.1038/nrg.2017.72] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multiple biological, behavioural and genetic determinants or correlates of obesity have been identified to date. Genome-wide association studies (GWAS) have contributed to the identification of more than 100 obesity-associated genetic variants, but their roles in causal processes leading to obesity remain largely unknown. Most variants are likely to have tissue-specific regulatory roles through joint contributions to biological pathways and networks, through changes in gene expression that influence quantitative traits, or through the regulation of the epigenome. The recent availability of large-scale functional genomics resources provides an opportunity to re-examine obesity GWAS data to begin elucidating the function of genetic variants. Interrogation of knockout mouse phenotype resources provides a further avenue to test for evidence of convergence between genetic variation and biological or behavioural determinants of obesity.
Collapse
|
27
|
Xu P, Zhu L, Saito K, Yang Y, Wang C, He Y, Yan X, Hyseni I, Tong Q, Xu Y. Melanocortin 4 receptor is not required for estrogenic regulations on energy homeostasis and reproduction. Metabolism 2017; 70:152-159. [PMID: 28403939 PMCID: PMC5407306 DOI: 10.1016/j.metabol.2016.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/02/2016] [Accepted: 12/05/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND Brain estrogen receptor-α (ERα) is essential for estrogenic regulation of energy homeostasis and reproduction. We previously showed that ERα expressed by pro-opiomelanocortin (POMC) neurons mediates estrogen's effects on food intake, body weight, negative regulation of hypothalamic-pituitary-gonadal axis (HPG axis) and fertility. RESULTS AND CONCLUSIONS We report here that global deletion of a key downstream receptor for POMC peptide, the melanocortin 4 receptor (MC4R), did not affect normal negative feedback regulation of estrogen on the HPG axis, estrous cyclicity and female fertility. Furthermore, loss of the MC4R did not influence estrogenic regulation on food intake and body weight. These results indicate that the MC4R is not required for estrogen's effects on metabolic and reproductive functions.
Collapse
Affiliation(s)
- Pingwen Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030.
| | - Liangru Zhu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Kenji Saito
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Yongjie Yang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Chunmei Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Yanlin He
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Xiaofeng Yan
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Ilirjana Hyseni
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030.
| |
Collapse
|
28
|
Clarifying the Ghrelin System's Ability to Regulate Feeding Behaviours Despite Enigmatic Spatial Separation of the GHSR and Its Endogenous Ligand. Int J Mol Sci 2017; 18:ijms18040859. [PMID: 28422060 PMCID: PMC5412441 DOI: 10.3390/ijms18040859] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/04/2017] [Accepted: 04/11/2017] [Indexed: 12/23/2022] Open
Abstract
Ghrelin is a hormone predominantly produced in and secreted from the stomach. Ghrelin is involved in many physiological processes including feeding, the stress response, and in modulating learning, memory and motivational processes. Ghrelin does this by binding to its receptor, the growth hormone secretagogue receptor (GHSR), a receptor found in relatively high concentrations in hypothalamic and mesolimbic brain regions. While the feeding and metabolic effects of ghrelin can be explained by the effects of this hormone on regions of the brain that have a more permeable blood brain barrier (BBB), ghrelin produced within the periphery demonstrates a limited ability to reach extrahypothalamic regions where GHSRs are expressed. Therefore, one of the most pressing unanswered questions plaguing ghrelin research is how GHSRs, distributed in brain regions protected by the BBB, are activated despite ghrelin’s predominant peripheral production and poor ability to transverse the BBB. This manuscript will describe how peripheral ghrelin activates central GHSRs to encourage feeding, and how central ghrelin synthesis and ghrelin independent activation of GHSRs may also contribute to the modulation of feeding behaviours.
Collapse
|
29
|
Demidowich AP, Jun JY, Yanovski JA. Polymorphisms and mutations in the melanocortin-3 receptor and their relation to human obesity. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2468-2476. [PMID: 28363697 DOI: 10.1016/j.bbadis.2017.03.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 12/18/2022]
Abstract
Inactivating mutations in the melanocortin 3 receptor (Mc3r) have been described as causing obesity in mice, but the physiologic effects of MC3R mutations in humans have been less clear. Here we review the MC3R polymorphisms and mutations identified in humans, and the in vitro, murine, and human cohort studies examining their putative effects. Some, but not all, studies suggest that the common human MC3R variant T6K+V81I, as well as several other rare, function-altering mutations, are associated with greater adiposity and hyperleptinemia with altered energy partitioning. In vitro, the T6K+V81I variant appears to decrease MC3R expression and therefore cAMP generation in response to ligand binding. Knockin mouse studies confirm that the T6K+V81I variant increases feeding efficiency and the avidity with which adipocytes derived from bone or adipose tissue stem cells store triglycerides. Other MC3R mutations occur too infrequently in the human population to make definitive conclusions regarding their clinical effects. This article is part of a Special Issue entitled: Melanocortin Receptors - edited by Ya-Xiong Tao.
Collapse
Affiliation(s)
- Andrew P Demidowich
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Joo Yun Jun
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Jack A Yanovski
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
30
|
Navarro M. The Role of the Melanocortin System in Drug and Alcohol Abuse. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 136:121-150. [DOI: 10.1016/bs.irn.2017.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
31
|
You P, Hu H, Chen Y, Zhao Y, Yang Y, Wang T, Xing R, Shao Y, Zhang W, Li D, Chen H, Liu M. Effects of Melanocortin 3 and 4 Receptor Deficiency on Energy Homeostasis in Rats. Sci Rep 2016; 6:34938. [PMID: 27713523 PMCID: PMC5054679 DOI: 10.1038/srep34938] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/20/2016] [Indexed: 01/08/2023] Open
Abstract
Melanocortin-3 and 4 receptors (MC3R and MC4R) can regulate energy homeostasis, but their respective roles especially the functions of MC3R need more exploration. Here Mc3r and Mc4r single and double knockout (DKO) rats were generated using CRISPR-Cas9 system. Metabolic phenotypes were examined and data were compared systematically. Mc3r KO rats displayed hypophagia and decreased body weight, while Mc4r KO and DKO exhibited hyperphagia and increased body weight. All three mutants showed increased white adipose tissue mass and adipocyte size. Interestingly, although Mc3r KO did not show a significant elevation in lipids as seen in Mc4r KO, DKO displayed even higher lipid levels than Mc4r KO. DKO also showed more severe glucose intolerance and hyperglycaemia than Mc4r KO. These data demonstrated MC3R deficiency caused a reduction of food intake and body weight, whereas at the same time exhibited additive effects on top of MC4R deficiency on lipid and glucose metabolism. This is the first phenotypic analysis and systematic comparison of Mc3r KO, Mc4r KO and DKO rats on a homogenous genetic background. These mutant rats will be important in defining the complicated signalling pathways of MC3R and MC4R. Both Mc4r KO and DKO are good models for obesity and diabetes research.
Collapse
Affiliation(s)
- Panpan You
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Handan Hu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Yuting Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Yongliang Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Yiqing Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Tongtong Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Roumei Xing
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Yanjiao Shao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Wen Zhang
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Huaqing Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China.,Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas 77030, USA
| |
Collapse
|
32
|
Leone S, Noera G, Bertolini A. Developments and new vistas in the field of melanocortins. Biomol Concepts 2016; 6:361-82. [PMID: 26479351 DOI: 10.1515/bmc-2015-0023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/25/2015] [Indexed: 12/14/2022] Open
Abstract
Melanocortins play a fundamental role in several basic functions of the organism (sexual activity, feeding, inflammation and immune responses, pain sensitivity, response to stressful situations, motivation, attention, learning, and memory). Moreover, a large body of animal data, some of which were also confirmed in humans, unequivocally show that melanocortins also have impressive therapeutic effects in several pathological conditions that are the leading cause of mortality and disability worldwide (hemorrhagic, or anyway hypovolemic, shock; septic shock; respiratory arrest; cardiac arrest; ischemia- and ischemia/reperfusion-induced damage of the brain, heart, intestine, and other organs; traumatic injury of brain, spinal cord, and peripheral nerves; neuropathic pain; toxic neuropathies; gouty arthritis; etc.). Recent data obtained in animal models seem to moreover confirm previous hypotheses and preliminary data concerning the neurotrophic activity of melanocortins in neurodegenerative diseases, in particular Alzheimer's disease. Our aim was (i) to critically reconsider the established extrahormonal effects of melanocortins (on sexual activity, feeding, inflammation, tissue hypoperfusion, and traumatic damage of central and peripheral nervous system) at the light of recent findings, (ii) to review the most recent advancements, particularly on the effects of melanocortins in models of neurodegenerative diseases, (iii) to discuss the reasons that support the introduction into clinical practice of melanocortins as life-saving agents in shock conditions and that suggest to verify in clinical setting the impressive results steadily obtained with melanocortins in different animal models of tissue ischemia and ischemia/reperfusion, and finally, (iv) to mention the advisable developments, particularly in terms of selectivity of action and of effects.
Collapse
|
33
|
Melanocortin 3 Receptor Signaling in Midbrain Dopamine Neurons Increases the Motivation for Food Reward. Neuropsychopharmacology 2016; 41:2241-51. [PMID: 26852738 PMCID: PMC4946052 DOI: 10.1038/npp.2016.19] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/31/2016] [Accepted: 02/01/2016] [Indexed: 01/21/2023]
Abstract
The central melanocortin (MC) system mediates its effects on food intake via MC3 (MC3R) and MC4 receptors (MC4R). Although the role of MC4R in meal size determination, satiation, food preference, and motivation is well established, the involvement of MC3R in the modulation of food intake has been less explored. Here, we investigated the role of MC3R on the incentive motivation for food, which is a crucial component of feeding behavior. Dopaminergic neurons within the ventral tegmental area (VTA) have a crucial role in the motivation for food. We here report that MC3Rs are expressed on VTA dopaminergic neurons and that pro-opiomelanocortinergic (POMC) neurons in the arcuate nucleus of the hypothalamus (Arc) innervate these VTA dopaminergic neurons. Our findings show that intracerebroventricular or intra-VTA infusion of the selective MC3R agonist γMSH increases responding for sucrose under a progressive ratio schedule of reinforcement, but not free sucrose consumption in rats. Furthermore, ex vivo electrophysiological recordings show increased VTA dopaminergic neuronal activity upon γMSH application. Consistent with a dopamine-mediated effect of γMSH, the increased motivation for sucrose after intra-VTA infusion of γMSH was blocked by pretreatment with the dopamine receptor antagonist α-flupenthixol. Taken together, we demonstrate an Arc POMC projection onto VTA dopaminergic neurons that modulates motivation for palatable food via activation of MC3R signaling.
Collapse
|
34
|
Gómez-SanMiguel AB, Villanúa MÁ, Martín AI, López-Calderón A. D-TRP(8)-γMSH Prevents the Effects of Endotoxin in Rat Skeletal Muscle Cells through TNFα/NF-KB Signalling Pathway. PLoS One 2016; 11:e0155645. [PMID: 27177152 PMCID: PMC4866687 DOI: 10.1371/journal.pone.0155645] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 05/02/2016] [Indexed: 02/07/2023] Open
Abstract
Sepsis induces anorexia and muscle wasting secondary to an increase in muscle proteolysis. Melanocyte stimulating hormones (MSH) is a family of peptides that have potent anti-inflammatory effects. Melanocortin receptor-3 (MC3-R) has been reported as the predominant anti-inflammatory receptor for melanocortins. The aim of this work was to analyse whether activation of MC3-R, by administration of its agonist D-Trp(8)-γMSH, is able to modify the response of skeletal muscle to inflammation induced by lipopolysaccharide endotoxin (LPS) or TNFα. Adult male rats were injected with 250 μg/kg LPS and/or 500 μg/kg D-Trp(8)-γMSH 17:00 h and at 8:00 h the following day, and euthanized 4 hours afterwards. D-Trp(8)-γMSH decreased LPS-induced anorexia and prevented the stimulatory effect of LPS on hypothalamic IL-1β, COX-2 and CRH as well as on serum ACTH and corticosterone. Serum IGF-I and its expression in liver and gastrocnemius were decreased in rats injected with LPS, but not in those that also received D-Trp(8)-γMSH. However, D-Trp(8)-γMSH was unable to modify the effect of LPS on IGFBP-3. In the gastrocnemius D-Trp(8)-γMSH blocked LPS-induced decrease in pAkt, pmTOR, MHC I and MCH II, as well as the increase in pNF-κB(p65), FoxO1, FoxO3, LC3b, Bnip-3, Gabarap1, atrogin-1, MuRF1 and in LC3a/b lipidation. In L6 myotube cultures, D-Trp(8)-γMSH was able to prevent TNFα-induced increase of NF-κB(p65) phosphorylation and decrease of Akt phosphorylation as well as of IGF-I and MHC I expression. These data suggest that MC3-R activation prevents the effect of endotoxin on skeletal wasting by modifying inflammation, corticosterone and IGF-I responses and also by directly acting on muscle cells through the TNFα/NF-κB(p65) pathway.
Collapse
Affiliation(s)
- Ana Belén Gómez-SanMiguel
- Department of Physiology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - María Ángeles Villanúa
- Department of Physiology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Ana Isabel Martín
- Department of Physiology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Asunción López-Calderón
- Department of Physiology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- * E-mail:
| |
Collapse
|
35
|
Gómez-SanMiguel AB, Martín AI, Nieto-Bona MP, Fernández-Galaz C, Villanúa MÁ, López-Calderón A. The melanocortin receptor type 3 agonist d-Trp(8)-γMSH decreases inflammation and muscle wasting in arthritic rats. J Cachexia Sarcopenia Muscle 2016; 7:79-89. [PMID: 27066320 PMCID: PMC4799854 DOI: 10.1002/jcsm.12036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 03/12/2015] [Accepted: 03/30/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Chronic inflammatory diseases induce cachexia that increases mortality and morbidity of the illness. Adjuvant-induced arthritis is an experimental model of rheumatoid arthritis that is associated with body weight loss and muscle wasting. Alpha-melanocyte stimulating hormone has an anti-inflammatory effect in arthritic rats and decreases muscle wasting. The aim of this work was to elucidate whether the anti-cachectic action of alpha-melanocyte stimulating hormone is mediated by the melanocortin receptor type 3 pathway. METHODS Arthritis was induced in male Wistar rats by intradermal injection of Freund's adjuvant, and 6 days afterwards, arthritic rats were injected with the selective melanocortin receptor type 3 agonist d-Trp(8)-gammaMSH ( d-Trp(8)-γMSH) 500 µg/kg subcutaneously. or saline twice a day, for 10 days. RESULTS d-Trp(8)-γMSH decreased the external signs of inflammation and body weight loss, but it was not able to modify the anorexigenic effect of arthritis or the increase in hypothalamic cyclooxygenase-2 (COX-2) expression. In contrast, d-Trp(8)-γMSH prevented arthritis-induced increase in hypothalamic IL-1β and serum corticosterone levels and the decrease in serum IGF-I levels. d-Trp(8)-γMSH treatment also prevented arthritis-induced NF-kB(p65) phosphorylation and tumour necrosis factor-α mRNA increase in the gastrocnemius. d-Trp(8)-γMSH administration to arthritic rats increased gastrocnemius mass, its cross-sectional area, and mean fast fibre area. Those effects of d-Trp(8)-γMSH were associated with a decreased expression of atrogin-1 and muscle ring-finger protein-1 in the gastrocnemius. In rats treated with saline, arthritis increased the expression of autophagy marker genes LC3b, Bnip-3, and Gabarap1 as well as the conversion of LC3b I to LC3b II by lipidation in the gastrocnemius. d-Trp(8)-γMSH decreased gastrocnemius LC3b, Bnip-3, and Gabarap1 mRNA expression and prevented the increase in LC3b II in arthritic rats. CONCLUSION These data suggest that d-Trp(8)-γMSH administration prevents the effect of arthritis on corticosterone and insulin-like growth factor-I serum levels and decreases muscle wasting, by down-regulating atrogenes and autophagy through modifying the NF-kB(p65)/tumour necrosis factor-α signalling transduction pathway.
Collapse
Affiliation(s)
| | - Ana Isabel Martín
- Department of Physiology, Faculty of Medicine Complutense University Madrid Spain
| | - María Paz Nieto-Bona
- Department of Basic Sciences in Health, Faculty of Health Sciences Rey Juan Carlos University Madrid Spain
| | | | | | | |
Collapse
|
36
|
Mutations in Melanocortin-3 Receptor Gene and Human Obesity. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 140:97-129. [DOI: 10.1016/bs.pmbts.2016.01.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
37
|
Cai M, Marelli UK, Bao J, Beck JG, Opperer F, Rechenmacher F, McLeod KR, Zingsheim MR, Doedens L, Kessler H, Hruby VJ. Systematic Backbone Conformational Constraints on a Cyclic Melanotropin Ligand Leads to Highly Selective Ligands for Multiple Melanocortin Receptors. J Med Chem 2015. [PMID: 26218460 DOI: 10.1021/acs.jmedchem.5b00102] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human melanocortin receptors (hMCRs) have been challenging targets to develop ligands that are explicitly selective for each of their subtypes. To modulate the conformational preferences of the melanocortin ligands and improve the biofunctional agonist/antagonist activities and selectivities, we have applied a backbone N-methylation approach on Ac-Nle-c[Asp-His-D-Nal(2')-Arg-Trp-Lys]-NH2 (Ac-Nle(4)-c[Asp(5),D-Nal(2')(7),Lys(10)]-NH2), a nonselective cyclic peptide antagonist at hMC3R and hMC4R and an agonist at hMC1R and hMC5R. Systematic N-methylated derivatives of Ac-Nle(4)-c[Asp(5),D-Nal(2')(7),Lys(10)]-NH2, with all possible backbone N-methylation combinations, have been synthesized and examined for their binding and functional activities toward melanocortin receptor subtypes 1, 3, 4, and 5 (hMCRs). Several N-methylated analogues are selective and potent agonists or antagonists for hMC1R or hMC5R or have selective antagonist activity for hMC3R. The selective hMC1R ligands show strong binding for human melanoma cells. We have also discovered the first universal antagonist (compound 19) for all subtypes of hMCRs.
Collapse
Affiliation(s)
- Minying Cai
- Department of Chemistry and Biochemistry, University of Arizona , Tucson, Arizona 85721, United States
| | - Udaya Kiran Marelli
- Institute for Advanced Study (IAS) and Center for Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München , 85747 Garching, Germany
| | - Jennifer Bao
- Department of Chemistry and Biochemistry, University of Arizona , Tucson, Arizona 85721, United States
| | - Johannes G Beck
- Institute for Advanced Study (IAS) and Center for Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München , 85747 Garching, Germany
| | - Florian Opperer
- Institute for Advanced Study (IAS) and Center for Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München , 85747 Garching, Germany
| | - Florian Rechenmacher
- Institute for Advanced Study (IAS) and Center for Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München , 85747 Garching, Germany
| | - Kaitlyn R McLeod
- Department of Chemistry and Biochemistry, University of Arizona , Tucson, Arizona 85721, United States
| | - Morgan R Zingsheim
- Department of Chemistry and Biochemistry, University of Arizona , Tucson, Arizona 85721, United States
| | - Lucas Doedens
- Institute for Advanced Study (IAS) and Center for Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München , 85747 Garching, Germany
| | - Horst Kessler
- Institute for Advanced Study (IAS) and Center for Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München , 85747 Garching, Germany.,Department of Chemistry, Faculty of Science, King Abdulaziz University , 21589 Jeddah, Saudi Arabia
| | - Victor J Hruby
- Department of Chemistry and Biochemistry, University of Arizona , Tucson, Arizona 85721, United States
| |
Collapse
|
38
|
Wang H, Semenova S, Kuusela S, Panula P, Lehtonen S. Tankyrases regulate glucoregulatory mechanisms and somatic growth via the central melanocortin system in zebrafish larvae. FASEB J 2015; 29:4435-48. [PMID: 26169937 DOI: 10.1096/fj.15-271817] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/30/2015] [Indexed: 11/11/2022]
Abstract
The central melanocortin system is a key regulator of energy homeostasis. Recent studies indicate that tankyrases (TNKSs), which poly(ADP-ribosyl)ate target proteins and direct them toward proteasomal degradation, affect overall metabolism, but the exact molecular mechanisms remain unclear. We used zebrafish larvae as a model to study the mechanisms by which TNKS1b, the zebrafish ortholog of mammalian TNKS1, regulates glucose homeostasis and somatic growth. In situ hybridization revealed that TNKS1b mRNA is prominently expressed in the hypothalamus and pituitary of the embryonic and larval brain. In the pituitary, TNKS1b is coexpressed with pro-opiomelanocortin a (pomca) gene in corticotropes and melanotropes. Knockdown of TNKS1b reduced the linear growth of the larvae, stimulated insulin gene and glucose transporter 4 protein, and suppressed gluconeogenic phosphoenolpyruvate carboxykinase 1 gene. This result indicates rapid glucose utilization and reduction of gluconeogenesis in TNKS1b-deficient larvae. Knockdown of TNKS1b down-regulated pomca expression and diminished α-melanocyte-stimulating hormone in the pars intermedia. Furthermore, down-regulation of TNKS1b suppressed the expression of melanocortin receptor 3 and increased the expression of melanocortin receptor 4. The collective data suggest that TNKS1b modulates glucoregulatory mechanisms and the somatic growth of zebrafish larvae via the central melanocortin system.
Collapse
Affiliation(s)
- Hong Wang
- *Department of Pathology and Institute of Biomedicine, Anatomy and Neuroscience Centre, University of Helsinki, Helsinki, Finland
| | - Svetlana Semenova
- *Department of Pathology and Institute of Biomedicine, Anatomy and Neuroscience Centre, University of Helsinki, Helsinki, Finland
| | - Sara Kuusela
- *Department of Pathology and Institute of Biomedicine, Anatomy and Neuroscience Centre, University of Helsinki, Helsinki, Finland
| | - Pertti Panula
- *Department of Pathology and Institute of Biomedicine, Anatomy and Neuroscience Centre, University of Helsinki, Helsinki, Finland
| | - Sanna Lehtonen
- *Department of Pathology and Institute of Biomedicine, Anatomy and Neuroscience Centre, University of Helsinki, Helsinki, Finland
| |
Collapse
|
39
|
|
40
|
Central vagal afferent endings mediate reduction of food intake by melanocortin-3/4 receptor agonist. J Neurosci 2014; 34:12636-45. [PMID: 25232103 DOI: 10.1523/jneurosci.1121-14.2014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Injection of the melanocortin-3/4 receptor agonist melanotan-II (MTII) into the nucleus of the solitary tract (NTS) produces rapid and sustained reduction of food intake. Melanocortin-4 receptors (MC4Rs) are expressed by vagal afferent endings in the NTS, but it is not known whether these endings participate in MTII-induced reduction of food intake. In experiments described here, we evaluated the contribution of central vagal afferent endings in MTII-induced reduction of food intake. Examination of rat hindbrain sections revealed that neuronal processes expressing immunoreactivity for the endogenous MC4R agonist α-melanoctyte-stimulating hormone course parallel and wrap around anterogradely labeled vagal afferent endings in the NTS and thus are aptly positioned to activate vagal afferent MC4Rs. Furthermore, MTII and endogenous MC4R agonists increased protein kinase A (PKA)-catalyzed phosphorylation of synapsin I in vagal afferent endings, an effect known to increase synaptic strength by enhancing neurotransmitter release in other neural systems. Hindbrain injection of a PKA inhibitor, KT5720, significantly attenuated MTII-induced reduction of food intake and the increase in synapsin I phosphorylation. Finally, unilateral nodose ganglion removal, resulting in degeneration of vagal afferent endings in the ipsilateral NTS, abolished MTII-induced synapsin I phosphorylation ipsilateral to nodose ganglion removal. Moreover, reduction of food intake following MTII injection into the NTS ipsilateral to nodose ganglion removal was significantly attenuated, whereas the response to MTII was not diminished when injected into the contralateral NTS. Altogether, our results suggest that reduction of food intake following hindbrain MC4R activation is mediated by central vagal afferent endings.
Collapse
|
41
|
Olney JJ, Navarro M, Thiele TE. Targeting central melanocortin receptors: a promising novel approach for treating alcohol abuse disorders. Front Neurosci 2014; 8:128. [PMID: 24917782 PMCID: PMC4042890 DOI: 10.3389/fnins.2014.00128] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 05/08/2014] [Indexed: 12/30/2022] Open
Abstract
The melanocortin (MC) peptides are produced centrally by propiomelanocortin (POMC) neurons within the arcuate nucleus of the hypothalamus and act through five seven-transmembrane G-protein coupled melanocortin receptor (MCR) subtypes. The MC3R and MC4R subtypes, the most abundant central MCRs, are widely expressed in brain regions known to modulate neurobiological responses to ethanol, including regions of the hypothalamus and extended amygdala. Agouti-related protein (AgRP), also produced in the arcuate nucleus, is secreted in terminals expressing MCRs and functions as an endogenous MCR antagonist. This review highlights recent genetic and pharmacological findings that have implicated roles for the MC and AgRP systems in modulating ethanol consumption. Ethanol consumption is associated with significant alterations in the expression levels of various MC peptides/protein, which suggests that ethanol-induced perturbations of MC/AgRP signaling may modulate excessive ethanol intake. Consistently, MCR agonists decrease, and AgRP increases, ethanol consumption in mice. MCR agonists fail to blunt ethanol intake in mutant mice lacking the MC4R, suggesting that the protective effects of MCR agonists are modulated by the MC4R. Interestingly, recent evidence reveals that MCR agonists are more effective at blunting binge-like ethanol intake in mutant mice lacking the MC3R, suggesting that the MC3R has opposing effects on the MC4R. Finally, mutant mice lacking AgRP exhibit blunted voluntary and binge-like ethanol drinking, consistent with pharmacological studies. Collectively, these preclinical observations provide compelling evidence that compounds that target the MC system may provide therapeutic value for treating alcohol abuse disorders and that the utilization of currently available MC-targeting compounds- such as those being used to treat eating disorders- may be used as effective treatments to this end.
Collapse
Affiliation(s)
- Jeffrey J Olney
- Department of Psychology, University of North Carolina Chapel Hill, NC, USA
| | - Montserrat Navarro
- Department of Psychology, University of North Carolina Chapel Hill, NC, USA
| | - Todd E Thiele
- Department of Psychology, University of North Carolina Chapel Hill, NC, USA ; Bowles Center for Alcohol Studies, University of North Carolina Chapel Hill, NC, USA
| |
Collapse
|
42
|
Olney JJ, Sprow GM, Navarro M, Thiele TE. The protective effects of the melanocortin receptor (MCR) agonist, melanotan-II (MTII), against binge-like ethanol drinking are facilitated by deletion of the MC3 receptor in mice. Neuropeptides 2014; 48:47-51. [PMID: 24290566 PMCID: PMC3946855 DOI: 10.1016/j.npep.2013.11.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 10/24/2013] [Accepted: 11/07/2013] [Indexed: 01/22/2023]
Abstract
Recent data have implicated the melanocortin (MC) system in modulating voluntary ethanol consumption. Administration of melanotan-II (MTII), a nonselective melanocortin receptor (MCR) agonist, reduces voluntary ethanol consumption in C57BL/6J mice. Previous studies have demonstrated that central infusion of MTII effectively reduced voluntary ethanol drinking in mutant mice lacking normal expression of MC3R (MC3R-/- mice) but failed to alter ethanol drinking in mice lacking expression of MC4R, demonstrating that central MTII administration reduces voluntary ethanol drinking by signaling through the MC4R. However, evidence shows that the neurocircuitry recruited during excessive binge-like ethanol drinking versus moderate ethanol drinking are not identical. Thus the present study sought to investigate the potential role of the MC3R in binge-like ethanol intake. To this end, the "drinking in the dark" (DID) procedure, a commonly used animal model of binge-like ethanol drinking, was employed. Wild-type MC3R+/+ and MC3R-/- mice were given intracerebroventricular (i.c.v.) infusion of MTII (0.0, 0.25, 0.50, or 1.0 μg) prior to the onset of a 4-h testing period in which mice were given access to 20% (v/v) ethanol. Immediately after the 4-h testing period, tail blood samples were collected from each animal in order to assess blood ethanol concentrations (BECs). Consistent with previous findings, central administration of MTII blunted binge-like ethanol drinking in both MC3R+/+ and MC3R-/- mice. Interestingly, all doses of MTII blunted binge-like ethanol drinking in MC3R-/- mice during the first hour of testing, while only the 1.0 μg dose reduced binge-like drinking in MC3R+/+ mice. Thus, MC3R-/- mice were more sensitive to the protective effects of MTII. These data suggest that MC3Rs oppose the protective effects of MTII against binge-like ethanol drinking, and thus selective MC3R antagonists may have potential therapeutic roles in treating excessive ethanol drinking.
Collapse
Affiliation(s)
- J J Olney
- Department of Psychology, University of North Carolina, Chapel Hill, NC, USA
| | - G M Sprow
- Department of Psychology, University of North Carolina, Chapel Hill, NC, USA
| | - M Navarro
- Department of Psychology, University of North Carolina, Chapel Hill, NC, USA
| | - T E Thiele
- Department of Psychology, University of North Carolina, Chapel Hill, NC, USA; Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
43
|
Jeong JK, Diano S. Prolyl carboxypeptidase and its inhibitors in metabolism. Trends Endocrinol Metab 2013; 24:61-7. [PMID: 23245768 PMCID: PMC3893043 DOI: 10.1016/j.tem.2012.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 10/30/2012] [Accepted: 11/04/2012] [Indexed: 01/09/2023]
Abstract
Proopiomelanocortin (POMC)-expressing neurons in the hypothalamus integrate a variety of central and peripheral metabolic inputs, and regulate energy homeostasis by controlling energy expenditure and food intake. To accomplish this, a precise balance of production and degradation of α-melanocyte-stimulating hormone (α-MSH), an anorexigenic neuropeptide and product of the POMC gene, in the hypothalamus, is crucial. Prolyl carboxypeptidase (PRCP) is a key enzyme that degrades α-MSH to an inactive form unable to inhibit food intake. Because it represents a new therapeutic target for the treatment of metabolic disorders, such as obesity and diabetes, efforts have been made to generate potent, brain-penetrant PRCP inhibitors. Here, we discuss the role of PRCP on energy metabolism and the development of PRCP inhibitors.
Collapse
Affiliation(s)
- Jin Kwon Jeong
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, CT 06520, USA
| | | |
Collapse
|
44
|
Park YG, Song J, Kim Y. Optimized purification and characterization of expressed hMC4R-TM2. JOURNAL OF THE KOREAN MAGNETIC RESONANCE SOCIETY 2012. [DOI: 10.6564/jkmrs.2012.16.2.147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
45
|
Yang F, Tao YX. Functional characterization of nine novel naturally occurring human melanocortin-3 receptor mutations. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1752-61. [PMID: 22884546 DOI: 10.1016/j.bbadis.2012.07.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Revised: 07/12/2012] [Accepted: 07/27/2012] [Indexed: 12/20/2022]
Abstract
The melanocortin-3 receptor (MC3R) is a member of family A rhodopsin-like G protein-coupled receptors. Mouse genetic studies suggested that MC3R and the related MC4R are non-redundant regulators of energy homeostasis. Lack of Mc3r leads to higher feed efficiency and fat mass. However, until now only a few MC3R mutations have been identified in humans and the role of MC3R in the pathogenesis of obesity was unclear. In the present study, we performed detailed functional studies on nine naturally occurring MC3R mutations recently reported. We found that all nine mutants had decreased cell surface expression. A260V, M275T, and L297V had decreased total expression whereas the other six mutants had normal total expression. Mutants S69C and T280S exhibited significant defects in ligand binding and signaling. The dramatic defects of T280S might be partially caused by decreased cell surface expression. In addition, we found mutants M134I and M275T had decreased maximal binding but displayed similar signaling properties as wild-type MC3R. All the other mutants had normal binding and signaling activities. Co-expression studies showed that all mutants except L297V did not affect wild-type MC3R signaling. Multiple mutations at T280 demonstrated the necessity of Thr for cell surface expression, ligand binding, and signaling. In summary, we provided detailed data of these novel human MC3R mutations leading to a better understanding of structure-function relationship of MC3R and the role of MC3R mutation in obesity.
Collapse
Affiliation(s)
- Fan Yang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | | |
Collapse
|
46
|
Electrophysiological analysis of circuits controlling energy homeostasis. Mol Neurobiol 2012; 45:258-78. [PMID: 22331510 DOI: 10.1007/s12035-012-8241-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 01/26/2012] [Indexed: 10/28/2022]
Abstract
Since the discovery of leptin and the central melanocortin circuit, electrophysiological studies have played a major role in elucidating mechanisms underlying energy homeostasis. This review highlights the contribution of findings made by electrophysiological measurements to the current understanding of hypothalamic neuronal networks involved in energy homeostasis with a specific focus on the arcuate-paraventricular nucleus circuit.
Collapse
|
47
|
Rediger A, Piechowski CL, Habegger K, Grüters A, Krude H, Tschöp MH, Kleinau G, Biebermann H. MC4R dimerization in the paraventricular nucleus and GHSR/MC3R heterodimerization in the arcuate nucleus: is there relevance for body weight regulation? Neuroendocrinology 2012; 95:277-88. [PMID: 22327910 DOI: 10.1159/000334903] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 11/06/2011] [Indexed: 01/21/2023]
Abstract
The worldwide obesity epidemic is increasing, yet at this time, no long-acting and specific pharmaceutical therapies are available. Peripheral hormonal signals communicate metabolic status to the hypothalamus by activating their corresponding receptors in the arcuate nucleus (ARC). In this brain region, a variety of G protein-coupled receptors (GPCRs) are expressed that are potentially involved in weight regulation, but so far, the detailed function of most hypothalamic GPCRs is only partially understood. An important and underappreciated feature of GPCRs is the capacity for regulation via di- and heterodimerization. Increasing evidence implicates that heterodimerization of GPCRs results in profound functional consequences. Recently, we could demonstrate that interaction of the melanocortin 3 receptor (MC3R) and the growth hormone secretagogue receptor (GHSR)-1a results in a modulation of function in both receptors. Although the physiological role of GPCR-GPCR interaction in the hypothalamus is yet to be elucidated, this concept promises new avenues for investigation and understanding of hypothalamic functions dependent on GPCR signaling. Since GPCRs are important targets for drugs to combat many diseases, identification of heterodimers may be a prerequisite for highly specific drugs. Therefore, a detailed understanding of the mechanisms and their involvement in weight regulation is necessary. Fundamental to this understanding is the interplay of GPCR-GPCR in the hypothalamic nuclei in energy metabolism. In this review, we summarize the current knowledge on melanocortin receptors and GHSR-1a in hypothalamic weight regulation, especially as they pertain to possible drug targets. Furthermore, we include available evidence for the participation and significance of GPCR dimerization.
Collapse
MESH Headings
- Animals
- Appetite Regulation/physiology
- Arcuate Nucleus of Hypothalamus/anatomy & histology
- Arcuate Nucleus of Hypothalamus/metabolism
- Arcuate Nucleus of Hypothalamus/physiology
- Body Weight/physiology
- Humans
- Models, Biological
- Paraventricular Hypothalamic Nucleus/anatomy & histology
- Paraventricular Hypothalamic Nucleus/metabolism
- Paraventricular Hypothalamic Nucleus/physiology
- Protein Multimerization/physiology
- Receptor, Melanocortin, Type 3/metabolism
- Receptor, Melanocortin, Type 3/physiology
- Receptor, Melanocortin, Type 4/metabolism
- Receptor, Melanocortin, Type 4/physiology
- Receptors, Ghrelin/metabolism
- Receptors, Ghrelin/physiology
Collapse
Affiliation(s)
- Anne Rediger
- Institute of Experimental Pediatric Endocrinology, Charité Universitätsmedizin Berlin, Humboldt University, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Begriche K, Levasseur PR, Zhang J, Rossi J, Skorupa D, Solt LA, Young B, Burris TP, Marks DL, Mynatt RL, Butler AA. Genetic dissection of the functions of the melanocortin-3 receptor, a seven-transmembrane G-protein-coupled receptor, suggests roles for central and peripheral receptors in energy homeostasis. J Biol Chem 2011; 286:40771-81. [PMID: 21984834 PMCID: PMC3220494 DOI: 10.1074/jbc.m111.278374] [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: 07/01/2011] [Revised: 09/05/2011] [Indexed: 01/29/2023] Open
Abstract
The melanocortin-3 receptor (MC3R) gene is pleiotropic, influencing body composition, natriuresis, immune function, and entrainment of circadian rhythms to nutrient intake. MC3Rs are expressed in hypothalamic and limbic regions of the brain and in peripheral tissues. To investigate the roles of central MC3Rs, we inserted a "lox-stop-lox" (LoxTB) 5' of the translation initiation codon of the mouse Mc3r gene and reactivated transcription using neuron-specific Cre transgenic mice. As predicted based on earlier observations of Mc3r knock-out mice, Mc3r(TB/TB) mice displayed reduced lean mass, increased fat mass, and accelerated diet-induced obesity. Surprisingly, rescuing Mc3r expression in the nervous system using the Nestin-Cre transgene only partially rescued obesity in chow-fed conditions and had no impact on the accelerated diet-induced obesity phenotype. The ventromedial hypothalamus (VMH), a critical node in the neural networks regulating feeding-related behaviors and metabolic homeostasis, exhibits dense Mc3r expression relative to other brain regions. To target VMH MC3R expression, we used the steroidogenic factor-1 Cre transgenic mouse. Although restoring VMH MC3R signaling also had a modest impact on obesity, marked improvements in metabolic homeostasis were observed. VMH MC3R signaling was not sufficient to rescue the lean mass phenotype or the regulation of behaviors anticipating food anticipation. These results suggest that actions of MC3Rs impacting on energy homeostasis involve both central and peripheral sites of action. The impact of central MC3Rs on behavior and metabolism involves divergent pathways; VMH MC3R signaling improves metabolic homeostasis but does not significantly impact on the expression of behaviors anticipating nutrient availability.
Collapse
Affiliation(s)
| | - Peter R. Levasseur
- the Department of Pediatrics, Oregon Health and Science University, Portland, Oregon 97239, and
| | - Jingying Zhang
- the Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808
| | - Jari Rossi
- From the Departments of Metabolism and Aging and
| | | | | | - Brandon Young
- the Genomics Core, The Scripps Research Institute, Jupiter, Florida 33458
| | | | - Daniel L. Marks
- the Department of Pediatrics, Oregon Health and Science University, Portland, Oregon 97239, and
| | - Randall L. Mynatt
- the Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808
| | | |
Collapse
|
49
|
Rediger A, Piechowski CL, Yi CX, Tarnow P, Strotmann R, Grüters A, Krude H, Schöneberg T, Tschöp MH, Kleinau G, Biebermann H. Mutually opposite signal modulation by hypothalamic heterodimerization of ghrelin and melanocortin-3 receptors. J Biol Chem 2011; 286:39623-31. [PMID: 21940628 PMCID: PMC3234785 DOI: 10.1074/jbc.m111.287607] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 09/03/2011] [Indexed: 12/22/2022] Open
Abstract
Interaction and cross-talk of G-protein-coupled receptors (GPCRs) are of considerable interest because an increasing number of examples implicate a profound functional and physiological relevance of homo- or hetero-oligomeric GPCRs. The ghrelin (growth hormone secretagogue receptor (GHSR)) and melanocortin-3 (MC3R) receptors are both known to have orexigenic effects on the hypothalamic control of body weight. Because in vitro studies indicate heterodimerization of GHSR and MC3R, we investigated their functional interplay. Combined in situ hybridization and immunohistochemistry indicated that the vast majority of GHSR-expressing neurons in the arcuate nucleus also express MC3R. In vitro coexpression of MC3R and GHSR promoted enhanced melanocortin-induced intracellular cAMP accumulation compared with activation of MC3R in the absence of GHSR. In contrast, agonist-independent basal signaling activity and ghrelin-induced signaling of GHSR were impaired, most likely due to interaction with MC3R. By taking advantage of naturally occurring GHSR mutations and an inverse agonist for GHSR, we demonstrate that the observed enhanced MC3R signaling capability depends directly on the basal activity of GHSR. In conclusion, we demonstrate a paradigm-shifting example of GPCR heterodimerization allowing for mutually opposite functional influence of two hypothalamic receptors controlling body weight. We found that the agonist-independent active conformation of one GPCR can determine the signaling modalities of another receptor in a heterodimer. Our discovery also implies that mutations within one of two interacting receptors might affect both receptors and different pathways simultaneously. These findings uncover mechanisms of important relevance for pharmacological targeting of GPCR in general and hypothalamic body weight regulation in particular.
Collapse
MESH Headings
- Animals
- Arcuate Nucleus of Hypothalamus/metabolism
- COS Cells
- Chlorocebus aethiops
- Cyclic AMP/genetics
- Cyclic AMP/metabolism
- Gene Expression Regulation/physiology
- Ghrelin/genetics
- Ghrelin/metabolism
- HEK293 Cells
- Humans
- Mice
- Mice, Knockout
- Mutation
- Nerve Tissue Proteins/agonists
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Neurons/metabolism
- Protein Multimerization/physiology
- Receptor, Melanocortin, Type 3/agonists
- Receptor, Melanocortin, Type 3/genetics
- Receptor, Melanocortin, Type 3/metabolism
- Receptors, Ghrelin/agonists
- Receptors, Ghrelin/genetics
- Receptors, Ghrelin/metabolism
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Anne Rediger
- From the Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Carolin L. Piechowski
- From the Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Chun-Xia Yi
- the Metabolic Diseases Institute, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio 45267, and
| | - Patrick Tarnow
- From the Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Rainer Strotmann
- the Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| | - Annette Grüters
- From the Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Heiko Krude
- From the Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Torsten Schöneberg
- the Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| | - Matthias H. Tschöp
- the Metabolic Diseases Institute, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio 45267, and
| | - Gunnar Kleinau
- From the Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Heike Biebermann
- From the Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| |
Collapse
|
50
|
De Jonghe BC, Hayes MR, Bence KK. Melanocortin control of energy balance: evidence from rodent models. Cell Mol Life Sci 2011; 68:2569-88. [PMID: 21553232 PMCID: PMC3135719 DOI: 10.1007/s00018-011-0707-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 04/19/2011] [Accepted: 04/20/2011] [Indexed: 01/18/2023]
Abstract
Regulation of energy balance is extremely complex, and involves multiple systems of hormones, neurotransmitters, receptors, and intracellular signals. As data have accumulated over the last two decades, the CNS melanocortin system is now identified as a prominent integrative network of energy balance controls in the mammalian brain. Here, we will review findings from rat and mouse models, which have provided an important framework in which to study melanocortin function. Perhaps most importantly, this review attempts for the first time to summarize recent advances in our understanding of the intracellular signaling pathways thought to mediate the action of melanocortin neurons and peptides in control of longterm energy balance. Special attention will be paid to the roles of MC4R/MC3R, as well as downstream neurotransmitters within forebrain and hindbrain structures that illustrate the distributed control of melanocortin signaling in energy balance. In addition, distinctions and controversy between rodent species will be discussed.
Collapse
Affiliation(s)
- Bart C. De Jonghe
- Dept. of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| | - Matthew R. Hayes
- Dept. of Psychiatry, School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| | - Kendra K. Bence
- Dept. of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| |
Collapse
|