1
|
Dai HC, Ji RL, Tao YX. SHU9119 and MBP10 are biased ligands at the human melanocortin-4 receptor. Biochem Pharmacol 2024; 228:116325. [PMID: 38815629 DOI: 10.1016/j.bcp.2024.116325] [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: 01/22/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
The melanocortin-4 receptor (MC4R), a G protein-coupled receptor, is critically involved in regulating energy homeostasis as well as modulation of reproduction and sexual function. Two peptide antagonists (SHU9119 and MBP10) were derived from the endogenous agonist α-melanocyte stimulating hormone. But their pharmacology at human MC4R is not fully understood. Herein, we performed detailed pharmacological studies of SHU9119 and MBP10 on wild-type (WT) and six naturally occurring constitutively active MC4Rs. Both ligands had no or negligible agonist activity in Gαs-cAMP signaling on WT MC4R, but stimulated extracellular signal-regulated kinases 1 and 2 (ERK1/2) activation on WT and mutant MC4Rs. Mechanistic studies revealed that SHU9119 and MBP10 stimulated ERK1/2 signaling of MC4R by different mechanisms, with SHU9119-stimulated ERK1/2 signaling mediated by phosphatidylinositol 3-kinase (PI3K) and MBP10-initiated ERK1/2 activation through PI3K and β-arrestin. In summary, our studies demonstrated that SHU9119 and MBP10 were biased ligands for MC4R, preferentially activating ERK1/2 signaling through different mechanisms. SHU9119 acted as a biased ligand and MBP10 behaved as a biased allosteric modulator.
Collapse
Affiliation(s)
- Han-Chuan Dai
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| | - Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
2
|
Rodríguez Rondón AV, Welling MS, van den Akker ELT, van Rossum EFC, Boon EMJ, van Haelst MM, Delhanty PJD, Visser JA. MC4R Variants Modulate α-MSH and Setmelanotide Induced Cellular Signaling at Multiple Levels. J Clin Endocrinol Metab 2024; 109:2452-2466. [PMID: 38567654 PMCID: PMC11403317 DOI: 10.1210/clinem/dgae210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/10/2024] [Indexed: 04/04/2024]
Abstract
CONTEXT The melanocortin-4 receptor (MC4R) plays an important role in body weight regulation. Pathogenic MC4R variants are the most common cause of monogenic obesity. OBJECTIVE We have identified 17 MC4R variants in adult and pediatric patients with obesity. Here we aimed to functionally characterize these variants by analyzing 4 different aspects of MC4R signaling. In addition, we aimed to analyze the effect of setmelanotide, a potent MC4R agonist, on these MC4R variants. MATERIALS AND METHODS Cell surface expression and α-melanocyte stimulating hormone (α-MSH)- or setmelanotide-induced cAMP response, β-arrestin-2 recruitment, and ERK activation were measured in cells expressing either wild type or variant MC4R. RESULTS We found a large heterogeneity in the function of these variants. We identified variants with a loss of response for all studied MC4R signaling, variants with no cAMP accumulation or ERK activation but normal β-arrestin-2 recruitment, and variants with normal cAMP accumulation and ERK activation but decreased β-arrestin-2 recruitment, indicating disrupted desensitization and signaling mechanisms. Setmelanotide displayed a greater potency and similar efficacy as α-MSH and induced significantly increased maximal cAMP responses of several variants compared to α-MSH. Despite the heterogeneity in functional response, there was no apparent difference in the obesity phenotype in our patients. CONCLUSION We show that these obesity-associated MC4R variants affect MC4R signaling differently yet lead to a comparable clinical phenotype. Our results demonstrate the clinical importance of assessing the effect of MC4R variants on a range of molecular signaling mechanisms to determine their association with obesity, which may aid in improving personalized treatment.
Collapse
Affiliation(s)
- Alejandra V Rodríguez Rondón
- Obesity Center CGG and Expertise Center Genetic Obesity, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Mila S Welling
- Obesity Center CGG and Expertise Center Genetic Obesity, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Pediatrics, Division of Endocrinology, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Erica L T van den Akker
- Obesity Center CGG and Expertise Center Genetic Obesity, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Pediatrics, Division of Endocrinology, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Elisabeth F C van Rossum
- Obesity Center CGG and Expertise Center Genetic Obesity, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Elles M J Boon
- Department of Human Genetics, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Mieke M van Haelst
- Department of Human Genetics, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Patric J D Delhanty
- Obesity Center CGG and Expertise Center Genetic Obesity, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Jenny A Visser
- Obesity Center CGG and Expertise Center Genetic Obesity, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
3
|
Ji RL, Jiang SS, Kleinau G, Scheerer P, Tao YX. Are Melanocortin Receptors Present in Extant Protochordates? Biomolecules 2024; 14:1120. [PMID: 39334886 PMCID: PMC11430673 DOI: 10.3390/biom14091120] [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: 08/03/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Functional melanocortin receptor (MCR) genes have been identified in the genomes of early chordates, e.g., the cyclostomata. Whether they appear in the most ancient chordates such as cephalochordate and urochordata, however, remains unclear due to missing genetic data. Herein, we studied five putative (from NCBI database), sequence-based predicted MCR-like receptors from urochordata and cephalochordate, including Styela clava, Ciona intestinalis, Branchiostoma floridae, and Branchiostoma belcheri. The BLAST and phylogenetic analyses suggested a relationship between these specific receptors and vertebrate MCRs. However, several essential residues for MCR functions in vertebrates were missing in these putative chordata MCRs. To test receptor functionality, several experimental studies were conducted. Binding assays and functional analyses showed no specific binding and no ligand-induced cAMP or ERK1/2 signaling (with either endogenous α-MSH or synthetic ligands for MC4R), despite successfully expressing four receptors in HEK 293T cells. These four receptors showed high basal cAMP signaling, likely mediated by ligand-independent Gs coupling. In summary, our results suggest that the five predicted MCR-like receptors are, indeed, class A G protein-coupled receptors (GPCRs), which in four cases show high constitutive activity in the Gs-cAMP signaling pathway but are not MCR-like receptors in terms of ligand recognition of known MCR ligands. These receptors might be ancient G protein-coupled receptors with so far unidentified ligands.
Collapse
Affiliation(s)
- Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Shan-Shan Jiang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Gunnar Kleinau
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, D-10117 Berlin, Germany
| | - Patrick Scheerer
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, D-10117 Berlin, Germany
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
4
|
Zhao Z, Yang Y, Liu P, Yan T, Li R, Pan C, Li Y, Lan X. A Critical Functional Missense Mutation (T117M) in Sheep MC4R Gene Significantly Leads to Gain-of-Function. Animals (Basel) 2024; 14:2207. [PMID: 39123733 PMCID: PMC11311007 DOI: 10.3390/ani14152207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
The melanocortin 4 receptor (MC4R) gene plays a central role in regulating energy homeostasis and food intake in livestock, thereby affecting their economic worth and growth. In a previous study, the p.T117M mutation in the sheep MC4R gene, which leads to the transition of threonine to methionine, was found to affect the body weight at six months and the average daily gain in Hu sheep. However, there are still limited studies on the frequency of the sheep p.T117M missense mutation globally, and the underlying cellular mechanism remains elusive. Therefore, this study first used WGS to investigate the distribution of the MC4R gene p.T117M mutation in 652 individuals across 22 breeds worldwide. The results showed that the mutation frequency was higher in European breeds compared with Chinese sheep breeds, particularly in Poll Dorset sheep (mutation frequency > 0.5). The p.T117M mutation occurs in the first extracellular loop of MC4R. Mechanistically, the basal activity of the mutated receptor is significantly increased. Specifically, upon treatment with α-MSH and ACTH ligands, the cAMP and MAPK/ERK signaling activation of M117 MC4R is enhanced. These results indicate that the T117M mutation may change the function of the gene by increasing the constitutive activity and signaling activation of cAMP and MAPK/ERK, and, thus, may regulate the growth traits of sheep. In conclusion, this study delved into the global distribution and underlying cellular mechanisms of the T117M mutation of the MC4R gene, establishing a scientific foundation for breeding sheep with superior growth, thereby contributing to the advancement of the sheep industry.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang 712100, China; (Z.Z.); (Y.Y.); (P.L.); (T.Y.); (R.L.); (C.P.)
| | - Xianyong Lan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang 712100, China; (Z.Z.); (Y.Y.); (P.L.); (T.Y.); (R.L.); (C.P.)
| |
Collapse
|
5
|
Wang S, Kahale F, Naderi A, Surico PL, Yin J, Dohlman T, Chen Y, Dana R. Therapeutic Effects of Stimulating the Melanocortin Pathway in Regulating Ocular Inflammation and Cell Death. Biomolecules 2024; 14:169. [PMID: 38397406 PMCID: PMC10886905 DOI: 10.3390/biom14020169] [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: 01/05/2024] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Alpha-melanocyte-stimulating hormone (α-MSH) and its binding receptors (the melanocortin receptors) play important roles in maintaining ocular tissue integrity and immune homeostasis. Particularly extensive studies have demonstrated the biological functions of α-MSH in both immunoregulation and cyto-protection. This review summarizes the current knowledge of both the physiological and pathological roles of α-MSH and its receptors in the eye. We focus on recent developments in the biology of α-MSH and the relevant clinical implications in treating ocular diseases.
Collapse
Affiliation(s)
- Shudan Wang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; (S.W.); (F.K.); (A.N.); (P.L.S.); (J.Y.); (T.D.)
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Francesca Kahale
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; (S.W.); (F.K.); (A.N.); (P.L.S.); (J.Y.); (T.D.)
| | - Amirreza Naderi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; (S.W.); (F.K.); (A.N.); (P.L.S.); (J.Y.); (T.D.)
| | - Pier Luigi Surico
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; (S.W.); (F.K.); (A.N.); (P.L.S.); (J.Y.); (T.D.)
| | - Jia Yin
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; (S.W.); (F.K.); (A.N.); (P.L.S.); (J.Y.); (T.D.)
| | - Thomas Dohlman
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; (S.W.); (F.K.); (A.N.); (P.L.S.); (J.Y.); (T.D.)
| | - Yihe Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; (S.W.); (F.K.); (A.N.); (P.L.S.); (J.Y.); (T.D.)
| | - Reza Dana
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; (S.W.); (F.K.); (A.N.); (P.L.S.); (J.Y.); (T.D.)
| |
Collapse
|
6
|
Orlandi P, Banchi M, Vaglini F, Carli M, Aringhieri S, Bandini A, Pardini C, Viaggi C, Lai M, Alì G, Ottani A, Vandini E, Guidi P, Bernardeschi M, La Rocca V, Francia G, Fontanini G, Pistello M, Frenzilli G, Giuliani D, Scarselli M, Bocci G. Melanocortin receptor 4 as a new target in melanoma therapy: Anticancer activity of the inhibitor ML00253764 alone and in association with B-raf inhibitor vemurafenib. Biochem Pharmacol 2024; 219:115952. [PMID: 38036189 DOI: 10.1016/j.bcp.2023.115952] [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: 09/09/2023] [Accepted: 11/27/2023] [Indexed: 12/02/2023]
Abstract
The aim of our study is to investigate in vitro and in vivo MC4R as a novel target in melanoma using the selective antagonist ML00253764 (ML) alone and in combination with vemurafenib, a B-rafV600E inhibitor. The human melanoma B-raf mutated A-2058 and WM 266-4 cell lines were used. An MC4R null A-2058 cell line was generated using a CRISPR/Cas9 system. MC4R protein expression was analysed by western blotting, immunohistochemistry, and immunofluorescence. Proliferation and apoptotic assays were performed with ML00253764, whereas the synergism with vemurafenib was evaluated by the combination index (CI) and Loewe methods. ERK1/2 phosphorylation and BCL-XL expression were quantified by western blot. In vivo experiments were performed in Athymic Nude-Foxn1nu male mice, injecting subcutaneously melanoma cells, and treating animals with ML, vemurafenib and their concomitant combination. Comet and cytome assays were performed. Our results show that human melanoma cell lines A-2058 and WM 266-4, and melanoma human tissue, express functional MC4R receptors on their surface. MC4R receptors on melanoma cells can be inhibited by the selective antagonist ML, causing antiproliferative and proapoptotic activity through the inhibition of phosphorylation of ERK1/2 and a reduction of BCL-XL. The concomitant combination of vemurafenib and ML caused a synergistic effect on melanoma cells in vitro and inhibited in vivo tumor growth in a preclinical model, without causing mouse weight loss or genotoxicity. Our original research contributes to the landscape of pharmacological treatments for melanoma, providing MC4R antagonists as drugs that can be added to established therapies.
Collapse
Affiliation(s)
- Paola Orlandi
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | - Marta Banchi
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | - Francesca Vaglini
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Pisa, Italy
| | - Marco Carli
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Pisa, Italy
| | - Stefano Aringhieri
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Pisa, Italy
| | - Arianna Bandini
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | - Carla Pardini
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Pisa, Italy
| | - Cristina Viaggi
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Pisa, Italy
| | - Michele Lai
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Pisa, Italy
| | - Greta Alì
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell'Area Critica, Università di Pisa, Pisa, Italy
| | - Alessandra Ottani
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze, Sezione di Farmacologia e Medicina Molecolare, Università di Modena e Reggio Emilia, Modena, Italy
| | - Eleonora Vandini
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze, Sezione di Farmacologia e Medicina Molecolare, Università di Modena e Reggio Emilia, Modena, Italy
| | - Patrizia Guidi
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | | | - Veronica La Rocca
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Pisa, Italy; Scuola Superiore Sant'Anna, Pisa, Italy
| | - Giulio Francia
- Border Biomedical Research Center, University of Texas at El Paso (UTEP), El Paso, TX, USA
| | - Gabriella Fontanini
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell'Area Critica, Università di Pisa, Pisa, Italy
| | - Mauro Pistello
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Pisa, Italy
| | - Giada Frenzilli
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | - Daniela Giuliani
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze, Sezione di Farmacologia e Medicina Molecolare, Università di Modena e Reggio Emilia, Modena, Italy
| | - Marco Scarselli
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Pisa, Italy
| | - Guido Bocci
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy.
| |
Collapse
|
7
|
Wei R, Li D, Jia S, Chen Y, Wang J. MC4R in Central and Peripheral Systems. Adv Biol (Weinh) 2023; 7:e2300035. [PMID: 37043700 DOI: 10.1002/adbi.202300035] [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: 01/21/2023] [Revised: 02/25/2023] [Indexed: 04/14/2023]
Abstract
Obesity has emerged as a critical and urgent health burden during the current global pandemic. Among multiple genetic causes, melanocortin receptor-4 (MC4R), involved in food intake and energy metabolism regulation through various signaling pathways, has been reported to be the lead genetic factor in severe and early onset obesity and hyperphagia disorders. Most previous studies have illustrated the roles of MC4R signaling in energy intake versus expenditure in the central system, while some evidence indicates that MC4R is also expressed in peripheral systems, such as the gut and endocrine organs. However, its physiopathological function remains poorly defined. This review aims to depict the central and peripheral roles of MC4R in energy metabolism and endocrine hormone homeostasis, the diversity of phenotypes, biased downstream signaling caused by distinct MC4R mutations, and current drug development targeting the receptor.
Collapse
Affiliation(s)
- Ran Wei
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Danjie Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Sheng Jia
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| |
Collapse
|
8
|
Ji RL, Liu T, Hou ZS, Wen HS, Tao YX. Divergent Pharmacology and Biased Signaling of the Four Melanocortin-4 Receptor Isoforms in Rainbow Trout ( Oncorhynchus mykiss). Biomolecules 2023; 13:1248. [PMID: 37627313 PMCID: PMC10452266 DOI: 10.3390/biom13081248] [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: 07/03/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
The melanocortin-4 receptor (MC4R) is essential for the modulation of energy balance and reproduction in both fish and mammals. Rainbow trout (Oncorhynchus mykiss) has been extensively studied in various fields and provides a unique opportunity to investigate divergent physiological roles of paralogues. Herein we identified four trout mc4r (mc4ra1, mc4ra2, mc4rb1, and mc4rb2) genes. Four trout Mc4rs (omMc4rs) were homologous to those of teleost and mammalian MC4Rs. Multiple sequence alignments, a phylogenetic tree, chromosomal synteny analyses, and pharmacological studies showed that trout mc4r genes may have undergone different evolutionary processes. All four trout Mc4rs bound to two peptide agonists and elevated intracellular cAMP levels dose-dependently. High basal cAMP levels were observed at two omMc4rs, which were decreased by Agouti-related peptide. Only omMc4rb2 was constitutively active in the ERK1/2 signaling pathway. Ipsen 5i, ML00253764, and MCL0020 were biased allosteric modulators of omMc4rb1 with selective activation upon ERK1/2 signaling. ML00253764 behaved as an allosteric agonist in Gs-cAMP signaling of omMc4rb2. This study will lay the foundation for future physiological studies of various mc4r paralogs and reveal the evolution of MC4R in vertebrates.
Collapse
Affiliation(s)
- Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA; (R.-L.J.); (T.L.)
| | - Ting Liu
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA; (R.-L.J.); (T.L.)
| | - Zhi-Shuai Hou
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA; (R.-L.J.); (T.L.)
| | - Hai-Shen Wen
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266100, China;
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA; (R.-L.J.); (T.L.)
| |
Collapse
|
9
|
Wu CLS, Cioanca AV, Gelmi MC, Wen L, Di Girolamo N, Zhu L, Natoli R, Conway RM, Petsoglou C, Jager MJ, McCluskey PJ, Madigan MC. The multifunctional human ocular melanocortin system. Prog Retin Eye Res 2023; 95:101187. [PMID: 37217094 DOI: 10.1016/j.preteyeres.2023.101187] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/12/2023] [Accepted: 05/12/2023] [Indexed: 05/24/2023]
Abstract
Immune privilege in the eye involves physical barriers, immune regulation and secreted proteins that together limit the damaging effects of intraocular immune responses and inflammation. The neuropeptide alpha-melanocyte stimulating hormone (α-MSH) normally circulates in the aqueous humour of the anterior chamber and the vitreous fluid, secreted by iris and ciliary epithelium, and retinal pigment epithelium (RPE). α-MSH plays an important role in maintaining ocular immune privilege by helping the development of suppressor immune cells and by activating regulatory T-cells. α-MSH functions by binding to and activating melanocortin receptors (MC1R to MC5R) and receptor accessory proteins (MRAPs) that work in concert with antagonists, otherwise known as the melanocortin system. As well as controlling immune responses and inflammation, a broad range of biological functions is increasingly recognised to be orchestrated by the melanocortin system within ocular tissues. This includes maintaining corneal transparency and immune privilege by limiting corneal (lymph)angiogenesis, sustaining corneal epithelial integrity, protecting corneal endothelium and potentially enhancing corneal graft survival, regulating aqueous tear secretion with implications for dry eye disease, facilitating retinal homeostasis via maintaining blood-retinal barriers, providing neuroprotection in the retina, and controlling abnormal new vessel growth in the choroid and retina. The role of melanocortin signalling in uveal melanocyte melanogenesis however remains unclear compared to its established role in skin melanogenesis. The early application of a melanocortin agonist to downregulate systemic inflammation used adrenocorticotropic hormone (ACTH)-based repository cortisone injection (RCI), but adverse side effects including hypertension, edema, and weight gain, related to increased adrenal gland corticosteroid production, impacted clinical uptake. Compared to ACTH, melanocortin peptides that target MC1R, MC3R, MC4R and/or MC5R, but not adrenal gland MC2R, induce minimal corticosteroid production with fewer amdverse systemic effects. Pharmacological advances in synthesising MCR-specific targeted peptides provide further opportunities for treating ocular (and systemic) inflammatory diseases. Following from these observations and a renewed clinical and pharmacological interest in the diverse biological roles of the melanocortin system, this review highlights the physiological and disease-related involvement of this system within human eye tissues. We also review the emerging benefits and versatility of melanocortin receptor targeted peptides as non-steroidal alternatives for inflammatory eye diseases such as non-infectious uveitis and dry eye disease, and translational applications in promoting ocular homeostasis, for example, in corneal transplantation and diabetic retinopathy.
Collapse
Affiliation(s)
- Chieh-Lin Stanley Wu
- School of Optometry and Vision Science, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia; Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Optometry, Asia University, Taichung, Taiwan
| | - Adrian V Cioanca
- Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; John Curtin School of Medical Research, The Australian National University, ACT, Australia; ANU Medical School, The Australian National University, ACT, Australia
| | - Maria C Gelmi
- Department of Ophthalmology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Li Wen
- New South Wales Organ and Tissue Donation Service, Sydney Hospital and Sydney Eye Hospital, NSW, 2000, Australia
| | - Nick Di Girolamo
- School of Biomedical Sciences, Mechanisms of Disease and Translational Research, University of New South Wales, Sydney, Australia
| | - Ling Zhu
- Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Riccardo Natoli
- Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; John Curtin School of Medical Research, The Australian National University, ACT, Australia; ANU Medical School, The Australian National University, ACT, Australia
| | - R Max Conway
- Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Constantinos Petsoglou
- Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; New South Wales Organ and Tissue Donation Service, Sydney Hospital and Sydney Eye Hospital, NSW, 2000, Australia
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Peter J McCluskey
- Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Michele C Madigan
- School of Optometry and Vision Science, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia; Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| |
Collapse
|
10
|
Deng Y, Deng G, Grobe JL, Cui H. Hypothalamic GPCR Signaling Pathways in Cardiometabolic Control. Front Physiol 2021; 12:691226. [PMID: 34262481 PMCID: PMC8274634 DOI: 10.3389/fphys.2021.691226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 05/26/2021] [Indexed: 01/22/2023] Open
Abstract
Obesity is commonly associated with sympathetic overdrive, which is one of the major risk factors for the development of cardiovascular diseases, such as hypertension and heart failure. Over the past few decades, there has been a growing understanding of molecular mechanisms underlying obesity development with central origin; however, the relative contribution of these molecular changes to the regulation of cardiovascular function remains vague. A variety of G-protein coupled receptors (GPCRs) and their downstream signaling pathways activated in distinct hypothalamic neurons by different metabolic hormones, neuropeptides and monoamine neurotransmitters are crucial not only for the regulation of appetite and metabolic homeostasis but also for the sympathetic control of cardiovascular function. In this review, we will highlight the main GPCRs and associated hypothalamic nuclei that are important for both metabolic homeostasis and cardiovascular function. The potential downstream molecular mediators of these GPCRs will also be discussed.
Collapse
Affiliation(s)
- Yue Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Guorui Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, United States
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Huxing Cui
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
- FOE Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, United States
- Obesity Research and Educational Initiative, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| |
Collapse
|
11
|
Pereira SC, Martins AD, Monteiro MP, Pinto S, Barros A, Oliveira PF, Alves MG. Expression of obesity-related genes in human spermatozoa affects the outcomes of reproductive treatments. F&S SCIENCE 2021; 2:164-175. [PMID: 35559751 DOI: 10.1016/j.xfss.2021.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To study the abundance of obesity-related gene (ORG) mRNA in human spermatozoa and its association with sperm quality parameters, embryonic development, and pregnancy rates after assisted reproduction treatment (ART). DESIGN Cross-sectional study of spermatozoa ORG mRNA expression, and sperm and embryonic development parameters of infertile couples attending a single ART center. SETTING University, in collaboration with a medically assisted reproduction center. PATIENT(S) One hundred six couples seeking fertility treatment and receiving ART. INTERVENTION(S) Expression of spermatozoa ORG mRNA was assessed by quantitative reverse transcription-polymerase chain reaction. Sperm and embryonic development parameters were measured by board-certified embryologists. Serum β-human chorionic gonadotropin levels and fetal heartbeat detection on ultrasound were used to document biochemical and clinical pregnancy, respectively. MAIN OUTCOME MEASURE(S) Correlations between the abundance of ORG transcripts in spermatozoa and sperm quality, embryonic development, and achievement of pregnancy. RESULTS The abundance of spermatozoa FTO mRNA was positively correlated with total sperm count (r = 0.5030), fertilization rate (r = 0.4854), embryo cleavage rate (r = 0.5705), and high-quality embryo rate (r = 0.6982). The abundance of spermatozoa MC4R transcript was negatively correlated with sperm viability (r = -0.3111) and positively correlated with biochemical pregnancy (r = 0.4420). The abundance of MC4R and GNPDA2 transcripts was higher in spermatozoa of men with asthenozoospermia and teratozoospermia than in those with normozoospermia. CONCLUSION To our knowledge, this is the first report showing that the abundance of MC4R and FTO transcripts in spermatozoa is associated with sperm and embryo quality parameters, as well as pregnancy rates. Overall, these results further support the view that male factors beyond classic sperm quality parameters, namely the abundance of ORG transcripts, also affect the outcome of ART.
Collapse
Affiliation(s)
- Sara C Pereira
- Clinical and Experimental Endocrinology, Department of Anatomy, Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Ana D Martins
- Clinical and Experimental Endocrinology, Department of Anatomy, Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal; Química Orgânica, Produtos Naturais e Agroalimentares (QOPNA) and Laboratório Associado para a Química Verde (LAQV), Department of Chemistry, University of Aveiro, Portugal
| | - Mariana P Monteiro
- Clinical and Experimental Endocrinology, Department of Anatomy, Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Soraia Pinto
- Center for Reproductive Genetics Professor Alberto Barros, Porto, Portugal
| | - Alberto Barros
- Center for Reproductive Genetics Professor Alberto Barros, Porto, Portugal; Department of Genetics, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
| | - Pedro F Oliveira
- Química Orgânica, Produtos Naturais e Agroalimentares (QOPNA) and Laboratório Associado para a Química Verde (LAQV), Department of Chemistry, University of Aveiro, Portugal
| | - Marco G Alves
- Clinical and Experimental Endocrinology, Department of Anatomy, Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal.
| |
Collapse
|
12
|
Kumar SS, Ward ML, Mountjoy KG. Quantitative high-throughput assay to measure MC4R-induced intracellular calcium. J Mol Endocrinol 2021; 66:285-297. [PMID: 33739935 PMCID: PMC8111326 DOI: 10.1530/jme-20-0285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/19/2021] [Indexed: 12/17/2022]
Abstract
The melanocortin-4 receptor (MC4R), a critical G-protein-coupled receptor (GPCR) regulating energy homeostasis, activates multiple signalling pathways, including mobilisation of intracellular calcium ([Ca2+]i). However, very little is known about the physiological significance of MC4R-induced [Ca2+]i since few studies measure MC4R-induced [Ca2+]i. High-throughput, read-out assays for [Ca2+]i have proven unreliable for overexpressed GPCRs like MC4R, which exhibit low sensitivity mobilising [Ca2+]i. Therefore, we developed, optimised, and validated a robust quantitative high-throughput assay using Fura-2 ratio-metric calcium dye and HEK293 cells stably transfected with MC4R. The quantitation enables direct comparisons between assays and even between different research laboratories. Assay conditions were optimised step-by-step to eliminate interference from stretch-activated receptor increases in [Ca2+]i and to maximise ligand-activated MC4R-induced [Ca2+]i. Calcium imaging was performed using a PheraStar FS multi-well plate reader. Probenecid, included in the buffers to prevent extrusion of Fura-2 dye from cells, was found to interfere with the EGTA-chelation of calcium, required to determine Rmin for quantitation of [Ca2+]i. Therefore, we developed a method to determine Rmin in specific wells without probenecid, which was run in parallel with each assay. The validation of the assay was shown by reproducible α-melanocyte-stimulating hormone (α-MSH) concentration-dependent activation of the stably expressed human MC4R (hMC4R) and mouse MC4R (mMC4R), inducing increases in [Ca2+]i, for three independent experiments. This robust, reproducible, high-throughput assay that quantitatively measures MC4R-induced mobilisation of [Ca2+]i in vitro has potential to advance the development of therapeutic drugs and understanding of MC4R signalling associated with human obesity.
Collapse
Affiliation(s)
- Shree Senthil Kumar
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Marie-Louise Ward
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kathleen Grace Mountjoy
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
- Correspondence should be addressed to K G Mountjoy:
| |
Collapse
|
13
|
Huang YJ, Galen K, Zweifel B, Brooks LR, Wright AD. Distinct binding and signaling activity of Acthar Gel compared to other melanocortin receptor agonists. J Recept Signal Transduct Res 2020; 41:425-433. [PMID: 32938265 DOI: 10.1080/10799893.2020.1818094] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE To compare the binding and agonistic activity of Acthar® Gel and synthetic melanocortin receptor (MCR) agonists and examine how the activity of select agonists affects the in vivo production of corticosterone. MATERIALS AND METHODS In vitro binding was determined using concentration-dependent displacement of the ligand [125I]Nle4, D-Phe7-α-melanocyte-stimulating hormone (α-MSH) on cells expressing MC1R, MC3R, MC4R, or MC5R. Functional activity was determined using a time-resolved fluorescence cyclic adenosine monophosphate (cAMP) assay in cells expressing MC1R, MC2R, MC3R, MC4R, or MC5R. In vivo corticosterone analyses were performed by measuring plasma corticosterone levels in Sprague Dawley rats. RESULTS Acthar Gel and synthetic MCR agonists exhibited the highest binding at MC1R, lowest binding at MC5R, and moderate binding at MC3R and MC4R. Acthar Gel stimulated the production of cAMP in all 5 MCR-expressing cell lines, with MC2R displaying the lowest level of full agonist activity, 3-, 6.6-, and 10-fold lower than MC1R, MC3R, and MC4R, respectively. Acthar Gel was a partial agonist at MC5R. The synthetic MCR agonists induced full activity at all 5 MCRs, with the exception of α-MSH having no activity at MC2R. Acthar Gel treatment had less of an impact on in vivo production of corticosterone compared with synthetic ACTH1-24 depot. CONCLUSIONS Acthar Gel bound to and activated each MCR tested in this study, with partial agonist activity at MC5R and the lowest level of full agonist activity at MC2R, which distinguished it from synthetic MCR agonists. The minimal activity of Acthar Gel at MC2R corresponded to lower endogenous corticosteroid production.
Collapse
Affiliation(s)
- Y Joyce Huang
- Cellular and Molecular Biology, Mallinckrodt Pharmaceuticals, Hazelwood, MO, USA
| | - Karen Galen
- Immunology and Pharmacology, Mallinckrodt Pharmaceuticals, Hazelwood, MO, USA
| | - Ben Zweifel
- Immunology and Pharmacology, Mallinckrodt Pharmaceuticals, Hazelwood, MO, USA
| | - Leah R Brooks
- Medical Affairs, Mallinckrodt Pharmaceuticals, Hazelwood, MO, USA
| | - A Dale Wright
- Immunology and Pharmacology, Mallinckrodt Pharmaceuticals, Hazelwood, MO, USA
| |
Collapse
|
14
|
Yang Z, Liang XF, Li GL, Tao YX. Biased signaling in fish melanocortin-4 receptors (MC4Rs): Divergent pharmacology of four ligands on spotted scat (Scatophagus argus) and grass carp (Ctenopharyngodon idella) MC4Rs. Mol Cell Endocrinol 2020; 515:110929. [PMID: 32615281 DOI: 10.1016/j.mce.2020.110929] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 12/24/2022]
Abstract
The melanocortin-4 receptor (MC4R) plays a critical role in the regulation of energy homeostasis in both mammals and fish. Several fish MC4Rs recently characterized have high constitutive activities, potentially associated with food intake and growth rate. In the present study, we systematically investigated the effects of four human MC4R (hMC4R) antagonists, including agouti-related peptide (AgRP), Ipsen 5i, ML00253764, and MCL0020, on the cAMP and ERK1/2 signaling of two fish MC4Rs: spotted scat (Scatophagus argus) MC4R (saMC4R) and grass carp (Ctenopharyngodon idella) MC4R (ciMC4R), with hMC4R as a control. We showed that both saMC4R and ciMC4R were constitutively active with significantly increased basal cAMP levels. AgRP acted as an inverse agonist in cAMP signaling pathway in both fish MC4Rs whereas MCL0020 functioned as an inverse agonist for ciMC4R but a weak neutral antagonist for saMC4R. Ipsen 5i and MCL0020 behaved as neutral allosteric modulators in the cAMP signaling of fish MC4Rs. The saMC4R and ciMC4R had similar basal pERK1/2 levels as hMC4R and the pERK1/2 levels of the two fish MC4Rs were significantly increased upon stimulation with all four ligands. In summary, our studies demonstrated the existence of biased signaling in fish MC4R. We also showed dramatic pharmacological differences of human and fish MC4Rs with synthetic ligands. Our data provided novel insights and led to a better understanding of fish MC4R pharmacology.
Collapse
Affiliation(s)
- Zhao Yang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, 36849, United States
| | - Xu-Fang Liang
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Hubei Collaborative Innovation Center for Freshwater Aquaculture, Wuhan, Hubei, 430070, China
| | - Guang-Li Li
- Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, 36849, United States.
| |
Collapse
|
15
|
Differential Signaling Profiles of MC4R Mutations with Three Different Ligands. Int J Mol Sci 2020; 21:ijms21041224. [PMID: 32059383 PMCID: PMC7072973 DOI: 10.3390/ijms21041224] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/06/2020] [Accepted: 02/06/2020] [Indexed: 12/21/2022] Open
Abstract
The melanocortin 4 receptor (MC4R) is a key player in hypothalamic weight regulation and energy expenditure as part of the leptin–melanocortin pathway. Mutations in this G protein coupled receptor (GPCR) are the most common cause for monogenetic obesity, which appears to be mediated by changes in the anorectic action of MC4R via GS-dependent cyclic adenosine-monophosphate (cAMP) signaling as well as other signaling pathways. To study potential bias in the effects of MC4R mutations between the different signaling pathways, we investigated three major MC4R mutations: a GS loss-of-function (S127L) and a GS gain-of-function mutant (H158R), as well as the most common European single nucleotide polymorphism (V103I). We tested signaling of all four major G protein families plus extracellular regulated kinase (ERK) phosphorylation and β-arrestin2 recruitment, using the two endogenous agonists, α- and β-melanocyte stimulating hormone (MSH), along with a synthetic peptide agonist (NDP-α-MSH). The S127L mutation led to a full loss-of-function in all investigated pathways, whereas V103I and H158R were clearly biased towards the Gq/11 pathway when challenged with the endogenous ligands. These results show that MC4R mutations can cause vastly different changes in the various MC4R signaling pathways and highlight the importance of a comprehensive characterization of receptor mutations.
Collapse
|
16
|
Miyazaki T, Fukui M, Inagaki E, Miki K, Takabayashi S, Katoh H, Ohira Y, Noguchi M, Tokumoto T. Identification of Two Additional Genomic Loci Responsible for experimentally induced testicular teratoma 2 and 3 (ett2 and ett3). Zoolog Sci 2019; 35:172-178. [PMID: 29623786 DOI: 10.2108/zs170176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Experimental testicular teratomas (ETTs) can be induced in 129/Sv mouse by E12.5 fetal testes transplant into adult testes. Previously, we conducted linkage analysis to explore candidate genes possibly involved in ETT development using F2 intercross fetuses derived from F1[LTXBJ × 129/Sv- + /Ter (+ /+)] hybrids. By linkage analysis on Chr 18 and Chr 19, we identified the genomic locus for experimental testicular teratoma 1 (ett1) on Chr 18. In the present study, we conducted additional mapping and linkage analysis on teratoma susceptibility and genome composition on Chr 1-17. The results revealed two new candidate loci, experimental testicular teratoma 2 (ett2) and experimental testicular teratoma 3 (ett3), on Chr 3 and 7. Interestingly, the rates of ETT generation were increased in the case of ett2 and ett3 regions replaced with LTXBJ strain. To determine whether a polymorphic gene was present, we performed exome analysis of 129/Sv- + /Ter (+ /+) and LTXBJ. This revealed the presence of SNPs in all three loci, ett1 to ett3. ett1 contains polymorphic Mc4r; ett2 contains polymorphic Polr3c, Cd160, and Pdzk1; and ett3 contains polymorphic Prmt3. We found additional loci responsible for ETT formation, namely, ett2 and ett3, and identified candidate genes in these regions by exome analysis.
Collapse
Affiliation(s)
- Takehiro Miyazaki
- 1 Integrated Bioscience Section, Graduate School of Science and Technology, National University Corporation Shizuoka University, Ohya 836, Suruga-ku, Shizuoka 422-8529, Japan
| | - Manami Fukui
- 2 Department of Biological Science, Faculty of Science, National University Corporation Shizuoka University, Ohya 836, Suruga-ku, Shizuoka 422-8529, Japan
| | - Emi Inagaki
- 2 Department of Biological Science, Faculty of Science, National University Corporation Shizuoka University, Ohya 836, Suruga-ku, Shizuoka 422-8529, Japan
| | - Kenji Miki
- 2 Department of Biological Science, Faculty of Science, National University Corporation Shizuoka University, Ohya 836, Suruga-ku, Shizuoka 422-8529, Japan
| | - Shuji Takabayashi
- 3 Institute for Experimental Animals, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Hideki Katoh
- 3 Institute for Experimental Animals, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Yukio Ohira
- 2 Department of Biological Science, Faculty of Science, National University Corporation Shizuoka University, Ohya 836, Suruga-ku, Shizuoka 422-8529, Japan
| | - Motoko Noguchi
- 2 Department of Biological Science, Faculty of Science, National University Corporation Shizuoka University, Ohya 836, Suruga-ku, Shizuoka 422-8529, Japan
| | - Toshinobu Tokumoto
- 1 Integrated Bioscience Section, Graduate School of Science and Technology, National University Corporation Shizuoka University, Ohya 836, Suruga-ku, Shizuoka 422-8529, Japan.,2 Department of Biological Science, Faculty of Science, National University Corporation Shizuoka University, Ohya 836, Suruga-ku, Shizuoka 422-8529, Japan
| |
Collapse
|
17
|
Kühnen P, Krude H, Biebermann H. Melanocortin-4 Receptor Signalling: Importance for Weight Regulation and Obesity Treatment. Trends Mol Med 2019; 25:136-148. [PMID: 30642682 DOI: 10.1016/j.molmed.2018.12.002] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 01/08/2023]
Abstract
The melanocortin-4 receptor (MC4R) - embedded in the leptin-melanocortin pathway - is activated by proopiomelanocortin (POMC)-derived neuropeptides such as α- and β-melanocyte-stimulating hormone (MSH) and plays an important role in hypothalamic body-weight regulation. Accordingly, MC4R is a potential drug target to combat obesity. Previous attempts to develop MC4R agonists failed due to ineffectiveness or severe adverse events. Recently, a new generation of MC4R ligands was developed. Specifically, setmelanotide was found to be effective by inducing biased signalling of the MC4R and thereby reducing feelings of hunger and leading to substantial weight loss in patients with POMC or leptin receptor deficiency. This new potential pharmacological treatment option could be beneficial for further groups of obese patients with defects in the leptin-melanocortin signalling pathway.
Collapse
Affiliation(s)
- Peter Kühnen
- Institute for Experimental Pediatric Endocrinology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Heiko Krude
- Institute for Experimental Pediatric Endocrinology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Heike Biebermann
- Institute for Experimental Pediatric Endocrinology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
18
|
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: 64] [Impact Index Per Article: 12.8] [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
|
19
|
Saba J, Carniglia L, Ramírez D, Turati J, Imsen M, Durand D, Lasaga M, Caruso C. Melanocortin 4 receptor activation protects striatal neurons and glial cells from 3-nitropropionic acid toxicity. Mol Cell Neurosci 2018; 94:41-51. [PMID: 30529228 DOI: 10.1016/j.mcn.2018.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/23/2018] [Accepted: 12/02/2018] [Indexed: 01/18/2023] Open
Abstract
α-Melanocyte stimulating hormone (α-MSH) is a melanocortin which exerts potent anti-inflammatory and anti-apoptotic effects. Melanocortin 4 receptors (MC4R) are abundantly expressed in the brain and we previously demonstrated that [Nle(4), D-Phe(7)]melanocyte-stimulating hormone (NDP-MSH), an α-MSH analogue, increased expression of brain derived-neurotrophic factor (BDNF), and peroxisome proliferator-activated receptor-γ (PPAR-γ). We hypothesized that melanocortins could affect striatal cell survival through BDNF and PPAR-γ. First, we determined the expression of these factors in the striatum. Acute intraperitoneal administration (0.5 mg/kg) of α-MSH increased the levels of BDNF mRNA in rat striatum but not in rat cerebral cortex. Also, protein expression of PPAR-γ and MC4R was increased by acute treatment with α-MSH in striatum but not in cortex. No changes were observed by 48 h treatment. Next, we evaluated melanocortins effect on neuron and glial survival. 3-nitropropionic acid (3-NP), which is known to induce striatal degeneration, was used to induce cell death in the rat striatal cell line ST14A expressing mutant human huntingtin (Q120) or in ST14A cells expressing normal human huntingtin (Q15), in primary cultured astrocytes, and in BV2 cells. NDP-MSH protected Q15 cells, astrocytes and BV2 cells from death by 3-NP whereas it did not fully protect Q120 cells. Protection of Q15 cells and astrocytes was blocked by a MC4R specific inhibitor (JKC-363) and a PPAR-γ antagonist (GW9662). The BDNF receptor antagonist (ANA-12) abolished NDP-MSH protective effect in astrocytes but not in Q15 cells. We demonstrate for the first time that melanocortins, acting through PPAR-γ and BDNF, protect neurons and glial cells from 3-NP toxicity.
Collapse
Affiliation(s)
- Julieta Saba
- Instituto de Investigaciones Biomédicas (INBIOMED) UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Lila Carniglia
- Instituto de Investigaciones Biomédicas (INBIOMED) UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Delia Ramírez
- Instituto de Investigaciones Biomédicas (INBIOMED) UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Turati
- Instituto de Investigaciones Biomédicas (INBIOMED) UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mercedes Imsen
- Instituto de Investigaciones Biomédicas (INBIOMED) UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Daniela Durand
- Instituto de Investigaciones Biomédicas (INBIOMED) UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mercedes Lasaga
- Instituto de Investigaciones Biomédicas (INBIOMED) UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carla Caruso
- Instituto de Investigaciones Biomédicas (INBIOMED) UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
20
|
Corbalán V, Vicenzi N, Moreno Azócar D, Literas S. Chromatic variability and sexual dimorphism in the rocky lizard Phymaturus verdugo. CAN J ZOOL 2018. [DOI: 10.1139/cjz-2018-0107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The genus Phymaturus comprises lizards that inhabit cold environments at high altitudes or latitudes. Phymaturus verdugo Cei and Videla, 2003 is characterized by cephalic melanism, which is interpreted as a character present only in males and associated with sexual dimorphism. Using spectrophotometry and photography, we demonstrate that this species has high chromatic variability and that melanism is also present in females. By comparing colour variables on 15 patches of the body, we find significant differences between sexes in all patches, indicating strong sexual dichromatism for the entire body, not only the cephalic region. Body temperature had no effect on skin reflectance or melanism, which indicates that thermoregulation early in the day is not mediated by colour changes. Contrary to our predictions, we found no evidence of better body condition in darker individuals. Our study breaks some paradigms about the genus Phymaturus and suggests that human perception of colour, as well as working with small samples, can lead to misidentification or erroneous descriptions of species. We believe that future studies should explore heating rates and other functions of melanism such as crypsis, sexual selection, immune response, and UV protection to understand chromatic variability within this genus.
Collapse
Affiliation(s)
- V. Corbalán
- Instituto Argentino de Investigaciones de las Zonas Áridas (IDIZA–CONICET), Centro Científico y Tecnológico (CCT Mendoza–CONICET), Avenida Ruiz Leal s/n, CP 5500, Mendoza, Argentina
| | - N. Vicenzi
- Instituto Argentino de Investigaciones de las Zonas Áridas (IDIZA–CONICET), Centro Científico y Tecnológico (CCT Mendoza–CONICET), Avenida Ruiz Leal s/n, CP 5500, Mendoza, Argentina
| | - D.L. Moreno Azócar
- Laboratorio de Fotobiología, Instituto de Investigaciones en Biodiversidad y Medioambiente (INIBIOMA), CONICET–UNCOMA, Quintral 1250, Bariloche, 8400, Río Negro, Argentina
| | - S. Literas
- Facultad de Ciencias Agrarias, Universidad Nacional de Cuyo, Mendoza. Almirante Brown 500, M5528AHB Chacras de Coria-Luján de Cuyo, Mendoza
| |
Collapse
|
21
|
Benque IJ, Xia P, Shannon R, Ng TF, Taylor AW. The Neuropeptides of Ocular Immune Privilege, α-MSH and NPY, Suppress Phagosome Maturation in Macrophages. Immunohorizons 2018; 2:314-323. [PMID: 30613828 PMCID: PMC6319950 DOI: 10.4049/immunohorizons.1800049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The ocular microenvironment has evolutionarily adapted several mechanisms of immunosuppression to minimize the induction of inflammation. Neuropeptides produced by the retinal pigment epithelial cells regulate macrophage activity. Two neuropeptides, α-melanocyte–stimulating hormone (α -MSH) and neuropeptide Y (NPY), are constitutively expressed by the retinal pigment epithelial cells. Together these two neuropeptides induce anti-inflammatory cytokine production in endotoxin-stimulated macrophages and suppress phagocytosis of unopsonized bioparticles. These neuropeptides do not suppress the phagocytosis of opsonized bioparticles; however, they do suppress phagolysosome activation or formation. In this report, we studied the possibility that α-MSH with NPY suppress phagosome maturation within macrophages using opsonized OVA-coated magnetic beads to isolate and analyze the phagosomes. The magnetic bead–containing intercellular vesicles were isolated and assayed for Rab5, Rab7, LAMP1, Iad, and OVA. The macrophages cotreated with α-MSH and NPY were suppressed in Rab7 recruitment to the phagosome with suppression in LAMP1 expression but not in Iad expression. The results demonstrated that the α-MSH/NPY cotreatment suppressed phagosome maturation. In addition, the a-MSH/NPY–cotreated macrophages were suppressed in their ability to Ag stimulate CD4+ T cell proliferation. These results imply a potential mechanism of ocular immune privilege to divert Ag processing to prevent autoreactive effector T cells from binding their target cognate Ag within the ocular microenvironment.
Collapse
Affiliation(s)
- Isaac J Benque
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA 02118
| | - Pu Xia
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA 02118
| | - Robert Shannon
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA 02118
| | - Tat Fong Ng
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA 02118
| | - Andrew W Taylor
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA 02118
| |
Collapse
|
22
|
Understanding melanocortin-4 receptor control of neuronal circuits: Toward novel therapeutics for obesity syndrome. Pharmacol Res 2018; 129:10-19. [DOI: 10.1016/j.phrs.2018.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/08/2018] [Accepted: 01/08/2018] [Indexed: 01/25/2023]
|
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: 5.7] [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
|
Melanocortins, Melanocortin Receptors and Multiple Sclerosis. Brain Sci 2017; 7:brainsci7080104. [PMID: 28805746 PMCID: PMC5575624 DOI: 10.3390/brainsci7080104] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/07/2017] [Accepted: 08/08/2017] [Indexed: 02/07/2023] Open
Abstract
The melanocortins and their receptors have been extensively investigated for their roles in the hypothalamo-pituitary-adrenal axis, but to a lesser extent in immune cells and in the nervous system outside the hypothalamic axis. This review discusses corticosteroid dependent and independent effects of melanocortins on the peripheral immune system, central nervous system (CNS) effects mediated through neuronal regulation of immune system function, and direct effects on endogenous cells in the CNS. We have focused on the expression and function of melanocortin receptors in oligodendroglia (OL), the myelin producing cells of the CNS, with the goal of identifying new therapeutic approaches to decrease CNS damage in multiple sclerosis as well as to promote repair. It is clear that melanocortin signaling through their receptors in the CNS has potential for neuroprotection and repair in diseases like MS. Effects of melanocortins on the immune system by direct effects on the circulating cells (lymphocytes and monocytes) and by signaling through CNS cells in regions lacking a mature blood brain barrier are clear. However, additional studies are needed to develop highly effective MCR targeted therapies that directly affect endogenous cells of the CNS, particularly OL, their progenitors and neurons.
Collapse
|
25
|
Vaglini F, Pardini C, Di Desidero T, Orlandi P, Pasqualetti F, Ottani A, Pacini S, Giuliani D, Guarini S, Bocci G. Melanocortin Receptor-4 and Glioblastoma Cells: Effects of the Selective Antagonist ML00253764 Alone and in Combination with Temozolomide In Vitro and In Vivo. Mol Neurobiol 2017; 55:4984-4997. [DOI: 10.1007/s12035-017-0702-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/31/2017] [Indexed: 12/13/2022]
|
26
|
Yang LK, Tao YX. Biased signaling at neural melanocortin receptors in regulation of energy homeostasis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2486-2495. [PMID: 28433713 DOI: 10.1016/j.bbadis.2017.04.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/09/2017] [Accepted: 04/10/2017] [Indexed: 12/15/2022]
Abstract
The global prevalence of obesity highlights the importance of understanding on regulation of energy homeostasis. The central melanocortin system is an important intersection connecting the neural pathways controlling satiety and energy expenditure to regulate energy homeostasis by sensing and integrating the signals of external stimuli. In this system, neural melanocortin receptors (MCRs), melanocortin-3 and -4 receptors (MC3R and MC4R), play crucial roles in the regulation of energy homeostasis. Recently, multiple intracellular signaling pathways and biased signaling at neural MCRs have been discovered, providing new insights into neural MCR signaling. This review attempts to summarize biased signaling including biased receptor mutants (both naturally occurring and lab-generated) and biased ligands at neural MCRs, and to provide a better understanding of obesity pathogenesis and new therapeutic implications for obesity treatment.
Collapse
Affiliation(s)
- Li-Kun Yang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
27
|
Jiang DN, Li JT, Tao YX, Chen HP, Deng SP, Zhu CH, Li GL. Effects of melanocortin-4 receptor agonists and antagonists on expression of genes related to reproduction in spotted scat, Scatophagus argus. J Comp Physiol B 2017; 187:603-612. [PMID: 28197776 DOI: 10.1007/s00360-017-1062-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 01/04/2017] [Accepted: 01/20/2017] [Indexed: 11/26/2022]
Abstract
Melanocortin-4 receptor (Mc4r) function related to reproduction in fish has not been extensively investigated. Here, we report on gene expression changes by real-time PCR following treatment with Mc4r agonists and antagonists in the spotted scat (Scatophagus argus). Using in vitro incubated hypothalamus, the Mc4r nonselective agonist NDP-MSH ([Nle4, D-Phe7]-α-melanocyte stimulating hormone; 10-6 M) and selective agonist THIQ (N-[(3R)-1, 2, 3, 4-Tetrahydroisoquinolinium-3-ylcarbonyl]- (1R)-1-(4-chlorobenzyl)-2-[4-cyclohexyl-4-(1H-1,2,4-triazol-1-ylmethyl) piperidin-1-yl]-2-oxoethylamine; 10-7 M) significantly increased the expression of gnrh (Gonadotropin releasing hormone), while the Mc4r nonselective antagonist SHU9119 (Ac-Nle-[Asp-His-DPhe/DNal(2')-Arg-Trp-Lys]-NH2; 10-6 M) and selective antagonist Ipsen 5i (compound 5i synthesized in Ipsen Research Laboratories; 10-6 M) significantly inhibited gnrh expression after 3 h of incubation. In incubated pituitary tissue, NDP-MSH and THIQ significantly increased the expression of fshb (Follicle-stimulating hormone beta subunit) and lhb (Luteinizing hormone beta subunit), while SHU9119 and Ipsen 5i significantly decreased fshb and lhb expression after 3 h of incubation. During the in vivo experiment, THIQ (1 mg/kg bw) significantly increased gnrh expression in hypothalamic tissue, as well as the fshb and lhb expression in pituitary tissue 12 h after abdominal injection. Furthermore, Ipsen 5i (1 mg/kg bw) significantly inhibited gnrh expression in hypothalamic tissue, as well as fshb and lhb gene expression in pituitary tissue 12 h after abdominal injection. In summary, Mc4r singling appears to stimulate gnrh expression in the hypothalamus, thereby modulating the synthesis of Fsh and Lh in the pituitary. In addition, Mc4r also appears to directly regulate fshb and lhb levels in the pituitary in spotted scat. Our study suggests that Mc4r, through the hypothalamus and pituitary, participates in reproductive regulation in fish.
Collapse
Affiliation(s)
- Dong-Neng Jiang
- Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Jian-Tao Li
- Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, 36849, USA
| | - Hua-Pu Chen
- Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Si-Ping Deng
- Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Chun-Hua Zhu
- Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Guang-Li Li
- Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China.
| |
Collapse
|
28
|
Agosti F, Cordisco Gonzalez S, Martinez Damonte V, Tolosa MJ, Di Siervi N, Schioth HB, Davio C, Perello M, Raingo J. Melanocortin 4 receptor constitutive activity inhibits L-type voltage-gated calcium channels in neurons. Neuroscience 2017; 346:102-112. [PMID: 28093215 DOI: 10.1016/j.neuroscience.2017.01.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/16/2016] [Accepted: 01/05/2017] [Indexed: 11/19/2022]
Abstract
The melanocortin 4 receptor (MC4R) is a G protein-coupled receptor (GPCR) that is expressed in several brain nuclei playing a crucial role in the regulation of energy balance controlling the homeostasis of the organism. It displays both agonist-evoked and constitutive activity, and moreover, it can couple to different G proteins. Most of the research on MC4R has been focused on agonist-induced activity, while the molecular and cellular basis of MC4R constitutive activity remains scarcely studied. We have previously shown that neuronal N-type voltage-gated calcium channels (CaV2.2) are inhibited by MC4R agonist-dependent activation, while the CaV subtypes that carry L- and P/Q-type current are not. Here, we tested the hypothesis that MC4R constitutive activity can affect CaV, with focus on the channel subtypes that can control transcriptional activity coupled to depolarization (L-type, CaV1.2/1.3) and neurotransmitter release (N- and P/Q-type, CaV2.2 and CaV2.1). We found that MC4R constitutive activity inhibits specifically CaV1.2/1.3 and CaV2.1 subtypes of CaV. We also explored the signaling pathways mediating this inhibition, and thus propose that agonist-dependent and basal MC4R activation modes signal differentially through Gs and Gi/o pathways to impact on different CaV subtypes. In addition, we found that chronic incubation with MC4R endogenous inverse agonist, agouti and agouti-related peptide (AgRP), occludes CaV inhibition in a cell line and in amygdaloid complex cultured neurons as well. Thus, we define new mechanisms of control of the main mediators of depolarization-induced calcium entry into neurons by a GPCR that displays constitutive activity.
Collapse
Affiliation(s)
- F Agosti
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE) Universidad de La Plata-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comision de Investigaciones de la Provincia de buenos Aires (CIC), La Plata, Buenos Aires, Argentina
| | - S Cordisco Gonzalez
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE) Universidad de La Plata-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comision de Investigaciones de la Provincia de buenos Aires (CIC), La Plata, Buenos Aires, Argentina
| | - V Martinez Damonte
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE) Universidad de La Plata-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comision de Investigaciones de la Provincia de buenos Aires (CIC), La Plata, Buenos Aires, Argentina
| | - M J Tolosa
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE) Universidad de La Plata-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comision de Investigaciones de la Provincia de buenos Aires (CIC), La Plata, Buenos Aires, Argentina
| | - N Di Siervi
- Instituto de Investigaciones Farmacológicas, ININFA, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina
| | - H B Schioth
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden
| | - C Davio
- Instituto de Investigaciones Farmacológicas, ININFA, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina
| | - M Perello
- Neurophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE) Universidad de La Plata-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comision de Investigaciones de la Provincia de buenos Aires (CIC), La Plata, Buenos Aires, Argentina
| | - J Raingo
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE) Universidad de La Plata-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comision de Investigaciones de la Provincia de buenos Aires (CIC), La Plata, Buenos Aires, Argentina
| |
Collapse
|
29
|
Melanocortin-4 receptor-regulated energy homeostasis. Nat Neurosci 2016; 19:206-19. [PMID: 26814590 DOI: 10.1038/nn.4202] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/13/2015] [Indexed: 12/11/2022]
Abstract
The melanocortin system provides a conceptual blueprint for the central control of energetic state. Defined by four principal molecular components--two antagonistically acting ligands and two cognate receptors--this phylogenetically conserved system serves as a prototype for hierarchical energy balance regulation. Over the last decade the application of conditional genetic techniques has facilitated the neuroanatomical dissection of the melanocortinergic network and identified the specific neural substrates and circuits that underscore the regulation of feeding behavior, energy expenditure, glucose homeostasis and autonomic outflow. In this regard, the melanocortin-4 receptor is a critical coordinator of mammalian energy homeostasis and body weight. Drawing on recent advances in neuroscience and genetic technologies, we consider the structure and function of the melanocortin-4 receptor circuitry and its role in energy homeostasis.
Collapse
|
30
|
Yang Z, Tao YX. Biased signaling initiated by agouti-related peptide through human melanocortin-3 and -4 receptors. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1485-94. [PMID: 27208795 DOI: 10.1016/j.bbadis.2016.05.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 05/09/2016] [Accepted: 05/16/2016] [Indexed: 01/01/2023]
Abstract
The neural melanocortin receptors (MCRs), melanocortin-3 and -4 receptors (MC3R and MC4R), have been increasingly recognized as important regulators of energy homeostasis. The orexigenic agouti-related peptide (AgRP), initially identified as an endogenous antagonist for both neural MCRs, has been suggested to be a biased agonist of MC4R independent of its antagonizing effects. In the present study, we sought to determine the potential of AgRP to regulate the activation of intracellular kinases, including extracellular signal-regulated kinase 1 and 2 (ERK1/2), AKT and AMP-activated protein kinase (AMPK), through neural MCRs. We showed that AgRP acted as a biased agonist in human MC3R (hMC3R), decreasing cAMP activity of constitutively active mutant (F347A) hMC3R but stimulating ERK1/2 activation in both wide type and F347A hMC3Rs. AgRP-stimulated ERK1/2 phosphorylation through MC3R was abolished by protein kinase A (PKA) inhibitor H-89 but not Rp-cAMPS, whereas AgRP-initiated ERK1/2 activation through MC4R was inhibited by phosphatidylinositol 3-kinase (PI3K) inhibitors wortmannin and LY294002. Both NDP-MSH and AgRP treatment induced significant AKT phosphorylation in GT1-7 cells but not in MC3R- or MC4R-transfected HEK293T cells. The phosphorylated AMPK levels in both GT1-7 cells and HERK293T cells transfected with neural MCRs were significantly decreased upon stimulation with NDP-MSH but not with AgRP. In summary, we provided novel data for AgRP-initiated multiple intracellular signaling pathways, demonstrating biased agonism of AgRP in both neural MCRs, leading to a better understanding of neural MCR pharmacology.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/metabolism
- Agouti-Related Protein/metabolism
- Amino Acid Substitution
- Central Nervous System/metabolism
- Cyclic AMP/metabolism
- HEK293 Cells
- Humans
- Kinetics
- Ligands
- MAP Kinase Signaling System
- Mutagenesis, Site-Directed
- Peptide Fragments/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor, Melanocortin, Type 3/agonists
- Receptor, Melanocortin, Type 3/genetics
- Receptor, Melanocortin, Type 3/metabolism
- Receptor, Melanocortin, Type 4/agonists
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/metabolism
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Zhao Yang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
31
|
Escudero P, Minoli I, González Marín M, Morando M, Avila L. Melanism and ontogeny: a case study in lizards of the Liolaemus fitzingerii group (Squamata: Liolaemini). CAN J ZOOL 2016. [DOI: 10.1139/cjz-2015-0112] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Color polymorphisms in general and melanism in particular have been the focus of many evolutionary adaptation studies. In lizards of the genus Liolaemus Wiegmann, 1834, patterns of melanism have been poorly studied, although they have been used as diagnostic characters for identifying and describing new species. We investigated the relationships between melanism, body size, sex, and time in a population of Rawson Lizard (Liolaemus xanthoviridis Cei and Scolaro, 1980) with extensive ventral melanism. The study took place in Bahía Isla Escondida, Chubut (Argentina), during three summer seasons (2012 to 2014). We tagged each individual, recorded body measurements and sex, and took ventral photographs to estimate the proportion of melanism. Our results showed that ventral melanism increased over time as each individual increased its snout–vent length (SVL). Body size explained 44% of the variation in melanism and males were more melanistic than females. Previous comparative studies of lizards in this species group showed no relationship between melanism and different taxonomic units or with thermal functionality. Here, we present evidence suggesting that melanism might be a character with an ontogenetic origin that is strongly associated with sex and body size. This pattern could be shared among species of this group of lizards, and even more importantly, it may be related to variable selection forces occurring throughout ontogeny.
Collapse
Affiliation(s)
- P.C. Escudero
- Grupo de Herpetología Patagónica, Instituto Patagónico para el Estudio de los Ecosistemas Continentales, Centro Nacional Patagónico–CONICET, Puerto Madryn, Bld. Alte. Brown 2915, (U9120ACD), Chubut, Argentina
- Grupo de Herpetología Patagónica, Instituto Patagónico para el Estudio de los Ecosistemas Continentales, Centro Nacional Patagónico–CONICET, Puerto Madryn, Bld. Alte. Brown 2915, (U9120ACD), Chubut, Argentina
| | - I. Minoli
- Grupo de Herpetología Patagónica, Instituto Patagónico para el Estudio de los Ecosistemas Continentales, Centro Nacional Patagónico–CONICET, Puerto Madryn, Bld. Alte. Brown 2915, (U9120ACD), Chubut, Argentina
- Grupo de Herpetología Patagónica, Instituto Patagónico para el Estudio de los Ecosistemas Continentales, Centro Nacional Patagónico–CONICET, Puerto Madryn, Bld. Alte. Brown 2915, (U9120ACD), Chubut, Argentina
| | - M.A. González Marín
- Grupo de Herpetología Patagónica, Instituto Patagónico para el Estudio de los Ecosistemas Continentales, Centro Nacional Patagónico–CONICET, Puerto Madryn, Bld. Alte. Brown 2915, (U9120ACD), Chubut, Argentina
- Grupo de Herpetología Patagónica, Instituto Patagónico para el Estudio de los Ecosistemas Continentales, Centro Nacional Patagónico–CONICET, Puerto Madryn, Bld. Alte. Brown 2915, (U9120ACD), Chubut, Argentina
| | - M. Morando
- Grupo de Herpetología Patagónica, Instituto Patagónico para el Estudio de los Ecosistemas Continentales, Centro Nacional Patagónico–CONICET, Puerto Madryn, Bld. Alte. Brown 2915, (U9120ACD), Chubut, Argentina
- Grupo de Herpetología Patagónica, Instituto Patagónico para el Estudio de los Ecosistemas Continentales, Centro Nacional Patagónico–CONICET, Puerto Madryn, Bld. Alte. Brown 2915, (U9120ACD), Chubut, Argentina
| | - L.J. Avila
- Grupo de Herpetología Patagónica, Instituto Patagónico para el Estudio de los Ecosistemas Continentales, Centro Nacional Patagónico–CONICET, Puerto Madryn, Bld. Alte. Brown 2915, (U9120ACD), Chubut, Argentina
- Grupo de Herpetología Patagónica, Instituto Patagónico para el Estudio de los Ecosistemas Continentales, Centro Nacional Patagónico–CONICET, Puerto Madryn, Bld. Alte. Brown 2915, (U9120ACD), Chubut, Argentina
| |
Collapse
|
32
|
Clemson CM, Yost J, Taylor AW. The Role of Alpha-MSH as a Modulator of Ocular Immunobiology Exemplifies Mechanistic Differences between Melanocortins and Steroids. Ocul Immunol Inflamm 2016; 25:179-189. [PMID: 26807874 PMCID: PMC5769144 DOI: 10.3109/09273948.2015.1092560] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Melanocortins are a highly conserved family of peptides and receptors that includes multiple proopiomelanocortin-derived peptides and five defined melanocortin receptors. The melanocortins have an important role in maintaining immune homeostasis and in suppressing inflammation. Within the healthy eye, the melanocortins have a central role in preventing inflammation and maintaining immune privilege. A central mediator of the anti-inflammatory activity is the non-steroidogenic melanocortin peptide alpha-melanocyte stimulating hormone. In this review we summarize the major findings of melanocortin regulation of ocular immunobiology with particular interest in the ability of melanocortin to induce immune tolerance and cytoprotection. The melanocortins have therapeutic potential because their mechanisms of action in regulating immunity are distinctly different from the actions of steroids.
Collapse
Affiliation(s)
- Christine M Clemson
- a Autoimmune and Rare Diseases , Mallinckrodt Pharmaceuticals , Hayward , CA , USA
| | - John Yost
- a Autoimmune and Rare Diseases , Mallinckrodt Pharmaceuticals , Hayward , CA , USA
| | - Andrew W Taylor
- b Department of Ophthalmology , Boston University School of Medicine , Boston , MA , USA
| |
Collapse
|
33
|
Müller A, Niederstadt L, Jonas W, Yi CX, Meyer F, Wiedmer P, Fischer J, Grötzinger C, Schürmann A, Tschöp M, Kleinau G, Grüters A, Krude H, Biebermann H. Ring Finger Protein 11 Inhibits Melanocortin 3 and 4 Receptor Signaling. Front Endocrinol (Lausanne) 2016; 7:109. [PMID: 27551276 PMCID: PMC4976663 DOI: 10.3389/fendo.2016.00109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 07/26/2016] [Indexed: 01/07/2023] Open
Abstract
Intact melanocortin signaling via the G protein-coupled receptors (GPCRs), melanocortin receptor 4 (MC4R), and melanocortin receptor 3 (MC3R) is crucial for body weight maintenance. So far, no connection between melanocortin signaling and hypothalamic inflammation has been reported. Using a bimolecular fluorescence complementation library screen, we identified a new interaction partner for these receptors, ring finger protein 11 (RNF11). RNF11 participates in the constitution of the A20 complex that is involved in reduction of tumor necrosis factor α (TNFα)-induced NFκB signaling, an important pathway in hypothalamic inflammation. Mice treated with high-fat diet (HFD) for 3 days demonstrated a trend toward an increase in hypothalamic Rnf11 expression, as shown for other inflammatory markers under HFD. Furthermore, Gs-mediated signaling of MC3/4R was demonstrated to be strongly reduced to 20-40% by co-expression of RNF11 despite unchanged total receptor expression. Cell surface expression was not affected for MC3R but resulted in a significant reduction of MC4R to 61% by co-expression with RNF11. Mechanisms linking HFD, inflammation, and metabolism remain partially understood. In this study, a new axis between signaling of specific body weight regulating GPCRs and factors involved in hypothalamic inflammation is suggested.
Collapse
Affiliation(s)
- Anne Müller
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lars Niederstadt
- Tumor Targeting Laboratory, Department of Hepatology and Gastroenterology, Molecular Cancer Research Center (MKFZ), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Wenke Jonas
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- German Center of Diabetes Research, Neuherberg, Germany
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Netherlands
| | - Franziska Meyer
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Wiedmer
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
| | - Jana Fischer
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Carsten Grötzinger
- Tumor Targeting Laboratory, Department of Hepatology and Gastroenterology, Molecular Cancer Research Center (MKFZ), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Annette Schürmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- German Center of Diabetes Research, Neuherberg, Germany
| | - Matthias Tschöp
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Germany, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
- Technische Universität München, München, Germany
| | - Gunnar Kleinau
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Annette Grüters
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Heiko Krude
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Heike Biebermann
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
- *Correspondence: Heike Biebermann,
| |
Collapse
|
34
|
Lisak RP, Nedelkoska L, Bealmear B, Benjamins JA. Melanocortin receptor agonist ACTH 1-39 protects rat forebrain neurons from apoptotic, excitotoxic and inflammation-related damage. Exp Neurol 2015; 273:161-7. [PMID: 26300474 DOI: 10.1016/j.expneurol.2015.08.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/12/2015] [Accepted: 08/14/2015] [Indexed: 12/22/2022]
Abstract
Patients with relapsing-remitting multiple sclerosis (RRMS) are commonly treated with high doses of intravenous corticosteroids (CS). ACTH 1-39, a member of the melanocortin family, stimulates production of CS by the adrenals, but melanocortin receptors are also found in the central nervous system (CNS) and on immune cells. ACTH is produced within the CNS and may have direct protective effects on glia and neurons independent of CS. We previously reported that ACTH 1-39 protected oligodendroglia (OL) and their progenitors (OPC) from a panel of excitotoxic and inflammation-related agents. Neurons are the most vulnerable cells in the CNS. They are terminally differentiated, and sensitive to inflammatory and excitotoxic insults. For potential therapeutic protection of gray matter, it is important to investigate the direct effects of ACTH on neurons. Cultures highly enriched in neurons were isolated from 2-3 day old rat brain. After 4-7 days in culture, the neurons were treated for 24h with selected toxic agents with or without ACTH 1-39. ACTH 1-39 protected neurons from death induced by staurosporine, glutamate, NMDA, AMPA, kainate, quinolinic acid, reactive oxygen species and, to a modest extent, from rapidly released NO, but did not protect against kynurenic acid or slowly released nitric oxide. Our results show that ACTH 1-39 protects neurons in vitro from several apoptotic, excitotoxic and inflammation-related insults.
Collapse
Affiliation(s)
- Robert P Lisak
- Department of Neurology, Wayne State University School of Medicine, 8D University Health Center, 4201 St. Antoine St., Detroit, MI 48201, USA.
| | - Liljana Nedelkoska
- Department of Neurology, Wayne State University School of Medicine, 8D University Health Center, 4201 St. Antoine St., Detroit, MI 48201, USA
| | - Beverly Bealmear
- Department of Neurology, Wayne State University School of Medicine, 8D University Health Center, 4201 St. Antoine St., Detroit, MI 48201, USA
| | - Joyce A Benjamins
- Department of Neurology, Wayne State University School of Medicine, 8D University Health Center, 4201 St. Antoine St., Detroit, MI 48201, USA
| |
Collapse
|
35
|
Ramírez D, Saba J, Carniglia L, Durand D, Lasaga M, Caruso C. Melanocortin 4 receptor activates ERK-cFos pathway to increase brain-derived neurotrophic factor expression in rat astrocytes and hypothalamus. Mol Cell Endocrinol 2015; 411:28-37. [PMID: 25892444 DOI: 10.1016/j.mce.2015.04.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 04/09/2015] [Accepted: 04/09/2015] [Indexed: 01/08/2023]
Abstract
Melanocortins are neuropeptides with well recognized anti-inflammatory and anti-apoptotic effects in the brain. Of the five melanocortin receptors (MCR), MC4R is abundantly expressed in the brain and is the only MCR present in astrocytes. We have previously shown that MC4R activation by the α-melanocyte stimulating hormone (α-MSH) analog, NDP-MSH, increased brain-derived neurotrophic factor (BDNF) expression through the classic cAMP-Protein kinase A-cAMP responsive element binding protein pathway in rat astrocytes. Now, we examined the participation of the mitogen activated protein kinases pathway in MC4R signaling. Rat cultured astrocytes treated with NDP-MSH 1 µM for 1 h showed increased BDNF expression. Inhibition of extracellular signal-regulated kinase (ERK) and ribosomal p90 S6 kinase (RSK), an ERK substrate, but not of p38 or JNK, prevented the increase in BDNF expression induced by NDP-MSH. Activation of MC4R increased cFos expression, a target of both ERK and RSK. ERK activation by MC4R involves cAMP, phosphoinositide-3 kinase (PI3K) and the non receptor tyrosine kinase, Src. Both PI3K and Src inhibition abolished NDP-MSH-induced BDNF expression. Moreover, we found that intraperitoneal injection of α-MSH induces BDNF and MC4R expression and activates ERK and cFos in male rat hypothalamus. Our results show for the first time that MC4R-induced BDNF expression in astrocytes involves ERK-RSK-cFos pathway which is dependent on PI3K and Src, and that melanocortins induce BDNF expression and ERK-cFos activation in rat hypothalamus.
Collapse
Affiliation(s)
- D Ramírez
- INBIOMED UBA-CONICET, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - J Saba
- INBIOMED UBA-CONICET, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - L Carniglia
- INBIOMED UBA-CONICET, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - D Durand
- INBIOMED UBA-CONICET, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - M Lasaga
- INBIOMED UBA-CONICET, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - C Caruso
- INBIOMED UBA-CONICET, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
36
|
Mountjoy KG. Pro-Opiomelanocortin (POMC) Neurones, POMC-Derived Peptides, Melanocortin Receptors and Obesity: How Understanding of this System has Changed Over the Last Decade. J Neuroendocrinol 2015; 27:406-18. [PMID: 25872650 DOI: 10.1111/jne.12285] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 04/04/2015] [Accepted: 04/07/2015] [Indexed: 12/19/2022]
Abstract
Following the cloning of the melanocortin receptor and agouti protein genes, a model was developed for the central melanocortin system with respect to the regulation of energy and glucose homeostasis. This model comprised leptin regulation of melanocortin peptides and agouti-related peptide (AgRP) produced from central pro-opiomelanocortin (POMC) and AgRP neurones, respectively, as well as AgRP competitive antagonism of melanocortin peptides activating melanocortin 4 receptor (MC4R) to Gαs and the cAMP signalling pathway. In the last decade, there have been paradigm shifts in our understanding of the central melanocortin system as a result of the application of advanced new technologies, including Cre-LoxP transgenic mouse technology, pharmacogenetics and optogenetics. During this period, our understanding of G protein coupled receptor signal transduction has also dramatically changed, such that these receptors are now known to exist in the plasma membrane oscillating between various inactive and active conformational states, and the active states signal through G protein-dependent and G protein-independent pathways. The present review focuses on evidence obtained over the past decade that has changed our understanding of POMC gene expression and regulation in the central nervous system, POMC and AgRP neuronal circuitry, neuroanatomical functions of melanocortin receptors, melanocortin 3 receptor (MC3R) and MC4R, and signal transduction through MC3R and MC4R.
Collapse
Affiliation(s)
- K G Mountjoy
- Departments of Physiology and Molecular Medicine and Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences and Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| |
Collapse
|
37
|
Rodrigues AR, Almeida H, Gouveia AM. Intracellular signaling mechanisms of the melanocortin receptors: current state of the art. Cell Mol Life Sci 2015; 72:1331-45. [PMID: 25504085 PMCID: PMC11113477 DOI: 10.1007/s00018-014-1800-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 11/07/2014] [Accepted: 12/01/2014] [Indexed: 12/28/2022]
Abstract
The melanocortin system is composed by the agonists adrenocorticotropic hormone and α, β and γ-melanocyte-stimulating hormone, and two naturally occurring antagonists, agouti and agouti-related protein. These ligands act by interaction with a family of five melanocortin receptors (MCRs), assisted by MCRs accessory proteins (MRAPs). MCRs stimulation activates different signaling pathways that mediate a diverse array of physiological processes, including pigmentation, energy metabolism, inflammation and exocrine secretion. This review focuses on the regulatory mechanisms of MCRs signaling, highlighting the differences among the five receptors. MCRs signal through G-dependent and independent mechanisms and their functional coupling to agonists at the cell surface is regulated by interacting proteins, namely MRAPs and β-arrestins. The knowledge of the distinct modulation pattern of MCRs signaling and function may be helpful for the future design of novel drugs able to combine specificity, safety and effectiveness in the course of their therapeutic use.
Collapse
Affiliation(s)
- Adriana R Rodrigues
- Department of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal,
| | | | | |
Collapse
|
38
|
Abstract
Although anti-inflammatory drugs are among the most common class of marketed drugs, chronic inflammatory conditions such as rheumatoid arthritis, multiple sclerosis or inflammatory bowel disease still represent unmet needs. New first-in-class drugs might be discovered in the future but the repurpose and further development of old drugs also offers promise for these conditions. This is the case of the melanocortin adrenocorticotropin hormone, ACTH, used in patients since 1952 but regarded as the last therapeutic option when other medications, such as glucocorticoids, cannot be used. Better understanding on its physiological and pharmacological mechanisms of actions and new insights on melanocortin receptors biology have revived the interest on rescuing this old and effective drug. ACTH does not only induce cortisol production, as previously assumed, but it also exerts anti-inflammatory actions by targeting melanocortin receptors present on immune cells. The endogenous agonists for these receptors (ACTH, α-, β-, and γ-melanocyte stimulating hormones), are also produced locally by immune cells, indicating the existence of an endogenous anti-inflammatory tissue-protective circuit involving the melanocortin system. These findings suggested that new ACTH-like melanocortin drugs devoid of steroidogenic actions, and hence side effects, could be developed. This review summarizes the actions of ACTH and melanocortin drugs, their role as endogenous pro-resolving mediators, their current clinical use and provides an overview on how recent advances on GPCR functioning may lead to a novel class of drugs.
Collapse
|
39
|
Tao YX. Constitutive activity in melanocortin-4 receptor: biased signaling of inverse agonists. ADVANCES IN PHARMACOLOGY 2015; 70:135-54. [PMID: 24931195 DOI: 10.1016/b978-0-12-417197-8.00005-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The melanocortin-4 receptor (MC4R) is a critical regulator of energy homeostasis, including both energy intake and energy expenditure. It mediates the actions of a number of hormones on energy balance. The endogenous ligands for MC4R include peptide agonists derived from processing of proopiomelanocortin and the antagonist Agouti-related peptide (AgRP). Wild-type MC4R has some basal (constitutive) activity. Naturally occurring and laboratory-generated mutations have been identified, which results in either increased or decreased basal activities. Impaired basal signaling has been suggested to be a cause of dysregulated energy homeostasis and early-onset obesity, although several constitutively active mutations have also been identified from obese patients. AgRP and several small-molecule antagonists have been shown to be inverse agonists in the Gs-cAMP pathway. However, in the extracellular signal-regulated kinase (ERK) 1/2 pathway, we showed that these inverse agonists are potent agonists, demonstrating convincingly that they are biased ligands. We also showed that some mutations that do not cause constitutive activation in the Gs-cAMP pathway cause constitutive activation in the ERK1/2 pathway, suggesting that they are biased receptors. The physiological and potential pathophysiological relevance of the biased constitutive signaling in MC4R and therapeutic potential remain to be investigated.
Collapse
Affiliation(s)
- Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA.
| |
Collapse
|
40
|
Abstract
Background:Neuropathic pain is characterised by spontaneous ongoing or shooting pain and evoked amplified pain responses after noxious or non-noxious stimuli. Neuropathic pain develops as a result of lesions or disease affecting the somatosensory nervous system either in the periphery or centrally. Melanocortin 4 receptor (MC4R) plays an important role in the initiation of neuropathic pain but the underlying mechanisms are still unclear.Methods:Adult male Wistar rats were given chronic constriction injury (CCI) or sham operations. Part of CCI rats were intrathecally treated with HS014 (MC4R antagonist) or SB203580 (p38MAPK inhibitor). On the third, seventh and fourteenth day, the thermal threshold of operated paws was tested. In addition, the MC4R or phosphorylated p38MAPK (p-p38MAPK) levels of lumbar spinal cord were tested with ELISA (enzyme-linked immunosorbent assay), western blot and immunohistochemistry.Results:Here we demonstrate that (1) both HS014 and SB203580 reduced CCI reduced hyperalgesia (2) p-p38MAPK was increased after CCI with a time course parallel to that of the MC4R change, (3) The p38 activation was prevented by blocking MC4R with an antagonist HS014, but MC4R-IR was not prevented by SB203580. (4) MC4R and p-p38MAPK were located in the same cells.Conclusion:The mechanisms of neuropathic pain mediated by MC4R is related to the inhibition of p38MAPK activation. P38MAPK may be a downstream of MC4R.
Collapse
|
41
|
He S, Tao YX. Defect in MAPK signaling as a cause for monogenic obesity caused by inactivating mutations in the melanocortin-4 receptor gene. Int J Biol Sci 2014; 10:1128-37. [PMID: 25332687 PMCID: PMC4202029 DOI: 10.7150/ijbs.10359] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 09/15/2014] [Indexed: 12/22/2022] Open
Abstract
The melanocortin-4 receptor (MC4R) is a Family A G protein-coupled receptor that plays an essential role in regulating energy homeostasis, including both energy intake and expenditure. Mutations leading to a reduced MC4R function confer a major gene effect for obesity. More than 170 distinct mutations have been identified in humans. In addition to the conventional Gs-stimulated cAMP pathway, the MC4R also activates MAPKs, especially ERK1/2. We also showed there is biased signaling in the two signaling pathways, with inverse agonists in the Gs-cAMP pathway acting as agonists for the ERK1/2 pathway. In the current study, we sought to determine whether defects in basal or agonist-induced ERK1/2 activation in MC4R mutants might potentially contribute to obesity pathogenesis in patients carrying these mutations. The constitutive and ligand-stimulated ERK1/2 activation were measured in wild type and 73 naturally occurring MC4R mutations. We showed that nineteen mutants had significantly decreased basal pERK1/2 level, and five Class V variants (where no functional defects have been identified previously), C40R, V50M, T112M, A154D and S295P, had impaired ligand-stimulated ERK1/2 activation. Our studies demonstrated for the first time that decreased basal or ligand-stimulated ERK1/2 signaling might contribute to obesity pathogenesis caused by mutations in the MC4R gene. We also observed biased signaling in 25 naturally occurring mutations in the Gs-cAMP and ERK1/2 pathways.
Collapse
Affiliation(s)
- Shan He
- 1. Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama 36849-5519, USA. ; 2. Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Hubei Collaborative Innovation Center for Freshwater Aquaculture, Wuhan, Hubei 430070, China
| | - Ya-Xiong Tao
- 1. Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama 36849-5519, USA
| |
Collapse
|
42
|
Gonzalez P, Machado I, Vilcaes A, Caruso C, Roth GA, Schiöth H, Lasaga M, Scimonelli T. Molecular mechanisms involved in interleukin 1-beta (IL-1β)-induced memory impairment. Modulation by alpha-melanocyte-stimulating hormone (α-MSH). Brain Behav Immun 2013; 34:141-50. [PMID: 23968970 DOI: 10.1016/j.bbi.2013.08.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 08/05/2013] [Accepted: 08/14/2013] [Indexed: 11/26/2022] Open
Abstract
Pro-inflammatory cytokines can affect cognitive processes such as learning and memory. Particularly, interleukin-1β (IL-1β) influences the consolidation of hippocampus-dependent memories. We previously reported that administration of IL-1β in dorsal hippocampus impaired contextual fear memory consolidation. Different mechanisms have been implicated in the action of IL-1β on long-term potentiation (LTP), but the processes by which this inhibition occurs in vivo remain to be elucidated. We herein report that intrahippocampal injection of IL-1β induced a significant increase in p38 phosphorylation after contextual fear conditioning. Also, treatment with SB203580, an inhibitor of p38, reversed impairment induced by IL-1β on conditioned fear behavior, indicating that this MAPK would be involved in the effect of the cytokine. We also showed that IL-1β administration produced a decrease in glutamate release from dorsal hippocampus synaptosomes and that treatment with SB203580 partially reversed this effect. Our results indicated that IL-1β-induced impairment in memory consolidation could be mediated by a decrease in glutamate release. This hypothesis is sustained by the fact that treatment with d-cycloserine (DCS), a partial agonist of the NMDA receptor, reversed the effect of IL-1β on contextual fear memory. Furthermore, we demonstrated that IL-1β produced a temporal delay in ERK phosphorylation and that DCS administration reversed this effect. We also observed that intrahippocampal injection of IL-1β decreased BDNF expression after contextual fear conditioning. We previously demonstrated that α-MSH reversed the detrimental effect of IL-1β on memory consolidation. The present results demonstrate that α-MSH administration did not modify the decrease in glutamate release induced by IL-1β. However, intrahippocampal injection of α-MSH prevented the effect on ERK phosphorylation and BDNF expression induced by IL-1β after contextual fear conditioning. Therefore, in the present study we determine possible molecular mechanisms involved in the impairment induced by IL-1β on fear memory consolidation. We also established how this effect could be modulated by α-MSH.
Collapse
Affiliation(s)
- P Gonzalez
- IFEC-CONICET, Depto. Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Caruso C, Carniglia L, Durand D, Scimonelli TN, Lasaga M. Astrocytes: new targets of melanocortin 4 receptor actions. J Mol Endocrinol 2013; 51:R33-50. [PMID: 23881919 DOI: 10.1530/jme-13-0064] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Astrocytes exert a wide variety of functions with paramount importance in brain physiology. After injury or infection, astrocytes become reactive and they respond by producing a variety of inflammatory mediators that help maintain brain homeostasis. Loss of astrocyte functions as well as their excessive activation can contribute to disease processes; thus, it is important to modulate reactive astrocyte response. Melanocortins are peptides with well-recognized anti-inflammatory and neuroprotective activity. Although melanocortin efficacy was shown in systemic models of inflammatory disease, mechanisms involved in their effects have not yet been fully elucidated. Central anti-inflammatory effects of melanocortins and their mechanisms are even less well known, and, in particular, the effects of melanocortins in glial cells are poorly understood. Of the five known melanocortin receptors (MCRs), only subtype 4 is present in astrocytes. MC4R has been shown to mediate melanocortin effects on energy homeostasis, reproduction, inflammation, and neuroprotection and, recently, to modulate astrocyte functions. In this review, we will describe MC4R involvement in anti-inflammatory, anorexigenic, and anti-apoptotic effects of melanocortins in the brain. We will highlight MC4R action in astrocytes and discuss their possible mechanisms of action. Melanocortin effects on astrocytes provide a new means of treating inflammation, obesity, and neurodegeneration, making them attractive targets for therapeutic interventions in the CNS.
Collapse
Affiliation(s)
- Carla Caruso
- School of Medicine, Biomedical Research Institute (UBA-CONICET), University of Buenos Aires, Paraguay 2155 piso 10, 1121ABG Buenos Aires, Argentina IFEC (CONICET) Department of Pharmacology, School of Chemistry, National University of Córdoba, Córdoba, Argentina
| | | | | | | | | |
Collapse
|
44
|
Taylor AW. Alpha-melanocyte stimulating hormone (α-MSH) is a post-caspase suppressor of apoptosis in RAW 264.7 macrophages. PLoS One 2013; 8:e74488. [PMID: 24009773 PMCID: PMC3757010 DOI: 10.1371/journal.pone.0074488] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 08/01/2013] [Indexed: 01/04/2023] Open
Abstract
The neuropeptide alpha-melanocyte stimulating hormone (α-MSH) is an important regulator of immune cell activity within the immunosuppressive ocular microenvironment. Its constitutive presence not only suppresses macrophage inflammatory activity, it also participates in retinal pigment epithelial cell (RPE) mediated activation of macrophages to function similar to myeloid suppressor cells. In addition, α-MSH promotes survival of the alternatively activated macrophages where without α-MSH RPE induce apoptosis in the macrophages, which is seen as increased TUNEL stained cells. Since there is little know about α-MSH as an anti-apoptotic factor, the effects of α-MSH on caspase activity, mitochondrial membrane potential, Bcl2 to BAX expression, along with TUNEL staining, and Annexin V binding were examined in RAW 264.7 macrophages under serum-starved conditions that trigger apoptosis. There was no effect of α-MSH on activated Caspase 9 and Caspase 3 while there was suppression of Caspase 8 activity. In addition, α-MSH did not improve mitochondrial membrane potential, change the ratio between Bcl-2 and BAX, nor reduce Annexin V binding. These results demonstrate that the diminution in TUNEL staining by α-MSH is through α-MSH mediating suppression of the apoptotic pathway that is post-Caspase 3, but before DNA fragmentation. Therefore, as α-MSH promotes the alternative activation of macrophages it also provides a survival signal, and the potential for the caspases to participate in non-apoptotic activities that can contribute to an immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Andrew W Taylor
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, USA.
| |
Collapse
|
45
|
Schaible EV, Steinsträßer A, Jahn-Eimermacher A, Luh C, Sebastiani A, Kornes F, Pieter D, Schäfer MK, Engelhard K, Thal SC. Single administration of tripeptide α-MSH(11-13) attenuates brain damage by reduced inflammation and apoptosis after experimental traumatic brain injury in mice. PLoS One 2013; 8:e71056. [PMID: 23940690 PMCID: PMC3733710 DOI: 10.1371/journal.pone.0071056] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 06/24/2013] [Indexed: 12/29/2022] Open
Abstract
Following traumatic brain injury (TBI) neuroinflammatory processes promote neuronal cell loss. Alpha-melanocyte-stimulating hormone (α-MSH) is a neuropeptide with immunomodulatory properties, which may offer neuroprotection. Due to short half-life and pigmentary side-effects of α-MSH, the C-terminal tripeptide α-MSH(11-13) may be an anti-inflammatory alternative. The present study investigated the mRNA concentrations of the precursor hormone proopiomelanocortin (POMC) and of melanocortin receptors 1 and 4 (MC1R/MC4R) in naive mice and 15 min, 6, 12, 24, and 48 h after controlled cortical impact (CCI). Regulation of POMC and MC4R expression did not change after trauma, while MC1R levels increased over time with a 3-fold maximum at 12 h compared to naive brain tissue. The effect of α-MSH(11-13) on secondary lesion volume determined in cresyl violet stained sections (intraperitoneal injection 30 min after insult of 1 mg/kg α-MSH(11-13) or 0.9% NaCl) showed a considerable smaller trauma in α-MSH(11-13) injected mice. The expression of the inflammatory markers TNF-α and IL-1β as well as the total amount of Iba-1 positive cells were not reduced. However, cell branch counting of Iba-1 positive cells revealed a reduced activation of microglia. Furthermore, tripeptide injection reduced neuronal apoptosis analyzed by cleaved caspase-3 and NeuN staining. Based on the results single α-MSH(11-13) administration offers a promising neuroprotective property by modulation of inflammation and prevention of apoptosis after traumatic brain injury.
Collapse
Affiliation(s)
- Eva-Verena Schaible
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Arne Steinsträßer
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Antje Jahn-Eimermacher
- Institute of Medical Biostatistics, Epidemiology and Informatics, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Clara Luh
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Anne Sebastiani
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Frida Kornes
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Dana Pieter
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Michael K. Schäfer
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Focus Program Translational Neuroscience, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Kristin Engelhard
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Focus Program Translational Neuroscience, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Serge C. Thal
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Focus Program Translational Neuroscience, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- * E-mail:
| |
Collapse
|
46
|
Mo XL, Tao YX. Activation of MAPK by inverse agonists in six naturally occurring constitutively active mutant human melanocortin-4 receptors. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1939-48. [PMID: 23791567 DOI: 10.1016/j.bbadis.2013.06.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 05/17/2013] [Accepted: 06/05/2013] [Indexed: 01/14/2023]
Abstract
The melanocortin-4 receptor (MC4R) is a G protein-coupled receptor that plays an essential role in regulating energy homeostasis. Defects in MC4R are the most common monogenic form of obesity, with about 170 distinct mutations identified in human. In addition to the conventional Gs-stimulated adenylyl cyclase pathway, it has been recently demonstrated that MC4R also activates mitogen-activated protein kinases, extracellular signal-regulated kinases 1 and 2 (ERK1/2). Herein, we investigated the potential of four MC4R ligands that are inverse agonists at the Gs-cAMP signaling pathway, including agouti-related peptide (AgRP), MCL0020, Ipsen 5i and ML00253764, to regulate ERK1/2 activation (pERK1/2) in wild type and six naturally occurring constitutively active mutant (CAM) MC4Rs. We showed that these four inverse agonists acted as agonists for the ERK1/2 signaling cascade in wild type and CAM MC4Rs. Three mutants (P230L, L250Q and F280L) had significantly increased pERK1/2 level upon stimulation with all four inverse agonists, with maximal induction ranging from 1.6 to 4.2-fold. D146N had significantly increased pERK1/2 level upon stimulation with AgRP, MCL0020 or ML00253764, but not Ipsen 5i. The pERK1/2 levels of H76R and S127L were significantly increased only upon stimulation with AgRP or MCL0020. In summary, our studies demonstrated for the first time that MC4R inverse agonists at the Gs-cAMP pathway could serve as agonists in the MAPK pathway. These results suggested that there were multiple activation states of MC4R with ligand-specific and/or mutant-specific conformations capable of differentially coupling the MC4R to distinct signaling pathways.
Collapse
Affiliation(s)
- Xiu-Lei Mo
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | | |
Collapse
|
47
|
Kaneva MK, Kerrigan MJP, Grieco P, Curley GP, Locke IC, Getting SJ. Chondroprotective and anti-inflammatory role of melanocortin peptides in TNF-α activated human C-20/A4 chondrocytes. Br J Pharmacol 2013; 167:67-79. [PMID: 22471953 DOI: 10.1111/j.1476-5381.2012.01968.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Melanocortin MC(1) and MC(3 ) receptors, mediate the anti-inflammatory effects of melanocortin peptides. Targeting these receptors could therefore lead to development of novel anti-inflammatory therapeutic agents. We investigated the expression of MC(1) and MC(3) receptors on chondrocytes and the role of α-melanocyte-stimulating hormone (α-MSH) and the selective MC(3) receptor agonist, [DTRP(8) ]-γ-MSH, in modulating production of inflammatory cytokines, tissue-destructive proteins and induction of apoptotic pathway(s) in the human chondrocytic C-20/A4 cells. EXPERIMENTAL APPROACH Effects of α-MSH, [DTRP(8) ]-γ-MSH alone or in the presence of the MC(3/4) receptor antagonist, SHU9119, on TNF-α induced release of pro-inflammatory cytokines, MMPs, apoptotic pathway(s) and cell death in C-20/A4 chondrocytes were investigated, along with their effect on the release of the anti-inflammatory cytokine IL-10. KEY RESULTS C-20/A4 chondrocytes expressed functionally active MC(1,3) receptors. α-MSH and [DTRP(8) ]-γ-MSH treatment, for 30 min before TNF-α stimulation, provided a time-and-bell-shaped concentration-dependent decrease in pro-inflammatory cytokines (IL-1β, IL-6 and IL-8) release and increased release of the chondroprotective and anti-inflammatory cytokine, IL-10, whilst decreasing expression of MMP1, MMP3, MMP13 genes.α-MSH and [DTRP(8) ]-γ-MSH treatment also inhibited TNF-α-induced caspase-3/7 activation and chondrocyte death. The effects of [DTRP(8) ]-γ-MSH, but not α-MSH, were abolished by the MC(3/4) receptor antagonist, SHU9119. CONCLUSION AND IMPLICATIONS Activation of MC(1) /MC(3) receptors in C-20/A4 chondrocytes down-regulated production of pro-inflammatory cytokines and cartilage-destroying proteinases, inhibited initiation of apoptotic pathways and promoted release of chondroprotective and anti-inflammatory cytokines. Developing small molecule agonists to MC(1) /MC(3) receptors could be a viable approach for developing chondroprotective and anti-inflammatory therapies in rheumatoid and osteoarthritis.
Collapse
|
48
|
Muceniece R, Dambrova M. Melanocortins in brain inflammation: the role of melanocortin receptor subtypes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 681:61-70. [PMID: 21222260 DOI: 10.1007/978-1-4419-6354-3_5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The melanocortins (MC) are released from neurons and paracrine cells in the CNS where they are involved in important physiological functions, including regulation of body temperature and immune responses. MC bind to melanocortin receptors, a class of cell surface G-protein-coupled receptors. Of the five subtypes of MC receptors that have been cloned in mammals, the MC1, MC3, MC4 and MC5 receptors are expressed in brain tissues. Expression of MC receptors in both brain cells and cells of the immune system suggests direct involvement of MC in regulation of inflammatory processes in the brain. The binding of MC to MC receptors induces activation of adenylate cyclase, increase in intracellular cAMP level and, consequently, inhibition of the nuclear transcription factor kappaB (NF-κB) signalling. Inflammatory processes contribute to development of severe CNS diseases, both in acute and chronic conditions. Thus far, the anti-inflammatory effects of MC in the CNS have been mainly studied using peptides that are relatively unselective for individual MC receptor subtypes. Consequently, these studies do not allow identification of specific MC receptor(s) involved in the regulation of inflammatory processes. However, recently synthesized ligands selective for individual MC receptors indicated that both MC4 and MC3 agonists are promising anti-inflammatory agents in treatment of brain inflammation.
Collapse
Affiliation(s)
- Ruta Muceniece
- Faculty of Medicine, University of Latvia, Sarlotes St. 1a, Riga, LV-1001, Latvia.
| | | |
Collapse
|
49
|
Israel DD, Sheffer-Babila S, de Luca C, Jo YH, Liu SM, Xia Q, Spergel DJ, Dun SL, Dun NJ, Chua SC. Effects of leptin and melanocortin signaling interactions on pubertal development and reproduction. Endocrinology 2012; 153:2408-19. [PMID: 22408174 PMCID: PMC3381095 DOI: 10.1210/en.2011-1822] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Leptin and melanocortin signaling control ingestive behavior, energy balance, and substrate utilization, but only leptin signaling defects cause hypothalamic hypogonadism and infertility. Although GnRH neurons do not express leptin receptors, leptin influences GnRH neuron activity via regulation of immediate downstream mediators including the neuropeptides neuropeptide Y and the melanocortin agonist and antagonist, α-MSH, agouti-related peptide, respectively. Here we show that modulation of melanocortin signaling in female db/db mice through ablation of agouti-related peptide, or heterozygosity of melanocortin 4 receptor, restores the timing of pubertal onset, fertility, and lactation. Additionally, melanocortin 4 receptor activation increases action potential firing and induces c-Fos expression in GnRH neurons, providing further evidence that melanocortin signaling influences GnRH neuron activity. These studies thus establish melanocortin signaling as an important component in the leptin-mediated regulation of GnRH neuron activity, initiation of puberty and fertility.
Collapse
Affiliation(s)
- Davelene D Israel
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Damm E, Buech TRH, Gudermann T, Breit A. Melanocortin-induced PKA activation inhibits AMPK activity via ERK-1/2 and LKB-1 in hypothalamic GT1-7 cells. Mol Endocrinol 2012; 26:643-54. [PMID: 22361823 DOI: 10.1210/me.2011-1218] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
α-Melanocyte-stimulating hormone (α-MSH)-induced activation of the melanocortin-4 receptor in hypothalamic neurons increases energy expenditure and inhibits food intake. Active hypothalamic AMP-activated protein kinase (AMPK) has recently been reported to enhance food intake, and in vivo experiments suggested that intrahypothalamic injection of melanocortins decreased food intake due to the inhibition of AMPK activity. However, it is not clear whether α-MSH affects AMPK via direct intracellular signaling cascades or if the release of paracrine factors is involved. Here, we used a murine, hypothalamic cell line (GT1-7 cells) and monitored AMPK phosphorylation at Thr(172), which has been suggested to increase AMPK activity. We found that α-MSH dephosphorylated AMPK at Thr(172) and consequently decreased phosphorylation of the established AMPK substrate acetyl-coenzyme A-carboxylase at Ser(79). Inhibitory effects of α-MSH on AMPK were blocked by specific inhibitors of protein kinase A (PKA) or ERK-1/2, pointing to an important role of both kinases in this process. Because α-MSH-induced activation of ERK-1/2 was blunted by PKA inhibitors, we propose that ERK-1/2 serves as a link between PKA and AMPK in GT1-7 cells. Furthermore, down-regulation of liver kinase B-1, but not inhibition of calcium-calmodulin-dependent kinase kinase-β or TGFβ-activated kinase-1 decreased basal phosphorylation of AMPK and its dephosphorylation induced by α-MSH. Thus, we propose that α-MSH inhibits AMPK activity via a linear pathway, including PKA, ERK-1/2, and liver kinase B-1 in GT1-7 cells. Given the importance of the melanocortin system in the formation of adipositas, detailed knowledge about this pathway might help to develop drugs targeting obesity.
Collapse
Affiliation(s)
- Ellen Damm
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität, Goethestrasse 33, 80336 München, Germany
| | | | | | | |
Collapse
|