1
|
Strnadová V, Pačesová A, Charvát V, Šmotková Z, Železná B, Kuneš J, Maletínská L. Anorexigenic neuropeptides as anti-obesity and neuroprotective agents: exploring the neuroprotective effects of anorexigenic neuropeptides. Biosci Rep 2024; 44:BSR20231385. [PMID: 38577975 PMCID: PMC11043025 DOI: 10.1042/bsr20231385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/26/2024] [Accepted: 04/05/2024] [Indexed: 04/06/2024] Open
Abstract
Since 1975, the incidence of obesity has increased to epidemic proportions, and the number of patients with obesity has quadrupled. Obesity is a major risk factor for developing other serious diseases, such as type 2 diabetes mellitus, hypertension, and cardiovascular diseases. Recent epidemiologic studies have defined obesity as a risk factor for the development of neurodegenerative diseases, such as Alzheimer's disease (AD) and other types of dementia. Despite all these serious comorbidities associated with obesity, there is still a lack of effective antiobesity treatment. Promising candidates for the treatment of obesity are anorexigenic neuropeptides, which are peptides produced by neurons in brain areas implicated in food intake regulation, such as the hypothalamus or the brainstem. These peptides efficiently reduce food intake and body weight. Moreover, because of the proven interconnection between obesity and the risk of developing AD, the potential neuroprotective effects of these two agents in animal models of neurodegeneration have been examined. The objective of this review was to explore anorexigenic neuropeptides produced and acting within the brain, emphasizing their potential not only for the treatment of obesity but also for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Veronika Strnadová
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Andrea Pačesová
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Vilém Charvát
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Zuzana Šmotková
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Blanka Železná
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Jaroslav Kuneš
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
- Department of Biochemistry and Molecular Biology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Lenka Maletínská
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
2
|
Garnsey MR, Smith AC, Polivkova J, Arons AL, Bai G, Blakemore C, Boehm M, Buzon LM, Campion SN, Cerny M, Chang SC, Coffman K, Farley KA, Fonseca KR, Ford KK, Garren J, Kong JX, Koos MRM, Kung DW, Lian Y, Li MM, Li Q, Martinez-Alsina LA, O'Connor R, Ogilvie K, Omoto K, Raymer B, Reese MR, Ryder T, Samp L, Stevens KA, Widlicka DW, Yang Q, Zhu K, Fortin JP, Sammons MF. Discovery of the Potent and Selective MC4R Antagonist PF-07258669 for the Potential Treatment of Appetite Loss. J Med Chem 2023; 66:3195-3211. [PMID: 36802610 DOI: 10.1021/acs.jmedchem.2c02012] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
The melanocortin-4 receptor (MC4R) is a centrally expressed, class A GPCR that plays a key role in the regulation of appetite and food intake. Deficiencies in MC4R signaling result in hyperphagia and increased body mass in humans. Antagonism of MC4R signaling has the potential to mitigate decreased appetite and body weight loss in the setting of anorexia or cachexia due to underlying disease. Herein, we report on the identification of a series of orally bioavailable, small-molecule MC4R antagonists using a focused hit identification effort and the optimization of these antagonists to provide clinical candidate 23. Introduction of a spirocyclic conformational constraint allowed for simultaneous optimization of MC4R potency and ADME attributes while avoiding the production of hERG active metabolites observed in early series leads. Compound 23 is a potent and selective MC4R antagonist with robust efficacy in an aged rat model of cachexia and has progressed into clinical trials.
Collapse
Affiliation(s)
| | - Aaron C Smith
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | - Jana Polivkova
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | - Autumn L Arons
- Pfizer, Incorporated, Cambridge, Massachusetts 02139, United States
| | - Guoyun Bai
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | | | - Markus Boehm
- Pfizer, Incorporated, Cambridge, Massachusetts 02139, United States
| | - Leanne M Buzon
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | - Sarah N Campion
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | - Matthew Cerny
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | - Shiao-Chi Chang
- Pfizer, Incorporated, Cambridge, Massachusetts 02139, United States
| | - Karen Coffman
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | | | - Kari R Fonseca
- Pfizer, Incorporated, Cambridge, Massachusetts 02139, United States
| | - Kristen K Ford
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | - Jeonifer Garren
- Pfizer, Incorporated, Cambridge, Massachusetts 02139, United States
| | - Jimmy X Kong
- Pfizer, Incorporated, Cambridge, Massachusetts 02139, United States
| | - Martin R M Koos
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | - Daniel W Kung
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | - Yajing Lian
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | - Monica M Li
- Pfizer, Incorporated, Cambridge, Massachusetts 02139, United States
| | - Qifang Li
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | | | | | - Kevin Ogilvie
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | - Kiyoyuki Omoto
- Pfizer, Incorporated, Cambridge, Massachusetts 02139, United States
| | - Brian Raymer
- Pfizer, Incorporated, Cambridge, Massachusetts 02139, United States
| | - Matthew R Reese
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | - Tim Ryder
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | - Lacey Samp
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | | | | | - Qingyi Yang
- Pfizer, Incorporated, Cambridge, Massachusetts 02139, United States
| | - Kaicheng Zhu
- Pfizer, Incorporated, Groton, Connecticut 06340, United States
| | | | | |
Collapse
|
3
|
Kurtzhals P, Østergaard S, Nishimura E, Kjeldsen T. Derivatization with fatty acids in peptide and protein drug discovery. Nat Rev Drug Discov 2023; 22:59-80. [PMID: 36002588 DOI: 10.1038/s41573-022-00529-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2022] [Indexed: 01/28/2023]
Abstract
Peptides and proteins are widely used to treat a range of medical conditions; however, they often have to be injected and their effects are short-lived. These shortcomings of the native structure can be addressed by molecular engineering, but this is a complex undertaking. A molecular engineering technology initially applied to insulin - and which has now been successfully applied to several biopharmaceuticals - entails the derivatization of peptides and proteins with fatty acids. Various protraction mechanisms are enabled by the specific characteristics and positions of the attached fatty acid. Furthermore, the technology can ensure a long half-life following oral administration of peptide drugs, can alter the distribution of peptides and may hold potential for tissue targeting. Due to the inherent safety and well-defined chemical nature of the fatty acids, this technology provides a versatile approach to peptide and protein drug discovery.
Collapse
|
4
|
Yuan XC, Tao YX. Ligands for Melanocortin Receptors: Beyond Melanocyte-Stimulating Hormones and Adrenocorticotropin. Biomolecules 2022; 12:biom12101407. [PMID: 36291616 PMCID: PMC9599618 DOI: 10.3390/biom12101407] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
The discovery of melanocortins in 1916 has resulted in more than 100 years of research focused on these peptides. Extensive studies have elucidated well-established functions of melanocortins mediated by cell surface receptors, including MSHR (melanocyte-stimulating hormone receptor) and ACTHR (adrenocorticotropin receptor). Subsequently, three additional melanocortin receptors (MCRs) were identified. Among these five MCRs, MC3R and MC4R are expressed primarily in the central nervous system, and are therefore referred to as the neural MCRs. Since the central melanocortin system plays important roles in regulating energy homeostasis, targeting neural MCRs is emerging as a therapeutic approach for treating metabolic conditions such as obesity and cachexia. Early efforts modifying endogenous ligands resulted in the development of many potent and selective ligands. This review focuses on the ligands for neural MCRs, including classical ligands (MSH and agouti-related peptide), nonclassical ligands (lipocalin 2, β-defensin, small molecules, and pharmacoperones), and clinically approved ligands (ACTH, setmelanotide, bremelanotide, and several repurposed drugs).
Collapse
Affiliation(s)
- Xiao-Chen Yuan
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230061, China
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
- Correspondence:
| |
Collapse
|
5
|
Yeo GSH, Chao DHM, Siegert AM, Koerperich ZM, Ericson MD, Simonds SE, Larson CM, Luquet S, Clarke I, Sharma S, Clément K, Cowley MA, Haskell-Luevano C, Van Der Ploeg L, Adan RAH. The melanocortin pathway and energy homeostasis: From discovery to obesity therapy. Mol Metab 2021; 48:101206. [PMID: 33684608 PMCID: PMC8050006 DOI: 10.1016/j.molmet.2021.101206] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 02/28/2021] [Accepted: 03/03/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Over the past 20 years, insights from human and mouse genetics have illuminated the central role of the brain leptin-melanocortin pathway in controlling mammalian food intake, with genetic disruption resulting in extreme obesity, and more subtle polymorphic variations influencing the population distribution of body weight. At the end of 2020, the U.S. Food and Drug Administration (FDA) approved setmelanotide, a melanocortin 4 receptor agonist, for use in individuals with severe obesity due to either pro-opiomelanocortin (POMC), proprotein convertase subtilisin/kexin type 1 (PCSK1), or leptin receptor (LEPR) deficiency. SCOPE OF REVIEW Herein, we chart the melanocortin pathway's history, explore its pharmacology, genetics, and physiology, and describe how a neuropeptidergic circuit became an important druggable obesity target. MAJOR CONCLUSIONS Unravelling the genetics of the subset of severe obesity has revealed the importance of the melanocortin pathway in appetitive control; coupling this with studying the molecular pharmacology of compounds that bind melanocortin receptors has brought a new obesity drug to the market. This process provides a drug discovery template for complex disorders, which for setmelanotide took 25 years to transform from a single gene into an approved drug.
Collapse
Affiliation(s)
- Giles S H Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| | | | - Anna-Maria Siegert
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| | - Zoe M Koerperich
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA 55455.
| | - Mark D Ericson
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA 55455.
| | - Stephanie E Simonds
- Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute, and Department of Physiology, Monash University, Clayton, Victoria, Australia.
| | - Courtney M Larson
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA 55455.
| | - Serge Luquet
- Université de Paris, BFA, UMR 8251, CNRS, Paris, France.
| | - Iain Clarke
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC 3010, Australia.
| | | | - Karine Clément
- Assistance Publique Hôpitaux de Paris, Nutrition Department, Pitié-Salpêtrière Hospital, Paris, France, Sorbonne Université, INSERM, Nutrition and Obesity: Systemic Approaches (NutriOmics) Research Unit, Paris, France.
| | - Michael A Cowley
- Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute, and Department of Physiology, Monash University, Clayton, Victoria, Australia.
| | - Carrie Haskell-Luevano
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA 55455.
| | | | - Roger A H Adan
- Department of Translational Neuroscience, UMCU Brain Centre, University Medical Centre Utrecht, Utrecht University, the Netherlands; Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Sweden.
| |
Collapse
|
6
|
Baldini G, Phelan KD. The melanocortin pathway and control of appetite-progress and therapeutic implications. J Endocrinol 2019; 241:R1-R33. [PMID: 30812013 PMCID: PMC6500576 DOI: 10.1530/joe-18-0596] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 01/22/2019] [Indexed: 12/19/2022]
Abstract
The initial discovery that ob/ob mice become obese because of a recessive mutation of the leptin gene has been crucial to discover the melanocortin pathway to control appetite. In the melanocortin pathway, the fed state is signaled by abundance of circulating hormones such as leptin and insulin, which bind to receptors expressed at the surface of pro-opiomelanocortin (POMC) neurons to promote processing of POMC to the mature hormone α-melanocyte-stimulating hormone (α-MSH). The α-MSH released by POMC neurons then signals to decrease energy intake by binding to melanocortin-4 receptor (MC4R) expressed by MC4R neurons to the paraventricular nucleus (PVN). Conversely, in the 'starved state' activity of agouti-related neuropeptide (AgRP) and of neuropeptide Y (NPY)-expressing neurons is increased by decreased levels of circulating leptin and insulin and by the orexigenic hormone ghrelin to promote food intake. This initial understanding of the melanocortin pathway has recently been implemented by the description of the complex neuronal circuit that controls the activity of POMC, AgRP/NPY and MC4R neurons and downstream signaling by these neurons. This review summarizes the progress done on the melanocortin pathway and describes how obesity alters this pathway to disrupt energy homeostasis. We also describe progress on how leptin and insulin receptors signal in POMC neurons, how MC4R signals and how altered expression and traffic of MC4R change the acute signaling and desensitization properties of the receptor. We also describe how the discovery of the melanocortin pathway has led to the use of melanocortin agonists to treat obesity derived from genetic disorders.
Collapse
Affiliation(s)
- Giulia Baldini
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kevin D. Phelan
- Department of Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
7
|
Ericson MD, Lensing CJ, Fleming KA, Schlasner KN, Doering SR, Haskell-Luevano C. Bench-top to clinical therapies: A review of melanocortin ligands from 1954 to 2016. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2414-2435. [PMID: 28363699 PMCID: PMC5600687 DOI: 10.1016/j.bbadis.2017.03.020] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/21/2017] [Accepted: 03/27/2017] [Indexed: 10/19/2022]
Abstract
The discovery of the endogenous melanocortin agonists in the 1950s have resulted in sixty years of melanocortin ligand research. Early efforts involved truncations or select modifications of the naturally occurring agonists leading to the development of many potent and selective ligands. With the identification and cloning of the five known melanocortin receptors, many ligands were improved upon through bench-top in vitro assays. Optimization of select properties resulted in ligands adopted as clinical candidates. A summary of every melanocortin ligand is outside the scope of this review. Instead, this review will focus on the following topics: classic melanocortin ligands, selective ligands, small molecule (non-peptide) ligands, ligands with sex-specific effects, bivalent and multivalent ligands, and ligands advanced to clinical trials. Each topic area will be summarized with current references to update the melanocortin field on recent progress. This article is part of a Special Issue entitled: Melanocortin Receptors - edited by Ya-Xiong Tao.
Collapse
Affiliation(s)
- Mark D Ericson
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Cody J Lensing
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Katlyn A Fleming
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Katherine N Schlasner
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Skye R Doering
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | | |
Collapse
|
8
|
Pražienková V, Holubová M, Pelantová H, Bugáňová M, Pirník Z, Mikulášková B, Popelová A, Blechová M, Haluzík M, Železná B, Kuzma M, Kuneš J, Maletínská L. Impact of novel palmitoylated prolactin-releasing peptide analogs on metabolic changes in mice with diet-induced obesity. PLoS One 2017; 12:e0183449. [PMID: 28820912 PMCID: PMC5562305 DOI: 10.1371/journal.pone.0183449] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 08/06/2017] [Indexed: 01/07/2023] Open
Abstract
Analogs of anorexigenic neuropeptides, such as prolactin-releasing peptide (PrRP), have a potential as new anti-obesity drugs. In our previous study, palmitic acid attached to the N-terminus of PrRP enabled its central anorexigenic effects after peripheral administration. In this study, two linkers, γ-glutamic acid at Lys11 and a short, modified polyethylene glycol at the N-terminal Ser and/or Lys11, were applied for the palmitoylation of PrRP31 to improve its bioavailability. These analogs had a high affinity and activation ability to the PrRP receptor GPR10 and the neuropeptide FF2 receptor, as well as short-term anorexigenic effect similar to PrRP palmitoylated at the N-terminus. Two-week treatment with analogs that were palmitoylated through linkers to Lys11 (analogs 1 and 2), but not with analog modified both at the N-terminus and Lys11 (analog 3) decreased body and liver weights, insulin, leptin, triglyceride, cholesterol and free fatty acid plasma levels in a mouse model of diet-induced obesity. Moreover, the expression of uncoupling protein-1 was increased in brown fat suggesting an increase in energy expenditure. In addition, treatment with analogs 1 and 2 but not analog 3 significantly decreased urinary concentrations of 1-methylnicotinamide and its oxidation products N-methyl-2-pyridone-5-carboxamide and N-methyl-4-pyridone-3-carboxamide, as shown by NMR-based metabolomics. This observation confirmed the previously reported increase in nicotinamide derivatives in obesity and type 2 diabetes mellitus and the effectiveness of analogs 1 and 2 in the treatment of these disorders.
Collapse
Affiliation(s)
- Veronika Pražienková
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Martina Holubová
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Helena Pelantová
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Martina Bugáňová
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- Faculty of Chemical Technology, University of Chemistry and Technology Prague, Prague, Czech Republic
| | - Zdenko Pirník
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak Republic
- Department of Human and Clinical Pharmacology, University of Veterinary Medicine, Košice, Slovak Republic
| | - Barbora Mikulášková
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Andrea Popelová
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Miroslava Blechová
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Martin Haluzík
- Institute of Medical Biochemistry and Laboratory Diagnostics, Charles University in Prague and General University Hospital, Prague, Czech Republic
- Centre of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Marek Kuzma
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- * E-mail:
| |
Collapse
|
9
|
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
|
10
|
Secher A, Kelstrup CD, Conde-Frieboes KW, Pyke C, Raun K, Wulff BS, Olsen JV. Analytic framework for peptidomics applied to large-scale neuropeptide identification. Nat Commun 2016; 7:11436. [PMID: 27142507 PMCID: PMC4857386 DOI: 10.1038/ncomms11436] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 03/24/2016] [Indexed: 01/03/2023] Open
Abstract
Large-scale mass spectrometry-based peptidomics for drug discovery is relatively unexplored because of challenges in peptide degradation and identification following tissue extraction. Here we present a streamlined analytical pipeline for large-scale peptidomics. We developed an optimized sample preparation protocol to achieve fast, reproducible and effective extraction of endogenous peptides from sub-dissected organs such as the brain, while diminishing unspecific protease activity. Each peptidome sample was analysed by high-resolution tandem mass spectrometry and the resulting data set was integrated with publically available databases. We developed and applied an algorithm that reduces the peptide complexity for identification of biologically relevant peptides. The developed pipeline was applied to rat hypothalamus and identifies thousands of neuropeptides and their post-translational modifications, which is combined in a resource format for visualization, qualitative and quantitative analyses. Neuropeptide research is challenged by technical difficulties in identifying new bioactive peptides. Here the authors present an analytical pipeline for large-scale peptidomics applied to the rat hypothalamus, identifying thousands of endogenous neuropeptides and their post-translational modifications.
Collapse
Affiliation(s)
- Anna Secher
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark.,Histology and Imaging, Novo Nordisk A/S, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Christian D Kelstrup
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark
| | | | - Charles Pyke
- Histology and Imaging, Novo Nordisk A/S, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Kirsten Raun
- Incretin &Obesity Pharmacology, Novo Nordisk A/S, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Birgitte S Wulff
- Incretin &Obesity Research, Novo Nordisk A/S, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Jesper V Olsen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark
| |
Collapse
|
11
|
Brain signaling systems in the Type 2 diabetes and metabolic syndrome: promising target to treat and prevent these diseases. Future Sci OA 2015; 1:FSO25. [PMID: 28031898 PMCID: PMC5137856 DOI: 10.4155/fso.15.23] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The changes in the brain signaling systems play an important role in etiology and pathogenesis of Type 2 diabetes mellitus (T2DM) and metabolic syndrome (MS), being a possible cause of these diseases. Therefore, their restoration at the early stages of T2DM and MS can be regarded as a promising way to treat and prevent these diseases and their complications. The data on the functional state of the brain signaling systems regulated by insulin, IGF-1, leptin, dopamine, serotonin, melanocortins and glucagon-like peptide-1, in T2DM and MS, are analyzed. The pharmacological approaches to restoration of these systems and improvement of insulin sensitivity, energy expenditure, lipid metabolism, and to prevent diabetic complications are discussed.
Collapse
|
12
|
Møller CL, Pedersen SB, Richelsen B, Conde-Frieboes KW, Raun K, Grove KL, Wulff BS. Melanocortin agonists stimulate lipolysis in human adipose tissue explants but not in adipocytes. BMC Res Notes 2015; 8:559. [PMID: 26459134 PMCID: PMC4604100 DOI: 10.1186/s13104-015-1539-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 10/02/2015] [Indexed: 01/12/2023] Open
Abstract
Background The central melanocortin system is broadly involved in the regulation of mammalian nutrient utilization. However, the function of melanocortin receptors (MCRs) expressed directly in peripheral metabolic tissues is still unclear. The objective of this study was to investigate the lipolytic capacity of MC1-5R in differentiated adipocytes versus intact white adipose tissue. Results Non-selective MCR agonist α-MSH, MC5R-selective agonist PG-901 and MC4R-selective agonist LY2112688 significantly stimulated lipolysis in intact white adipose tissue, whereas stimulation of MCRs in differentiated adipocytes failed to do so. The lipolytic response of MC5R was decreased in intact human white adipose tissue when co-treating with β-adrenergic antagonist propranolol, suggesting that the effect may be dependent on neuronal innervation via noradrenalin release. Conclusion When developing an anti-obesity therapeutic drug with selective MC4R/MC5R properties, effects on lipolysis in white adipose tissue may be physiologically relevant.
Collapse
Affiliation(s)
- Cathrine Laustrup Møller
- Diabetes and Obesity Biology, Novo Nordisk A/S, 2760, Maaloev, Denmark. .,Steno Diabetes Center, Niels Steensensvej 2-4, 2820, Gentofte, Denmark.
| | - Steen B Pedersen
- Department of Endocrinology MEA, Aarhus University Hospital, 8000, Aarhus, Denmark.
| | - Bjørn Richelsen
- Department of Endocrinology MEA, Aarhus University Hospital, 8000, Aarhus, Denmark.
| | | | - Kirsten Raun
- Type 2 Diabetes, Novo Nordisk A/S, 2760, Maaloev, Denmark.
| | - Kevin L Grove
- Diabetes, Obesity and Metabolism, Oregon National Primate Research Centre, Oregon Health & Science University, Portland, OR, 97006, USA. .,Obesity Research, Novo Nordisk A/S, Seattle, WA, 98109, USA.
| | | |
Collapse
|
13
|
Biased signalling: the instinctive skill of the cell in the selection of appropriate signalling pathways. Biochem J 2015; 470:155-67. [DOI: 10.1042/bj20150358] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
GPCRs (G-protein-coupled receptors) are members of a family of proteins which are generally regarded as the largest group of therapeutic drug targets. Ligands of GPCRs do not usually activate all cellular signalling pathways linked to a particular seven-transmembrane receptor in a uniform manner. The fundamental idea behind this concept is that each ligand has its own ability, while interacting with the receptor, to activate different signalling pathways (or a particular set of signalling pathways) and it is this concept which is known as biased signalling. The importance of biased signalling is that it may selectively activate biological responses to favour therapeutically beneficial signalling pathways and to avoid adverse effects. There are two levels of biased signalling. First, bias can arise from the ability of GPCRs to couple to a subset of the available G-protein subtypes: Gαs, Gαq/11, Gαi/o or Gα12/13. These subtypes produce the diverse effects of GPCRs by targeting different effectors. Secondly, biased GPCRs may differentially activate G-proteins or β-arrestins. β-Arrestins are ubiquitously expressed and function to terminate or inhibit classic G-protein signalling and initiate distinct β-arrestin-mediated signalling processes. The interplay of G-protein and β-arrestin signalling largely determines the cellular consequences of the administration of GPCR-targeted drugs. In the present review, we highlight the particular functionalities of biased signalling and discuss its biological effects subsequent to GPCR activation. We consider that biased signalling is potentially allowing a choice between signalling through ‘beneficial’ pathways and the avoidance of ‘harmful’ ones.
Collapse
|
14
|
Sahin T, Vairaprakash P, Borbas KE, Balasubramanian T, Lindsey JS. Hydrophilic bioconjugatable trans-AB-porphyrins and peptide conjugates. J PORPHYR PHTHALOCYA 2015. [DOI: 10.1142/s1088424615500121] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Porphyrins bearing a single bioconjugatable group and a single water-solubilization motif in a trans-AB-architecture (with no other substituents) provide a compact design of value for studies in diverse disciplines. Established synthetic methods have been employed to prepare four new free base porphyrins and one Mn ( III ) chelate. The hydrophilic motif includes 4-N-methylpyridinium, 2,4,6-tris(carboxymethoxy)phenyl, 2,6-bis(phosphonomethoxy)phenyl, and carboxy; the bioconjugatable unit includes carboxy, maleimido, and N-hydroxysuccinimido (NHS) ester. Bioconjugation experiments with a protected porphyrin-diphosphate or unprotected porphyrin-diphosphonate were examined in organic solution or water, respectively. Both approaches were employed to conjugate to the ε-amino group of Lys11 in AcKPV- NH 2, a tripeptide fragment [ Ac -α-MSH(11-13)- NH 2] of melanocyte stimulating hormone, yielding porphyrin-peptide conjugates.
Collapse
Affiliation(s)
- Tuba Sahin
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695-8204, USA
| | | | - K. Eszter Borbas
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695-8204, USA
| | | | - Jonathan S. Lindsey
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695-8204, USA
| |
Collapse
|
15
|
Ghamari-Langroudi M, Digby GJ, Sebag JA, Millhauser GL, Palomino R, Matthews R, Gillyard T, Panaro BL, Tough IR, Cox HM, Denton JS, Cone RD. G-protein-independent coupling of MC4R to Kir7.1 in hypothalamic neurons. Nature 2015; 520:94-8. [PMID: 25600267 PMCID: PMC4383680 DOI: 10.1038/nature14051] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 11/10/2014] [Indexed: 02/04/2023]
Abstract
The regulated release of anorexigenic α-melanocyte stimulating hormone (α-MSH) and orexigenic Agouti-related protein (AgRP) from discrete hypothalamic arcuate neurons onto common target sites in the central nervous system has a fundamental role in the regulation of energy homeostasis. Both peptides bind with high affinity to the melanocortin-4 receptor (MC4R); existing data show that α-MSH is an agonist that couples the receptor to the Gαs signalling pathway, while AgRP binds competitively to block α-MSH binding and blocks the constitutive activity mediated by the ligand-mimetic amino-terminal domain of the receptor. Here we show that, in mice, regulation of firing activity of neurons from the paraventricular nucleus of the hypothalamus (PVN) by α-MSH and AgRP can be mediated independently of Gαs signalling by ligand-induced coupling of MC4R to closure of inwardly rectifying potassium channel, Kir7.1. Furthermore, AgRP is a biased agonist that hyperpolarizes neurons by binding to MC4R and opening Kir7.1, independently of its inhibition of α-MSH binding. Consequently, Kir7.1 signalling appears to be central to melanocortin-mediated regulation of energy homeostasis within the PVN. Coupling of MC4R to Kir7.1 may explain unusual aspects of the control of energy homeostasis by melanocortin signalling, including the gene dosage effect of MC4R and the sustained effects of AgRP on food intake.
Collapse
Affiliation(s)
- Masoud Ghamari-Langroudi
- Department of Molecular Physiology &Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Gregory J Digby
- Department of Molecular Physiology &Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Julien A Sebag
- Department of Molecular Physiology &Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Glenn L Millhauser
- Department of Chemistry &Biochemistry, University of California, Santa Cruz, California 95064, USA
| | - Rafael Palomino
- Department of Chemistry &Biochemistry, University of California, Santa Cruz, California 95064, USA
| | - Robert Matthews
- Department of Molecular Physiology &Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Taneisha Gillyard
- 1] Department of Molecular Physiology &Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA [2] Department of Pharmacology, Meharry Medical College, Nashville, Tennessee 37208, USA
| | - Brandon L Panaro
- Department of Molecular Physiology &Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Iain R Tough
- King's College London, Wolfson Centre for Age-Related Diseases, Guy's Campus, London SE1 1UL, UK
| | - Helen M Cox
- King's College London, Wolfson Centre for Age-Related Diseases, Guy's Campus, London SE1 1UL, UK
| | - Jerod S Denton
- 1] Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA [2] Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Roger D Cone
- Department of Molecular Physiology &Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| |
Collapse
|
16
|
Melanocortin-4 receptor modulators for the treatment of obesity: a patent analysis (2008–2014). Pharm Pat Anal 2015; 4:95-107. [DOI: 10.4155/ppa.15.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The central melanocortin system and particularly the melanocortin-4 receptor (MC4R) subtype, plays an important role in the regulation of body weight. The discovery of orally active MC4R agonists suitable for evaluation in human clinical trials as weight loss agents has attracted considerable interest over the past decade, but has proved challenging, in part because of cardiovascular and behavioral side effects. Currently, the only MC4R agonist in clinical trials is a peptide identified as RM-493. To avoid some of the undesirable side effects associated with MC4R activation, new pharmacological approaches for modulating the MC system have been investigated. In this article, we provide a review of the MC4R patent landscape from 2008 to 2014 and analyze the physicochemical properties of compounds described herein.
Collapse
|
17
|
Lute B, Jou W, Lateef DM, Goldgof M, Xiao C, Piñol RA, Kravitz AV, Miller NR, Huang YG, Girardet C, Butler AA, Gavrilova O, Reitman ML. Biphasic effect of melanocortin agonists on metabolic rate and body temperature. Cell Metab 2014; 20:333-45. [PMID: 24981835 PMCID: PMC4126889 DOI: 10.1016/j.cmet.2014.05.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/03/2014] [Accepted: 05/22/2014] [Indexed: 11/30/2022]
Abstract
The melanocortin system regulates metabolic homeostasis and inflammation. Melanocortin agonists have contradictorily been reported to both increase and decrease metabolic rate and body temperature. We find two distinct physiologic responses occurring at similar doses. Intraperitoneal administration of the nonselective melanocortin agonist MTII causes a melanocortin-4 receptor (Mc4r)-mediated hypermetabolism/hyperthermia. This is preceded by a profound, transient hypometabolism/hypothermia that is preserved in mice lacking any one of Mc1r, Mc3r, Mc4r, or Mc5r. Three other melanocortin agonists also caused hypothermia, which is actively achieved via seeking a cool environment, vasodilation, and inhibition of brown adipose tissue thermogenesis. These results suggest that the hypometabolic/hypothermic effect of MTII is not due to a failure of thermoregulation. The hypometabolism/hypothermia was prevented by dopamine antagonists, and MTII selectively activated arcuate nucleus dopaminergic neurons, suggesting that these neurons may contribute to the hypometabolism/hypothermia. We propose that the hypometabolism/hypothermia is a regulated response, potentially beneficial during extreme physiologic stress.
Collapse
Affiliation(s)
- Beth Lute
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - William Jou
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Dalya M Lateef
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Margalit Goldgof
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Cuiying Xiao
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Ramón A Piñol
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Alexxai V Kravitz
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Nicole R Miller
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Yuning George Huang
- Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Clemence Girardet
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Andrew A Butler
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Marc L Reitman
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
18
|
Yi X, Kabanov AV. Brain delivery of proteins via their fatty acid and block copolymer modifications. J Drug Target 2014; 21:940-55. [PMID: 24160902 DOI: 10.3109/1061186x.2013.847098] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
It is well known that hydrophobic small molecules penetrate cell membranes better than hydrophilic molecules. Amphiphilic molecules that dissolve both in lipid and aqueous phases are best suited for membrane transport. Transport of biomacromolecules across physiological barriers, e.g. the blood-brain barrier, is greatly complicated by the unique structure and function of such barriers. Two decades ago we adopted a simple philosophy that to increase protein delivery to the brain one needs to modify this protein with hydrophobic moieties. With this general idea we began modifying proteins (antibodies, enzymes, hormones, etc.) with either hydrophobic fatty acid residues or amphiphilic block copolymer moieties, such as poy(ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide) (pluronics or poloxamers) and more recently, poly(2-oxasolines). This simple approach has resulted in impressive successes in CNS drug delivery. We present a retrospective overview of these works initiated in the Soviet Union in 1980s, and then continued in the United States and other countries. Notably some of the early findings were later corroborated by brain pharmacokinetic data. Industrial development of several drug candidates employing these strategies has followed. Overall modification by hydrophobic fatty acids residues or amphiphilic block copolymers represents a promising and relatively safe strategy to deliver proteins to the brain.
Collapse
Affiliation(s)
- Xiang Yi
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA and
| | | |
Collapse
|
19
|
Royalty JE, Konradsen G, Eskerod O, Wulff BS, Hansen BS. Investigation of safety, tolerability, pharmacokinetics, and pharmacodynamics of single and multiple doses of a long-acting α-MSH analog in healthy overweight and obese subjects. J Clin Pharmacol 2014; 54:394-404. [PMID: 24166760 PMCID: PMC4263154 DOI: 10.1002/jcph.211] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 10/17/2013] [Indexed: 11/07/2022]
Abstract
MC4-NN2-0453 is a novel, long-acting, selective, melanocortin-4-receptor agonist developed for treatment of obesity. This first-human-dose, randomized, double-blind, placebo-controlled trial investigated the safety, tolerability, pharmacokinetics, and pharmacodynamics of single and multiple doses of MC4-NN2-0453 in overweight to obese but otherwise healthy subjects. The trial included a single-dose part of ascending subcutaneous 0.03-1.50 mg/kg doses in overweight to obese but otherwise healthy men, and a multiple-dose part of ascending subcutaneous 0.75-3.0 mg/day doses in obese but otherwise healthy men/women. The single-dose part included 7 cohorts of 8 subjects, randomized 6:2 to active drug/placebo; the multiple-dose part included 4 cohorts of 20 subjects, randomized 16:4 to active drug/placebo. MC4-NN2-0453 was well tolerated and raised no safety concerns except for nonserious skin-related adverse events, this along with lack of weight loss effect led to premature termination of the trial. Headache, sexual-arousal disturbance, and penile erection were also reported. Single-dose pharmacokinetics showed dose-linearity and dose-proportionality. Maximum plasma concentration was observed after 50-100 hours, which then declined with a of approximately 250 hours. Plasma concentration reached steady state after 4 weeks for 0.75 and 1.5 mg/day multiple-dose cohorts, and the was similar to single dose. There were no significant pharmacodynamic effects, including effect on body weight.
Collapse
|
20
|
Fosgerau K, Raun K, Nilsson C, Dahl K, Wulff BS. Novel α-MSH analog causes weight loss in obese rats and minipigs and improves insulin sensitivity. J Endocrinol 2014; 220:97-107. [PMID: 24204009 PMCID: PMC3888513 DOI: 10.1530/joe-13-0284] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Obesity is a major burden to people and to health care systems around the world. The aim of the study was to characterize the effect of a novel selective α-MSH analog on obesity and insulin sensitivity. The subchronic effects of the selective MC4-R peptide agonist MC4-NN1-0182 were investigated in diet-induced obese (DIO) rats and DIO minipigs by assessing the effects on food intake, energy consumption, and body weight. The acute effect of MC4-NN1-0182 on insulin sensitivity was assessed by a euglycemic-hyperinsulinemic clamp study in normal rats. Three weeks of treatment of DIO rats with MC4-NN1-0182 caused a decrease in food intake and a significant decrease in body weight 7±1%, P<0.05 compared with 3±1% increase with the vehicle control. In DIO minipigs, 8 weeks of treatment with MC4-NN1-0182 resulted in a body weight loss of 13.3±2.5 kg (13±3%), whereas the vehicle control group had gained 3.7±1.4 kg (4±1%). Finally, clamp studies in normal rats showed that acute treatment with MC4-NN1-0182 caused a significant increase in glucose disposal (Rd) compared with vehicle control (Rd, mg/kg per min, 17.0±0.7 vs 13.9±0.6, P<0.01). We demonstrate that treatment of DIO rats or minipigs with a selective MC4-R peptide agonist causes weight loss. Moreover, we have demonstrated weight-independent effects on insulin sensitivity. Our observations identify MC4 agonism as a viable target for the treatment of obesity and insulin resistance.
Collapse
Affiliation(s)
- Keld Fosgerau
- Novo Nordisk Diabetes Research Unit, Novo Nordisk A/S, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | | | | | | | | |
Collapse
|
21
|
Abstract
G-protein–coupled receptors (GPCRs) still offer enormous scope for new therapeutic targets. Currently marketed agents are dominated by those with activity at aminergic receptors and yet they account for only ~10% of the family. Progress up until now with other subfamilies, notably orphans, Family A/peptide, Family A/lipid, Family B, Family C, and Family F, has been, at best, patchy. This may be attributable to the heterogeneous nature of GPCRs, their endogenous ligands, and consequently their binding sites. Our appreciation of receptor similarity has arguably been too simplistic, and screening collections have not necessarily been well suited to identifying leads in new areas. Despite the relative shortage of high-quality tool molecules in a number of cases, there is an emerging, and increasingly substantial, body of evidence associating many as yet “undrugged” receptors with a very wide range of diseases. Significant advances in our understanding of receptor pharmacology and technical advances in screening, protein X-ray crystallography, and ligand design methods are paving the way for new successes in the area. Exploitation of allosteric mechanisms; alternative signaling pathways such as G12/13, Gβγ, and β-arrestin; the discovery of “biased” ligands; and the emergence of GPCR-protein complexes as potential drug targets offer scope for new and much improved drugs.
Collapse
|
22
|
Eliasen R, Daly NL, Wulff BS, Andresen TL, Conde-Frieboes KW, Craik DJ. Design, synthesis, structural and functional characterization of novel melanocortin agonists based on the cyclotide kalata B1. J Biol Chem 2012; 287:40493-501. [PMID: 23012369 DOI: 10.1074/jbc.m112.395442] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Cyclotides are useful scaffolds to stabilize bioactive peptides. RESULTS Four melanocortin analogues of kalata B1 were synthesized. One is a selective MC4R agonist. CONCLUSION The analogues retain the native kalata B1 scaffold and introduce a designed pharmacological activity, validating cyclotides as protein engineering scaffolds. SIGNIFICANCE A novel type of melanocortin agonist has been developed, with potential as a drug lead for treating obesity. Obesity is an increasingly important global health problem that lacks current treatment options. The melanocortin receptor 4 (MC4R) is a target for obesity therapies because its activation triggers appetite suppression and increases energy expenditure. Cyclotides have been suggested as scaffolds for the insertion and stabilization of pharmaceutically active peptides. In this study, we explored the development of appetite-reducing peptides by synthesizing MC4R agonists based on the insertion of the His-Phe-Arg-Trp sequence into the cyclotide kalata B1. The ability of the analogues to fold similarly to kalata B1 but display MC4R activity were investigated. Four peptides were synthesized using t-butoxycarbonyl peptide chemistry with a C-terminal thioester to facilitate backbone cyclization. The structures of the peptides were found to be similar to kalata B1, evaluated by Hα NMR chemical shifts. KB1(GHFRWG;23-28) had a K(i) of 29 nm at the MC4R and was 107 or 314 times more selective over this receptor than MC1R or MC5R, respectively, and had no detectable binding to MC3R. The peptide had higher affinity for the MC4R than the endogenous agonist, α-melanocyte stimulation hormone, but it was less potent at the MC4R, with an EC(50) of 580 nm for activation of the MC4R. In conclusion, we synthesized melanocortin analogues of kalata B1 that preserve the structural scaffold and display receptor binding and functional activity. KB1(GHFRWG;23-28) is potent and selective for the MC4R. This compound validates the use of cyclotides as scaffolds and has the potential to be a new lead for the treatment of obesity.
Collapse
|