1
|
Rahim M, Bednarski TK, Hasenour CM, Banerjee DR, Trenary I, Young JD. Simultaneous in vivo multi-organ fluxomics reveals divergent metabolic adaptations in liver, heart, and skeletal muscle during obesity. Cell Rep 2025; 44:115591. [PMID: 40244853 DOI: 10.1016/j.celrep.2025.115591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 02/23/2025] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
We present an isotope-based metabolic flux analysis (MFA) approach to simultaneously quantify metabolic fluxes in the liver, heart, and skeletal muscle of individual mice. The platform was scaled to examine metabolic flux adaptations in age-matched cohorts of mice exhibiting varying levels of chronic obesity. We found that severe obesity increases hepatic gluconeogenesis and citric acid cycle flux, accompanied by elevated glucose oxidation in the heart that compensates for impaired fatty acid oxidation. In contrast, skeletal muscle fluxes exhibit an overall reduction in substrate oxidation. These findings demonstrate the dichotomy in fuel utilization between cardiac and skeletal muscle during worsening metabolic disease and demonstrate the divergent effects of obesity on metabolic fluxes in different organs. This multi-tissue MFA technology can be extended to address important questions about in vivo regulation of metabolism and its dysregulation in disease, which cannot be fully answered through studies of single organs or isolated cells/tissues.
Collapse
Affiliation(s)
- Mohsin Rahim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Tomasz K Bednarski
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Clinton M Hasenour
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Deveena R Banerjee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Irina Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
2
|
Patel TP, Jun JY, Seo AY, Levi NJ, Elizondo DM, Chen J, Wong AM, Tugarinov N, Altman EK, Gehle DB, Jung SM, Patel P, Ericson M, Haskell-Luevano C, Demby TC, Cougnoux A, Wolska A, Yanovski JA. Melanocortin 3 receptor regulates hepatic autophagy and systemic adiposity. Nat Commun 2025; 16:1690. [PMID: 39956805 PMCID: PMC11830824 DOI: 10.1038/s41467-025-56936-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/05/2025] [Indexed: 02/18/2025] Open
Abstract
Systemic lipid homeostasis requires hepatic autophagy, a major cellular program for intracellular fat recycling. Here, we find melanocortin 3 receptor (MC3R) regulates hepatic autophagy in addition to its previously established CNS role in systemic energy partitioning and puberty. Mice with Mc3r deficiency develop obesity with hepatic triglyceride accumulation and disrupted hepatocellular autophagosome turnover. Mice with partially inactive human MC3R due to obesogenic variants demonstrate similar hepatic autophagic dysfunction. In vitro and in vivo activation of hepatic MC3R upregulates autophagy through LC3II activation, TFEB cytoplasmic-to-nuclear translocation, and subsequent downstream gene activation. MC3R-deficient hepatocytes had blunted autophagosome-lysosome docking and lipid droplet clearance. Finally, the liver-specific rescue of Mc3r was sufficient to restore hepatocellular autophagy, improve hepatocyte mitochondrial function and systemic energy expenditures, reduce adipose tissue lipid accumulation, and partially restore body weight in both male and female mice. We thus report a role for MC3R in regulating hepatic autophagy and systemic adiposity.
Collapse
Affiliation(s)
- Tushar P Patel
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Joo Yun Jun
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Arnold Y Seo
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Noah J Levi
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Diana M Elizondo
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Jocelyn Chen
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Adrian M Wong
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Nicol Tugarinov
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Elizabeth K Altman
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Daniel B Gehle
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Sun Min Jung
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Pooja Patel
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Mark Ericson
- Department of Medicinal Chemistry, University of Minnesota College of Pharmacy, Minneapolis, MN, USA
| | - Carrie Haskell-Luevano
- Department of Medicinal Chemistry, University of Minnesota College of Pharmacy, Minneapolis, MN, USA
| | - Tamar C Demby
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Antony Cougnoux
- Section on Molecular Dysmorphology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Anna Wolska
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD, USA
| | - Jack A Yanovski
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA.
| |
Collapse
|
3
|
Jamaluddin A, Wyatt RA, Lee J, Dowsett GK, Tadross JA, Broichhagen J, Yeo GS, Levitz J, Gorvin CM. The MRAP2 accessory protein directly interacts with melanocortin-3 receptor to enhance signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.06.622243. [PMID: 39574659 PMCID: PMC11580913 DOI: 10.1101/2024.11.06.622243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
The central melanocortin system links nutrition to energy expenditure, with melanocortin-4 receptor (MC4R) controlling appetite and food intake, and MC3R regulating timing of sexual maturation, rate of linear growth and lean mass accumulation. Melanocortin-2 receptor accessory protein-2 (MRAP2) is a single transmembrane protein that interacts with MC4R to potentiate it's signalling, and human mutations in MRAP2 cause obesity. Previous studies have been unable to consistently show whether MRAP2 affects MC3R activity. Here we used single-molecule pull-down (SiMPull) to confirm that MC3R and MRAP2 interact in HEK293 cells. Analysis of fluorescent photobleaching steps showed that MC3R and MRAP2 readily form heterodimers most commonly with a 1:1 stoichiometry. Human single-nucleus and spatial transcriptomics show MRAP2 is co-expressed with MC3R in hypothalamic neurons with important roles in energy homeostasis and appetite control. Functional analyses showed MRAP2 enhances MC3R cAMP signalling, impairs β-arrestin recruitment, and reduces internalization in HEK293 cells. Structural homology models revealed putative interactions between the two proteins and alanine mutagenesis of five MRAP2 and three MC3R transmembrane residues significantly reduced MRAP2 effects on MC3R signalling. Finally, we showed genetic variants in MRAP2 that have been identified in individuals that are overweight or obese prevent MRAP2's enhancement of MC3R-driven signalling. Thus, these studies reveal MRAP2 as an important regulator of MC3R function and provide further evidence for the crucial role of MRAP2 in energy homeostasis.
Collapse
Affiliation(s)
- Aqfan Jamaluddin
- Department of Metabolism and Systems Science, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - Rachael A. Wyatt
- Department of Metabolism and Systems Science, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - Joon Lee
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Georgina K.C. Dowsett
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - John A. Tadross
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
- East Genomics Laboratory Hub, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Johannes Broichhagen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany
| | - Giles S.H. Yeo
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Caroline M. Gorvin
- Department of Metabolism and Systems Science, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| |
Collapse
|
4
|
Stark R. The olfactory bulb: A neuroendocrine spotlight on feeding and metabolism. J Neuroendocrinol 2024; 36:e13382. [PMID: 38468186 DOI: 10.1111/jne.13382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 03/13/2024]
Abstract
Olfaction is the most ancient sense and is needed for food-seeking, danger protection, mating and survival. It is often the first sensory modality to perceive changes in the external environment, before sight, taste or sound. Odour molecules activate olfactory sensory neurons that reside on the olfactory epithelium in the nasal cavity, which transmits this odour-specific information to the olfactory bulb (OB), where it is relayed to higher brain regions involved in olfactory perception and behaviour. Besides odour processing, recent studies suggest that the OB extends its function into the regulation of food intake and energy balance. Furthermore, numerous hormone receptors associated with appetite and metabolism are expressed within the OB, suggesting a neuroendocrine role outside the hypothalamus. Olfactory cues are important to promote food preparatory behaviours and consumption, such as enhancing appetite and salivation. In addition, altered metabolism or energy state (fasting, satiety and overnutrition) can change olfactory processing and perception. Similarly, various animal models and human pathologies indicate a strong link between olfactory impairment and metabolic dysfunction. Therefore, understanding the nature of this reciprocal relationship is critical to understand how olfactory or metabolic disorders arise. This present review elaborates on the connection between olfaction, feeding behaviour and metabolism and will shed light on the neuroendocrine role of the OB as an interface between the external and internal environments. Elucidating the specific mechanisms by which olfactory signals are integrated and translated into metabolic responses holds promise for the development of targeted therapeutic strategies and interventions aimed at modulating appetite and promoting metabolic health.
Collapse
Affiliation(s)
- Romana Stark
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
5
|
Gunraj RE, Yang C, Liu L, Larochelle J, Candelario-Jalil E. Protective roles of adropin in neurological disease. Am J Physiol Cell Physiol 2023; 324:C674-C678. [PMID: 36717106 PMCID: PMC10027081 DOI: 10.1152/ajpcell.00318.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 02/01/2023]
Abstract
Adropin is a highly conserved secreted peptide encoded by the Energy Homeostasis Associated gene (Enho). It is expressed in many tissues throughout the body, including the liver and brain, and plays a crucial role in maintaining lipid homeostasis and regulating insulin sensitivity. Adropin also participates in several other pathophysiological processes of multiple central nervous system (CNS) diseases. There is strong evidence of the protective effects of adropin in stroke, heart disease, aging, and other diseases. The peptide has been shown to reduce the risk of disease, attenuate histological alterations, and reduce cognitive decline associated with neurological disorders. Recent findings support its critical role in regulating endothelial cells and maintaining blood-brain barrier integrity through an endothelial nitric oxide synthase (eNOS)-dependent mechanism. Here we discuss current evidence of the protective effects of adropin in CNS diseases specifically involving the cerebrovasculature and highlight potential mechanisms through which the peptide exhibits these effects.
Collapse
Affiliation(s)
- Rachel E Gunraj
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, United States
| | - Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, United States
| | - Lei Liu
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, United States
| | - Jonathan Larochelle
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, United States
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
6
|
Ai ZL, Zhang X, Ge W, Zhong YB, Wang HY, Zuo ZY, Liu DY. Salvia miltiorrhiza extract may exert an anti-obesity effect in rats with high-fat diet-induced obesity by modulating gut microbiome and lipid metabolism. World J Gastroenterol 2022; 28:6131-6156. [PMID: 36483153 PMCID: PMC9724488 DOI: 10.3748/wjg.v28.i43.6131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/21/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Studies have shown that a high-fat diet (HFD) can alter gut microbiota (GM) homeostasis and participate in lipid metabolism disorders associated with obesity. Therefore, regulating the construction of GM with the balance of lipid metabolism has become essential for treating obesity. Salvia miltiorrhiza extract (Sal), a common traditional Chinese medicine, has been proven effective against atherosclerosis, hyperlipidemia, obesity, and other dyslipidemia-related diseases. AIM To investigate the anti-obesity effects of Sal in rats with HFD-induced obesity, and explore the underlying mechanism by focusing on GM and lipid metabolism. METHODS Obesity was induced in rats with an HFD for 7 wk, and Sal (0.675 g/1.35 g/2.70 g/kg/d) was administered to treat obese rats for 8 wk. The therapeutic effect was evaluated by body weight, body fat index, waistline, and serum lipid level. Lipid factors (cAMP, PKA, and HSL) in liver and fat homogenates were analyzed by ELISA. The effect of Sal on GM and lipid metabolism was assessed by 16S rRNA-based microbiota analysis and untargeted lipidomic analysis (LC-MS/MS), respectively. RESULTS Sal treatment markedly reduced weight, body fat index, serum triglycerides (TG), total cholesterol (TC), low-density lipoprotein, glucose, free fatty acid, hepatic lipid accumulation, and adipocyte vacuolation, and increased serum high-density lipoprotein (HDL-C) in rats with HFD-induced obesity. These effects were associated with increased concentrations of lipid factors such as cAMP, PKA, and HSL in the liver and adipose tissues, enhanced gut integrity, and improved lipid metabolism. GM analysis revealed that Sal could reverse HFD-induced dysbacteriosis by promoting the abundance of Actinobacteriota and Proteobacteria, and decreasing the growth of Firmicutes and Desulfobacterita. Furthermore, LC-MS/MS analysis indicated that Sal decreased TGs (TG18:2/18:2/20:4, TG16:0/18:2/22:6), DGs (DG14:0/22:6, DG22:6/22:6), CL (18:2/ 18:1/18:1/20:0), and increased ceramides (Cers; Cer d16:0/21:0, Cer d16:1/24:1), (O-acyl)-ω-hydroxy fatty acids (OAHFAs; OAHFA18:0/14:0) in the feces of rats. Spearman's correlation analysis further indicated that TGs, DGs, and CL were negatively related to the abundance of Facklamia and Dubosiella, and positively correlated with Blautia and Quinella, while OAHFAs and Cers were the opposite. CONCLUSION Sal has an anti-obesity effect by regulating the GM and lipid metabolism.
Collapse
Affiliation(s)
- Zi-Li Ai
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Xian Zhang
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Wei Ge
- Department of Proctology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi Province, China
| | - You-Bao Zhong
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Hai-Yan Wang
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Zheng-Yun Zuo
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Duan-Yong Liu
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| |
Collapse
|
7
|
Bedenbaugh MN, Brener SC, Maldonado J, Lippert RN, Sweeney P, Cone RD, Simerly RB. Organization of neural systems expressing melanocortin-3 receptors in the mouse brain: Evidence for sexual dimorphism. J Comp Neurol 2022; 530:2835-2851. [PMID: 35770983 PMCID: PMC9724692 DOI: 10.1002/cne.25379] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/24/2022] [Accepted: 05/27/2022] [Indexed: 01/13/2023]
Abstract
The central melanocortin system is fundamentally important for controlling food intake and energy homeostasis. Melanocortin-3 receptor (MC3R) is one of two major receptors of the melanocortin system found in the brain. In contrast to the well-characterized melanocortin-4 receptor (MC4R), little is known regarding the organization of MC3R-expressing neural circuits. To increase our understanding of the intrinsic organization of MC3R neural circuits, identify specific differences between males and females, and gain a neural systems level perspective of this circuitry, we conducted a brain-wide mapping of neurons labeled for MC3R and characterized the distribution of their projections. Analysis revealed MC3R neuronal and terminal labeling in multiple brain regions that control a diverse range of physiological functions and behavioral processes. Notably, dense labeling was observed in the hypothalamus, as well as areas that share considerable connections with the hypothalamus, including the cortex, amygdala, thalamus, and brainstem. Additionally, MC3R neuronal labeling was sexually dimorphic in several areas, including the anteroventral periventricular area, arcuate nucleus, principal nucleus of the bed nucleus of the stria terminalis, and ventral premammillary region. Altogether, anatomical evidence reported here suggests that MC3R has the potential to influence several different classes of motivated behavior that are essential for survival, including ingestive, reproductive, defensive, and arousal behaviors, and is likely to modulate these behaviors differently in males and females.
Collapse
Affiliation(s)
- Michelle N. Bedenbaugh
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Samantha C. Brener
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Jose Maldonado
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Rachel N. Lippert
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition Potsdam-Rehbruecke, Potsdam, Germany
| | - Patrick Sweeney
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
- Department of Molecular and Integrative Physiology, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Roger D. Cone
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
- Department of Molecular and Integrative Physiology, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Richard B. Simerly
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
8
|
Phung HH, Lee CH. Mouse models of nonalcoholic steatohepatitis and their application to new drug development. Arch Pharm Res 2022; 45:761-794. [DOI: 10.1007/s12272-022-01410-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022]
|
9
|
Melanocortin-5 Receptor: Pharmacology and Its Regulation of Energy Metabolism. Int J Mol Sci 2022; 23:ijms23158727. [PMID: 35955857 PMCID: PMC9369360 DOI: 10.3390/ijms23158727] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 12/12/2022] Open
Abstract
As the most recent melanocortin receptor (MCR) identified, melanocortin-5 receptor (MC5R) has unique tissue expression patterns, pharmacological properties, and physiological functions. Different from the other four MCR subtypes, MC5R is widely distributed in both the central nervous system and peripheral tissues and is associated with multiple functions. MC5R in sebaceous and preputial glands regulates lipid production and sexual behavior, respectively. MC5R expressed in immune cells is involved in immunomodulation. Among the five MCRs, MC5R is the predominant subtype expressed in skeletal muscle and white adipose tissue, tissues critical for energy metabolism. Activated MC5R triggers lipid mobilization in adipocytes and glucose uptake in skeletal muscle. Therefore, MC5R is a potential target for treating patients with obesity and diabetes mellitus. Melanocortin-2 receptor accessory proteins can modulate the cell surface expression, dimerization, and pharmacology of MC5R. This minireview summarizes the molecular and pharmacological properties of MC5R and highlights the progress made on MC5R in energy metabolism. We poInt. out knowledge gaps that need to be explored in the future.
Collapse
|
10
|
Mouat MA, Wilkins BP, Ding E, Govindaraju H, Coleman JLJ, Graham RM, Turner N, Smith NJ. Metabolic Profiling of Mice with Deletion of the Orphan G Protein-Coupled Receptor, GPR37L1. Cells 2022; 11:cells11111814. [PMID: 35681509 PMCID: PMC9180194 DOI: 10.3390/cells11111814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/18/2022] [Accepted: 05/30/2022] [Indexed: 02/01/2023] Open
Abstract
Understanding the neurogenic causes of obesity may reveal novel drug targets to counter the obesity crisis and associated sequelae. Here, we investigate whether the deletion of GPR37L1, an astrocyte-specific orphan G protein-coupled receptor, affects whole-body energy homeostasis in mice. We subjected male Gpr37l1−/− mice and littermate wildtype (Gpr37l1+/+, C57BL/6J background) controls to either 12 weeks of high-fat diet (HFD) or chow feeding, or to 1 year of chow diet, with body composition quantified by EchoMRI, glucose handling by glucose tolerance test and metabolic rate by indirect calorimetry. Following an HFD, Gpr37l1−/− mice had similar glucose handling, body weight and fat mass compared with wildtype controls. Interestingly, we observed a significantly elevated respiratory exchange ratio in HFD- and chow-fed Gpr37l1−/− mice during daylight hours. After 1 year of chow feeding, we again saw no differences in glucose and insulin tolerance or body weight between genotypes, nor in energy expenditure or respiratory exchange ratio. However, there was significantly lower fat mass accumulation, and higher ambulatory activity in the Gpr37l1−/− mice during night hours. Overall, these results indicate that while GPR37L1 may play a minor role in whole-body metabolism, it is not a viable clinical target for the treatment of obesity.
Collapse
Affiliation(s)
- Margaret A. Mouat
- Orphan Receptor Laboratory, School of Medical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia; (M.A.M.); (B.P.W.)
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (J.L.J.C.); (R.M.G.)
| | - Brendan P. Wilkins
- Orphan Receptor Laboratory, School of Medical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia; (M.A.M.); (B.P.W.)
| | - Eileen Ding
- Mitochondrial Bioenergetics Laboratory, School of Medical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia; (E.D.); (H.G.)
| | - Hemna Govindaraju
- Mitochondrial Bioenergetics Laboratory, School of Medical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia; (E.D.); (H.G.)
| | - James L. J. Coleman
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (J.L.J.C.); (R.M.G.)
| | - Robert M. Graham
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (J.L.J.C.); (R.M.G.)
| | - Nigel Turner
- Mitochondrial Bioenergetics Laboratory, School of Medical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia; (E.D.); (H.G.)
- Cellular Bioenergetics Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- Correspondence: (N.T.); (N.J.S.)
| | - Nicola J. Smith
- Orphan Receptor Laboratory, School of Medical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia; (M.A.M.); (B.P.W.)
- Correspondence: (N.T.); (N.J.S.)
| |
Collapse
|
11
|
Powell DR, Revelli JP, Doree DD, DaCosta CM, Desai U, Shadoan MK, Rodriguez L, Mullens M, Yang QM, Ding ZM, Kirkpatrick LL, Vogel P, Zambrowicz B, Sands AT, Platt KA, Hansen GM, Brommage R. High-Throughput Screening of Mouse Gene Knockouts Identifies Established and Novel High Body Fat Phenotypes. Diabetes Metab Syndr Obes 2021; 14:3753-3785. [PMID: 34483672 PMCID: PMC8409770 DOI: 10.2147/dmso.s322083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/04/2021] [Indexed: 01/05/2023] Open
Abstract
PURPOSE Obesity is a major public health problem. Understanding which genes contribute to obesity may better predict individual risk and allow development of new therapies. Because obesity of a mouse gene knockout (KO) line predicts an association of the orthologous human gene with obesity, we reviewed data from the Lexicon Genome5000TM high throughput phenotypic screen (HTS) of mouse gene KOs to identify KO lines with high body fat. MATERIALS AND METHODS KO lines were generated using homologous recombination or gene trapping technologies. HTS body composition analyses were performed on adult wild-type and homozygous KO littermate mice from 3758 druggable mouse genes having a human ortholog. Body composition was measured by either DXA or QMR on chow-fed cohorts from all 3758 KO lines and was measured by QMR on independent high fat diet-fed cohorts from 2488 of these KO lines. Where possible, comparisons were made to HTS data from the International Mouse Phenotyping Consortium (IMPC). RESULTS Body fat data are presented for 75 KO lines. Of 46 KO lines where independent external published and/or IMPC KO lines are reported as obese, 43 had increased body fat. For the remaining 29 novel high body fat KO lines, Ksr2 and G2e3 are supported by data from additional independent KO cohorts, 6 (Asnsd1, Srpk2, Dpp8, Cxxc4, Tenm3 and Kiss1) are supported by data from additional internal cohorts, and the remaining 21 including Tle4, Ak5, Ntm, Tusc3, Ankk1, Mfap3l, Prok2 and Prokr2 were studied with HTS cohorts only. CONCLUSION These data support the finding of high body fat in 43 independent external published and/or IMPC KO lines. A novel obese phenotype was identified in 29 additional KO lines, with 27 still lacking the external confirmation now provided for Ksr2 and G2e3 KO mice. Undoubtedly, many mammalian obesity genes remain to be identified and characterized.
Collapse
Affiliation(s)
- David R Powell
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Jean-Pierre Revelli
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Deon D Doree
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Christopher M DaCosta
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Urvi Desai
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Melanie K Shadoan
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Lawrence Rodriguez
- Department of Information Technology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Michael Mullens
- Department of Information Technology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Qi M Yang
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Zhi-Ming Ding
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Laura L Kirkpatrick
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Peter Vogel
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Brian Zambrowicz
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
- Department of Information Technology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Arthur T Sands
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
- Department of Information Technology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Kenneth A Platt
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Gwenn M Hansen
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Robert Brommage
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| |
Collapse
|
12
|
Cázarez‐Márquez F, Eliveld J, Ritsema WIGR, Foppen E, Bossenbroek Y, Pelizzari S, Simonneaux V, Kalsbeek A. Role of central kisspeptin and RFRP-3 in energy metabolism in the male Wistar rat. J Neuroendocrinol 2021; 33:e12973. [PMID: 33960524 PMCID: PMC8365661 DOI: 10.1111/jne.12973] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/12/2021] [Accepted: 03/24/2021] [Indexed: 02/01/2023]
Abstract
Kisspeptin (Kp) and (Arg)(Phe) related peptide 3 (RFRP-3) are two RF-amides acting in the hypothalamus to control reproduction. In the past 10 years, it has become clear that, apart from their role in reproductive physiology, both neuropeptides are also involved in the control of food intake, as well as glucose and energy metabolism. To investigate further the neural mechanisms responsible for these metabolic actions, we assessed the effect of acute i.c.v. administration of Kp or RFRP-3 in ad lib. fed male Wistar rats on feeding behaviour, glucose and energy metabolism, circulating hormones (luteinising hormone, testosterone, insulin and corticosterone) and hypothalamic neuronal activity. Kp increased plasma testosterone levels, had an anorexigenic effect and increased lipid catabolism, as attested by a decreased respiratory exchange ratio (RER). RFRP-3 also increased plasma testosterone levels but did not modify food intake or energy metabolism. Both RF-amides increased endogenous glucose production, yet with no change in plasma glucose levels, suggesting that these peptides provoke not only a release of hepatic glucose, but also a change in glucose utilisation. Finally, plasma insulin and corticosterone levels did not change after the RF-amide treatment. The Kp effects were associated with an increased c-Fos expression in the median preoptic area and a reduction in pro-opiomelanocortin immunostaining in the arcuate nucleus. No effects on neuronal activation were found for RFRP-3. Our results provide further evidence that Kp is not only a very potent hypothalamic activator of reproduction, but also part of the hypothalamic circuit controlling energy metabolism.
Collapse
Affiliation(s)
- Fernando Cázarez‐Márquez
- Institute of Cellular and Integrative Neurosciences (INCI)StrasbourgFrance
- Netherlands Institute for Neuroscience (NIN)AmsterdamThe Netherlands
- Laboratory of EndocrinologyAmsterdam UMCAmsterdam Gastroenterology & MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
| | - Jitske Eliveld
- Netherlands Institute for Neuroscience (NIN)AmsterdamThe Netherlands
- Laboratory of EndocrinologyAmsterdam UMCAmsterdam Gastroenterology & MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
| | - Wayne I. G. R. Ritsema
- Netherlands Institute for Neuroscience (NIN)AmsterdamThe Netherlands
- Laboratory of EndocrinologyAmsterdam UMCAmsterdam Gastroenterology & MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
| | - Ewout Foppen
- Netherlands Institute for Neuroscience (NIN)AmsterdamThe Netherlands
- Laboratory of EndocrinologyAmsterdam UMCAmsterdam Gastroenterology & MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
| | - Yvonne Bossenbroek
- Laboratory of EndocrinologyAmsterdam UMCAmsterdam Gastroenterology & MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
| | - Simone Pelizzari
- Netherlands Institute for Neuroscience (NIN)AmsterdamThe Netherlands
| | - Valérie Simonneaux
- Institute of Cellular and Integrative Neurosciences (INCI)StrasbourgFrance
| | - Andries Kalsbeek
- Netherlands Institute for Neuroscience (NIN)AmsterdamThe Netherlands
- Laboratory of EndocrinologyAmsterdam UMCAmsterdam Gastroenterology & MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Endocrinology and MetabolismAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
13
|
Micioni Di Bonaventura E, Botticelli L, Tomassoni D, Tayebati SK, Micioni Di Bonaventura MV, Cifani C. The Melanocortin System behind the Dysfunctional Eating Behaviors. Nutrients 2020; 12:E3502. [PMID: 33202557 PMCID: PMC7696960 DOI: 10.3390/nu12113502] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022] Open
Abstract
The dysfunction of melanocortin signaling has been associated with obesity, given the important role in the regulation of energy homeostasis, food intake, satiety and body weight. In the hypothalamus, the melanocortin-3 receptor (MC3R) and melanocortin-4 receptor (MC4R) contribute to the stability of these processes, but MC3R and MC4R are also localized in the mesolimbic dopamine system, the region that responds to the reinforcing properties of highly palatable food (HPF) and where these two receptors seem to affect food reward and motivation. Loss of function of the MC4R, resulting from genetic mutations, leads to overeating in humans, but to date, a clear understanding of the underlying mechanisms and behaviors that promote overconsumption of caloric foods remains unknown. Moreover, the MC4R demonstrated to be a crucial modulator of the stress response, factor that is known to be strictly related to binge eating behavior. In this review, we will explore the preclinical and clinical studies, and the controversies regarding the involvement of melanocortin system in altered eating patterns, especially binge eating behavior, food reward and motivation.
Collapse
Affiliation(s)
| | - Luca Botticelli
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| | - Daniele Tomassoni
- School of Bioscience and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy;
| | - Seyed Khosrow Tayebati
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| | | | - Carlo Cifani
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| |
Collapse
|
14
|
Lutgens E, Atzler D, Döring Y, Duchene J, Steffens S, Weber C. Immunotherapy for cardiovascular disease. Eur Heart J 2020; 40:3937-3946. [PMID: 31121017 DOI: 10.1093/eurheartj/ehz283] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/11/2019] [Accepted: 04/17/2019] [Indexed: 12/21/2022] Open
Abstract
The outcomes of the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) trial have unequivocally proven that inflammation is a key driver of atherosclerosis and that targeting inflammation, in this case by using an anti-interleukin-1β antibody, improves cardiovascular disease (CVD) outcomes. This is especially true for CVD patients with a pro-inflammatory constitution. Although CANTOS has epitomized the importance of targeting inflammation in atherosclerosis, treatment with canakinumab did not improve CVD mortality, and caused an increase in infections. Therefore, the identification of novel drug targets and development of novel therapeutics that block atherosclerosis-specific inflammatory pathways and exhibit limited immune-suppressive side effects, as pursued in our collaborative research centre, are required to optimize immunotherapy for CVD. In this review, we will highlight the potential of novel immunotherapeutic targets that are currently considered to become a future treatment for CVD.
Collapse
Affiliation(s)
- Esther Lutgens
- Institute for Cardiovascular Prevention (IPEK), CRC 1123 Atherosclerosis - Mechanisms and Networks of novel therapeutic Targets, Ludwig-Maximilians-Universität, Ludwig-Maximilians-University Munich, Pettenkoferstraße 9, Munich 80336, Germany.,Department of Medical Biochemistry, Amsterdam University Medical Centers, Location AMC, Amsterdam Cardiovascular Sciences (ACS), University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, the Netherlands.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention (IPEK), CRC 1123 Atherosclerosis - Mechanisms and Networks of novel therapeutic Targets, Ludwig-Maximilians-Universität, Ludwig-Maximilians-University Munich, Pettenkoferstraße 9, Munich 80336, Germany.,Department of Medical Biochemistry, Amsterdam University Medical Centers, Location AMC, Amsterdam Cardiovascular Sciences (ACS), University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, the Netherlands.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.,Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Goethestraße 33, Munich 80336, Germany
| | - Yvonne Döring
- Institute for Cardiovascular Prevention (IPEK), CRC 1123 Atherosclerosis - Mechanisms and Networks of novel therapeutic Targets, Ludwig-Maximilians-Universität, Ludwig-Maximilians-University Munich, Pettenkoferstraße 9, Munich 80336, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Johan Duchene
- Institute for Cardiovascular Prevention (IPEK), CRC 1123 Atherosclerosis - Mechanisms and Networks of novel therapeutic Targets, Ludwig-Maximilians-Universität, Ludwig-Maximilians-University Munich, Pettenkoferstraße 9, Munich 80336, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), CRC 1123 Atherosclerosis - Mechanisms and Networks of novel therapeutic Targets, Ludwig-Maximilians-Universität, Ludwig-Maximilians-University Munich, Pettenkoferstraße 9, Munich 80336, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), CRC 1123 Atherosclerosis - Mechanisms and Networks of novel therapeutic Targets, Ludwig-Maximilians-Universität, Ludwig-Maximilians-University Munich, Pettenkoferstraße 9, Munich 80336, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitsingel 50, 6229 ER Maastricht, the Netherlands
| |
Collapse
|
15
|
Holland J, Sorrell J, Yates E, Smith K, Arbabi S, Arnold M, Rivir M, Morano R, Chen J, Zhang X, Dimarchi R, Woods SC, Sanchez-Gurmaches J, Wohleb E, Perez-Tilve D. A Brain-Melanocortin-Vagus Axis Mediates Adipose Tissue Expansion Independently of Energy Intake. Cell Rep 2020; 27:2399-2410.e6. [PMID: 31116984 PMCID: PMC6550338 DOI: 10.1016/j.celrep.2019.04.089] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 03/18/2019] [Accepted: 04/18/2019] [Indexed: 02/07/2023] Open
Abstract
The melanocortin system is a brain circuit that influences energy balance by regulating energy intake and expenditure. In addition, the brain-melanocortin system controls adipose tissue metabolism to optimize fuel mobilization and storage. Specifically, increased brain-melanocortin signaling or negative energy balance promotes lipid mobilization by increasing sympathetic nervous system input to adipose tissue. In contrast, calorie-independent mechanisms favoring energy storage are less understood. Here, we demonstrate that reduction of brain-melanocortin signaling actively promotes fat mass gain by activating the lipogenic program and adipocyte and endothelial cell proliferation in white fat depots independently of caloric intake via efferent nerve fibers conveyed by the common hepatic branch of the vagus nerve. Those vagally regulated obesogenic signals also contribute to the fat mass gain following chronic high-fat diet feeding. These data reveal a physiological mechanism whereby the brain controls energy stores that may contribute to increased susceptibility to obesity. Brain-melanocortin signaling controls fat mass indirectly by regulating energy balance and by direct control of lipid mobilization from adipose tissue via sympathetic nervous system activity. Holland et al. show that reduced brain-melanocortin signaling promotes white adipose tissue expansion via signals conveyed by efferent innervation of the vagus nerve.
Collapse
Affiliation(s)
- Jenna Holland
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joyce Sorrell
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Emily Yates
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kathleen Smith
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Shahriar Arbabi
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Myrtha Arnold
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Marita Rivir
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rachel Morano
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jenny Chen
- Genomics, Epigenomics and Sequencing Core, Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Xiang Zhang
- Genomics, Epigenomics and Sequencing Core, Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Richard Dimarchi
- Novo Nordisk Research Center Indianapolis, IN, USA; Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - Stephen C Woods
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joan Sanchez-Gurmaches
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Endocrinology and Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, USA
| | - Eric Wohleb
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Diego Perez-Tilve
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
16
|
Powell DR, Doree DD, DaCosta CM, Platt KA, Hansen GM, van Sligtenhorst I, Ding ZM, Revelli JP, Brommage R. Obesity of G2e3 Knockout Mice Suggests That Obesity-Associated Variants Near Human G2E3 Decrease G2E3 Activity. Diabetes Metab Syndr Obes 2020; 13:2641-2652. [PMID: 32801815 PMCID: PMC7394505 DOI: 10.2147/dmso.s259546] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
PURPOSE In humans, single nucleotide polymorphisms (SNPs) near the adjacent protein kinase D1 (PRKD1) and G2/M-phase-specific E3 ubiquitin protein ligase (G2E3) genes on chromosome 14 are associated with obesity. To date, no published evidence links inactivation of either gene to changes in body fat. These two genes are also adjacent on mouse chromosome 12. Because obesity genes are highly conserved between humans and mice, we analyzed body fat in adult G2e3 and Prkd1 knockout (KO) mice to determine whether inactivating either gene leads to obesity in mice and, by inference, probably in humans. METHODS The G2e3 and Prkd1 KO lines were generated by gene trapping and by homologous recombination methodologies, respectively. Body fat was measured by DEXA in adult mice fed chow from weaning and by QMR in a separate cohort of mice fed high-fat diet (HFD) from weaning. Glucose homeostasis was evaluated with oral glucose tolerance tests (OGTTs) performed on adult mice fed HFD from weaning. RESULTS Body fat was increased in multiple cohorts of G2e3 KO mice relative to their wild-type (WT) littermates. When data from all G2e3 KO (n=32) and WT (n=31) mice were compared, KO mice showed increases of 11% in body weight (P<0.01), 65% in body fat (P<0.001), 48% in % body fat (P<0.001), and an insignificant 3% decrease in lean body mass. G2e3 KO mice were also glucose intolerant during an OGTT (P<0.05). In contrast, Prkd1 KO and WT mice had comparable body fat levels and glucose tolerance. CONCLUSION Significant obesity and glucose intolerance were observed in G2e3, but not Prkd1, KO mice. The conservation of obesity genes between mice and humans strongly suggests that the obesity-associated SNPs located near the human G2E3 and PRKD1 genes are linked to variants that decrease the amount of functional human G2E3.
Collapse
Affiliation(s)
- David R Powell
- Lexicon Pharmaceuticals Inc, The Woodlands, TX, 77381, USA
- Correspondence: David R Powell Lexicon Pharmaceuticals Inc., 8800 Technology Forest Place, The Woodlands, TX77381, USATel +1 281 863 3060Fax +1 281 863 8115 Email
| | - Deon D Doree
- Lexicon Pharmaceuticals Inc, The Woodlands, TX, 77381, USA
| | | | | | - Gwenn M Hansen
- Lexicon Pharmaceuticals Inc, The Woodlands, TX, 77381, USA
| | | | - Zhi-Ming Ding
- Lexicon Pharmaceuticals Inc, The Woodlands, TX, 77381, USA
| | | | | |
Collapse
|
17
|
Kwon EJ, Lee HA, You YA, Yoo JY, Park H, Park EA, Ha EH, Kim YJ. MC4R and HNF4α promoter methylation at birth contribute to triglyceride levels in childhood: A prospective cohort study. Medicine (Baltimore) 2019; 98:e16424. [PMID: 31305461 PMCID: PMC6641802 DOI: 10.1097/md.0000000000016424] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Although the changes in DNA methylation are assumed to be due to the association between adverse intrauterine conditions and adult metabolic health, evidence from human studies is rare. Little is known about the changes in DNA methylation present at birth that affect metabolic profiles in childhood. Previous studies have shown that the melanocortin 4 receptor (MC4R) and hepatocyte nuclear factor 4 alpha (HNF4α) genes are associated with obesity and metabolic disorders. Thus, we investigated the associations of the DNA methylation statuses of MC4R and HNF4α in cord blood with metabolic profiles in childhood.We collected data from 90 children 7 to 9 years of age included in the Ewha Birth & Growth Cohort Study in Korea. DNA methylation was analyzed by pyrosequencing. The children were split into 2 groups according to the cutoff triglyceride (TG) levels (<110 and ≥110 mg/dL).The methylation statuses of MC4R and HNF4α at birth were significantly associated with the TG level in childhood (P < .05). It was interesting to note that the methylation statuses of MC4R and HNF4α in cord blood were significantly decreased, whereas childhood body mass index was significantly increased, in children with high TG levels compared with children with low TG levels (P < .05).Our findings show that the methylation statuses of MC4R and HNF4α at birth are associated with metabolic profiles in childhood. These epigenetic modifications occurring in early life may contribute to subsequent metabolic-related disorders. Thus, we suggest that DNA methylation status in cord blood may be predictive of the risk of developing metabolic syndrome.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eun Hee Ha
- Department of Occupational and Environmental Medicine
| | - Young Ju Kim
- Department of Obstetrics and Gynecology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
18
|
Yang LK, Zhang ZR, Wen HS, Tao YX. Characterization of channel catfish (Ictalurus punctatus) melanocortin-3 receptor reveals a potential network in regulation of energy homeostasis. Gen Comp Endocrinol 2019; 277:90-103. [PMID: 30905760 DOI: 10.1016/j.ygcen.2019.03.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 03/12/2019] [Accepted: 03/18/2019] [Indexed: 11/19/2022]
Abstract
The melanocortin-3 receptor (MC3R) is known to be involved in regulation of energy homeostasis, regulating feed efficiency and nutrient partitioning in mammals. Its physiological roles in non-mammalian vertebrates, especially economically important aquaculture species, are not well understood. Channel catfish (Ictalurus punctatus) is the main freshwater aquaculture species in North America. In this study, we characterized the channel catfish MC3R. The mc3r of channel catfish encoded a putative protein (ipMC3R) of 367 amino acids. We transfected HEK293T cells with ipMC3R plasmid for functional studies. Five agonists, including adrenocorticotropin, α-melanocyte stimulating hormone (α-MSH), β-MSH, [Nle4, D-Phe7]-α-MSH, and D-Trp8-γ-MSH, were used in the pharmacological studies. Our results showed that ipMC3R bound β-MSH with higher affinity and D-Trp8-γ-MSH with lower affinity compared with human MC3R. All agonists could stimulate ipMC3R and increase intracellular cAMP production with sub-nanomolar potencies. The extracellular signal-regulated kinases 1 and 2 (ERK1/2) activation could also be triggered by ipMC3R. The ipMC3R exhibited constitutive activities in both cAMP and ERK1/2 pathways, and Agouti-related protein served as an inverse agonist at ipMC3R, potently inhibiting the high basal cAMP level. Moreover, we showed that melanocortin receptor accessory protein 2 (MRAP2) preferentially modulated ipMC3R in cAMP production rather than ERK1/2 activation. Our study will assist further investigation of the physiological roles of the ipMC3R, especially in energy homeostasis, in channel catfish.
Collapse
Affiliation(s)
- Li-Kun Yang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Zheng-Rui Zhang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States; Key Laboratory of Marine Genetics and Breeding, Ministry of Education, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Hai-Shen Wen
- College of Fisheries, Ocean University of China, Qingdao, China
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
19
|
Hubbard K, Shome A, Sun B, Pontré B, McGregor A, Mountjoy KG. Chronic High-Fat Diet Exacerbates Sexually Dimorphic Pomctm1/tm1 Mouse Obesity. Endocrinology 2019; 160:1081-1096. [PMID: 30997487 PMCID: PMC6469954 DOI: 10.1210/en.2018-00924] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/04/2019] [Indexed: 01/16/2023]
Abstract
Mice with a targeted mutation in the pro-opiomelanocortin (Pomc) gene (Pomctm1/tm1 mice) are unable to synthesize desacetyl-α-MSH and α-MSH and they develop obesity when fed chow diet. In this study, we hypothesized that a chronic high-fat (HF) diet exacerbates Pomctm1/tm1 mouse obesity. Male and female Pomcwt/wt and Pomctm1/tm1 mice were fed low-fat (LF) (10 kcal percent fat) or HF (45 kcal percent fat) diets from weaning for 23 weeks. We show that Pomctm1/tm1 mouse obesity is sexually dimorphic and exacerbated by an HF diet. Male Pomctm1/tm1 mice develop obesity because they are hyperphagic compared with Pomcwt/wt mice when fed an LF or HF diet. Female Pomctm1/tm1 mice develop obesity when feeding on an LF or HF diet because they exhibit signs of reduced energy expenditure (no change in feed efficiency; body weight gained exceeding energy intake) compared with Pomcwt/wt mice. A chronic HF diet exacerbates male Pomctm1/tm1 and Pomcwt/wt mouse obesity, and the increased energy intake fully accounts for increased weight gain. In contrast, female Pomcwt/wt mice are protected from chronic HF diet-induced obesity because they reduce the amount of HF diet eaten, and they appear to increase their energy expenditure (no change in feed efficiency but energy intake exceeding body weight gained). A chronic HF diet exacerbates female Pomctm1/tm1 mouse obesity due to impaired ability to reduce the amount of HF diet eaten and apparent impaired HF diet-induced adaptive thermogenesis. Our data show that desacetyl-α-MSH and α-MSH are required for sexually dimorphic HF diet-induced C57BL/6J obesity. In conclusion, desacetyl-α-MSH and α-MSH play salutary roles in sexually dimorphic melanocortin obesity and sexually dimorphic HF diet-induced C57BL/6J obesity.
Collapse
Affiliation(s)
- Kristina Hubbard
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Avik Shome
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Bo Sun
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Beau Pontré
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Ailsa McGregor
- Department of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kathleen G 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 Biodiscovery, University of Auckland, Auckland, New Zealand
| |
Collapse
|
20
|
Pei H, Patterson CM, Sutton AK, Burnett KH, Myers MG, Olson DP. Lateral Hypothalamic Mc3R-Expressing Neurons Modulate Locomotor Activity, Energy Expenditure, and Adiposity in Male Mice. Endocrinology 2019; 160:343-358. [PMID: 30541071 PMCID: PMC6937456 DOI: 10.1210/en.2018-00747] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/04/2018] [Indexed: 02/05/2023]
Abstract
The central melanocortin system plays a crucial role in the control of energy balance. Although the decreased energy expenditure and increased adiposity of melanocortin-3 receptor (Mc3R)-null mice suggest the importance of Mc3R-regulated neurons in energy homeostasis, the roles for specific subsets of Mc3R neurons in energy balance have yet to be determined. Because the lateral hypothalamic area (LHA) contributes to the control of energy expenditure and feeding, we generated Mc3rcre mice to determine the roles of LHA Mc3R (Mc3RLHA) neurons in energy homeostasis. We found that Mc3RLHA neurons overlap extensively with LHA neuron markers that contribute to the control of energy balance (neurotensin, galanin, and leptin receptor) and project to brain areas involved in the control of feeding, locomotion, and energy expenditure, consistent with potential roles for Mc3RLHA neurons in these processes. Indeed, selective chemogenetic activation of Mc3RLHA neurons increased locomotor activity and augmented refeeding after a fast. Although the ablation of Mc3RLHA neurons did not alter food intake, mice lacking Mc3RLHA neurons displayed decreased energy expenditure and locomotor activity, along with increased body mass and adiposity. Thus, Mc3R neurons lie within LHA neurocircuitry that modulates locomotor activity and energy expenditure and contribute to energy balance control.
Collapse
Affiliation(s)
- Hongjuan Pei
- Division of Endocrinology, Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan
| | | | - Amy K Sutton
- Molecular and Integrative Physiology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Korri H Burnett
- Division of Endocrinology, Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan
| | - Martin G Myers
- Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan
- Molecular and Integrative Physiology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - David P Olson
- Division of Endocrinology, Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan
- Molecular and Integrative Physiology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
- Correspondence: David P. Olson, MD, PhD, University of Michigan, 1000 Wall Street, Brehm Tower 6329, Ann Arbor, Michigan 48105. E-mail:
| |
Collapse
|
21
|
Ding W, Chang WG, Guo XC, Liu Y, Xiao DD, Ding D, Wang JX, Zhang XJ. Exenatide Protects Against Cardiac Dysfunction by Attenuating Oxidative Stress in the Diabetic Mouse Heart. Front Endocrinol (Lausanne) 2019; 10:202. [PMID: 31024445 PMCID: PMC6459897 DOI: 10.3389/fendo.2019.00202] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/12/2019] [Indexed: 01/01/2023] Open
Abstract
Cardiovascular disease is the major cause of death in patients with diabetes. Current treatment strategies for diabetes rely on lifestyle changes and glucose control to prevent angiopathy and organ failure. Exenatide, a glucagon-like peptide-1 (GLP-1) receptor agonist, is used as an add-on therapy to insulin treatment. Exenatide also has multiple beneficial effects in addition to its hypoglycemic effects, such as preventing hepatic steatosis and protecting against cardiac injury from doxorubicin-induced cardiotoxicity or ischemic reperfusion. However, the mechanisms underlying the cardioprotective effects of exenatide in diabetes have not been fully clarified. To address this issue, we investigated the cardioprotective effects of exenatide in type 1 and type 2 diabetic mice. We found that exenatide simultaneously attenuated reactive oxidative species (ROS) production through increases in the antioxidant enzymes manganese dependent superoxide dismutase (MnSOD) and catalase. Moreover, exenatide decreased tumor protein P53 (p53) expression and prevented cell apoptosis in H9c2 cells. The presence of the catalase inhibitor 3-AT attenuated the effects of exenatide. Overall, the results strongly indicate that exenatide treatment may be protective against the development of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Wei Ding
- Department of General Medicine, The Affiliated Hospital, Qingdao University, Qingdao, China
| | - Wen-guang Chang
- Center for Regenerative Medicine, Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Xiao-ci Guo
- Department of General Medicine, The Affiliated Hospital, Qingdao University, Qingdao, China
| | - Ying Liu
- Center for Regenerative Medicine, Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Dan-dan Xiao
- Center for Regenerative Medicine, Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Dan Ding
- Center for Regenerative Medicine, Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Jian-xun Wang
- Center for Regenerative Medicine, Institute for Translational Medicine, Qingdao University, Qingdao, China
- School of Basic Medical Sciences, Qingdao University, Qingdao, China
- *Correspondence: Jian-xun Wang
| | - Xue-juan Zhang
- Department of General Medicine, The Affiliated Hospital, Qingdao University, Qingdao, China
- Xue-juan Zhang
| |
Collapse
|
22
|
Yamada T, Kashiwagi Y, Rokugawa T, Kato H, Konishi H, Hamada T, Nagai R, Masago Y, Itoh M, Suganami T, Ogawa Y, Abe K. Evaluation of hepatic function using dynamic contrast-enhanced magnetic resonance imaging in melanocortin 4 receptor-deficient mice as a model of nonalcoholic steatohepatitis. Magn Reson Imaging 2018; 57:210-217. [PMID: 30465867 DOI: 10.1016/j.mri.2018.11.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/09/2018] [Accepted: 11/17/2018] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Melanocortin 4 receptor-deficient (MC4R-KO) mice fed a high-fat diet (HFD) develop liver pathology similar to human nonalcoholic steatohepatitis (NASH). However, although liver histology and blood biochemistry have been reported, hepatic function has not been evaluated. In the present study, we evaluated hepatic function in MC4R-KO mice fed an HFD using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) with gadolinium‑ethoxybenzyl‑diethylenetriamine pentaacetic acid (Gd-EOB-DTPA). MATERIALS AND METHODS Wild type (WT) mice and MC4R-KO mice were fed a standard diet (SD) or an HFD for 20 weeks. The hepatic signal intensity was obtained from DCE-MRI images, and relative enhancement (RE), the time to maximum RE (Tmax), and the half-life of RE elimination (T1/2) were calculated. Histopathological analysis was then performed. RESULTS Histological analysis with nonalcoholic fatty liver disease activity score (NAS) revealed that MC4R-KO mice fed an HFD achieved the NAS of 5. There was moderate fibrosis in MC4R-KO mice fed an HFD. DCE-MRI with Gd-EOB-DTPA showed that Tmax and T1/2 were significantly longer in MC4R-KO mice fed an HFD compared with wild type (WT) mice (Tmax, WT, 3.9 ± 0.4 min; MC4R-KO, 7.4 ± 1.5 min; T1/2, WT, 23.7 ± 1.9 min; MC4R-KO, 62.5 ± 18.5 min). Tmax and T1/2 were significantly correlated with histopathologic score (steatosis vs. Tmax, rho = 0.48, P = 0.04; steatosis vs. T1/2, rho = 0.50, P = 0.03; inflammation vs. Tmax, rho = 0.55, P = 0.02; inflammation vs. T1/2, rho = 0.61, P < 0.01; ballooning vs. T1/2, rho = 0.51, P = 0.03;fibrosis vs Tmax, rho = 0.72, P < 0.01; fibrosis vs T1/2, rho = 0.75, P < 0.01). CONCLUSIONS MC4R-KO mice fed an HFD developed obesity and NASH. The liver kinetics of Gd-EOB-DTPA were significantly different in MC4R-KO mice fed an HFD from WT mice, and correlated with the histopathologic score. These results suggest that MC4R-KO mice fed an HFD mimic the hepatic pathology and liver function of human NASH, and therefore might be useful for the study of hepatic dysfunction during the fibrotic stage of NASH.
Collapse
Affiliation(s)
- Tomomi Yamada
- Biomarker R&D Department, Shionogi & Co., Ltd., Osaka, Japan.
| | - Yuto Kashiwagi
- Biomarker R&D Department, Shionogi & Co., Ltd., Osaka, Japan
| | - Takemi Rokugawa
- Biomarker R&D Department, Shionogi & Co., Ltd., Osaka, Japan
| | - Hideaki Kato
- Drug Discovery& Disease Research Laboratory, Shionogi & Co., Ltd., Osaka, Japan
| | - Haruyo Konishi
- Drug Discovery& Disease Research Laboratory, Shionogi & Co., Ltd., Osaka, Japan
| | - Tadateru Hamada
- Drug Discovery& Disease Research Laboratory, Shionogi & Co., Ltd., Osaka, Japan
| | - Ryohei Nagai
- Drug Discovery& Disease Research Laboratory, Shionogi & Co., Ltd., Osaka, Japan
| | - Yusaku Masago
- Drug Discovery& Disease Research Laboratory, Shionogi & Co., Ltd., Osaka, Japan
| | - Michiko Itoh
- Department of Organ Network and Metabolism, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Yoshihiro Ogawa
- Department of Medical and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Molecular and Cellular Metabolism, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Japan Agency for Medical Research and Development, CREST, Tokyo, Japan
| | - Kohji Abe
- Biomarker R&D Department, Shionogi & Co., Ltd., Osaka, Japan
| |
Collapse
|
23
|
Rapkin J, Jensen K, House CM, Wilson AJ, Hunt J. Genotype-by-sex-by-diet interactions for nutritional preference, dietary consumption, and lipid deposition in a field cricket. Heredity (Edinb) 2018; 121:361-373. [PMID: 30089778 DOI: 10.1038/s41437-018-0130-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 07/05/2018] [Accepted: 07/14/2018] [Indexed: 12/13/2022] Open
Abstract
Changes in feeding behaviour, especially the overconsumption of calories, has led to a rise in the rates of obesity, diabetes, and other associated disorders in humans and a range of animals inhabiting human-influenced environments. However, understanding the relative contribution of genes, the nutritional environment, and their interaction to dietary intake and lipid deposition in the sexes still remains a major challenge. By combining nutritional geometry with quantitative genetics, we determined the effect of genes, the nutritional environment, and their interaction on the total nutritional preference (TP), total diet eaten (TE), and lipid mass (LM) of male and female black field crickets (Teleogryllus commodus) fed one of four diet pairs (DPs) differing in the ratio of protein to carbohydrate and total nutritional content. We found abundant additive genetic variance for TP, TE, and LM in both sexes and across all four DPs, with significant genetic correlations between TE and TP and between TP and LM in males. We also found significant genotype-by-DP and genotype-by-sex-by-DP interactions for each trait and significant genotype-by-sex interactions for TE and LM. Complex interactions between genes, sex, and the nutritional environment, therefore, play an important role in nutrient regulation and lipid deposition in T. commodus. This finding may also help explain the increasing rate of obesity and the maintenance of sex differences in obesity observed across many animal species, including humans.
Collapse
Affiliation(s)
- James Rapkin
- Centre for Ecology and Conservation, University of Exeter, Cornwall Campus, Penryn, Cornwall, TR10 9FE, UK
| | - Kim Jensen
- Department of Bioscience, Terrestrial Ecology, Aarhus University, Vejlsøvej 25, 8600, Silkeborg, Denmark
| | - Clarissa M House
- Centre for Ecology and Conservation, University of Exeter, Cornwall Campus, Penryn, Cornwall, TR10 9FE, UK.,School of Science and Health, Western Sydney University, Locked Bag 1797, Penrith, NSW, 2751, Australia.,Hawkesbury Institute for the Environment, University of Western Sydney, Locked Bay 1797, Penrith, NSW, 2751, Australia
| | - Alastair J Wilson
- Centre for Ecology and Conservation, University of Exeter, Cornwall Campus, Penryn, Cornwall, TR10 9FE, UK
| | - John Hunt
- Centre for Ecology and Conservation, University of Exeter, Cornwall Campus, Penryn, Cornwall, TR10 9FE, UK. .,School of Science and Health, Western Sydney University, Locked Bag 1797, Penrith, NSW, 2751, Australia. .,Hawkesbury Institute for the Environment, University of Western Sydney, Locked Bay 1797, Penrith, NSW, 2751, Australia.
| |
Collapse
|
24
|
Liang J, Li L, Jin X, Xu B, Pi L, Liu S, Zhu W, Zhang C, Luan B, Gong L, Zhang C. Pharmacological effect of human melanocortin-2 receptor accessory protein 2 variants on hypothalamic melanocortin receptors. Endocrine 2018; 61:94-104. [PMID: 29704154 DOI: 10.1007/s12020-018-1596-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 04/09/2018] [Indexed: 10/17/2022]
Abstract
PURPOSE Melanocortin-3 receptor (MC3R), melanocortin-4 receptor (MC4R), and a recently identified melanocortin-2 receptor accessory protein 2 (MRAP2), are highly expressed in hypothalamus and coordinately regulate energy homeostasis, but the single cellular transcriptome of melanocortin system remains unknown. Several infrequent MRAP2 variants are reported from severe obese human patients but the mechanisms on how they affect melanocortin signaling are unclear. METHODS First, we performed in silico analysis of mouse hypothalamus RNA sequencing datasets at single-cell resolution from two independent studies. Next, we inspected the three-dimensional conformational alteration of three mutations on MRAP2 protein. Finally, the influence of MRAP2 variants on MC3R and MC4R signaling was analyzed in vitro. RESULTS (1) We confirmed the actual co-expression of Mrap2 with Mc3r and Mc4r, and demonstrated more broad distribution of Mrap2-positive neuronal populations than Mc3r or Mc4r in mouse hypothalamus. (2) Compared with wild-type MRAP2, MRAP2N88Y, and MRAP2R125C showed impaired α-MSH-induced MC4R or MC3R stimulation. (3) MRAP2N88Yexhibited enhanced interaction with MC4R protein and its own. CONCLUSIONS This is the first dedicated description of single-cell transcriptome signature of Mrap2, Mc3r, and Mc4r in the central nerve system and the first evidence describing the unique dimer formation, conformational change, and pharmacological effect of MRAP2 mutations on MC3R signaling.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- Carrier Proteins/pharmacology
- Computer Simulation
- Genetic Variation
- Humans
- Hypothalamus/drug effects
- Hypothalamus/metabolism
- Mice
- Mutation/genetics
- Neurons/metabolism
- Nucleic Acid Conformation
- Plasmids
- RNA/genetics
- Receptor, Melanocortin, Type 3/genetics
- Receptor, Melanocortin, Type 3/metabolism
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/metabolism
- Receptors, Melanocortin/drug effects
- Signal Transduction/genetics
- alpha-MSH/pharmacology
Collapse
Affiliation(s)
- Jinye Liang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Lei Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xuanxuan Jin
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Bingxin Xu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Linyu Pi
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shangyun Liu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Wei Zhu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Cong Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Bing Luan
- Department of Endocrinology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lulu Gong
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.
| | - Chao Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.
| |
Collapse
|
25
|
Ghoshal S, Stevens JR, Billon C, Girardet C, Sitaula S, Leon AS, Rao DC, Skinner JS, Rankinen T, Bouchard C, Nuñez MV, Stanhope KL, Howatt DA, Daugherty A, Zhang J, Schuelke M, Weiss EP, Coffey AR, Bennett BJ, Sethupathy P, Burris TP, Havel PJ, Butler AA. Adropin: An endocrine link between the biological clock and cholesterol homeostasis. Mol Metab 2017; 8:51-64. [PMID: 29331507 PMCID: PMC5985041 DOI: 10.1016/j.molmet.2017.12.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/28/2017] [Accepted: 12/02/2017] [Indexed: 01/13/2023] Open
Abstract
Objective Identify determinants of plasma adropin concentrations, a secreted peptide translated from the Energy Homeostasis Associated (ENHO) gene linked to metabolic control and vascular function. Methods Associations between plasma adropin concentrations, demographics (sex, age, BMI) and circulating biomarkers of lipid and glucose metabolism were assessed in plasma obtained after an overnight fast in humans. The regulation of adropin expression was then assessed in silico, in cultured human cells, and in animal models. Results In humans, plasma adropin concentrations are inversely related to atherogenic LDL-cholesterol (LDL-C) levels in men (n = 349), but not in women (n = 401). Analysis of hepatic Enho expression in male mice suggests control by the biological clock. Expression is rhythmic, peaking during maximal food consumption in the dark correlating with transcriptional activation by RORα/γ. The nadir in the light phase coincides with the rest phase and repression by Rev-erb. Plasma adropin concentrations in nonhuman primates (rhesus monkeys) also exhibit peaks coinciding with feeding times (07:00 h, 15:00 h). The ROR inverse agonists SR1001 and the 7-oxygenated sterols 7-β-hydroxysterol and 7-ketocholesterol, or the Rev-erb agonist SR9009, suppress ENHO expression in cultured human HepG2 cells. Consumption of high-cholesterol diets suppress expression of the adropin transcript in mouse liver. However, adropin over expression does not prevent hypercholesterolemia resulting from a high cholesterol diet and/or LDL receptor mutations. Conclusions In humans, associations between plasma adropin concentrations and LDL-C suggest a link with hepatic lipid metabolism. Mouse studies suggest that the relationship between adropin and cholesterol metabolism is unidirectional, and predominantly involves suppression of adropin expression by cholesterol and 7-oxygenated sterols. Sensing of fatty acids, cholesterol and oxysterols by the RORα/γ ligand-binding domain suggests a plausible functional link between adropin expression and cellular lipid metabolism. Furthermore, the nuclear receptors RORα/γ and Rev-erb may couple adropin synthesis with circadian rhythms in carbohydrate and lipid metabolism. In male humans, plasma adropin concentrations are inversely related to low-density circulating cholesterol (LDL-C) levels. Adropin expression is regulated by core elements of the biological clock (RORA/G, Rev-Erb). Sterol-sensing by the ROR ligand-binding domain provides a plausible link between adropin expression and lipid metabolism. In mouse liver, adropin expression is rhythmic and suppressed by exogenous (dietary) cholesterol.
Collapse
Affiliation(s)
- Sarbani Ghoshal
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Joseph R Stevens
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Cyrielle Billon
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Clemence Girardet
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Sadichha Sitaula
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Arthur S Leon
- School of Kinesiology and Leisure Studies, University of Minnesota, Minneapolis, MN, USA
| | - D C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - James S Skinner
- Department of Kinesiology, Indiana University, Bloomington, IN, USA
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Marinelle V Nuñez
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA; Department of Nutrition, School of Medicine, University of California-Davis, Davis, CA, USA
| | - Kimber L Stanhope
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA; Department of Nutrition, School of Medicine, University of California-Davis, Davis, CA, USA
| | - Deborah A Howatt
- Saha Cardiovascular Research Center, Department of Physiology, University of Kentucky, KY, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center, Department of Physiology, University of Kentucky, KY, USA
| | - Jinsong Zhang
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Matthew Schuelke
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Edward P Weiss
- Department of Nutrition and Dietetics, Doisy College of Health Sciences, Saint Louis University, St. Louis, MO, USA
| | - Alisha R Coffey
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC, USA
| | - Brian J Bennett
- Obesity and Metabolism Unit, Western Human Nutrition Center, USDA-ARS, Davis, CA, USA
| | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Thomas P Burris
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Peter J Havel
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA; Department of Nutrition, School of Medicine, University of California-Davis, Davis, CA, USA
| | - Andrew A Butler
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, USA.
| |
Collapse
|
26
|
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: 6.9] [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
|
27
|
Differential body weight, blood pressure and placental inflammatory responses to normal versus high-fat diet in melanocortin-4 receptor-deficient pregnant rats. J Hypertens 2017; 34:1998-2007. [PMID: 27467764 DOI: 10.1097/hjh.0000000000001059] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Although obesity increases the risk for hypertensive disorders of pregnancy, the mechanisms remain unclear. Neural melanocortin-4 receptor (MC4R) deficiency causes hyperphagia and obesity. Effects of MC4R deficiency on body weight, blood pressure (BP) and placental inflammatory responses to high-fat diet (HFD) are unknown. We tested two hypotheses: MC4R deficiency results in higher body weight, BP and placental inflammation under normal-fat diet (NFD) conditions and HFD exaggerates these responses in MC4R-deficient pregnant rats. METHODS MC4R and MC4R rats were maintained on NFD (13% kcal fat) or HFD (40% kcal fat) for ∼15 weeks, then measurements made on gestational day 19. RESULTS MC4R pregnant rats had greater body mass and total body fat and visceral adipose tissue weights along with greater circulating total cholesterol (TC) and leptin levels than MC4R rats regardless of diet. On NFD, circulating adiponectin levels were lower and placental TNFα levels and BP (conscious with carotid catheter) were higher in these heavier rats. Circulating adiponectin levels were lower and placental TNFα levels and BP were higher in MC4R rats compared with NFD controls. These parameters were not affected by HFD in the already heavier and hypertensive MC4R pregnant rats. CONCLUSION Obesity in MC4R deficiency and HFD in MC4R rats result in higher BP and placental inflammation during pregnancy. However, HFD did not exaggerate these responses in already obese MC4R pregnant rats. These data suggest that obesity and HFD are independently related to hypertension and placental inflammation in pregnancy.
Collapse
|
28
|
Miller GD. Appetite Regulation: Hormones, Peptides, and Neurotransmitters and Their Role in Obesity. Am J Lifestyle Med 2017; 13:586-601. [PMID: 31662725 DOI: 10.1177/1559827617716376] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/18/2017] [Accepted: 05/31/2017] [Indexed: 12/29/2022] Open
Abstract
Understanding body weight regulation will aid in the development of new strategies to combat obesity. This review examines energy homeostasis and food intake behaviors, specifically with regards to hormones, peptides, and neurotransmitters in the periphery and central nervous system, and their potential role in obesity. Dysfunction in feeding signals by the brain is a factor in obesity. The hypothalamic (arcuate nucleus) and brainstem (nucleus tractus solitaris) areas integrate behavioral, endocrine, and autonomic responses via afferent and efferent pathways from and to the brainstem and peripheral organs. Neurons present in the arcuate nucleus express pro-opiomelanocortin, Neuropeptide Y, and Agouti Related Peptide, with the former involved in lowering food intake, and the latter two acutely increasing feeding behaviors. Action of peripheral hormones from the gut, pancreas, adipose, and liver are also involved in energy homeostasis. Vagal afferent neurons are also important in regulating energy homeostasis. Peripheral signals respond to the level of stored and currently available fuel. By studying their actions, new agents maybe developed that disable orexigenic responses and enhance anorexigenic signals. Although there are relatively few medications currently available for obesity treatment, a number of agents are in development that work through these pathways.
Collapse
Affiliation(s)
- Gary D Miller
- Department of Health and Exercise Science, Wake Forest University, Winston-Salem, North Carolina
| |
Collapse
|
29
|
Demidowich AP, Jun JY, Yanovski JA. Polymorphisms and mutations in the melanocortin-3 receptor and their relation to human obesity. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2468-2476. [PMID: 28363697 DOI: 10.1016/j.bbadis.2017.03.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 12/18/2022]
Abstract
Inactivating mutations in the melanocortin 3 receptor (Mc3r) have been described as causing obesity in mice, but the physiologic effects of MC3R mutations in humans have been less clear. Here we review the MC3R polymorphisms and mutations identified in humans, and the in vitro, murine, and human cohort studies examining their putative effects. Some, but not all, studies suggest that the common human MC3R variant T6K+V81I, as well as several other rare, function-altering mutations, are associated with greater adiposity and hyperleptinemia with altered energy partitioning. In vitro, the T6K+V81I variant appears to decrease MC3R expression and therefore cAMP generation in response to ligand binding. Knockin mouse studies confirm that the T6K+V81I variant increases feeding efficiency and the avidity with which adipocytes derived from bone or adipose tissue stem cells store triglycerides. Other MC3R mutations occur too infrequently in the human population to make definitive conclusions regarding their clinical effects. This article is part of a Special Issue entitled: Melanocortin Receptors - edited by Ya-Xiong Tao.
Collapse
Affiliation(s)
- Andrew P Demidowich
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Joo Yun Jun
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Jack A Yanovski
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
30
|
Butler AA, Girardet C, Mavrikaki M, Trevaskis JL, Macarthur H, Marks DL, Farr SA. A Life without Hunger: The Ups (and Downs) to Modulating Melanocortin-3 Receptor Signaling. Front Neurosci 2017; 11:128. [PMID: 28360832 PMCID: PMC5352694 DOI: 10.3389/fnins.2017.00128] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 03/01/2017] [Indexed: 11/13/2022] Open
Abstract
Melanocortin neurons conserve body mass in hyper- or hypo-caloric conditions by conveying signals from nutrient sensors into areas of the brain governing appetite and metabolism. In mice, melanocortin-3 receptor (MC3R) deletion alters nutrient partitioning independently of hyperphagia, promoting accumulation of fat over muscle mass. Enhanced rhythms in insulin and insulin-responsive metabolic genes during hypocaloric feeding suggest partial insulin resistance and enhanced lipogenesis. However, exactly where and how MC3Rs affect metabolic control to alter nutrient partitioning is not known. The behavioral phenotypes exhibited by MC3R-deficient mice suggest a contextual role in appetite control. The impact of MC3R-deficiency on feeding behavior when food is freely available is minor. However, homeostatic responses to hypocaloric conditioning involving increased expression of appetite-stimulating (orexigenic) neuropeptides, binge-feeding, food anticipatory activity (FAA), entrainment to nutrient availability and enhanced feeding-related motivational responses are compromised with MC3R-deficiency. Rescuing Mc3r transcription in hypothalamic and limbic neurons improves appetitive responses during hypocaloric conditioning while having minor effects on nutrient partitioning, suggesting orexigenic functions. Rescuing hypothalamic MC3Rs also restores responses of fasting-responsive hypothalamic orexigenic neurons in hypocaloric conditions, suggesting actions that sensitize fasting-responsive neurons to signals from nutrient sensors. MC3R signaling in ventromedial hypothalamic SF1(+ve) neurons improves metabolic control, but does not restore appetitive responses or nutrient partitioning. In summary, desensitization of fasting-responsive orexigenic neurons may underlie attenuated appetitive responses of MC3R-deficient mice in hypocaloric situations. Further studies are needed to identify the specific location(s) of MC3Rs controlling appetitive responses and partitioning of nutrients between fat and lean tissues.
Collapse
Affiliation(s)
- Andrew A Butler
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine St. Louis, MO, USA
| | - Clemence Girardet
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine St. Louis, MO, USA
| | - Maria Mavrikaki
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine St. Louis, MO, USA
| | - James L Trevaskis
- In vivo Pharmacology, Cardiovascular and Metabolic Disease, Medimmune Gaithersburg, MD, USA
| | - Heather Macarthur
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine St. Louis, MO, USA
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health and Science University Portland, OR, USA
| | - Susan A Farr
- Department of Internal Medicine, Division of Geriatrics, Saint Louis University School of MedicineSt. Louis, MO, USA; VA Medical CenterSt. Louis, MO, USA
| |
Collapse
|
31
|
Melanocortin-3 receptors expressed in Nkx2.1(+ve) neurons are sufficient for controlling appetitive responses to hypocaloric conditioning. Sci Rep 2017; 7:44444. [PMID: 28294152 PMCID: PMC5353610 DOI: 10.1038/srep44444] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 02/08/2017] [Indexed: 01/12/2023] Open
Abstract
Melanocortin-3 receptors (MC3R) have a contextual role in appetite control that is amplified with hypocaloric conditioning. C57BL/6J (B6) mice subjected to hypocaloric feeding schedules (HFS) exhibit compulsive behavioral responses involving food anticipatory activity (FAA) and caloric loading following food access. These homeostatic responses to calorie-poor environs are attenuated in B6 mice in which Mc3r transcription is suppressed by a lox-stop-lox sequence in the 5'UTR (Mc3rTB/TB). Here, we report that optimization of caloric loading in B6 mice subject to HFS, characterized by increased meal size and duration, is not observed in Mc3rTB/TB mice. Analysis of hypothalamic and neuroendocrine responses to HFS throughout the light-dark cycle suggests uncoupling of hypothalamic responses involving appetite-stimulating fasting-responsive hypothalamic neurons expressing agouti-related peptide (AgRP) and neuropeptide Y (Npy). Rescuing Mc3rs expression in Nkx2.1(+ve) neurons is sufficient to restore normal hypothalamic responses to negative energy balance. In addition, Mc3rs expressed in Nkx2.1(+ve) neurons are also sufficient to restore FAA and caloric loading of B6 mice subjected to HFS. In summary, MC3Rs expressed in Nkx2.1(+ve) neurons are sufficient to coordinate hypothalamic response and expression of compulsive behavioral responses involving meal anticipation and consumption of large meals during situations of prolonged negative energy balance.
Collapse
|
32
|
Kato M, Huang YY, Matsuo M, Takashina Y, Sasaki K, Horai Y, Juni A, Kamijo SI, Saigo K, Ui-Tei K, Tei H. RNAi-mediated knockdown of mouse melanocortin-4 receptor in vitro and in vivo, using an siRNA expression construct based on the mir-187 precursor. Exp Anim 2017; 66:41-50. [PMID: 27725374 PMCID: PMC5301000 DOI: 10.1538/expanim.16-0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
RNA interference (RNAi) is a powerful tool for the study of gene function in mammalian
systems, including transgenic mice. Here, we report a gene knockdown system based on the
human mir-187 precursor. We introduced small interfering RNA (siRNA) sequences against the
mouse melanocortin-4 receptor (mMc4r) to alter the targeting of miR-187.
The siRNA-expressing cassette was placed under the control of the cytomegalovirus (CMV)
early enhancer/chicken β-actin promoter. In vitro, the construct
efficiently knocked down the gene expression of a co-transfected
mMc4r-expression vector in cultured mammalian cells. Using this
construct, we generated a transgenic mouse line which exhibited partial but significant
knockdown of mMc4r mRNA in various brain regions. Northern blot analysis
detected transgenic expression of mMc4r siRNA in these regions.
Furthermore, the transgenic mice fed a normal diet ate 9% more and were 30% heavier than
wild-type sibs. They also developed hyperinsulinemia and fatty liver as do
mMc4r knockout mice. We determined that this siRNA expression construct
based on mir-187 is a practical and useful tool for gene functional studies in
vitro as well as in vivo.
Collapse
Affiliation(s)
- Minoru Kato
- Research Unit/Neuroscience, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Hill JW, Faulkner LD. The Role of the Melanocortin System in Metabolic Disease: New Developments and Advances. Neuroendocrinology 2017; 104:330-346. [PMID: 27728914 PMCID: PMC5724371 DOI: 10.1159/000450649] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/01/2016] [Indexed: 12/17/2022]
Abstract
Obesity is increasing in prevalence across all sectors of society, and with it a constellation of associated ailments including hypertension, type 2 diabetes, and eating disorders. The melanocortin system is a critical neural system underlying the control of body weight and other functions. Deficits in the melanocortin system may promote or exacerbate the comorbidities of obesity. This system has therefore generated great interest as a potential target for treatment of obesity. However, drugs targeting melanocortin receptors are plagued by problematic side effects, including undesirable increases in sympathetic nervous system activity, heart rate, and blood pressure. Circumnavigating this roadblock will require a clearer picture of the precise neural circuits that mediate the functions of melanocortins. Recent, novel experimental approaches have significantly advanced our understanding of these pathways. We here review the latest advances in our understanding of the role of melanocortins in food intake, reward pathways, blood pressure, glucose control, and energy expenditure. The evidence suggests that downstream melanocortin-responsive circuits responsible for different physiological actions do diverge. Ultimately, a more complete understanding of melanocortin pathways and their myriad roles should allow treatments tailored to the mix of metabolic disorders in the individual patient.
Collapse
Affiliation(s)
- Jennifer W Hill
- Department of Physiology and Pharmacology, College of Medicine, The University of Toledo, Toledo, OH, USA
| | | |
Collapse
|
34
|
Severe Atherosclerosis and Hypercholesterolemia in Mice Lacking Both the Melanocortin Type 4 Receptor and Low Density Lipoprotein Receptor. PLoS One 2016; 11:e0167888. [PMID: 28030540 PMCID: PMC5193345 DOI: 10.1371/journal.pone.0167888] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 11/22/2016] [Indexed: 11/19/2022] Open
Abstract
Dysfunction of the melanocortin system can result in severe obesity accompanied with dyslipidemia and symptoms of the metabolic syndrome but the effect on vascular atherogenesis is not known. To study the impact of obesity and dyslipidemia on the cardiovascular system, we generated mice double-deficient for the melanocortin type 4 receptor (Mc4rmut mice) and the LDL receptor (Ldlr-/- mice). Mc4rmut mice develop obesity due to hyperphagia. Double-mutant mice (Mc4rmut;Ldlr-/-) exhibited massive increases in body weight, plasma cholesterol and triacylglycerol levels and developed atherosclerosis. Atherosclerotic lesion size was affected throughout the aortic root and brachiocephalic artery not only under semisynthetic, cholesterol-containing diet but also under cholesterol-free standard chow. The Mc4rmut mice developed a hepatic steatosis which contributes to increased plasma cholesterol levels even under cholesterol-free standard chow. Transcripts of cholesterol biosynthesis components and liver cholesterol levels did not significantly differ between wild-type and all mutant mouse strains but RNA sequencing data and biochemical measurements point to an altered bile acid elimination in Mc4rmut;Ldlr-/-. Therefore, the unchanged endogenous cholesterol biosynthesis together with a reduced hepatic VLDL and LDL-cholesterol clearance most likely led to increased plasma lipid levels and consequently to atherosclerosis in this animal model. Our data indicate that dysfunction of the melanocortin-regulated food intake and the resulting obesity significantly add to the proatherogenic lipoprotein profile caused by LDL receptor deficiency and, therefore, can be regarded as relevant risk factor for atherosclerosis.
Collapse
|
35
|
You P, Hu H, Chen Y, Zhao Y, Yang Y, Wang T, Xing R, Shao Y, Zhang W, Li D, Chen H, Liu M. Effects of Melanocortin 3 and 4 Receptor Deficiency on Energy Homeostasis in Rats. Sci Rep 2016; 6:34938. [PMID: 27713523 PMCID: PMC5054679 DOI: 10.1038/srep34938] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/20/2016] [Indexed: 01/08/2023] Open
Abstract
Melanocortin-3 and 4 receptors (MC3R and MC4R) can regulate energy homeostasis, but their respective roles especially the functions of MC3R need more exploration. Here Mc3r and Mc4r single and double knockout (DKO) rats were generated using CRISPR-Cas9 system. Metabolic phenotypes were examined and data were compared systematically. Mc3r KO rats displayed hypophagia and decreased body weight, while Mc4r KO and DKO exhibited hyperphagia and increased body weight. All three mutants showed increased white adipose tissue mass and adipocyte size. Interestingly, although Mc3r KO did not show a significant elevation in lipids as seen in Mc4r KO, DKO displayed even higher lipid levels than Mc4r KO. DKO also showed more severe glucose intolerance and hyperglycaemia than Mc4r KO. These data demonstrated MC3R deficiency caused a reduction of food intake and body weight, whereas at the same time exhibited additive effects on top of MC4R deficiency on lipid and glucose metabolism. This is the first phenotypic analysis and systematic comparison of Mc3r KO, Mc4r KO and DKO rats on a homogenous genetic background. These mutant rats will be important in defining the complicated signalling pathways of MC3R and MC4R. Both Mc4r KO and DKO are good models for obesity and diabetes research.
Collapse
Affiliation(s)
- Panpan You
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Handan Hu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Yuting Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Yongliang Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Yiqing Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Tongtong Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Roumei Xing
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Yanjiao Shao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Wen Zhang
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Huaqing Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, Shanghai 200241, China.,Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas 77030, USA
| |
Collapse
|
36
|
McConn BR, Matias J, Wang G, Cline MA, Gilbert ER. Dietary macronutrient composition affects hypothalamic appetite regulation in chicks. Nutr Neurosci 2016; 21:49-58. [PMID: 27686011 DOI: 10.1080/1028415x.2016.1219103] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Betty R. McConn
- Department of Animal and Poultry Sciences, Virginia Tech, 175 West Campus Drive, Blacksburg, VA 24061, USA
| | - Justin Matias
- Department of Animal and Poultry Sciences, Virginia Tech, 175 West Campus Drive, Blacksburg, VA 24061, USA
| | - Guoqing Wang
- Department of Animal and Poultry Sciences, Virginia Tech, 175 West Campus Drive, Blacksburg, VA 24061, USA
| | - Mark A. Cline
- Department of Animal and Poultry Sciences, Virginia Tech, 175 West Campus Drive, Blacksburg, VA 24061, USA
| | - Elizabeth R. Gilbert
- Department of Animal and Poultry Sciences, Virginia Tech, 175 West Campus Drive, Blacksburg, VA 24061, USA
| |
Collapse
|
37
|
Melanocortin-4 receptor-regulated energy homeostasis. Nat Neurosci 2016; 19:206-19. [PMID: 26814590 DOI: 10.1038/nn.4202] [Citation(s) in RCA: 231] [Impact Index Per Article: 25.7] [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
|
38
|
Mavrikaki M, Girardet C, Kern A, Faruzzi Brantley A, Miller CA, Macarthur H, Marks DL, Butler AA. Melanocortin-3 receptors in the limbic system mediate feeding-related motivational responses during weight loss. Mol Metab 2016; 5:566-579. [PMID: 27408780 PMCID: PMC4921936 DOI: 10.1016/j.molmet.2016.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 04/28/2016] [Accepted: 05/04/2016] [Indexed: 12/21/2022] Open
Abstract
Objective Appetitive responses to weight loss are mediated by a nutrient-sensing neural network comprised of melanocortin neurons. The role of neural melanocortin-3 receptors (MC3R) in mediating these responses is enigmatic. Mc3r knockout mice exhibit a paradoxical phenotype of obesity and reduced feeding-related behaviors in situations of nutrient scarcity. Here we examined whether MC3Rs expressed in mesolimbic neurons regulate feeding-related motivational responses. Methods Interactions between Mc3r genotype, cognitive function and energy balance on food self-administration were assessed using operant conditioning with fixed- and progressive ratio (FR1/PR1) settings. Inhibition of Mc3r transcription by a loxP-flanked transcriptional blocker (TB) in C57BL/6JN mice (Mc3rTB/TB) was reversed in mesolimbic neurons using DAT-Cre (DAT-MC3R). Results Caloric restriction (CR) caused 10–15% weight loss and increased motivation to acquire food rewards during training sessions. c-Fos-expression in the nucleus accumbens was increased 1 h following food presentation. While exhibiting weight loss, total food self-administration, enhanced motivation to self-administer food rewards in training sessions held during CR and c-Fos-activation in the nucleus accumbens following re-feeding were all markedly attenuated in Mc3rTB/TB mice. In contrast, cognitive abilities were normal in Mc3rTB/TB mice. Total food self-administration during FR1 sessions was not rescued in DAT-MC3R mice, however enhanced motivational responses to self-administer food rewards in PR1 conditions were restored. The nutrient-partitioning phenotype observed with Mc3r-deficiency was not rescued in DAT-MC3R mice. Conclusions Mesolimbic MC3Rs mediate enhanced motivational responses during CR. However, they are insufficient to restore normal caloric loading when food is presented during CR and do not affect metabolic conditions altering nutrient partitioning. Food-related motivational responses in mice increase with caloric restriction (CR). Melanocortin-3 receptors (MC3R) are required for food-related motivational responses. MC3Rs role in food-related motivational responses depends on metabolic condition. Mesolimbic MC3Rs increase food-related motivational responses during CR.
Collapse
Affiliation(s)
- Maria Mavrikaki
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL 33458, USA; Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Clemence Girardet
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL 33458, USA; Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Andras Kern
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Alicia Faruzzi Brantley
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA; Behavioral Core, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Courtney A Miller
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL 33458, USA; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Heather Macarthur
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Andrew A Butler
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL 33458, USA; Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA.
| |
Collapse
|
39
|
Anderson EJP, Çakir I, Carrington SJ, Cone RD, Ghamari-Langroudi M, Gillyard T, Gimenez LE, Litt MJ. 60 YEARS OF POMC: Regulation of feeding and energy homeostasis by α-MSH. J Mol Endocrinol 2016; 56:T157-74. [PMID: 26939593 PMCID: PMC5027135 DOI: 10.1530/jme-16-0014] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/01/2016] [Indexed: 12/20/2022]
Abstract
The melanocortin peptides derived from pro-opiomelanocortin (POMC) were originally understood in terms of the biological actions of α-melanocyte-stimulating hormone (α-MSH) on pigmentation and adrenocorticotrophic hormone on adrenocortical glucocorticoid production. However, the discovery of POMC mRNA and melanocortin peptides in the CNS generated activities directed at understanding the direct biological actions of melanocortins in the brain. Ultimately, discovery of unique melanocortin receptors expressed in the CNS, the melanocortin-3 (MC3R) and melanocortin-4 (MC4R) receptors, led to the development of pharmacological tools and genetic models leading to the demonstration that the central melanocortin system plays a critical role in the regulation of energy homeostasis. Indeed, mutations in MC4R are now known to be the most common cause of early onset syndromic obesity, accounting for 2-5% of all cases. This review discusses the history of these discoveries, as well as the latest work attempting to understand the molecular and cellular basis of regulation of feeding and energy homeostasis by the predominant melanocortin peptide in the CNS, α-MSH.
Collapse
Affiliation(s)
- Erica J P Anderson
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Isin Çakir
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Sheridan J Carrington
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Roger D Cone
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Masoud Ghamari-Langroudi
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Taneisha Gillyard
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA Meharry Medical CollegeDepartment of Neuroscience and Pharmacology, Nashville, Tennessee, USA
| | - Luis E Gimenez
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Michael J Litt
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
40
|
Abstract
Obesity is a disorder that develops from the interaction between genotype and environment involving social, behavioral, cultural, and physiological factors. Obesity increases the risk for type 2 diabetes mellitus, hypertension, cardiovascular disease, cancer, musculoskeletal disorders, chronic kidney and pulmonary disease. Although obesity is clearly associated with an increased prevalence of hypertension, many obese individuals may not develop hypertension. Protecting factors may exist and it is important to understand why obesity is not always related to hypertension. The aim of this review is to highlight the knowledge gap for the association between obesity, hypertension, and potential genetic and racial differences or environmental factors that may protect obese patients against the development of hypertension and other co-morbidities. Specific mutations in the leptin and the melaninocortin receptor genes in animal models of obesity without hypertension, the actions of α-melanocyte stimulating hormone, and SNS activity in obesity-related hypertension may promote recognition of protective and promoting factors for hypertension in obesity. Furthermore, gene-environment interactions may have the potential to modify gene expression and epigenetic mechanisms could also contribute to the heritability of obesity-induced hypertension. Finally, differences in nutrition, gut microbiota, exposure to sun light and exercise may play an important role in the presence or absence of hypertension in obesity.
Collapse
|
41
|
Mutations in Melanocortin-3 Receptor Gene and Human Obesity. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 140:97-129. [DOI: 10.1016/bs.pmbts.2016.01.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
42
|
Jackson DS, Ramachandrappa S, Clark AJ, Chan LF. Melanocortin receptor accessory proteins in adrenal disease and obesity. Front Neurosci 2015; 9:213. [PMID: 26113808 PMCID: PMC4461818 DOI: 10.3389/fnins.2015.00213] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 05/28/2015] [Indexed: 12/02/2022] Open
Abstract
Melanocortin receptor accessory proteins (MRAPs) are regulators of the melanocortin receptor family. MRAP is an essential accessory factor for the functional expression of the MC2R/ACTH receptor. The importance of MRAP in adrenal gland physiology is demonstrated by the clinical condition familial glucocorticoid deficiency type 2. The role of its paralog melanocortin-2-receptor accessory protein 2 (MRAP2), which is predominantly expressed in the hypothalamus including the paraventricular nucleus, has recently been linked to mammalian obesity. Whole body deletion and targeted brain specific deletion of the Mrap2 gene result in severe obesity in mice. Interestingly, Mrap2 complete knockout (KO) mice have increased body weight without detectable changes to food intake or energy expenditure. Rare heterozygous variants of MRAP2 have been found in humans with severe, early-onset obesity. In vitro data have shown that Mrap2 interaction with the melanocortin-4-receptor (Mc4r) affects receptor signaling. However, the mechanism by which Mrap2 regulates body weight in vivo is not fully understood and differences between the phenotypes of Mrap2 and Mc4r KO mice may point toward Mc4r independent mechanisms.
Collapse
Affiliation(s)
- David S Jackson
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London London, UK
| | - Shwetha Ramachandrappa
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London London, UK
| | - Adrian J Clark
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London London, UK
| | - Li F Chan
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London London, UK
| |
Collapse
|
43
|
Morgan DA, McDaniel LN, Yin T, Khan M, Jiang J, Acevedo MR, Walsh SA, Ponto LLB, Norris AW, Lutter M, Rahmouni K, Cui H. Regulation of glucose tolerance and sympathetic activity by MC4R signaling in the lateral hypothalamus. Diabetes 2015; 64:1976-87. [PMID: 25605803 PMCID: PMC4439564 DOI: 10.2337/db14-1257] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 01/10/2015] [Indexed: 02/06/2023]
Abstract
Melanocortin 4 receptor (MC4R) signaling mediates diverse physiological functions, including energy balance, glucose homeostasis, and autonomic activity. Although the lateral hypothalamic area (LHA) is known to express MC4Rs and to receive input from leptin-responsive arcuate proopiomelanocortin neurons, the physiological functions of MC4Rs in the LHA are incompletely understood. We report that MC4R(LHA) signaling regulates glucose tolerance and sympathetic nerve activity. Restoring expression of MC4Rs specifically in the LHA improves glucose intolerance in obese MC4R-null mice without affecting body weight or circulating insulin levels. Fluorodeoxyglucose-mediated tracing of whole-body glucose uptake identifies the interscapular brown adipose tissue (iBAT) as a primary source where glucose uptake is increased in MC4R(LHA) mice. Direct multifiber sympathetic nerve recording further reveals that sympathetic traffic to iBAT is significantly increased in MC4R(LHA) mice, which accompanies a significant elevation of Glut4 expression in iBAT. Finally, bilateral iBAT denervation prevents the glucoregulatory effect of MC4R(LHA) signaling. These results identify a novel role for MC4R(LHA) signaling in the control of sympathetic nerve activity and glucose tolerance independent of energy balance.
Collapse
Affiliation(s)
- Donald A Morgan
- Department of Pharmacology, University of Iowa, Carver College of Medicine, Iowa City, IA
| | - Latisha N McDaniel
- Department of Psychiatry, University of Iowa, Carver College of Medicine, Iowa City, IA
| | - Terry Yin
- Department of Psychiatry, University of Iowa, Carver College of Medicine, Iowa City, IA
| | - Michael Khan
- Department of Psychiatry, University of Iowa, Carver College of Medicine, Iowa City, IA
| | - Jingwei Jiang
- Department of Pharmacology, University of Iowa, Carver College of Medicine, Iowa City, IA
| | - Michael R Acevedo
- Small Animal Imaging Core, University of Iowa, Carver College of Medicine, Iowa City, IA
| | - Susan A Walsh
- Small Animal Imaging Core, University of Iowa, Carver College of Medicine, Iowa City, IA
| | - Laura L Boles Ponto
- Department of Radiology, University of Iowa, Carver College of Medicine, Iowa City, IA Department of Pediatrics, University of Iowa, Carver College of Medicine, Iowa City, IA
| | - Andrew W Norris
- Department of Pediatrics, University of Iowa, Carver College of Medicine, Iowa City, IA
| | - Michael Lutter
- Department of Psychiatry, University of Iowa, Carver College of Medicine, Iowa City, IA Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Carver College of Medicine, Iowa City, IA Obesity Research and Education Initiative, University of Iowa, Carver College of Medicine, Iowa City, IA
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa, Carver College of Medicine, Iowa City, IA Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Carver College of Medicine, Iowa City, IA Obesity Research and Education Initiative, University of Iowa, Carver College of Medicine, Iowa City, IA
| | - Huxing Cui
- Department of Pharmacology, University of Iowa, Carver College of Medicine, Iowa City, IA Department of Psychiatry, University of Iowa, Carver College of Medicine, Iowa City, IA Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Carver College of Medicine, Iowa City, IA
| |
Collapse
|
44
|
Lemus MB, Bayliss JA, Lockie SH, Santos VV, Reichenbach A, Stark R, Andrews ZB. A stereological analysis of NPY, POMC, Orexin, GFAP astrocyte, and Iba1 microglia cell number and volume in diet-induced obese male mice. Endocrinology 2015; 156:1701-13. [PMID: 25742051 DOI: 10.1210/en.2014-1961] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The hypothalamic arcuate nucleus (ARC) contains 2 key neural populations, neuropeptide Y (NPY) and proopiomelanocortin (POMC), and, together with orexin neurons in the lateral hypothalamus, plays an integral role in energy homeostasis. However, no studies have examined total neuronal number and volume after high-fat diet (HFD) exposure using sophisticated stereology. We used design-based stereology to estimate NPY and POMC neuronal number and volume, as well as glial fibrillary acidic protein (astrocyte marker) and ionized calcium-binding adapter molecule 1 (microglia marker) cell number in the ARC; as well as orexin neurons in the lateral hypothalamus. Stereological analysis indicated approximately 8000 NPY and approximately 9000 POMC neurons in the ARC, and approximately 7500 orexin neurons in the lateral hypothalamus. HFD exposure did not affect total neuronal number in any population. However, HFD significantly increased average NPY cell volume and affected NPY and POMC cell volume distribution. HFD reduced orexin cell volume but had a bimodal effect on volume distribution with increased cells at relatively small volumes and decreased cells with relatively large volumes. ARC glial fibrillary acidic protein cells increased after 2 months on a HFD, although no significant difference after 6 months on chow diet or HFD was observed. No differences in ARC ionized calcium-binding adapter molecule 1 cell number were observed in any group. Thus, HFD affects ARC NPY or POMC neuronal cell volume number not cell number. Our results demonstrate the importance of stereology to perform robust unbiased analysis of cell number and volume. These data should be an empirical baseline reference to which future studies are compared.
Collapse
Affiliation(s)
- Moyra B Lemus
- Department of Physiology, Monash University, Clayton, Victoria 3800, Australia
| | | | | | | | | | | | | |
Collapse
|
45
|
Growth Hormone Secretagogue Receptor Dimers: A New Pharmacological Target. eNeuro 2015; 2:eN-REV-0053-14. [PMID: 26464979 PMCID: PMC4596092 DOI: 10.1523/eneuro.0053-14.2015] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 03/25/2015] [Accepted: 04/07/2015] [Indexed: 01/04/2023] Open
Abstract
The growth hormone secretagogue receptor (GHSR1a), the target of the ghrelin peptide, is widely distributed throughout the brain, and, while studies have often reported very low or absent levels of central ghrelin, it is now known that GHSR1a, even in the absence of a natural ligand, has physiological roles. Not only do these roles originate from the receptor's constitutive activity, but recent data indicate that GHSR1a dimerizes with a wide array of other receptors. These include the dopamine 1 receptor (D1R), the dopamine 2 receptor (D2R), the melanocortin-3 receptor (MC3R), the serotonin 2C receptor (5-HT2C), and possibly the cannabinoid type 1 receptor (CB1). Within these dimers, signaling of the protomers involved are modified through facilitation, inhibition, and even modification of signaling pathways resulting in physiological consequences not seen in the absence of these dimers. While in some cases the ghrelin peptide is not required for these modifications to occur, in others, the presence is necessary for these changes to take effect. These heterodimers demonstrate the broad array of roles and complexity of the ghrelin system. By better understanding how these dimers work, it is hoped that improved treatments for a variety of disorders, including Parkinson's disease, schizophrenia, addiction, obesity, diabetes, and more, can be devised. In this review, we examine the current state of knowledge surrounding GHSR heterodimers, and how we can apply this knowledge to various pharmacological treatments.
Collapse
|
46
|
Hughey CC, Wasserman DH, Lee-Young RS, Lantier L. Approach to assessing determinants of glucose homeostasis in the conscious mouse. Mamm Genome 2014; 25:522-38. [PMID: 25074441 DOI: 10.1007/s00335-014-9533-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/04/2014] [Indexed: 01/11/2023]
Abstract
Obesity and type 2 diabetes lessen the quality of life of those afflicted and place considerable burden on the healthcare system. Furthermore, the detrimental impact of these pathologies is expected to persist or even worsen. Diabetes is characterized by impaired insulin action and glucose homeostasis. This has led to a rapid increase in the number of mouse models of metabolic disease being used in the basic sciences to assist in facilitating a greater understanding of the metabolic dysregulation associated with obesity and diabetes, the identification of therapeutic targets, and the discovery of effective treatments. This review briefly describes the most frequently utilized models of metabolic disease. A presentation of standard methods and technologies on the horizon for assessing metabolic phenotypes in mice, with particular emphasis on glucose handling and energy balance, is provided. The article also addresses issues related to study design, selection and execution of metabolic tests of glucose metabolism, the presentation of data, and interpretation of results.
Collapse
Affiliation(s)
- Curtis C Hughey
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, 823 Light Hall, 2215 Garland Ave, Nashville, TN, 37232, USA,
| | | | | | | |
Collapse
|
47
|
Girardet C, Begriche K, Ptitsyn A, Koza RA, Butler AA. Unravelling the mysterious roles of melanocortin-3 receptors in metabolic homeostasis and obesity using mouse genetics. INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2014; 4:S37-44. [PMID: 27152165 DOI: 10.1038/ijosup.2014.10] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The central nervous melanocortin system maintains body mass and adiposity within a 'healthy' range by regulating satiety and metabolic homeostasis. Neural melanocortin-4 receptors (MC4R) modulate satiety signals and regulate autonomic outputs governing glucose and lipid metabolism in the periphery. The functions of melanocortin-3 receptors (MC3R) have been less well defined. We have observed that food anticipatory activity (FAA) is attenuated in Mc3r-/- mice housed in light:dark or constant dark conditions. Mc3r-/- mice subjected to the restricted feeding protocol that was used to induce FAA also developed insulin resistance, dyslipidaemia, impaired glucose tolerance and evidence of a cellular stress response in the liver. MC3Rs may thus function as modulators of oscillator systems that govern circadian rhythms, integrating signals from nutrient sensors to facilitate synchronizing peak foraging behaviour and metabolic efficiency with nutrient availability. To dissect the functions of MC3Rs expressed in hypothalamic and extra-hypothalamic structures, we inserted a 'lox-stop-lox' (TB) sequence into the Mc3r gene. Mc3r (TB/TB) mice recapitulate the phenotype reported for Mc3r-/- mice: increased adiposity, accelerated diet-induced obesity and attenuated FAA. The ventromedial hypothalamus exhibits high levels of Mc3r expression; however, restoring the expression of the LoxTB Mc3r allele in this nucleus did not restore FAA. However, a surprising outcome came from studies using Nestin-Cre to restore the expression of the LoxTB Mc3r allele in the nervous system. These data suggest that 'non-neural' MC3Rs have a role in the defence of body weight. Future studies examining the homeostatic functions of MC3Rs should therefore consider actions outside the central nervous system.
Collapse
Affiliation(s)
- C Girardet
- Department of Metabolism and Aging, The Scripps Research Institute , Jupiter, FL, USA
| | - K Begriche
- Department of Metabolism and Aging, The Scripps Research Institute , Jupiter, FL, USA
| | - A Ptitsyn
- The Pennington Biomedical Research Center, Louisiana State University System , Baton Rouge, LA, USA
| | - R A Koza
- The Pennington Biomedical Research Center, Louisiana State University System , Baton Rouge, LA, USA
| | - A A Butler
- Department of Metabolism and Aging, The Scripps Research Institute , Jupiter, FL, USA
| |
Collapse
|
48
|
Seli E, Babayev E, Collins SC, Nemeth G, Horvath TL. Minireview: Metabolism of female reproduction: regulatory mechanisms and clinical implications. Mol Endocrinol 2014; 28:790-804. [PMID: 24678733 DOI: 10.1210/me.2013-1413] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Female fertility is highly dependent on successful regulation of energy metabolism. Central processes in the hypothalamus monitor the metabolic state of the organism and, together with metabolic hormones, drive the peripheral availability of energy for cellular functions. In the ovary, the oocyte and neighboring somatic cells of the follicle work in unison to achieve successful metabolism of carbohydrates, amino acids, and lipids. Metabolic disturbances such as anorexia nervosa, obesity, and diabetes mellitus have clinically important consequences on human reproduction. In this article, we review the metabolic determinants of female reproduction and their role in infertility.
Collapse
Affiliation(s)
- Emre Seli
- Department of Obstetrics, Gynecology, and Reproductive Sciences (E.S., E.B., S.C., T.L.H.), Yale School of Medicine, New Haven, Connecticut 06520; Department of Obstetrics and Gynecology (G.N., T.L.H.), University of Szeged, Faculty of Medicine, Szeged, Hungary 6701; Department of Comparative Medicine (T.L.H.), Yale School of Medicine, New Haven, Connecticut 06520; and the Department of Neurobiology (T.L.H.), Yale School of Medicine, New Haven, Connecticut 06520
| | | | | | | | | |
Collapse
|
49
|
Zorrilla EP, Conti B. Interleukin-18 null mutation increases weight and food intake and reduces energy expenditure and lipid substrate utilization in high-fat diet fed mice. Brain Behav Immun 2014; 37:45-53. [PMID: 24316258 PMCID: PMC4219830 DOI: 10.1016/j.bbi.2013.12.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 12/01/2013] [Accepted: 12/01/2013] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE The proinflammatory cytokine interleukin-18 (IL-18) putatively modulates food intake and energy metabolism, but the effects of IL-18 in high-fat diet fed animals are unknown. Whether IL-18 alters basal metabolic rate or metabolic processes of living is unknown. Here, we tested the hypothesis that IL-18 modulates weight gain, energy intake, whole-body energy expenditure, and utilization of lipid as a fuel substrate in high-fat diet fed mice. METHODS Food intake, whole-body metabolism, and motor activity of IL-18 knockout mice were compared to those of wildtype littermates; anorectic effects of intracerebroventricular IL-18 administration were compared between IL-18 receptor knockout, IL-18/IL-18R knockout and wildtype mice. RESULTS Chow-reared IL-18 knockout mice were overweight at 6 months of age and then gained excess weight on both low-fat and high-fat diets, ate more high-fat diet, and showed reduced whole-body energy expenditure and increased respiratory exchange ratios. Reductions in energy expenditure of IL-18 knockout mice were seen across fasting vs. feeding conditions, low- vs. high-fat diets, high vs. low levels of physical activity and times of day, suggesting actions on basal metabolic rate. The circadian amplitude of energy expenditure, but not respiratory exchange ratio, food intake, or motor activity, also was blunted in IL-18 knockout mice. Central IL-18 administration reduced high-fat diet intake in wildtype mice, but not in mice lacking the IL-18 receptor. CONCLUSION The loss-of-function results support the hypothesis that endogenous IL-18 suppresses appetite and promote energy expenditure and lipid fuel substrate utilization not only during sickness, but also in healthy adults consuming high-fat diets.
Collapse
Affiliation(s)
- Eric P. Zorrilla
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, 10550 N. Torrey Pines Rd. La Jolla, California, 92037, USA,Molecular and Integrative Neurosciences Department, The Scripps Research Institute, 10550 N. Torrey Pines Rd. La Jolla, California, 92037, USA
| | - Bruno Conti
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, 10550 N. Torrey Pines Rd. La Jolla, California, 92037, USA
| |
Collapse
|
50
|
Khalifeh-Soltani A, McKleroy W, Sakuma S, Cheung YY, Tharp K, Qiu Y, Turner SM, Chawla A, Stahl A, Atabai K. Mfge8 promotes obesity by mediating the uptake of dietary fats and serum fatty acids. Nat Med 2014; 20:175-83. [PMID: 24441829 DOI: 10.1038/nm.3450] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/11/2013] [Indexed: 12/14/2022]
Abstract
Fatty acids are integral mediators of energy storage, membrane formation and cell signaling. The pathways that orchestrate uptake of fatty acids remain incompletely understood. Expression of the integrin ligand Mfge8 is increased in human obesity and in mice on a high-fat diet, but its role in obesity is unknown. We show here that Mfge8 promotes the absorption of dietary triglycerides and the cellular uptake of fatty acid and that Mfge8-deficient (Mfge8(-/-)) mice are protected from diet-induced obesity, steatohepatitis and insulin resistance. Mechanistically, we found that Mfge8 coordinates fatty acid uptake through αvβ3 integrin- and αvβ5 integrin-dependent phosphorylation of Akt by phosphatidylinositide-3 kinase and mTOR complex 2, leading to translocation of Cd36 and Fatp1 from cytoplasmic vesicles to the cell surface. Collectively, our results imply a role for Mfge8 in regulating the absorption and storage of dietary fats, as well as in the development of obesity and its complications.
Collapse
Affiliation(s)
- Amin Khalifeh-Soltani
- 1] Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA. [2] Lung Biology Center, University of California, San Francisco, San Francisco, California, USA. [3] Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - William McKleroy
- 1] Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA. [2] Lung Biology Center, University of California, San Francisco, San Francisco, California, USA. [3] Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Stephen Sakuma
- 1] Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA. [2] Lung Biology Center, University of California, San Francisco, San Francisco, California, USA. [3] Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Yuk Yin Cheung
- 1] Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA. [2] Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Kevin Tharp
- 1] Metabolic Biology, University of California, Berkeley, Berkeley, California, USA. [2] Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, California, USA
| | - Yifu Qiu
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | | | - Ajay Chawla
- 1] Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA. [2] Department of Medicine, University of California, San Francisco, San Francisco, California, USA. [3] Department of Physiology, University of California, San Francisco, San Francisco, California, USA
| | - Andreas Stahl
- 1] Metabolic Biology, University of California, Berkeley, Berkeley, California, USA. [2] Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, California, USA
| | - Kamran Atabai
- 1] Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA. [2] Lung Biology Center, University of California, San Francisco, San Francisco, California, USA. [3] Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|