1
|
Sun X, Liu B, Yuan Y, Rong Y, Pang R, Li Q. Neural and hormonal mechanisms of appetite regulation during eating. Front Nutr 2025; 12:1484827. [PMID: 40201582 PMCID: PMC11977392 DOI: 10.3389/fnut.2025.1484827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Numerous animal and clinical studies have demonstrated that the arcuate nucleus of the hypothalamus, a central regulator of appetite, plays a significant role in modulating feeding behavior. However, current research primarily focuses on long-term dietary changes and their effects on the body, with limited investigation into neuroendocrine dynamics during individual meals across diverse populations. In contrast to long-term dietary adjustments, directives for dietary behavior during a specific meal are more actionable, potentially enhancing patient adherence and achieving better outcomes in dietary behavior interventions. This review aimed to explore the neural pathways and endocrine changes activated by gastrointestinal expansion and variations in blood nutrient levels during a single meal, with the goal of informing dietary behavior guidance.
Collapse
Affiliation(s)
- Xurui Sun
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Binghan Liu
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuan Yuan
- Department of Clinical Nutrition, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ying Rong
- Department of Clinical Nutrition, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Rui Pang
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qiu Li
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
2
|
Guo X, Asthana P, Zhai L, Cheng KW, Gurung S, Huang J, Wu J, Zhang Y, Mahato AK, Saarma M, Ustav M, Kwan HY, Lyu A, Chan KM, Xu P, Bian ZX, Wong HLX. Artesunate treats obesity in male mice and non-human primates through GDF15/GFRAL signalling axis. Nat Commun 2024; 15:1034. [PMID: 38310105 PMCID: PMC10838268 DOI: 10.1038/s41467-024-45452-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/23/2024] [Indexed: 02/05/2024] Open
Abstract
Obesity, a global health challenge, is a major risk factor for multiple life-threatening diseases, including diabetes, fatty liver, and cancer. There is an ongoing need to identify safe and tolerable therapeutics for obesity management. Herein, we show that treatment with artesunate, an artemisinin derivative approved by the FDA for the treatment of severe malaria, effectively reduces body weight and improves metabolic profiles in preclinical models of obesity, including male mice with overnutrition-induced obesity and male cynomolgus macaques with spontaneous obesity, without inducing nausea and malaise. Artesunate promotes weight loss and reduces food intake in obese mice and cynomolgus macaques by increasing circulating levels of Growth Differentiation Factor 15 (GDF15), an appetite-regulating hormone with a brainstem-restricted receptor, the GDNF family receptor α-like (GFRAL). Mechanistically, artesunate induces the expression of GDF15 in multiple organs, especially the liver, in mice through a C/EBP homologous protein (CHOP)-directed integrated stress response. Inhibition of GDF15/GFRAL signalling by genetic ablation of GFRAL or tissue-specific knockdown of GDF15 abrogates the anti-obesity effect of artesunate in mice with diet-induced obesity, suggesting that artesunate controls bodyweight and appetite in a GDF15/GFRAL signalling-dependent manner. These data highlight the therapeutic benefits of artesunate in the treatment of obesity and related comorbidities.
Collapse
Affiliation(s)
- Xuanming Guo
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Pallavi Asthana
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| | - Lixiang Zhai
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ka Wing Cheng
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Centre for Chinese Herbal Medicine Drug Development Limited, Hong Kong Baptist University, Hong Kong SAR, China
| | - Susma Gurung
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jiangang Huang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Jiayan Wu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yijing Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Arun Kumar Mahato
- Institute of Biotechnology-HILIFE, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology-HILIFE, University of Helsinki, Helsinki, Finland
| | | | - Hiu Yee Kwan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Aiping Lyu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Kui Ming Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Pingyi Xu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhao-Xiang Bian
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Centre for Chinese Herbal Medicine Drug Development Limited, Hong Kong Baptist University, Hong Kong SAR, China.
| | | |
Collapse
|
3
|
Hammerschmidt P, Steculorum SM, Bandet CL, Del Río-Martín A, Steuernagel L, Kohlhaas V, Feldmann M, Varela L, Majcher A, Quatorze Correia M, Klar RFU, Bauder CA, Kaya E, Porniece M, Biglari N, Sieben A, Horvath TL, Hornemann T, Brodesser S, Brüning JC. CerS6-dependent ceramide synthesis in hypothalamic neurons promotes ER/mitochondrial stress and impairs glucose homeostasis in obese mice. Nat Commun 2023; 14:7824. [PMID: 38016943 PMCID: PMC10684560 DOI: 10.1038/s41467-023-42595-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/17/2023] [Indexed: 11/30/2023] Open
Abstract
Dysregulation of hypothalamic ceramides has been associated with disrupted neuronal pathways in control of energy and glucose homeostasis. However, the specific ceramide species promoting neuronal lipotoxicity in obesity have remained obscure. Here, we find increased expression of the C16:0 ceramide-producing ceramide synthase (CerS)6 in cultured hypothalamic neurons exposed to palmitate in vitro and in the hypothalamus of obese mice. Conditional deletion of CerS6 in hypothalamic neurons attenuates high-fat diet (HFD)-dependent weight gain and improves glucose metabolism. Specifically, CerS6 deficiency in neurons expressing pro-opiomelanocortin (POMC) or steroidogenic factor 1 (SF-1) alters feeding behavior and alleviates the adverse metabolic effects of HFD feeding on insulin sensitivity and glucose tolerance. POMC-expressing cell-selective deletion of CerS6 prevents the diet-induced alterations of mitochondrial morphology and improves cellular leptin sensitivity. Our experiments reveal functions of CerS6-derived ceramides in hypothalamic lipotoxicity, altered mitochondrial dynamics, and ER/mitochondrial stress in the deregulation of food intake and glucose metabolism in obesity.
Collapse
Affiliation(s)
- Philipp Hammerschmidt
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Sophie M Steculorum
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Max Planck Institute for Metabolism Research, Research Group Neurocircuit Wiring and Function, Cologne, Germany
- National Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Cécile L Bandet
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Almudena Del Río-Martín
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Vivien Kohlhaas
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Marvin Feldmann
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Luis Varela
- Yale Center for Molecular and Systems Metabolism, Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., BML 330, New Haven, CT, 06520, USA
- Laboratory of Glia-Neuron Interactions in the Control of Hunger. Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain
- Ikerbasque-Basque Foundation for Science, Bilbao, 48013, Spain
| | - Adam Majcher
- Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland
- Institute of Clinical Chemistry, University Hospital, Zürich, Switzerland
| | - Marta Quatorze Correia
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
| | - Rhena F U Klar
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
| | - Corinna A Bauder
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Ecem Kaya
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Marta Porniece
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Nasim Biglari
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Anna Sieben
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Tamas L Horvath
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Yale Center for Molecular and Systems Metabolism, Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., BML 330, New Haven, CT, 06520, USA
- Laboratory of Glia-Neuron Interactions in the Control of Hunger. Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain
- Ikerbasque-Basque Foundation for Science, Bilbao, 48013, Spain
| | - Thorsten Hornemann
- Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland
- Institute of Clinical Chemistry, University Hospital, Zürich, Switzerland
| | - Susanne Brodesser
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
- National Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany.
| |
Collapse
|
4
|
Rupp AC, Tomlinson AJ, Affinati AH, Yacawych WT, Duensing AM, True C, Lindsley SR, Kirigiti MA, MacKenzie A, Polex-Wolf J, Li C, Knudsen LB, Seeley RJ, Olson DP, Kievit P, Myers MG. Suppression of food intake by Glp1r/Lepr-coexpressing neurons prevents obesity in mouse models. J Clin Invest 2023; 133:e157515. [PMID: 37581939 PMCID: PMC10541203 DOI: 10.1172/jci157515] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 08/03/2023] [Indexed: 08/17/2023] Open
Abstract
The adipose-derived hormone leptin acts via its receptor (LepRb) in the brain to control energy balance. A potentially unidentified population of GABAergic hypothalamic LepRb neurons plays key roles in the restraint of food intake and body weight by leptin. To identify markers for candidate populations of LepRb neurons in an unbiased manner, we performed single-nucleus RNA-Seq of enriched mouse hypothalamic LepRb cells, identifying several previously unrecognized populations of hypothalamic LepRb neurons. Many of these populations displayed strong conservation across species, including GABAergic Glp1r-expressing LepRb (LepRbGlp1r) neurons, which expressed more Lepr than other LepRb cell populations. Ablating Lepr from LepRbGlp1r cells provoked hyperphagic obesity without impairing energy expenditure. Similarly, improvements in energy balance caused by Lepr reactivation in GABA neurons of otherwise Lepr-null mice required Lepr expression in GABAergic Glp1r-expressing neurons. Furthermore, restoration of Glp1r expression in LepRbGlp1r neurons in otherwise Glp1r-null mice enabled food intake suppression by the GLP1R agonist, liraglutide. Thus, the conserved GABAergic LepRbGlp1r neuron population plays crucial roles in the suppression of food intake by leptin and GLP1R agonists.
Collapse
Affiliation(s)
| | | | | | - Warren T. Yacawych
- Department of Internal Medicine and
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Allison M. Duensing
- Department of Internal Medicine and
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Cadence True
- Oregon National Primate Research Center, Beaverton, Oregon, USA
| | | | | | | | | | - Chien Li
- Novo Nordisk, Copenhagen, Denmark
| | | | | | - David P. Olson
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Paul Kievit
- Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Martin G. Myers
- Department of Internal Medicine and
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
5
|
Lee NJ, Oraha J, Qi Y, Enriquez RF, Tasan R, Herzog H. Altered function of arcuate leptin receptor expressing neuropeptide Y neurons depending on energy balance. Mol Metab 2023; 76:101790. [PMID: 37562743 PMCID: PMC10460992 DOI: 10.1016/j.molmet.2023.101790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 08/12/2023] Open
Abstract
OBJECTIVE One of leptin's main targets in the hypothalamus are neuropeptide Y (NPY) neurons, with selective deletion of leptin receptors (Lepr) specifically in Npy neurons resulting in major alterations of energy partitioning between fat and bone mass. However, the specific action of these Npy+/Lepr+ neurons compared to Npy-negative Lepr (Npy-/Lepr+) neurons in regard to energy homeostasis regulation is unknown. METHODS Specific AAV viral vectors were generated using DREADD and INTRSECT technology and used in male LeprCre/+ and LeprCre/+;NpyFlp/+ mice to assess the effect of activating either all Lepr neurons or specifically Npy+/Lepr+ or Npy-/Lepr+ neurons only on feeding, energy homeostasis control, and body composition. RESULTS Selective stimulation of Npy+/Lepr+ neurons led to an immediate decrease in respiratory quotient followed by a delayed increase in food intake in standard chow fed, but interestingly not in high fat diet (HFD) fed mice. In addition, stimulation of Npy+/Lepr+ neurons led to a robust increase in brown adipose tissue thermogenesis and improved glucose tolerance. These effects were not observed in standard chow fed mice when Npy-/Lepr+ expressing neurons were specifically activated, suggesting the effects of leptin on these parameters are driven by NPY. However, under HFD condition when leptin levels are elevated, the stimulation of the Npy-/Lepr+ neurons increased food intake, physical activity and energy expenditure. Interestingly, chronic stimulation of Npy-positive Lepr neurons was able to increase bone mass independently of bodyweight, whilst chronic stimulation of the Npy-/Lepr+ neurons resulted in increased bodyweight and fat mass with proportionate increases in bone mass. CONCLUSIONS Together, these data indicate that leptin signalling through Npy-positive Lepr-expressing neurons controls energy partitioning via stimulation of thermogenesis, energy expenditure, and the use of fat as a fuel source. However, under prolonged HFD, leptin resistance may occur and actions of leptin signalling through Npy-negative Lepr hypothalamic neurons may exacerbate excess food intake.
Collapse
Affiliation(s)
- Nicola J Lee
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Garvan Institute of Medical Research, NSW, Australia; St Vincent's Clinical School, UNSW Sydney, NSW, Australia.
| | - Jennifer Oraha
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Garvan Institute of Medical Research, NSW, Australia
| | - Yue Qi
- Garvan Institute of Medical Research, NSW, Australia
| | | | - Ramon Tasan
- Institute of Pharmacology, University of Innsbruck, Austria
| | - Herbert Herzog
- Garvan Institute of Medical Research, NSW, Australia; St Vincent's Clinical School, UNSW Sydney, NSW, Australia
| |
Collapse
|
6
|
Mancini M, Patel JC, Affinati AH, Witkovsky P, Rice ME. Leptin Promotes Striatal Dopamine Release via Cholinergic Interneurons and Regionally Distinct Signaling Pathways. J Neurosci 2022; 42:6668-6679. [PMID: 35906070 PMCID: PMC9436012 DOI: 10.1523/jneurosci.0238-22.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 07/06/2022] [Accepted: 07/14/2022] [Indexed: 11/21/2022] Open
Abstract
Dopamine (DA) is a critical regulator of striatal network activity and is essential for motor activation and reward-associated behaviors. Previous work has shown that DA is influenced by the reward value of food, as well as by hormonal factors that reguate food intake and energy expenditure. Changes in striatal DA signaling also have been linked to aberrant eating patterns. Here we test the effect of leptin, an adipocyte-derived hormone involved in feeding and energy homeostasis regulation, on striatal DA release and uptake. Immunohistochemical evaluation identified leptin receptor (LepR) expression throughout mouse striatum, including on striatal cholinergic interneurons (ChIs) and their extensive processes. Using fast-scan cyclic voltammetry (FSCV), we found that leptin causes a concentration-dependent increase in evoked extra-cellular DA concentration ([DA]o) in dorsal striatum (dStr) and nucleus accumbens (NAc) core and shell in male mouse striatal slices, and also an increase in the rate of DA uptake. Further, we found that leptin increases ChI excitability, and that the enhancing effect of leptin on evoked [DA]o is lost when nicotinic acetylcholine (ACh) receptors are antagonized or when examined in striatal slices from mice lacking ACh synthesis. Evaluation of signaling pathways underlying leptin's action revealed a requirement for intracellular Ca2+, and the involvement of different downstream pathways in dStr and NAc core versus NAc shell. These results provide the first evidence for dynamic regulation of DA release and uptake by leptin within brain motor and reward pathways, and highlight the involvement of ChIs in this process.SIGNIFICANCE STATEMENT Given the importance of striatal dopamine (DA) in reward, motivation, motor behavior and food intake, identifying the actions of metabolic hormones on DA release in striatal subregions should provide new insight into factors that influence DA-dependent motivated behaviors. We find that one of these hormones, leptin, boosts striatal DA release through a process involving striatal cholinergic interneurons (ChIs) and nicotinic acetylcholine (ACh) receptors. Moreover, we find that the intracellular cascades downstream from leptin receptor (LepR) activation that lead to enhanced DA release differ among striatal subregions. Thus, we not only show that leptin regulates DA release, but also identify characteristics of this process that could be harnessed to alter pathologic eating behaviors.
Collapse
Affiliation(s)
- Maria Mancini
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, New York 10016
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York 10016
| | - Jyoti C Patel
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, New York 10016
| | - Alison H Affinati
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - Paul Witkovsky
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, New York 10016
| | - Margaret E Rice
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, New York 10016
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York 10016
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, New York 10016
| |
Collapse
|
7
|
Lockridge A, Hanover JA. A nexus of lipid and O-Glcnac metabolism in physiology and disease. Front Endocrinol (Lausanne) 2022; 13:943576. [PMID: 36111295 PMCID: PMC9468787 DOI: 10.3389/fendo.2022.943576] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Although traditionally considered a glucose metabolism-associated modification, the O-linked β-N-Acetylglucosamine (O-GlcNAc) regulatory system interacts extensively with lipids and is required to maintain lipid homeostasis. The enzymes of O-GlcNAc cycling have molecular properties consistent with those expected of broad-spectrum environmental sensors. By direct protein-protein interactions and catalytic modification, O-GlcNAc cycling enzymes may provide both acute and long-term adaptation to stress and other environmental stimuli such as nutrient availability. Depending on the cell type, hyperlipidemia potentiates or depresses O-GlcNAc levels, sometimes biphasically, through a diversity of unique mechanisms that target UDP-GlcNAc synthesis and the availability, activity and substrate selectivity of the glycosylation enzymes, O-GlcNAc Transferase (OGT) and O-GlcNAcase (OGA). At the same time, OGT activity in multiple tissues has been implicated in the homeostatic regulation of systemic lipid uptake, storage and release. Hyperlipidemic patterns of O-GlcNAcylation in these cells are consistent with both transient physiological adaptation and feedback uninhibited obesogenic and metabolic dysregulation. In this review, we summarize the numerous interconnections between lipid and O-GlcNAc metabolism. These links provide insights into how the O-GlcNAc regulatory system may contribute to lipid-associated diseases including obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Amber Lockridge
- Laboratory of Cell and Molecular Biology, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - John A. Hanover
- Laboratory of Cell and Molecular Biology, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
8
|
Cheng W, Gordian D, Ludwig MQ, Pers TH, Seeley RJ, Myers MG. Hindbrain circuits in the control of eating behaviour and energy balance. Nat Metab 2022; 4:826-835. [PMID: 35879458 DOI: 10.1038/s42255-022-00606-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Abstract
Body weight and adiposity represent biologically controlled parameters that are influenced by a combination of genetic, developmental and environmental variables. Although the hypothalamus plays a crucial role in matching caloric intake with energy expenditure to achieve a stable body weight, it is now recognized that neuronal circuits in the hindbrain not only serve to produce nausea and to terminate feeding in response to food consumption or during pathological states, but also contribute to the long-term control of body weight. Additionally, recent work has identified hindbrain neurons that are capable of suppressing food intake without producing aversive responses like those associated with nausea. Here we review recent advances in our understanding of the hindbrain neurons that control feeding, particularly those located in the area postrema and the nucleus tractus solitarius. We frame this information in the context of new atlases of hindbrain neuronal populations and develop a model of the hindbrain circuits that control food intake and energy balance, suggesting important areas for additional research.
Collapse
Affiliation(s)
- Wenwen Cheng
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Desiree Gordian
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA
| | - Mette Q Ludwig
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Tune H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Martin G Myers
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
9
|
Kaneko K, Takekuma Y, Goto T, Ohinata K. An orally active plant Rubisco-derived peptide increases neuronal leptin responsiveness. Sci Rep 2022; 12:8599. [PMID: 35597815 PMCID: PMC9124197 DOI: 10.1038/s41598-022-12595-6] [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] [Received: 01/20/2022] [Accepted: 05/13/2022] [Indexed: 11/25/2022] Open
Abstract
Nutrient excess, such as the intake of a high-fat diet, reduces hypothalamic responses to exogenously administered leptin and induces dietary obesity; however, orally active components that attenuate neural leptin dysregulation have yet to be identified. We herein demonstrated that YHIEPV, derived from the pepsin-pancreatin digestion of the green leaf protein Rubisco, increased the leptin-induced phosphorylation of STAT3 in ex vivo hypothalamic slice cultures. We also showed that YHIEPV mitigated palmitic acid-induced decreases in leptin responsiveness. Furthermore, orally administered YHIEPV promoted leptin-induced reductions in body weight and food intake in obese mice. In addition, dietary-induced body weight gain was significantly less in mice orally or centrally administered YHIEPV daily than in saline-control mice. Cellular leptin sensitivity and the levels of proinflammatory-related factors, such as IL1β and Socs-3, in the hypothalamus of obese mice were also restored by YHIEPV. YHIEPV blocked cellular leptin resistance induced by forskolin, which activates Epac-Rap1 signaling, and reduced the level of the GTP-bound active form of Rap1 in the brains of obese mice. Collectively, the present results demonstrated that the orally active peptide YHIEPV derived from a major green leaf protein increased neural leptin responsiveness and reduced body weight gain in mice with dietary obesity.
Collapse
Affiliation(s)
- Kentaro Kaneko
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Gokasho, Uji, Kyoto, 611-0011, Japan. .,Department of Agricultural Chemistry, School of Agriculture, Meiji University, 1-1-1, Higashimita, Tama-ku, Kawasaki-shi, Kanagawa, 214-8571, Japan.
| | - Yukihiro Takekuma
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Gokasho, Uji, Kyoto, 611-0011, Japan
| | - Tsuyoshi Goto
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Gokasho, Uji, Kyoto, 611-0011, Japan
| | - Kousaku Ohinata
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Gokasho, Uji, Kyoto, 611-0011, Japan.
| |
Collapse
|
10
|
Badini I, Coleman JR, Hagenaars SP, Hotopf M, Breen G, Lewis CM, Fabbri C. Depression with atypical neurovegetative symptoms shares genetic predisposition with immuno-metabolic traits and alcohol consumption. Psychol Med 2022; 52:726-736. [PMID: 32624019 PMCID: PMC8961332 DOI: 10.1017/s0033291720002342] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/03/2020] [Accepted: 06/12/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Depression is a highly prevalent and heterogeneous disorder. This study aims to determine whether depression with atypical features shows different heritability and different degree of overlap with polygenic risk for psychiatric and immuno-metabolic traits than other depression subgroups. METHODS Data included 30 069 European ancestry individuals from the UK Biobank who met criteria for lifetime major depression. Participants reporting both weight gain and hypersomnia were classified as ↑WS depression (N = 1854) and the others as non-↑WS depression (N = 28 215). Cases with non-↑WS depression were further classified as ↓WS depression (i.e. weight loss and insomnia; N = 10 142). Polygenic risk scores (PRS) for 22 traits were generated using genome-wide summary statistics (Bonferroni corrected p = 2.1 × 10-4). Single-nucleotide polymorphism (SNP)-based heritability of depression subgroups was estimated. RESULTS ↑WS depression had a higher polygenic risk for BMI [OR = 1.20 (1.15-1.26), p = 2.37 × 10-14] and C-reactive protein [OR = 1.11 (1.06-1.17), p = 8.86 × 10-06] v. non-↑WS depression and ↓WS depression. Leptin PRS was close to the significance threshold (p = 2.99 × 10-04), but the effect disappeared when considering GWAS summary statistics of leptin adjusted for BMI. PRS for daily alcohol use was inversely associated with ↑WS depression [OR = 0.88 (0.83-0.93), p = 1.04 × 10-05] v. non-↑WS depression. SNP-based heritability was not significantly different between ↑WS depression and ↓WS depression (14.3% and 12.2%, respectively). CONCLUSIONS ↑WS depression shows evidence of distinct genetic predisposition to immune-metabolic traits and alcohol consumption. These genetic signals suggest that biological targets including immune-cardio-metabolic pathways may be relevant to therapies in individuals with ↑WS depression.
Collapse
Affiliation(s)
- Isabella Badini
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Jonathan R.I. Coleman
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- National Institute for Health Research (NIHR) Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Saskia P. Hagenaars
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Matthew Hotopf
- National Institute for Health Research (NIHR) Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
| | - Gerome Breen
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- National Institute for Health Research (NIHR) Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Cathryn M. Lewis
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- National Institute for Health Research (NIHR) Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Chiara Fabbri
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
11
|
Maffei M, Giordano A. Leptin, the brain and energy homeostasis: From an apparently simple to a highly complex neuronal system. Rev Endocr Metab Disord 2022; 23:87-101. [PMID: 33822303 DOI: 10.1007/s11154-021-09636-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2021] [Indexed: 12/14/2022]
Abstract
Leptin, produced and secreted by white adipose tissue in tight relationship with adipose mass, informs the brain about the status of the energy stores serving as the main peripheral signal for energy balance regulation through interaction with a multitude of highly interconnected neuronal populations. Most obese patients display resistance to the anorectic effect of the hormone. The present review unravels the multiple levels of complexity that trigger hypothalamic response to leptin with the objective of highlighting those critical hubs that, mainly in the hypothalamic arcuate nucleus, may undergo obesity-induced alterations and create an obstacle to leptin action. Several mechanisms underlying leptin resistance have been proposed, possibly representing useful targets to empower leptin effects. Among these, a special focus is herein dedicated to detail how leptin gains access into the brain and how neuronal plasticity may interfere with leptin function.
Collapse
Affiliation(s)
- Margherita Maffei
- Institute of Clinical Physiology, CNR, Via Moruzzi 1, 56124, Pisa, Italy.
- Obesity and Lipodystrophy Center, University Hospital of Pisa, Via Paradisa 2, 56124, Pisa, Italy.
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020, Ancona, Italy.
| |
Collapse
|
12
|
Quaresma PGF, Wasinski F, Mansano NS, Furigo IC, Teixeira PDS, Gusmao DO, Frazao R, Donato J. Leptin Receptor Expression in GABAergic Cells is Not Sufficient to Normalize Metabolism and Reproduction in Mice. Endocrinology 2021; 162:6353267. [PMID: 34402859 DOI: 10.1210/endocr/bqab168] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Indexed: 12/12/2022]
Abstract
Previous studies indicate that leptin receptor (LepR) expression in GABAergic neurons is necessary for the biological effects of leptin. However, it is not clear whether LepR expression only in GABAergic neurons is sufficient to prevent the metabolic and neuroendocrine imbalances caused by LepR deficiency. In the present study, we produced mice that express the LepR exclusively in GABAergic cells (LepRVGAT mice) and compared them with wild-type (LepR+/+) and LepR-deficient (LepRNull/Null) mice. Although LepRVGAT mice showed a pronounced reduction in body weight and fat mass, as compared with LepRNull/Null mice, male and female LepRVGAT mice exhibited an obese phenotype relative to LepR+/+ mice. Food intake was normalized in LepRVGAT mice; however, LepRVGAT mice still exhibited lower energy expenditure in both sexes and reduced ambulatory activity in the females, compared with LepR+/+ mice. The acute anorexigenic effect of leptin and hedonic feeding were normalized in LepRVGAT mice despite the hyperleptinemia they present. Although LepRVGAT mice showed improved glucose homeostasis compared with LepRNull/Null mice, both male and female LepRVGAT mice exhibited insulin resistance. In contrast, LepR expression only in GABAergic cells was sufficient to normalize the density of agouti-related peptide (AgRP) and α-MSH immunoreactive fibers in the paraventricular nucleus of the hypothalamus. However, LepRVGAT mice exhibited reproductive dysfunctions, including subfertility in males and alterations in the estrous cycle of females. Taken together, our findings indicate that LepR expression in GABAergic cells, although critical to the physiology of leptin, is insufficient to normalize several metabolic aspects and the reproductive function in mice.
Collapse
Affiliation(s)
- Paula G F Quaresma
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| | - Frederick Wasinski
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| | - Naira S Mansano
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Isadora C Furigo
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| | - Pryscila D S Teixeira
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| | - Daniela O Gusmao
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| | - Renata Frazao
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Jose Donato
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| |
Collapse
|
13
|
Kaneko K. Appetite regulation by plant-derived bioactive peptides for promoting health. Peptides 2021; 144:170608. [PMID: 34265369 DOI: 10.1016/j.peptides.2021.170608] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/20/2021] [Accepted: 06/28/2021] [Indexed: 11/30/2022]
Abstract
Appetite is closely regulated not only by gut hormonal and neuronal peptides but also by exogenous peptides derived from food proteins. Food proteins are now recognized to contain many thousands of bioactive compounds that provide additional health benefits beyond their nutritional effects. Bioactive peptides are beneficial to the life and/or to regulate physiological functions. Although animal protein products have been widely applied in the food industry, exploring the possibilities of developing functional foods based on plant protein-derived peptides is considered attractive for achieving sustainable development goals. In addition, peptides from plant proteins have the potential to treat numerous diseases or risk factors and may therefore facilitate a healthy life expectancy. In this review, we discuss the identified plant-based bioactive peptides and their appetite regulating effects. Plant-based bioactive peptides may provide new opportunities to discover novel approaches that can improve and prevent diseases in a sustainable environment.
Collapse
Affiliation(s)
- Kentaro Kaneko
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Gokasho, Uji, Kyoto 611-0011, Japan.
| |
Collapse
|
14
|
Butiaeva LI, Slutzki T, Swick HE, Bourguignon C, Robins SC, Liu X, Storch KF, Kokoeva MV. Leptin receptor-expressing pericytes mediate access of hypothalamic feeding centers to circulating leptin. Cell Metab 2021; 33:1433-1448.e5. [PMID: 34129812 DOI: 10.1016/j.cmet.2021.05.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/19/2021] [Accepted: 05/21/2021] [Indexed: 12/18/2022]
Abstract
Knowledge of how leptin receptor (LepR) neurons of the mediobasal hypothalamus (MBH) access circulating leptin is still rudimentary. Employing intravital microscopy, we found that almost half of the blood-vessel-enwrapping pericytes in the MBH express LepR. Selective disruption of pericytic LepR led to increased food intake, increased fat mass, and loss of leptin-dependent signaling in nearby LepR neurons. When delivered intravenously, fluorescently tagged leptin accumulated at hypothalamic LepR pericytes, which was attenuated upon pericyte-specific LepR loss. Because a paracellular tracer was also preferentially retained at LepR pericytes, we pharmacologically targeted regulators of inter-endothelial junction tightness and found that they affect LepR neuronal signaling and food intake. Optical imaging in MBH slices revealed a long-lasting, tonic calcium increase in LepR pericytes in response to leptin, suggesting pericytic contraction and vessel constriction. Together, our data indicate that LepR pericytes facilitate localized, paracellular blood-brain barrier leaks, enabling MBH LepR neurons to access circulating leptin.
Collapse
Affiliation(s)
- Liliia I Butiaeva
- Division of Endocrinology, Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal QC H4A 3J1, Canada; Integrated Program in Neuroscience, McGill University, Montreal QC H3A 2B4, Canada
| | - Tal Slutzki
- Division of Endocrinology, Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal QC H4A 3J1, Canada; Integrated Program in Neuroscience, McGill University, Montreal QC H3A 2B4, Canada
| | - Hannah E Swick
- Division of Endocrinology, Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal QC H4A 3J1, Canada; Integrated Program in Neuroscience, McGill University, Montreal QC H3A 2B4, Canada
| | - Clément Bourguignon
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal QC H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Montreal QC H3A 2B4, Canada
| | - Sarah C Robins
- Division of Endocrinology, Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal QC H4A 3J1, Canada
| | - Xiaohong Liu
- Division of Endocrinology, Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal QC H4A 3J1, Canada
| | - Kai-Florian Storch
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal QC H4H 1R3, Canada
| | - Maia V Kokoeva
- Division of Endocrinology, Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal QC H4A 3J1, Canada.
| |
Collapse
|
15
|
Fatty acids role on obesity induced hypothalamus inflammation: From problem to solution – A review. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.03.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
16
|
Voigtmann F, Wolf P, Landgraf K, Stein R, Kratzsch J, Schmitz S, Abou Jamra R, Blüher M, Meiler J, Beck-Sickinger AG, Kiess W, Körner A. Identification of a novel leptin receptor (LEPR) variant and proof of functional relevance directing treatment decisions in patients with morbid obesity. Metabolism 2021; 116:154438. [PMID: 33221380 DOI: 10.1016/j.metabol.2020.154438] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Deficiency in the leptin-leptin receptor (LEPR) axis leads to severe, and potentially treatable, obesity in humans. To guide clinical decision-making, the functional relevance of variants in the LEPR gene needs to be carefully investigated. CASES AND METHODS We characterized the functional impact of LEPR variants identified in two patients with severe early-onset obesity (1: compound heterozygous for the novel variant p.Tyr411del and p.Trp664Arg; 2: heterozygous for p.Arg612His) by investigating leptin-mediated signaling, leptin binding, receptor expression on cell surfaces, and receptor dimerization and activation for either wild-type and/or mutant LEPR. RESULTS Leptin-induced STAT3-phosphorylation was blunted the novel p.Tyr411del or the p.Trp664Arg variant and mildly reduced with the p.Arg612His variant. Computational structure prediction suggested impaired leptin binding for all three LEPR variants. Experimentally, reduced leptin binding of all mutant proteins was due to diminished LEPR expression on the cell surface, with the p.Trp664Arg mutations being the most affected. Considering the heterozygosity in our patients, we assessed the heterodimerization capacity with the wild-type LEPR, which was retained for the p.Tyr411del and p.Arg612His variants. Finally, mimicking (compound) heterozygosity, we confirmed abolished STAT3-phosphorylation for the variant combination [p.Tyr411del + p.Trp664Arg] as found in patient 1, whereas it was retained for [p.Arg612His + wilde type] as found in patient 2. CONCLUSIONS The novel p.Tyr411del mutation causes complete loss of function alone (and combined with p.Trp664Arg) and is likely the cause for the early onset obesity, qualifying the patient for pharmacologic treatment. Heterozygosity for the p.Arg612His variant, however, appears unlikely to be solely responsible for the phenotype.
Collapse
Affiliation(s)
- Franziska Voigtmann
- Center of Pediatric Research Leipzig, University Hospital for Children & Adolescents, Medical Faculty, University of Leipzig, Germany
| | - Philipp Wolf
- Institute of Biochemistry, Faculty of Life Sciences, Pharmacy and Psychology, University of Leipzig, Germany
| | - Kathrin Landgraf
- Center of Pediatric Research Leipzig, University Hospital for Children & Adolescents, Medical Faculty, University of Leipzig, Germany
| | - Robert Stein
- Center of Pediatric Research Leipzig, University Hospital for Children & Adolescents, Medical Faculty, University of Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Germany
| | - Jürgen Kratzsch
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnosis, University Medical Center Leipzig, Medical Faculty, University of Leipzig, Germany
| | - Samuel Schmitz
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, TN, USA
| | - Rami Abou Jamra
- Institute of Human Genetics, University Medical Center Leipzig, Medical Faculty, University of Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Germany; Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig, Germany
| | - Jens Meiler
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, TN, USA; Institute of Drug Discovery, Medical Faculty, University of Leipzig, Germany
| | - Annette G Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Pharmacy and Psychology, University of Leipzig, Germany
| | - Wieland Kiess
- Center of Pediatric Research Leipzig, University Hospital for Children & Adolescents, Medical Faculty, University of Leipzig, Germany
| | - Antje Körner
- Center of Pediatric Research Leipzig, University Hospital for Children & Adolescents, Medical Faculty, University of Leipzig, Germany.
| |
Collapse
|
17
|
Zaghlool SB, Sharma S, Molnar M, Matías-García PR, Elhadad MA, Waldenberger M, Peters A, Rathmann W, Graumann J, Gieger C, Grallert H, Suhre K. Revealing the role of the human blood plasma proteome in obesity using genetic drivers. Nat Commun 2021; 12:1279. [PMID: 33627659 PMCID: PMC7904950 DOI: 10.1038/s41467-021-21542-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/29/2021] [Indexed: 12/21/2022] Open
Abstract
Blood circulating proteins are confounded readouts of the biological processes that occur in different tissues and organs. Many proteins have been linked to complex disorders and are also under substantial genetic control. Here, we investigate the associations between over 1000 blood circulating proteins and body mass index (BMI) in three studies including over 4600 participants. We show that BMI is associated with widespread changes in the plasma proteome. We observe 152 replicated protein associations with BMI. 24 proteins also associate with a genome-wide polygenic score (GPS) for BMI. These proteins are involved in lipid metabolism and inflammatory pathways impacting clinically relevant pathways of adiposity. Mendelian randomization suggests a bi-directional causal relationship of BMI with LEPR/LEP, IGFBP1, and WFIKKN2, a protein-to-BMI relationship for AGER, DPT, and CTSA, and a BMI-to-protein relationship for another 21 proteins. Combined with animal model and tissue-specific gene expression data, our findings suggest potential therapeutic targets further elucidating the role of these proteins in obesity associated pathologies.
Collapse
Affiliation(s)
- Shaza B Zaghlool
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Sapna Sharma
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Megan Molnar
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
| | - Pamela R Matías-García
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Mohamed A Elhadad
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- German Research Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- German Research Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Wolfgang Rathmann
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Biometrics and Epidemiology, German Diabetes Center, Düsseldorf, Germany
| | - Johannes Graumann
- Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, W.G. Kerckhoff Institute, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Max Planck Institute of Heart and Lung Research, Bad Nauheim, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar.
| |
Collapse
|
18
|
Zheng R, Liu R, Wu M, Wang H, Xie L. Effects of sodium perchlorate and exogenous L-thyroxine on growth, development and leptin signaling pathway of Bufo gargarizans tadpoles during metamorphosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 206:111410. [PMID: 33007540 DOI: 10.1016/j.ecoenv.2020.111410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/20/2020] [Accepted: 09/23/2020] [Indexed: 06/11/2023]
Abstract
Sodium perchlorate (NaClO4) and exogenous L-thyroxine (T4), two kinds of endocrine-disrupting chemicals (EDCs), mainly affect the circulating thyroid hormones, which regulate the initiation and rate of metamorphosis in amphibian. The aim of this study is to evaluate the potential role of EDCs in regulating the development of tadpoles and leptin signaling pathway of liver during the metamorphosis of Bufo gargarizans. There was completely opposite result of average development stage of tadpoles and morphological parameters between the NaClO4 and T4 exposure groups. Histological analysis revealed that NaClO4 and T4 exposure both caused liver injury, such as the decreased size of hepatocytes, atrophy of nucleus, increased melanomacrophage centres and disappearance of hepatocyte membranes. In addition, the results of RT-qPCR revealed that NaClO4 treatment significantly inhibited the transcript levels of genes related to thyroid hormone (D2, TRα and TRβ) and leptin signaling pathway (LepR, JAK1, JAK2, and TYK2), while there was an increase of mRNA expression of these genes in the liver of tadpoles administrated with T4 compared with control. This work lays an important foundation for assessing the risk of EDCs in relation to amphibian development during metamorphosis.
Collapse
Affiliation(s)
- Rui Zheng
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China
| | - Rong Liu
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China
| | - Minyao Wu
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China
| | - Hongyuan Wang
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China.
| | - Lei Xie
- National and Local Joint Engineering Research Center of Ecological Treatment Technology for Urban Water Pollution, Wenzhou University, 325035, Wenzhou, China; College of Life and Environmental Science, Wenzhou University, 325035, Wenzhou, China.
| |
Collapse
|
19
|
FOXO activity adaptation safeguards the hematopoietic stem cell compartment in hyperglycemia. Blood Adv 2020; 4:5512-5526. [PMID: 33166407 DOI: 10.1182/bloodadvances.2020001826] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 10/05/2020] [Indexed: 12/17/2022] Open
Abstract
Hematopoietic stem cell (HSC) activity is tightly controlled to ensure the integrity of the hematopoietic system during the organism's lifetime. How the HSC compartment maintains its long-term fitness in conditions of chronic stresses associated with systemic metabolic disorders is poorly understood. In this study, we show that obesity functionally affects the long-term function of the most immature engrafting HSC subpopulation. We link this altered regenerative activity to the oxidative stress and the aberrant constitutive activation of the AKT signaling pathway that characterized the obese environment. In contrast, we found minor disruptions of the HSC function in obese mice at steady state, suggesting that active mechanisms could protect the HSC compartment from its disturbed environment. Consistent with this idea, we found that FOXO proteins in HSCs isolated from obese mice become insensitive to their normal upstream regulators such as AKT, even during intense oxidative stress. We established that hyperglycemia, a key condition associated with obesity, is directly responsible for the alteration of the AKT-FOXO axis in HSCs and their abnormal oxidative stress response. As a consequence, we observed that HSCs isolated from a hyperglycemic environment display enhanced resistance to oxidative stress and DNA damage. Altogether, these results indicate that chronic metabolic stresses associated with obesity and/or hyperglycemia affect the wiring of the HSCs and modify their oxidative stress response. These data suggest that the uncoupling of FOXO from its environmental regulators could be a key adaptive strategy that promotes the survival of the HSC compartment in obesity.
Collapse
|
20
|
Milaneschi Y, Lamers F, Berk M, Penninx BWJH. Depression Heterogeneity and Its Biological Underpinnings: Toward Immunometabolic Depression. Biol Psychiatry 2020; 88:369-380. [PMID: 32247527 DOI: 10.1016/j.biopsych.2020.01.014] [Citation(s) in RCA: 230] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/03/2019] [Accepted: 01/18/2020] [Indexed: 12/14/2022]
Abstract
Epidemiological evidence indicates the presence of dysregulated homeostatic biological pathways in depressed patients, such as increased inflammation and disrupted energy-regulating neuroendocrine signaling (e.g., leptin, insulin). Alterations in these biological pathways may explain the considerable comorbidity between depression and cardiometabolic conditions (e.g., obesity, metabolic syndrome, diabetes) and represent a promising target for intervention. This review describes how immunometabolic dysregulations vary as a function of depression heterogeneity by illustrating that such biological dysregulations map more consistently to atypical behavioral symptoms reflecting altered energy intake/expenditure balance (hyperphagia, weight gain, hypersomnia, fatigue, and leaden paralysis) and may moderate the antidepressant effects of standard or novel (e.g., anti-inflammatory) therapeutic approaches. These lines of evidence are integrated in a conceptual model of immunometabolic depression emerging from the clustering of immunometabolic biological dysregulations and specific behavioral symptoms. The review finally elicits questions to be answered by future research and describes how the immunometabolic depression dimension could be used to dissect the heterogeneity of depression and potentially to match subgroups of patients to specific treatments with higher likelihood of clinical success.
Collapse
Affiliation(s)
- Yuri Milaneschi
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam University Medical Center/Vrije Universiteit & GGZinGeest, Amsterdam, The Netherlands.
| | - Femke Lamers
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam University Medical Center/Vrije Universiteit & GGZinGeest, Amsterdam, The Netherlands
| | - Michael Berk
- Institute for Mental and Physical Health and Clinical Treatment, School of Medicine, Deakin University and Barwon Health, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Department of Psychiatry, University of Melbourne, Melbourne, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam University Medical Center/Vrije Universiteit & GGZinGeest, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Pedroso JAB, Wasinski F, Donato J. Prolonged fasting induces long-lasting metabolic consequences in mice. J Nutr Biochem 2020; 84:108457. [PMID: 32738733 DOI: 10.1016/j.jnutbio.2020.108457] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022]
Abstract
To endure prolonged fasting, animals undergo important acute physiological adjustments. However, whether severe fasting also leads to long-term metabolic adaptations is largely unknown. Forty-eight-hour fasting caused a pronounced weight loss in adult C57BL/6 male mice. Seven days of refeeding increased body adiposity to levels above baseline, whereas fasting-induced reductions in lean body mass and energy expenditure were not fully recovered. Respiratory exchange ratio and locomotor activity also remained altered. A fasting/refeeding cycle led to persistent suppression of Pomc mRNA levels and significant changes in the expression of histone deacetylases and DNA methyltransferases in the hypothalamus. Additionally, histone acetylation in the ventromedial nucleus of the hypothalamus was reduced by prolonged fasting and remained suppressed after refeeding. Mice subjected to 48-h fasting 30 days earlier exhibited higher body weight and fat mass compared to aged-matched animals that were never food-deprived. Furthermore, a previous fasting experience altered the changes in body weight, lean mass, energy expenditure and locomotor activity induced by a second cycle of fasting and refeeding. Notably, when acutely exposed to high-palatable/high-fat diet, mice that went through cumulative fasting episodes presented higher calorie intake and reduced energy expenditure and fat oxidation, compared to mice that had never been subjected to fasting. When chronically exposed to high-fat diet, mice that experienced cumulative fasting episodes showed higher gain of body and fat mass and reduced energy expenditure and calorie intake. In summary, cumulative episodes of prolonged fasting lead to hypothalamic epigenetic changes and long-lasting metabolic adaptations in mice.
Collapse
Affiliation(s)
- João A B Pedroso
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Fisiologia e Biofísica, São Paulo, 05508-000, Brazil
| | - Frederick Wasinski
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Fisiologia e Biofísica, São Paulo, 05508-000, Brazil
| | - Jose Donato
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Fisiologia e Biofísica, São Paulo, 05508-000, Brazil.
| |
Collapse
|
22
|
Nabil M, El Demellawy MA, Mahmoud MF, Mahmoud AAA. Prolonged overnutrition with fructose or fat induces metabolic derangements in rats by disrupting the crosstalk between the hypothalamus and periphery: Possible amelioration with fenofibrate. Eur J Pharmacol 2020; 879:173136. [PMID: 32360834 DOI: 10.1016/j.ejphar.2020.173136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 11/15/2022]
Abstract
Metabolic Syndrome (MetS) increases the risk of developing type 2 diabetes mellitus and cardiovascular complications. The crosstalk between the hypothalamus and periphery is vital for regulating food intake and energy homeostasis. However, it is impaired during MetS. The present study aimed to compare the distinct central and peripheral metabolic derangements induced by a high-fructose drink or high-fat diet, as well as the possible intervention by fenofibrate. Rats were divided into five groups: standard chow diet (SCD) group, high-fructose group (FR), high-fat group (HF), FR plus fenofibrate group (FR-F), and HF plus fenofibrate group (HF-F). FR and HF groups showed hyperglycemia, hyperinsulinemia, hypertriglyceridemia, hyperleptinemia, steatosis, and adipocyte hypertrophy. This was associated with elevated circulating levels of proinflammatory cytokines and free fatty acids (FFAs). The latter mediators are involved in the hypothalamic inflammation and dysregulation of signaling cascades that control food intake and glucose homeostasis. The effects were more pronounced in the HF group than FR group, which were matched with the observed higher levels of plasma FFAs and cytokines. Fenofibrate administration improved not only the peripheral metabolic disturbances, but also the central disturbances associated with insulin resistance induced by FR or HF diet. This study sheds light on the pivotal role of the hypothalamus in diet-induced MetS. Furthermore, the study suggests the utmost importance of developing a standardized model of metabolic syndrome in place of the great diversity between available models, which can induce different effects and negatively impact the validity of prospective studies.
Collapse
Affiliation(s)
- Mohamed Nabil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt; Pharmaceutical and Fermentation Industries Development Center (PFIDC), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab, Alexandria, 21934, Egypt
| | - Maha A El Demellawy
- Department of Medical Biotechnology, Genetic Engineering & Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab, Alexandria, 21934, Egypt
| | - Mona F Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Amr A A Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
23
|
Barnes TM, Shah K, Allison MB, Steinl GK, Gordian D, Sabatini PV, Tomlinson AJ, Cheng W, Jones JC, Zhu Q, Faber C, Myers MG. Identification of the leptin receptor sequences crucial for the STAT3-Independent control of metabolism. Mol Metab 2020; 32:168-175. [PMID: 32029227 PMCID: PMC6992902 DOI: 10.1016/j.molmet.2019.12.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/12/2019] [Accepted: 12/29/2019] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Leptin acts via its receptor, LepRb, on specialized neurons in the brain to modulate energy balance and glucose homeostasis. LepRb→STAT3 signaling plays a crucial role in leptin action, but LepRb also mediates an additional as-yet-unidentified signal (Signal 2) that is important for leptin action. Signal 2 requires LepRb regions in addition to those required for JAK2 activation but operates independently of STAT3 and LepRb phosphorylation sites. METHODS To identify LepRb sequences that mediate Signal 2, we used CRISPR/Cas9 to generate five novel mouse lines containing COOH-terminal truncation mutants of LepRb. We analyzed the metabolic phenotype and measures of hypothalamic function for these mouse lines. RESULTS We found that deletion of LepRb sequences between residues 921 and 960 dramatically worsens metabolic control and alters hypothalamic function relative to smaller truncations. We also found that deletion of the regions including residues 1013-1053 and 960-1013 each decreased obesity compared to deletions that included additional COOH-terminal residues. CONCLUSIONS LepRb sequences between residues 921 and 960 mediate the STAT3 and LepRb phosphorylation-independent second signal that contributes to the control of energy balance and metabolism by leptin/LepRb. In addition to confirming the inhibitory role of the region (residues 961-1013) containing Tyr985, we also identified the region containing residues 1013-1053 (which contains no Tyr residues) as a second potential mediator of LepRb inhibition. Thus, the intracellular domain of LepRb mediates multiple Tyr-independent signals.
Collapse
Affiliation(s)
- Tammy M Barnes
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kimi Shah
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Margaret B Allison
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Gabrielle K Steinl
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Desiree Gordian
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Paul V Sabatini
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Wenwen Cheng
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Justin C Jones
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Qing Zhu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Chelsea Faber
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Martin G Myers
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
24
|
Chellappa K, Brinkman JA, Mukherjee S, Morrison M, Alotaibi MI, Carbajal KA, Alhadeff AL, Perron IJ, Yao R, Purdy CS, DeFelice DM, Wakai MH, Tomasiewicz J, Lin A, Meyer E, Peng Y, Arriola Apelo SI, Puglielli L, Betley JN, Paschos GK, Baur JA, Lamming DW. Hypothalamic mTORC2 is essential for metabolic health and longevity. Aging Cell 2019; 18:e13014. [PMID: 31373126 PMCID: PMC6718533 DOI: 10.1111/acel.13014] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 05/26/2019] [Accepted: 07/03/2019] [Indexed: 12/16/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) is an evolutionarily conserved protein kinase that regulates growth and metabolism. mTOR is found in two protein complexes, mTORC1 and mTORC2, that have distinct components and substrates and are both inhibited by rapamycin, a macrolide drug that robustly extends lifespan in multiple species including worms and mice. Although the beneficial effect of rapamycin on longevity is generally attributed to reduced mTORC1 signaling, disruption of mTORC2 signaling can also influence the longevity of worms, either positively or negatively depending on the temperature and food source. Here, we show that loss of hypothalamic mTORC2 signaling in mice decreases activity level, increases the set point for adiposity, and renders the animals susceptible to diet-induced obesity. Hypothalamic mTORC2 signaling normally increases with age, and mice lacking this pathway display higher fat mass and impaired glucose homeostasis throughout life, become more frail with age, and have decreased overall survival. We conclude that hypothalamic mTORC2 is essential for the normal metabolic health, fitness, and lifespan of mice. Our results have implications for the use of mTORC2-inhibiting pharmaceuticals in the treatment of brain cancer and diseases of aging.
Collapse
Affiliation(s)
- Karthikeyani Chellappa
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Jacqueline A. Brinkman
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
| | - Sarmistha Mukherjee
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Mark Morrison
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
| | - Mohammed I. Alotaibi
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
- Endocrinology and Reproductive Physiology Graduate Training ProgramUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Kathryn A. Carbajal
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
| | - Amber L. Alhadeff
- Department of Biology, School of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Isaac J. Perron
- Center for Sleep and Circadian Neurobiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Rebecca Yao
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Cole S. Purdy
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Denise M. DeFelice
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Matthew H. Wakai
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
| | - Jay Tomasiewicz
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Amy Lin
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Emma Meyer
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Yajing Peng
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Sebastian I. Arriola Apelo
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Luigi Puglielli
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - J. Nicholas Betley
- Department of Biology, School of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Georgios K. Paschos
- Center for Sleep and Circadian Neurobiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
- The Institute for Translational Medicine and Therapeutics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Joseph A. Baur
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Dudley W. Lamming
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
- Endocrinology and Reproductive Physiology Graduate Training ProgramUniversity of Wisconsin‐MadisonMadisonWIUSA
| |
Collapse
|
25
|
Feeding circuit development and early-life influences on future feeding behaviour. Nat Rev Neurosci 2019; 19:302-316. [PMID: 29662204 DOI: 10.1038/nrn.2018.23] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A wide range of maternal exposures - undernutrition, obesity, diabetes, stress and infection - are associated with an increased risk of metabolic disease in offspring. Developmental influences can cause persistent structural changes in hypothalamic circuits regulating food intake in the service of energy balance. The physiological relevance of these alterations has been called into question because maternal impacts on daily caloric intake do not persist to adulthood. Recent behavioural and epidemiological studies in humans provide evidence that the relative contribution of appetitive traits related to satiety, reward and the emotional aspects of food intake regulation changes across the lifespan. This Opinion article outlines a neurodevelopmental framework to explore the possibility that crosstalk between developing circuits regulating different modalities of food intake shapes future behavioural responses to environmental challenges.
Collapse
|
26
|
Perry RJ, Resch JM, Douglass AM, Madara JC, Rabin-Court A, Kucukdereli H, Wu C, Song JD, Lowell BB, Shulman GI. Leptin's hunger-suppressing effects are mediated by the hypothalamic-pituitary-adrenocortical axis in rodents. Proc Natl Acad Sci U S A 2019; 116:13670-13679. [PMID: 31213533 PMCID: PMC6613139 DOI: 10.1073/pnas.1901795116] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Leptin informs the brain about sufficiency of fuel stores. When insufficient, leptin levels fall, triggering compensatory increases in appetite. Falling leptin is first sensed by hypothalamic neurons, which then initiate adaptive responses. With regard to hunger, it is thought that leptin-sensing neurons work entirely via circuits within the central nervous system (CNS). Very unexpectedly, however, we now show this is not the case. Instead, stimulation of hunger requires an intervening endocrine step, namely activation of the hypothalamic-pituitary-adrenocortical (HPA) axis. Increased corticosterone then activates AgRP neurons to fully increase hunger. Importantly, this is true for 2 forms of low leptin-induced hunger, fasting and poorly controlled type 1 diabetes. Hypoglycemia, which also stimulates hunger by activating CNS neurons, albeit independently of leptin, similarly recruits and requires this pathway by which HPA axis activity stimulates AgRP neurons. Thus, HPA axis regulation of AgRP neurons is a previously underappreciated step in homeostatic regulation of hunger.
Collapse
Affiliation(s)
- Rachel J Perry
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520
| | - Jon M Resch
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Amelia M Douglass
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Joseph C Madara
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Aviva Rabin-Court
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Hakan Kucukdereli
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Chen Wu
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Joongyu D Song
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215;
- Program in Neuroscience, Harvard Medical School, Boston, MA 02215
| | - Gerald I Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520;
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|
27
|
Arunagiri A, Haataja L, Pottekat A, Pamenan F, Kim S, Zeltser LM, Paton AW, Paton JC, Tsai B, Itkin-Ansari P, Kaufman RJ, Liu M, Arvan P. Proinsulin misfolding is an early event in the progression to type 2 diabetes. eLife 2019; 8:44532. [PMID: 31184302 PMCID: PMC6559786 DOI: 10.7554/elife.44532] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/09/2019] [Indexed: 02/06/2023] Open
Abstract
Biosynthesis of insulin – critical to metabolic homeostasis – begins with folding of the proinsulin precursor, including formation of three evolutionarily conserved intramolecular disulfide bonds. Remarkably, normal pancreatic islets contain a subset of proinsulin molecules bearing at least one free cysteine thiol. In human (or rodent) islets with a perturbed endoplasmic reticulum folding environment, non-native proinsulin enters intermolecular disulfide-linked complexes. In genetically obese mice with otherwise wild-type islets, disulfide-linked complexes of proinsulin are more abundant, and leptin receptor-deficient mice, the further increase of such complexes tracks with the onset of islet insulin deficiency and diabetes. Proinsulin-Cys(B19) and Cys(A20) are necessary and sufficient for the formation of proinsulin disulfide-linked complexes; indeed, proinsulin Cys(B19)-Cys(B19) covalent homodimers resist reductive dissociation, highlighting a structural basis for aberrant proinsulin complex formation. We conclude that increased proinsulin misfolding via disulfide-linked complexes is an early event associated with prediabetes that worsens with ß-cell dysfunction in type two diabetes. Our body fine-tunes the amount of sugar in our blood thanks to specialized ‘beta cells’ in the pancreas, which can release a hormone called insulin. To produce insulin, the beta cells first need to build an early version of the molecule – known as proinsulin – inside a cellular compartment called the endoplasmic reticulum. This process involves the formation of internal staples that keep the molecule of proinsulin folded correctly. Individuals developing type 2 diabetes have spikes of sugar in their blood, and so their bodies often respond by trying to make large amounts of insulin. After a while, the beta cells can fail to keep up, which brings on the full-blown disease. However, scientists have discovered that early in type 2 diabetes, the endoplasmic reticulum of beta cells can already show signs of stress; yet, the exact causes of this early damage are still unknown. To investigate this, Arunagiri et al. looked into whether proinsulin folds correctly during the earliest stages of type 2 diabetes. Biochemical experiments showed that even healthy beta cells contained some misfolded proinsulin molecules, where the molecular staples that should fold proinsulin internally were instead abnormally linking proinsulin molecules together. Further work revealed that the misfolded proinsulin was accumulating inside the endoplasmic reticulum. Finally, obese mice that were in the earliest stages of type 2 diabetes had the highest levels of abnormal proinsulin in their beta cells. Overall, the work by Arunagiri et al. suggests that large amounts of proinsulin molecules stapling themselves to each other in the endoplasmic reticulum of beta cells could be an early hallmark of the disease, and could make it get worse. A separate study by Jang et al. also shows that a protein that limits the misfolding of proinsulin is key to maintain successful insulin production in animals eating a Western-style, high fat diet. Hundreds of millions of people around the world have type 2 diabetes, and this number is rising quickly. Detecting and then fixing early problems associated with the condition may help to stop the disease in its track.
Collapse
Affiliation(s)
- Anoop Arunagiri
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, United States
| | - Leena Haataja
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, United States
| | - Anita Pottekat
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - Fawnnie Pamenan
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, United States
| | - Soohyun Kim
- Department of Biomedical Science and Technology, Konkuk University, Gwangjin-gu, Republic of Korea
| | - Lori M Zeltser
- Department of Pathology and Cell Biology, Naomi Berrie Diabetes Center, Columbia University, New York, United States
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Billy Tsai
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Pamela Itkin-Ansari
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - Ming Liu
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, United States.,Department of Endocrinology and Metabolism, Tianjin Medical University, Tianjin, China
| | - Peter Arvan
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, United States
| |
Collapse
|
28
|
Pan W, Allison MB, Sabatini P, Rupp A, Adams J, Patterson C, Jones JC, Olson DP, Myers MG. Transcriptional and physiological roles for STAT proteins in leptin action. Mol Metab 2019; 22:121-131. [PMID: 30718218 PMCID: PMC6437596 DOI: 10.1016/j.molmet.2019.01.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/14/2019] [Accepted: 01/17/2019] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES Leptin acts via its receptor LepRb on specialized neurons in the brain to modulate food intake, energy expenditure, and body weight. LepRb activates signal transducers and activators of transcription (STATs, including STAT1, STAT3, and STAT5) to control gene expression. METHODS Because STAT3 is crucial for physiologic leptin action, we used TRAP-seq to examine gene expression in LepRb neurons of mice ablated for Stat3 in LepRb neurons (Stat3LepRbKO mice), revealing the STAT3-dependent transcriptional targets of leptin. To understand roles for STAT proteins in leptin action, we also ablated STAT1 or STAT5 from LepRb neurons and expressed a constitutively-active STAT3 (CASTAT3) in LepRb neurons. RESULTS While we also found increased Stat1 expression and STAT1-mediated transcription of leptin-regulated genes in Stat3LepRbKO mice, ablating Stat1 in LepRb neurons failed to alter energy balance (even on the Stat3LepRbKO background); ablating Stat5 in LepRb neurons also failed to alter energy balance. Importantly, expression of a constitutively-active STAT3 (CASTAT3) in LepRb neurons decreased food intake and body weight and improved metabolic parameters in leptin-deficient (ob/ob) mice, as well as in wild-type animals. CONCLUSIONS Thus, STAT3 represents the unique STAT protein required for leptin action and STAT3 suffices to mediate important components of leptin action in the absence of other LepRb signals.
Collapse
Affiliation(s)
- Warren Pan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA
| | - Margaret B Allison
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Paul Sabatini
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Alan Rupp
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jessica Adams
- Division of Endocrinology, Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Christa Patterson
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Justin C Jones
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - David P Olson
- Division of Endocrinology, Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Martin G Myers
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
29
|
Sahu M, Anamthathmakula P, Sahu A. Hypothalamic PDE3B deficiency alters body weight and glucose homeostasis in mouse. J Endocrinol 2018; 239:93-105. [PMID: 30307157 PMCID: PMC6190684 DOI: 10.1530/joe-18-0304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Pharmacological studies have suggested hypothalamic phosphodiesterase-3B to mediate leptin and insulin action in regulation of energy homeostasis. Whereas Pde3b-null mice show altered energy homeostasis, it is unknown whether this is due to ablation of Pde3b in the hypothalamus. Thus, to address the functional significance of hypothalamic phosphodiesterase-3B, we used Pde3bflox/flox and Nkx2.1-Cre mice to generate Pde3b Nkx2.1KD mice that showed 50% reduction of phosphodiesterase-3B in the hypothalamus. To determine the effect of partial ablation of phosphodiesterase-3B in the hypothalamus on energy and glucose homeostasis, males and females were subjected to either a low- or high-fat diet for 19–21 weeks. Only female but not male Pde3b Nkx2.1KD mice on the low-fat diet showed increased body weight from 13 weeks onward with increased food intake, decreased fat pad weights and hypoleptinemia. Glucose tolerance was improved in high-fat diet-fed male Pde3b Nkx2.1KD mice in association with decreased phosphoenolpyruvate carboxykinase-1 and glucose-6-phosphatase mRNA levels in the liver. Also, insulin sensitivity was increased in male Pde3b Nkx2.1KD mice on the low-fat diet. Changes in body weight or in glucose homeostasis were not associated with any alteration in hypothalamic proopiomelanocortin, neuropepide Y and agouti-related peptide mRNA levels. These results suggest that partial loss of phosphodiesterase-3B in the hypothalamus produces a sex-specific response in body weight and glucose homeostasis, and support a role, at least in part, for hypothalamic phosphodiesterase-3B in regulation of energy and glucose homeostasis in mice.
Collapse
Affiliation(s)
- Maitrayee Sahu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Prashanth Anamthathmakula
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Abhiram Sahu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
30
|
Iqbal NJ, Lu Z, Liu SM, Schwartz GJ, Chua S, Zhu L. Cyclin-dependent kinase 4 is a preclinical target for diet-induced obesity. JCI Insight 2018; 3:123000. [PMID: 30185666 PMCID: PMC6171799 DOI: 10.1172/jci.insight.123000] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 07/26/2018] [Indexed: 12/12/2022] Open
Abstract
When obesity is caused by consumption of a high-fat diet, the tumor suppressor pRb is phosphoinactivated in the neurons of the mediobasal hypothalamus, a brain area critical for energy-balance regulation. However, the functional relevance of pRb phosphoinactivation in the mediobasal hypothalamus to diet-induced obesity remains unknown. Here, we show that inhibiting pRb phosphorylation in the mediobasal hypothalamus can prevent and treat diet-induced obesity in mice. Expressing an unphosphorylable pRb nonselectively in the mediobasal hypothalamus or conditionally in anorexigenic POMC neurons inhibits diet-induced obesity. Intracerebroventricular delivery of US Food and Drug Administration–approved (FDA-approved) cyclin-dependent kinase 4 (CDK4) inhibitor abemaciclib inhibits pRb phosphorylation in the mediobasal hypothalamus and prevents diet-induced obesity. Oral administration of abemaciclib at doses approved for human use reduces fat mass in diet-induced obese mice by increasing lipid oxidation without significantly reducing lean mass. With analysis of recent literature identifying CDK4 as the most abundantly expressed neuronal CDK in the mediobasal hypothalamus, our work uncovers CDK4 as the major kinase for hypothalamic pRb phosphoinactivation and a highly effective central antiobesity target. As three CDK4/6 inhibitors have recently received FDA approval for life-long breast cancer therapy, our study provides a preclinical basis for their expedient repurposing for obesity management. Inhibiting pRb phosphorylation in the mediobasal hypothalamus can prevent and treat diet-induced obesity in mice.
Collapse
Affiliation(s)
| | - Zhonglei Lu
- Department of Developmental and Molecular Biology and
| | - Shun Mei Liu
- Department of Medicine, Albert Einstein College of Medicine, New York, New York, USA
| | - Gary J Schwartz
- Department of Medicine, Albert Einstein College of Medicine, New York, New York, USA
| | - Streamson Chua
- Department of Medicine, Albert Einstein College of Medicine, New York, New York, USA
| | - Liang Zhu
- Department of Developmental and Molecular Biology and.,Department of Medicine, Albert Einstein College of Medicine, New York, New York, USA
| |
Collapse
|
31
|
Allison MB, Pan W, MacKenzie A, Patterson C, Shah K, Barnes T, Cheng W, Rupp A, Olson DP, Myers MG. Defining the Transcriptional Targets of Leptin Reveals a Role for Atf3 in Leptin Action. Diabetes 2018; 67:1093-1104. [PMID: 29535089 PMCID: PMC5961413 DOI: 10.2337/db17-1395] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/27/2018] [Indexed: 12/22/2022]
Abstract
Leptin acts via its receptor (LepRb) to modulate gene expression in hypothalamic LepRb-expressing neurons, thereby controlling energy balance and glucose homeostasis. Despite the importance of the control of gene expression in hypothalamic LepRb neurons for leptin action, the transcriptional targets of LepRb signaling have remained undefined because LepRb cells contribute a small fraction to the aggregate transcriptome of the brain regions in which they reside. We thus employed translating ribosome affinity purification followed by RNA sequencing to isolate and analyze mRNA from the hypothalamic LepRb neurons of wild-type or leptin-deficient (Lepob/ob) mice treated with vehicle or exogenous leptin. Although the expression of most of the genes encoding the neuropeptides commonly considered to represent the main targets of leptin action were altered only following chronic leptin deprivation, our analysis revealed other transcripts that were coordinately regulated by leptin under multiple treatment conditions. Among these, acute leptin treatment increased expression of the transcription factor Atf3 in LepRb neurons. Furthermore, ablation of Atf3 from LepRb neurons (Atf3LepRbKO mice) decreased leptin efficacy and promoted positive energy balance in mice. Thus, this analysis revealed the gene targets of leptin action, including Atf3, which represents a cellular mediator of leptin action.
Collapse
MESH Headings
- Activating Transcription Factor 3/agonists
- Activating Transcription Factor 3/chemistry
- Activating Transcription Factor 3/genetics
- Activating Transcription Factor 3/metabolism
- Animals
- Crosses, Genetic
- Diabetes Mellitus/drug therapy
- Diabetes Mellitus/metabolism
- Diabetes Mellitus/pathology
- Energy Metabolism/drug effects
- Female
- Gene Expression Profiling
- Gene Expression Regulation/drug effects
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Hypothalamus/cytology
- Hypothalamus/drug effects
- Hypothalamus/metabolism
- Hypothalamus/pathology
- Leptin/analogs & derivatives
- Leptin/metabolism
- Leptin/pharmacology
- Leptin/therapeutic use
- Lipotropic Agents/pharmacology
- Lipotropic Agents/therapeutic use
- Male
- Mice, Knockout
- Mice, Mutant Strains
- Mice, Transgenic
- Nerve Tissue Proteins/agonists
- Nerve Tissue Proteins/chemistry
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Neurons/cytology
- Neurons/drug effects
- Neurons/metabolism
- Neurons/pathology
- Obesity/drug therapy
- Obesity/metabolism
- Obesity/pathology
- RNA, Messenger/chemistry
- RNA, Messenger/metabolism
- Receptors, Leptin/agonists
- Receptors, Leptin/genetics
- Receptors, Leptin/metabolism
- Sequence Analysis, RNA
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Margaret B Allison
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
| | - Warren Pan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI
| | | | - Christa Patterson
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Kimi Shah
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Tammy Barnes
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Wenwen Cheng
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Alan Rupp
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - David P Olson
- Division of Pediatric Endocrinology, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI
| | - Martin G Myers
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
32
|
Pan W, Adams JM, Allison MB, Patterson C, Flak JN, Jones J, Strohbehn G, Trevaskis J, Rhodes CJ, Olson DP, Myers MG. Essential Role for Hypothalamic Calcitonin Receptor‒Expressing Neurons in the Control of Food Intake by Leptin. Endocrinology 2018; 159:1860-1872. [PMID: 29522093 PMCID: PMC5888224 DOI: 10.1210/en.2017-03259] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/28/2018] [Indexed: 01/07/2023]
Abstract
The adipocyte-derived hormone leptin acts via its receptor (LepRb) on central nervous system neurons to communicate the repletion of long-term energy stores, to decrease food intake, and to promote energy expenditure. We generated mice that express Cre recombinase from the calcitonin receptor (Calcr) locus (Calcrcre mice) to study Calcr-expressing LepRb (LepRbCalcr) neurons, which reside predominantly in the arcuate nucleus (ARC). Calcrcre-mediated ablation of LepRb in LepRbCalcrknockout (KO) mice caused hyperphagic obesity. Because LepRb-mediated transcriptional control plays a crucial role in leptin action, we used translating ribosome affinity purification followed by RNA sequencing to define the transcriptome of hypothalamic Calcr neurons, along with its alteration in LepRbCalcrKO mice. We found that ARC LepRbCalcr cells include neuropeptide Y (NPY)/agouti-related peptide (AgRP)/γ-aminobutyric acid (GABA) ("NAG") cells as well as non-NAG cells that are distinct from pro-opiomelanocortin cells. Furthermore, although LepRbCalcrKO mice exhibited dysregulated expression of several genes involved in energy balance, neither the expression of Agrp and Npy nor the activity of NAG cells was altered in vivo. Thus, although direct leptin action via LepRbCalcr cells plays an important role in leptin action, our data also suggest that leptin indirectly, as well as directly, regulates these cells.
Collapse
Affiliation(s)
- Warren Pan
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan
| | - Jessica M Adams
- Division of Endocrinology, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan
| | - Margaret B Allison
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Christa Patterson
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Jonathan N Flak
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Justin Jones
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Garth Strohbehn
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | | | | | - David P Olson
- Division of Endocrinology, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan
| | - Martin G Myers
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
33
|
Mancuso P, Curtis JL, Freeman CM, Peters-Golden M, Weinberg JB, Myers MG. Ablation of the leptin receptor in myeloid cells impairs pulmonary clearance of Streptococcus pneumoniae and alveolar macrophage bactericidal function. Am J Physiol Lung Cell Mol Physiol 2018; 315:L78-L86. [PMID: 29565180 DOI: 10.1152/ajplung.00447.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Leptin is a pleiotropic hormone produced by white adipose tissue that regulates appetite and many physiological functions, including the immune response to infection. Genetic leptin deficiency in humans and mice impairs host defenses against respiratory tract infections. Since leptin deficiency is associated with obesity and other metabolic abnormalities, we generated mice that lack the leptin receptor (LepRb) in cells of the myeloid linage (LysM-LepRb-KO) to evaluate its impact in lean metabolically normal mice in a murine model of pneumococcal pneumonia. We observed higher lung and spleen bacterial burdens in LysM-LepRb-KO mice following an intratracheal challenge with Streptococcus pneumoniae. Although numbers of leukocytes recovered from bronchoalveolar lavage fluid did not differ between groups, we did observe higher levels of pulmonary IL-13 and TNFα in LysM-LepRb-KO mice 48 h post infection. Phagocytosis and killing of ingested S. pneumoniae were also impaired in alveolar macrophages (AMs) from LysM-LepRb-KO mice in vitro and were associated with reduced LTB4 and enhanced PGE2 synthesis in vitro. Pretreatment of AMs with LTB4 and the cyclooxygenase inhibitor, indomethacin, restored phagocytosis but not bacterial killing in vitro. These results confirm our previous observations in leptin-deficient ( ob/ob) and fasted mice and demonstrate that decreased leptin action, as opposed to metabolic irregularities associated with obesity or starvation, is responsible for the defective host defense against pneumococcal pneumonia. They also provide novel targets for therapeutic intervention in humans with bacterial pneumonia.
Collapse
Affiliation(s)
- Peter Mancuso
- Department of Environmental Health Sciences, University of Michigan , Ann Arbor, Michigan.,Department of Nutritional Sciences, School of Public Health, University of Michigan , Ann Arbor, Michigan.,Graduate Program in Immunology, University of Michigan , Ann Arbor, Michigan
| | - Jeffrey L Curtis
- Graduate Program in Immunology, University of Michigan , Ann Arbor, Michigan.,Division of Pulmonary and Critical Care Medicine, University of Michigan , Ann Arbor, Michigan.,Veterans Affairs, Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Christine M Freeman
- Graduate Program in Immunology, University of Michigan , Ann Arbor, Michigan.,Division of Pulmonary and Critical Care Medicine, University of Michigan , Ann Arbor, Michigan.,Veterans Affairs, Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Marc Peters-Golden
- Graduate Program in Immunology, University of Michigan , Ann Arbor, Michigan.,Division of Pulmonary and Critical Care Medicine, University of Michigan , Ann Arbor, Michigan
| | - Jason B Weinberg
- Department of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan.,Department of Microbiology and Immunology, University of Michigan , Ann Arbor, Michigan
| | - Martin G Myers
- Department of Integrative and Molecular Physiology, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
34
|
Leptin resistance was involved in susceptibility to overweight in the striped hamster re-fed with high fat diet. Sci Rep 2018; 8:920. [PMID: 29343842 PMCID: PMC5772526 DOI: 10.1038/s41598-017-18158-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 12/06/2017] [Indexed: 02/03/2023] Open
Abstract
Food restriction (FR) is the most commonly used intervention to prevent the overweight. However, the lost weight is usually followed by “compensatory growth” when FR ends, resulting in overweight. The present study was aimed to examining the behavior patterns and hormones mechanisms underpinning the over-weight. Energy budget and body fat content, and several endocrine markers related to leptin signals were examined in the striped hamsters under 20% FR refed by either low-fat diet (LF group) or high-fat diet (HF group). Body mass and fat content significantly regained when FR ended, and the hamsters in HF group showed 49.1% more body fat than in LF group (P < 0.01). Digestive energy intake was higher by 20.1% in HF than LF group, while metabolic thermogenesis and behavior patterns did not differed between the two groups. Gene expression of leptin receptor and anorexigenic peptides of pro-opiomelanocortin and cocaine- and amphetamine-regulated transcript in hypothalamus were significantly up-regulated in LF group, but down-regulated in HF group. It suggests that effective leptin signals to the brain were involved in attenuation of hyperphagia in hamsters refed with LF. However, “leptin resistance” probably occurred in hamsters refed with HF, which impaired the control of hyperphagia, resulting in development of over-weight.
Collapse
|
35
|
Lee JM, Govindarajah V, Goddard B, Hinge A, Muench DE, Filippi MD, Aronow B, Cancelas JA, Salomonis N, Grimes HL, Reynaud D. Obesity alters the long-term fitness of the hematopoietic stem cell compartment through modulation of Gfi1 expression. J Exp Med 2017; 215:627-644. [PMID: 29282250 PMCID: PMC5789409 DOI: 10.1084/jem.20170690] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/31/2017] [Accepted: 11/16/2017] [Indexed: 12/13/2022] Open
Abstract
Lee et al. show that established obesity alters the composition and long-term fitness of the hematopoietic stem cell (HSC) compartment, in part through a Gfi1-dependent HSC regulatory program that is activated by the chronic oxidative stress associated with this condition. Obesity is a chronic organismal stress that disrupts multiple systemic and tissue-specific functions. In this study, we describe the impact of obesity on the activity of the hematopoietic stem cell (HSC) compartment. We show that obesity alters the composition of the HSC compartment and its activity in response to hematopoietic stress. The impact of obesity on HSC function is progressively acquired but persists after weight loss or transplantation into a normal environment. Mechanistically, we establish that the oxidative stress induced by obesity dysregulates the expression of the transcription factor Gfi1 and that increased Gfi1 expression is required for the abnormal HSC function induced by obesity. These results demonstrate that obesity produces durable changes in HSC function and phenotype and that elevation of Gfi1 expression in response to the oxidative environment is a key driver of the altered HSC properties observed in obesity. Altogether, these data provide phenotypic and mechanistic insight into durable hematopoietic dysregulations resulting from obesity.
Collapse
Affiliation(s)
- Jung-Mi Lee
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Vinothini Govindarajah
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Bryan Goddard
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Ashwini Hinge
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - David E Muench
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Marie-Dominique Filippi
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Bruce Aronow
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Jose A Cancelas
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.,Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - H Leighton Grimes
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Damien Reynaud
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
36
|
Milaneschi Y, Lamers F, Peyrot WJ, Baune BT, Breen G, Dehghan A, Forstner AJ, Grabe HJ, Homuth G, Kan C, Lewis C, Mullins N, Nauck M, Pistis G, Preisig M, Rivera M, Rietschel M, Streit F, Strohmaier J, Teumer A, Van der Auwera S, Wray NR, Boomsma DI, Penninx BWJH. Genetic Association of Major Depression With Atypical Features and Obesity-Related Immunometabolic Dysregulations. JAMA Psychiatry 2017; 74:1214-1225. [PMID: 29049554 PMCID: PMC6396812 DOI: 10.1001/jamapsychiatry.2017.3016] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
IMPORTANCE The association between major depressive disorder (MDD) and obesity may stem from shared immunometabolic mechanisms particularly evident in MDD with atypical features, characterized by increased appetite and/or weight (A/W) during an active episode. OBJECTIVE To determine whether subgroups of patients with MDD stratified according to the A/W criterion had a different degree of genetic overlap with obesity-related traits (body mass index [BMI] and levels of C-reactive protein [CRP] and leptin). DESIGN, SETTING, AND PATIENTS This multicenter study assembled genome-wide genotypic and phenotypic measures from 14 data sets of the Psychiatric Genomics Consortium. Data sets were drawn from case-control, cohort, and population-based studies, including 26 628 participants with established psychiatric diagnoses and genome-wide genotype data. Data on BMI were available for 15 237 participants. Data were retrieved and analyzed from September 28, 2015, through May 20, 2017. MAIN OUTCOMES AND MEASURES Lifetime DSM-IV MDD was diagnosed using structured diagnostic instruments. Patients with MDD were stratified into subgroups according to change in the DSM-IV A/W symptoms as decreased or increased. RESULTS Data included 11 837 participants with MDD and 14 791 control individuals, for a total of 26 628 participants (59.1% female and 40.9% male). Among participants with MDD, 5347 (45.2%) were classified in the decreased A/W and 1871 (15.8%) in the increased A/W subgroups. Common genetic variants explained approximately 10% of the heritability in the 2 subgroups. The increased A/W subgroup showed a strong and positive genetic correlation (SE) with BMI (0.53 [0.15]; P = 6.3 × 10-4), whereas the decreased A/W subgroup showed an inverse correlation (-0.28 [0.14]; P = .06). Furthermore, the decreased A/W subgroup had a higher polygenic risk for increased BMI (odds ratio [OR], 1.18; 95% CI, 1.12-1.25; P = 1.6 × 10-10) and levels of CRP (OR, 1.08; 95% CI, 1.02-1.13; P = 7.3 × 10-3) and leptin (OR, 1.09; 95% CI, 1.06-1.12; P = 1.7 × 10-3). CONCLUSIONS AND RELEVANCE The phenotypic associations between atypical depressive symptoms and obesity-related traits may arise from shared pathophysiologic mechanisms in patients with MDD. Development of treatments effectively targeting immunometabolic dysregulations may benefit patients with depression and obesity, both syndromes with important disability.
Collapse
Affiliation(s)
- Yuri Milaneschi
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Vrije Universiteit Medical Center and GGZ inGeest, Amsterdam, the Netherlands
| | - Femke Lamers
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Vrije Universiteit Medical Center and GGZ inGeest, Amsterdam, the Netherlands
| | - Wouter J. Peyrot
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Vrije Universiteit Medical Center and GGZ inGeest, Amsterdam, the Netherlands
| | - Bernhard T. Baune
- Discipline of Psychiatry, University of Adelaide, Adelaide, Australia
| | - Gerome Breen
- Medical Research Council Social Genetic and Developmental Psychiatry Centre, King’s College London, London, England,National Institute for Health Research Biomedical Research Centre for Mental Health, King’s College London, London, England
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, Imperial College London, London, England
| | - Andreas J. Forstner
- Institute of Human Genetics, University of Bonn, Bonn, Germany,Life Brain Center, Department of Genomics, University of Bonn, Bonn, Germany,Department of Psychiatry, University of Basel, Basel, Switzerland,Human Genomics Research Group, Department of Biomedicine, University of Basel, Basel, Switzerland,Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Hans J. Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine and Ernst Moritz Arndt University Greifswald, Greifswald, Germany
| | - Carol Kan
- Department of Psychological Medicine, King’s College London, London, England,South London and Maudsley National Health Service Foundation, London, England
| | - Cathryn Lewis
- Medical Research Council Social Genetic and Developmental Psychiatry Centre, King’s College London, London, England
| | - Niamh Mullins
- Medical Research Council Social Genetic and Developmental Psychiatry Centre, King’s College London, London, England
| | - Matthias Nauck
- German Centre for Cardiovascular Research, Partner Site Greifswald, University Medicine, University Medicine Greifswald, Greifswald, Germany,Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Giorgio Pistis
- Department of Psychiatry, University Hospital of Lausanne, Prilly, Switzerland
| | - Martin Preisig
- Department of Psychiatry, University Hospital of Lausanne, Prilly, Switzerland
| | - Margarita Rivera
- Medical Research Council Social Genetic and Developmental Psychiatry Centre, King’s College London, London, England,Department of Biochemistry and Molecular Biology II, Institute of Neurosciences, Center for Biomedical Research, University of Granada, Granada, Spain
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Fabian Streit
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jana Strohmaier
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Sandra Van der Auwera
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Naomi R. Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia,Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - Dorret I. Boomsma
- Department of Biological Psychology, VU University Amsterdam, Amsterdam, the Netherlands
| | - Brenda W. J. H. Penninx
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Vrije Universiteit Medical Center and GGZ inGeest, Amsterdam, the Netherlands
| | | |
Collapse
|
37
|
Andermann ML, Lowell BB. Toward a Wiring Diagram Understanding of Appetite Control. Neuron 2017; 95:757-778. [PMID: 28817798 DOI: 10.1016/j.neuron.2017.06.014] [Citation(s) in RCA: 373] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 06/02/2017] [Accepted: 06/07/2017] [Indexed: 01/26/2023]
Abstract
Prior mouse genetic research has set the stage for a deep understanding of appetite regulation. This goal is now being realized through the use of recent technological advances, such as the ability to map connectivity between neurons, manipulate neural activity in real time, and measure neural activity during behavior. Indeed, major progress has been made with regard to meal-related gut control of appetite, arcuate nucleus-based hypothalamic circuits linking energy state to the motivational drive, hunger, and, finally, limbic and cognitive processes that bring about hunger-mediated increases in reward value and perception of food. Unexpected findings are also being made; for example, the rapid regulation of homeostatic neurons by cues that predict future food consumption. The aim of this review is to cover the major underpinnings of appetite regulation, describe recent advances resulting from new technologies, and synthesize these findings into an updated view of appetite regulation.
Collapse
Affiliation(s)
- Mark L Andermann
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
38
|
Flak JN, Arble D, Pan W, Patterson C, Lanigan T, Goforth PB, Sacksner J, Joosten M, Morgan DA, Allison MB, Hayes J, Feldman E, Seeley RJ, Olson DP, Rahmouni K, Myers MG. A leptin-regulated circuit controls glucose mobilization during noxious stimuli. J Clin Invest 2017; 127:3103-3113. [PMID: 28714862 DOI: 10.1172/jci90147] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 06/02/2017] [Indexed: 12/23/2022] Open
Abstract
Adipocytes secrete the hormone leptin to signal the sufficiency of energy stores. Reductions in circulating leptin concentrations reflect a negative energy balance, which augments sympathetic nervous system (SNS) activation in response to metabolically demanding emergencies. This process ensures adequate glucose mobilization despite low energy stores. We report that leptin receptor-expressing neurons (LepRb neurons) in the periaqueductal gray (PAG), the largest population of LepRb neurons in the brain stem, mediate this process. Application of noxious stimuli, which often signal the need to mobilize glucose to support an appropriate response, activated PAG LepRb neurons, which project to and activate parabrachial nucleus (PBN) neurons that control SNS activation and glucose mobilization. Furthermore, activating PAG LepRb neurons increased SNS activity and blood glucose concentrations, while ablating LepRb in PAG neurons augmented glucose mobilization in response to noxious stimuli. Thus, decreased leptin action on PAG LepRb neurons augments the autonomic response to noxious stimuli, ensuring sufficient glucose mobilization during periods of acute demand in the face of diminished energy stores.
Collapse
Affiliation(s)
| | | | - Warren Pan
- Department of Internal Medicine.,Graduate Program in Cellular and Molecular Biology, and
| | | | | | - Paulette B Goforth
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | - Donald A Morgan
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, USA
| | - Margaret B Allison
- Department of Internal Medicine.,Department of Molecular and Integrative Physiology
| | | | | | | | - David P Olson
- Division of Endocrinology, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, USA
| | - Martin G Myers
- Department of Internal Medicine.,Department of Surgery.,Department of Molecular and Integrative Physiology
| |
Collapse
|
39
|
Milaneschi Y, Lamers F, Bot M, Drent ML, Penninx BWJH. Leptin Dysregulation Is Specifically Associated With Major Depression With Atypical Features: Evidence for a Mechanism Connecting Obesity and Depression. Biol Psychiatry 2017; 81:807-814. [PMID: 26742925 DOI: 10.1016/j.biopsych.2015.10.023] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 10/27/2015] [Accepted: 10/27/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Obesity-related dysregulation of leptin signaling (e.g., hyperleptinemia due to central functional resistance) may affect mood. However, evidence for leptin dysregulation in major depressive disorder (MDD) is conflicting. Inconclusive findings may be attributable to heterogeneity of MDD, aggregating biologically different subtypes. We examined the relationship of leptin with MDD, its common subtypes (typical and atypical), and clinical features. METHODS The sample consisted of participants (aged 18 to 65 years) from the Netherlands Study of Depression and Anxiety with current (n = 1062) or remitted (n = 711) MDD and healthy control subjects (n = 497). Diagnoses of MDD and subtypes were based on DSM-IV symptoms. Additional symptoms were measured with the Inventory of Depressive Symptomatology. Blood levels of leptin and adiposity indexes (body mass index and waist circumference) were assessed. RESULTS As compared to control subjects, higher leptin was associated with the atypical MDD subtype both for remitted (n = 144, odds ratio = 1.53, 95% confidence interval = 1.16-2.03, p = .003) and current (n = 270, odds ratio = 1.90, 95% confidence interval = 1.51-2.93, p = 5.3e-8) cases. This association was stronger for increasing adiposity levels (leptin by body mass index interaction, p < .02), strengthening the hypothesis of the involvement of leptin resistance. No association with leptin was found for overall MDD or the typical subtype. Among currently depressed patients, higher leptin was associated with key symptoms identifying the atypical subtype, such as hyperphagia, increased weight, and leaden paralysis. CONCLUSIONS Leptin dysregulation (resistance) may represent an underlying mechanism connecting obesity and MDD with atypical features. Development of treatment effectively targeting leptin resistance may benefit patients with atypical depression characterized by obesity-related metabolic alterations.
Collapse
Affiliation(s)
- Yuri Milaneschi
- Department of Psychiatry, EMGO Institute for Health and Care Research and Neuroscience Campus Amsterdam, VU University Medical Center/GGZ ingest, Amsterdam, The Netherlands.
| | - Femke Lamers
- Department of Psychiatry, EMGO Institute for Health and Care Research and Neuroscience Campus Amsterdam, VU University Medical Center/GGZ ingest, Amsterdam, The Netherlands
| | - Mariska Bot
- Department of Psychiatry, EMGO Institute for Health and Care Research and Neuroscience Campus Amsterdam, VU University Medical Center/GGZ ingest, Amsterdam, The Netherlands
| | - Madeleine L Drent
- Department of Internal Medicine, Endocrine Section, VU University Medical Center, Department of Clinical Neuropsychology, Faculty of Psychology and Education, VU University, Neuroscience Campus Amsterdam, Amsterdam, The Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, EMGO Institute for Health and Care Research and Neuroscience Campus Amsterdam, VU University Medical Center/GGZ ingest, Amsterdam, The Netherlands
| |
Collapse
|
40
|
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
|
41
|
do Carmo JM, da Silva AA, Romero DG, Hall JE. Changes in ambient temperature elicit divergent control of metabolic and cardiovascular actions by leptin. FASEB J 2017; 31:2418-2428. [PMID: 28228474 DOI: 10.1096/fj.201601224r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 01/30/2017] [Indexed: 11/11/2022]
Abstract
Interactions of hypothalamic signaling pathways that control body temperature (BT), blood pressure (BP), and energy balance are poorly understood. We investigated whether the chronic BP and metabolic actions of leptin are differentially modulated by changes in ambient temperature (TA ). Mean arterial pressure (MAP), heart rate (HR), BT, motor activity (MA), and oxygen consumption (Vo2) were measured 24 h/d at normal laboratory TA (23°C), at thermoneutral zone (TNZ, 30°C) for mice or during cold exposure (15°C) in male wild-type mice. After control measurements, leptin (4 μg/kg/min) or saline vehicle was infused for 7 d. At TNZ, leptin reduced food intake (-11.0 ± 0.5 g cumulative deficit) and body weight by 6% but caused no changes in MAP or HR. At 15°C, leptin infusion did not alter food intake but increased MAP and HR (8 ± 1 mmHg and 33 ± 7 bpm), while Vo2 increased by ∼10%. Leptin reduced plasma glucose and insulin levels at 15°C but not at 30°C. These results demonstrate that the chronic anorexic effects of leptin are enhanced at TNZ, while its effects on insulin and glucose levels are attenuated and its effects on BP and HR are abolished. Conversely, cold TA caused resistance to leptin's anorexic effects but amplified its effects to raise BP and reduce insulin and glucose levels. Thus, the brain circuits by which leptin regulates food intake and cardiovascular function are differentially influenced by changes in TA -Do Carmo, J. M., da Silva, A. A., Romero, D. G., Hall, J. E. Changes in ambient temperature elicit divergent control of metabolic and cardiovascular actions by leptin.
Collapse
Affiliation(s)
- Jussara M do Carmo
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi, USA;
| | - Alexandre A da Silva
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi, USA.,Centro Universitário Barão de Mauá, Ribeirão Preto, São Paulo, Brazil
| | - Damian G Romero
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - John E Hall
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
42
|
Burmeister MA, Ayala JE, Smouse H, Landivar-Rocha A, Brown JD, Drucker DJ, Stoffers DA, Sandoval DA, Seeley RJ, Ayala JE. The Hypothalamic Glucagon-Like Peptide 1 Receptor Is Sufficient but Not Necessary for the Regulation of Energy Balance and Glucose Homeostasis in Mice. Diabetes 2017; 66:372-384. [PMID: 27908915 PMCID: PMC5248999 DOI: 10.2337/db16-1102] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/23/2016] [Indexed: 12/16/2022]
Abstract
Pharmacological activation of the hypothalamic glucagon-like peptide 1 (GLP-1) receptor (GLP-1R) promotes weight loss and improves glucose tolerance. This demonstrates that the hypothalamic GLP-1R is sufficient but does not show whether it is necessary for the effects of exogenous GLP-1R agonists (GLP-1RA) or endogenous GLP-1 on these parameters. To address this, we crossed mice harboring floxed Glp1r alleles to mice expressing Nkx2.1-Cre to knock down Glp1r expression throughout the hypothalamus (GLP-1RKDΔNkx2.1cre). We also generated mice lacking Glp1r expression specifically in two GLP-1RA-responsive hypothalamic feeding nuclei/cell types, the paraventricular nucleus (GLP-1RKDΔSim1cre) and proopiomelanocortin neurons (GLP-1RKDΔPOMCcre). Chow-fed GLP-1RKDΔNkx2.1cre mice exhibited increased food intake and energy expenditure with no net effect on body weight. When fed a high-fat diet, these mice exhibited normal food intake but elevated energy expenditure, yielding reduced weight gain. None of these phenotypes were observed in GLP-1RKDΔSim1cre and GLP-1RKDΔPOMCcre mice. The acute anorectic and glucose tolerance effects of peripherally dosed GLP-1RA exendin-4 and liraglutide were preserved in all mouse lines. Chronic liraglutide treatment reduced body weight in chow-fed GLP-1RKDΔNkx2.1cre mice, but this effect was attenuated with high-fat diet feeding. In sum, classic homeostatic control regions are sufficient but not individually necessary for the effects of GLP-1RA on nutrient homeostasis.
Collapse
Affiliation(s)
- Melissa A Burmeister
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL
| | - Jennifer E Ayala
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL
| | - Hannah Smouse
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL
| | - Adriana Landivar-Rocha
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL
| | - Jacob D Brown
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL
| | - Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Doris A Stoffers
- Department of Medicine, University of Pennsylvania, Pennsylvania, PA
| | - Darleen A Sandoval
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI
| | - Randy J Seeley
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI
| | - Julio E Ayala
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL
| |
Collapse
|
43
|
A role for leptin-regulated neurocircuitry in subordination stress. Physiol Behav 2016; 178:144-150. [PMID: 27887997 DOI: 10.1016/j.physbeh.2016.11.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 11/07/2016] [Accepted: 11/18/2016] [Indexed: 02/07/2023]
Abstract
The visible burrow system produces a distinct combination of psychological and metabolic stress on, primarily, subordinate individuals that results in pronounced physiologic and behavioral dysfunction. However, the mechanisms underlying the consequences of chronic subordination stress are largely unknown. The simplest mechanistic explanation is that adaptations within brain systems with overlapping functions of both psychological and metabolic control provide immediate benefits that result in lasting susceptibility to diseases, disorders, and increased mortality rates in subordinates. Circuits regulated by leptin adapt to fluctuating levels of energy storage, such that the loss of leptin action within leptin-regulated neurocircuitry results in dysfunction in physiologic and behavioral systems implicated in the consequences of chronic social subordination. Thus, leptin-regulated neurocircuitry may provide a window into understanding the consequences of social subordination stress. This review examines the neural systems of leptin physiology implicated in social subordination stress: energy balance, motivation, HPA axis, and glycemic control.
Collapse
|
44
|
Abstract
The brain is considered a major site for microRNA (miRNA) expression; as evidenced by several studies reporting microarray data of different brain substructures. The hypothalamus is among the brain regions that plays a crucial role in integrating signals from other brain nuclei as well as environmental, hormonal, metabolic and neuronal signals from the periphery in order to deliver an adequate response. The hypothalamus controls vital functions such as reproduction, energy homeostasis, water balance, circadian rhythm and stress. These functions need a high neuronal plasticity to adequately respond to physiological, environmental and psychological stimuli that could be limited to a specific temporal period during life or are cyclic events. In this context, miRNAs constitute major regulators and coordinators of gene expression. Indeed, in response to specific stimuli, changes in miRNA expression profiles finely tune specific mRNA targets to adequately fit to the immediate needs through mainly the modulation of neuronal plasticity.
Collapse
Affiliation(s)
- Mohammed Taouis
- Molecular Neuroendocrinology of Food Intake (NMPA), UMR 9197, University Paris-Sud, Orsay, France; NMPA, Neurosciences Paris Saclay Institute (NeuroPSI), Department Molecules & Circuits, CNRS UMR 9197, Orsay, France.
| |
Collapse
|
45
|
Aujla PK, Bogdanovic V, Naratadam GT, Raetzman LT. Persistent expression of activated notch in the developing hypothalamus affects survival of pituitary progenitors and alters pituitary structure. Dev Dyn 2016; 244:921-34. [PMID: 25907274 DOI: 10.1002/dvdy.24283] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 04/10/2015] [Accepted: 04/13/2015] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND As the pituitary gland develops, signals from the hypothalamus are necessary for pituitary induction and expansion. Little is known about the control of cues that regulate early signaling between the two structures. Ligands and receptors of the Notch signaling pathway are found in both the hypothalamus and Rathke's pouch. The downstream Notch effector gene Hes1 is required for proper pituitary formation; however, these effects could be due to the action of Hes1 in the hypothalamus, Rathke's pouch, or both. To determine the contribution of hypothalamic Notch signaling to pituitary organogenesis, we used mice with loss and gain of Notch function within the developing hypothalamus. RESULTS We demonstrate that loss of Notch signaling by conditional deletion of Rbpj in the hypothalamus does not affect expression of Hes1 within the posterior hypothalamus or expression of Hes5. In contrast, expression of activated Notch within the hypothalamus results in ectopic Hes5 expression and increased Hes1 expression, which is sufficient to disrupt pituitary development and postnatal expansion. CONCLUSIONS Taken together, our results indicate that Rbpj-dependent Notch signaling within the developing hypothalamus is not necessary for pituitary development, but persistent Notch signaling and ectopic Hes5 expression in hypothalamic progenitors affects pituitary induction and expansion.
Collapse
Affiliation(s)
- Paven K Aujla
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Vedran Bogdanovic
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - George T Naratadam
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Lori T Raetzman
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
46
|
Zhao C, Castonguay TW. Effects of free access to sugar solutions on the control of energy intake. FOOD REVIEWS INTERNATIONAL 2016. [DOI: 10.1080/87559129.2016.1149863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Changhui Zhao
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland, USA
| | - Thomas W. Castonguay
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
47
|
Djogo T, Robins SC, Schneider S, Kryzskaya D, Liu X, Mingay A, Gillon CJ, Kim JH, Storch KF, Boehm U, Bourque CW, Stroh T, Dimou L, Kokoeva MV. Adult NG2-Glia Are Required for Median Eminence-Mediated Leptin Sensing and Body Weight Control. Cell Metab 2016; 23:797-810. [PMID: 27166944 DOI: 10.1016/j.cmet.2016.04.013] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/12/2016] [Accepted: 04/13/2016] [Indexed: 12/31/2022]
Abstract
While leptin is a well-known regulator of body fat mass, it remains unclear how circulating leptin is sensed centrally to maintain energy homeostasis. Here we show that genetic and pharmacological ablation of adult NG2-glia (also known as oligodendrocyte precursors), but not microglia, leads to primary leptin resistance and obesity in mice. We reveal that NG2-glia contact the dendritic processes of arcuate nucleus leptin receptor (LepR) neurons in the median eminence (ME) and that these processes degenerate upon NG2-glia elimination, which explains the consequential attenuation of these neurons' molecular and electrical responses to leptin. Our data therefore indicate that LepR dendrites in the ME represent the principal conduits of leptin's anorexigenic action and that NG2-glia are essential for their maintenance. Given that ME-directed X-irradiation confirmed the pharmacological and genetically mediated ablation effects on body weight, our findings provide a rationale for the known obesity risk associated with cranial radiation therapy.
Collapse
Affiliation(s)
- Tina Djogo
- Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal, QC H4A 3J1, Canada
| | - Sarah C Robins
- Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal, QC H4A 3J1, Canada
| | - Sarah Schneider
- Physiological Genomics, Institute of Physiology, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Darya Kryzskaya
- Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal, QC H4A 3J1, Canada
| | - Xiaohong Liu
- Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal, QC H4A 3J1, Canada
| | - Andrew Mingay
- Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal, QC H4A 3J1, Canada
| | - Colleen J Gillon
- Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal, QC H4A 3J1, Canada
| | - Joo Hyun Kim
- Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal, QC H4A 3J1, Canada
| | - Kai-Florian Storch
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
| | - Ulrich Boehm
- Department of Pharmacology and Toxicology, University of Saarland School of Medicine, 66421 Homburg, Germany
| | - Charles W Bourque
- Centre for Research in Neuroscience, Montreal General Hospital, McGill University, Montreal, QC H3G 1A4, Canada
| | - Thomas Stroh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Leda Dimou
- Physiological Genomics, Institute of Physiology, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Maia V Kokoeva
- Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal, QC H4A 3J1, Canada.
| |
Collapse
|
48
|
Zhang ZY, Dodd GT, Tiganis T. Protein Tyrosine Phosphatases in Hypothalamic Insulin and Leptin Signaling. Trends Pharmacol Sci 2016; 36:661-674. [PMID: 26435211 DOI: 10.1016/j.tips.2015.07.003] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/01/2015] [Accepted: 07/10/2015] [Indexed: 12/22/2022]
Abstract
The hypothalamus is critical to the coordination of energy balance and glucose homeostasis. It responds to peripheral factors, such as insulin and leptin, that convey to the brain the degree of adiposity and the metabolic status of the organism. The development of leptin and insulin resistance in hypothalamic neurons appears to have a key role in the exacerbation of diet-induced obesity. In rodents, this has been attributed partly to the increased expression of the tyrosine phosphatases Protein Tyrosine Phosphatase 1B (PTP1B) and T cell protein tyrosine phosphatase (TCPTP), which attenuate leptin and insulin signaling. Deficiencies in PTP1B and TCPTP in the brain, or specific neurons, promote insulin and leptin signaling and prevent diet-induced obesity, type 2 diabetes mellitus (T2DM), and fatty liver disease. Although targeting phosphatases and hypothalamic circuits remains challenging, recent advances indicate that such hurdles might be overcome. Here, we focus on the roles of PTP1B and TCPTP in insulin and leptin signaling and explore their potential as therapeutic targets.
Collapse
Affiliation(s)
- Zhong-Yin Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202-5126, USA
| | - Garron T Dodd
- Department of Biochemistry and Molecular Biology, Monash University, VIC 3800, Australia
| | - Tony Tiganis
- Department of Biochemistry and Molecular Biology, Monash University, VIC 3800, Australia.
| |
Collapse
|
49
|
Lerea JS, Ring LE, Hassouna R, Chong ACN, Szigeti-Buck K, Horvath TL, Zeltser LM. Reducing Adiposity in a Critical Developmental Window Has Lasting Benefits in Mice. Endocrinology 2016; 157:666-78. [PMID: 26587784 PMCID: PMC4733128 DOI: 10.1210/en.2015-1753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although most adults can lose weight by dieting, a well-characterized compensatory decrease in energy expenditure promotes weight regain more than 90% of the time. Using mice with impaired hypothalamic leptin signaling as a model of early-onset hyperphagia and obesity, we explored whether this unfavorable response to weight loss could be circumvented by early intervention. Early-onset obesity was associated with impairments in the structure and function of brown adipose tissue mitochondria, which were ameliorated by weight loss at any age. Although decreased sympathetic tone in weight-reduced adults resulted in net reductions in brown adipose tissue thermogenesis and energy expenditure that promoted rapid weight regain, this was not the case when dietary interventions were initiated at weaning. Enhanced energy expenditure persisted even after mice were allowed to resume overeating, leading to lasting reductions in adiposity. These findings reveal a time window when dietary interventions can produce metabolic improvements that are stably maintained.
Collapse
Affiliation(s)
- Jaclyn S Lerea
- Institute of Human Nutrition (J.S.L., A.C.N.N.), Columbia University, New York, New York 10032; Department of Anesthesiology (L.E.R.), Columbia University, New York, New York 10032; Naomi Berrie Diabetes Center (R.H., L.M.Z.), Columbia University, New York, New York 10032; Department of Obstetrics, Gynecology, and Reproductive Sciences (K.S.-B., T.L.H.), Yale University School of Medicine, New Haven, Connecticut 06510; Department of Neurobiology (T.L.H.), Yale University School of Medicine, New Haven, Connecticut 06510; and Department of Pathology and Cell Biology (L.M.Z.), Columbia University, New York, New York 10032
| | - Laurence E Ring
- Institute of Human Nutrition (J.S.L., A.C.N.N.), Columbia University, New York, New York 10032; Department of Anesthesiology (L.E.R.), Columbia University, New York, New York 10032; Naomi Berrie Diabetes Center (R.H., L.M.Z.), Columbia University, New York, New York 10032; Department of Obstetrics, Gynecology, and Reproductive Sciences (K.S.-B., T.L.H.), Yale University School of Medicine, New Haven, Connecticut 06510; Department of Neurobiology (T.L.H.), Yale University School of Medicine, New Haven, Connecticut 06510; and Department of Pathology and Cell Biology (L.M.Z.), Columbia University, New York, New York 10032
| | - Rim Hassouna
- Institute of Human Nutrition (J.S.L., A.C.N.N.), Columbia University, New York, New York 10032; Department of Anesthesiology (L.E.R.), Columbia University, New York, New York 10032; Naomi Berrie Diabetes Center (R.H., L.M.Z.), Columbia University, New York, New York 10032; Department of Obstetrics, Gynecology, and Reproductive Sciences (K.S.-B., T.L.H.), Yale University School of Medicine, New Haven, Connecticut 06510; Department of Neurobiology (T.L.H.), Yale University School of Medicine, New Haven, Connecticut 06510; and Department of Pathology and Cell Biology (L.M.Z.), Columbia University, New York, New York 10032
| | - Angie C N Chong
- Institute of Human Nutrition (J.S.L., A.C.N.N.), Columbia University, New York, New York 10032; Department of Anesthesiology (L.E.R.), Columbia University, New York, New York 10032; Naomi Berrie Diabetes Center (R.H., L.M.Z.), Columbia University, New York, New York 10032; Department of Obstetrics, Gynecology, and Reproductive Sciences (K.S.-B., T.L.H.), Yale University School of Medicine, New Haven, Connecticut 06510; Department of Neurobiology (T.L.H.), Yale University School of Medicine, New Haven, Connecticut 06510; and Department of Pathology and Cell Biology (L.M.Z.), Columbia University, New York, New York 10032
| | - Klara Szigeti-Buck
- Institute of Human Nutrition (J.S.L., A.C.N.N.), Columbia University, New York, New York 10032; Department of Anesthesiology (L.E.R.), Columbia University, New York, New York 10032; Naomi Berrie Diabetes Center (R.H., L.M.Z.), Columbia University, New York, New York 10032; Department of Obstetrics, Gynecology, and Reproductive Sciences (K.S.-B., T.L.H.), Yale University School of Medicine, New Haven, Connecticut 06510; Department of Neurobiology (T.L.H.), Yale University School of Medicine, New Haven, Connecticut 06510; and Department of Pathology and Cell Biology (L.M.Z.), Columbia University, New York, New York 10032
| | - Tamas L Horvath
- Institute of Human Nutrition (J.S.L., A.C.N.N.), Columbia University, New York, New York 10032; Department of Anesthesiology (L.E.R.), Columbia University, New York, New York 10032; Naomi Berrie Diabetes Center (R.H., L.M.Z.), Columbia University, New York, New York 10032; Department of Obstetrics, Gynecology, and Reproductive Sciences (K.S.-B., T.L.H.), Yale University School of Medicine, New Haven, Connecticut 06510; Department of Neurobiology (T.L.H.), Yale University School of Medicine, New Haven, Connecticut 06510; and Department of Pathology and Cell Biology (L.M.Z.), Columbia University, New York, New York 10032
| | - Lori M Zeltser
- Institute of Human Nutrition (J.S.L., A.C.N.N.), Columbia University, New York, New York 10032; Department of Anesthesiology (L.E.R.), Columbia University, New York, New York 10032; Naomi Berrie Diabetes Center (R.H., L.M.Z.), Columbia University, New York, New York 10032; Department of Obstetrics, Gynecology, and Reproductive Sciences (K.S.-B., T.L.H.), Yale University School of Medicine, New Haven, Connecticut 06510; Department of Neurobiology (T.L.H.), Yale University School of Medicine, New Haven, Connecticut 06510; and Department of Pathology and Cell Biology (L.M.Z.), Columbia University, New York, New York 10032
| |
Collapse
|
50
|
Abstract
Leptin is an adipocytokine that circulates in proportion to body fat to signal the repletion of long-term energy stores. Leptin acts via its receptor, LepRb, on specialized neuronal populations in the brain (mainly in the hypothalamus and brainstem) to alter motivation and satiety, as well as to permit energy expenditure and appropriate glucose homeostasis. Decreased leptin, as with prolonged caloric restriction, promotes a powerful orexigenic signal, decreases energy use via a number of neuroendocrine and autonomic axes, and disrupts glucose homeostasis. Here, we review what is known about cellular leptin action and focus on the roles for specific populations of LepRb-expressing neurons for leptin action.
Collapse
Affiliation(s)
- Jonathan N Flak
- Division of Metabolism, Endocrinology and Diabetes (J.N.F., M.G.M.), Department of Internal Medicine, and Department of Molecular and Integrative Physiology (M.G.M.), University of Michigan, Ann Arbor, Michigan 48109
| | - Martin G Myers
- Division of Metabolism, Endocrinology and Diabetes (J.N.F., M.G.M.), Department of Internal Medicine, and Department of Molecular and Integrative Physiology (M.G.M.), University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|