1
|
Reddy P, Narayan Prajapati J, Chaterji S, Varughese A, Chaudhary Y, Sathyamurthy A, Barik A. Converging inputs compete at the lateral parabrachial nuclei to dictate the affective-motivational responses to cold pain. Pain 2025; 166:1105-1117. [PMID: 39715193 DOI: 10.1097/j.pain.0000000000003468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 09/05/2024] [Indexed: 12/25/2024]
Abstract
ABSTRACT The neural mechanisms of the affective-motivational symptoms of chronic pain are poorly understood. In chronic pain, our innate coping mechanisms fail to provide relief. Hence, these behaviors are manifested at higher frequencies. In laboratory animals, such as mice and rats, licking the affected areas is a behavioral coping mechanism and it is sensitized in chronic pain. Hence, we have focused on delineating the brain circuits mediating licking in mice with chemotherapy-induced peripheral neuropathy (CIPN). Mice with CIPN develop intense cold hypersensitivity and lick their paws upon contact with cold stimuli. We studied how the lateral parabrachial nucleus (LPBN) neurons facilitate licking behavior when mice are exposed to noxious thermal stimuli. Taking advantage of transsynaptic viral, optogenetic, and chemogenetic strategies, we observed that the LPBN neurons become hypersensitive to cold in mice with CIPN and facilitate licks. Furthermore, we found that the expression of licks depends on competing excitatory and inhibitory inputs from the spinal cord and lateral hypothalamus (LHA), respectively. We anatomically traced the postsynaptic targets of the spinal cord and LHA in the LPBN and found that they synapse onto overlapping populations. Activation of this LPBN population was sufficient to promote licking due to cold allodynia. In sum, our data indicate that the nociceptive inputs from the spinal cord and information on brain states from the hypothalamus impinge on overlapping LPBN populations to modulate their activity and, in turn, regulate the elevated affective-motivational responses in CIPN.
Collapse
Affiliation(s)
- Prannay Reddy
- Center for Neuroscience, Indian Institute of Science, Bengaluru, Karnataka, India
| | | | | | | | | | | | | |
Collapse
|
2
|
Chen J, Cai M, Zhan C. Neuronal Regulation of Feeding and Energy Metabolism: A Focus on the Hypothalamus and Brainstem. Neurosci Bull 2025; 41:665-675. [PMID: 39704987 PMCID: PMC11978587 DOI: 10.1007/s12264-024-01335-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 09/19/2024] [Indexed: 12/21/2024] Open
Abstract
In the face of constantly changing environments, the central nervous system (CNS) rapidly and accurately calculates the body's needs, regulates feeding behavior, and maintains energy homeostasis. The arcuate nucleus of the hypothalamus (ARC) plays a key role in this process, serving as a critical brain region for detecting nutrition-related hormones and regulating appetite and energy homeostasis. Agouti-related protein (AgRP)/neuropeptide Y (NPY) neurons in the ARC are core elements that interact with other brain regions through a complex appetite-regulating network to comprehensively control energy homeostasis. In this review, we explore the discovery and research progress of AgRP neurons in regulating feeding and energy metabolism. In addition, recent advances in terms of feeding behavior and energy homeostasis, along with the redundant neural mechanisms involved in energy metabolism, are discussed. Finally, the challenges and opportunities in the field of neural regulation of feeding and energy metabolism are briefly discussed.
Collapse
Affiliation(s)
- Jing Chen
- Department of Neurosurgery, Xuanwu Hospital, China International Neuroscience Institute, Capital Medical University, Beijing, 100053, China
| | - Meiting Cai
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Key Laboratory of Immune Response and Immunotherapy, CAS Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Cheng Zhan
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Key Laboratory of Immune Response and Immunotherapy, CAS Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
3
|
Mendoza-Romero HN, Biddinger JE, Bedenbaugh MN, Simerly RB. Microglia are Required for Developmental Specification of AgRP Innervation in the Hypothalamus of Offspring Exposed to Maternal High-Fat Diet During Lactation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.12.607566. [PMID: 39185162 PMCID: PMC11343114 DOI: 10.1101/2024.08.12.607566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Agouti-related peptide (AgRP) neurons in the arcuate nucleus of the hypothalamus respond to multiple metabolic signals and distribute neuroendocrine information to other brain regions such as the paraventricular hypothalamic nucleus (PVH), which plays a central role in metabolic homeostasis. Neural projections from AgRP neurons to the PVH form during the postnatal lactational period in mice and these projections are reduced in offspring of dams that consumed a high-fat diet (HFD) during lactation (MHFD-L). Here we used immunohistochemistry to visualize microglial morphology in MHFD-L offspring and identified changes that were regionally localized to the PVH and appeared temporally restricted to the period when AgRP neurons innervate this region. In addition, axon labeling experiments revealed that microglia engulf AgRP terminals in the PVH, and that the density of AgRP innervation to the PVH in MHFD-L offspring may be dependent on microglia, because microglial depletion blocked the decrease in PVH AgRP innervation observed in MHFD-L offspring, as well as prevented the increased body weight exhibited at weaning. Together, these findings suggest that microglia are activated by exposure to MHFD-L and interact directly with AgRP axons during postnatal development to permanently alter innervation of the PVH, with implications for developmental programming of metabolic phenotype.
Collapse
Affiliation(s)
| | - Jessica E. Biddinger
- Dept of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Michelle N. Bedenbaugh
- Dept of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Richard B. Simerly
- Dept of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
4
|
Bernecker M, Lin A, Feuchtinger A, Molenaar A, Schriever SC, Pfluger PT. Weight cycling exacerbates glucose intolerance and hepatic triglyceride storage in mice with a history of chronic high fat diet exposure. J Transl Med 2025; 23:7. [PMID: 39754229 PMCID: PMC11699648 DOI: 10.1186/s12967-024-06039-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/25/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Obese subjects undergoing weight loss often fear the Yoyo dieting effect, which involves regaining or even surpassing their initial weight. To date, our understanding of such long-term obesity and weight cycling effects is still limited and often based on only short-term murine weight gain and loss studies. This study aimed to investigate the long-term impacts of weight cycling on glycemic control and metabolic health, focusing on adipose tissue, liver, and hypothalamus. METHODS Chow-fed mice and mice subjected to prolonged high-fat diet (HFD) consumption for 20 weeks, followed by 24 weeks of dietary interventions to either induce weight gain, weight loss, or weight cycling were monitored for perturbations in feeding efficiency and glucose homeostasis. Post-mortem analyses included qPCR, Western Blotting, biochemical and microscopical assessments for hepatic steatosis and insulin resistance, hypothalamic and adipose tissue inflammation, and circulating lipid, leptin and IL-6 levels. RESULTS Weight cycling led to hyperphagia and rapid weight regain, matching the weights of mice continuously on HFD. Despite weight loss, adipose tissue inflammation persisted with elevated pro-inflammatory markers, macrophage infiltration, and impaired Glut4 expression. HFD-induced dysregulation in hypothalamic expression of orexigenic peptides and synaptic plasticity markers persisted also after weight normalization suggesting long-lasting neural alterations. Weight-cycled mice exhibited higher circulating IL-6 and leptin levels, increased hepatic lipid storage, and dysregulated glucose metabolism compared to those with consistent diets, indicating worsened metabolic effects by Yoyo dieting. CONCLUSION In sum, our study highlights significant metabolic risks associated with weight cycling, particularly following prolonged obesity. Persistent adipose tissue inflammation, perturbed neural peptide and plasticity markers and impaired glucose tolerance emphasize the need for effective and sustainable weight loss strategies to mitigate the adverse outcomes of weight regain and improve long-term metabolic health.
Collapse
Affiliation(s)
- Miriam Bernecker
- Research Unit NeuroBiology of Diabetes, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- Division of NeuroBiology of Diabetes, TUM School of Medicine & Health, Technical University of Munich, Munich, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Anna Lin
- Research Unit NeuroBiology of Diabetes, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Annette Feuchtinger
- Core Facility Pathology and Tissue Analytics, Helmholtz Munich, Neuherberg, Germany
| | - Anna Molenaar
- Research Unit NeuroBiology of Diabetes, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- Division of NeuroBiology of Diabetes, TUM School of Medicine & Health, Technical University of Munich, Munich, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Sonja C Schriever
- Research Unit NeuroBiology of Diabetes, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Paul T Pfluger
- Research Unit NeuroBiology of Diabetes, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany.
- Division of NeuroBiology of Diabetes, TUM School of Medicine & Health, Technical University of Munich, Munich, Germany.
- German Center for Diabetes Research, Neuherberg, Germany.
| |
Collapse
|
5
|
Yang S, Li Y, Tian M, Deng W, Liu D, Chen C, Zhu Z, Zheng H, Yang G, Li L, Yang M. Hypothalamic P62 (SQSTM1) regulates energy balance by modulating leptin signaling. Theranostics 2024; 14:6605-6624. [PMID: 39479445 PMCID: PMC11519807 DOI: 10.7150/thno.96480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 09/22/2024] [Indexed: 10/30/2024] Open
Abstract
RATIONALE The multifaceted functions of p62 (SQSTM1) are increasingly recognized, but its role in hypothalamic metabolism-associated neurons for energy balance has yet to be elucidated. METHODS Single-nucleus RNA sequencing (snRNA-Seq) was performed on hypothalamic tissues from db/db and db/m mice to explore p62 expression. Overexpression and knockout of p62 in hypothalamic POMC neurons were performed via AAV-mediated gene delivery and Cre-loxP systems. Metabolic outcomes were assessed under normal chow (NCD) and high-fat diet (HFD) conditions. The co-immunoprecipitation and luciferase reporter assays were used to investigate the interaction between p62 and STAT3. RESULTS The snRNA-Seq analysis found that p62 was ubiquitously expressed in hypothalamic neurons, with significantly higher levels in POMC neurons of db/db mice compared to db/m controls. Under NCD or HFD conditions, the absence of p62 in POMC neurons led to increased body weight, decreased energy expenditure and leptin sensitivity, while its overexpression in POMC neurons produced the opposite phenotype. Mechanistically, p62 interacts with STAT3, facilitating its phosphorylation to initiate POMC transcription and amplify leptin sensitivity. CONCLUSION This study demonstrated the capacity of p62 to monogenically regulate the obesity phenotype and emphasized its dual role in managing energy homeostasis through direct modulation of STAT3/POMC signaling and amplification of leptin sensitivity.
Collapse
Affiliation(s)
- Shan Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yang Li
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Mingyuan Tian
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Clinical Biochemistry and the Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Wuquan Deng
- Department of Endocrinology and Metabolism, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Jiankang Road, Yuzhong District, Chongqing, China
| | - Dongfang Liu
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Chen Chen
- Endocrinology, SBMS, Faculty of Medicine, University of Queensland, Brisbane, 4072, Australia
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Hongting Zheng
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Gangyi Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ling Li
- Department of Clinical Biochemistry and the Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Mengliu Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Berbegal-Sáez P, Gallego-Landin I, Macía J, Alegre-Zurano L, Castro-Zavala A, Welz PS, Benitah SA, Valverde O. Lack of Bmal1 leads to changes in rhythmicity and impairs motivation towards natural stimuli. Open Biol 2024; 14:240051. [PMID: 39045857 PMCID: PMC11267724 DOI: 10.1098/rsob.240051] [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: 04/15/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 07/25/2024] Open
Abstract
Maintaining proper circadian rhythms is essential for coordinating biological functions in mammals. This study investigates the effects of daily arrhythmicity using Bmal1-knockout (KO) mice as a model, aiming to understand behavioural and motivational implications. By employing a new mathematical analysis based on entropy divergence, we identified disrupted intricate activity patterns in mice derived by the complete absence of BMAL1 and quantified the difference regarding the activity oscillation's complexity. Changes in locomotor activity coincided with disturbances in circadian gene expression patterns. Additionally, we found a dysregulated gene expression profile particularly in brain nuclei like the ventral striatum, impacting genes related to reward and motivation. Further investigation revealed that arrhythmic mice exhibited heightened motivation for food and water rewards, indicating a link between circadian disruptions and the reward system. This research sheds light on how circadian clock alterations impact the gene expression regulating the reward system and how this, in turn, can lead to altered seeking behaviour and motivation for natural rewards. In summary, the present study contributes to our understanding of how reward processing is under the regulation of circadian clock machinery.
Collapse
Affiliation(s)
- Paula Berbegal-Sáez
- Department of Medicine and Life Sciences (MELIS), Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Universitat Pompeu Fabra, Barcelona, Spain
| | - Ines Gallego-Landin
- Department of Medicine and Life Sciences (MELIS), Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Universitat Pompeu Fabra, Barcelona, Spain
| | - Javier Macía
- Department of Medicine and Life Sciences (MELIS), Synthetic Biology for Biomedical Applications, Universitat Pompeu Fabra, Barcelona, Spain
| | - Laia Alegre-Zurano
- Department of Medicine and Life Sciences (MELIS), Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Universitat Pompeu Fabra, Barcelona, Spain
| | - Adriana Castro-Zavala
- Department of Medicine and Life Sciences (MELIS), Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Universitat Pompeu Fabra, Barcelona, Spain
| | - Patrick-Simon Welz
- Program in Cancer Research, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Salvador A. Benitah
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Barcelon08028, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Olga Valverde
- Department of Medicine and Life Sciences (MELIS), Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Universitat Pompeu Fabra, Barcelona, Spain
- Neuroscience Research Program, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| |
Collapse
|
7
|
Lee HG, Jung IH, Park BS, Yang HR, Kim KK, Tu TH, Yeh JY, Lee S, Yang S, Lee BJ, Kim JG, Nam-Goong IS. Altered Metabolic Phenotypes and Hypothalamic Neuronal Activity Triggered by Sodium-Glucose Cotransporter 2 Inhibition. Diabetes Metab J 2023; 47:784-795. [PMID: 37915185 PMCID: PMC10695712 DOI: 10.4093/dmj.2022.0261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 01/17/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGRUOUND Sodium-glucose cotransporter 2 (SGLT-2) inhibitors are currently used to treat patients with diabetes. Previous studies have demonstrated that treatment with SGLT-2 inhibitors is accompanied by altered metabolic phenotypes. However, it has not been investigated whether the hypothalamic circuit participates in the development of the compensatory metabolic phenotypes triggered by the treatment with SGLT-2 inhibitors. METHODS Mice were fed a standard diet or high-fat diet and treated with dapagliflozin, an SGLT-2 inhibitor. Food intake and energy expenditure were observed using indirect calorimetry system. The activity of hypothalamic neurons in response to dapagliflozin treatment was evaluated by immunohistochemistry with c-Fos antibody. Quantitative real-time polymerase chain reaction was performed to determine gene expression patterns in the hypothalamus of dapagliflozin-treated mice. RESULTS Dapagliflozin-treated mice displayed enhanced food intake and reduced energy expenditure. Altered neuronal activities were observed in multiple hypothalamic nuclei in association with appetite regulation. Additionally, we found elevated immunosignals of agouti-related peptide neurons in the paraventricular nucleus of the hypothalamus. CONCLUSION This study suggests the functional involvement of the hypothalamus in the development of the compensatory metabolic phenotypes induced by SGLT-2 inhibitor treatment.
Collapse
Affiliation(s)
- Ho Gyun Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea
| | - Il Hyeon Jung
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea
| | - Byong Seo Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea
| | - Hye Rim Yang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea
| | - Kwang Kon Kim
- Department of Biological Science, University of Ulsan, Ulsan, Korea
| | - Thai Hien Tu
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea
| | - Jung-Yong Yeh
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea
| | - Sewon Lee
- Division of Sport Science, College of Arts & Physical Education, Incheon National University, Incheon, Korea
- Research Center of Brain-Machine Interface, Incheon National University, Incheon, Korea
| | - Sunggu Yang
- Research Center of Brain-Machine Interface, Incheon National University, Incheon, Korea
- Department of Nano-Bioengineering, College of Life Science and Technology, Incheon National University, Incheon, Korea
| | - Byung Ju Lee
- Department of Biological Science, University of Ulsan, Ulsan, Korea
| | - Jae Geun Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea
- Research Center of Brain-Machine Interface, Incheon National University, Incheon, Korea
| | - Il Seong Nam-Goong
- Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| |
Collapse
|
8
|
Kim KK, Lee TH, Park BS, Kang D, Kim DH, Jeong B, Kim JW, Yang HR, Kim HR, Jin S, Back SH, Park JW, Kim JG, Lee BJ. Bridging Energy Need and Feeding Behavior: The Impact of eIF2α Phosphorylation in AgRP Neurons. Diabetes 2023; 72:1384-1396. [PMID: 37478284 DOI: 10.2337/db23-0004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023]
Abstract
Eukaryotic translation initiation factor 2α (eIF2α) is a key mediator of the endoplasmic reticulum (ER) stress-induced unfolded protein response (UPR). In mammals, eIF2α is phosphorylated by overnutrition-induced ER stress and is related to the development of obesity. Here, we studied the function of phosphorylated eIF2α (p-eIF2α) in agouti-related peptide (AgRP) neurons using a mouse model (AgRPeIF2αA/A) with an AgRP neuron-specific substitution from Ser 51 to Ala in eIF2α, which impairs eIF2α phosphorylation in AgRP neurons. These AgRPeIF2αA/A mice had decreases in starvation-induced AgRP neuronal activity and food intake and an increased responsiveness to leptin. Intriguingly, impairment of eIF2α phosphorylation produced decreases in the starvation-induced expression of UPR and autophagy genes in AgRP neurons. Collectively, these findings suggest that eIF2α phosphorylation regulates AgRP neuronal activity by affecting intracellular responses such as the UPR and autophagy during starvation, thereby participating in the homeostatic control of whole-body energy metabolism. ARTICLE HIGHLIGHTS This study examines the impact of eukaryotic translation initiation factor 2α (eIF2α) phosphorylation, triggered by an energy deficit, on hypothalamic AgRP neurons and its subsequent influence on whole-body energy homeostasis. Impaired eIF2α phosphorylation diminishes the unfolded protein response and autophagy, both of which are crucial for energy deficit-induced activation of AgRP neurons. This study highlights the significance of eIF2α phosphorylation as a cellular marker indicating the availability of energy in AgRP neurons and as a molecular switch that regulates homeostatic feeding behavior.
Collapse
Affiliation(s)
- Kwang Kon Kim
- Department of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea
| | - Tae Hwan Lee
- Department of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea
| | - Byong Seo Park
- Department of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Dasol Kang
- Department of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea
| | - Dong Hee Kim
- Department of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea
| | - Bora Jeong
- Department of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea
| | - Jin Woo Kim
- Department of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea
| | - Hye Rim Yang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Han Rae Kim
- Department of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, George Washington University, Washington, DC
| | - Sungho Jin
- Department of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY
| | - Sung Hoon Back
- Department of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea
- Basic-Clinical Convergence Research Center, University of Ulsan, Ulsan, Republic of Korea
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea
- Basic-Clinical Convergence Research Center, University of Ulsan, Ulsan, Republic of Korea
| | - Jae Geun Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea
- Basic-Clinical Convergence Research Center, University of Ulsan, Ulsan, Republic of Korea
| |
Collapse
|
9
|
Zagmutt S, Mera P, González-García I, Ibeas K, Romero MDM, Obri A, Martin B, Esteve-Codina A, Soler-Vázquez MC, Bastias-Pérez M, Cañes L, Augé E, Pelegri C, Vilaplana J, Ariza X, García J, Martinez-González J, Casals N, López M, Palmiter R, Sanz E, Quintana A, Herrero L, Serra D. CPT1A in AgRP neurons is required for sex-dependent regulation of feeding and thirst. Biol Sex Differ 2023; 14:14. [PMID: 36966335 PMCID: PMC10040140 DOI: 10.1186/s13293-023-00498-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/10/2023] [Indexed: 03/27/2023] Open
Abstract
BACKGROUND Fatty acid metabolism in the hypothalamus has an important role in food intake, but its specific role in AgRP neurons is poorly understood. Here, we examined whether carnitinea palmitoyltransferase 1A (CPT1A), a key enzyme in mitochondrial fatty acid oxidation, affects energy balance. METHODS To obtain Cpt1aKO mice and their control littermates, Cpt1a(flox/flox) mice were crossed with tamoxifen-inducible AgRPCreERT2 mice. Food intake and body weight were analyzed weekly in both males and females. At 12 weeks of age, metabolic flexibility was determined by ghrelin-induced food intake and fasting-refeeding satiety tests. Energy expenditure was analyzed by calorimetric system and thermogenic activity of brown adipose tissue. To study fluid balance the analysis of urine and water intake volumes; osmolality of urine and plasma; as well as serum levels of angiotensin and components of RAAS (renin-angiotensin-aldosterone system) were measured. At the central level, changes in AgRP neurons were determined by: (1) analyzing specific AgRP gene expression in RiboTag-Cpt1aKO mice obtained by crossing Cpt1aKO mice with RiboTag mice; (2) measuring presynaptic terminal formation in the AgRP neurons with the injection of the AAV1-EF1a-DIO-synaptophysin-GFP in the arcuate nucleus of the hypothalamus; (3) analyzing AgRP neuronal viability and spine formations by the injection AAV9-EF1a-DIO-mCherry in the arcuate nucleus of the hypothalamus; (4) analyzing in situ the specific AgRP mitochondria in the ZsGreen-Cpt1aKO obtained by breeding ZsGreen mice with Cpt1aKO mice. Two-way ANOVA analyses were performed to determine the contributions of the effect of lack of CPT1A in AgRP neurons in the sex. RESULTS Changes in food intake were just seen in male Cpt1aKO mice while only female Cpt1aKO mice increased energy expenditure. The lack of Cpt1a in the AgRP neurons enhanced brown adipose tissue activity, mainly in females, and induced a substantial reduction in fat deposits and body weight. Strikingly, both male and female Cpt1aKO mice showed polydipsia and polyuria, with more reduced serum vasopressin levels in females and without osmolality alterations, indicating a direct involvement of Cpt1a in AgRP neurons in fluid balance. AgRP neurons from Cpt1aKO mice showed a sex-dependent gene expression pattern, reduced mitochondria and decreased presynaptic innervation to the paraventricular nucleus, without neuronal viability alterations. CONCLUSIONS Our results highlight that fatty acid metabolism and CPT1A in AgRP neurons show marked sex differences and play a relevant role in the neuronal processes necessary for the maintenance of whole-body fluid and energy balance.
Collapse
Affiliation(s)
- Sebastián Zagmutt
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Paula Mera
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Ismael González-García
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Kevin Ibeas
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - María Del Mar Romero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Arnaud Obri
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Beatriz Martin
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - M Carmen Soler-Vázquez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Marianela Bastias-Pérez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Laia Cañes
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Elisabeth Augé
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Carme Pelegri
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Neurosciences of the Universitat de Barcelona, Barcelona, Spain
| | - Jordi Vilaplana
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Neurosciences of the Universitat de Barcelona, Barcelona, Spain
| | - Xavier Ariza
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Inorganic & Organic Chemistry, Faculty of Chemistry, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Jordi García
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Inorganic & Organic Chemistry, Faculty of Chemistry, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - José Martinez-González
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Núria Casals
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Basic Sciences, Faculty of Medicine & Health Sciences, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Spain
| | - Miguel López
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Richard Palmiter
- Department of Biochemistry, Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Elisenda Sanz
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Albert Quintana
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain.
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
10
|
Lee YH, Kim YB, Kim KS, Jang M, Song HY, Jung SH, Ha DS, Park JS, Lee J, Kim KM, Cheon DH, Baek I, Shin MG, Lee EJ, Kim SJ, Choi HJ. Lateral hypothalamic leptin receptor neurons drive hunger-gated food-seeking and consummatory behaviours in male mice. Nat Commun 2023; 14:1486. [PMID: 36932069 PMCID: PMC10023672 DOI: 10.1038/s41467-023-37044-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/01/2023] [Indexed: 03/19/2023] Open
Abstract
For survival, it is crucial for eating behaviours to be sequenced through two distinct seeking and consummatory phases. Heterogeneous lateral hypothalamus (LH) neurons are known to regulate motivated behaviours, yet which subpopulation drives food seeking and consummatory behaviours have not been fully addressed. Here, in male mice, fibre photometry recordings demonstrated that LH leptin receptor (LepR) neurons are correlated explicitly in both voluntary seeking and consummatory behaviours. Further, micro-endoscope recording of the LHLepR neurons demonstrated that one subpopulation is time-locked to seeking behaviours and the other subpopulation time-locked to consummatory behaviours. Seeking or consummatory phase specific paradigm revealed that activation of LHLepR neurons promotes seeking or consummatory behaviours and inhibition of LHLepR neurons reduces consummatory behaviours. The activity of LHLepR neurons was increased via Neuropeptide Y (NPY) which acted as a tonic permissive gate signal. Our results identify neural populations that mediate seeking and consummatory behaviours and may lead to therapeutic targets for maladaptive food seeking and consummatory behaviours.
Collapse
Affiliation(s)
- Young Hee Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Yu-Been Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Kyu Sik Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Mirae Jang
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Ha Young Song
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Sang-Ho Jung
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Dong-Soo Ha
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Joon Seok Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Jaegeon Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Kyung Min Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Deok-Hyeon Cheon
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Inhyeok Baek
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Min-Gi Shin
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Eun Jeong Lee
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Sang Jeong Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Wide River Institute of Immunology, Seoul National University, 101 Dabyeonbat-gil, Hwachon-myeon, Gangwon-do, 25159, Republic of Korea
| | - Hyung Jin Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Neuroscience Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Wide River Institute of Immunology, Seoul National University, 101 Dabyeonbat-gil, Hwachon-myeon, Gangwon-do, 25159, Republic of Korea.
| |
Collapse
|
11
|
Tsuneki H, Sugiyama M, Ito T, Sato K, Matsuda H, Onishi K, Yubune K, Matsuoka Y, Nagai S, Yamagishi T, Maeda T, Honda K, Okekawa A, Watanabe S, Yaku K, Okuzaki D, Otsubo R, Nomoto M, Inokuchi K, Nakagawa T, Wada T, Yasui T, Sasaoka T. Food odor perception promotes systemic lipid utilization. Nat Metab 2022; 4:1514-1531. [PMID: 36376564 DOI: 10.1038/s42255-022-00673-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 09/30/2022] [Indexed: 11/16/2022]
Abstract
Food cues during fasting elicit Pavlovian conditioning to adapt for anticipated food intake. However, whether the olfactory system is involved in metabolic adaptations remains elusive. Here we show that food-odor perception promotes lipid metabolism in male mice. During fasting, food-odor stimulation is sufficient to increase serum free fatty acids via adipose tissue lipolysis in an olfactory-memory-dependent manner, which is mediated by the central melanocortin and sympathetic nervous systems. Additionally, stimulation with a food odor prior to refeeding leads to enhanced whole-body lipid utilization, which is associated with increased sensitivity of the central agouti-related peptide system, reduced sympathetic activity and peripheral tissue-specific metabolic alterations, such as an increase in gastrointestinal lipid absorption and hepatic cholesterol turnover. Finally, we show that intermittent fasting coupled with food-odor stimulation improves glycemic control and prevents insulin resistance in diet-induced obese mice. Thus, olfactory regulation is required for maintaining metabolic homeostasis in environments with either an energy deficit or energy surplus, which could be considered as part of dietary interventions against metabolic disorders.
Collapse
Affiliation(s)
- Hiroshi Tsuneki
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan.
| | - Masanori Sugiyama
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Toshihiro Ito
- Laboratory of Proteome Research, National Institutes of Biomedical Innovation, Health, and Nutrition, Osaka, Japan
| | - Kiyofumi Sato
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Hiroki Matsuda
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Kengo Onishi
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Koharu Yubune
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Yukina Matsuoka
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Sanaka Nagai
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Towa Yamagishi
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Takahiro Maeda
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Kosuke Honda
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Akira Okekawa
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Shiro Watanabe
- Division of Nutritional Biochemistry, University of Toyama, Toyama, Japan
| | - Keisuke Yaku
- Department of Molecular and Medical Pharmacology, University of Toyama, Toyama, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Ryota Otsubo
- Laboratory of Infectious Diseases and Immunity, National Institutes of Biomedical Innovation, Health, and Nutrition, Osaka, Japan
- Laboratory of Immunobiologics Evaluation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health, and Nutrition, Osaka, Japan
| | - Masanori Nomoto
- Department of Biochemistry, University of Toyama, Toyama, Japan
- Research Centre for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Kaoru Inokuchi
- Department of Biochemistry, University of Toyama, Toyama, Japan
- Research Centre for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Takashi Nakagawa
- Department of Molecular and Medical Pharmacology, University of Toyama, Toyama, Japan
| | - Tsutomu Wada
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Teruhito Yasui
- Laboratory of Infectious Diseases and Immunity, National Institutes of Biomedical Innovation, Health, and Nutrition, Osaka, Japan.
- Laboratory of Immunobiologics Evaluation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health, and Nutrition, Osaka, Japan.
- Laboratory of Pharmaceutical Integrated Omics, Department of Pharmaceutical Engineering, Facility of Engineering, Toyama Prefectural University, Toyama, Japan.
| | - Toshiyasu Sasaoka
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan.
| |
Collapse
|
12
|
Angelidi AM, Belanger MJ, Kokkinos A, Koliaki CC, Mantzoros CS. Novel Noninvasive Approaches to the Treatment of Obesity: From Pharmacotherapy to Gene Therapy. Endocr Rev 2022; 43:507-557. [PMID: 35552683 PMCID: PMC9113190 DOI: 10.1210/endrev/bnab034] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Indexed: 02/08/2023]
Abstract
Recent insights into the pathophysiologic underlying mechanisms of obesity have led to the discovery of several promising drug targets and novel therapeutic strategies to address the global obesity epidemic and its comorbidities. Current pharmacologic options for obesity management are largely limited in number and of modest efficacy/safety profile. Therefore, the need for safe and more efficacious new agents is urgent. Drugs that are currently under investigation modulate targets across a broad range of systems and tissues, including the central nervous system, gastrointestinal hormones, adipose tissue, kidney, liver, and skeletal muscle. Beyond pharmacotherapeutics, other potential antiobesity strategies are being explored, including novel drug delivery systems, vaccines, modulation of the gut microbiome, and gene therapy. The present review summarizes the pathophysiology of energy homeostasis and highlights pathways being explored in the effort to develop novel antiobesity medications and interventions but does not cover devices and bariatric methods. Emerging pharmacologic agents and alternative approaches targeting these pathways and relevant research in both animals and humans are presented in detail. Special emphasis is given to treatment options at the end of the development pipeline and closer to the clinic (ie, compounds that have a higher chance to be added to our therapeutic armamentarium in the near future). Ultimately, advancements in our understanding of the pathophysiology and interindividual variation of obesity may lead to multimodal and personalized approaches to obesity treatment that will result in safe, effective, and sustainable weight loss until the root causes of the problem are identified and addressed.
Collapse
Affiliation(s)
- Angeliki M Angelidi
- Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Matthew J Belanger
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alexander Kokkinos
- First Department of Propaedeutic Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Chrysi C Koliaki
- First Department of Propaedeutic Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Christos S Mantzoros
- Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
NPFF Decreases Activity of Human Arcuate NPY Neurons: A Study in Embryonic-Stem-Cell-Derived Model. Int J Mol Sci 2022; 23:ijms23063260. [PMID: 35328681 PMCID: PMC8948797 DOI: 10.3390/ijms23063260] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 12/10/2022] Open
Abstract
Restoring the control of food intake is the key to obesity management and prevention. The arcuate nucleus (ARC) of the hypothalamus is extensively being studied as a potential anti-obesity target. Animal studies showed that neuropeptide FF (NPFF) reduces food intake by its action in neuropeptide Y (NPY) neurons of the hypothalamic ARC, but the detailed mode of action observed in human neurons is missing, due to the lack of a human-neuron-based model for pharmacology testing. Here, we validated and utilized a human-neural-stem-cell-based (hNSC) model of ARC to test the effects of NPFF on cellular pathways and neuronal activity. We found that in the human neurons, decreased cAMP levels by NPFF resulted in a reduced rate of cytoplasmic calcium oscillations, indicating an inhibition of ARC NPY neurons. This suggests the therapeutic potential of NPFFR2 in obesity. In addition, we demonstrate the use of human-stem-cell-derived neurons in pharmacological applications and the potential of this model to address functional aspects of human hypothalamic neurons.
Collapse
|
14
|
Naito M, Iwakoshi-Ukena E, Moriwaki S, Narimatsu Y, Kato M, Furumitsu M, Miyamoto Y, Esumi S, Ukena K. Immunohistochemical Analysis of Neurotransmitters in Neurosecretory Protein GL-Producing Neurons of the Mouse Hypothalamus. Biomedicines 2022; 10:biomedicines10020454. [PMID: 35203663 PMCID: PMC8962320 DOI: 10.3390/biomedicines10020454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 01/03/2023] Open
Abstract
We recently discovered a novel neuropeptide of 80 amino acid residues: neurosecretory protein GL (NPGL), in the hypothalamus of birds and rodents. NPGL is localized in the lateral posterior part of the arcuate nucleus (ArcLP), and it enhances feeding behavior and fat accumulation in mice. Various neurotransmitters, such as catecholamine, glutamate, and γ-aminobutyric acid (GABA), produced in the hypothalamus are also involved in energy metabolism. The colocalization of neurotransmitters and NPGL in neurons of the ArcLP leads to the elucidation of the regulatory mechanism of NPGL neurons. In this study, we performed double immunofluorescence staining to elucidate the relationship between NPGL and neurotransmitters in mice. The present study revealed that NPGL neurons did not co-express tyrosine hydroxylase as a marker of catecholaminergic neurons and vesicular glutamate transporter-2 as a marker of glutamatergic neurons. In contrast, NPGL neurons co-produced glutamate decarboxylase 67, a marker for GABAergic neurons. In addition, approximately 50% of NPGL neurons were identical to GABAergic neurons. These results suggest that some functions of NPGL neurons may be related to those of GABA. This study provides insights into the neural network of NPGL neurons that regulate energy homeostasis, including feeding behavior and fat accumulation.
Collapse
Affiliation(s)
- Mana Naito
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (M.N.); (E.I.-U.); (S.M.); (Y.N.); (M.K.); (M.F.)
| | - Eiko Iwakoshi-Ukena
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (M.N.); (E.I.-U.); (S.M.); (Y.N.); (M.K.); (M.F.)
| | - Shogo Moriwaki
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (M.N.); (E.I.-U.); (S.M.); (Y.N.); (M.K.); (M.F.)
| | - Yuki Narimatsu
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (M.N.); (E.I.-U.); (S.M.); (Y.N.); (M.K.); (M.F.)
| | - Masaki Kato
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (M.N.); (E.I.-U.); (S.M.); (Y.N.); (M.K.); (M.F.)
| | - Megumi Furumitsu
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (M.N.); (E.I.-U.); (S.M.); (Y.N.); (M.K.); (M.F.)
| | - Yuta Miyamoto
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (Y.M.); (S.E.)
| | - Shigeyuki Esumi
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (Y.M.); (S.E.)
| | - Kazuyoshi Ukena
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (M.N.); (E.I.-U.); (S.M.); (Y.N.); (M.K.); (M.F.)
- Correspondence:
| |
Collapse
|
15
|
Al-Massadi O, Dieguez C, Schneeberger M, López M, Schwaninger M, Prevot V, Nogueiras R. Multifaceted actions of melanin-concentrating hormone on mammalian energy homeostasis. Nat Rev Endocrinol 2021; 17:745-755. [PMID: 34608277 DOI: 10.1038/s41574-021-00559-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2021] [Indexed: 12/12/2022]
Abstract
Melanin-concentrating hormone (MCH) is a small cyclic peptide expressed in all mammals, mainly in the hypothalamus. MCH acts as a robust integrator of several physiological functions and has crucial roles in the regulation of sleep-wake rhythms, feeding behaviour and metabolism. MCH signalling has a very broad endocrine context and is involved in physiological functions and emotional states associated with metabolism, such as reproduction, anxiety, depression, sleep and circadian rhythms. MCH mediates its functions through two receptors (MCHR1 and MCHR2), of which only MCHR1 is common to all mammals. Owing to the wide variety of MCH downstream signalling pathways, MCHR1 agonists and antagonists have great potential as tools for the directed management of energy balance disorders and associated metabolic complications, and translational strategies using these compounds hold promise for the development of novel treatments for obesity. This Review provides an overview of the numerous roles of MCH in energy and glucose homeostasis, as well as in regulation of the mesolimbic dopaminergic circuits that encode the hedonic component of food intake.
Collapse
Affiliation(s)
- Omar Al-Massadi
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Spain.
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain.
| | - Carlos Dieguez
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Marc Schneeberger
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Miguel López
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience and Cognition, Laboratory of Development and Plasticity of the Neuroendocrine Brain, UMR-S1172, EGID, Lille, France
| | - Ruben Nogueiras
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain.
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain.
- Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain.
| |
Collapse
|
16
|
Morville T, Madsen KH, Siebner HR, Hulme OJ. Reward signalling in brainstem nuclei under fluctuating blood glucose. PLoS One 2021; 16:e0243899. [PMID: 33826633 PMCID: PMC8026025 DOI: 10.1371/journal.pone.0243899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 12/01/2020] [Indexed: 11/18/2022] Open
Abstract
Phasic dopamine release from mid-brain dopaminergic neurons is thought to signal errors of reward prediction (RPE). If reward maximisation is to maintain homeostasis, then the value of primary rewards should be coupled to the homeostatic errors they remediate. This leads to the prediction that RPE signals should be configured as a function of homeostatic state and thus diminish with the attenuation of homeostatic error. To test this hypothesis, we collected a large volume of functional MRI data from five human volunteers on four separate days. After fasting for 12 hours, subjects consumed preloads that differed in glucose concentration. Participants then underwent a Pavlovian cue-conditioning paradigm in which the colour of a fixation-cross was stochastically associated with the delivery of water or glucose via a gustometer. This design afforded computation of RPE separately for better- and worse-than expected outcomes during ascending and descending trajectories of serum glucose fluctuations. In the parabrachial nuclei, regional activity coding positive RPEs scaled positively with serum glucose for both ascending and descending glucose levels. The ventral tegmental area and substantia nigra became more sensitive to negative RPEs when glucose levels were ascending. Together, the results suggest that RPE signals in key brainstem structures are modulated by homeostatic trajectories of naturally occurring glycaemic flux, revealing a tight interplay between homeostatic state and the neural encoding of primary reward in the human brain.
Collapse
Affiliation(s)
- Tobias Morville
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Kristoffer H. Madsen
- DTU Compute, Department of Informatics and Mathematical Modelling, Technical University of Denmark, Copenhagen, Denmark
| | - Hartwig R. Siebner
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Department of Neurology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Oliver J. Hulme
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| |
Collapse
|
17
|
Mehdipour M, Mehdipour T, Skinner CM, Wong N, Liu C, Chen CC, Jeon OH, Zuo Y, Conboy MJ, Conboy IM. Plasma dilution improves cognition and attenuates neuroinflammation in old mice. GeroScience 2020; 43:1-18. [PMID: 33191466 PMCID: PMC8050203 DOI: 10.1007/s11357-020-00297-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/01/2020] [Indexed: 02/06/2023] Open
Abstract
Our recent study has established that young blood factors are not causal, nor necessary, for the systemic rejuvenation of mammalian tissues. Instead, a procedure referred to as neutral blood exchange (NBE) that resets signaling milieu to a pro-regenerative state through dilution of old plasma, enhanced the health and repair of the muscle and liver, and promoted better hippocampal neurogenesis in 2-year-old mice (Mehdipour et al., Aging 12:8790–8819, 2020). Here we expand the rejuvenative phenotypes of NBE, focusing on the brain. Namely, our results demonstrate that old mice perform much better in novel object and novel texture (whisker discrimination) tests after a single NBE, which is accompanied by reduced neuroinflammation (less-activated CD68+ microglia). Evidence against attenuation/dilution of peripheral senescence-associated secretory phenotype (SASP) as the main mechanism behind NBE was that the senolytic ABT 263 had limited effects on neuroinflammation and did not enhance hippocampal neurogenesis in the old mice. Interestingly, peripherally acting ABT 263 and NBE both diminished SA-βGal signal in the old brain, demonstrating that peripheral senescence propagates to the brain, but NBE was more robustly rejuvenative than ABT 263, suggesting that rejuvenation was not simply by reducing senescence. Explaining the mechanism of the positive effects of NBE on the brain, our comparative proteomics analysis demonstrated that dilution of old blood plasma yields an increase in the determinants of brain maintenance and repair in mice and in people. These findings confirm the paradigm of rejuvenation through dilution of age-elevated systemic factors and extrapolate it to brain health and function.
Collapse
Affiliation(s)
- Melod Mehdipour
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA, USA
| | - Taha Mehdipour
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA, USA
| | - Colin M Skinner
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA, USA
| | - Nathan Wong
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA, USA
| | - Chao Liu
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA, USA
| | - Chia-Chien Chen
- Department of Molecular and Cellular Biology and QB3, UCSC, Santa Cruz, CA, USA
| | - Ok Hee Jeon
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, USA.,Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yi Zuo
- Department of Molecular and Cellular Biology and QB3, UCSC, Santa Cruz, CA, USA
| | - Michael J Conboy
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA, USA
| | - Irina M Conboy
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA, USA.
| |
Collapse
|
18
|
Gervais M, Labouèbe G, Picard A, Thorens B, Croizier S. EphrinB1 modulates glutamatergic inputs into POMC-expressing progenitors and controls glucose homeostasis. PLoS Biol 2020; 18:e3000680. [PMID: 33253166 PMCID: PMC7728393 DOI: 10.1371/journal.pbio.3000680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 12/10/2020] [Accepted: 11/05/2020] [Indexed: 12/25/2022] Open
Abstract
Proopiomelanocortin (POMC) neurons are major regulators of energy balance and glucose homeostasis. In addition to being regulated by hormones and nutrients, POMC neurons are controlled by glutamatergic input originating from multiple brain regions. However, the factors involved in the formation of glutamatergic inputs and how they contribute to bodily functions remain largely unknown. Here, we show that during the development of glutamatergic inputs, POMC neurons exhibit enriched expression of the Efnb1 (EphrinB1) and Efnb2 (EphrinB2) genes, which are known to control excitatory synapse formation. In vivo loss of Efnb1 in POMC-expressing progenitors decreases the amount of glutamatergic inputs, associated with a reduced number of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-D-aspartate (NMDA) receptor subunits and excitability of these cells. We found that mice lacking Efnb1 in POMC-expressing progenitors display impaired glucose tolerance due to blunted vagus nerve activity and decreased insulin secretion. However, despite reduced excitatory inputs, mice lacking Efnb2 in POMC-expressing progenitors showed no deregulation of insulin secretion and only mild alterations in feeding behavior and gluconeogenesis. Collectively, our data demonstrate the role of ephrins in controlling excitatory input amount into POMC-expressing progenitors and show an isotype-specific role of ephrins on the regulation of glucose homeostasis and feeding.
Collapse
Affiliation(s)
- Manon Gervais
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Gwenaël Labouèbe
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Alexandre Picard
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Sophie Croizier
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
19
|
Li P, Shan H, Nie B, Liu H, Dong G, Guo Y, Du J, Gao H, Ma L, Li D, Shan B. Sleeve Gastrectomy Rescuing the Altered Functional Connectivity of Lateral but Not Medial Hypothalamus in Subjects with Obesity. Obes Surg 2020; 29:2191-2199. [PMID: 30895508 DOI: 10.1007/s11695-019-03822-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Lateral and medial hypothalamus (LH and MH) play important roles in energy balance. Changed hypothalamic function has been found in subjects with obesity. However, the effect of bariatric surgery on the function of the two sub-regions has been poorly investigated. METHODS Thirty-eight subjects with obesity and 34 age- and sex-matched normal-weight controls were included. Seventeen of the 38 subjects underwent laparoscopic sleeve gastrectomy. Functional magnetic resonance imaging data and metabolic parameters were collected to investigate functional connectivity networks of the two hypothalamic sub-regions as well as the influence of sleeve gastrectomy on the two networks in subjects with obesity. RESULTS Compared to normal-weight controls, pre-surgical subjects had increased functional connectivity (FC) in the reward region (putamen) within the LH network, and increased FC in somatosensory cortical area (insula), as well as decreased FC in the cognitive control regions (prefrontal regions) within the MH network. After the surgery, post-surgical FC of the putamen within the LH network changed towards the patterns found in the control group. Furthermore, the changes in fasting glucose before and after the surgery were associated with the changes in FC of the putamen within the LH network. CONCLUSIONS The FC within the LH and MH networks were changed in subjects with obesity. Part of these altered FC was rescued after the surgery.
Collapse
Affiliation(s)
- Panlong Li
- Department of Physics, Zhengzhou University, Zhengzhou, 450001, Henan, China.,Division of Nuclear Technology and Applications, Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China.,School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Han Shan
- Department of Radiology, Chinese People's Liberation Army General Hospital, Beijing, 100853, China
| | - Binbin Nie
- Division of Nuclear Technology and Applications, Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China.,School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hua Liu
- Division of Nuclear Technology and Applications, Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China.,School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guanglong Dong
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yulin Guo
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jin Du
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hongkai Gao
- Department of General Surgery, The General Hospital of Chinese People's Armed Police Forces, Beijing, 100039, China
| | - Lin Ma
- Department of Radiology, Chinese People's Liberation Army General Hospital, Beijing, 100853, China.
| | - Demin Li
- Department of Physics, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| | - Baoci Shan
- Division of Nuclear Technology and Applications, Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China. .,School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, 100049, China. .,CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, China.
| |
Collapse
|
20
|
Cowen N, Bhatnagar A. The Potential Role of Activating the ATP-Sensitive Potassium Channel in the Treatment of Hyperphagic Obesity. Genes (Basel) 2020; 11:genes11040450. [PMID: 32326226 PMCID: PMC7230375 DOI: 10.3390/genes11040450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
To evaluate the potential role of ATP-sensitive potassium (KATP) channel activation in the treatment of hyperphagic obesity, a PubMed search was conducted focused on the expression of genes encoding the KATP channel, the response to activating the KATP channel in tissues regulating appetite and the establishment and maintenance of obesity, the evaluation of KATP activators in obese hyperphagic animal models, and clinical studies on syndromic obesity. KATP channel activation is mechanistically involved in the regulation of appetite in the arcuate nucleus; the regulation of hyperinsulinemia, glycemic control, appetite and satiety in the dorsal motor nucleus of vagus; insulin secretion by β-cells; and the synthesis and β-oxidation of fatty acids in adipocytes. KATP channel activators have been evaluated in hyperphagic obese animal models and were shown to reduce hyperphagia, induce fat loss and weight loss in older animals, reduce the accumulation of excess body fat in growing animals, reduce circulating and hepatic lipids, and improve glycemic control. Recent experience with a KATP channel activator in Prader-Willi syndrome is consistent with the therapeutic responses observed in animal models. KATP channel activation, given the breadth of impact and animal model and clinical results, is a viable target in hyperphagic obesity.
Collapse
|
21
|
Guia RM, Hassing AS, Skov LJ, Ratner C, Plucińska K, Madsen S, Diep TA, Dela Cruz GV, Trammell SA, Sustarsic EG, Emanuelli B, Gillum MP, Gerhart‐Hines Z, Holst B, Treebak JT. Fasting- and ghrelin-induced food intake is regulated by NAMPT in the hypothalamus. Acta Physiol (Oxf) 2020; 228:e13437. [PMID: 31900990 DOI: 10.1111/apha.13437] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 11/18/2019] [Accepted: 12/29/2019] [Indexed: 12/13/2022]
Abstract
AIM Neurons in the arcuate nucleus of the hypothalamus are involved in regulation of food intake and energy expenditure, and dysregulation of signalling in these neurons promotes development of obesity. The role of the rate-limiting enzyme in the NAD+ salvage pathway, nicotinamide phosphoribosyltransferase (NAMPT), for regulation energy homeostasis by the hypothalamus has not been extensively studied. METHODS We determined whether Nampt mRNA or protein levels in the hypothalamus of mice were affected by diet-induced obesity, by fasting and re-feeding, and by leptin and ghrelin treatment. Primary hypothalamic neurons were treated with FK866, a selective inhibitor of NAMPT, or rAAV carrying shRNA directed against Nampt, and levels of reactive oxygen species (ROS) and mitochondrial respiration were assessed. Fasting and ghrelin-induced food intake was measured in mice in metabolic cages after intracerebroventricular (ICV)-mediated FK866 administration. RESULTS NAMPT levels in the hypothalamus were elevated by administration of ghrelin and leptin. In diet-induced obese mice, both protein and mRNA levels of NAMPT decreased in the hypothalamus. NAMPT inhibition in primary hypothalamic neurons significantly reduced levels of NAD+ , increased levels of ROS, and affected the expression of Agrp, Pomc and genes related to mitochondrial function. Finally, ICV-induced NAMPT inhibition by FK866 did not cause malaise or anhedonia, but completely ablated fasting- and ghrelin-induced increases in food intake. CONCLUSION Our findings indicate that regulation of NAMPT levels in hypothalamic neurons is important for the control of fasting- and ghrelin-induced food intake.
Collapse
Affiliation(s)
- Roldan M. Guia
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Anna S. Hassing
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Louise J. Skov
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Cecilia Ratner
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Kaja Plucińska
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Søren Madsen
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Thi A. Diep
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Gelo V. Dela Cruz
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Stem Cell Biology University of Copenhagen Copenhagen Denmark
| | - Samuel A.J. Trammell
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Elahu G. Sustarsic
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Brice Emanuelli
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Matthew P. Gillum
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Zach Gerhart‐Hines
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Birgitte Holst
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Jonas T. Treebak
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| |
Collapse
|
22
|
Chen J, Chen V, Kawamura T, Hoang I, Yang Y, Wong AT, McBride R, Repunte-Canonigo V, Millhauser GL, Sanna PP. Charge Characteristics of Agouti-Related Protein Implicate Potent Involvement of Heparan Sulfate Proteoglycans in Metabolic Function. iScience 2019; 22:557-570. [PMID: 31863782 PMCID: PMC6928319 DOI: 10.1016/j.isci.2019.10.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/21/2019] [Accepted: 10/25/2019] [Indexed: 12/05/2022] Open
Abstract
The endogenous melanocortin peptide agouti-related protein (AgRP) plays a well-known role in foraging, but its contribution to metabolic regulation is less understood. Mature AgRP(83-132) has distinct residues for melanocortin receptor binding and heparan sulfate interactions. Here, we show that AgRP increases ad libitum feeding and operant responding for food in mice, decreases oxygen consumption, and lowers body temperature and activity, indicating lower energy expenditure. AgRP increased the respiratory exchange ratio, indicating a reduction of fat oxidation and a shift toward carbohydrates as the primary fuel source. The duration and intensity of AgRP's effects depended on the density of its positively charged amino acids, suggesting that its orexigenic and metabolic effects depend on its affinity for heparan sulfate. These findings may have major clinical implications by unveiling the critical involvement of interactions between AgRP and heparan sulfate to the central regulation of energy expenditure, fat utilization, and possibly their contribution to metabolic disease. AgRP increases both ad libitum and operant food intake and reduces energy expenditure AgRP reduces fat utilization as a fuel source, which promotes body fat accumulation These actions of AgRP depend on the positive charges, outside its ICK motif, that bind heparan sulfate
Collapse
Affiliation(s)
- Jihuan Chen
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Valerie Chen
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064, USA
| | - Tomoya Kawamura
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ivy Hoang
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yang Yang
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ashley Tess Wong
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064, USA
| | - Ryan McBride
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; Genomics Core, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Vez Repunte-Canonigo
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Glenn L Millhauser
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064, USA.
| | - Pietro Paolo Sanna
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
23
|
Reilly AM, Zhou S, Panigrahi SK, Yan S, Conley JM, Sheets PL, Wardlaw SL, Ren H. Gpr17 deficiency in POMC neurons ameliorates the metabolic derangements caused by long-term high-fat diet feeding. Nutr Diabetes 2019; 9:29. [PMID: 31611548 PMCID: PMC6791877 DOI: 10.1038/s41387-019-0096-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/12/2019] [Accepted: 08/23/2019] [Indexed: 11/19/2022] Open
Abstract
Background Proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARH) control energy homeostasis by sensing hormonal and nutrient cues and activating secondary melanocortin sensing neurons. We identified the expression of a G protein-coupled receptor, Gpr17, in the ARH and hypothesized that it contributes to the regulatory function of POMC neurons on metabolism. Methods In order to test this hypothesis, we generated POMC neuron-specific Gpr17 knockout (PGKO) mice and determined their energy and glucose metabolic phenotypes on normal chow diet (NCD) and high-fat diet (HFD). Results Adult PGKO mice on NCD displayed comparable body composition and metabolic features measured by indirect calorimetry. By contrast, PGKO mice on HFD demonstrated a sexually dimorphic phenotype with female PGKO mice displaying better metabolic homeostasis. Notably, female PGKO mice gained significantly less body weight and adiposity (p < 0.01), which was associated with increased energy expenditure, locomotor activity, and respiratory quotient, while males did not have an overt change in energy homeostasis. Though PGKO mice of both sexes had comparable glucose and insulin tolerance, detailed analyses of liver gene expression and serum metabolites indicate that PGKO mice could have reduced gluconeogenesis and increased lipid utilization on HFD. To elucidate the central-based mechanism(s) underlying the better-preserved energy and glucose homeostasis in PGKO mice on HFD, we examined the electrophysiological properties of POMC neurons and found Gpr17 deficiency led to increased spontaneous action potentials. Moreover, PGKO mice, especially female knockouts, had increased POMC-derived alpha-melanocyte stimulating hormone and beta-endorphin despite a comparable level of prohormone POMC in their hypothalamic extracts. Conclusions Gpr17 deficiency in POMC neurons protects metabolic homeostasis in a sex-dependent manner during dietary and aging challenges, suggesting that Gpr17 could be an effective anti-obesity target in specific populations with poor metabolic control.
Collapse
Affiliation(s)
- Austin M Reilly
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, 320 W. 15th Street Indianapolis, Indianapolis, IN, 46202, USA
| | - Shudi Zhou
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, 320 W. 15th Street Indianapolis, Indianapolis, IN, 46202, USA
| | - Sunil K Panigrahi
- Department of Medicine, Division of Endocrinology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Shijun Yan
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA.,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA
| | - Jason M Conley
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA.,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA
| | - Patrick L Sheets
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, 320 W. 15th Street Indianapolis, Indianapolis, IN, 46202, USA.,Department of Pharmacology & Toxicology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA
| | - Sharon L Wardlaw
- Department of Medicine, Division of Endocrinology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Hongxia Ren
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, 320 W. 15th Street Indianapolis, Indianapolis, IN, 46202, USA. .,Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA. .,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA. .,Department of Pharmacology & Toxicology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA. .,Department of Cellular & Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA. .,Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA.
| |
Collapse
|
24
|
Seicol BJ, Bejarano S, Behnke N, Guo L. Neuromodulation of metabolic functions: from pharmaceuticals to bioelectronics to biocircuits. J Biol Eng 2019; 13:67. [PMID: 31388355 PMCID: PMC6676523 DOI: 10.1186/s13036-019-0194-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/01/2019] [Indexed: 12/18/2022] Open
Abstract
Neuromodulation of central and peripheral neural circuitry brings together neurobiologists and neural engineers to develop advanced neural interfaces to decode and recapitulate the information encoded in the nervous system. Dysfunctional neuronal networks contribute not only to the pathophysiology of neurological diseases, but also to numerous metabolic disorders. Many regions of the central nervous system (CNS), especially within the hypothalamus, regulate metabolism. Recent evidence has linked obesity and diabetes to hyperactive or dysregulated autonomic nervous system (ANS) activity. Neural regulation of metabolic functions provides access to control pathology through neuromodulation. Metabolism is defined as cellular events that involve catabolic and/or anabolic processes, including control of systemic metabolic functions, as well as cellular signaling pathways, such as cytokine release by immune cells. Therefore, neuromodulation to control metabolic functions can be used to target metabolic diseases, such as diabetes and chronic inflammatory diseases. Better understanding of neurometabolic circuitry will allow for targeted stimulation to modulate metabolic functions. Within the broad category of metabolic functions, cellular signaling, including the production and release of cytokines and other immunological processes, is regulated by both the CNS and ANS. Neural innervations of metabolic (e.g. pancreas) and immunologic (e.g. spleen) organs have been understood for over a century, however, it is only now becoming possible to decode the neuronal information to enable exogenous controls of these systems. Future interventions taking advantage of this progress will enable scientists, engineering and medical doctors to more effectively treat metabolic diseases.
Collapse
Affiliation(s)
- Benjamin J. Seicol
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH USA
- Department of Neuroscience, The Ohio State University, Columbus, OH USA
| | | | - Nicholas Behnke
- Department of Food, Agricultural, and Biological Engineering, The Ohio State University, Columbus, OH USA
| | - Liang Guo
- Department of Neuroscience, The Ohio State University, Columbus, OH USA
- Department of Electrical and Computer Engineering, The Ohio State University, Columbus, OH USA
| |
Collapse
|
25
|
Torres DJ, Pitts MW, Hashimoto AC, Berry MJ. Agrp-Specific Ablation of Scly Protects against Diet-Induced Obesity and Leptin Resistance. Nutrients 2019; 11:nu11071693. [PMID: 31340540 PMCID: PMC6682868 DOI: 10.3390/nu11071693] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/18/2019] [Accepted: 07/20/2019] [Indexed: 01/25/2023] Open
Abstract
Selenium, an essential trace element known mainly for its antioxidant properties, is critical for proper brain function and regulation of energy metabolism. Whole-body knockout of the selenium recycling enzyme, selenocysteine lyase (Scly), increases susceptibility to metabolic syndrome and diet-induced obesity in mice. Scly knockout mice also have decreased selenoprotein expression levels in the hypothalamus, a key regulator of energy homeostasis. This study investigated the role of selenium in whole-body metabolism regulation using a mouse model with hypothalamic knockout of Scly. Agouti-related peptide (Agrp) promoter-driven Scly knockout resulted in reduced weight gain and adiposity while on a high-fat diet (HFD). Scly-Agrp knockout mice had reduced Agrp expression in the hypothalamus, as measured by Western blot and immunohistochemistry (IHC). IHC also revealed that while control mice developed HFD-induced leptin resistance in the arcuate nucleus, Scly-Agrp knockout mice maintained leptin sensitivity. Brown adipose tissue from Scly-Agrp knockout mice had reduced lipid deposition and increased expression of the thermogenic marker uncoupled protein-1. This study sheds light on the important role of selenium utilization in energy homeostasis, provides new information on the interplay between the central nervous system and whole-body metabolism, and may help identify key targets of interest for therapeutic treatment of metabolic disorders.
Collapse
Affiliation(s)
- Daniel J Torres
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI 96813, USA
| | - Matthew W Pitts
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI 96813, USA
| | - Ann C Hashimoto
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI 96813, USA
| | - Marla J Berry
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI 96813, USA.
| |
Collapse
|
26
|
Wu CS, Bongmba OYN, Lee JH, Tuchaai E, Zhou Y, Li DP, Xue B, Chen Z, Sun Y. Ghrelin receptor in agouti-related peptide neurones regulates metabolic adaptation to calorie restriction. J Neuroendocrinol 2019; 31:e12763. [PMID: 31251830 PMCID: PMC7233797 DOI: 10.1111/jne.12763] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/24/2019] [Accepted: 06/25/2019] [Indexed: 12/16/2022]
Abstract
Ghrelin is a gut hormone that signals to the hypothalamus to stimulate growth hormone release, increase food intake and promote fat deposition. The ghrelin receptor, also known as growth hormone secretagogue receptor (GHS-R), is highly expressed in the brain, with the highest expression in agouti-related peptide (AgRP) neurones in the hypothalamus. Compelling evidence indicates that ghrelin serves as a survival hormone with respect to maintaining blood glucose and body weight during nutritional deficiencies. Recent studies have demonstrated that AgRP neurones are involved in metabolic and behavioural adaptation to an energy deficit to improve survival. In the present study, we used a neuronal subtype-specific GHS-R knockout mouse (AgRP-Cre;Ghsrf/f ) to investigate the role of GHS-R in hypothalamic AgRP neurones in metabolic and behavioural adaptation to hypocaloric restricted feeding. We subjected the mice to a restricted feeding regimen of 40% mild calorie restriction (CR), with one-quarter of food allotment given in the beginning of the light cycle and three-quarters given at the beginning of the dark cycle, to mimic normal mouse intake pattern. The CR-fed AgRP-Cre;Ghsrf/f mice exhibited reductions in body weight, fat mass and blood glucose. Metabolic profiling of these CR-fed AgRP-Cre;Ghsrf/f mice showed a trend toward reduced basal metabolic rate, significantly reduced core body temperature and a decreased expression of thermogenic genes in brown adipose tissue. This suggests a metabolic reset to a lower threshold. Significantly increased physical activity, a trend toward increased food anticipatory behaviour and altered fuel preferences were also observed in these mice. In addition, these CR-fed AgRP-Cre;Ghsrf/f mice exhibited a decreased counter-regulatory response, showing impaired hepatic glucose production. Lastly, hypothalamic gene expression in AgRP-Cre;Ghsrf/f mice revealed increased AgRP expression and a decreased expression of genes in β-oxidation pathways. In summary, our data suggest that GHS-R in AgRP neurones is a key component of the neurocircuitry involved in metabolic adaptation to calorie restriction.
Collapse
Affiliation(s)
- Chia-Shan Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Odelia Y. N. Bongmba
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jong Han Lee
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
- College of Pharmacy, Gachon University, Incheon, 21936, Korea
| | - Ellie Tuchaai
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Yu Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
| | - De-Pei Li
- Center for precision medicine, School of Medicine, University of Missouri. Columbia, MO 65212, USA
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Zheng Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Yuxiang Sun
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
27
|
Opazo R, Plaza-Parrochia F, Cardoso dos Santos GR, Carneiro GRA, Sardela VF, Romero J, Valladares L. Fasting Upregulates npy, agrp, and ghsr Without Increasing Ghrelin Levels in Zebrafish ( Danio rerio) Larvae. Front Physiol 2019; 9:1901. [PMID: 30733682 PMCID: PMC6353792 DOI: 10.3389/fphys.2018.01901] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/18/2018] [Indexed: 11/24/2022] Open
Abstract
Food intake in fish and mammals is orchestrated by hypothalamic crosstalk between orexigenic (food intake stimulation) and anorexigenic (food intake inhibition) signals. Some of these signals are released by peripheral tissues that are associated with energy homeostasis or nutrient availability. During the fish larva stage, orexigenic stimulation plays a critical role in individual viability. The goal of this study was to assess the mRNA levels of the main neuropeptides involved in food intake regulation (npy, agrp, carppt, and pomc), in concert with the mRNA levels and peptide levels of ghrelin, under a fasting intervention at the larval stage in zebrafish (Danio rerio). Prior to the fasting intervention, the zebrafish larva cohort was reared for 20 days post fertilization (dpf) and then randomly divided into two groups of 20 individuals. One group was subjected to a fasting intervention for 5 days (fasted group), and the other group was fed normally (fed group); this experimental protocol was performed twice independently. At the end of the fasting period, individuals from each experimental group were divided into different analysis groups, for evaluations such as relative gene expression, immunohistochemistry, and liquid chromatography coupled to nano high-resolution mass spectrometry (nLC-HRMS) analyses. The relative expression levels of the following genes were assessed: neuropeptide Y (npy), agouti-related peptide (agrp), proopiomelanocortin (pomc), cocaine and amphetamine-regulated transcript (cartpt), ghrelin (ghrl), ghrelin O-acyltransferase (mboat4), growth hormone secretagogue receptor (ghsr), and glucokinase (gck). In the fasted group, significant upregulation of orexigenic peptides (npy - agrp) and ghsr was observed, which was associated with significant downregulation of gck. The anorexigenic peptides (pomc and cartpt) did not show any significant modulation between the groups, similar to mboat4. Contrary to what was expected, the relative mRNA upregulation of the orexigenic peptides observed in the fasted experimental group could not be associated with significant ghrelin modulation as assessed by three different approaches: qPCR (relative gene expression of ghrelin), nLC-HRMS (des-acyl-ghrelin levels), and immunohistochemistry (integrated optical density of prepropeptides in intestinal and hepatopancreas tissues). Our results demonstrate that zebrafish larvae at 25 dpf exhibit suitable modulation of the relative mRNA levels of orexigenic peptides (npy and agrp) in response to fasting intervention; nevertheless, ghrelin was not coregulated by fasting. Therefore, it can be suggested that ghrelin is not an essential peptide for an increase in appetite in the zebrafish larva stage. These results give rise to new questions about food intake regulation factors in the early stages of fish.
Collapse
Affiliation(s)
- Rafael Opazo
- Laboratorio de Biotecnología INTA, Universidad de Chile, Santiago, Chile
| | - Francisca Plaza-Parrochia
- Laboratorio de Endocrinología y Biología de la Reproducción, Hospital Clínico, Universidad de Chile, Santiago, Chile
| | - Gustavo R. Cardoso dos Santos
- Laboratorio de Pesquisa, Desenvolvimento e Inovação (LPDI-LADETEC), Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriel R. A. Carneiro
- Laboratorio de Pesquisa, Desenvolvimento e Inovação (LPDI-LADETEC), Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vinicius F. Sardela
- Laboratorio de Pesquisa, Desenvolvimento e Inovação (LPDI-LADETEC), Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jaime Romero
- Laboratorio de Biotecnología INTA, Universidad de Chile, Santiago, Chile
| | - Luis Valladares
- Laboratorio de Hormonas y Receptores INTA, Universidad de Chile, Santiago, Chile
| |
Collapse
|
28
|
Schier LA, Spector AC. The Functional and Neurobiological Properties of Bad Taste. Physiol Rev 2019; 99:605-663. [PMID: 30475657 PMCID: PMC6442928 DOI: 10.1152/physrev.00044.2017] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 05/18/2018] [Accepted: 06/30/2018] [Indexed: 12/12/2022] Open
Abstract
The gustatory system serves as a critical line of defense against ingesting harmful substances. Technological advances have fostered the characterization of peripheral receptors and have created opportunities for more selective manipulations of the nervous system, yet the neurobiological mechanisms underlying taste-based avoidance and aversion remain poorly understood. One conceptual obstacle stems from a lack of recognition that taste signals subserve several behavioral and physiological functions which likely engage partially segregated neural circuits. Moreover, although the gustatory system evolved to respond expediently to broad classes of biologically relevant chemicals, innate repertoires are often not in register with the actual consequences of a food. The mammalian brain exhibits tremendous flexibility; responses to taste can be modified in a specific manner according to bodily needs and the learned consequences of ingestion. Therefore, experimental strategies that distinguish between the functional properties of various taste-guided behaviors and link them to specific neural circuits need to be applied. Given the close relationship between the gustatory and visceroceptive systems, a full reckoning of the neural architecture of bad taste requires an understanding of how these respective sensory signals are integrated in the brain.
Collapse
Affiliation(s)
- Lindsey A Schier
- Department of Biological Sciences, University of Southern California , Los Angeles, California ; and Department of Psychology and Program in Neuroscience, Florida State University , Tallahassee, Florida
| | - Alan C Spector
- Department of Biological Sciences, University of Southern California , Los Angeles, California ; and Department of Psychology and Program in Neuroscience, Florida State University , Tallahassee, Florida
| |
Collapse
|
29
|
Wen S, Wang C, Gong M, Zhou L. An overview of energy and metabolic regulation. SCIENCE CHINA-LIFE SCIENCES 2018; 62:771-790. [PMID: 30367342 DOI: 10.1007/s11427-018-9371-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/23/2018] [Indexed: 12/21/2022]
Abstract
The physiology and behaviors related to energy balance are monitored by the nervous and humoral systems. Because of the difficulty in treating diabetes and obesity, elucidating the energy balance mechanism and identifying critical targets for treatment are important research goals. Therefore, the purpose of this article is to describe energy regulation by the central nervous system (CNS) and peripheral humoral pathway. Homeostasis and rewarding are the basis of CNS regulation. Anorexigenic or orexigenic effects reflect the activities of the POMC/CART or NPY/AgRP neurons within the hypothalamus. Neurotransmitters have roles in food intake, and responsive brain nuclei have different functions related to food intake, glucose monitoring, reward processing. Peripheral gut- or adipose-derived hormones are the major source of peripheral humoral regulation systems. Nutrients or metabolites and gut microbiota affect metabolism via a discrete pathway. We also review the role of peripheral organs, the liver, adipose tissue, and skeletal muscle in peripheral regulation. We discuss these topics and how the body regulates metabolism.
Collapse
Affiliation(s)
- Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Chaoxun Wang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Min Gong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China.
| |
Collapse
|
30
|
p53 in AgRP neurons is required for protection against diet-induced obesity via JNK1. Nat Commun 2018; 9:3432. [PMID: 30143607 PMCID: PMC6109113 DOI: 10.1038/s41467-018-05711-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/19/2018] [Indexed: 12/13/2022] Open
Abstract
p53 is a well-known tumor suppressor that has emerged as an important player in energy balance. However, its metabolic role in the hypothalamus remains unknown. Herein, we show that mice lacking p53 in agouti-related peptide (AgRP), but not proopiomelanocortin (POMC) or steroidogenic factor-1 (SF1) neurons, are more prone to develop diet-induced obesity and show reduced brown adipose tissue (BAT) thermogenic activity. AgRP-specific ablation of p53 resulted in increased hypothalamic c-Jun N-terminal kinase (JNK) activity before the mice developed obesity, and central inhibition of JNK reversed the obese phenotype of these mice. The overexpression of p53 in the ARC or specifically in AgRP neurons of obese mice decreased body weight and stimulated BAT thermogenesis, resulting in body weight loss. Finally, p53 in AgRP neurons regulates the ghrelin-induced food intake and body weight. Overall, our findings provide evidence that p53 in AgRP neurons is required for normal adaptations against diet-induced obesity. Emerging studies suggest that p53 is an important regulator of energy metabolism, yet there is little known about the metabolic function of this tumor suppressor in the hypothalamus. Here, authors illustrate that p53, specifically in AgRP neurons, is required for adaptation to diet-induced obesity.
Collapse
|
31
|
Calarco CA, Li Z, Taylor SR, Lee S, Zhou W, Friedman JM, Mineur YS, Gotti C, Picciotto MR. Molecular and cellular characterization of nicotinic acetylcholine receptor subtypes in the arcuate nucleus of the mouse hypothalamus. Eur J Neurosci 2018; 48:10.1111/ejn.13966. [PMID: 29791746 PMCID: PMC6251769 DOI: 10.1111/ejn.13966] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/26/2018] [Accepted: 05/09/2018] [Indexed: 12/22/2022]
Abstract
Nicotine, acting through nicotinic acetylcholine receptors (nAChRs), increases the firing rate of both orexigenic agouti-related peptide (AgRP) and anorexigenic pro-opiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARC), yet nicotine and other nAChR agonists decrease food intake in mice. Viral-mediated knockdown of the β4 nAChR subunit in all neuronal cell types in the ARC prevents the nicotinic agonist cytisine from decreasing food intake, but it is not known whether the β4 subunit is selectively expressed in anorexigenic neurons or how other nAChR subtypes are distributed in this nucleus. Using translating ribosome affinity purification (TRAP) on ARC tissue from mice with ribosomes tagged in either AgRP or POMC cells, we examined nAChR subunit mRNA levels using real-time PCR. Both AgRP and POMC cells express a comparable panel of nAChR subunits with differences in α7 mRNA levels and a trend for difference in α4 levels, but no differences in β4 expression. Immunoprecipitation of assembled nAChRs revealed that the β4 subunit forms assembled channels with α3, β2 and α4, but not other subunits found in the ARC. Finally, using cell type-selective, virally delivered small hairpin RNAs targeting either the β4 or α7 subunit, we examined the contribution of each subunit in either AgRP or POMC cells to the behavioural response to nicotine, refining the understanding of nicotinic regulation of this feeding circuit. These experiments identify a more complex set of nAChRs expressed in ARC than in other hypothalamic regions. Thus, the ARC appears to be a particular target of nicotinic modulation.
Collapse
Affiliation(s)
- Cali A. Calarco
- Department of Psychiatry and Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06508, USA
| | | | - Seth R. Taylor
- Department of Psychiatry and Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06508, USA
| | - Somin Lee
- Department of Psychiatry and Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06508, USA
| | - Wenliang Zhou
- Department of Psychiatry and Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06508, USA
| | | | - Yann S. Mineur
- Department of Psychiatry and Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06508, USA
| | | | - Marina R. Picciotto
- Department of Psychiatry and Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06508, USA
| |
Collapse
|
32
|
Goldsammler M, Merhi Z, Buyuk E. Role of hormonal and inflammatory alterations in obesity-related reproductive dysfunction at the level of the hypothalamic-pituitary-ovarian axis. Reprod Biol Endocrinol 2018; 16:45. [PMID: 29743077 PMCID: PMC5941782 DOI: 10.1186/s12958-018-0366-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 05/03/2018] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Besides being a risk factor for multiple metabolic disorders, obesity could affect female reproduction. While increased adiposity is associated with hormonal changes that could disrupt the function of the hypothalamus and the pituitary, compelling data suggest that obesity-related hormonal and inflammatory changes could directly impact ovarian function. OBJECTIVE To review the available data related to the mechanisms by which obesity, and its associated hormonal and inflammatory changes, could affect the female reproductive function with a focus on the hypothalamic-pituitary-ovarian (HPO) axis. METHODS PubMed database search for publications in English language until October 2017 pertaining to obesity and female reproductive function was performed. RESULTS The obesity-related changes in hormone levels, in particular leptin, adiponectin, ghrelin, neuropeptide Y and agouti-related protein, are associated with reproductive dysfunction at both the hypothalamic-pituitary and the ovarian levels. The pro-inflammatory molecules advanced glycation end products (AGEs) and monocyte chemotactic protein-1 (MCP-1) are emerging as relatively new players in the pathophysiology of obesity-related ovarian dysfunction. CONCLUSION There is an intricate crosstalk between the adipose tissue and the inflammatory system with the HPO axis function. Understanding the mechanisms behind this crosstalk could lead to potential therapies for the common obesity-related reproductive dysfunction.
Collapse
Affiliation(s)
- Michelle Goldsammler
- Montefiore’s Institute for Reproductive Medicine and Health, Department of Obstetrics & Gynecology and Women’s Health, Albert Einstein College of Medicine, Montefiore Medical Center, Hartsdale, NY USA
| | - Zaher Merhi
- 0000 0004 1936 8753grid.137628.9Department of Obstetrics and Gynecology, Division of Reproductive Biology, NYU School of Medicine, New York, NY USA
- 0000000121791997grid.251993.5Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY USA
| | - Erkan Buyuk
- Montefiore’s Institute for Reproductive Medicine and Health, Department of Obstetrics & Gynecology and Women’s Health, Albert Einstein College of Medicine, Montefiore Medical Center, Hartsdale, NY USA
| |
Collapse
|
33
|
Electroacupuncture Reduces Weight in Diet-Induced Obese Rats via Hypothalamic Tsc1 Promoter Demethylation and Inhibition of the Activity of mTORC1 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:3039783. [PMID: 29853949 PMCID: PMC5944273 DOI: 10.1155/2018/3039783] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 03/09/2018] [Accepted: 04/01/2018] [Indexed: 12/12/2022]
Abstract
Subject The study aimed to investigate the mechanism of electroacupuncture reducing weight via tuberous sclerosis complex 1 (Tsc1) promoter methylation, inhibiting the mammalian target of rapamycin complex 1 (mTORC1) pathway. Materials and Methods Male Sprague-Dawley rats were divided into chow-fed group (chow group) or high-fat diet group (HF group) for 14 weeks. The obesity rats in HF group were randomly divided into electroacupuncture group (EA group) and diet-induced obesity (DIO) group, which received EA stimulation on bilateral ST25, RN12, SP6, and ST36 for 4 weeks or no further treatment, respectively. Methylation of the Tsc1 gene promoter and expression of agouti-related protein (AgRP), neuropeptide Y (NPY), and proopiomelanocortin (PoMC) were detected at the 18th week. Results At week 18, weight, body fat, and the body fat rate in DIO group were significantly higher than those of the chow and EA group. Compared with the chow group, the DIO group had increased methylation of the Tsc1 gene promoter and expression of mTORC1, AgRP, and NPY gene and decreased PoMC in the hypothalamus; after EA, methylation of the Tsc1 gene promoter, mRNA, and protein of the mTORC1 and expression of AgRP and NPY gene decreased and PoMC increased significantly. Conclusions Our study could shed light on the potential pathway where EA exerts effects on the mechanism of EA treatment for obesity through the hypothalamic Tsc1 promoter demethylation and inhibition of the activity of mTORC1 signaling pathway.
Collapse
|
34
|
The physiological and neuroendocrine correlates of hunger in the Red Junglefowl (Gallus gallus). Sci Rep 2017; 7:17984. [PMID: 29269733 PMCID: PMC5740172 DOI: 10.1038/s41598-017-17922-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 12/02/2017] [Indexed: 12/12/2022] Open
Abstract
The ability to regulate food intake is critical to survival. The hypothalamus is central to this regulation, integrating peripheral signals of energy availability. Although our understanding of hunger in rodents is advanced, an equivalent understanding in birds is lacking. In particular, the relationship between peripheral energy indices and hypothalamic 'hunger' peptides, agouti-related protein (AgRP), pro-opiomelanocortin (POMC) and neuropeptide Y (NPY) is poorly understood. Here, we compare AgRP, POMC and NPY RNA levels in the hypothalamus of Red Junglefowl chicks raised under ad libitum, chronic restriction and intermittent feeding regimens. Hypothalamic gene expression differed between chronically and intermittently restricted birds, confirming that different restriction regimens elicit different patterns of hunger. By assessing the relationship between hypothalamic gene expression and carcass traits, we show for the first time in birds that AgRP and POMC are responsive to fat-related measures and therefore represent long-term energy status. Chronically restricted birds, having lower indices of fat, show elevated hunger according to AgRP and POMC. NPY was elevated in intermittently fasted birds during fasting, suggesting a role as a short-term index of hunger. The different physiological and neuroendocrine responses to quantitative versus temporal feed restriction provide novel insights into the divergent roles of avian hunger neuropeptides.
Collapse
|
35
|
|
36
|
Thomas MA, Xue B. Mechanisms for AgRP neuron-mediated regulation of appetitive behaviors in rodents. Physiol Behav 2017; 190:34-42. [PMID: 29031550 DOI: 10.1016/j.physbeh.2017.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 08/29/2017] [Accepted: 10/06/2017] [Indexed: 12/19/2022]
Abstract
The obesity epidemic is a major health and economic burden facing both developed and developing countries worldwide. Interrogation of the central and peripheral mechanisms regulating ingestive behaviors have primarily focused on food intake, and in the process uncovered a detailed neuroanatomical framework controlling this behavior. However, these studies have largely ignored the behaviors that bring animals, including humans, in contact with food. It is therefore useful to dichotomize ingestive behaviors as appetitive (motivation to find and store food) and consummatory (consumption of food once found), and utilize an animal model that naturally displays these behaviors. Recent advances in genetics have facilitated the identification of several neuronal populations critical for regulating ingestive behaviors in mice, and novel functions of these neurons and neuropeptides in regulating appetitive behaviors in Siberian hamsters, a natural model of food foraging and food hoarding, have been identified. To this end, hypothalamic agouti-related protein/neuropeptide Y expressing neurons (AgRP neurons) have emerged as a critical regulator of ingestive behaviors. Recent studies by Dr. Timothy Bartness and others have identified several discrete mechanisms through which peripheral endocrine signals regulate AgRP neurons to control food foraging, food hoarding, and food intake. We review here recent advances in our understanding of the neuroendocrine control of ingestive behaviors in Siberian hamsters and other laboratory rodents, and identify novel mechanisms through which AgRP neurons mediate appetitive and consummatory behaviors.
Collapse
Affiliation(s)
- M Alex Thomas
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA; Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302, USA
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA; Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302, USA; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA.
| |
Collapse
|
37
|
Liver ERα regulates AgRP neuronal activity in the arcuate nucleus of female mice. Sci Rep 2017; 7:1194. [PMID: 28446774 PMCID: PMC5430776 DOI: 10.1038/s41598-017-01393-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/27/2017] [Indexed: 01/22/2023] Open
Abstract
Recent work revealed the major role played by liver Estrogen Receptor α (ERα) in the regulation of metabolic and reproductive functions. By using mutant mice with liver-specific ablation of Erα, we here demonstrate that the hepatic ERα is essential for the modulation of the activity of Agouti Related Protein (AgRP) neurons in relation to the reproductive cycle and diet. Our results suggest that the alterations of hepatic lipid metabolism due to the lack of liver ERα activity are responsible for a neuroinflammatory status that induces refractoriness of AgRP neurons to reproductive and dietary stimuli. The study therefore points to the liver ERα as a necessary sensor for the coordination of systemic energy metabolism and reproductive functions.
Collapse
|
38
|
Almundarij TI, Gavini CK, Novak CM. Suppressed sympathetic outflow to skeletal muscle, muscle thermogenesis, and activity energy expenditure with calorie restriction. Physiol Rep 2017; 5:5/4/e13171. [PMID: 28242830 PMCID: PMC5328781 DOI: 10.14814/phy2.13171] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 01/29/2017] [Indexed: 12/21/2022] Open
Abstract
During weight loss, adaptive thermogenesis occurs where energy expenditure (EE) is suppressed beyond that predicted for the smaller body size. Here, we investigated the contributions of resting and nonresting EE to the reduced total EE seen after 3 weeks of 50% calorie restriction (CR) in rats, focusing on activity‐associated EE, muscle thermogenesis, and sympathetic outflow. Prolonged food restriction resulted in a 42% reduction in daily EE, through a 40% decrease in resting EE, and a 48% decline in nonresting EE. These decreases in EE were significant even when the reductions in body weight and lean mass were taken into account. Along with a decreased caloric need for low‐to‐moderate‐intensity treadmill activity with 50% CR, baseline and activity‐related muscle thermogenesis were also suppressed, though the ability to increase muscle thermogenesis above baseline levels was not compromised. When sympathetic drive was measured by assessing norepinephrine turnover (NETO), 50% CR was found to decrease NETO in three of the four muscle groups examined, whereas elevated NETO was found in white adipose tissue of food‐restricted rats. Central activation of melanocortin 4 receptors in the ventromedial hypothalamus stimulated this pathway, enhancing activity EE; this was not compromised by 50% CR. These data suggest that suppressed activity EE contributes to adaptive thermogenesis during energy restriction. This may stem from decreased sympathetic drive to skeletal muscle, increasing locomotor efficiency and reducing skeletal muscle thermogenesis. The capacity to increase activity EE in response to central stimuli is retained, however, presenting a potential target for preventing weight regain.
Collapse
Affiliation(s)
- Tariq I Almundarij
- College of Agriculture and Veterinary Medicine, Al Qassim University, Buraydah, Al-Qassim Province, Saudi Arabia.,Department of Biological Sciences, Kent State University, Kent, Ohio
| | - Chaitanya K Gavini
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois.,School of Biomedical Sciences, Kent State University, Kent, Ohio
| | - Colleen M Novak
- Department of Biological Sciences, Kent State University, Kent, Ohio .,School of Biomedical Sciences, Kent State University, Kent, Ohio
| |
Collapse
|
39
|
Bojanowska E, Ciosek J. Can We Selectively Reduce Appetite for Energy-Dense Foods? An Overview of Pharmacological Strategies for Modification of Food Preference Behavior. Curr Neuropharmacol 2016; 14:118-42. [PMID: 26549651 PMCID: PMC4825944 DOI: 10.2174/1570159x14666151109103147] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 09/19/2015] [Accepted: 10/31/2015] [Indexed: 12/11/2022] Open
Abstract
Excessive intake of food, especially palatable and energy-dense carbohydrates and fats, is
largely responsible for the growing incidence of obesity worldwide. Although there are a number of
candidate antiobesity drugs, only a few of them have been proven able to inhibit appetite for palatable
foods without the concurrent reduction in regular food consumption. In this review, we discuss the
interrelationships between homeostatic and hedonic food intake control mechanisms in promoting
overeating with palatable foods and assess the potential usefulness of systemically administered pharmaceuticals that
impinge on the endogenous cannabinoid, opioid, aminergic, cholinergic, and peptidergic systems in the modification of
food preference behavior. Also, certain dietary supplements with the potency to reduce specifically palatable food intake
are presented. Based on human and animal studies, we indicate the most promising therapies and agents that influence the
effectiveness of appetite-modifying drugs. It should be stressed, however, that most of the data included in our review
come from preclinical studies; therefore, further investigations aimed at confirming the effectiveness and safety of the
aforementioned medications in the treatment of obese humans are necessary.
Collapse
Affiliation(s)
- Ewa Bojanowska
- Department of Behavioral Pathophysiology, Institute of General and Experimental Pathology, Medical University of Lodz, 60 Narutowicza Street, 90-136 Lodz, Poland.
| | | |
Collapse
|
40
|
Thongrod S, Changklungmoa N, Chansela P, Siangcham T, Kruangkum T, Suwansa-Ard S, Saetan J, Sroyraya M, Tinikul Y, Wanichanon C, Sobhon P. Characterization and tissue distribution of neuropeptide F in the eyestalk and brain of the male giant freshwater prawn, Macrobrachium rosenbergii. Cell Tissue Res 2016; 367:181-195. [DOI: 10.1007/s00441-016-2538-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 11/16/2016] [Indexed: 10/20/2022]
|
41
|
Schlegel P, Texada MJ, Miroschnikow A, Schoofs A, Hückesfeld S, Peters M, Schneider-Mizell CM, Lacin H, Li F, Fetter RD, Truman JW, Cardona A, Pankratz MJ. Synaptic transmission parallels neuromodulation in a central food-intake circuit. eLife 2016; 5:16799. [PMID: 27845623 PMCID: PMC5182061 DOI: 10.7554/elife.16799] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 11/14/2016] [Indexed: 02/06/2023] Open
Abstract
NeuromedinU is a potent regulator of food intake and activity in mammals. In Drosophila, neurons producing the homologous neuropeptide hugin regulate feeding and locomotion in a similar manner. Here, we use EM-based reconstruction to generate the entire connectome of hugin-producing neurons in the Drosophila larval CNS. We demonstrate that hugin neurons use synaptic transmission in addition to peptidergic neuromodulation and identify acetylcholine as a key transmitter. Hugin neuropeptide and acetylcholine are both necessary for the regulatory effect on feeding. We further show that subtypes of hugin neurons connect chemosensory to endocrine system by combinations of synaptic and peptide-receptor connections. Targets include endocrine neurons producing DH44, a CRH-like peptide, and insulin-like peptides. Homologs of these peptides are likewise downstream of neuromedinU, revealing striking parallels in flies and mammals. We propose that hugin neurons are part of an ancient physiological control system that has been conserved at functional and molecular level. DOI:http://dx.doi.org/10.7554/eLife.16799.001
Collapse
Affiliation(s)
- Philipp Schlegel
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Bonn, Germany
| | - Michael J Texada
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Anton Miroschnikow
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Bonn, Germany
| | - Andreas Schoofs
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Bonn, Germany
| | - Sebastian Hückesfeld
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Bonn, Germany
| | - Marc Peters
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Bonn, Germany
| | | | - Haluk Lacin
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Feng Li
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Richard D Fetter
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - James W Truman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Albert Cardona
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Michael J Pankratz
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
42
|
Schlenker EH. Sexual dimorphism of cardiopulmonary regulation in the arcuate nucleus of the hypothalamus. Respir Physiol Neurobiol 2016; 245:37-44. [PMID: 27756648 DOI: 10.1016/j.resp.2016.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 10/11/2016] [Accepted: 10/12/2016] [Indexed: 11/19/2022]
Abstract
The arcuate nucleus of the hypothalamus (ANH) interacts with other hypothalamic nuclei, forebrain regions, and downstream brain sites to affect autonomic nervous system outflow, energy balance, temperature regulation, sleep, arousal, neuroendocrine function, reproduction, and cardiopulmonary regulation. Compared to studies of other ANH functions, how the ANH regulates cardiopulmonary function is less understood. Importantly, the ANH exhibits structural and functional sexually dimorphic characteristics and contains numerous neuroactive substances and receptors including leptin, neuropeptide Y, glutamate, acetylcholine, endorphins, orexin, kisspeptin, insulin, Agouti-related protein, cocaine and amphetamine-regulated transcript, dopamine, somatostatin, components of renin-angiotensin system and gamma amino butyric acid that modulate physiological functions. Moreover, several clinically relevant disorders are associated with ANH ventilatory control dysfunction. This review highlights how ANH neurotransmitter systems and receptors modulate breathing differently in male and female rodents. Results highlight the significance of the ANH in cardiopulmonary regulation. The paucity of studies in this area that will hopefully spark investigations of sexually dimorphic ANH-modulation of breathing.
Collapse
Affiliation(s)
- Evelyn H Schlenker
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, 414 East Clark St., Vermillion, SD, 57069, United States.
| |
Collapse
|
43
|
Kakizawa K, Watanabe M, Mutoh H, Okawa Y, Yamashita M, Yanagawa Y, Itoi K, Suda T, Oki Y, Fukuda A. A novel GABA-mediated corticotropin-releasing hormone secretory mechanism in the median eminence. SCIENCE ADVANCES 2016; 2:e1501723. [PMID: 27540587 PMCID: PMC4988769 DOI: 10.1126/sciadv.1501723] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 07/19/2016] [Indexed: 05/13/2023]
Abstract
Corticotropin-releasing hormone (CRH), which is synthesized in the paraventricular nucleus (PVN) of the hypothalamus, plays an important role in the endocrine stress response. The excitability of CRH neurons is regulated by γ-aminobutyric acid (GABA)-containing neurons projecting to the PVN. We investigated the role of GABA in the regulation of CRH release. The release of CRH was impaired, accumulating in the cell bodies of CRH neurons in heterozygous GAD67-GFP (green fluorescent protein) knock-in mice (GAD67(+/GFP)), which exhibited decreased GABA content. The GABAA receptor (GABAAR) and the Na(+)-K(+)-2Cl(-) cotransporter (NKCC1), but not the K(+)-Cl(-) cotransporter (KCC2), were expressed in the terminals of the CRH neurons at the median eminence (ME). In contrast, CRH neuronal somata were enriched with KCC2 but not with NKCC1. Thus, intracellular Cl(-) concentrations ([Cl(-)]i) may be increased at the terminals of CRH neurons compared with concentrations in the cell body. Moreover, GABAergic terminals projecting from the arcuate nucleus were present in close proximity to CRH-positive nerve terminals. Furthermore, a GABAAR agonist increased the intracellular calcium (Ca(2+)) levels in the CRH neuron terminals but decreased the Ca(2+) levels in their somata. In addition, the increases in Ca(2+) concentrations were prevented by an NKCC1 inhibitor. We propose a novel mechanism by which the excitatory action of GABA maintains a steady-state CRH release from axon terminals in the ME.
Collapse
Affiliation(s)
- Keisuke Kakizawa
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
- Second Division, Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Miho Watanabe
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Hiroki Mutoh
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Yuta Okawa
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
- Second Division, Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Miho Yamashita
- Second Division, Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Keiichi Itoi
- Laboratory of Information Biology, Graduate School of Information Sciences, Tohoku University, Sendai, Miyagi 980-8579, Japan
| | - Takafumi Suda
- Second Division, Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Yutaka Oki
- Department of Family and Community Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| |
Collapse
|
44
|
Sprow GM, Rinker JA, Lowery-Gointa EG, Sparrow AM, Navarro M, Thiele TE. Lateral hypothalamic melanocortin receptor signaling modulates binge-like ethanol drinking in C57BL/6J mice. Addict Biol 2016; 21:835-46. [PMID: 25975524 DOI: 10.1111/adb.12264] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Binge ethanol drinking is a highly pervasive and destructive behavior yet the underlying neurobiological mechanisms remain poorly understood. Recent work suggests that overlapping neurobiological mechanisms modulate feeding disorders and excessive ethanol intake, and converging evidence indicates that the melanocortin (MC) system may be a promising candidate. The aims of the present work were to examine how repeated binge-like ethanol drinking, using the 'drinking in the dark' (DID) protocol, impacts key peptides within the MC system and if site-specific manipulation of MC receptor (MCR) signaling modulates binge-like ethanol drinking. Male C57BL/6J mice were exposed to one, three or six cycles of binge-like ethanol, sucrose or water drinking, after which brain tissue was processed via immunohistochemistry (IHC) for analysis of key MC peptides, including alpha-melanocyte stimulating hormone (α-MSH) and agouti-related protein (AgRP). Results indicated that α-MSH expression was selectively decreased, while AgRP expression was selectively increased, within specific hypothalamic subregions following repeated binge-like ethanol drinking. To further explore this relationship, we used site-directed drug delivery techniques to agonize or antagonize MCRs within the lateral hypothalamus (LH). We found that the nonselective MCR agonist melanotan-II (MTII) blunted, while the nonselective MCR antagonist AgRP augmented, binge-like ethanol consumption when delivered into the LH. As these effects were region-specific, the present results suggest that a more thorough understanding of the MC neurocircuitry within the hypothalamus will help provide novel insight into the mechanisms that modulate excessive binge-like ethanol intake and may help uncover new therapeutic targets aimed at treating alcohol abuse disorders.
Collapse
Affiliation(s)
- Gretchen M. Sprow
- Department of Psychology and Neuroscience; University of North Carolina at Chapel Hill; Chapel Hill NC USA
| | - Jennifer A. Rinker
- Department of Psychology and Neuroscience; University of North Carolina at Chapel Hill; Chapel Hill NC USA
- Bowles Center for Alcohol Studies; University of North Carolina at Chapel Hill; Chapel Hill NC USA
| | - Emily G. Lowery-Gointa
- Department of Psychology and Neuroscience; University of North Carolina at Chapel Hill; Chapel Hill NC USA
- Bowles Center for Alcohol Studies; University of North Carolina at Chapel Hill; Chapel Hill NC USA
| | - Angela M. Sparrow
- Department of Psychology and Neuroscience; University of North Carolina at Chapel Hill; Chapel Hill NC USA
| | - Montserrat Navarro
- Department of Psychology and Neuroscience; University of North Carolina at Chapel Hill; Chapel Hill NC USA
- Bowles Center for Alcohol Studies; University of North Carolina at Chapel Hill; Chapel Hill NC USA
| | - Todd E. Thiele
- Department of Psychology and Neuroscience; University of North Carolina at Chapel Hill; Chapel Hill NC USA
- Bowles Center for Alcohol Studies; University of North Carolina at Chapel Hill; Chapel Hill NC USA
| |
Collapse
|
45
|
Knudsen LB, Secher A, Hecksher-Sørensen J, Pyke C. Long-acting glucagon-like peptide-1 receptor agonists have direct access to and effects on pro-opiomelanocortin/cocaine- and amphetamine-stimulated transcript neurons in the mouse hypothalamus. J Diabetes Investig 2016; 7 Suppl 1:56-63. [PMID: 27186357 PMCID: PMC4854506 DOI: 10.1111/jdi.12463] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/21/2015] [Indexed: 12/17/2022] Open
Abstract
Liraglutide is a glucagon‐like peptide‐1 receptor (GLP‐1R) agonist marketed for the treatment of type 2 diabetes. Besides lowering blood glucose, liraglutide reduces bodyweight, and has recently also been approved for the obesity indication. Acutely, GLP‐1 markedly reduces gastric emptying, and this effect was previously believed to at least partly explain the effect on bodyweight loss. However, recent studies in both humans and animals have shown that GLP‐1R agonists, such as liraglutide, that lead to pharmacological concentrations for 24 h/day only have a minor effect on gastric emptying; such an effect is unlikely to have lasting effects on appetite reduction. Liraglutide has been shown to have direct effects in the arcuate nucleus of the rodent brain, activating pro‐opiomelanocortin neurons and increasing levels of the cocaine‐ and amphetamine‐stimulated transcript neuropeptide messenger ribonucleic acid, which correlate nicely to clinical studies where liraglutide was shown to increase feelings of satiety. However, despite the lack of a GLP‐1R on agouti‐related peptide/neuropeptide Y neurons, liraglutide also was able to prevent a hunger associated increase in agouti‐related peptide and neuropeptide Y neuropeptide messenger ribonucleic acid, again with a strong correlation to clinical studies that document reduced hunger feelings in patients while taking liraglutide. Studies using fluorescent labeled liraglutide, as well as other GLP‐1R agonists, and analysis using single‐plane illumination microscopy show that such medium‐sized peptide‐based compounds can directly access not only circumventricular organs of the brain, but also directly access discrete regions in the hypothalamus. The direct effects of long‐acting GLP‐1R agonists in the hypothalamus are likely to be an important new pathway in understanding GLP‐1R agonist mediated weight loss.
Collapse
Affiliation(s)
| | - Anna Secher
- Novo Nordisk A/S Novo Nordisk Park Maaloev Denmark
| | | | - Charles Pyke
- Novo Nordisk A/S Novo Nordisk Park Maaloev Denmark
| |
Collapse
|
46
|
Food intake inhibition in rainbow trout induced by activation of serotonin 5-HT2C receptors is associated with increases in POMC, CART and CRF mRNA abundance in hypothalamus. J Comp Physiol B 2016; 186:313-21. [PMID: 26832922 DOI: 10.1007/s00360-016-0961-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/05/2016] [Accepted: 01/15/2016] [Indexed: 11/27/2022]
Abstract
In rainbow trout, the food intake inhibition induced by serotonin occurs through 5-HT2C and 5-HT1A receptors, though the mechanisms involved are still unknown. Therefore, we assessed if a direct stimulation of 5-HT2C and 5-HT1A serotonin receptors (resulting in decreased food intake in rainbow trout), affects gene expression of neuropeptides involved in the control of food intake, such as pro-opiomelanocortin (POMC), cocaine- and amphetamine-regulated transcript (CART), corticotrophin releasing factor (CRF), and agouti-related peptide (AgRP). In a first set of experiments, the injection of the 5-HT2C receptor agonists MK212 (60 μg kg(-1) icv) and WAY 161503 (1 mg kg(-1) ip), and of the 5-HT1A receptor agonist 8-OH-DPAT (1 mg kg(-1) ip and 30 μg kg(-1) icv) induced food intake inhibition. In a second set of experiments, we observed that the injection of MK212 or WAY 161503 (1 and 3 mg kg(-1)) significantly increased hypothalamic POMC mRNA abundance. CART mRNA abundance in hypothalamus was enhanced by treatment with MK212 and unaffected by WAY 161503. The administration of the 5-HT1A receptor agonist 8-OH-DPAT did not induce any significant variation in the hypothalamic POMC or CART mRNA levels. CRF mRNA abundance was only affected by MK212 that increased hypothalamic values. Finally, hypothalamic AgRP mRNA abundance was only evaluated with the agonist 5-HT2C MK212 resulting in no significant effects. The results show that the reduction in food intake mediated by 5-HT2C receptors is associated with increases in hypothalamic POMC, CART and CRF mRNA abundance.
Collapse
|
47
|
Interacting Neural Processes of Feeding, Hyperactivity, Stress, Reward, and the Utility of the Activity-Based Anorexia Model of Anorexia Nervosa. Harv Rev Psychiatry 2016; 24:416-436. [PMID: 27824637 PMCID: PMC5485261 DOI: 10.1097/hrp.0000000000000111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Anorexia nervosa (AN) is a psychiatric illness with minimal effective treatments and a very high rate of mortality. Understanding the neurobiological underpinnings of the disease is imperative for improving outcomes and can be aided by the study of animal models. The activity-based anorexia rodent model (ABA) is the current best parallel for the study of AN. This review describes the basic neurobiology of feeding and hyperactivity seen in both ABA and AN, and compiles the research on the role that stress-response and reward pathways play in modulating the homeostatic drive to eat and to expend energy, which become dysfunctional in ABA and AN.
Collapse
|
48
|
Yang L, McKnight GS. Hypothalamic PKA regulates leptin sensitivity and adiposity. Nat Commun 2015; 6:8237. [PMID: 26381935 PMCID: PMC4576457 DOI: 10.1038/ncomms9237] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/30/2015] [Indexed: 12/17/2022] Open
Abstract
Mice lacking the RIIβ regulatory subunit of cyclic AMP-dependent protein kinase A (PKA) display reduced adiposity and resistance to diet-induced obesity. Here we show that RIIβ knockout (KO) mice have enhanced sensitivity to leptin's effects on both feeding and energy metabolism. After administration of a low dose of leptin, the duration of hypothalamic JAK/STAT3 signalling is increased, resulting in enhanced POMC mRNA induction. Consistent with the extended JAK/STAT3 activation, we find that the negative feedback regulator of leptin receptor signalling, Socs3, is inhibited in the hypothalamus of RIIβ KO mice. During fasting, RIIβ–PKA is activated and this correlates with an increase in CREB phosphorylation. The increase in CREB phosphorylation is absent in the fasted RIIβ KO hypothalamus. Selective inhibition of PKA activity in AgRP neurons partially recapitulates the leanness and resistance to diet-induced obesity of RIIβ KO mice. Our findings suggest that RIIβ–PKA modulates the duration of leptin receptor signalling and therefore the magnitude of the catabolic response to leptin. Mice lacking RIIβ, a regulatory subunit of protein kinase A, are lean and resistant to diet-induced obesity. Here, the authors show that RIIβ regulates leptin sensitivity, acting as a physiological brake on leptin responsiveness and the duration of leptin signalling in the hypothalamus.
Collapse
Affiliation(s)
- Linghai Yang
- Department of Pharmacology, University of Washington School of Medicine, 1959 North East Pacific Street, Box 357280, Seattle, Washington 98195, USA
| | - G Stanley McKnight
- Department of Pharmacology, University of Washington School of Medicine, 1959 North East Pacific Street, Box 357280, Seattle, Washington 98195, USA
| |
Collapse
|
49
|
Abstract
Prolactin (PRL) released from lactotrophs of the anterior pituitary gland in response to the suckling by the offspring is the major hormonal signal responsible for stimulation of milk synthesis in the mammary glands. PRL secretion is under chronic inhibition exerted by dopamine (DA), which is released from neurons of the arcuate nucleus of the hypothalamus into the hypophyseal portal vasculature. Suckling by the young activates ascending systems that decrease the release of DA from this system, resulting in enhanced responsiveness to one or more PRL-releasing hormones, such as thyrotropin-releasing hormone. The neuropeptide oxytocin (OT), synthesized in magnocellular neurons of the hypothalamic supraoptic, paraventricular, and several accessory nuclei, is responsible for contracting the myoepithelial cells of the mammary gland to produce milk ejection. Electrophysiological recordings demonstrate that shortly before each milk ejection, the entire neurosecretory OT population fires a synchronized burst of action potentials (the milk ejection burst), resulting in release of OT from nerve terminals in the neurohypophysis. Both of these neuroendocrine systems undergo alterations in late gestation that prepare them for the secretory demands of lactation, and that reduce their responsiveness to stimuli other than suckling, especially physical stressors. The demands of milk synthesis and release produce a condition of negative energy balance in the suckled mother, and, in laboratory rodents, are accompanied by a dramatic hyperphagia. The reduction in secretion of the adipocyte hormone, leptin, a hallmark of negative energy balance, may be an important endocrine signal to hypothalamic systems that integrate lactation-associated food intake with neuroendocrine systems.
Collapse
Affiliation(s)
- William R Crowley
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah Health Sciences Center, Salt Lake City, Utah
| |
Collapse
|
50
|
Thomas AL, Davis SM, Dierick HA. Of Fighting Flies, Mice, and Men: Are Some of the Molecular and Neuronal Mechanisms of Aggression Universal in the Animal Kingdom? PLoS Genet 2015; 11:e1005416. [PMID: 26312756 PMCID: PMC4551476 DOI: 10.1371/journal.pgen.1005416] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Aggressive behavior is widespread in the animal kingdom, but the degree of molecular conservation between distantly related species is still unclear. Recent reports suggest that at least some of the molecular mechanisms underlying this complex behavior in flies show remarkable similarities with such mechanisms in mice and even humans. Surprisingly, some aspects of neuronal control of aggression also show remarkable similarity between these distantly related species. We will review these recent findings, address the evolutionary implications, and discuss the potential impact for our understanding of human diseases characterized by excessive aggression.
Collapse
Affiliation(s)
- Amanda L. Thomas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Shaun M. Davis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Herman A. Dierick
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|