1
|
Loera-Lopez AL, Lord MN, Noble EE. Astrocytes of the hippocampus and responses to periprandial neuroendocrine hormones. Physiol Behav 2025; 295:114913. [PMID: 40209869 DOI: 10.1016/j.physbeh.2025.114913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/15/2025] [Accepted: 04/08/2025] [Indexed: 04/12/2025]
Abstract
Astrocytes have risen as stars in the field of energy homeostasis and neurocognitive function, acting as a bridge of communication between the periphery and the brain, providing metabolic support, signaling via gliotransmitters, and altering synaptic communication. Dietary factors and energy state have a profound influence on hippocampal function, and the hippocampus is critical for appropriate behavioral responses associated with feeding and internal hunger cues (being in the fasted or full state), but how the hippocampus senses periprandial status and is impacted by diet is largely unknown. Periprandial hormones act within the hippocampus to modulate processes involved in hippocampal-dependent learning and memory function and astrocytes likely play an important role in modulating this signaling. In addition to periprandial hormones, astrocytes are positioned to respond to changes in circulating nutrients like glucose. Here, we review literature investigating how astrocytes mediate changes in hippocampal function, highlighting astrocyte location, morphology, and function in the context of integrating glucose metabolism, neuroendocrine hormone action, and/or cognitive function in the hippocampus. Specifically, we discuss research findings on the effects of insulin, ghrelin, leptin, and GLP-1 on glucose homeostasis, neural activity, astrocyte function, and behavior in the hippocampus. Because obesogenic diets impact neuroendocrine hormones, astrocytes, and cognitive function, we also discuss the effects of diet and diet-induced obesity on these parameters.
Collapse
Affiliation(s)
- Ana L Loera-Lopez
- Neuroscience Graduate Program, University of Georgia, Athens, GA, 30606, USA; Department of Nutritional Sciences, University of Georgia, Athens, GA, 30606, USA
| | - Magen N Lord
- Department of Nutritional Sciences, University of Georgia, Athens, GA, 30606, USA
| | - Emily E Noble
- Neuroscience Graduate Program, University of Georgia, Athens, GA, 30606, USA; Department of Nutritional Sciences, University of Georgia, Athens, GA, 30606, USA.
| |
Collapse
|
2
|
Pearl AJ, Maddern XJ, Pinares-Garcia P, Ursich LT, Anversa RG, Shesham A, Brown RM, Reed FM, Giardino WJ, Lawrence AJ, Walker LC. Midbrain ghrelin receptor signalling regulates binge drinking in a sex specific manner. Nat Commun 2025; 16:2568. [PMID: 40089486 PMCID: PMC11910522 DOI: 10.1038/s41467-025-57880-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Risky drinking rates are rising, particularly in women, yet sex as a biological variable has only recently gained traction. The centrally projecting Edinger-Westphal (EWcp) nucleus has emerged as a key regulator of alcohol consumption. Here we found that EWcppeptidergic cells reduce binge drinking specifically in female mice. We show this effect is mediated by the ghrelin receptor (GHSR), with EWcppeptidergic inhibition blocking ghrelin-induced drinking and Ghsr knockdown in EWcppeptidergic, but not EWcpglutamatergic or ventral tegmental area cells, reducing binge drinking in females, independent of circulating sex hormones. Female mice showed higher EWcp Ghsr expression, and EWcppeptidergic neurons were more sensitive to ghrelin. Moreover, intra-EWcp delivery of GHSR inverse agonist and antagonist reduced binge drinking, suggesting direct actions of ghrelin. These findings highlight the EWcp as a critical mediator of excessive alcohol consumption via GHSR in female mice, offering insights into the ghrelin system's role in alcohol consumption.
Collapse
Affiliation(s)
- Amy J Pearl
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
| | - Xavier J Maddern
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Paulo Pinares-Garcia
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
| | - Lauren T Ursich
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Roberta G Anversa
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Arnav Shesham
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Robyn M Brown
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Felicia M Reed
- Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| | - William J Giardino
- Dept. of Psychiatry and Behavioural Sciences, Stanford University School of Medicine, Stanford, CA, 94305-5453, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, 94305-5453, USA
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Leigh C Walker
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia.
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia.
| |
Collapse
|
3
|
Edwards A, DeSante S, Spencer CD, Hyland L, Smith A, Sankhe AS, Szilvásy-Szabó A, Fekete C, Hill MN, Chee MJ, Abizaid A. Ghrelin Recruits the Endocannabinoid System to Modulate Food Reward. J Neurosci 2025; 45:e1620242024. [PMID: 39779372 DOI: 10.1523/jneurosci.1620-24.2024] [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: 08/26/2024] [Revised: 12/05/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
Ghrelin enhances feeding by activating the growth hormone secretagogue receptor (GHSR). In the brain, GHSRs are expressed in regions responsible for regulating food motivation including the ventral tegmental area (VTA). Endogenous cannabinoids also promote food-seeking behaviors through the cannabinoid receptor-1 (CB-1Rs) in brain regions including the VTA. It is not known, however, if ghrelin and endocannabinoids interact in the VTA to produce these effects. We therefore examined if GHSR and CB-1R interact within the VTA to enhance food motivation. Results show that GHSR and CB-1R mRNA are expressed in the VTA cells in male and female rats and mice, with the GHSR being expressed in dopamine cells and the CB-1R being expressed primarily in nondopaminergic cells with no obvious sex differences. Ghrelin directly activated and increased excitatory tone onto dopamine cells of male and female mice. Male rats lacking fully functional GHSR signaling showed disrupted gene expression of transcripts important for regulating the synthesis, release, and degradation of endocannabinoids and lowered the levels of 2-arachidonoylglycerol (2-AG) within the VTA. Moreover, pharmacological antagonism of VTA CB-1Rs attenuates the orexigenic and appetitive effects of intra-VTA ghrelin in rats and blocks the ability of ghrelin to promote excitatory drive to VTA dopamine neurons. Finally, blocking the breakdown of cannabinoids in the VTA enhances the effects of ghrelin on food motivation. Together, our data show that ghrelin stimulates VTA dopamine cells and ultimately food motivation in part through a mechanism that involves endocannabinoid signaling at the CB-1R.
Collapse
Affiliation(s)
- Alexander Edwards
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Stephanie DeSante
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Carl Duncan Spencer
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Lindsay Hyland
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Andrea Smith
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Aditi S Sankhe
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Anett Szilvásy-Szabó
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Budapest H-1083, Hungary
| | - Csaba Fekete
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Budapest H-1083, Hungary
| | - Matthew N Hill
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N4T1, Canada
| | - Melissa J Chee
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Alfonso Abizaid
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| |
Collapse
|
4
|
Yang M, Singh A, de Araujo A, McDougle M, Ellis H, Décarie-Spain L, Kanoski SE, de Lartigue G. Separate orexigenic hippocampal ensembles shape dietary choice by enhancing contextual memory and motivation. Nat Metab 2025; 7:276-296. [PMID: 39815079 PMCID: PMC11860247 DOI: 10.1038/s42255-024-01194-6] [Citation(s) in RCA: 1] [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: 10/23/2023] [Accepted: 11/28/2024] [Indexed: 01/18/2025]
Abstract
The hippocampus (HPC) has emerged as a critical player in the control of food intake, beyond its well-known role in memory. While previous studies have primarily associated the HPC with food intake inhibition, recent research suggests a role in appetitive processes. Here we identified spatially distinct neuronal populations within the dorsal HPC (dHPC) that respond to either fats or sugars, potent natural reinforcers that contribute to obesity development. Using activity-dependent genetic capture of nutrient-responsive dHPC neurons, we demonstrate a causal role of both populations in promoting nutrient-specific intake through different mechanisms. Sugar-responsive neurons encoded spatial memory for sugar location, whereas fat-responsive neurons selectively enhanced the preference and motivation for fat intake. Importantly, stimulation of either nutrient-responsive dHPC neurons increased food intake, while ablation differentially impacted obesogenic diet consumption and prevented diet-induced weight gain. Collectively, these findings uncover previously unknown orexigenic circuits underlying macronutrient-specific consumption and provide a foundation for developing potential obesity treatments.
Collapse
Affiliation(s)
- Mingxin Yang
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arashdeep Singh
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alan de Araujo
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Molly McDougle
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hillary Ellis
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Léa Décarie-Spain
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Scott E Kanoski
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Guillaume de Lartigue
- Monell Chemical Senses Center, Philadelphia, PA, USA.
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Conde KM, Wong H, Fang S, Li Y, Yu M, Deng Y, Liu Q, Fang X, Wang M, Shi Y, Ginnard OZ, Yang Y, Tu L, Liu H, Liu H, Yin N, Bean JC, Han J, Burt ME, Jossy SV, Yang Y, Tong Q, Arenkiel BR, Wang C, He Y, Xu Y. Serotonin neurons integrate GABA and dopamine inputs to regulate meal initiation. Metabolism 2025; 163:156099. [PMID: 39667432 PMCID: PMC11924950 DOI: 10.1016/j.metabol.2024.156099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/04/2024] [Accepted: 12/07/2024] [Indexed: 12/14/2024]
Abstract
Obesity is a growing global health epidemic with limited orally administered therapeutics. Serotonin (5-HT) is one neurotransmitter which remains an excellent target for new weight-loss therapies, but a gap remains in understanding the mechanisms involved in 5-HT produced in the dorsal Raphe nucleus (DRN) and its involvement in meal initiation. Using an optogenetic feeding paradigm, we showed that the 5-HTDRN➔arcuate nucleus (ARH) circuit plays a role in meal initiation. Incorporating electrophysiology and ChannelRhodopsin-2-Assisted Circuit Mapping, we demonstrated that 5-HTDRN neurons receive inhibitory input partially from GABAergic neurons in the DRN, and the 5-HT response can be enhanced by hunger. Additionally, deletion of the GABAA receptor subunit in 5-HT neurons inhibits meal initiation with no effect on the satiation process. Finally, we identified the role of dopaminergic inputs via dopamine receptor D2 in enhancing the response to GABA-induced feeding. Thus, our results indicate that 5-HTDRN neurons are inhibited by synergistic inhibitory actions of GABA and dopamine, for the initiation of a meal.
Collapse
Affiliation(s)
- Kristine M Conde
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - HueyZhong Wong
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Shuzheng Fang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yongxiang Li
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Meng Yu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yue Deng
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Qingzhuo Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Xing Fang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Mengjie Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yuhan Shi
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Olivia Z Ginnard
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yuxue Yang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Longlong Tu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hesong Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hailan Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Na Yin
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jonathan C Bean
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Junying Han
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Megan E Burt
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Sanika V Jossy
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yongjie Yang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chunmei Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yang He
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
6
|
Faulkner ML, Farokhnia M, Lee MR, Farinelli L, Browning BD, Abshire K, Daurio AM, Munjal V, Deschaine SL, Boukabara SR, Fortney C, Sherman G, Schwandt M, Akhlaghi F, Momenan R, Ross TJ, Persky S, Leggio L. A randomized, double-blind, placebo-controlled study of a GHSR blocker in people with alcohol use disorder. JCI Insight 2024; 9:e182331. [PMID: 39704175 PMCID: PMC11665556 DOI: 10.1172/jci.insight.182331] [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: 04/23/2024] [Accepted: 10/30/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUNDStudies have demonstrated the role of ghrelin in alcohol-related behaviors and consumption. Blockade of the growth hormone secretagogue receptor (GHSR), which is the ghrelin receptor, has been shown to decrease alcohol drinking and reward-related behaviors across several animal models. We previously conducted a human study testing a GHSR inverse agonist/competitive antagonist, PF-5190457, in individuals who are heavy drinkers and showed its safety when coadministered with alcohol. Here, we conducted a phase IIa experimental medicine study in patients with alcohol use disorder (AUD) to investigate the effects of PF-5190457 on alcohol- and food-related outcomes.METHODSForty-two individuals with AUD (n = 29 completers) participated in a randomized, double-blind, placebo-controlled study where they received PF-5190457 100mg b.i.d. (or placebo) in 2 counterbalanced, within-subject stages. Participants completed an alcohol cue-reactivity (CR) experiment in a bar-like laboratory and a virtual food choice experiment in a cafeteria-like virtual reality (VR) environment. A subset of participants (n = 12) performed a CR task during a brain functional MRI (fMRI) experiment.RESULTSPF-5190457 did not reduce cue-elicited alcohol craving. PF-5190457 reduced virtual calories selected (P = 0.04) in the VR environment. PF-5190457 did not influence neural activation during CR task in the fMRI experiment.CONCLUSIONThis study provides human evidence of the role of GHSR blockade in behaviors related to food selection and highlights the need for future investigations into targeting the ghrelin system in AUD.TRIAL REGISTRATIONClinicalTrials.gov (accession no. NCT02707055).FUNDINGNIDA and NIAAA ZIA-DA000635; National Center for Advancing Translational Sciences UH2/UH3-TR000963.
Collapse
Affiliation(s)
- Monica L. Faulkner
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Mary R. Lee
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Lisa Farinelli
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Brittney D. Browning
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Kelly Abshire
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Allison M. Daurio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Vikas Munjal
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Sara L. Deschaine
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Selim R. Boukabara
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Christopher Fortney
- Immersive Simulation Program, Social and Behavioral Research Branch, National Human Genome Research Institute (NHGRI), NIH, Bethesda, Maryland, USA
| | - Garrick Sherman
- Office of the Clinical Director, NIDA, Intramural Research Program, NIH, Baltimore, Maryland, USA
| | - Melanie Schwandt
- Office of the Clinical Director, NIAAA Division of Intramural Clinical and Biological Research, NIH, Bethesda, Maryland, USA
| | - Fatemeh Akhlaghi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - Reza Momenan
- Clinical NeuroImaging Research Core, NIAAA, NIH, Bethesda, Maryland, USA
| | - Thomas J. Ross
- Neuroimaging Core, NIDA Intramural Research Program, NIH, Baltimore, Maryland, USA
| | - Susan Persky
- Immersive Simulation Program, Social and Behavioral Research Branch, National Human Genome Research Institute (NHGRI), NIH, Bethesda, Maryland, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island, USA
- Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Neuroscience, Georgetown University Medical Center, Washington DC, USA
| |
Collapse
|
7
|
Saenz C, Fernandez G, Llovera R, Tolosa MJ, Cantel S, Fehrentz JA, Mackie K, Leggio L, Zigman J, De Francesco PN, Perello M. Growth hormone secretagogue receptor and cannabinoid receptor type 1 intersection in the mouse brain. Brain Struct Funct 2024; 230:15. [PMID: 39702649 PMCID: PMC11659360 DOI: 10.1007/s00429-024-02876-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/14/2024] [Indexed: 12/21/2024]
Abstract
The growth hormone secretagogue receptor (GHSR) and the cannabinoid receptor type 1 (CB1R) are G-protein coupled receptors highly expressed in the brain and involved in critical regulatory processes, such as energy homeostasis, appetite control, reward, and stress responses. GHSR mediates the effects of both ghrelin and liver-expressed antimicrobial peptide 2, while CB1R is targeted by cannabinoids. Strikingly, both receptors mediate their effects by acting on common brain areas and their individual roles have been well characterized. However, the potential for their co-expression in the same neuronal subsets remains largely unexplored. Here, we aim to map the cell populations where GHSR and CB1R might converge, hypothesizing that their co-expression in specific brain circuits could mediate integrated physiological responses. By utilizing two complementary labeling techniques-GHSR-eGFP mice and Fr-ghrelin labeling of GHSR+ cells-along with specific CB1R immunostaining, we sought to visualize and quantify potential areas of overlap. Also, we analyzed several cell RNA sequencing datasets to estimate the fraction of brain cells expressing both GPCRs and their phenotype. Our neuroanatomical studies revealed evident overlap of GHSR+ and CB1R+ signals in specific neuronal subsets mainly located in the cerebral cortex, hippocampus and the amygdala. Transcriptomic analysis revealed specific subsets of Ghsr+/Cnr1+ glutamatergic neurons in the hippocampus and amygdala, as well as different subtypes of Ghsr+/Cnr1+ neurons in the midbrain, hypothalamus, pons, and medulla. Thus, we revealed that GHSR and CB1R interact differentially across specific regions of the mouse brain, providing new insights into how these receptors' actions are integrated. Current findings may open new avenues for dual therapeutic interventions in metabolic disorders, obesity, and psychiatric conditions.
Collapse
Affiliation(s)
- Camila Saenz
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Gimena Fernandez
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Ramiro Llovera
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - María J Tolosa
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Kenneth Mackie
- Department of Psychological & Brain Sciences, Indiana University, Bloomington Indiana, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Jeffrey Zigman
- Center for Hypothalamic Research and Division of Endocrinology & Metabolism, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pablo N De Francesco
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina.
| | - Mario Perello
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina.
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden.
| |
Collapse
|
8
|
Bechtel W. Hierarchy or Heterarchy of Mammalian Circadian Timekeepers? J Biol Rhythms 2024; 39:513-534. [PMID: 39449278 PMCID: PMC11613639 DOI: 10.1177/07487304241286573] [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] [Indexed: 10/26/2024]
Abstract
Mammalian circadian biologists commonly characterize the relation between circadian clocks as hierarchical, with the clock in the suprachiasmatic nucleus at the top of the hierarchy. The lineage of research since the suprachiasmatic nucleus (SCN) was first identified as the clock in mammals has challenged this perspective, revealing clocks in peripheral tissues, showing that they respond to their own zeitgebers, coordinate oscillations among themselves, and in some cases modify the behavior of the SCN. Increasingly circadian timekeepers appear to constitute a heterarchical network, with control distributed and operating along multiple pathways. One reason for the continued invocation of hierarchy in mammalian circadian biology is that it is difficult to understand how a heterarchical system could operate effectively so as to maintain the organism. Evolved mechanisms, however, need not respect hierarchy and those that have survived have demonstrated the ability of heterarchical organizaton to maintain organisms.
Collapse
Affiliation(s)
- William Bechtel
- Department of Philosophy, University of California, San Diego, La Jolla, California
| |
Collapse
|
9
|
Aguggia J, Fernandez G, Cassano D, Mustafá ER, Rodríguez SS, Cantel S, Fehrentz JA, Raingo J, Schiöth HB, Habib AM, De Francesco PN, Perello M. Selective Colocalization of GHSR and GLP-1R in a Subset of Hypothalamic Neurons and Their Functional Interaction. Endocrinology 2024; 166:bqae160. [PMID: 39737802 DOI: 10.1210/endocr/bqae160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Indexed: 01/01/2025]
Abstract
The GH secretagogue receptor (GHSR) and the glucagon-like peptide-1 receptor (GLP-1R) are G protein-coupled receptors with critical, yet opposite, roles in regulating energy balance. Interestingly, these receptors are expressed in overlapping brain regions. However, the extent to which they target the same neurons and engage in molecular crosstalk remains unclear. To explore the potential colocalization of GHSR and GLP-1R in specific neurons, we performed detailed mapping of cells positive for both receptors using GHSR-eGFP reporter mice or wild-type mice infused with fluorescent ghrelin, alongside an anti-GLP-1R antibody. We found that GHSR+ and GLP-1R+ cells are largely segregated in the mouse brain. The highest overlap was observed in the hypothalamic arcuate nucleus, where 15% to 20% of GHSR+ cells were also GLP-1R+ cells. Additionally, we examined RNA-sequencing datasets from mouse and human brains to assess the fraction and distribution of neurons expressing both receptors, finding that double-positive Ghsr+/Glp1r+ cells are highly segregated, with a small subset of double-positive Ghsr+/Glp1r+ cells representing <10% of all Ghsr+ or Glp1r+ cells, primarily enriched in the hypothalamus. Furthermore, we conducted functional studies using patch-clamp recordings in a heterologous expression system to assess potential crosstalk in regulating presynaptic calcium channels. We provide the first evidence that liraglutide-evoked GLP-1R activity inhibits presynaptic channels, and that the presence of one GPCR attenuates the inhibitory effects of ligand-evoked activity mediated by the other on presynaptic calcium channels. In conclusion, while GHSR and GLP-1R can engage in molecular crosstalk, they are largely segregated across most neuronal types within the brain.
Collapse
Affiliation(s)
- Julieta Aguggia
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Gimena Fernandez
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Daniela Cassano
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Emilio R Mustafá
- Laboratory of Electrophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Silvia S Rodríguez
- Laboratory of Electrophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, 34293 Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, 34293 Montpellier, France
| | - Jesica Raingo
- Laboratory of Electrophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, 751 24 Uppsala, Sweden
| | - Abdella M Habib
- College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Pablo N De Francesco
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, 751 24 Uppsala, Sweden
| |
Collapse
|
10
|
Yakabi K, Yamaguchi N, Takayama K, Hosomi E, Hori Y, Ro S, Ochiai M, Maezawa K, Yakabi S, Harada Y, Fujitsuka N, Nagoshi S. Rikkunshito improves anorexia through ghrelin- and orexin-dependent activation of the brain hypothalamus and mesolimbic dopaminergic pathway in rats. Neurogastroenterol Motil 2024; 36:e14900. [PMID: 39164871 DOI: 10.1111/nmo.14900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 07/17/2024] [Accepted: 07/29/2024] [Indexed: 08/22/2024]
Abstract
BACKGROUND Rikkunshito (RKT), a traditional Japanese medicine, can relieve epigastric discomfort and anorexia in patients with functional dyspepsia. RKT enhances the orexigenic hormone, ghrelin. Ghrelin regulates food motivation by stimulating the appetite control center in the hypothalamus and the brain mesolimbic dopaminergic pathway (MDPW). However, the effect of RKT on MDPW remains unclear. Here, we aimed to investigate the central neural mechanisms underlying the orexigenic effects of RKT, focusing on the MDPW. METHODS We examined the effects of RKT on food intake and neuronal c-Fos expression in restraint stress- and cholecystokinin octapeptide-induced anorexia in male rats. KEY RESULTS RKT treatment significantly restored stress- and cholecystokinin octapeptide-induced decreased food intake. RKT increased c-Fos expression in the ventral tegmental area (VTA), especially in tyrosine hydroxylase-immunoreactive neurons, and nucleus accumbens (NAc). The effects of RKT were suppressed by the ghrelin receptor antagonist [D-Lys3]-GHRP-6. RKT increased the number of c-Fos/orexin-double-positive neurons in the lateral hypothalamus (LH), which project to the VTA. The orexin receptor antagonist, SB334867, suppressed RKT-induced increase in food intake and c-Fos expression in the LH, VTA, and NAc. RKT increased c-Fos expression in the arcuate nucleus and nucleus of the solitary tract of the medulla, which was inhibited by [D-Lys3]-GHRP-6. CONCLUSIONS & INFERENCES RKT may restore appetite in subjects with anorexia through ghrelin- and orexin-dependent activation of neurons regulating the brain appetite control network, including the hypothalamus and MDPW.
Collapse
Affiliation(s)
- Koji Yakabi
- Department of Gastroenterology and Hepatology, Saitama Medical Center, Saitama Medical University, Kawagoe City, Saitama, Japan
| | - Naomi Yamaguchi
- Department of Gastroenterology and Hepatology, Saitama Medical Center, Saitama Medical University, Kawagoe City, Saitama, Japan
| | - Kiyoshige Takayama
- Department of Gastroenterology and Hepatology, Saitama Medical Center, Saitama Medical University, Kawagoe City, Saitama, Japan
| | - Eriko Hosomi
- Department of Gastroenterology and Hepatology, Saitama Medical Center, Saitama Medical University, Kawagoe City, Saitama, Japan
| | - Yutaro Hori
- Department of Gastroenterology and Hepatology, Saitama Medical Center, Saitama Medical University, Kawagoe City, Saitama, Japan
| | - Shoki Ro
- Department of Gastroenterology and Hepatology, Saitama Medical Center, Saitama Medical University, Kawagoe City, Saitama, Japan
| | - Mitsuko Ochiai
- Department of Gastroenterology and Hepatology, Saitama Medical Center, Saitama Medical University, Kawagoe City, Saitama, Japan
| | - Kosuke Maezawa
- Department of Gastroenterology and Hepatology, Saitama Medical Center, Saitama Medical University, Kawagoe City, Saitama, Japan
| | - Seiichi Yakabi
- Department of Gastroenterology and Hepatology, Saitama Medical Center, Saitama Medical University, Kawagoe City, Saitama, Japan
- Department of Gastroenterology, University of Tokyo Hospital, Tokyo, Japan
| | - Yumi Harada
- TSUMURA Kampo Research Laboratories, TSUMURA & CO., Ibaraki, Japan
| | - Naoki Fujitsuka
- TSUMURA Kampo Research Laboratories, TSUMURA & CO., Ibaraki, Japan
| | - Sumiko Nagoshi
- Department of Gastroenterology and Hepatology, Saitama Medical Center, Saitama Medical University, Kawagoe City, Saitama, Japan
| |
Collapse
|
11
|
Stark R, Dempsey H, Kleeman E, Sassi M, Osborne-Lawrence S, Sheybani-Deloui S, Rushby HJ, Mirth CK, Austin-Muttitt K, Mullins J, Zigman JM, Davies JS, Andrews ZB. Hunger signalling in the olfactory bulb primes exploration, food-seeking and peripheral metabolism. Mol Metab 2024; 89:102025. [PMID: 39236785 PMCID: PMC11471258 DOI: 10.1016/j.molmet.2024.102025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/15/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024] Open
Abstract
OBJECTIVE Although the metabolic state of an organism affects olfactory function, the precise mechanisms and their impact on behavior and metabolism remain unknown. Here, we assess whether ghrelin receptors (GHSRs) in the olfactory bulb (OB) increase olfactory function and influence foraging behaviors and metabolism. METHODS We performed a detailed behavioural and metabolic analysis in mice lacking GHSRs in the OB (OBGHSR deletion). We also analsyed OB scRNA-seq and spatial transcriptomic datasets to assess GHSR+ cells in the main and accessory olfactory bulbs, as well as the anterior olfactory nucleus. RESULTS OBGHSR deletion affected olfactory discrimination and habituation to both food and non-food odors. Anxiety-like and depression-like behaviors were significantly greater after OBGHSR deletion, whereas exploratory behavior was reduced, with the greatest effect under fasted conditions. OBGHSR deletion impacted feeding behavior as evidenced by altered bout number and duration, as well as buried food-seeking. OBGHSR deletion increased body weight and fat mass, spared fat utilisation on a chow diet and impaired glucose metabolism indicating metabolic dysfunction. Cross referenced analysis of OB scRNA-seq and spatial transcriptomic datasets revealed GHSR+ glutamate neurons in the main and accessory olfactory bulbs, as well as the anterior olfactory nucleus. Ablation of glutamate neurons in the OB reduced ghrelin-induced food finding and phenocopied results seen after OBGHSR deletion. CONCLUSIONS OBGHSRs help to maintain olfactory function, particularly during hunger, and facilitate behavioral adaptations that optimise food-seeking in anxiogenic environments, priming metabolic pathways in preparation for food consumption.
Collapse
Affiliation(s)
- Romana Stark
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia.
| | - Harry Dempsey
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Elizabeth Kleeman
- The Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, Australia
| | - Martina Sassi
- Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Sherri Osborne-Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sepideh Sheybani-Deloui
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Helen J Rushby
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| | - Christen K Mirth
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| | - Karl Austin-Muttitt
- Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Jonathan Mullins
- Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey S Davies
- Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Zane B Andrews
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
12
|
Zhang M, Yang L, Jia J, Xu F, Gao S, Han F, Deng M, Wang J, Li V, Yu M, Sun Y, Yuan H, Zhou Y, Li N. Increased GHS-R1a expression in the hippocampus impairs memory encoding and contributes to AD-associated memory deficits. Commun Biol 2024; 7:1334. [PMID: 39415032 PMCID: PMC11484987 DOI: 10.1038/s42003-024-06914-y] [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: 03/21/2024] [Accepted: 09/17/2024] [Indexed: 10/18/2024] Open
Abstract
Growth hormone secretagogue receptor 1a (GHS-R1a), also known as the ghrelin receptor, is an important nutrient sensor and metabolic regulator in both humans and rodents. Increased GHS-R1a expression is observed in the hippocampus of both Alzheimer's disease (AD) patients and AD model mice. However, the causal relationship between GHS-R1a elevation in the hippocampus and AD memory deficits remains uncertain. Here, we find that increasing GHS-R1a expression in dCA1 pyramidal neurons impairs hippocampus-dependent memory formation, which is abolished by local administration of the endogenous antagonist LEAP2. GHS-R1a elevation in dCA1 pyramidal neurons suppresses excitability and blocks memory allocation in these neurons. Chemogenetic activation of those high GHS-R1a neurons during training rescues GHS-R1a overexpression-induced memory impairment. Moreover, we demonstrate that increasing GHS-R1a expression in dCA1 pyramidal neurons hampers these neurons' ability to encode spatial memory and reduces engram size in the dCA1 region. Finally, we show that GHS-R1a deletion mitigates spatial memory deficits in APP/PS1 mice with increased GHS-R1a expression in the hippocampus. Our findings reveal a negative, causal relationship between hippocampal GHS-R1a expression and memory encoding, and suggest that blocking the abnormal increase in GHS-R1a activity/expression may be a promising approach to improve memory and treat cognitive decline in AD.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266000, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
- College of Agriculture and Bioengineering, Heze University, Heze, Shandong, 274000, China
| | - Liu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Jiajia Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Fenghua Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Shanshan Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Fubing Han
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, China
| | - Mingru Deng
- Department of Neurology, Affiliated Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong, 266042, China
| | - Jiwei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Vincent Li
- Beverly Hills High School, Beverly Hills, CA, 90212, USA
| | - Ming Yu
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266000, China
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
| | - Haicheng Yuan
- Department of Neurology, Affiliated Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong, 266042, China
| | - Yu Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China.
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266000, China.
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China.
| | - Nan Li
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266000, China.
| |
Collapse
|
13
|
Sturaro C, Ruzza C, Ferrari F, Pola P, Argentieri M, Frezza A, Marzola E, Bettegazzi B, Cattaneo S, Pietra C, Malfacini D, Calò G. In vitro pharmacological characterization of growth hormone secretagogue receptor ligands using the dynamic mass redistribution and calcium mobilization assays. Eur J Pharmacol 2024; 981:176880. [PMID: 39128804 DOI: 10.1016/j.ejphar.2024.176880] [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: 05/13/2024] [Revised: 07/25/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Ghrelin modulates several biological functions via selective activation of the growth hormone secretagogue receptor (GHSR). GHSR agonists may be useful for the treatment of anorexia and cachexia, while antagonists and inverse agonists may represent new drugs for the treatment of metabolic and substance use disorders. Thus, the identification and pharmacodynamic characterization of new GHSR ligands is of high interest. In the present work the label-free dynamic mass redistribution (DMR) assay has been used to evaluate the pharmacological activity of a panel of GHSR ligands. This includes the endogenous peptides ghrelin, desacyl-ghrelin and LEAP2(1-14). Among synthetic compounds, the agonists anamorelin and HM01, the antagonists HM04 and YIL-781, and the inverse agonist PF-05190457 have been tested, together with HM03, R011, and H1498 from patent literature. The DMR results have been compared to those obtained in parallel experiments with the calcium mobilization assay. Ghrelin, anamorelin, HM01, and HM03 behaved as potent full GHSR agonists. YIL-781 behaved as a partial GHSR agonist and R011 as antagonist in both the assays. LEAP2(1-14) resulted a GHSR inverse agonist in DMR but not in calcium mobilization assay. PF-05190457, HM04, and H1498 behaved as GHSR inverse agonists in DMR experiments, while they acted as antagonists in calcium mobilization studies. In conclusion, this study provided a systematic pharmacodynamic characterization of several GHSR ligands in two different pharmacological assays. It demonstrated that the DMR assay can be successfully used particularly to discriminate between antagonists and inverse agonists. This study may be useful for the selection of the most appropriate compounds to be used in future studies.
Collapse
Affiliation(s)
- Chiara Sturaro
- U.O. Neurological Clinic of the University Hospital of Ferrara, Italy
| | - Chiara Ruzza
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy; LTTA Laboratory for Advanced Therapies, Technopole of Ferrara, Ferrara, Italy.
| | - Federica Ferrari
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Pietro Pola
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Michela Argentieri
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Alessia Frezza
- U.O. Neurological Clinic of the University Hospital of Ferrara, Italy
| | - Erika Marzola
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | | | | | | | - Davide Malfacini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Girolamo Calò
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| |
Collapse
|
14
|
Zhang Z, Su D, Lai M, Song Y, Li H, Yang M, Zhu G, Liu H, Ai Z. New antidepressant mechanism of Yueju Pill: Increasing ghrelin level by inhibiting gastric mTOR/S6K signaling pathway and sensitizing hippocampal GHS-R. Heliyon 2024; 10:e37038. [PMID: 39296021 PMCID: PMC11407933 DOI: 10.1016/j.heliyon.2024.e37038] [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: 03/22/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/21/2024] Open
Abstract
Background and aim Yueju Pill (YJ) not only has good antidepressant effect but also can effectively treat digestive system diseases. However,it remains unclear whether the mechanism of antidepressant action of YJ is related to the peripheral digestive system. The purpose of this study was to elucidate the antidepressant mechanism of YJ on ghrelin level based on gastric mTOR/S6K signal pathway and sensitized hippocampal Ghrelin/GHS-R system in CUMS mice. Experimental procedure The depression model was induced by chronic unpredictable mild stress (CUMS) and social isolation. The antidepressant effect of YJ was observed by behavioral experiment and hemodynamic experiments. Ghrelin levels in in hippocampus and blood were measured by Elisa kit, and the mRNA of ghrelin in mice stomach was measured by Real-time Quantitative PCR (RT-qPCR). The activation level of gastric mTOR/S6K signal pathway was detected by Western Blot (WB). Rapamycin (Rapa) and L-Leucine (L-Leu) were used to verify the effects of YJ on the synthesis and release of ghrelin. The activity of GHS-R in hippocampus was observed by immunofluorescence. Hippocampal neuronal damage was evaluated by HE staining and Nissl staining. The level of central neurotransmitter was measured by liquid chromatograph mass spectrometer (LC-MS). Results and conclusion YJ ameliorates CUMS-induced depressive-like behavior by inhibiting the gastric mTOR/S6K signaling pathway and increasing GHR expression in the mouse stomach. However, these effects of YJ could be resisted by L-Leu (a mTOR receptor agonist). Further studies have shown that YJ can sensitize the Ghrelin/GHS-R system in the hippocampus, with significant neuroprotective effects, and is also involved in regulating the levels of key neurotransmitters (5-hydroxytryptamine, Dopamine and γ-aminobutyric acid) in depressive-like states.
Collapse
Affiliation(s)
- Zhentao Zhang
- Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Diseases with Mental Disorders), Key Laboratory of Depression Animal Model Based on TCM Syndrome, Jiangxi Administration of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang, 330006, China
| | - Dan Su
- Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Diseases with Mental Disorders), Key Laboratory of Depression Animal Model Based on TCM Syndrome, Jiangxi Administration of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang, 330006, China
| | - Meixizi Lai
- Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Diseases with Mental Disorders), Key Laboratory of Depression Animal Model Based on TCM Syndrome, Jiangxi Administration of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang, 330006, China
| | - Yonggui Song
- Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Diseases with Mental Disorders), Key Laboratory of Depression Animal Model Based on TCM Syndrome, Jiangxi Administration of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang, 330006, China
| | - Huizhen Li
- Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Diseases with Mental Disorders), Key Laboratory of Depression Animal Model Based on TCM Syndrome, Jiangxi Administration of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang, 330006, China
| | - Ming Yang
- Jiangxi Guxiang Jinyun Comprehensive Health Industry Co., Ltd., Nanchang, China
| | - Genhua Zhu
- Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Diseases with Mental Disorders), Key Laboratory of Depression Animal Model Based on TCM Syndrome, Jiangxi Administration of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang, 330006, China
| | - Hong Liu
- Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang, 330006, China
| | - Zhifu Ai
- Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Diseases with Mental Disorders), Key Laboratory of Depression Animal Model Based on TCM Syndrome, Jiangxi Administration of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang, 330006, China
| |
Collapse
|
15
|
Hewedy WA, Darwish IE. Activation of bitter taste receptors (TAS2R) protects against rotenone-induced neurotoxicity: Could ghrelin have a role? Eur J Pharmacol 2024; 978:176802. [PMID: 38945288 DOI: 10.1016/j.ejphar.2024.176802] [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: 03/11/2024] [Revised: 06/05/2024] [Accepted: 06/28/2024] [Indexed: 07/02/2024]
Abstract
AIMS Bitter taste receptors (TAS2Rs) and their downstream signaling pathways are expressed not only in the oral tissues but also in extraoral tissues. Emerging data has demonstrated the beneficial effect of ghrelin in neurodegenerative diseases. Gaining more insight into the interaction between TAS2Rs and gut hormones may expand their therapeutic applications. Herein, we aimed to assess the possible effect of TAS2R activation by denatonium benzoate (DB) in modulating functional and neurobiochemical alterations in a model of Parkinson's disease (PD). MAIN METHODS PD model was induced by daily injection of rotenone (2 mg/kg). Rats received DB (5 mg/kg), atenolol (10 mg/kg), or both concomitantly with rotenone, daily for 28 days. Evaluation of the motor abnormalities and histological examination of brain tissues were conducted. In addition, striatal dopamine contents, immunohistochemical expression of tyrosine hydroxylase, plasma ghrelin level, and biochemical analysis of markers of inflammation and oxidative stress were assessed. KEY FINDINGS Treatment with DB increased serum levels of ghrelin and striatal dopamine contents with consequent amelioration of oxidative stress and attenuation of inflammatory cytokines. Moreover, DB treatment significantly ameliorated motor disturbance and histological abnormalities compared to untreated rats. Atenolol inhibited ghrelin release and abolished the positive effect of DB suggesting the involvement of ghrelin on such effects. SIGNIFICANCE The current study suggests that TAS2Rs agonists are promising candidates for ameliorating rotenone-induced PD pathology in rats, an action that could be linked to the enhancement of ghrelin release with consequent antioxidant and anti-inflammatory activities.
Collapse
Affiliation(s)
- Wafaa A Hewedy
- Clinical Pharmacology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Inas E Darwish
- Clinical Pharmacology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
16
|
Casado S, Varela-Miguéns M, de Oliveira Diz T, Quintela-Vilariño C, Nogueiras R, Diéguez C, Tovar S. The effects of ghrelin and LEAP-2 in energy homeostasis are modulated by thermoneutrality, high-fat diet and aging. J Endocrinol Invest 2024; 47:2061-2074. [PMID: 38337094 PMCID: PMC11266414 DOI: 10.1007/s40618-024-02307-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/09/2024] [Indexed: 02/12/2024]
Abstract
PURPOSE Liver-expressed antimicrobial peptide 2 (LEAP-2) has been recently identified as the endogenous non-competitive allosteric antagonist of the growth hormone secretagogue receptor 1a (GHSR1a). In rodents, LEAP-2 blunts ghrelin-induced feeding and its plasma levels are modulated in response to nutritional status, being decreased upon fasting and increased in high-fat diet (HFD) fed mice. Clinical data support the regulation of circulating LEAP-2 by nutrient availability in humans. In this work, our primary objective was to examine the chronic effects of ghrelin and LEAP-2 administration on food intake, adiposity, and energy expenditure in young mice subjected to standard and HFD at both room temperature and at thermoneutrality. Furthermore, we aimed to assess the impact of these two hormones on aging mice. RESULTS Our results indicate that LEAP-2 produces a significant decrease of body weight and adiposity, an increase in energy expenditure, and activation of the thermogenic program in white and brown adipose tissue depots. However, this effect is not maintained under HFD or under thermoneutral conditions and is only partially observed in aging mice. CONCLUSION In summary our studies describe the central effects of LEAP-2 within distinct experimental contexts, and contribute to the comprehension of LEAP-2's role in energy metabolism.
Collapse
Affiliation(s)
- S Casado
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain
| | - M Varela-Miguéns
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain
| | - T de Oliveira Diz
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain
| | - C Quintela-Vilariño
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain
| | - R Nogueiras
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029, Madrid, Spain
| | - C Diéguez
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029, Madrid, Spain.
| | - S Tovar
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029, Madrid, Spain.
| |
Collapse
|
17
|
Englund A, Gilliam-Vigh H, Suppli MP, Gasbjerg LS, Vilsbøll T, Knop FK. Intestinal expression profiles and hepatic expression of LEAP2, ghrelin and their common receptor, GHSR, in humans. Peptides 2024; 177:171227. [PMID: 38657907 DOI: 10.1016/j.peptides.2024.171227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 04/26/2024]
Abstract
Liver-expressed antimicrobial peptide 2 (LEAP2) and ghrelin have reciprocal effects on their common receptor, the growth hormone secretagogue receptor (GHSR). Ghrelin is considered a gastric hormone and LEAP2 a liver-derived hormone and both have been proposed to be involved in the pathophysiology of obesity and type 2 diabetes (T2D). We investigated the mRNA expression of LEAP2, ghrelin and GHSR along the intestinal tract of individuals with and without TD2, and in the liver of men with and without obesity. Mucosal biopsies retrieved with 30-cm intervals throughout the small intestine and from 7 well-defined locations along the large intestine from 12 individuals with T2D and 12 healthy controls together with liver biopsies from 15 men with obesity and 15 lean men were subjected to bulk transcriptomics analysis. Both in individuals with and without T2D, mRNA expression of LEAP2 increased through the small intestine until dropping at the ileocecal valve, with little LEAP2 mRNA expression in the large intestine. Pronounced LEAP2 expression was observed in the liver of men with and without obesity. Robust ghrelin mRNA expression was observed in the duodenum of individuals with and without T2D, gradually decreasing along the small intestine with little expression in the large intestine. Ghrelin mRNA expression was not detected in the liver biopsies, and GHSR mRNA expression was not. In conclusion, we provide unique mRNA expression profiles of LEAP2, ghrelin and GHSR along the human intestinal tract showing no T2D-associated changes, and in the liver showing no differences between men with and without obesity.
Collapse
Affiliation(s)
- Anders Englund
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark.
| | - Hannah Gilliam-Vigh
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark.
| | - Malte P Suppli
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark.
| | - Lærke S Gasbjerg
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Tina Vilsbøll
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Herlev, Denmark.
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Herlev, Denmark; Novo Nordisk A/S, Novo Allé, Bagsværd, Denmark.
| |
Collapse
|
18
|
Smith A, MacAulay B, Scheufen J, Hudak A, Abizaid A. Chronic Social Defeat Stress Increases Brain Permeability to Ghrelin in Male Mice. eNeuro 2024; 11:ENEURO.0093-24.2024. [PMID: 38937108 PMCID: PMC11253241 DOI: 10.1523/eneuro.0093-24.2024] [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: 03/04/2024] [Revised: 06/05/2024] [Accepted: 06/23/2024] [Indexed: 06/29/2024] Open
Abstract
Ghrelin is a stomach-derived hormone that increases feeding and is elevated in response to chronic psychosocial stressors. The effects of ghrelin on feeding are mediated by the binding of ghrelin to the growth hormone secretagogue receptor (GHSR), a receptor located in hypothalamic and extrahypothalamic regions important for regulating food intake and metabolic rate. The ability of ghrelin to enter the brain, however, seems to be restricted to circumventricular organs like the median eminence and the brainstem area postrema, whereas ghrelin does not readily enter other GHSR-expressing regions like the ventral tegmental area (VTA). Interestingly, social stressors result in increased blood-brain barrier permeability, and this could therefore facilitate the entry of ghrelin into the brain. To investigate this, we exposed mice to social defeat stress for 21 d and then peripherally injected a Cy5-labelled biologically active ghrelin analog. The results demonstrate that chronically stressed mice exhibit higher Cy5-ghrelin fluorescence in several hypothalamic regions in addition to the ARC, including the hippocampus and midbrain. Furthermore, Cy5-ghrelin injections resulted in increased FOS expression in regions associated with the reward system in chronically stressed mice. Further histologic analyses identified a reduction in the branching of hypothalamic astrocytes in the ARC-median eminence junction, suggesting increased blood-brain barrier permeability. These data support the hypothesis that during metabolically challenging conditions like chronic stress, ghrelin may be more able to cross the blood-brain barrier and diffuse throughout the brain to target GHSR-expressing brain regions away from circumventricular organs.
Collapse
Affiliation(s)
- Andrea Smith
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Brenna MacAulay
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Jessica Scheufen
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Abagael Hudak
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Alfonso Abizaid
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S5B6, Canada
| |
Collapse
|
19
|
Lee S, Cai WT, Yoon HS, Kim JH. Visualization of the existence of growth hormone secretagogue receptor in the rat nucleus accumbens. Mol Brain 2024; 17:37. [PMID: 38872222 PMCID: PMC11177452 DOI: 10.1186/s13041-024-01109-2] [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: 05/09/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024] Open
Abstract
The potential role of the ghrelin receptor, also known as the growth hormone secretagogue receptor (GHSR), within the nucleus accumbens (NAcc) in regulating drug addiction and feeding has been documented; however, the pattern of its expression in this site remains elusive. In this study, we characterized the expression patterns of GHSR1a and 1b, two subtypes of GHSRs, within the NAcc of the rat brain by immunohistochemistry. We visually detected GHSR signals, for the first time, at the protein level in the NAcc in which they were mostly expressed in neurons including both medium spiny neurons (MSNs) and non-MSNs. Furthermore, GHSR1a was found expressed as localized near the cellular membrane or some in the cytoplasm, whereas GHSR1b expressed solely throughout the large cytoplasmic area. The existence and subcellular expression pattern of GHSRs in the NAcc identified in this study will contribute to improving our understanding about the role of GHSR-mediated neurosignaling in feeding and drug addiction.
Collapse
Affiliation(s)
- Seohyeon Lee
- Department of Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Wen Ting Cai
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Hyung Shin Yoon
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Jeong-Hoon Kim
- Department of Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, South Korea.
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
20
|
Zhang L, Liu J, Gao D, Li D. Role of ghrelin in promoting catch-up growth and maintaining metabolic homeostasis in small-for-gestational-age infants. Front Pediatr 2024; 12:1395571. [PMID: 38903769 PMCID: PMC11187245 DOI: 10.3389/fped.2024.1395571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/27/2024] [Indexed: 06/22/2024] Open
Abstract
Small-for-gestational age (SGA) has been a great concern in the perinatal period as it leads to adverse perinatal outcomes and increased neonatal morbidity and mortality, has an impact on long-term health outcomes, and increases the risk of metabolic disorders, cardiovascular, and endocrine diseases in adulthood. As an endogenous ligand of the growth hormone secretagotor (GHS-R), ghrelin may play an important role in regulating growth and energy metabolic homeostasis from fetal to adult life. We reviewed the role of ghrelin in catch-up growth and energy metabolism of SGA in recent years. In addition to promoting SGA catch-up growth, ghrelin may also participate in SGA energy metabolism and maintain metabolic homeostasis. The causes of small gestational age infants are very complex and may be related to a variety of metabolic pathway disorders. The related signaling pathways regulated by ghrelin may help to identify high-risk groups of SGA metabolic disorders and formulate targeted interventions to prevent the occurrence of adult dwarfism, insulin resistance-related metabolic syndrome and other diseases.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pediatrics, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jingfei Liu
- Department of Neonatology, Dalian Women and Children’s Medical Group, Dalian, China
| | - Dianyong Gao
- Department of Orthopedics, Lushunkou District People’s Hospital, Dalian, China
| | - Dong Li
- Department of Neonatology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
21
|
Cornejo MP, Fernandez G, Cabral A, Barrile F, Heredia F, García Romero G, Zubimendi Sampieri JP, Quelas JI, Cantel S, Fehrentz JA, Alonso A, Pla R, Ferran JL, Andreoli MF, De Francesco PN, Perelló M. GHSR in a Subset of GABA Neurons Controls Food Deprivation-Induced Hyperphagia in Male Mice. Endocrinology 2024; 165:bqae061. [PMID: 38815068 DOI: 10.1210/endocr/bqae061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
The growth hormone secretagogue receptor (GHSR), primarily known as the receptor for the hunger hormone ghrelin, potently controls food intake, yet the specific Ghsr-expressing cells mediating the orexigenic effects of this receptor remain incompletely characterized. Since Ghsr is expressed in gamma-aminobutyric acid (GABA)-producing neurons, we sought to investigate whether the selective expression of Ghsr in a subset of GABA neurons is sufficient to mediate GHSR's effects on feeding. First, we crossed mice that express a tamoxifen-dependent Cre recombinase in the subset of GABA neurons that express glutamic acid decarboxylase 2 (Gad2) enzyme (Gad2-CreER mice) with reporter mice, and found that ghrelin mainly targets a subset of Gad2-expressing neurons located in the hypothalamic arcuate nucleus (ARH) and that is predominantly segregated from Agouti-related protein (AgRP)-expressing neurons. Analysis of various single-cell RNA-sequencing datasets further corroborated that the primary subset of cells coexpressing Gad2 and Ghsr in the mouse brain are non-AgRP ARH neurons. Next, we crossed Gad2-CreER mice with reactivable GHSR-deficient mice to generate mice expressing Ghsr only in Gad2-expressing neurons (Gad2-GHSR mice). We found that ghrelin treatment induced the expression of the marker of transcriptional activation c-Fos in the ARH of Gad2-GHSR mice, yet failed to induce food intake. In contrast, food deprivation-induced refeeding was higher in Gad2-GHSR mice than in GHSR-deficient mice and similar to wild-type mice, suggesting that ghrelin-independent roles of GHSR in a subset of GABA neurons is sufficient for eliciting full compensatory hyperphagia in mice.
Collapse
Affiliation(s)
- María Paula Cornejo
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Gimena Fernandez
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Agustina Cabral
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Franco Barrile
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Florencia Heredia
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Guadalupe García Romero
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | | | | | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Antonia Alonso
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia 30100, Spain
- Institute of Biomedical Research of Murcia-IMIB, Virgen de la Arrixaca University Hospital, Murcia 30100, Spain
| | - Ramon Pla
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia 30100, Spain
- Institute of Biomedical Research of Murcia-IMIB, Virgen de la Arrixaca University Hospital, Murcia 30100, Spain
| | - José Luis Ferran
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia 30100, Spain
- Institute of Biomedical Research of Murcia-IMIB, Virgen de la Arrixaca University Hospital, Murcia 30100, Spain
| | - María Florencia Andreoli
- Instituto de Desarrollo e Investigaciones Pediátricas (IDIP), HIAEP Sor María Ludovica de La Plata, Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala 751 24, Sweden
| | - Pablo Nicolas De Francesco
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Mario Perelló
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala 751 24, Sweden
| |
Collapse
|
22
|
Lékó AH, Gregory-Flores A, Marchette RCN, Gomez JL, Vendruscolo JCM, Repunte-Canonigo V, Choung V, Deschaine SL, Whiting KE, Jackson SN, Cornejo MP, Perello M, You ZB, Eckhaus M, Rasineni K, Janda KD, Zorman B, Sumazin P, Koob GF, Michaelides M, Sanna PP, Vendruscolo LF, Leggio L. Genetic or pharmacological GHSR blockade has sexually dimorphic effects in rodents on a high-fat diet. Commun Biol 2024; 7:632. [PMID: 38796563 PMCID: PMC11127961 DOI: 10.1038/s42003-024-06303-5] [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: 09/29/2023] [Accepted: 05/08/2024] [Indexed: 05/28/2024] Open
Abstract
The stomach-derived hormone ghrelin regulates essential physiological functions. The ghrelin receptor (GHSR) has ligand-independent actions; therefore, GHSR gene deletion may be a reasonable approach to investigate the role of this system in feeding behaviors and diet-induced obesity (DIO). Here, we investigate the effects of a long-term (12-month) high-fat (HFD) versus regular diet on obesity-related measures in global GHSR-KO and wild-type (WT) Wistar male and female rats. Our main findings are that the GHSR gene deletion protects against DIO and decreases food intake during HFD in male but not in female rats. GHSR gene deletion increases thermogenesis and brain glucose uptake in male rats and modifies the effects of HFD on brain glucose metabolism in a sex-specific manner, as assessed with small animal positron emission tomography. We use RNA-sequencing to show that GHSR-KO rats have upregulated expression of genes responsible for fat oxidation in brown adipose tissue. Central administration of a novel GHSR inverse agonist, PF-5190457, attenuates ghrelin-induced food intake, but only in male, not in female mice. HFD-induced binge-like eating is reduced by inverse agonism in both sexes. Our results support GHSR as a promising target for new pharmacotherapies for obesity.
Collapse
Affiliation(s)
- András H Lékó
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
- Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Adriana Gregory-Flores
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Renata C N Marchette
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Juan L Gomez
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Janaina C M Vendruscolo
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Vez Repunte-Canonigo
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Vicky Choung
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Sara L Deschaine
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
| | - Kimberly E Whiting
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Shelley N Jackson
- Translational Analytical Core, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Maria Paula Cornejo
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata, Argentina
| | - Mario Perello
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata, Argentina
| | - Zhi-Bing You
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Michael Eckhaus
- Pathology Service, Division of Veterinary Resources, Office of Research Services, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Karuna Rasineni
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kim D Janda
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Barry Zorman
- Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Pavel Sumazin
- Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - George F Koob
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Pietro P Sanna
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Leandro F Vendruscolo
- Stress and Addiction Neuroscience Unit, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA.
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA.
- Translational Analytical Core, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI, USA.
- Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
- Department of Neuroscience, Georgetown University Medical Center, Washington DC, USA.
| |
Collapse
|
23
|
Gupta D, Burstein AW, Shankar K, Varshney S, Singh O, Osborne-Lawrence S, Richard CP, Zigman JM. Impact of Ghrelin on Islet Size in Nonpregnant and Pregnant Female Mice. Endocrinology 2024; 165:bqae048. [PMID: 38626085 PMCID: PMC11075791 DOI: 10.1210/endocr/bqae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/03/2024] [Accepted: 04/12/2024] [Indexed: 04/18/2024]
Abstract
Reducing ghrelin by ghrelin gene knockout (GKO), ghrelin-cell ablation, or high-fat diet feeding increases islet size and β-cell mass in male mice. Here we determined if reducing ghrelin also enlarges islets in females and if pregnancy-associated changes in islet size are related to reduced ghrelin. Islet size and β-cell mass were larger (P = .057 for β-cell mass) in female GKO mice. Pregnancy was associated with reduced ghrelin and increased liver-expressed antimicrobial peptide-2 (LEAP2; a ghrelin receptor antagonist) in wild-type mice. Ghrelin deletion and pregnancy each increased islet size (by ∼19.9-30.2% and ∼34.9-46.4%, respectively), percentage of large islets (>25 µm2×103, by ∼21.8-42% and ∼21.2-41.2%, respectively), and β-cell mass (by ∼15.7-23.8% and ∼65.2-76.8%, respectively). Neither islet cross-sectional area, β-cell cross-sectional area, nor β-cell mass correlated with plasma ghrelin, although all positively correlated with LEAP2 (P = .081 for islet cross-sectional area). In ad lib-fed mice, there was an effect of pregnancy, but not ghrelin deletion, to change (raise) plasma insulin without impacting blood glucose. Similarly, there was an effect of pregnancy, but not ghrelin deletion, to change (lower) blood glucose area under the curve during a glucose tolerance test. Thus, genetic deletion of ghrelin increases islet size and β-cell cross-sectional area in female mice, similar to males. Yet, despite pregnancy-associated reductions in ghrelin, other factors appear to govern islet enlargement and changes to insulin sensitivity and glucose tolerance in the setting of pregnancy. In the case of islet size and β-cell mass, one of those factors may be the pregnancy-associated increase in LEAP2.
Collapse
Affiliation(s)
- Deepali Gupta
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Avi W Burstein
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kripa Shankar
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Salil Varshney
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Omprakash Singh
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sherri Osborne-Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Corine P Richard
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Division of Endocrinology & Metabolism, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
24
|
Nie M, Zhang J, Bal M, Duran C, An SW, Zigman JM, Baum M, Hiremath C, Marciano DK, Wolf MTF. Ghrelin enhances tubular magnesium absorption in the kidney. Front Physiol 2024; 15:1363708. [PMID: 38638279 PMCID: PMC11024433 DOI: 10.3389/fphys.2024.1363708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/07/2024] [Indexed: 04/20/2024] Open
Abstract
Osteoporosis after bariatric surgery is an increasing health concern as the rate of bariatric surgery has risen. In animal studies mimicking bariatric procedures, bone disease, together with decreased serum levels of Ca2+, Mg2+ and the gastric hormone Ghrelin were described. Ghrelin regulates metabolism by binding to and activating the growth hormone secretagogue receptor (GHSR) which is also expressed in the kidney. As calcium and magnesium are key components of bone, we tested the hypothesis that Ghrelin-deficiency contributes to osteoporosis via reduced upregulation of the renal calcium channel TRPV5 and the heteromeric magnesium channel TRPM6/7. We expressed GHSR with TRPV5 or TRPM6/7 channel in HEK293 cells and treated them with purified Ghrelin. Whole-cell current density was analyzed by patch-clamp recording. Nephron-specific gene expression was performed by tubular microdissection followed by qPCR in wild-type (WT) mice, and immunofluorescent imaging of GHSR-eGFP mice. Tubular magnesium homeostasis was analyzed in GHSR-null and WT mice at baseline and after caloric restriction. After Ghrelin exposure, whole-cell current density did not change for TRPV5 but increased for TRPM6/7 in a dose-dependent fashion. Applying the Ghrelin-mimetic (D-Trp7, Ala8,D-Phe10)-α-MSH (6-11) amide without and with the GHSR antagonist (D-Lys3)-GHRP6, we confirmed the stimulatory role of Ghrelin towards TRPM6/7. As GHSR initiates downstream signaling via protein kinase A (PKA), we found that the PKA inhibitor H89 abrogated TRPM6/7 stimulation by Ghrelin. Similarly, transfected Gαs, but not the Gαs mutant Q227L, nor Gαi2, Gαq, or Gα13 upregulated TRPM6/7 current density. In microdissected TALs and DCTs similar levels of GHSR mRNA were detected. In contrast, TRPM6 mRNA was expressed in the DCT and also detected in the TAL at 25% expression compared to DCT. Immunofluorescent studies using reporter GHSR-eGFP mice showed a strong eGFP signal in the TAL but surprisingly displayed no eGFP signal in the DCT. In 3-, 6-, and 9-month-old GHSR-null and WT mice, baseline serum magnesium was not significantly different, but 24-h urinary magnesium excretion was elevated in 9-month-old GHSR-null mice. In calorically restricted GHSR-null mice, we detected excess urinary magnesium excretion and reduced serum magnesium levels compared to WT mice. The kidneys from calorically restricted WT mice showed upregulated gene expression of magnesiotropic genes Hnf1b, Cldn-16, Cldn-19, Fxyd-2b, and Parvalbumin compared to GHSR-null mice. Our in vitro studies show that Ghrelin stimulates TRPM6/7 via GHSR and Gαs-PKA signaling. The murine studies are consistent with Ghrelin-GHSR signaling inducing reduced urinary magnesium excretion, particularly in calorically restricted mice when Ghrelin levels are elevated. This effect may be mediated by Ghrelin-upregulation of TRPM6 in the TAL and/or upregulation of other magnesiotropic genes. We postulate that rising Ghrelin levels with hunger contribute to increased renal Mg2+ reabsorption to compensate for lack of enteral Mg2+ uptake.
Collapse
Affiliation(s)
- Mingzhu Nie
- Pediatric Nephrology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jing Zhang
- Pediatric Nephrology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Manjot Bal
- Pediatric Nephrology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Claudia Duran
- Pediatric Nephrology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Sung Wan An
- Pediatric Nephrology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Pediatric Nephrology, Department of Pediatrics, University of Michigan, Ann Arbor, MI, United States
| | - Jeffrey M. Zigman
- Department of Internal Medicine, Center for Hypothalamic Research, UTSW Medical Center, Dallas, TX, United States
| | - Michel Baum
- Pediatric Nephrology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Chitkale Hiremath
- Department of Internal Medicine, Nephrology, and Department of Cell Biology, UTSW Medical Center, Dallas, TX, United States
| | - Denise K. Marciano
- Department of Internal Medicine, Nephrology, and Department of Cell Biology, UTSW Medical Center, Dallas, TX, United States
| | - Matthias T. F. Wolf
- Pediatric Nephrology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Pediatric Nephrology, Department of Pediatrics, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
25
|
Kulkarni SS, Singh O, Zigman JM. The intersection between ghrelin, metabolism and circadian rhythms. Nat Rev Endocrinol 2024; 20:228-238. [PMID: 38123819 PMCID: PMC11760189 DOI: 10.1038/s41574-023-00927-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2023] [Indexed: 12/23/2023]
Abstract
Despite the growing popular interest in sleep and diet, many gaps exist in our scientific understanding of the interaction between circadian rhythms and metabolism. In this Review, we explore a promising, bidirectional role for ghrelin in mediating this interaction. Ghrelin both influences and is influenced by central and peripheral circadian systems. Specifically, we focus on how ghrelin impacts outputs of circadian rhythm, including neuronal activity, circulating growth hormone levels, locomotor activity and eating behaviour. We also consider the effects of circadian rhythms on ghrelin expression and the consequences of disrupted circadian patterns, such as shift work and jet lag, on ghrelin secretion. Our Review is aimed at both the casual reader interested in gaining more insight into the scientific context surrounding the trending topics of sleep and metabolism, as well as experienced scientists in the fields of ghrelin and circadian biology seeking inspiration and a comprehensive overview of how these fields are related.
Collapse
Affiliation(s)
- Soumya S Kulkarni
- Medical Scientist Training Program, UT Southwestern Medical Center, Dallas, TX, USA
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Omprakash Singh
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.
- Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
26
|
Skowronski AA, Leibel RL, LeDuc CA. Neurodevelopmental Programming of Adiposity: Contributions to Obesity Risk. Endocr Rev 2024; 45:253-280. [PMID: 37971140 PMCID: PMC10911958 DOI: 10.1210/endrev/bnad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 11/19/2023]
Abstract
This review analyzes the published evidence regarding maternal factors that influence the developmental programming of long-term adiposity in humans and animals via the central nervous system (CNS). We describe the physiological outcomes of perinatal underfeeding and overfeeding and explore potential mechanisms that may mediate the impact of such exposures on the development of feeding circuits within the CNS-including the influences of metabolic hormones and epigenetic changes. The perinatal environment, reflective of maternal nutritional status, contributes to the programming of offspring adiposity. The in utero and early postnatal periods represent critically sensitive developmental windows during which the hormonal and metabolic milieu affects the maturation of the hypothalamus. Maternal hyperglycemia is associated with increased transfer of glucose to the fetus driving fetal hyperinsulinemia. Elevated fetal insulin causes increased adiposity and consequently higher fetal circulating leptin concentration. Mechanistic studies in animal models indicate important roles of leptin and insulin in central and peripheral programming of adiposity, and suggest that optimal concentrations of these hormones are critical during early life. Additionally, the environmental milieu during development may be conveyed to progeny through epigenetic marks and these can potentially be vertically transmitted to subsequent generations. Thus, nutritional and metabolic/endocrine signals during perinatal development can have lifelong (and possibly multigenerational) impacts on offspring body weight regulation.
Collapse
Affiliation(s)
- Alicja A Skowronski
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rudolph L Leibel
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Charles A LeDuc
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
27
|
Wang H, Shen Z, Wu CS, Ji P, Noh JY, Geoffroy CG, Kim S, Threadgill D, Li J, Zhou Y, Xiao X, Zheng H, Sun Y. Neuronal ablation of GHSR mitigates diet-induced depression and memory impairment via AMPK-autophagy signaling-mediated inflammation. Front Immunol 2024; 15:1339937. [PMID: 38464534 PMCID: PMC10920242 DOI: 10.3389/fimmu.2024.1339937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/06/2024] [Indexed: 03/12/2024] Open
Abstract
Obesity is associated with chronic inflammation in the central nervous system (CNS), and neuroinflammation has been shown to have detrimental effects on mood and cognition. The growth hormone secretagogue receptor (GHSR), the biologically relevant receptor of the orexigenic hormone ghrelin, is primarily expressed in the brain. Our previous study showed that neuronal GHSR deletion prevents high-fat diet-induced obesity (DIO). Here, we investigated the effect of neuronal GHSR deletion on emotional and cognitive functions in DIO. The neuron-specific GHSR-deficient mice exhibited reduced depression and improved spatial memory compared to littermate controls under DIO. We further examined the cortex and hippocampus, the major regions regulating cognitive and emotional behaviors, and found that the neuronal deletion of GHSR reduced DIO-induced neuroinflammation by suppressing proinflammatory chemokines/cytokines and decreasing microglial activation. Furthermore, our data showed that neuronal GHSR deletion suppresses neuroinflammation by downregulating AMPK-autophagy signaling in neurons. In conclusion, our data reveal that neuronal GHSR inhibition protects against DIO-induced depressive-like behavior and spatial cognitive dysfunction, at least in part, through AMPK-autophagy signaling-mediated neuroinflammation.
Collapse
Affiliation(s)
- Hongying Wang
- Department of Nutrition, Texas A&M University, College Station, TX, United States
- Department of Endocrinology, Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zheng Shen
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Chia-Shan Wu
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Pengfei Ji
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Ji Yeon Noh
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Cédric G. Geoffroy
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University, College Station, TX, United States
| | - Sunja Kim
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, TX, United States
| | - David Threadgill
- Department of Nutrition, Texas A&M University, College Station, TX, United States
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, TX, United States
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, United States
| | - Jianrong Li
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States
| | - Yu Zhou
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Xiaoqiu Xiao
- Department of Endocrinology, Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, United States
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| |
Collapse
|
28
|
Athar F, Karmani M, Templeman N. Metabolic hormones are integral regulators of female reproductive health and function. Biosci Rep 2024; 44:BSR20231916. [PMID: 38131197 PMCID: PMC10830447 DOI: 10.1042/bsr20231916] [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: 11/03/2023] [Revised: 11/29/2023] [Accepted: 12/21/2023] [Indexed: 12/23/2023] Open
Abstract
The female reproductive system is strongly influenced by nutrition and energy balance. It is well known that food restriction or energy depletion can induce suppression of reproductive processes, while overnutrition is associated with reproductive dysfunction. However, the intricate mechanisms through which nutritional inputs and metabolic health are integrated into the coordination of reproduction are still being defined. In this review, we describe evidence for essential contributions by hormones that are responsive to food intake or fuel stores. Key metabolic hormones-including insulin, the incretins (glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1), growth hormone, ghrelin, leptin, and adiponectin-signal throughout the hypothalamic-pituitary-gonadal axis to support or suppress reproduction. We synthesize current knowledge on how these multifaceted hormones interact with the brain, pituitary, and ovaries to regulate functioning of the female reproductive system, incorporating in vitro and in vivo data from animal models and humans. Metabolic hormones are involved in orchestrating reproductive processes in healthy states, but some also play a significant role in the pathophysiology or treatment strategies of female reproductive disorders. Further understanding of the complex interrelationships between metabolic health and female reproductive function has important implications for improving women's health overall.
Collapse
Affiliation(s)
- Faria Athar
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Muskan Karmani
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Nicole M. Templeman
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| |
Collapse
|
29
|
Wee RWS, Mishchanchuk K, AlSubaie R, Church TW, Gold MG, MacAskill AF. Internal-state-dependent control of feeding behavior via hippocampal ghrelin signaling. Neuron 2024; 112:288-305.e7. [PMID: 37977151 DOI: 10.1016/j.neuron.2023.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/13/2023] [Accepted: 10/12/2023] [Indexed: 11/19/2023]
Abstract
Hunger is an internal state that not only invigorates feeding but also acts as a contextual cue for higher-order control of anticipatory feeding-related behavior. The ventral hippocampus is crucial for differentiating optimal behavior across contexts, but how internal contexts such as hunger influence hippocampal circuitry is unknown. In this study, we investigated the role of the ventral hippocampus during feeding behavior across different states of hunger in mice. We found that activity of a unique subpopulation of neurons that project to the nucleus accumbens (vS-NAc neurons) increased when animals investigated food, and this activity inhibited the transition to begin eating. Increases in the level of the peripheral hunger hormone ghrelin reduced vS-NAc activity during this anticipatory phase of feeding via ghrelin-receptor-dependent increases in postsynaptic inhibition and promoted the initiation of eating. Together, these experiments define a ghrelin-sensitive hippocampal circuit that informs the decision to eat based on internal state.
Collapse
Affiliation(s)
- Ryan W S Wee
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Karyna Mishchanchuk
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Rawan AlSubaie
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Timothy W Church
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Matthew G Gold
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Andrew F MacAskill
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK.
| |
Collapse
|
30
|
Kim DM, Lee JH, Pan Q, Han HW, Shen Z, Eshghjoo S, Wu CS, Yang W, Noh JY, Threadgill DW, Guo S, Wright G, Alaniz R, Sun Y. Nutrient-sensing growth hormone secretagogue receptor in macrophage programming and meta-inflammation. Mol Metab 2024; 79:101852. [PMID: 38092245 PMCID: PMC10772824 DOI: 10.1016/j.molmet.2023.101852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/03/2023] [Accepted: 12/08/2023] [Indexed: 12/20/2023] Open
Abstract
OBJECTIVE Obesity-associated chronic inflammation, aka meta-inflammation, is a key pathogenic driver for obesity-associated comorbidity. Growth hormone secretagogue receptor (GHSR) is known to mediate the effects of nutrient-sensing hormone ghrelin in food intake and fat deposition. We previously reported that global Ghsr ablation protects against diet-induced inflammation and insulin resistance, but the site(s) of action and mechanism are unknown. Macrophages are key drivers of meta-inflammation. To unravel the role of GHSR in macrophages, we generated myeloid-specific Ghsr knockout mice (LysM-Cre;Ghsrf/f). METHODS LysM-Cre;Ghsrf/f and control Ghsrf/f mice were subjected to 5 months of high-fat diet (HFD) feeding to induce obesity. In vivo, metabolic profiling of food intake, physical activity, and energy expenditure, as well as glucose and insulin tolerance tests (GTT and ITT) were performed. At termination, peritoneal macrophages (PMs), epididymal white adipose tissue (eWAT), and liver were analyzed by flow cytometry and histology. For ex vivo studies, bone marrow-derived macrophages (BMDMs) were generated from the mice and treated with palmitic acid (PA) or lipopolysaccharide (LPS). For in vitro studies, macrophage RAW264.7 cells with Ghsr overexpression or Insulin receptor substrate 2 (Irs2) knockdown were studied. RESULTS We found that Ghsr expression in PMs was increased under HFD feeding. In vivo, HFD-fed LysM-Cre;Ghsrf/f mice exhibited significantly attenuated systemic inflammation and insulin resistance without affecting food intake or body weight. Tissue analysis showed that HFD-fed LysM-Cre;Ghsrf/f mice have significantly decreased monocyte/macrophage infiltration, pro-inflammatory activation, and lipid accumulation, showing elevated lipid-associated macrophages (LAMs) in eWAT and liver. Ex vivo, Ghsr-deficient macrophages protected against PA- or LPS-induced pro-inflammatory polarization, showing reduced glycolysis, increased fatty acid oxidation, and decreased NF-κB nuclear translocation. At molecular level, GHSR metabolically programs macrophage polarization through PKA-CREB-IRS2-AKT2 signaling pathway. CONCLUSIONS These novel results demonstrate that macrophage GHSR plays a key role in the pathogenesis of meta-inflammation, and macrophage GHSR promotes macrophage infiltration and induces pro-inflammatory polarization. These exciting findings suggest that GHSR may serve as a novel immunotherapeutic target for the treatment of obesity and its associated comorbidity.
Collapse
Affiliation(s)
- Da Mi Kim
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Jong Han Lee
- Department of Marine Bioindustry, Hanseo University, Seosan 31962, South Korea; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA
| | - Quan Pan
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Hye Won Han
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Zheng Shen
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Sahar Eshghjoo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Agilent technologies, Aanta Clara, CA 95051, USA
| | - Chia-Shan Wu
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA
| | - Wanbao Yang
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Ji Yeon Noh
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - David W Threadgill
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; Texas A&M Institute for Genome Sciences and Society, Department of Cell Biology and Genetics, Texas A&M University, College Station, TX 77843, USA
| | - Shaodong Guo
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Gus Wright
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 77843, USA
| | - Robert Alaniz
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Tlaloc Therapeutics Inc., College Station, TX 77845, USA
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA.
| |
Collapse
|
31
|
Singh O, Ogden SB, Varshney S, Shankar K, Gupta D, Paul S, Osborne-Lawrence S, Richard CP, Metzger NP, Lawrence C, Leon Mercado L, Zigman JM. Ghrelin-responsive mediobasal hypothalamic neurons mediate exercise-associated food intake and exercise endurance. JCI Insight 2023; 8:e172549. [PMID: 37962950 PMCID: PMC10807726 DOI: 10.1172/jci.insight.172549] [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: 05/24/2023] [Accepted: 11/08/2023] [Indexed: 11/16/2023] Open
Abstract
Previous studies have implicated the orexigenic hormone ghrelin as a mediator of exercise endurance and the feeding response postexercise. Specifically, plasma ghrelin levels nearly double in mice when they are subjected to an hour-long bout of high-intensity interval exercise (HIIE) using treadmills. Also, growth hormone secretagogue receptor-null (GHSR-null) mice exhibit decreased food intake following HIIE and diminished running distance (time until exhaustion) during a longer, stepwise exercise endurance protocol. To investigate whether ghrelin-responsive mediobasal hypothalamus (MBH) neurons mediate these effects, we stereotaxically delivered the inhibitory designer receptor exclusively activated by designer drugs virus AAV2-hSyn-DIO-hM4(Gi)-mCherry to the MBH of Ghsr-IRES-Cre mice, which express Cre recombinase directed by the Ghsr promoter. We found that chemogenetic inhibition of GHSR-expressing MBH neurons (upon delivery of clozapine-N-oxide) 1) suppressed food intake following HIIE, 2) reduced maximum running distance and raised blood glucose and blood lactate levels during an exercise endurance protocol, 3) reduced food intake following ghrelin administration, and 4) did not affect glucose tolerance. Further, HIIE increased MBH Ghsr expression. These results indicate that activation of ghrelin-responsive MBH neurons is required for the normal feeding response to HIIE and the usual amount of running exhibited during an exercise endurance protocol.
Collapse
Affiliation(s)
- Omprakash Singh
- Center for Hypothalamic Research, Department of Internal Medicine
| | - Sean B. Ogden
- Center for Hypothalamic Research, Department of Internal Medicine
| | - Salil Varshney
- Center for Hypothalamic Research, Department of Internal Medicine
| | - Kripa Shankar
- Center for Hypothalamic Research, Department of Internal Medicine
| | - Deepali Gupta
- Center for Hypothalamic Research, Department of Internal Medicine
| | - Subhojit Paul
- Center for Hypothalamic Research, Department of Internal Medicine
| | | | | | | | - Connor Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine
| | | | - Jeffrey M. Zigman
- Center for Hypothalamic Research, Department of Internal Medicine
- Division of Endocrinology & Metabolism, Department of Internal Medicine; and
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
32
|
Tatum SM, Holland WL. Germline and conditional ghrelin knockout increases islet size. J Clin Invest 2023; 133:e175799. [PMID: 38099493 PMCID: PMC10721140 DOI: 10.1172/jci175799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
Conflicting studies in recent years report that genetic or pharmacological increases or decreases in ghrelin either increase or have no effect on islet size. In this issue of the JCI, Gupta, Burstein, and colleagues applied a rigorous approach to determine the effects of reducing ghrelin on islet size in germline and conditional ghrelin-knockout mice as well as across varying ages and weight. Both germline and conditional ghrelin-knockout mice associated with increased islet size, which was further exacerbated by older age and diet-induced obesity. These findings suggest that modulation of ghrelin may open a therapeutic window to prevent or treat diabetes.
Collapse
|
33
|
Gupta D, Burstein AW, Schwalbe DC, Shankar K, Varshney S, Singh O, Paul S, Ogden SB, Osborne-Lawrence S, Metzger NP, Richard CP, Campbell JN, Zigman JM. Ghrelin deletion and conditional ghrelin cell ablation increase pancreatic islet size in mice. J Clin Invest 2023; 133:e169349. [PMID: 38099492 PMCID: PMC10721155 DOI: 10.1172/jci169349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 10/05/2023] [Indexed: 12/18/2023] Open
Abstract
Ghrelin exerts key effects on islet hormone secretion to regulate blood glucose levels. Here, we sought to determine whether ghrelin's effects on islets extend to the alteration of islet size and β cell mass. We demonstrate that reducing ghrelin - by ghrelin gene knockout (GKO), conditional ghrelin cell ablation, or high-fat diet (HFD) feeding - was associated with increased mean islet size (up to 62%), percentage of large islets (up to 854%), and β cell cross-sectional area (up to 51%). In GKO mice, these effects were more apparent in 10- to 12-week-old mice than in 4-week-old mice. Higher β cell numbers from decreased β cell apoptosis drove the increase in β cell cross-sectional area. Conditional ghrelin cell ablation in adult mice increased the β cell number per islet by 40% within 4 weeks. A negative correlation between islet size and plasma ghrelin in HFD-fed plus chow-fed WT mice, together with even larger islet sizes in HFD-fed GKO mice than in HFD-fed WT mice, suggests that reduced ghrelin was not solely responsible for diet-induced obesity-associated islet enlargement. Single-cell transcriptomics revealed changes in gene expression in several GKO islet cell types, including upregulation of Manf, Dnajc3, and Gnas expression in β cells, which supports decreased β cell apoptosis and/or increased β cell proliferation. These effects of ghrelin reduction on islet morphology might prove useful when designing new therapies for diabetes.
Collapse
Affiliation(s)
- Deepali Gupta
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Avi W. Burstein
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Dana C. Schwalbe
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Kripa Shankar
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Salil Varshney
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Omprakash Singh
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Subhojit Paul
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Sean B. Ogden
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Sherri Osborne-Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Nathan P. Metzger
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Corine P. Richard
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - John N. Campbell
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Jeffrey M. Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
- Division of Endocrinology and Metabolism, Department of Internal Medicine and
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
34
|
Ibrahim M, Khalife L, Abdel-Latif R, Faour WH. Ghrelin hormone a new molecular modulator between obesity and glomerular damage. Mol Biol Rep 2023; 50:10525-10533. [PMID: 37924451 DOI: 10.1007/s11033-023-08866-8] [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: 06/30/2023] [Accepted: 09/27/2023] [Indexed: 11/06/2023]
Abstract
The incidence of glomerular diseases is increasing worldwide due to increased prevalence of obesity which is a major risk factor for type-2 diabetes mellitus and cardiovascular disorders.Ghrelin, an orexigenic peptide hormone, has been implicated in obesity, and its impact on the pathology and function of the kidneys was found to be significant. Ghrelin known to regulate energy homeostasis and growth hormone release, has been shown to modulate critical signaling pathways involved in the health and survival of podocytes. These derangements directly affect glomerular function and manifest as impaired glomerular filtration barrier and leakage of albumin into urine. Although the pathological features of the above-mentioned disorders are different, they interestingly lead to similar clinical features of glomerular damage. The pathological events are majorly initiated by endocrine imbalance leading to abnormal activation of downstream signaling pathways involved in the development of glomerulosclerosis. In fact, obesity increases the risk of developing chronic kidney disease by altering the secretion of pro-inflammatory cytokines and adipokines, activating the renin-angiotensin-aldosterone system (RAAS), promoting lipotoxicity, oxidative stress and fibrosis within the kidneys. Whilst these bioregulators are well described, their direct involvement in renal homeostasis is still mostly elusive. This review summarized previous and recent evidence on the endocrine properties of ghrelin and perivascular adipose tissue involved in modulating kidney physiology.
Collapse
Affiliation(s)
- Maroun Ibrahim
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Lynn Khalife
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Rania Abdel-Latif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Miniya, Egypt
| | - Wissam H Faour
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon.
| |
Collapse
|
35
|
So WL, Hu J, Jeffs L, Dempsey H, Lockie SH, Zigman JM, Stark R, Reichenbach A, Andrews ZB. Ghrelin signalling in AgRP neurons links metabolic state to the sensory regulation of AgRP neural activity. Mol Metab 2023; 78:101826. [PMID: 37898450 PMCID: PMC10643323 DOI: 10.1016/j.molmet.2023.101826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/30/2023] Open
Abstract
OBJECTIVE The sensory detection of food and food cues suppresses Agouti related peptide (AgRP) neuronal activity prior to consumption with greatest suppression occurring in response to highly caloric food or interoceptive energy need. However, the interoceptive mechanisms priming an appropriate AgRP neural response to external sensory information of food availability remain unexplored. Since hunger increases plasma ghrelin, we hypothesized that ghrelin receptor (GHSR) signalling on AgRP neurons is a key interoceptive mechanism integrating energy need with external sensory cues predicting caloric availability. METHODS We used in vivo photometry to measure the effects of ghrelin administration or fasting on AgRP neural activity with GCaMP6s and dopamine release in the nucleus accumbens with GRAB-DA in mice lacking ghrelin receptors in AgRP neurons. RESULTS The deletion of GHSR on AgRP neurons prevented ghrelin-induced food intake, motivation and AgRP activity. The presentation of food (peanut butter pellet) or a wooden dowel suppressed AgRP activity in fasted WT but not mice lacking GHSRs in AgRP neurons. Similarly, peanut butter and a wooden dowel increased dopamine release in the nucleus accumbens after ip ghrelin injection in WT but not mice lacking GHSRs in AgRP neurons. No difference in dopamine release was observed in fasted mice. Finally, ip ghrelin administration did not directly increase dopamine neural activity in the ventral tegmental area. CONCLUSIONS Our results suggest that AgRP GHSRs integrate an interoceptive state of energy need with external sensory information to produce an optimal change in AgRP neural activity. Thus, ghrelin signalling on AgRP neurons is more than just a feedback signal to increase AgRP activity during hunger.
Collapse
Affiliation(s)
- Wang Lok So
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton 3800, Victoria, Australia
| | - Jiachen Hu
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton 3800, Victoria, Australia
| | - Lotus Jeffs
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton 3800, Victoria, Australia
| | - Harry Dempsey
- The Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, Australia
| | - Sarah H Lockie
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton 3800, Victoria, Australia
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Romana Stark
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton 3800, Victoria, Australia
| | - Alex Reichenbach
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton 3800, Victoria, Australia
| | - Zane B Andrews
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton 3800, Victoria, Australia.
| |
Collapse
|
36
|
Alex Thomas M, Cui X, Artinian LR, Cao Q, Jing J, Silva FC, Wang S, Zigman JM, Sun Y, Shi H, Xue B. Crosstalk between Gut Sensory Ghrelin Signaling and Adipose Tissue Sympathetic Outflow Regulates Metabolic Homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.25.568689. [PMID: 38076894 PMCID: PMC10705268 DOI: 10.1101/2023.11.25.568689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
The stomach-derived orexigenic hormone ghrelin is a key regulator of energy homeostasis and metabolism in humans. The ghrelin receptor, growth hormone secretagogue receptor 1a (GHSR), is widely expressed in the brain and gastrointestinal vagal sensory neurons, and neuronal GHSR knockout results in a profoundly beneficial metabolic profile and protects against diet-induced obesity (DIO) and insulin resistance. Here we show that in addition to the well characterized vagal GHSR, GHSR is robustly expressed in gastrointestinal sensory neurons emanating from spinal dorsal root ganglia. Remarkably, sensory neuron GHSR deletion attenuates DIO through increased energy expenditure and sympathetic outflow to adipose tissue independent of food intake. In addition, neuronal viral tract tracing reveals prominent crosstalk between gut non-vagal sensory afferents and adipose sympathetic outflow. Hence, these findings demonstrate a novel gut sensory ghrelin signaling pathway critical for maintaining energy homeostasis.
Collapse
Affiliation(s)
- M. Alex Thomas
- Department of Biology, Georgia State University, Atlanta, GA
| | - Xin Cui
- Department of Biology, Georgia State University, Atlanta, GA
| | | | - Qiang Cao
- Department of Biology, Georgia State University, Atlanta, GA
| | - Jia Jing
- Department of Biology, Georgia State University, Atlanta, GA
| | - Felipe C. Silva
- Department of Biology, Georgia State University, Atlanta, GA
| | - Shirong Wang
- Department of Biology, Georgia State University, Atlanta, GA
| | - Jeffrey M. Zigman
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Yuxiang Sun
- Department of Nutrition, Texas A & M University, College Station, TX
| | - Hang Shi
- Department of Biology, Georgia State University, Atlanta, GA
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA
| |
Collapse
|
37
|
Willmore L, Minerva AR, Engelhard B, Murugan M, McMannon B, Oak N, Thiberge SY, Peña CJ, Witten IB. Overlapping representations of food and social stimuli in mouse VTA dopamine neurons. Neuron 2023; 111:3541-3553.e8. [PMID: 37657441 PMCID: PMC11672631 DOI: 10.1016/j.neuron.2023.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 05/17/2023] [Accepted: 08/03/2023] [Indexed: 09/03/2023]
Abstract
Dopamine neurons of the ventral tegmental area (VTADA) respond to food and social stimuli and contribute to both forms of motivation. However, it is unclear whether the same or different VTADA neurons encode these different stimuli. To address this question, we performed two-photon calcium imaging in mice presented with food and conspecifics and found statistically significant overlap in the populations responsive to both stimuli. Both hunger and opposite-sex social experience further increased the proportion of neurons that respond to both stimuli, implying that increasing motivation for one stimulus increases overlap. In addition, single-nucleus RNA sequencing revealed significant co-expression of feeding- and social-hormone-related genes in individual VTADA neurons. Taken together, our functional and transcriptional data suggest overlapping VTADA populations underlie food and social motivation.
Collapse
Affiliation(s)
- Lindsay Willmore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Adelaide R Minerva
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Ben Engelhard
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA; Faculty of Medicine, Technion, Haifa 3525433, Israel.
| | - Malavika Murugan
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Brenna McMannon
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Nirja Oak
- Faculty of Medicine, Technion, Haifa 3525433, Israel
| | - Stephan Y Thiberge
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Catherine J Peña
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA.
| | - Ilana B Witten
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
38
|
Maric I, López-Ferreras L, Bhat Y, Asker M, Börchers S, Bellfy L, Byun S, Kwapis JL, Skibicka KP. From the stomach to locus coeruleus: new neural substrate for ghrelin's effects on ingestive, motivated and anxiety-like behaviors. Front Pharmacol 2023; 14:1286805. [PMID: 38026980 PMCID: PMC10679437 DOI: 10.3389/fphar.2023.1286805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Ghrelin, a stomach-derived orexigenic hormone, has a well-established role in energy homeostasis, food reward, and emotionality. Noradrenergic neurons of the locus coeruleus (LC) are known to play an important role in arousal, emotion, cognition, but recently have also been implicated in control of feeding behavior. Ghrelin receptors (the growth hormone secretagogue receptor, GHSR) may be found in the LC, but the behavioral effects of ghrelin signaling in this area are still unexplored. Here, we first determined whether GHSR are present in the rat LC, and demonstrate that GHSR are expressed on noradrenergic neurons in both sexes. We next investigated whether ghrelin controls ingestive and motivated behaviors as well as anxiety-like behavior by acting in the LC. To pursue this idea, we examined the effects of LC GHSR stimulation and blockade on food intake, operant responding for a palatable food reward and, anxiety-like behavior in the open field (OF) and acoustic startle response (ASR) tests in male and female rats. Our results demonstrate that intra-LC ghrelin administration increases chow intake and motivated behavior for sucrose in both sexes. Additionally, females, but not males, exhibited a potent anxiolytic response in the ASR. In order to determine whether activation of GHSR in the LC was necessary for feeding and anxiety behavior control, we utilized liver-expressed antimicrobial peptide 2 (LEAP2), a newly identified endogenous GHSR antagonist. LEAP2 delivered specifically into the LC was sufficient to reduce fasting-induced chow hyperphagia in both sexes, but food reward only in females. Moreover, blockade of GHSR in the LC increased anxiety-like behavior measured in the ASR test in both sexes. Taken together, these results indicate that ghrelin acts in the LC to alter ingestive, motivated and anxiety-like behaviors, with a degree of sex divergence.
Collapse
Affiliation(s)
- Ivana Maric
- Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA, United States
| | - Lorena López-Ferreras
- Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Yashaswini Bhat
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA, United States
| | - Mohammed Asker
- Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Stina Börchers
- Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA, United States
| | - Lauren Bellfy
- Department of Biology, Pennsylvania State University, State College, PA, United States
- Huck Institutes of the Life Sciences, Pennsylvania State University, State College, PA, United States
| | - Suyeun Byun
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA, United States
| | - Janine L. Kwapis
- Department of Biology, Pennsylvania State University, State College, PA, United States
- Huck Institutes of the Life Sciences, Pennsylvania State University, State College, PA, United States
| | - Karolina P. Skibicka
- Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA, United States
- Huck Institutes of the Life Sciences, Pennsylvania State University, State College, PA, United States
| |
Collapse
|
39
|
Barton JR, Londregan AK, Alexander TD, Entezari AA, Covarrubias M, Waldman SA. Enteroendocrine cell regulation of the gut-brain axis. Front Neurosci 2023; 17:1272955. [PMID: 38027512 PMCID: PMC10662325 DOI: 10.3389/fnins.2023.1272955] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Enteroendocrine cells (EECs) are an essential interface between the gut and brain that communicate signals about nutrients, pain, and even information from our microbiome. EECs are hormone-producing cells expressed throughout the gastrointestinal epithelium and have been leveraged by pharmaceuticals like semaglutide (Ozempic, Wegovy), terzepatide (Mounjaro), and retatrutide (Phase 2) for diabetes and weight control, and linaclotide (Linzess) to treat irritable bowel syndrome (IBS) and visceral pain. This review focuses on role of intestinal EECs to communicate signals from the gut lumen to the brain. Canonically, EECs communicate information about the intestinal environment through a variety of hormones, dividing EECs into separate classes based on the hormone each cell type secretes. Recent studies have revealed more diverse hormone profiles and communication modalities for EECs including direct synaptic communication with peripheral neurons. EECs known as neuropod cells rapidly relay signals from gut to brain via a direct communication with vagal and primary sensory neurons. Further, this review discusses the complex information processing machinery within EECs, including receptors that transduce intraluminal signals and the ion channel complement that govern initiation and propagation of these signals. Deeper understanding of EEC physiology is necessary to safely treat devastating and pervasive conditions like irritable bowel syndrome and obesity.
Collapse
Affiliation(s)
- Joshua R. Barton
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Annie K. Londregan
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Tyler D. Alexander
- Department of Neurosciences, Thomas Jefferson University, Philadelphia, PA, United States
| | - Ariana A. Entezari
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Manuel Covarrubias
- Department of Neurosciences, Thomas Jefferson University, Philadelphia, PA, United States
| | - Scott A. Waldman
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
40
|
Leggio L, Leko A, Gregory-Flores A, Marchette R, Gomez J, Vendruscolo J, Repunte-Canonigo V, Chuong V, Deschaine S, Whiting K, Jackson S, Cornejo M, Perello M, You ZB, Eckhaus M, Janda K, Zorman B, Sumazin P, Koob G, Michaelides M, Sanna PP, Vendruscolo L. Genetic or pharmacological GHSR blockade has sexually dimorphic effects in rodents on a high-fat diet. RESEARCH SQUARE 2023:rs.3.rs-3236045. [PMID: 37886546 PMCID: PMC10602167 DOI: 10.21203/rs.3.rs-3236045/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
The stomach-derived hormone ghrelin regulates essential physiological functions. The ghrelin receptor (GHSR) has ligand-independent actions, therefore, GHSR gene deletion may be a reasonable approach to investigate the role of this system in feeding behaviors and diet-induced obesity (DIO). Here we investigated the effects of a long-term (12 month) high-fat (HFD) versus regular diet on obesity-related measures in global GHSR-KO and wild type (WT) Wistar male and female rats. Our main findings were that the GHSR gene deletion protects against DIO and decreases food intake during HFD in male but not in female rats. GHSR gene deletion increased thermogenesis and brain glucose uptake in male rats and modified the effects of HFD on brain glucose metabolism in a sex-specific manner, as assessed with small animal positron emission tomography. RNA-sequencing was also used to show that GHSR-KO rats had upregulated expression of genes responsible for fat oxidation in brown adipose tissue. Central administration of a novel GHSR inverse agonist, PF-5190457, attenuated ghrelin-induced food intake, but only in male, not in female mice. HFD-induced binge-like eating was reduced by inverse agonism in both sexes. Our results support GHSR as a promising target for new pharmacotherapies for obesity.
Collapse
|
41
|
Barrile F, Cassano D, Fernandez G, De Francesco PN, Reynaldo M, Cantel S, Fehrentz JA, Donato J, Schiöth HB, Zigman JM, Perello M. Ghrelin's orexigenic action in the lateral hypothalamic area involves indirect recruitment of orexin neurons and arcuate nucleus activation. Psychoneuroendocrinology 2023; 156:106333. [PMID: 37454647 PMCID: PMC10530520 DOI: 10.1016/j.psyneuen.2023.106333] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/30/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
OBJECTIVE Ghrelin is a potent orexigenic hormone, and the lateral hypothalamic area (LHA) has been suggested as a putative target mediating ghrelin's effects on food intake. Here, we aimed to investigate the presence of neurons expressing ghrelin receptor (a.k.a. growth hormone secretagogue receptor, GHSR) in the mouse LHA (LHAGHSR neurons), its physiological implications and the neuronal circuit recruited by local ghrelin action. METHODS We investigated the distribution of LHAGHSR neurons using different histologic strategies, including the use of a reporter mice expressing enhanced green fluorescent protein under the control of the GHSR promoter. Also, we investigated the physiological implications of local injections of ghrelin within the LHA, and the extent to which the orexigenic effect of intra-LHA-injected ghrelin involves the arcuate nucleus (ARH) and orexin neurons of the LHA (LHAorexin neurons) RESULTS: We found that: 1) LHAGHSR neurons are homogeneously distributed throughout the entire LHA; 2) intra-LHA injections of ghrelin transiently increase food intake and locomotor activity; 3) ghrelin's orexigenic effect in the LHA involves the indirect recruitment of LHAorexin neurons and the activation of ARH neurons; and 4) LHAGHSR neurons are not targeted by plasma ghrelin. CONCLUSIONS We provide a compelling neuroanatomical and functional characterization of LHAGHSR neurons in male mice that indicates that LHAGHSR cells are part of a hypothalamic neuronal circuit that potently induces food intake.
Collapse
Affiliation(s)
- Franco Barrile
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Daniela Cassano
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Gimena Fernandez
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Pablo N De Francesco
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Mirta Reynaldo
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - José Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Mario Perello
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina; Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden.
| |
Collapse
|
42
|
Fernandez G, De Francesco PN, Cornejo MP, Cabral A, Aguggia JP, Duque VJ, Sayar N, Cantel S, Burgos JI, Fehrentz JA, Rorato R, Atasoy D, Mecawi AS, Perello M. Ghrelin Action in the PVH of Male Mice: Accessibility, Neuronal Targets, and CRH Neurons Activation. Endocrinology 2023; 164:bqad154. [PMID: 37823477 PMCID: PMC11491828 DOI: 10.1210/endocr/bqad154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/08/2023] [Accepted: 10/11/2023] [Indexed: 10/13/2023]
Abstract
The hormone ghrelin displays several well-characterized functions, including some with pharmaceutical interest. The receptor for ghrelin, the growth hormone secretagogue receptor (GHSR), is expressed in the hypothalamic paraventricular nucleus (PVH), a critical hub for the integration of metabolic, neuroendocrine, autonomic, and behavioral functions. Here, we performed a neuroanatomical and functional characterization of the neuronal types mediating ghrelin actions in the PVH of male mice. We found that fluorescent ghrelin mainly labels PVH neurons immunoreactive for nitric oxide synthase 1 (NOS1), which catalyze the production of nitric oxide [NO]). Centrally injected ghrelin increases c-Fos in NOS1 PVH neurons and NOS1 phosphorylation in the PVH. We also found that a high dose of systemically injected ghrelin increases the ghrelin level in the cerebrospinal fluid and in the periventricular PVH, and induces c-Fos in NOS1 PVH neurons. Such a high dose of systemically injected ghrelin activates a subset of NOS1 PVH neurons, which do not express oxytocin, via an arcuate nucleus-independent mechanism. Finally, we found that pharmacological inhibition of NO production fully abrogates ghrelin-induced increase of calcium concentration in corticotropin-releasing hormone neurons of the PVH whereas it partially impairs ghrelin-induced increase of plasma glucocorticoid levels. Thus, plasma ghrelin can directly target a subset of NO-producing neurons of the PVH that is involved in ghrelin-induced activation of the hypothalamic-pituitary-adrenal neuroendocrine axis.
Collapse
Affiliation(s)
- Gimena Fernandez
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Pablo N De Francesco
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - María P Cornejo
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Agustina Cabral
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Julieta P Aguggia
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Victor J Duque
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, CEP: 04023-062, Brazil
| | - Nilufer Sayar
- Department of Neuroscience and Pharmacology, Carver College of Medicine, Iowa Neuroscience Institute and Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa, Iowa City, IA 52242, USA
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, Montpellier cedex 5 34293, France
| | - Juan I Burgos
- Centro de Investigaciones Cardiovasculares “Dr. Horacio Eugenio Cingolani” (CONICET and National University of La Plata), La Plata 1900, Buenos Aires, Argentina
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, Montpellier cedex 5 34293, France
| | - Rodrigo Rorato
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, CEP: 04023-062, Brazil
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, Carver College of Medicine, Iowa Neuroscience Institute and Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa, Iowa City, IA 52242, USA
| | - André S Mecawi
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, CEP: 04023-062, Brazil
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala 751 05, Sweden
| |
Collapse
|
43
|
Merritt CR, Garcia EJ, Brehm VD, Fox RG, Moeller FG, Anastasio NC, Cunningham KA. Ghrelin receptor antagonist JMV2959 blunts cocaine and oxycodone drug-seeking, but not self-administration, in male rats. Front Pharmacol 2023; 14:1268366. [PMID: 37795028 PMCID: PMC10545966 DOI: 10.3389/fphar.2023.1268366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/06/2023] [Indexed: 10/06/2023] Open
Abstract
The drug overdose crisis has spawned serious health consequences, including the increased incidence of substance use disorders (SUDs), conditions manifested by escalating medical and psychological impairments. While medication management is a key adjunct in SUD treatment, this crisis has crystallized the need to develop additional therapeutics to facilitate extended recovery from SUDs. The "hunger hormone" ghrelin acts by binding to the growth hormone secretagogue receptor 1α (GHS1αR) to control homeostatic and hedonic aspects of food intake and has been implicated in the mechanisms underlying SUDs. Preclinical studies indicate that GHS1αR antagonists and inverse agonists suppress reward-related signaling associated with cocaine and opioids. In the present study, we found that the GHS1αR antagonist JMV2959 was efficacious to suppress both cue-reinforced cocaine and oxycodone drug-seeking, but not cocaine or oxycodone self-administration in male Sprague-Dawley rats. These data suggest a role of the ghrelin-GHS1αR axis in mediating overlapping reward-related aspects of cocaine and oxycodone and premises the possibility that a GHS1αR antagonist may be a valuable therapeutic strategy for relapse vulnerability in SUDs.
Collapse
Affiliation(s)
- Christina R. Merritt
- Center for Addiction Sciences and Therapeutics and Department of Pharmacology and Toxicology, John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Erik J. Garcia
- Center for Addiction Sciences and Therapeutics and Department of Pharmacology and Toxicology, John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Victoria D. Brehm
- Center for Addiction Sciences and Therapeutics and Department of Pharmacology and Toxicology, John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Robert G. Fox
- Center for Addiction Sciences and Therapeutics and Department of Pharmacology and Toxicology, John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - F. Gerard Moeller
- C. Kenneth and Dianne Wright Center for Clinical and Translational Research, Departments of Psychiatry and Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Noelle C. Anastasio
- Center for Addiction Sciences and Therapeutics and Department of Pharmacology and Toxicology, John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Kathryn A. Cunningham
- Center for Addiction Sciences and Therapeutics and Department of Pharmacology and Toxicology, John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
44
|
Geisler CE, Hayes MR. Metabolic hormone action in the VTA: Reward-directed behavior and mechanistic insights. Physiol Behav 2023; 268:114236. [PMID: 37178855 PMCID: PMC10330780 DOI: 10.1016/j.physbeh.2023.114236] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/10/2023] [Accepted: 05/10/2023] [Indexed: 05/15/2023]
Abstract
Dysfunctional signaling in midbrain reward circuits perpetuates diseases characterized by compulsive overconsumption of rewarding substances such as substance abuse, binge eating disorder, and obesity. Ventral tegmental area (VTA) dopaminergic activity serves as an index for how rewarding stimuli are perceived and triggers behaviors necessary to obtain future rewards. The evolutionary linking of reward with seeking and consuming palatable foods ensured an organism's survival, and hormone systems that regulate appetite concomitantly developed to regulate motivated behaviors. Today, these same mechanisms serve to regulate reward-directed behavior around food, drugs, alcohol, and social interactions. Understanding how hormonal regulation of VTA dopaminergic output alters motivated behaviors is essential to leveraging therapeutics that target these hormone systems to treat addiction and disordered eating. This review will outline our current understanding of the mechanisms underlying VTA action of the metabolic hormones ghrelin, glucagon-like peptide-1, amylin, leptin, and insulin to regulate behavior around food and drugs of abuse, highlighting commonalities and differences in how these five hormones ultimately modulate VTA dopamine signaling.
Collapse
Affiliation(s)
- Caroline E Geisler
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
45
|
Chae Y, Lee IS. Central Regulation of Eating Behaviors in Humans: Evidence from Functional Neuroimaging Studies. Nutrients 2023; 15:3010. [PMID: 37447336 PMCID: PMC10347214 DOI: 10.3390/nu15133010] [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: 06/12/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Neuroimaging has great potential to provide insight into the neural response to food stimuli. Remarkable advances have been made in understanding the neural activity underlying food perception, not only in normal eating but also in obesity, eating disorders, and disorders of gut-brain interaction in recent decades. In addition to the abnormal brain function in patients with eating disorders compared to healthy controls, new therapies, such as neurofeedback and neurostimulation techniques, have been developed that target the malfunctioning brain regions in patients with eating disorders based on the results of neuroimaging studies. In this review, we present an overview of early and more recent research on the central processing and regulation of eating behavior in healthy and patient populations. In order to better understand the relationship between the gut and the brain as well as the neural mechanisms underlying abnormal ingestive behaviors, we also provide suggestions for future directions to enhance our current methods used in food-related neuroimaging studies.
Collapse
Affiliation(s)
- Younbyoung Chae
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - In-Seon Lee
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
46
|
Khouma A, Moeini MM, Plamondon J, Richard D, Caron A, Michael NJ. Histaminergic regulation of food intake. Front Endocrinol (Lausanne) 2023; 14:1202089. [PMID: 37448468 PMCID: PMC10338010 DOI: 10.3389/fendo.2023.1202089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/06/2023] [Indexed: 07/15/2023] Open
Abstract
Histamine is a biogenic amine that acts as a neuromodulator within the brain. In the hypothalamus, histaminergic signaling contributes to the regulation of numerous physiological and homeostatic processes, including the regulation of energy balance. Histaminergic neurons project extensively throughout the hypothalamus and two histamine receptors (H1R, H3R) are strongly expressed in key hypothalamic nuclei known to regulate energy homeostasis, including the paraventricular (PVH), ventromedial (VMH), dorsomedial (DMH), and arcuate (ARC) nuclei. The activation of different histamine receptors is associated with differential effects on neuronal activity, mediated by their different G protein-coupling. Consequently, activation of H1R has opposing effects on food intake to that of H3R: H1R activation suppresses food intake, while H3R activation mediates an orexigenic response. The central histaminergic system has been implicated in atypical antipsychotic-induced weight gain and has been proposed as a potential therapeutic target for the treatment of obesity. It has also been demonstrated to interact with other major regulators of energy homeostasis, including the central melanocortin system and the adipose-derived hormone leptin. However, the exact mechanisms by which the histaminergic system contributes to the modification of these satiety signals remain underexplored. The present review focuses on recent advances in our understanding of the central histaminergic system's role in regulating feeding and highlights unanswered questions remaining in our knowledge of the functionality of this system.
Collapse
Affiliation(s)
- Axelle Khouma
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Moein Minbashi Moeini
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Julie Plamondon
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
| | - Denis Richard
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Medicine, Université Laval, Québec, QC, Canada
| | - Alexandre Caron
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
- Montreal Diabetes Research Center, Montreal, QC, Canada
| | - Natalie Jane Michael
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| |
Collapse
|
47
|
Vaudry H, Schoofs L, Civelli O, Kojima M. Editorial: Neuropeptide GPCRs in neuroendocrinology, Volume II. Front Endocrinol (Lausanne) 2023; 14:1219530. [PMID: 37415662 PMCID: PMC10321770 DOI: 10.3389/fendo.2023.1219530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/25/2023] [Indexed: 07/08/2023] Open
Affiliation(s)
- Hubert Vaudry
- Institute of Biomedical Research and Innovation, University of Rouen Normandy, Mont-Saint-Aignan, France
| | | | - Olivier Civelli
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States
| | - Masayasu Kojima
- Institute of Life Science, Kurume University, Fukuoka, Japan
| |
Collapse
|
48
|
Weaver KJ, Raju S, Rucker RA, Chakraborty T, Holt RA, Pletcher SD. Behavioral dissection of hunger states in Drosophila. eLife 2023; 12:RP84537. [PMID: 37326496 PMCID: PMC10328523 DOI: 10.7554/elife.84537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023] Open
Abstract
Hunger is a motivational drive that promotes feeding, and it can be generated by the physiological need to consume nutrients as well as the hedonic properties of food. Brain circuits and mechanisms that regulate feeding have been described, but which of these contribute to the generation of motive forces that drive feeding is unclear. Here, we describe our first efforts at behaviorally and neuronally distinguishing hedonic from homeostatic hunger states in Drosophila melanogaster and propose that this system can be used as a model to dissect the molecular mechanisms that underlie feeding motivation. We visually identify and quantify behaviors exhibited by hungry flies and find that increased feeding duration is a behavioral signature of hedonic feeding motivation. Using a genetically encoded marker of neuronal activity, we find that the mushroom body (MB) lobes are activated by hedonic food environments, and we use optogenetic inhibition to implicate a dopaminergic neuron cluster (protocerebral anterior medial [PAM]) to α'/β' MB circuit in hedonic feeding motivation. The identification of discrete hunger states in flies and the development of behavioral assays to measure them offers a framework to begin dissecting the molecular and circuit mechanisms that generate motivational states in the brain.
Collapse
Affiliation(s)
- Kristina J Weaver
- Department of Molecular and Integrative Physiology and Geriatrics Center, Biomedical Sciences and Research Building, University of MichiganAnn ArborUnited States
| | - Sonakshi Raju
- College of Literature, Science, and the Arts, Biomedical Sciences and Research Building, University of MichiganAnn ArborUnited States
| | - Rachel A Rucker
- Neuroscience Graduate Program, University of Michigan, University of MichiganAnn ArborUnited States
| | - Tuhin Chakraborty
- Department of Molecular and Integrative Physiology and Geriatrics Center, Biomedical Sciences and Research Building, University of MichiganAnn ArborUnited States
| | - Robert A Holt
- College of Literature, Science, and the Arts, Biomedical Sciences and Research Building, University of MichiganAnn ArborUnited States
| | - Scott D Pletcher
- Department of Molecular and Integrative Physiology and Geriatrics Center, Biomedical Sciences and Research Building, University of MichiganAnn ArborUnited States
| |
Collapse
|
49
|
Tian J, Guo L, Wang T, Jia K, Swerdlow RH, Zigman JM, Du H. Liver-expressed antimicrobial peptide 2 elevation contributes to age-associated cognitive decline. JCI Insight 2023; 8:166175. [PMID: 37212281 DOI: 10.1172/jci.insight.166175] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/31/2023] [Indexed: 05/23/2023] Open
Abstract
Elderly individuals frequently report cognitive decline, while various studies indicate hippocampal functional declines with advancing age. Hippocampal function is influenced by ghrelin through hippocampus-expressed growth hormone secretagogue receptor (GHSR). Liver-expressed antimicrobial peptide 2 (LEAP2) is an endogenous GHSR antagonist that attenuates ghrelin signaling. Here, we measured plasma ghrelin and LEAP2 levels in a cohort of cognitively normal individuals older than 60 and found that LEAP2 increased with age while ghrelin (also referred to in literature as "acyl-ghrelin") marginally declined. In this cohort, plasma LEAP2/ghrelin molar ratios were inversely associated with Mini-Mental State Examination scores. Studies in mice showed an age-dependent inverse relationship between plasma LEAP2/ghrelin molar ratio and hippocampal lesions. In aged mice, restoration of the LEAP2/ghrelin balance to youth-associated levels with lentiviral shRNA Leap2 downregulation improved cognitive performance and mitigated various age-related hippocampal deficiencies such as CA1 region synaptic loss, declines in neurogenesis, and neuroinflammation. Our data collectively suggest that LEAP2/ghrelin molar ratio elevation may adversely affect hippocampal function and, consequently, cognitive performance; thus, it may serve as a biomarker of age-related cognitive decline. Moreover, targeting LEAP2 and ghrelin in a manner that lowers the plasma LEAP2/ghrelin molar ratio could benefit cognitive performance in elderly individuals for rejuvenation of memory.
Collapse
Affiliation(s)
- Jing Tian
- Department of Pharmacology and Toxicology and
| | - Lan Guo
- Higuchi Biosciences Center, University of Kansas, Lawrence, Kansas, USA
| | - Tienju Wang
- Department of Pharmacology and Toxicology and
| | - Kun Jia
- Department of Pharmacology and Toxicology and
| | - Russell H Swerdlow
- Alzheimer's Disease Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jeffrey M Zigman
- Departments of Internal Medicine and Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Heng Du
- Department of Pharmacology and Toxicology and
- Higuchi Biosciences Center, University of Kansas, Lawrence, Kansas, USA
- Alzheimer's Disease Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
50
|
Willmore L, Minerva AR, Engelhard B, Murugan M, McMannon B, Oak N, Thiberge SY, Peña CJ, Witten IB. Overlapping representations of food and social stimuli in VTA dopamine neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541104. [PMID: 37293057 PMCID: PMC10245666 DOI: 10.1101/2023.05.17.541104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Dopamine neurons of the ventral tegmental area (VTA DA ) respond to food and social stimuli and contribute to both forms of motivation. However, it is unclear if the same or different VTA DA neurons encode these different stimuli. To address this question, we performed 2-photon calcium imaging in mice presented with food and conspecifics, and found statistically significant overlap in the populations responsive to both stimuli. Both hunger and opposite-sex social experience further increased the proportion of neurons that respond to both stimuli, implying that modifying motivation for one stimulus affects responses to both stimuli. In addition, single-nucleus RNA sequencing revealed significant co-expression of feeding- and social-hormone related genes in individual VTA DA neurons. Taken together, our functional and transcriptional data suggest overlapping VTA DA populations underlie food and social motivation.
Collapse
Affiliation(s)
- Lindsay Willmore
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Adelaide R. Minerva
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Ben Engelhard
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
- Department of Medicine, Technion, Haifa, 3525433, Israel
| | - Malavika Murugan
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Brenna McMannon
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Nirja Oak
- Department of Medicine, Technion, Haifa, 3525433, Israel
| | - Stephan Y. Thiberge
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Catherine J. Peña
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Ilana B. Witten
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| |
Collapse
|