1
|
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
|
2
|
Moran KM, Enstrom AE, Jarrell L, Khashchuluun M, Tran A, Delville Y. Adolescent social stress alters the role of orexin innervation in the hindbrain in male hamsters. J Neuroendocrinol 2024:e13457. [PMID: 39462511 DOI: 10.1111/jne.13457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/05/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024]
Abstract
Juvenile male hamsters exposed to chronic social stress eat more, gain weight, and have larger fat pads. The purpose of the present study was to address possible changes in food hoarding and orexin/hypocretin innervation in response to social stress. Male hamsters in early adolescence were exposed to a resident-intruder social stress paradigm or control condition daily for 2 weeks. Metabolism-related physiological measures and behaviors were tracked, and brains were immunocytochemically labeled for orexin-A. Our data confirm our previous observations on appetite, weight gain, and obesity, and showed a strong trend toward enhanced food hoarding as in prior studies. In addition, there were no statistically significant differences in orexin innervation in any brain area analyzed. However, unique correlation patterns were observed between orexin innervation and appetite or metabolic outcome. In particular, opposite correlations were observed between groups within the dorsal raphe nucleus, lateral parabrachial nucleus, and nucleus of the solitary tract. These opposite patterns of correlations suggest chronic social stress causes site-specific alterations in synaptic activity in relation with these behaviors.
Collapse
Affiliation(s)
- Kevin M Moran
- Psychology Department, The University of Texas at Austin, Austin, Texas, USA
| | - Ava Elana Enstrom
- Psychology Department, The University of Texas at Austin, Austin, Texas, USA
| | - Leah Jarrell
- Psychology Department, The University of Texas at Austin, Austin, Texas, USA
| | | | - Anna Tran
- Psychology Department, The University of Texas at Austin, Austin, Texas, USA
| | - Yvon Delville
- Psychology Department, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
3
|
Kore MS, Mamsa R, Patil D, Bhatt LK. Ghrelin in Depression: A Promising Therapeutic Target. Mol Neurobiol 2024:10.1007/s12035-024-04554-1. [PMID: 39424690 DOI: 10.1007/s12035-024-04554-1] [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: 08/16/2023] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Depression is a widespread disease affecting over 300 million individuals of various ethnicities and socioeconomic backgrounds globally. It frequently strikes early in life and becomes a chronic or recurring lifelong illness. Out of the various hypotheses for the pathophysiology of depression, the gut-brain axis and stress hypothesis are the ones that need to be researched, as psychological stress impairs one or more pathways of the brain-gut axis and is likely to cause brain-gut axis dysfunction and depression. A dysfunctional reciprocal gut-brain relationship may contribute to many diseases, including inflammatory disorders, abnormal stress responses, impaired behavior, and metabolic changes. The hormone ghrelin is a topic of interest concerning the gut-brain axis as it interacts with the gut-brain axis indirectly via the central nervous system or via crossing the blood-brain barrier. Ghrelin release is also affected by the gut microbes, which has also been discussed in the review. This review elaborates on Ghrelin's role in depression and its effect on various aspects like neurogenesis, HPA axis, and neuroinflammation. Furthermore, this review focuses on ghrelin as a potential target for alleviation of depressive symptoms.
Collapse
Affiliation(s)
- Mikhil Santosh Kore
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Rumaiza Mamsa
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Dipti Patil
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India.
| |
Collapse
|
4
|
Richardson RS, Kryszak LA, Vendruscolo JCM, Koob GF, Vendruscolo LF, Leggio L. GHSR blockade, but not reduction of peripherally circulating ghrelin via β 1-adrenergic receptor antagonism, decreases binge-like alcohol drinking in mice. Mol Psychiatry 2024:10.1038/s41380-024-02713-3. [PMID: 39232198 DOI: 10.1038/s41380-024-02713-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 09/06/2024]
Abstract
Alcohol use disorder (AUD) and binge drinking are highly prevalent public health issues. The stomach-derived peptide ghrelin, and its receptor, the growth hormone secretagogue receptor (GHSR), both of which are expressed in the brain and periphery, are implicated in alcohol-related outcomes. We previously found that systemic and central administration of GHSR antagonists reduced binge-like alcohol drinking, whereas a ghrelin vaccine did not. Thus, we hypothesized that central GHSR drives binge-like alcohol drinking independently of peripheral ghrelin. To investigate this hypothesis, we antagonized β1-adrenergic receptors (β1ARs), which are required for peripheral ghrelin release, and combined them with GHSR blockers. We found that both systemic β1AR antagonism with atenolol (peripherally restricted) and metoprolol (brain permeable) robustly decreased plasma ghrelin levels. Also, ICV administration of atenolol had no effect on peripheral endogenous ghrelin levels. However, only metoprolol, but not atenolol, decreased binge-like alcohol drinking. The β1AR antagonism also did not prevent the effects of the GHSR blockers JMV2959 and PF-5190457 in decreasing binge-like alcohol drinking. These results suggest that the GHSR rather than peripheral endogenous ghrelin is involved in binge-like alcohol drinking. Thus, GHSRs and β1ARs represent possible targets for therapeutic intervention for AUD, including the potential combination of drugs that target these two systems.
Collapse
Affiliation(s)
- Rani S Richardson
- 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 and Bethesda, MD, USA
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
- University of North Carolina School of Medicine MD/PhD Program, University of North Carolina, Chapel Hill, NC, USA
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
- 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
| | - Lindsay A Kryszak
- National Institute on Drug Abuse Intramural Research Program Translational Analytical Core, 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
- 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
| | - George F Koob
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, 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 and Bethesda, MD, USA.
- National Institute on Drug Abuse Intramural Research Program Translational Analytical Core, National Institutes of Health, Baltimore, MD, USA.
- Department of Behavioral and Social Sciences, Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA.
- Medication Development Program, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, 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
|
5
|
Pan X, Gao Y, Guan K, Chen J, Ji B. Ghrelin/GHSR System in Depressive Disorder: Pathologic Roles and Therapeutic Implications. Curr Issues Mol Biol 2024; 46:7324-7338. [PMID: 39057075 PMCID: PMC11275499 DOI: 10.3390/cimb46070434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Depression is the most common chronic mental illness and is characterized by low mood, insomnia, and affective disorders. However, its pathologic mechanisms remain unclear. Numerous studies have suggested that the ghrelin/GHSR system may be involved in the pathophysiologic process of depression. Ghrelin plays a dual role in experimental animals, increasing depressed behavior and decreasing anxiety. By combining several neuropeptides and traditional neurotransmitter systems to construct neural networks, this hormone modifies signals connected to depression. The present review focuses on the role of ghrelin in neuritogenesis, astrocyte protection, inflammatory factor production, and endocrine disruption in depression. Furthermore, ghrelin/GHSR can activate multiple signaling pathways, including cAMP/CREB/BDNF, PI3K/Akt, Jak2/STAT3, and p38-MAPK, to produce antidepressant effects, given which it is expected to become a potential therapeutic target for the treatment of depression.
Collapse
Affiliation(s)
- Xingli Pan
- School of Biological Sciences, Jining Medical University, Jining 272067, China;
| | - Yuxin Gao
- School of Clinical Medicine, Jining Medical University, Jining 272067, China; (Y.G.); (K.G.)
| | - Kaifu Guan
- School of Clinical Medicine, Jining Medical University, Jining 272067, China; (Y.G.); (K.G.)
| | - Jing Chen
- Neurobiology Institute, Jining Medical University, Jining 272067, China
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Bingyuan Ji
- Institute of Precision Medicine, Jining Medical University, Jining 272067, China
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Smith A, Rodrigues T, Wallace C, Mezher K, MacAulay B, Prowse R, Hyland L, Abizaid A. Growth Hormone Secretagogue Receptor (GHSR) Signaling in the Ventral Tegmental Area (VTA) Mediates Feeding Produced by Chronic Social Defeat Stress in Male Mice. Neuroscience 2024; 547:17-27. [PMID: 38583506 DOI: 10.1016/j.neuroscience.2024.03.022] [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: 12/20/2023] [Revised: 03/04/2024] [Accepted: 03/18/2024] [Indexed: 04/09/2024]
Abstract
Ghrelin, a hormone secreted by the stomach, binds to the growth hormone secretagogue receptor (GHSR) in various brain regions to produce a number of behavioral effects that include increased feeding motivation. During social defeat stress, ghrelin levels rise in correlation with increased feeding and potentially play a role in attenuating the anxiogenic effects of social defeat. One region implicated in the feeding effects of ghrelin is the ventral tegmental area (VTA), a region implicated in reward seeking behaviors, and linked to social defeat in mice. Here we examined the role of GHSR signaling in the VTA in feeding behavior in mice exposed to social defeat stress. Male C57BL/J6 mice that were socially defeated once daily for 3 weeks ate more, had higher plasma ghrelin level and increased GHSR expression in the VTA compared to non-stressed mice. Socially defeated GHSR KO mice failed to increase their caloric intake in response to this stressor but rescue of GHSR expression in the VTA restored feeding responses. Finally, we pharmacologically blocked VTA GHSR signalling with JMV2959 infused via an indwelling VTA cannula connected to a minipump. Vehicle-treated mice increased their caloric intake during social defeat, but JMV2959-infusions attenuated feeding responses and increased anxiety-like behaviors. The data suggest that GHSR signalling in the VTA is critical for the increases in appetite observed during chronic social defeat stress. Furthermore, these data support the idea that GHSR signaling in the VTA may also have anxiolytic effects, and blocking GHSR in this region may result in an anxiety-like phenotype.
Collapse
Affiliation(s)
- Andrea Smith
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Trevor Rodrigues
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Caroline Wallace
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Karen Mezher
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Brenna MacAulay
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Rebecca Prowse
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Lindsay Hyland
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Alfonso Abizaid
- Carleton University, Neuroscience Department, Ottawa, ON, Canada.
| |
Collapse
|
9
|
Chang L, Niu F, Li B. Ghrelin/GHSR signaling in the lateral septum ameliorates chronic stress-induced depressive-like behaviors. Prog Neuropsychopharmacol Biol Psychiatry 2024; 131:110953. [PMID: 38278286 DOI: 10.1016/j.pnpbp.2024.110953] [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: 08/26/2023] [Revised: 01/11/2024] [Accepted: 01/23/2024] [Indexed: 01/28/2024]
Abstract
Ghrelin is a gastrointestinal hormone on feeding and metabolism regulation, and acts through its receptor-growth hormone secretagogue receptor (GHSR), which is widely distributed throughout the central nervous system. Recent studies have suggested that ghrelin plays an important role in the regulation of depression, but the underlying mechanisms remain uncertain. Lateral septum (LS) is a critical brain region in modulating depression. Therefore, we investigated the role of ghrelin/GHSR signaling in the LS on the depressive-like behaviors of mice under conditions of chronic stress by using behavioral tests, neuropharmacology, and molecular biology techniques. We found that infusion of ghrelin into the LS produced antidepressant-like responses in mice. Activation of LS GABAergic neurons was involved in the antidepressant effect of ghrelin. Importantly, GHSR was highly expressed and distributed in the LS neurons. Blockade of GHSR in the LS reversed the ghrelin-induced antidepressant-like effects. Molecular knockdown of GHSR in the LS induced depressive-like symptoms in mice. Furthermore, administration of ghrelin into the LS alleviated depressive-like behaviors induced by chronic social defeat stress (CSDS). Consistent with the neuropharmacological results, overexpression of GHSR in the LS reversed CSDS-induced depressive-like behaviors. Our findings clarify a key role for ghrelin/GHSR signaling in the regulation of chronic stress-induced depressive-like behaviors, which could provide new strategies for the treatment of depression.
Collapse
Affiliation(s)
- Leilei Chang
- Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Fengnan Niu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Bin Li
- Women and Children's Medical Research Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
10
|
Moran KM, Delville Y. A hamster model for stress-induced weight gain. Horm Behav 2024; 160:105488. [PMID: 38306877 DOI: 10.1016/j.yhbeh.2024.105488] [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: 09/05/2023] [Revised: 12/18/2023] [Accepted: 01/18/2024] [Indexed: 02/04/2024]
Abstract
This review addresses the translational relevance of animal models of stress and their effects on body weight. In humans, stress, whether chronic or acute, has often been associated with increased food intake and weight gain. In view of the current obesity epidemic, this phenomenon is especially relevant. Such observations contrast with reports with commonly used laboratory animals, especially rats and mice. In these species, it is common to find individuals gaining less weight under stress, even with potent social stressors. However, there are laboratory species that present increased appetite and weight gain under stress, such as golden hamsters. Furthermore, these animals also include metabolic and behavioral similarities with humans, including hoarding behavior which is also enhanced under stress. Consequently, we propose that our comparative perspective provides useful insights for future research on the development of obesity in humans as a consequence of chronic stress exposure.
Collapse
Affiliation(s)
- Kevin M Moran
- Psychology Department, The University of Texas at Austin, USA.
| | - Yvon Delville
- Psychology Department, The University of Texas at Austin, USA
| |
Collapse
|
11
|
Kulkarni SS, Singh O, Zigman JM. The intersection between ghrelin, metabolism and circadian rhythms. Nat Rev Endocrinol 2024; 20:228-238. [PMID: 38123819 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
|
12
|
Wilbrecht L, Lin WC, Callahan K, Bateson M, Myers K, Ross R. Experimental biology can inform our understanding of food insecurity. J Exp Biol 2024; 227:jeb246215. [PMID: 38449329 PMCID: PMC10949070 DOI: 10.1242/jeb.246215] [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] [Indexed: 03/08/2024]
Abstract
Food insecurity is a major public health issue. Millions of households worldwide have intermittent and unpredictable access to food and this experience is associated with greater risk for a host of negative health outcomes. While food insecurity is a contemporary concern, we can understand its effects better if we acknowledge that there are ancient biological programs that evolved to respond to the experience of food scarcity and uncertainty, and they may be particularly sensitive to food insecurity during development. Support for this conjecture comes from common findings in several recent animal studies that have modeled insecurity by manipulating predictability of food access in various ways. Using different experimental paradigms in different species, these studies have shown that experience of insecure access to food can lead to changes in weight, motivation and cognition. Some of these studies account for changes in weight through changes in metabolism, while others observe increases in feeding and motivation to work for food. It has been proposed that weight gain is an adaptive response to the experience of food insecurity as 'insurance' in an uncertain future, while changes in motivation and cognition may reflect strategic adjustments in foraging behavior. Animal studies also offer the opportunity to make in-depth controlled studies of mechanisms and behavior. So far, there is evidence that the experience of food insecurity can impact metabolic efficiency, reproductive capacity and dopamine neuron synapses. Further work on behavior, the central and peripheral nervous system, the gut and liver, along with variation in age of exposure, will be needed to better understand the full body impacts of food insecurity at different stages of development.
Collapse
Affiliation(s)
- Linda Wilbrecht
- Department of Psychology, University of California, Berkeley, Berkeley, CA 94720-1650, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Wan Chen Lin
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kathryn Callahan
- Psychiatric Research Institute of Montefiore and Einstein, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Melissa Bateson
- Bioscience Institute, University of Newcastle, Newcastle upon Tyne, NE2 4HH, UK
| | - Kevin Myers
- Department of Psychology and Programs in Animal Behavior and Neuroscience, Bucknell University, Lewisburg, PA 17837, USA
| | - Rachel Ross
- Psychiatric Research Institute of Montefiore and Einstein, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Department of Psychiatry, Montefiore Medical Center, Bronx, New York, NY 10467, USA
| |
Collapse
|
13
|
Zhang C, Dong Y, Li S, Li M, Gao Y, Liu Y, Liu X, Zhou C, Li J. Ghrelin and depressive symptoms in patients with first-episode drug-naïve major depressive disorder: The mediating role of hypothalamic-pituitary-adrenal axis. Asia Pac Psychiatry 2024; 16:e12552. [PMID: 38348641 DOI: 10.1111/appy.12552] [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: 08/27/2023] [Revised: 12/13/2023] [Accepted: 01/19/2024] [Indexed: 02/15/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) is one of the global burdens of disease, and its pathogenesis remains unclear. An increasing amount of research indicates that ghrelin regulates mood in patients with MDD. Still, current results are inconsistent, and the mechanisms underlying how ghrelin modulates depressive symptoms are inconclusive, especially in first-episode drug-naïve MDD patients. Therefore, this study aims to investigate the relationship and potential mechanism between ghrelin and first-episode drug-naïve MDD. METHODS Ninety first-episode drug-naïve MDD patients and 65 healthy controls (HCs) were included. Hamilton Depression Scale (HAMD-17) as a measure of depressive symptoms. Plasma levels of ghrelin and hypothalamic-pituitary-adrenal axis (HPA-axis) hormones were measured in all participants. RESULTS Compared to HCs, the ghrelin levels were higher in the MDD (p < .001) and still showed significance after covarying for sex, age, and Body Mass Index (BMI). Ghrelin was positively related to corticotropin-releasing-hormone (CRH) levels (r = .867, p < .001), adrenocorticotropic hormone (ACTH) levels (r = .830, p < .001), and cortisol levels (r = .902, p < .001) in partial correlation analysis. In addition, there was a positive correlation between HAMD total score and ghrelin levels (r = .240, p = .026). Other than that, the HAMD total score also had a positive correlation with the CRH (r = .333, p = .002) and cortisol (r = .307, p = .004) levels. Further mediation analysis demonstrated that the relationship between ghrelin and HAMD total score was mediated by CRH (ab-path; β = .4457, 95% CI = 0.0780-1.0253, c-path; β = .2447, p = .0260, c'-path; β = -.2009, p = .3427). CONCLUSIONS These findings revealed that plasma ghrelin provides a pivotal link to depressive symptoms in first-episode drug-naive MDD patients. CRH mediated the relationship between ghrelin and HAMD total score. It might provide new insights into understanding the pathogenesis of MDD, contributing to intervention and treatment from this approach.
Collapse
Affiliation(s)
- Chuhao Zhang
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Yeqing Dong
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - ShuHua Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
- Department of Clinical Psychology, Tianjin TEDA Hospital, Tianjin, China
| | - Meijuan Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Ying Gao
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Yuan Liu
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Xueying Liu
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Chi Zhou
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Jie Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| |
Collapse
|
14
|
Martínez-Magaña JJ, Genis-Mendoza AD, Gallegos-Silva I, López-Narváez ML, Juárez-Rojop IE, Diaz-Zagoya JC, Tovilla-Zárate CA, González-Castro TB, Nicolini H, Solis-Medina A. Differential Alterations of Expression of the Serotoninergic System Genes and Mood-Related Behavior by Consumption of Aspartame or Potassium Acesulfame in Rats. Nutrients 2024; 16:490. [PMID: 38398814 PMCID: PMC10892058 DOI: 10.3390/nu16040490] [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: 01/08/2024] [Revised: 01/18/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
The use of aspartame (ASP) and potassium acesulfame (ACK) to reduce weight gain is growing; however, contradictory effects in body mass index control and neurobiological alterations resulting from artificial sweeteners consumption have been reported. This study aimed to evaluate the impact of the chronic consumption of ASP and ACK on mood-related behavior and the brain expression of serotonin genes in male Wistar rats. Mood-related behaviors were evaluated using the swim-forced test and defensive burying at two time points: 45 days (juvenile) and 95 days (adult) postweaning. Additionally, the mRNA expression of three serotoninergic genes (Slc6a4, Htr1a, and Htr2c) was measured in the brain areas (prefrontal cortex, hippocampus, and hypothalamus) involved in controlling mood-related behaviors. In terms of mood-related behaviors, rats consuming ACK exhibited anxiety-like behavior only during the juvenile stage. In contrast, rats consuming ASP showed a reduction in depressive-like behavior during the juvenile stage but an increase in the adult stage. The expression of Slc6a4 mRNA increased in the hippocampus of rats consuming artificial sweeteners during the juvenile stage. In the adult stage, there was an upregulation in the relative expression of Slc6a4 and Htr1a in the hypothalamus, while Htr2c expression decreased in the hippocampus of rats consuming ASP. Chronic consumption of ASP and ACK appears to have differential effects during neurodevelopmental stages in mood-related behavior, potentially mediated by alterations in serotoninergic gene expression.
Collapse
Affiliation(s)
- José Jaime Martínez-Magaña
- Laboratorio de Genómica de Enfermedades Psiquiátricas y Neurodegenerativas, Instituto Nacional de Medicina Genómica, Ciudad de México 14610, Mexico; (J.J.M.-M.); (A.D.G.-M.); (I.G.-S.); (A.S.-M.)
| | - Alma Delia Genis-Mendoza
- Laboratorio de Genómica de Enfermedades Psiquiátricas y Neurodegenerativas, Instituto Nacional de Medicina Genómica, Ciudad de México 14610, Mexico; (J.J.M.-M.); (A.D.G.-M.); (I.G.-S.); (A.S.-M.)
| | - Ileana Gallegos-Silva
- Laboratorio de Genómica de Enfermedades Psiquiátricas y Neurodegenerativas, Instituto Nacional de Medicina Genómica, Ciudad de México 14610, Mexico; (J.J.M.-M.); (A.D.G.-M.); (I.G.-S.); (A.S.-M.)
| | - María Lilia López-Narváez
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa 86100, Mexico; (M.L.L.-N.); (I.E.J.-R.)
| | - Isela Esther Juárez-Rojop
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa 86100, Mexico; (M.L.L.-N.); (I.E.J.-R.)
| | - Juan C. Diaz-Zagoya
- División de Investigación, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Carlos Alfonso Tovilla-Zárate
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa 86100, Mexico; (M.L.L.-N.); (I.E.J.-R.)
| | | | - Humberto Nicolini
- Laboratorio de Genómica de Enfermedades Psiquiátricas y Neurodegenerativas, Instituto Nacional de Medicina Genómica, Ciudad de México 14610, Mexico; (J.J.M.-M.); (A.D.G.-M.); (I.G.-S.); (A.S.-M.)
| | - Anayelly Solis-Medina
- Laboratorio de Genómica de Enfermedades Psiquiátricas y Neurodegenerativas, Instituto Nacional de Medicina Genómica, Ciudad de México 14610, Mexico; (J.J.M.-M.); (A.D.G.-M.); (I.G.-S.); (A.S.-M.)
| |
Collapse
|
15
|
Nwakama CA, Durand-de Cuttoli R, Oketokoun ZM, Brown SO, Haller JE, Méndez A, Farshbaf MJ, Cho YZ, Ahmed S, Leng S, Ables JL, Sweis BM. Diabetes alters neuroeconomically dissociable forms of mental accounting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.04.574210. [PMID: 38260368 PMCID: PMC10802482 DOI: 10.1101/2024.01.04.574210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Those with diabetes mellitus are at high-risk of developing psychiatric disorders, yet the link between hyperglycemia and alterations in motivated behavior has not been explored in detail. We characterized value-based decision-making behavior of a streptozocin-induced diabetic mouse model on a naturalistic neuroeconomic foraging paradigm called Restaurant Row. Mice made self-paced choices while on a limited time-budget accepting or rejecting reward offers as a function of cost (delays cued by tone-pitch) and subjective value (flavors), tested daily in a closed-economy system across months. We found streptozocin-treated mice disproportionately undervalued less-preferred flavors and inverted their meal-consumption patterns shifted toward a more costly strategy that overprioritized high-value rewards. We discovered these foraging behaviors were driven by impairments in multiple decision-making systems, including the ability to deliberate when engaged in conflict and cache the value of the passage of time in the form of sunk costs. Surprisingly, diabetes-induced changes in behavior depended not only on the type of choice being made but also the salience of reward-scarcity in the environment. These findings suggest complex relationships between glycemic regulation and dissociable valuation algorithms underlying unique cognitive heuristics and sensitivity to opportunity costs can disrupt fundamentally distinct computational processes and could give rise to psychiatric vulnerabilities.
Collapse
|
16
|
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
|
17
|
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
|
18
|
Moazzami K, Pearce BD, Gurel NZ, Wittbrodt MT, Levantsevych OM, Huang M, Shandhi MMH, Herring I, Murrah N, Driggers E, Alkhalaf ML, Soudan M, Shallenberger L, Hankus AN, Nye JA, Vaccarino V, Shah AJ, Inan OT, Bremner JD. Transcutaneous vagal nerve stimulation modulates stress-induced plasma ghrelin levels: A double-blind, randomized, sham-controlled trial. J Affect Disord 2023; 342:85-90. [PMID: 37714385 PMCID: PMC10698687 DOI: 10.1016/j.jad.2023.09.015] [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: 06/20/2023] [Revised: 08/22/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023]
Abstract
BACKGROUND Transcutaneous cervical vagus nerve stimulation (tcVNS) has emerged as a potential treatment strategy for patients with stress-related psychiatric disorders. Ghrelin is a hormone that has been postulated to be a biomarker of stress. While the mechanisms of action of tcVNS are unclear, we hypothesized that tcVNS reduces the levels of ghrelin in response to stress. METHODS Using a randomized double-blind approach, we studied the effects of tcVNS on ghrelin levels in individuals with a history of exposure to traumatic stress. Participants received either sham (n = 29) or active tcVNS (n = 26) after exposure to acute personalized traumatic script stress and mental stress challenges (public speech, mental arithmetic) over a three day period. RESULTS There were no significant differences in the levels of ghrelin between the tcVNS and sham stimulation groups at either baseline or in the absence of trauma scripts. However, tcVNS in conjunction with personalized traumatic scripts resulted in lower ghrelin levels compared to the sham stimulation group (265.2 ± 143.6 pg/ml vs 478.7 ± 349.2 pg/ml, P = 0.01). Additionally, after completing the public speaking and mental arithmetic tests, ghrelin levels were found to be lower in the group receiving tcVNS compared to the sham group (293.3 ± 102.4 pg/ml vs 540.3 ± 203.9 pg/ml, P = 0.009). LIMITATIONS Timing of ghrelin measurements, and stimulation of only left vagus nerve. CONCLUSION tcVNS decreases ghrelin levels in response to various stressful stimuli. These findings are consistent with a growing literature that tcVNS modulates hormonal and autonomic responses to stress.
Collapse
Affiliation(s)
- Kasra Moazzami
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA, United States of America; Emory Clinical Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America.
| | - Bradley D Pearce
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA, United States of America
| | - Nil Z Gurel
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Matthew T Wittbrodt
- Department of Psychiatry & Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Oleksiy M Levantsevych
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA, United States of America
| | - Minxuan Huang
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA, United States of America
| | - Md Mobashir H Shandhi
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA, United States of America
| | - Isaias Herring
- Department of Psychiatry & Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Nancy Murrah
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA, United States of America
| | - Emily Driggers
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA, United States of America; Department of Psychiatry & Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States of America
| | - MhmtJamil L Alkhalaf
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA, United States of America
| | - Majd Soudan
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA, United States of America
| | - Lucy Shallenberger
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA, United States of America
| | - Allison N Hankus
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA, United States of America
| | - Jonathon A Nye
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Viola Vaccarino
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA, United States of America; Emory Clinical Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Amit J Shah
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA, United States of America; Emory Clinical Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America; Atlanta VA Medical Center, Decatur, GA, United States of America
| | - Omer T Inan
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America; Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - J Douglas Bremner
- Department of Psychiatry & Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States of America; Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States of America; Atlanta VA Medical Center, Decatur, GA, United States of America
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
Heberden C, Maximin E, Rabot S, Naudon L. Male mice engaging differently in emotional eating present distinct plasmatic and neurological profiles. Nutr Neurosci 2023; 26:1034-1044. [PMID: 36154930 DOI: 10.1080/1028415x.2022.2122137] [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: 10/14/2022]
Abstract
Objective: Stressed individuals tend to turn to calorie-rich food, also known as 'comfort food' for the temporary relief it provides. The emotional eating drive is highly variable among subjects. Using a rodent model, we explored the plasmatic and neurobiological differences between 'high and low emotional eaters' (HEE and LEE).Methods: 40 male mice were exposed for 5 weeks to a protocol of unpredictable chronic mild stress. Every 3 or 4 days, they were submitted to a 1-h restraint stress, immediately followed by a 3-h period during which a choice between chow and chocolate sweet cereals was proposed. The dietary intake was measured by weighing. Plasmatic and neurobiological characteristics were compared in mice displaying high vs low intakes.Results: Out of 40 mice, 8 were considered as HEE because of their high post-stress eating score, and 8 as LEE because of their consistent low intake. LEE displayed higher plasma corticosterone and lower levels of NPY than HEE, but acylated and total ghrelin were similar in both groups. In the brain, the abundance of NPY neurons in the arcuate nucleus of the hypothalamus was similar in both groups, but was higher in the ventral hippocampus and the basal lateral amygdala of LEE. The lateral hypothalamus LEE had also more orexin (OX) positive neurons. Both NPY and OX are orexigenic peptides and mood regulators.Discussion: Emotional eating difference was reflected in plasma and brain structures implicated in emotion and eating regulation. These results concur with the psychological side of food consumption.
Collapse
Affiliation(s)
- Christine Heberden
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay Jouy-en-Josas, France
| | - Elise Maximin
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay Jouy-en-Josas, France
| | - Sylvie Rabot
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay Jouy-en-Josas, France
| | - Laurent Naudon
- INRAE, AgroParisTech, CNRS, Micalis Institute, Université Paris-Saclay Jouy-en-Josas, France
| |
Collapse
|
22
|
Smith A, Hyland L, Al-Ansari H, Watts B, Silver Z, Wang L, Dahir M, Akgun A, Telfer A, Abizaid A. Metabolic, neuroendocrine and behavioral effects of social defeat in male and female mice using the chronic non-discriminatory social defeat stress model. Horm Behav 2023; 155:105412. [PMID: 37633226 DOI: 10.1016/j.yhbeh.2023.105412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/28/2023]
Abstract
Stress-related disorders predominately affect females, yet preclinical models of chronic stress exclusively use males especially in models where social stressors are studied. Here, we implemented a 21-day novel social defeat paradigm in which a female and male C57 intruder are simultaneously placed in the cage of a territorial, resident CD-1 male mouse, and the resident proceeds to attack both intruders. Mice were given access to a regular laboratory diet, high in carbohydrates, and a palatable diet, high in fat. Chronic social defeat stress using this paradigm resulted in increased caloric intake in male and female mice, with the effects being more pronounced in females. We observed sex differences in high fat diet intake in response to stress, which was correlated with higher levels of plasma ghrelin observed in female mice but not male mice. Furthermore, females exposed to chronic stress displayed changes in growth hormone secretatogue receptor (ghsr) and neuropeptide-y (npy) expression in the arcuate nucleus of the hypothalamus, potentially increasing ghrelin sensitivity and inducing changes in diet choice and caloric intake. Behavioral results show that females tended to spend more time interacting during the social interaction test, compared to males who displayed higher vigilance towards the stranger mouse. Overall, our results highlight unique neurometabolic alterations in female mice in response to stress that is not present in male mice and may be important for coping with chronic stress and sustaining reproductive function.
Collapse
Affiliation(s)
- Andrea Smith
- Department of Neuroscience and Stress, Trauma and Resilience Work Group (STAR), Carleton University, Ottawa, Ontario, Canada
| | - Lindsay Hyland
- Department of Neuroscience and Stress, Trauma and Resilience Work Group (STAR), Carleton University, Ottawa, Ontario, Canada
| | - Hiyam Al-Ansari
- Department of Neuroscience and Stress, Trauma and Resilience Work Group (STAR), Carleton University, Ottawa, Ontario, Canada
| | - Bethany Watts
- Department of Neuroscience and Stress, Trauma and Resilience Work Group (STAR), Carleton University, Ottawa, Ontario, Canada
| | - Zachary Silver
- Department of Neuroscience and Stress, Trauma and Resilience Work Group (STAR), Carleton University, Ottawa, Ontario, Canada
| | - Longfei Wang
- Department of Neuroscience and Stress, Trauma and Resilience Work Group (STAR), Carleton University, Ottawa, Ontario, Canada
| | - Miski Dahir
- Department of Neuroscience and Stress, Trauma and Resilience Work Group (STAR), Carleton University, Ottawa, Ontario, Canada
| | - Aleyna Akgun
- Department of Neuroscience and Stress, Trauma and Resilience Work Group (STAR), Carleton University, Ottawa, Ontario, Canada
| | - Andre Telfer
- Department of Neuroscience and Stress, Trauma and Resilience Work Group (STAR), Carleton University, Ottawa, Ontario, Canada
| | - Alfonso Abizaid
- Department of Neuroscience and Stress, Trauma and Resilience Work Group (STAR), Carleton University, Ottawa, Ontario, Canada.
| |
Collapse
|
23
|
Wittekind DA, Kratzsch J, Mergl R, Wirkner K, Baber R, Sander C, Witte AV, Villringer A, Kluge M. Childhood sexual abuse is associated with higher total ghrelin serum levels in adulthood: results from a large, population-based study. Transl Psychiatry 2023; 13:219. [PMID: 37349303 DOI: 10.1038/s41398-023-02517-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/24/2023] Open
Abstract
Ghrelin is an orexigenic peptide hormone synthesized in times of stress and hunger and alterations of the ghrelin system following acute stressors could be repeatedly shown in humans. However, little data exists on long-term effects of trauma on the ghrelin system. We aimed to investigate the influence of childhood trauma on total ghrelin serum levels in a large, population-based study. Total serum ghrelin was measured in 1666 participants of a population-based cross-sectional study ('LIFE study'). The Childhood Trauma Screener (CTS) was used for the assessment of childhood trauma in the final sample (n = 1086; mean age: 57.10 ± 16.23 years; 632 males, 454 females). Multiple linear regression analyses and generalized linear models were chosen to examine the association between childhood trauma and total serum ghrelin concentrations. Childhood sexual abuse went along with significantly higher ghrelin serum levels in the total sample (β = 0.114, t = 3.958; p = 0.00008) and in women (β = 0.142, t = 3.115; p = 0.002), but not in men (β = 0.055; t = 1.388; p = 0.166). Women with severe emotional neglect in the childhood had higher ghrelin levels than those without (odds ratio = 1.204; p = 0.018). For the CTS Sum Score and other CTS sub-scale scores, no significant association with ghrelin serum levels was found. Our study is the first to show associations between childhood sexual trauma and total ghrelin levels in adults in a large, community-based sample. Our results should initiate further research of the role of ghrelin in human stress response in prospective study designs.
Collapse
Affiliation(s)
- Dirk Alexander Wittekind
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Leipzig, Germany.
| | - Jürgen Kratzsch
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Leipzig, Germany
| | - Roland Mergl
- Institute of Psychology, University of the Bundeswehr Munich, Neubiberg, Germany
| | - Kerstin Wirkner
- Leipzig Research Center for Civilization Diseases (LIFE), University of Leipzig, Leipzig, Germany
- Institute for Medical Informatics, Statistics and Epidemiology, Leipzig University, Leipzig, Germany
| | - Ronny Baber
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Leipzig, Germany
- Leipzig Research Center for Civilization Diseases (LIFE), University of Leipzig, Leipzig, Germany
| | - Christian Sander
- Department of Psychiatry and Psychotherapy, University of Leipzig, Leipzig, Germany
| | - A Veronica Witte
- Clinic of Cognitive Neurology, University of Leipzig, and Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany
| | - Arno Villringer
- Clinic of Cognitive Neurology, University of Leipzig, and Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany
| | - Michael Kluge
- Department of Psychiatry and Psychotherapy, University of Leipzig, Leipzig, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, Rudolf-Virchow-Hospital, Glauchau, Germany
| |
Collapse
|
24
|
Bhargava R, Luur S, Rodriguez Flores M, Emini M, Prechtl CG, Goldstone AP. Postprandial Increases in Liver-Gut Hormone LEAP2 Correlate with Attenuated Eating Behavior in Adults Without Obesity. J Endocr Soc 2023; 7:bvad061. [PMID: 37287649 PMCID: PMC10243873 DOI: 10.1210/jendso/bvad061] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Indexed: 06/09/2023] Open
Abstract
Background The novel liver-gut hormone liver-expressed antimicrobial peptide-2 (LEAP2) is a centrally acting inverse agonist, and competitive antagonist of orexigenic acyl ghrelin (AG), at the GH secretagogue receptor, reducing food intake in rodents. In humans, the effects of LEAP2 on eating behavior and mechanisms behind the postprandial increase in LEAP2 are unclear, though this is reciprocal to the postprandial decrease in plasma AG. Methods Plasma LEAP2 was measured in a secondary analysis of a previous study. Twenty-two adults without obesity attended after an overnight fast, consuming a 730-kcal meal without or with subcutaneous AG administration. Postprandial changes in plasma LEAP2 were correlated with postprandial changes in appetite, high-energy (HE) or low-energy (LE) food cue reactivity using functional magnetic resonance imaging, ad libitum food intake, and plasma/serum AG, glucose, insulin, and triglycerides. Results Postprandial plasma LEAP2 increased by 24.5% to 52.2% at 70 to 150 minutes, but was unchanged by exogenous AG administration. Postprandial increases in LEAP2 correlated positively with postprandial decreases in appetite, and cue reactivity to HE/LE and HE food in anteroposterior cingulate cortex, paracingulate cortex, frontal pole, and middle frontal gyrus, with similar trend for food intake. Postprandial increases in LEAP2 correlated negatively with body mass index, but did not correlate positively with increases in glucose, insulin, or triglycerides, nor decreases in AG. Conclusions These correlational findings are consistent with a role for postprandial increases in plasma LEAP2 in suppressing human eating behavior in adults without obesity. Postprandial increases in plasma LEAP2 are unrelated to changes in plasma AG and the mediator(s) remain uncertain.
Collapse
Affiliation(s)
- Raghav Bhargava
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Sandra Luur
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Marcela Rodriguez Flores
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Mimoza Emini
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Christina G Prechtl
- School of Public Health, Faculty of Medicine, Imperial College London, St. Mary's Hospital, London, W2 1PG, UK
| | - Anthony P Goldstone
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| |
Collapse
|
25
|
Jin R, Sun S, Hu Y, Zhang H, Sun X. Neuropeptides Modulate Feeding via the Dopamine Reward Pathway. Neurochem Res 2023:10.1007/s11064-023-03954-4. [PMID: 37233918 DOI: 10.1007/s11064-023-03954-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Dopamine (DA) is a catecholamine neurotransmitter widely distributed in the central nervous system. It participates in various physiological functions, such as feeding, anxiety, fear, sleeping and arousal. The regulation of feeding is exceptionally complex, involving energy homeostasis and reward motivation. The reward system comprises the ventral tegmental area (VTA), nucleus accumbens (NAc), hypothalamus, and limbic system. This paper illustrates the detailed mechanisms of eight typical orexigenic and anorexic neuropeptides that regulate food intake through the reward system. According to recent literature, neuropeptides released from the hypothalamus and other brain regions regulate reward feeding predominantly through dopaminergic neurons projecting from the VTA to the NAc. In addition, their effect on the dopaminergic system is mediated by the prefrontal cortex, paraventricular thalamus, laterodorsal tegmental area, amygdala, and complex neural circuits. Research on neuropeptides involved in reward feeding can help identify more targets to treat diseases with metabolic disorders, such as obesity.
Collapse
Affiliation(s)
- Ruijie Jin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Shanbin Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Yang Hu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Hongfei Zhang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xiangrong Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
26
|
Chovel Sella A, Hadaway N, Stern C, Becker KR, Holsen LM, Eddy KT, Micali N, Misra M, Thomas JJ, Lawson EA. Lower Ghrelin Levels Are Associated With Higher Anxiety Symptoms in Adolescents and Young Adults With Avoidant/Restrictive Food Intake Disorder. J Clin Psychiatry 2023; 84:22m14482. [PMID: 37134126 PMCID: PMC10336648 DOI: 10.4088/jcp.22m14482] [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] [Indexed: 05/04/2023]
Abstract
Objective: Avoidant/restrictive food intake disorder (ARFID) is associated with increased risk for anxiety, which may adversely affect prognosis. The appetite-stimulating hormone, ghrelin, increases in response to stress, and exogenous ghrelin decreases anxiety-like behaviors in animal models. The aim of this study was to evaluate the relationship between ghrelin levels and measures of anxiety in youth with ARFID. We hypothesized that lower ghrelin levels would be associated with increased anxiety symptoms. Methods: We studied a cross-sectional sample of 80 subjects with full and subthreshold ARFID diagnosed by DSM-5 criteria, aged 10-23 years (female, n = 39; male, n = 41). Subjects were enrolled in a study of the neurobiology of avoidant/restrictive eating conducted from August 2016 to January 2021. We assessed fasting ghrelin levels and anxiety symptoms (State-Trait Anxiety Inventory [STAI] and STAI for Children [STAI-C] measuring general trait anxiety; Beck Anxiety Inventory [BAI] and BAI for youth [BAI-Y] assessing cognitive, emotional, and somatic symptoms of anxiety; and Liebowitz Social Anxiety Scale [LSAS] assessing symptoms of social anxiety). Results: Consistent with our hypothesis, ghrelin levels were inversely associated with anxiety symptoms as assessed by STAI/STAI-C T scores (r = -0.28, P = .012), BAI/BAI-Y T scores (r = -0.28, P = .010), and LSAS scores (r = -0.3, P = .027), all with medium effect sizes. Findings held in the full threshold ARFID group when adjusting for body mass index z scores (STAI/STAI-C T scores, β = -0.27, P = .024; BAI/BAI-Y T scores, β = -0.26, P = .034; LSAS, β = -0.34, P = .024). Conclusions: These findings demonstrate that lower levels of ghrelin are associated with more severe anxiety symptoms in youth with ARFID and raise the question of whether ghrelin pathways could be targeted in the treatment of ARFID.
Collapse
Affiliation(s)
- Aluma Chovel Sella
- Neuroendocrine Unit, Massachusetts General Hospital, Boston, Massachusetts
- Division of Pediatric Endocrinology, Mass General for Children, Massachusetts General Hospital, Boston, Massachusetts
| | - Natalia Hadaway
- Neuroendocrine Unit, Massachusetts General Hospital, Boston, Massachusetts
| | - Casey Stern
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, Massachusetts
| | - Kendra R Becker
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, Massachusetts
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Laura M Holsen
- Division of Women's Health, Department of Medicine and Department of Psychiatry, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kamryn T Eddy
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, Massachusetts
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Nadia Micali
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- Department of Pediatrics Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Madhusmita Misra
- Neuroendocrine Unit, Massachusetts General Hospital, Boston, Massachusetts
- Division of Pediatric Endocrinology, Mass General for Children, Massachusetts General Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Indicates shared senior authorship
| | - Jennifer J Thomas
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, Massachusetts
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
- Indicates shared senior authorship
| | - Elizabeth A Lawson
- Neuroendocrine Unit, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Corresponding author: Elizabeth A. Lawson, MD, MMSc, Neuroendocrine Unit, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114
- Indicates shared senior authorship
| |
Collapse
|
27
|
Yang L, Shi LJ, Shen SY, Yang JY, Lv SS, Wang ZC, Huang Q, Xu WD, Yu J, Zhang YQ. Toward Antifragility: Social Defeat Stress Enhances Learning and Memory in Young Mice Via Hippocampal Synaptosome Associated Protein 25. Psychol Sci 2023; 34:616-632. [PMID: 37040450 DOI: 10.1177/09567976231160098] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023] Open
Abstract
Social adversity not only causes severe psychological diseases but also may improve people's ability to learn and grow. However, the beneficial effects of social adversity are often ignored. In this study, we investigated whether and how social adversity affects learning and memory in a mouse social defeat stress (SDS) model. A total of 652 mice were placed in experimental groups of six to 23 mice each. SDS enhanced spatial, novelty, and fear memory with increased synaptosome associated protein 25 (SNAP-25) level and dendritic spine density in hippocampal neurons among young but not middle-aged mice. Chemogenetic inhibition of hippocampal CaMK2A+ neurons blocked SDS-induced enhancement of learning or memory. Knockdown of SNAP-25 or blockade of N-methyl-D-aspartate (NMDA) receptor subunit GluN2B in the hippocampus prevented SDS-induced learning memory enhancement in an emotion-independent manner. These findings suggest that social adversity promotes learning and memory ability in youths and provide a neurobiological foundation for biopsychological antifragility.
Collapse
Affiliation(s)
- Liu Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University
| | - Li-Jun Shi
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University
| | - Shi-Yu Shen
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University
| | - Jing-Yan Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University
| | - Su-Su Lv
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University
| | - Zhe-Chen Wang
- Department of Psychology, School of Social Development and Public Policy, Fudan University
- School of Psychology, The University of Queensland
| | - Qian Huang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University
| | - Wen-Dong Xu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University
- Department of Hand Surgery, Huashan Hospital, Fudan University
| | - Jin Yu
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University
| | - Yu-Qiu Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University
| |
Collapse
|
28
|
Kuckuck S, van der Valk ES, Scheurink AJW, van der Voorn B, Iyer AM, Visser JA, Delhanty PJD, van den Berg SAA, van Rossum EFC. Glucocorticoids, stress and eating: The mediating role of appetite-regulating hormones. Obes Rev 2023; 24:e13539. [PMID: 36480471 PMCID: PMC10077914 DOI: 10.1111/obr.13539] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022]
Abstract
Disrupted hormonal appetite signaling plays a crucial role in obesity as it may lead to uncontrolled reward-related eating. Such disturbances can be induced not only by weight gain itself but also by glucocorticoid overexposure, for example, due to chronic stress, disease, or medication use. However, the exact pathways are just starting to be understood. Here, we present a conceptual framework of how glucocorticoid excess may impair hormonal appetite signaling and, consequently, eating control in the context of obesity. The evidence we present suggests that counteracting glucocorticoid excess can lead to improvements in appetite signaling and may therefore pose a crucial target for obesity prevention and treatment. In turn, targeting hormonal appetite signals may not only improve weight management and eating behavior but may also decrease detrimental effects of glucocorticoid excess on cardio-metabolic outcomes and mood. We conclude that gaining a better understanding of the relationship between glucocorticoid excess and circulating appetite signals will contribute greatly to improvements in personalized obesity prevention and treatment.
Collapse
Affiliation(s)
- Susanne Kuckuck
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Obesity Center CGG, Erasmus MC, Room Rg528, P.O. Box 2040, Rotterdam, 3000 CA, Netherlands
| | - Eline S van der Valk
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Obesity Center CGG, Erasmus MC, Room Rg528, P.O. Box 2040, Rotterdam, 3000 CA, Netherlands
| | - Anton J W Scheurink
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| | - Bibian van der Voorn
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Obesity Center CGG, Erasmus MC, Room Rg528, P.O. Box 2040, Rotterdam, 3000 CA, Netherlands
| | - Anand M Iyer
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Obesity Center CGG, Erasmus MC, Room Rg528, P.O. Box 2040, Rotterdam, 3000 CA, Netherlands
| | - Jenny A Visser
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands
| | - Patric J D Delhanty
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands
| | - Sjoerd A A van den Berg
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Department of Clinical Chemistry, Erasmus MC, Rotterdam, Netherlands
| | - Elisabeth F C van Rossum
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Obesity Center CGG, Erasmus MC, Room Rg528, P.O. Box 2040, Rotterdam, 3000 CA, Netherlands
| |
Collapse
|
29
|
Engel JA, Pålsson E, Vallöf D, Jerlhag E. Ghrelin activates the mesolimbic dopamine system via nitric oxide associated mechanisms in the ventral tegmental area. Nitric Oxide 2023; 131:1-7. [PMID: 36513266 DOI: 10.1016/j.niox.2022.12.001] [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: 09/02/2022] [Revised: 11/08/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
Besides enhanced feeding, the orexigenic peptide ghrelin activates the mesolimbic dopamine system to cause reward as measured by locomotor stimulation, dopamine release in nucleus accumbens shell (NAcS), and conditioned place preference. Although the ventral tegmental area (VTA) appears to be a central brain region for this ghrelin-reward, the underlying mechanisms within this area are unknown. The findings that the gaseous neurotransmitter nitric oxide (NO) modulate the ghrelin enhanced feeding, led us to hypothesize that ghrelin increases NO levels in the VTA, and thereby stimulates reward-related behaviors. We initially demonstrated that inhibition of NO synthesis blocked the ghrelin-induced activation of the mesolimbic dopamine system. We then established that antagonism of downstream signaling of NO in the VTA, namely sGC, prevents the ability of ghrelin to stimulate the mesolimbic dopamine system. The association of ghrelin to NO was further strengthened by in vivo electrochemical recordings showing that ghrelin enhances the NO release in the VTA. Besides a GABAB -receptor agonist, known to reduce NO and cGMP, blocks the stimulatory properties of ghrelin. The present series of experiments reveal that ablated NO signaling, through pharmacologically inhibiting the production of NO and/or cGMP, prevents the ability of ghrelin to induced reward-related behaviors.
Collapse
Affiliation(s)
- Jörgen A Engel
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Erik Pålsson
- Institute of Neuroscience and Physiology, Department of Neurochemistry and Psychiatry, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Daniel Vallöf
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
30
|
Impaired Ghrelin Signaling Does Not Lead to Alterations of Anxiety-like Behaviors in Adult Mice Chronically Exposed to THC during Adolescence. Biomedicines 2023; 11:biomedicines11010144. [PMID: 36672651 PMCID: PMC9855766 DOI: 10.3390/biomedicines11010144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/17/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
As marijuana use during adolescence has been increasing, the need to understand the effects of its long-term use becomes crucial. Previous research suggested that marijuana consumption during adolescence increases the risk of developing mental illnesses, such as schizophrenia, depression, and anxiety. Ghrelin is a peptide produced primarily in the gut and is important for feeding behavior. Recent studies have shown that ghrelin and its receptor, the growth hormone secretagogue receptor (GHSR), play important roles in mediating stress, as well as anxiety and depression-like behaviors in animal models. Here, we investigated the effects of chronic tetrahydrocannabinol (THC) administration during late adolescence (P42-55) in GHSR (GHSR -/-) knockout mice and their wild-type littermates in relation to anxiety-like behaviors. We determined that continuous THC exposure during late adolescence did not lead to any significant alterations in the anxiety-like behaviors of adult mice, regardless of genotype, following a prolonged period of no exposure (1 month). These data indicate that in the presence of intact or impaired ghrelin/GHSR signaling, THC exposure during late adolescence has limited if any long-term impact on anxiety-like behaviors in mice.
Collapse
|
31
|
Tian J, Du E, Jia K, Wang T, Guo L, Zigman JM, Du H. Elevated Ghrelin Promotes Hippocampal Ghrelin Receptor Defects in Humanized Amyloid-β Knockin Mice During Aging. J Alzheimers Dis 2023; 96:1579-1592. [PMID: 38007666 PMCID: PMC10841720 DOI: 10.3233/jad-231002] [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] [Indexed: 11/27/2023]
Abstract
BACKGROUND Emerging evidence has revealed that dysregulation of the hormone ghrelin and its receptor, growth hormone secretagogue receptor (GHSR), contributes to the pathogenesis of Alzheimer's disease (AD). Specifically, defective GHSR function and resultant hippocampal ghrelin resistance are linked to hippocampal synaptic injury in AD paradigms. Also, AD patients exhibit elevated ghrelin activation. However, the detailed molecular mechanisms of hippocampal GHSR dysfunction and the relevance of ghrelin elevation to hippocampal ghrelin resistance in AD-relevant pathological settings are not fully understood. OBJECTIVE In the current study, we employed a recently established mouse line of AD risk [humanized amyloid beta knockin (hAβ KI mice), also referred to as a mouse model of late-onset AD in previous literature] to further define the role of ghrelin system dysregulation in the development of AD. METHODS We employed multidisciplinary techniques to determine the change of plasma ghrelin and the functional status of GHSR in hAβ KI mice as well as primary neuron cultures. RESULTS We observed concurrent plasma ghrelin elevation and hippocampal GHSR desensitization with disease progression. Further examination excluded the possibility that ghrelin elevation is a compensatory change in response to GHSR dysfunction. In contrast, further in vitro and in vivo results show that agonist-mediated overstimulation potentiates GHSR desensitization through enhanced GHSR internalization. CONCLUSIONS These findings suggest that circulating ghrelin elevation is a pathological event underlying hippocampal GHSR dysfunction, culminating in hippocampal ghrelin resistance and resultant synaptic injury in late-onset AD-related settings.
Collapse
Affiliation(s)
- Jing Tian
- Department of Pharmacology and Toxicology, The University of Kansas, Lawrence, KS, USA
| | - Eric Du
- Department of Pharmacology and Toxicology, The University of Kansas, Lawrence, KS, USA
- Blue Valley West High School, Overland Park, KS, USA
| | - Kun Jia
- Department of Pharmacology and Toxicology, The University of Kansas, Lawrence, KS, USA
| | - Tienju Wang
- Department of Pharmacology and Toxicology, The University of Kansas, Lawrence, KS, USA
| | - Lan Guo
- Department of Pharmacology and Toxicology, The University of Kansas, Lawrence, KS, USA
| | - Jeffrey M. Zigman
- Department of Internal Medicine, Center for Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Heng Du
- Department of Pharmacology and Toxicology, The University of Kansas, Lawrence, KS, USA
- Alzheimer’s disease Research Center (ADRC), Department of Neurology, The University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
32
|
Wang CH, Tseng CY, Hsu WL, Tzen JTC. Establishment of a Cell Line Stably Expressing the Growth Hormone Secretagogue Receptor to Identify Crocin as a Ghrelin Agonist. Biomolecules 2022; 12:biom12121813. [PMID: 36551241 PMCID: PMC9775697 DOI: 10.3390/biom12121813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
The growth hormone secretagogue receptor-1a (GHSR1a) is the endogenous receptor for ghrelin. Activation of GHSR1a participates in many physiological processes including energy homeostasis and eating behavior. Due to its transitory half-life, the efficacy of ghrelin treatment in patients is restricted; hence the development of new adjuvant therapy is an urgent need. This study aimed to establish a cell line stably expressing GHSR1a, which could be employed to screen potential ghrelin agonists from natural compounds. First, by means of lentiviral transduction, the genome of a human HEK293T cell was modified, and a cell platform stably overexpressing GHSR1a was successfully established. In this platform, GHSR1a was expressed as a fusion protein tagged with mCherry, which allowed the monitoring of the dynamic cellular distribution of GHSR1a by fluorescent microscopy. Subsequently, the authenticity of the GHSR1a mediated signaling was further characterized by using ghrelin and teaghrelin, two molecules known to stimulate GHSR1a. The results indicated that both ghrelin and teaghrelin readily activated GHSR1a mediated signaling pathways, presumably via increasing phosphorylation levels of ERK. The specific GHSR1a signaling was further validated by using SP-analog, an antagonist of GHSR1a as well as using a cell model with the knockdown expression of GHSR1a. Molecular modeling predicted that crocin might be a potential ghrelin agonist, and this prediction was further confirmed by the established platform.
Collapse
Affiliation(s)
- Chia-Hao Wang
- Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung 402, Taiwan
| | - Ching-Yu Tseng
- Graduate Institute of Microbiology and Public Health, National Chung-Hsing University, Taichung 402, Taiwan
| | - Wei-Li Hsu
- Graduate Institute of Microbiology and Public Health, National Chung-Hsing University, Taichung 402, Taiwan
- Correspondence: (W.-L.H.); (J.T.C.T.)
| | - Jason T. C. Tzen
- Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung 402, Taiwan
- Correspondence: (W.-L.H.); (J.T.C.T.)
| |
Collapse
|
33
|
Linders LE, Patrikiou L, Soiza-Reilly M, Schut EHS, van Schaffelaar BF, Böger L, Wolterink-Donselaar IG, Luijendijk MCM, Adan RAH, Meye FJ. Stress-driven potentiation of lateral hypothalamic synapses onto ventral tegmental area dopamine neurons causes increased consumption of palatable food. Nat Commun 2022; 13:6898. [PMID: 36371405 PMCID: PMC9653441 DOI: 10.1038/s41467-022-34625-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 11/01/2022] [Indexed: 11/13/2022] Open
Abstract
Stress can cause overconsumption of palatable high caloric food. Despite the important role of stress eating in obesity and (binge) eating disorders, its underlying neural mechanisms remain unclear. Here we demonstrate in mice that stress alters lateral hypothalamic area (LHA) control over the ventral tegmental area (VTA), thereby promoting overconsumption of palatable food. Specifically, we show that glutamatergic LHA neurons projecting to the VTA are activated by social stress, after which their synapses onto dopamine neurons are potentiated via AMPA receptor subunit alterations. We find that stress-driven strengthening of these specific synapses increases LHA control over dopamine output in key target areas like the prefrontal cortex. Finally, we demonstrate that while inducing LHA-VTA glutamatergic potentiation increases palatable fat intake, reducing stress-driven potentiation of this connection prevents such stress eating. Overall, this study provides insights in the neural circuit adaptations caused by stress that drive overconsumption of palatable food.
Collapse
Affiliation(s)
- Louisa E. Linders
- grid.5477.10000000120346234Department of Translational Neuroscience, Brain Center, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Lefkothea Patrikiou
- grid.5477.10000000120346234Department of Translational Neuroscience, Brain Center, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mariano Soiza-Reilly
- grid.7345.50000 0001 0056 1981Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET, University of Buenos Aires, Buenos Aires, Argentina
| | - Evelien H. S. Schut
- grid.5477.10000000120346234Department of Translational Neuroscience, Brain Center, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Bram F. van Schaffelaar
- grid.5477.10000000120346234Department of Translational Neuroscience, Brain Center, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Leonard Böger
- grid.5477.10000000120346234Department of Translational Neuroscience, Brain Center, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Inge G. Wolterink-Donselaar
- grid.5477.10000000120346234Department of Translational Neuroscience, Brain Center, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mieneke C. M. Luijendijk
- grid.5477.10000000120346234Department of Translational Neuroscience, Brain Center, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Roger A. H. Adan
- grid.5477.10000000120346234Department of Translational Neuroscience, Brain Center, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Frank J. Meye
- grid.5477.10000000120346234Department of Translational Neuroscience, Brain Center, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
34
|
Wang M, Wang X, Jiang B, Zhai Y, Zheng J, Yang L, Tai X, Li Y, Fu S, Xu J, Lei X, Kuang Z, Zhang C, Bai X, Li M, Zan T, Qu S, Li Q, Zhang C. Identification of MRAP protein family as broad-spectrum GPCR modulators. Clin Transl Med 2022; 12:e1091. [PMID: 36314066 PMCID: PMC9619224 DOI: 10.1002/ctm2.1091] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The melanocortin receptor accessory proteins (MRAP1 and MRAP2) are well-known endocrine regulators for the trafficking and signalling of all five melanocortin receptors (MC1R-MC5R). The observation of MRAP2 on regulating several non-melanocortin G protein-coupled receptors (GPCRs) has been sporadically reported, whereas other endogenous GPCR partners of the MRAP protein family are largely unknown. METHODS Here, we performed single-cell transcriptome analysis and drew a fine GPCR blueprint and MRAPs-associated network of two major endocrine organs, the hypothalamus and adrenal gland at single-cell resolution. We also integrated multiple bulk RNA-seq profiles and single-cell datasets of human and mouse tissues, and narrowed down a list of 48 GPCRs with strong endogenous co-expression correlation with MRAPs. RESULTS 36 and 46 metabolic-related GPCRs were consequently identified as novel interacting partners of MRAP1 or MRAP2, respectively. MRAPs exhibited protein-protein interactions and varying pharmacological properties on the surface translocation, constitutive activities and ligand-stimulated downstream signalling of these GPCRs. Knockdown of MRAP2 expression by hypothalamic administration of adeno-associated virus (AAV) packed shRNA stimulated body weight gain in mouse model. Co-injection of corticotropinreleasing factor (CRF), the agonist of corticotropin releasing hormone receptor 1 (CRHR1), suppressed feeding behaviour in a MRAP2-dependent manner. CONCLUSIONS Collectively, our study has comprehensively elucidated the complex GPCR networks in two major endocrine organs and redefined the MRAP protein family as broad-spectrum GPCR modulators. MRAP proteins not only serve as a vital endocrine pivot on the regulation of global GPCR activities in vivo that could explain the composite physiological phenotypes of the MRAP2 null murine model but also provide us with new insights of the phenotyping investigation of GPCR-MRAP functional complexes.
Collapse
Affiliation(s)
- Meng Wang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaozhu Wang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bopei Jiang
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yue Zhai
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Jihong Zheng
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Liu Yang
- Department of Endocrinology and MetabolismNational Metabolic Management CenterShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
| | - Xiaolu Tai
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yunpeng Li
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Shaliu Fu
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Jing Xu
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Xiaowei Lei
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Zhe Kuang
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Cong Zhang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xuanxuan Bai
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Mingyu Li
- Fujian Provincial Key Laboratory of Innovative Drug Target ResearchSchool of Pharmaceutical SciencesXiamen UniversityXiamenChina
| | - Tao Zan
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shen Qu
- Department of Endocrinology and MetabolismNational Metabolic Management CenterShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
| | - Qingfeng Li
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chao Zhang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
35
|
Bales MB, Centanni SW, Luchsinger JR, Fathi P, Biddinger JE, Le TDV, Nwaba KG, Paldrmic IM, Winder DG, Ayala JE. High fat diet blunts stress-induced hypophagia and activation of Glp1r dorsal lateral septum neurons in male but not in female mice. Mol Metab 2022; 64:101571. [PMID: 35953023 PMCID: PMC9418981 DOI: 10.1016/j.molmet.2022.101571] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 07/19/2022] [Accepted: 08/02/2022] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE While stress typically reduces caloric intake (hypophagia) in chow-fed rodents, presentation of palatable, high calorie substances during stress can increase caloric consumption (i.e. "comfort feeding") and promote obesity. However, little is known about how obesity itself affects feeding behavior in response to stress and the mechanisms that can influence stress-associated feeding in the context of obesity. METHODS We assessed food intake and other metabolic parameters in lean and obese male and female mice following acute restraint stress. We also measured real-time activity of glucagon-like peptide-1 (Glp1) receptor (Glp1r)-expressing neurons in the dorsal lateral septum (dLS) during stress in lean and obese mice using fiber photometry. Glp1r activation in various brain regions, including the dLS, promotes hypophagia in response to stress. Finally, we used inhibitory Designer Receptors Activated Exclusively by Designer Drugs (DREADDs) to test whether activation of Glp1r-expressing neurons in the LS is required for stress-induced hypophagia. RESULTS Lean male mice display the expected hypophagic response following acute restraint stress, but obese male mice are resistant to this acute stress-induced hypophagia. Glp1r-positive neurons in the dLS are robustly activated during acute restraint stress in lean but not in obese male mice. This raises the possibility that activation of dLS Glp1r neurons during restraint stress contributes to subsequent hypophagia. Supporting this, we show that chemogenetic inhibition of LS Glp1r neurons attenuates acute restraint stress hypophagia in male mice. Surprisingly, we show that both lean and obese female mice are resistant to acute restraint stress-induced hypophagia and activation of dLS Glp1r neurons. CONCLUSIONS These results suggest that dLS Glp1r neurons contribute to the hypophagic response to acute restraint stress in male mice, but not in female mice, and that obesity disrupts this response in male mice. Broadly, these findings show sexually dimorphic mechanisms and feeding behaviors in lean vs. obese mice in response to acute stress.
Collapse
Affiliation(s)
- Michelle B Bales
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Samuel W Centanni
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Joseph R Luchsinger
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Payam Fathi
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Jessica E Biddinger
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Thao D V Le
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Kaitlyn Ginika Nwaba
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Isabella M Paldrmic
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Danny G Winder
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Julio E Ayala
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA; Vanderbilt Mouse Metabolic Phenotyping Center, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA.
| |
Collapse
|
36
|
Involvement of the ghrelin system in the maintenance and reinstatement of cocaine-motivated behaviors: a role of adrenergic action at peripheral β1 receptors. Neuropsychopharmacology 2022; 47:1449-1460. [PMID: 34923576 PMCID: PMC9206024 DOI: 10.1038/s41386-021-01249-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/22/2021] [Accepted: 11/28/2021] [Indexed: 12/30/2022]
Abstract
Cocaine addiction is a significant medical and public concern. Despite decades of research effort, development of pharmacotherapy for cocaine use disorder remains largely unsuccessful. This may be partially due to insufficient understanding of the complex biological mechanisms involved in the pathophysiology of this disorder. In the present study, we show that: (1) elevation of ghrelin by cocaine plays a critical role in maintenance of cocaine self-administration and cocaine-seeking motivated by cocaine-conditioned stimuli; (2) acquisition of cocaine-taking behavior is associated with the acquisition of stimulatory effects of cocaine by cocaine-conditioned stimuli on ghrelin secretion, and with an upregulation of ghrelin receptor mRNA levels in the ventral tegmental area (VTA); (3) blockade of ghrelin signaling by pretreatment with JMV2959, a selective ghrelin receptor antagonist, dose-dependently inhibits reinstatement of cocaine-seeking triggered by either cocaine or yohimbine in behaviorally extinguished animals with a history of cocaine self-administration; (4) JMV2959 pretreatment also inhibits brain stimulation reward (BSR) and cocaine-potentiated BSR maintained by optogenetic stimulation of VTA dopamine neurons in DAT-Cre mice; (5) blockade of peripheral adrenergic β1 receptors by atenolol potently attenuates the elevation in circulating ghrelin induced by cocaine and inhibits cocaine self-administration and cocaine reinstatement triggered by cocaine. These findings demonstrate that the endogenous ghrelin system plays an important role in cocaine-related addictive behaviors and suggest that manipulating and targeting this system may be viable for mitigating cocaine use disorder.
Collapse
|
37
|
Wittekind DA, Kratzsch J, Mergl R, Riedel-Heller S, Witte AV, Villringer A, Kluge M. Serum ghrelin is positively associated with physiological anxiety but negatively associated with pathological anxiety in humans: Data from a large community-based study. Psychoneuroendocrinology 2022; 140:105728. [PMID: 35305404 DOI: 10.1016/j.psyneuen.2022.105728] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/05/2022] [Accepted: 03/11/2022] [Indexed: 12/19/2022]
Abstract
The orexigenic hormone ghrelin is being increasingly recognized as a stress hormone being involved in anxiety regulation. In animals, ghrelin effects on, and responses to acute stress differed from those in chronic stress, an animal model for anxiety and depression. In humans, elevated ghrelin levels were reported in pathological anxiety (e.g. panic disorder). However, no reports exist on physiological anxiety in mentally healthy subjects. In addition, reports on generalized anxiety symptoms, both in mentally healthy subjects (e.g. worrying) or in adult patients, are lacking. Total serum ghrelin was determined in 1666 subjects of a population-based cross-sectional study ('LIFE'). The 7-item Generalized Anxiety Disorder Scale (GAD-7), detecting also other anxiety disorders, was administered. For multiple linear regression analyses, 1091 subjects were finally included. Serum ghrelin and GAD-7 scores were positively but not significantly associated in the total group (ß=0.00025, standardized β = 0.039, 95%CI: -0.00006;0.0006;p = 0.144), in subjects with no more than mild anxiety, there was a significant positive association (GAD-7 ≤9: n = 1061, 97.25%, β = 0.00032; standardized β = 0.060; 95%CI: 0.000023;0.00062;p = 0.036). In contrast, there was a negative association in subjects with anxiety symptoms above the GAD-7 cut-off (GAD-7 ≥10: n = 30, 2.75%, ß=-0.003, standardized β = -0.462; 95% CI:-0.006;0.0001;p = 0.045). Ghrelin levels were only numerically (p = 0.23) higher in subjects with clinically relevant anxiety symptoms (963.5 ± 399.6 pg/ml; mean±SD) than in those without (901.0 ± 416.4 pg/ml). In conclusion, the positive association between ghrelin and no more than mild anxiety is an initial indication for a role for ghrelin in the regulation of physiological anxiety in humans. This association and the opposed association in pathological anxiety resemble findings in animals showing diverging ghrelin effects in acute and chronic stress.
Collapse
Affiliation(s)
| | - Jürgen Kratzsch
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Leipzig, Germany
| | - Roland Mergl
- Institute of Psychology, Universität der Bundeswehr München, Neubiberg, Germany
| | - Steffi Riedel-Heller
- Faculty of Medicine, Institute of Social Medicine, Occupational Health and Public Health, University of Leipzig, Leipzig, Germany
| | - A Veronica Witte
- Clinic of Cognitive Neurology, University of Leipzig, and Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany
| | - Arno Villringer
- Clinic of Cognitive Neurology, University of Leipzig, and Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany
| | - Michael Kluge
- Department of Psychiatry and Psychotherapy, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
38
|
Fernandez G, Cabral A, De Francesco PN, Uriarte M, Reynaldo M, Castrogiovanni D, Zubiría G, Giovambattista A, Cantel S, Denoyelle S, Fehrentz JA, Tolle V, Schiöth HB, Perello M. GHSR controls food deprivation-induced activation of CRF neurons of the hypothalamic paraventricular nucleus in a LEAP2-dependent manner. Cell Mol Life Sci 2022; 79:277. [PMID: 35504998 PMCID: PMC11072678 DOI: 10.1007/s00018-022-04302-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Prolonged fasting is a major challenge for living organisms. An appropriate metabolic response to food deprivation requires the activation of the corticotropin-releasing factor-producing neurons of the hypothalamic paraventricular nucleus (PVHCRF neurons), which are a part of the hypothalamic-pituitary-adrenal axis (HPA), as well as the growth hormone secretagogue receptor (GHSR) signaling, whose activity is up- or down-regulated, respectively, by the hormones ghrelin and the liver-expressed antimicrobial peptide 2 (LEAP2). Since ghrelin treatment potently up-regulates the HPA axis, we studied the role of GHSR in mediating food deprivation-induced activation of the PVHCRF neurons in mice. METHODS We estimated the activation of the PVHCRF neurons, using immuno-staining against CRF and the marker of neuronal activation c-Fos in brain sections, and assessed plasma levels of corticosterone and glucose in different pharmacologically or genetically manipulated mouse models exposed, or not, to a 2-day food deprivation protocol. In particular, we investigated ad libitum fed or food-deprived male mice that: (1) lacked GHSR gene expression, (2) had genetic deletion of the ghrelin gene, (3) displayed neurotoxic ablation of the hypothalamic arcuate nucleus, (4) were centrally treated with an anti-ghrelin antibody to block central ghrelin action, (5) were centrally treated with a GHSR ligand that blocks ghrelin-evoked and constitutive GHSR activities, or (6) received a continuous systemic infusion of LEAP2(1-12). RESULTS We found that food deprivation results in the activation of the PVHCRF neurons and in a rise of the ghrelin/LEAP2 molar ratio. Food deprivation-induced activation of PVHCRF neurons required the presence and the signaling of GHSR at hypothalamic level, but not of ghrelin. Finally, we found that preventing the food deprivation-induced fall of LEAP2 reverses the activation of the PVHCRF neurons in food-deprived mice, although it has no effect on body weight or blood glucose. CONCLUSION Food deprivation-induced activation of the PVHCRF neurons involves ghrelin-independent actions of GHSR at hypothalamic level and requires a decrease of plasma LEAP2 levels. We propose that the up-regulation of the actions of GHSR associated to the fall of plasma LEAP2 level are physiologically relevant neuroendocrine signals during a prolonged fasting.
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 (UNLP)], Calle 526 S/N entre 10 y 11, 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 (UNLP)], Calle 526 S/N entre 10 y 11, 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 (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina
| | - Maia Uriarte
- 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 (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina
| | - Mirta Reynaldo
- 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 (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina
| | - Daniel Castrogiovanni
- Cell Culture Facility, 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 (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina
| | - Guillermina Zubiría
- Laboratory of Neuroendocrinology, 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 (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina
| | - Andrés Giovambattista
- Laboratory of Neuroendocrinology, 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 (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina
| | - Sonia Cantel
- Institut Des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM, Montpellier, France
| | - Severine Denoyelle
- Institut Des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut Des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM, Montpellier, France
| | - Virginie Tolle
- Institute of Psychiatry and Neuroscience of Paris, Université de Paris, UMR-S 1266 INSERM, Paris, France
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
- Institute for Translational Medicine and Biotechnology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - 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 (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina.
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
39
|
Schwenzer C, Voelz C, Kogel V, Schlösser A, Herpertz‐Dahlmann B, Beyer C, Seitz J, Trinh S. Fear and food: Anxiety-like behavior and the susceptibility to weight loss in an activity-based anorexia rat model. Clin Transl Sci 2022; 15:889-898. [PMID: 34793620 PMCID: PMC9010269 DOI: 10.1111/cts.13196] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/19/2021] [Accepted: 11/01/2021] [Indexed: 12/31/2022] Open
Abstract
Anorexia nervosa (AN) is a severe psychiatric disorder characterized by energy restriction, low body weight, a fear of gaining weight, and often excessive physical activity. Anxiety disorders appear to constitute a major risk factor for developing AN and are the most frequent comorbidity. Here, the influence of anxiety-like behavior prior to food restriction on increased physical activity, leading to greater susceptibility to weight loss, was tested in rats. Furthermore, the possible anxiolytic effect of starvation itself was analyzed. A chronic starvation model activity-based anorexia (ABA) was applied to mimic physiological and behavioral characteristics of AN. During the induction of starvation and acute starvation, food intake was reduced by 70% and the rats lost 25% of their body weight, which was kept stable to imitate chronic starvation. Anxiety-like behavior was quantified before and after chronic starvation using the elevated plus maze, based on rodents' aversion to open spaces. Anxiety-related behavior before food restriction was associated with increased running-wheel activity during habituation and during the induction of starvation, and predicted faster weight loss in ABA rats. Additionally, food-restricted animals showed less anxiety-like behavior after chronic starvation. Animals showing more anxiety-like behavior appear to be more susceptible to weight loss, partially mediated by increased physical activity. Anxiety-related behavior was associated with increased physical activity, which in turn was associated with more rapid weight loss. Our data let us assume that food restriction has an anxiolytic effect. These findings demonstrate the importance of considering anxiety disorders in patients with AN.
Collapse
Affiliation(s)
| | - Clara Voelz
- Institute of NeuroanatomyRWTH Aachen UniversityAachenGermany
| | - Vanessa Kogel
- Institute of NeuroanatomyRWTH Aachen UniversityAachenGermany
| | - Anna Schlösser
- Institute of NeuroanatomyRWTH Aachen UniversityAachenGermany
| | - Beate Herpertz‐Dahlmann
- Department of Child and Adolescent Psychiatry, Psychosomatics and PsychotherapyRWTH Aachen UniversityAachenGermany
| | - Cordian Beyer
- Institute of NeuroanatomyRWTH Aachen UniversityAachenGermany
| | - Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychosomatics and PsychotherapyRWTH Aachen UniversityAachenGermany
| | - Stefanie Trinh
- Institute of NeuroanatomyRWTH Aachen UniversityAachenGermany
| |
Collapse
|
40
|
Abdel Aziz K, Al-Mugaddam F, Sugathan S, Saseedharan P, Jouini T, Elamin ME, Moselhy H, Aly El-Gabry D, Arnone D, Karam SM. Decreased acylated and total ghrelin levels in bipolar disorder patients recovering from a manic episode. BMC Psychiatry 2022; 22:209. [PMID: 35313855 PMCID: PMC8935687 DOI: 10.1186/s12888-022-03842-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/02/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND To date, only few studies have investigated ghrelin levels in bipolar disorders, and all have exclusively measured acylated ghrelin, with none investigating total ghrelin (acylated and des-acylated). We aimed to investigate peripheral levels of acylated and total ghrelin in subjects experiencing a manic episode of bipolar disorder. METHODS Peripheral levels of acylated and total ghrelin were measured in hospitalised medicated individuals recovering from a manic episode. Enzyme-linked immunosorbent assays (ELISA) were used to measure ghrelin levels in patients and compared with healthy controls. The relationship between ghrelin levels in bipolar disorder, self-reported hunger measures, demographic and clinical parameters was investigated with correlational analyses. RESULTS Twenty-four subjects (15 males, 9 females) recovering from mania and 27 matched healthy controls (13 males, 14 females) were recruited for the study. Mean values of both acylated (187 vs.520 pg/mL) and total ghrelin (396 vs. 648 pg/mL) were significantly reduced in bipolar disorder (p = 0.001). Ghrelin levels correlated positively with markers of illness severity and negatively with prescribed mood stabilizers, second-generation antipsychotics, weight and body mass index. CONCLUSION Peripheral measurements of acylated and total ghrelin were both reduced in bipolar disorder patients compared to healthy controls. Whilst illness severity promotes higher ghrelin levels, pharmacological treatment and weight gain exercise the opposite effect.
Collapse
Affiliation(s)
- Karim Abdel Aziz
- Department of Psychiatry and Behavioural Sciences, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Fadwa Al-Mugaddam
- Department of Psychiatry and Behavioural Sciences, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Subi Sugathan
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Prashanth Saseedharan
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- Dept of Chemical Engineering and Biotechnology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Tarek Jouini
- Behavioural Science Institute, Al-Ain Hospital, Al-Ain, United Arab Emirates
| | | | - Hamdy Moselhy
- American Center for Psychiatry and Neurology (ACPN), Dubai, United Arab Emirates
| | - Dina Aly El-Gabry
- Okasha Institute of Psychiatry, Neuropsychiatry Department, Ain Shams University, Cairo, Egypt
| | - Danilo Arnone
- Department of Psychiatry and Behavioural Sciences, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.
- Institute of Psychiatry, Psychology and Neuroscience, Centre for Affective Disorders, Kings' College London, London, UK.
| | - Sherif M Karam
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
41
|
Crandall AK, McKay NJ, Khan AM, Lantyer MC, Temple JL. The effect of acute and chronic scarcity on acute stress: A dyadic developmental examination. Physiol Behav 2022; 246:113684. [PMID: 34929257 PMCID: PMC8821326 DOI: 10.1016/j.physbeh.2021.113684] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Food insecurity, obesity, and psychological stress are interrelated constructs which are thought to be connected through increased energy intake, but the underlying mechanisms for these relationships remain unclear. The current study used experimental methods to investigate how financial losses may influence acute stress in the context of food insecurity for both parents and offspring. This study also sought to examine the effect of acute stress related to financial losses on the reinforcing value of food (RRVfood) and delay discounting (DD). METHODS One hundred and six families stratified by both offspring age (53 children aged 7-10, 53 adolescents aged 15-17) and household financial resources, visited our laboratory for three separate appointments. Each appointment included the experimental manipulation of financial gains and losses, saliva samples for cortisol assay, continuous heart rate monitoring, self-rated tension, and computer-based DD and RRVfood tasks. Participants also completed surveys to report perceived life stress level and food insecurity status. RESULTS Among all participants, financial losses were related to decreased heart rates and increased self-rated tension. Among parents reporting food insecurity, acute financial losses resulted in an increase in cortisol levels. Changes in cortisol, heart rate, and tension were not related to RRVfood or DD. CONCLUSION Food insecure parents are sensitive to financial losses and respond with an increase in cortisol. However, we found no evidence for a relationship between cortisol and RRVfood or DD. This sensitivity to financial losses did not extend to children or adolescents.
Collapse
Affiliation(s)
- Amanda K Crandall
- Department of Community Health and Health Behavior, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY 14214.
| | - Naomi J McKay
- Department of Psychology, SUNY Buffalo State, Buffalo, NY 14222
| | - Ali M Khan
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY 14214
| | - Maria Catharina Lantyer
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY 14214
| | - Jennifer L Temple
- Department of Community Health and Health Behavior, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY 14214; Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY 14214
| |
Collapse
|
42
|
Chen X, Dong J, Jiao Q, Du X, Bi M, Jiang H. "Sibling" battle or harmony: crosstalk between nesfatin-1 and ghrelin. Cell Mol Life Sci 2022; 79:169. [PMID: 35239020 PMCID: PMC11072372 DOI: 10.1007/s00018-022-04193-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/17/2022]
Abstract
Ghrelin was first identified as an endogenous ligand of the growth hormone secretagogue receptor (GHSR) in 1999, with the function of stimulating the release of growth hormone (GH), while nesfatin-1 was identified in 2006. Both peptides are secreted by the same kind of endocrine cells, X/A-like cells in the stomach. Compared with ghrelin, nesfatin-1 exerts opposite effects on energy metabolism, glucose metabolism, gastrointestinal functions and regulation of blood pressure, but exerts similar effects on anti-inflammation and neuroprotection. Up to now, nesfatin-1 remains as an orphan ligand because its receptor has not been identified. Several studies have shown the effects of nesfatin-1 are dependent on the receptor of ghrelin. We herein compare the effects of nesfatin-1 and ghrelin in several aspects and explore the possibility of their interactions.
Collapse
Affiliation(s)
- Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Jing Dong
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Mingxia Bi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China.
| |
Collapse
|
43
|
Woodward ORM, Gribble FM, Reimann F, Lewis JE. Gut peptide regulation of food intake - evidence for the modulation of hedonic feeding. J Physiol 2022; 600:1053-1078. [PMID: 34152020 DOI: 10.1113/jp280581] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
The number of people living with obesity has tripled worldwide since 1975 with serious implications for public health, as obesity is linked to a significantly higher chance of early death from associated comorbidities (metabolic syndrome, type 2 diabetes, cardiovascular disease and cancer). As obesity is a consequence of food intake exceeding the demands of energy expenditure, efforts are being made to better understand the homeostatic and hedonic mechanisms governing food intake. Gastrointestinal peptides are secreted from enteroendocrine cells in response to nutrient and energy intake, and modulate food intake either via afferent nerves, including the vagus nerve, or directly within the central nervous system, predominantly gaining access at circumventricular organs. Enteroendocrine hormones modulate homeostatic control centres at hypothalamic nuclei and the dorso-vagal complex. Additional roles of these peptides in modulating hedonic food intake and/or preference via the neural systems of reward are starting to be elucidated, with both peripheral and central peptide sources potentially contributing to central receptor activation. Pharmacological interventions and gastric bypass surgery for the treatment of type 2 diabetes and obesity elevate enteroendocrine hormone levels and also alter food preference. Hence, understanding of the hedonic mechanisms mediated by gut peptide action could advance development of potential therapeutic strategies for the treatment of obesity and its comorbidities.
Collapse
Affiliation(s)
- Orla R M Woodward
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Fiona M Gribble
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Frank Reimann
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Jo E Lewis
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| |
Collapse
|
44
|
Lugilde J, Casado S, Beiroa D, Cuñarro J, Garcia-Lavandeira M, Álvarez CV, Nogueiras R, Diéguez C, Tovar S. LEAP-2 Counteracts Ghrelin-Induced Food Intake in a Nutrient, Growth Hormone and Age Independent Manner. Cells 2022; 11:cells11030324. [PMID: 35159134 PMCID: PMC8834077 DOI: 10.3390/cells11030324] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 12/30/2022] Open
Abstract
Data gleaned recently shows that ghrelin, a stomach derived peptide, and liver-expressed-antimicrobial peptide 2 (LEAP-2) play opposite roles on food intake. However, the data available with LEAP-2 in relation to in vivo studies are still very scanty and some key questions regarding the interplay among ghrelin and LEAP-2 remain to be answered. In this work, using rats and mice, we study fasting-induced food intake as well as testing the effect of diet exposure, e.g., standard diet and high fat diet, in terms of ghrelin-induced food intake. The anorexigenic effect of LEAP-2 on fasting induced food intake appears to be dependent on energy stores, being more evident in ob/ob than in wild type mice and also in animals exposed to high fat diet. On the other hand, LEAP-2 administration markedly inhibited ghrelin-induced food intake in lean, obese (ob/ob and DIO) mice, aged rats and GH-deficient dwarf rats. In contrast, the inhibitory effect on glucose levels can only be observed in some specific experimental models indicating that the mechanisms involved are likely to be quite different. Taken together from these data, LEAP-2 emerged as a potential candidate to be therapeutically useful in obesity.
Collapse
Affiliation(s)
- Javier Lugilde
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (J.L.); (S.C.); (D.B.); (J.C.); (R.N.)
| | - Sabela Casado
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (J.L.); (S.C.); (D.B.); (J.C.); (R.N.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Daniel Beiroa
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (J.L.); (S.C.); (D.B.); (J.C.); (R.N.)
| | - Juan Cuñarro
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (J.L.); (S.C.); (D.B.); (J.C.); (R.N.)
| | - Montserrat Garcia-Lavandeira
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), 15782 Santiago de Compostela, Spain; (M.G.-L.); (C.V.Á.)
| | - Clara V. Álvarez
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), 15782 Santiago de Compostela, Spain; (M.G.-L.); (C.V.Á.)
| | - Rubén Nogueiras
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (J.L.); (S.C.); (D.B.); (J.C.); (R.N.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Carlos Diéguez
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (J.L.); (S.C.); (D.B.); (J.C.); (R.N.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
- Correspondence: (C.D.); (S.T.)
| | - Sulay Tovar
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (J.L.); (S.C.); (D.B.); (J.C.); (R.N.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
- Correspondence: (C.D.); (S.T.)
| |
Collapse
|
45
|
Smith A, Woodside B, Abizaid A. Ghrelin and the Control of Energy Balance in Females. Front Endocrinol (Lausanne) 2022; 13:904754. [PMID: 35909536 PMCID: PMC9334675 DOI: 10.3389/fendo.2022.904754] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Ghrelin is considered one of the most potent orexigenic peptide hormones and one that promotes homeostatic and hedonic food intake. Research on ghrelin, however, has been conducted predominantly in males and particularly in male rodents. In female mammals the control of energy metabolism is complex and it involves the interaction between ovarian hormones like estrogen and progesterone, and metabolic hormones. In females, the role that ghrelin plays in promoting feeding and how this is impacted by ovarian hormones is not well understood. Basal ghrelin levels are higher in females than in males, and ghrelin sensitivity changes across the estrus cycle. Yet, responses to ghrelin are lower in female and seem dependent on circulating levels of ovarian hormones. In this review we discuss the role that ghrelin plays in regulating homeostatic and hedonic food intake in females, and how the effects of ghrelin interact with those of ovarian hormones to regulate feeding and energy balance.
Collapse
Affiliation(s)
- Andrea Smith
- Department of Neuroscience, Carleton Unversity, Ottawa, ON, Canada
| | - Barbara Woodside
- Department of Neuroscience, Carleton Unversity, Ottawa, ON, Canada
| | - Alfonso Abizaid
- Department of Neuroscience, Carleton Unversity, Ottawa, ON, Canada
- Stress, Trauma and Relience (STAR) Work Group Carleton University, Ottawa, ON, Canada
- *Correspondence: Alfonso Abizaid,
| |
Collapse
|
46
|
Masule MV, Rathod S, Agrawal Y, Patil CR, Nakhate KT, Ojha S, Goyal SN, Mahajan UB. Ghrelin mediated regulation of neurosynaptic transmitters in depressive disorders. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100113. [PMID: 35782191 PMCID: PMC9240712 DOI: 10.1016/j.crphar.2022.100113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022] Open
Abstract
Ghrelin is a peptide released by the endocrine cells of the stomach and the neurons in the arcuate nucleus of the hypothalamus. It modulates both peripheral and central functions. Although ghrelin has emerged as a potent stimulator of growth hormone release and as an orexigenic neuropeptide, the wealth of literature suggests its involvement in the pathophysiology of affective disorders including depression. Ghrelin exhibits a dual role through the advancement and reduction of depressive behavior with nervousness in the experimental animals. It modulates depression-related signals by forming neuronal networks with various neuropeptides and classical neurotransmitter systems. The present review emphasizes the integration and signaling of ghrelin with other neuromodulatory systems concerning depressive disorders. The role of ghrelin in the regulation of neurosynaptic transmission and depressive illnesses implies that the ghrelin system modulation can yield promising antidepressive therapies. Ghrelin is the orexigenic type of neuropeptide. It binds with the growth hormone secretagogue receptor (GHSR). GHSR is ubiquitously present in the various brain regions. Ghrelin is involved in the regulation of depression-related behavior. The review focuses on the neurotransmission and signaling of ghrelin in neuropsychiatric and depressive disorders.
Collapse
Affiliation(s)
- Milind V. Masule
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, 425405, Maharashtra, India
| | - Sumit Rathod
- Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Yogeeta Agrawal
- Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Chandragouda R. Patil
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, 425405, Maharashtra, India
| | - Kartik T. Nakhate
- Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sameer N. Goyal
- Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
- Corresponding author.
| | - Umesh B. Mahajan
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, 425405, Maharashtra, India
- Corresponding author.
| |
Collapse
|
47
|
Deschaine SL, Leggio L. From "Hunger Hormone" to "It's Complicated": Ghrelin Beyond Feeding Control. Physiology (Bethesda) 2022; 37:5-15. [PMID: 34964687 PMCID: PMC8742734 DOI: 10.1152/physiol.00024.2021] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Discovered as a peptide involved in releasing growth hormone, ghrelin was initially characterized as the "hunger hormone." However, emerging research indicates that ghrelin appears to play an important part in relaying information regarding nutrient availability and value and adjusting physiological and motivational processes accordingly. These functions make ghrelin an interesting therapeutic candidate for metabolic and neuropsychiatric diseases involving disrupted nutrition that can further potentiate the rewarding effect of maladaptive behaviors.
Collapse
Affiliation(s)
- Sara L. Deschaine
- 1Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore and Bethesda, Maryland
| | - Lorenzo Leggio
- 1Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore and Bethesda, Maryland,2Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland,3Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island,4Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland,5Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia
| |
Collapse
|
48
|
Choi W, Kim JW, Kang HJ, Kim HK, Kang HC, Lee JY, Kim SW, Stewart R, Kim JM. Synergistic effects of resilience and serum ghrelin levels on the 12-week pharmacotherapeutic response in patients with depressive disorders. J Affect Disord 2021; 295:1489-1493. [PMID: 34565598 DOI: 10.1016/j.jad.2021.09.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/25/2021] [Accepted: 09/12/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND This study investigated the individual and combined effects of self-reported resilience and serum ghrelin levels on 12-week remission in outpatients with depressive disorders who received antidepressant treatment. METHODS The Connor-Davidson Resilience Scale (CDRS) score and serum ghrelin levels were assessed at baseline in 1,094 patients. The patients initially received antidepressant monotherapy. Patients with an insufficient response or uncomfortable side effects received alternative treatments every 3 weeks (at 3, 6, and 9 weeks). Subsequently, 12-week remission, defined as a Hamilton Depression Rating Scale (HAMD) score of ≤ 7, was evaluated. The individual and combined effects of the CDRS score (low vs. high) and serum ghrelin level (low vs. high) on 12-week remission were analyzed using logistic regression models after adjusting for relevant covariates. RESULTS The individual effects of the CDRS score and serum ghrelin level on 12-week remission were not statistically significant. However, the high-CDRS, high-ghrelin group had a significantly higher 12-week remission rate compared to the low-CDRS, low-ghrelin group. CONCLUSIONS The combination of the CDRS score and serum ghrelin level is useful for predicting 12-week remission in patients with depressive disorders receiving pharmacotherapy.
Collapse
Affiliation(s)
- Wonsuk Choi
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Ju-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Korea
| | - Hee-Ju Kang
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Korea
| | - Hee Kyung Kim
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Ho-Cheol Kang
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Ju-Yeon Lee
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Korea
| | - Sung-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Korea
| | - Robert Stewart
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK; South London and Maudsley NHS Foundation Trust, London, UK
| | - Jae-Min Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Korea.
| |
Collapse
|
49
|
Peris-Sampedro F, Le May MV, Stoltenborg I, Schéle E, Dickson SL. A skeleton in the cupboard in ghrelin research: Where are the skinny dwarfs? J Neuroendocrinol 2021; 33:e13025. [PMID: 34427011 DOI: 10.1111/jne.13025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/09/2021] [Accepted: 08/05/2021] [Indexed: 12/15/2022]
Abstract
Based on studies delivering ghrelin or ghrelin receptor agonists, we have learned a great deal about the importance of the brain ghrelin signalling system for a wide range of physiological processes that include feeding behaviours, growth hormone secretion and glucose homeostasis. Because these processes can be considered as essential to life, the question arises as to why mouse models of depleted ghrelin signalling are not all skinny dwarfs with a host of behavioural and metabolic problems. Here, we provide a systematic detailed review of the phenotype of mice with deficient ghrelin signalling to help better understand the relevance and importance of the brain ghrelin signalling system, with a particular emphasis on those questions that remain unanswered.
Collapse
Affiliation(s)
- Fiona Peris-Sampedro
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Marie V Le May
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Iris Stoltenborg
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Erik Schéle
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Suzanne L Dickson
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
50
|
A review of sex differences in the mechanisms and drivers of overeating. Front Neuroendocrinol 2021; 63:100941. [PMID: 34454955 DOI: 10.1016/j.yfrne.2021.100941] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/23/2021] [Accepted: 08/17/2021] [Indexed: 12/16/2022]
Abstract
Disordered eating is often associated with marked psychological and emotional distress, and severe adverse impact on quality of life. Several factors can influence eating behavior and drive food consumption in excess of energy requirements for homeostasis. It is well established that stress and negative affect contribute to the aetiology of eating disorders and weight gain, and there is substantial evidence suggesting sex differences in sub-clinical and clinical types of overeating. This review will examine how negative affect and stress shape eating behaviors, and how the relationship between the physiological, endocrine, and neural responses to stress and eating behaviors differs between men and women. We will examine several drivers of overeating and explore possible mechanisms underlying sex differences in eating behavior.
Collapse
|