1
|
Zhang C, Liu Y, Gao Y, Li M, Dong Y, Liu X, Li J. The sex-specific relationship of ghrelin and cognition in Chinese han first-episode drug-naive major depressive disorder. J Neural Transm (Vienna) 2025; 132:699-707. [PMID: 39849211 DOI: 10.1007/s00702-025-02880-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/08/2025] [Indexed: 01/25/2025]
Abstract
In major depressive disorder (MDD), alterations in ghrelin levels and cognitive impairment coexist, yet their association has remained largely elusive. This study aimed to investigate the association between ghrelin levels and cognition in both MDD patients and healthy controls (HCs) while also exploring sex-specific differences in this correlation. A total of 155 Chinese Han subjects, including 90 first-episode drug-naive MDD patients and 65 HCs, were enrolled. Ghrelin levels were measured using ELISA kits, and neurocognitive assessments were conducted using the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS). MDD patients exhibited significantly higher ghrelin levels and lower cognitive scores of RBANS compared to HCs. However, there was no significant correlation between ghrelin levels and cognitive function in both MDD patients and HCs. Exploratory analyses revealed sex-specific associations between ghrelin and cognitive function, particularly in MDD patients. Females with MDD showed distinct patterns of association between ghrelin levels and cognitive domains such as attention and language, which were not observed in healthy controls or male MDD patients. The relationship between ghrelin and cognition only existed in MDD patients, not in the HCs, and there was a sex-specific difference in this association. Further research on the mechanism of ghrelin in the cognitive function of MDD should focus on sex differences.
Collapse
Affiliation(s)
- Chuhao Zhang
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Yuan Liu
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Ying Gao
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Meijuan Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Yeqing Dong
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Xueying Liu
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Jie Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China.
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China.
| |
Collapse
|
2
|
Beuker C, Schreiner U, Strecker JK, Altach E, Rätzel V, Schmidt-Pogoda A, Wiendl H, Minnerup J, Diederich K. Ghrelin promotes neurologic recovery and neurogenesis in the chronic phase after experimental stroke. Neurol Res Pract 2025; 7:14. [PMID: 40025613 PMCID: PMC11921976 DOI: 10.1186/s42466-025-00371-6] [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: 11/25/2024] [Accepted: 01/24/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND The neuroprotective and proangiogenic potential of ghrelin in acute ischemic stroke has been demonstrated in experimental studies. However, the transferability of these results is limited as ghrelin was administered either before or very early after stroke onset and follow-up was limited to the first days after stroke. The aim of this study was therefore to close and extend this knowledge gap. To this end, we investigated the effect of ghrelin in two different translational animal models, one investigating acute and one investigating long-term structural and functional recovery after experimental stroke. METHODS Middle cerebral artery occlusion (MCAO) or photothrombotic stroke was induced in 65 adult male Wistar rats. Eleven sham-operated animals served as controls. The rats were treated with either ghrelin, the ghrelin receptor antagonist [D-Lys]-GHRP-6 or a control substance. Up to four weeks after ischemia, behavioral tests such as the cylinder test, the tape removal test, and the rotarod test were performed to examine sensorimotor deficits, and the Morris water maze was performed to examine effects on the acquisition and consolidation of new memories. The structural outcome was determined by a differential analysis of neurogenesis in relation to survival and proliferation of newborn neurons in the post-ischemic brain, angiogenesis and determination of infarct size. RESULTS Ghrelin treatment improved motor and somatosensory functions and preserved the consolidation of new memories after photothrombotic stroke. As a structural correlate, long-term survival and sustained proliferation of neuronal cells after stroke was significantly increased in ghrelin-treated rats, while angiogenesis remained unaffected. In contrast to these neuroregenerative mechanisms, ghrelin did not induce immediate neuroprotective effects after MCAO. CONCLUSIONS Our results suggest that ghrelin has a significant pro-neuroregenerative effect by enhancing long-term survival and sustained proliferation of neurons in the dentate gyrus and peri-infarct area, thus promoting functional recovery. Overall, ghrelin represents a promising target in the subacute and chronic phase after ischemic stroke.
Collapse
Affiliation(s)
- Carolin Beuker
- Department of Neurology with Institute of Translational Neurology, Medical Faculty, University Hospital Muenster, 48149, Muenster, Germany.
| | - Ulrike Schreiner
- Department of Neurology with Institute of Translational Neurology, Medical Faculty, University Hospital Muenster, 48149, Muenster, Germany
| | - Jan-Kolja Strecker
- Department of Neurology with Institute of Translational Neurology, Medical Faculty, University Hospital Muenster, 48149, Muenster, Germany
| | - Elena Altach
- Department of Neurology with Institute of Translational Neurology, Medical Faculty, University Hospital Muenster, 48149, Muenster, Germany
| | - Verena Rätzel
- Department of Neurology with Institute of Translational Neurology, Medical Faculty, University Hospital Muenster, 48149, Muenster, Germany
| | - Antje Schmidt-Pogoda
- Department of Neurology with Institute of Translational Neurology, Medical Faculty, University Hospital Muenster, 48149, Muenster, Germany
| | - Heinz Wiendl
- Clinic of Neurology and Neurophysiology, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Jens Minnerup
- Department of Neurology with Institute of Translational Neurology, Medical Faculty, University Hospital Muenster, 48149, Muenster, Germany
- Department of Neurology, University of Lübeck and University Medical Center of Schleswig- Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Kai Diederich
- Department of Neurology with Institute of Translational Neurology, Medical Faculty, University Hospital Muenster, 48149, Muenster, Germany
| |
Collapse
|
3
|
van Gils P, Nutma S, Meeske K, van Heugten C, van den Bergh W, Foudraine N, le Feber J, Filius M, van Putten M, Beishuizen B, Hofmeijer J. Ghrelin for neuroprotection in post-cardiac arrest coma: a 1-year follow-up of cognitive and psychosocial outcomes. EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2025; 14:5-11. [PMID: 39445445 PMCID: PMC11783279 DOI: 10.1093/ehjacc/zuae119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/27/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
AIMS Effective treatments to improve brain recovery after cardiac arrest are needed. Ghrelin showed efficacy in experimental models and was associated with lower neuron-specific enolase levels in the clinical Ghrelin in Coma (GRECO) trial. Here, we present cognitive and psychosocial outcomes at 1-year follow-up. METHODS AND RESULTS GRECO was a Phase 2 multicentre, double-blind, randomized, placebo-controlled trial in comatose patients after cardiac arrest. The intervention was intravenous acyl-ghrelin 600 μg twice daily or placebo for 1 week, starting within 12 h after the arrest. Patients were assessed after 1 year using cognitive tests and questionnaires measuring participation, health-related quality of life, mood, and caregiver strain. Composite z-scores of the cognitive tests were computed by comparing the scores with those of a norm population and averaging the tests for memory, attention, and executive functioning separately. Groups were compared based on composite z-scores and cut-off scores for psychosocial outcomes. Of the 160 participants originally included, 66 of the 85 participants who survived to 1 year after OHCA completed the psychosocial and cognitive follow-up. The intervention group scored numerically higher across the cognitive domains compared with the control group, but the differences were not statistically significant (memory median = -0.850 vs. -1.385, U = 424.5, P = 0.587; attention median = -0.733 vs. -0.717, U = 420.5, P = 0.548; and executive functioning median = -0.311 vs. -0.482, U = 408.5, P = 0.323). There were significantly fewer signs of depression in the intervention group (U = 322.5, P = 0.014). CONCLUSION This predefined secondary analysis found that ghrelin treatment was associated with non-significantly but consistently better cognitive outcomes and significantly fewer signs of depression. This is in line with the primary outcomes. CLINICAL TRIAL REGISTRATION Clinicaltrialsregister.eu: EUCTR2018-000005-23-NL.
Collapse
Affiliation(s)
- Pauline van Gils
- Department of Clinical Neurophysiology, Technical Medical Center, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
- Limburg Brain Injury Center, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Sjoukje Nutma
- Department of Clinical Neurophysiology, Technical Medical Center, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
- Department of Neurology, Medisch Spectrum Twente, Koningstraat 1, 7512 KZ Enschede, The Netherlands
| | - Karen Meeske
- Department of Medical Psychology, Medisch Spectrum Twente, Koningstraat 1, 7512 KZ Enschede, The Netherlands
| | - Caroline van Heugten
- Limburg Brain Injury Center, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Walter van den Bergh
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Norbert Foudraine
- Department of Critical Care, VieCuri Medical Center, Tegelseweg 210, 5912 BL Venlo, The Netherlands
| | - Joost le Feber
- Department of Clinical Neurophysiology, Technical Medical Center, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - Margreet Filius
- Department of Clinical Pharmacy, Rijnstate Hospital, Wagnerlaan 55, 6815 AD Arnhem, The Netherlands
| | - Michel van Putten
- Department of Clinical Neurophysiology, Technical Medical Center, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
- Department of Neurology, Medisch Spectrum Twente, Koningstraat 1, 7512 KZ Enschede, The Netherlands
| | - Bert Beishuizen
- Department of Critical Care, Medisch Spectrum Twente, Koningstraat 1, 7512 KZ Enschede, The Netherlands
| | - Jeannette Hofmeijer
- Department of Clinical Neurophysiology, Technical Medical Center, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
- Department of Neurology, Rijnstate Hospital, Wagnerlaan 55, 6815 AD Arnhem, The Netherlands
| |
Collapse
|
4
|
Jászberényi M, Thurzó B, Jayakumar AR, Schally AV. The Aggravating Role of Failing Neuropeptide Networks in the Development of Sporadic Alzheimer's Disease. Int J Mol Sci 2024; 25:13086. [PMID: 39684795 DOI: 10.3390/ijms252313086] [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: 10/12/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease imposes an increasing burden on aging Western societies. The disorder most frequently appears in its sporadic form, which can be caused by environmental and polygenic factors or monogenic conditions of incomplete penetrance. According to the authors, in the majority of cases, Alzheimer's disease represents an aggravated form of the natural aging of the central nervous system. It can be characterized by the decreased elimination of amyloid β1-42 and the concomitant accumulation of degradation-resistant amyloid plaques. In the present paper, the dysfunction of neuropeptide regulators, which contributes to the pathophysiologic acceleration of senile dementia, is reviewed. However, in the present review, exclusively those neuropeptides or neuropeptide families are scrutinized, and the authors' investigations into their physiologic and pathophysiologic activities have made significant contributions to the literature. Therefore, the pathophysiologic role of orexins, neuromedins, RFamides, corticotrope-releasing hormone family, growth hormone-releasing hormone, gonadotropin-releasing hormone, ghrelin, apelin, and natriuretic peptides are discussed in detail. Finally, the therapeutic potential of neuropeptide antagonists and agonists in the inhibition of disease progression is discussed here.
Collapse
Affiliation(s)
- Miklós Jászberényi
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
| | - Balázs Thurzó
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
- Emergency Patient Care Unit, Albert Szent-Györgyi Health Centre, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Arumugam R Jayakumar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Andrew V Schally
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
5
|
Mao Q, Wang J, Yang Z, Ding R, Lv S, Ji X. The Pathologic Roles and Therapeutic Implications of Ghrelin/GHSR System in Mental Disorders. Depress Anxiety 2024; 2024:5537319. [PMID: 40226675 PMCID: PMC11919235 DOI: 10.1155/2024/5537319] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 04/15/2025] Open
Abstract
Ghrelin is a hormone consisting of 28 amino acids. Growth hormone secretagogue receptor (GHSR) is a receptor for ghrelin, which is expressed in the brain, pituitary gland, and adrenal glands, especially in the hypothalamus. The binding of ghrelin to the receptor 1a subtype mediates most of the biological effects of ghrelin. Ghrelin has a close relationship with the onset of psychosis. Ghrelin can affect the onset of psychosis by regulating neurotransmitters such as dopamine, γ-aminobutyric acid (GABA), and 5-hydroxytryptamine (5-HT) through the hypothalamus-pituitary-adrenal (HPA) axis, brain-gut axis, the mesolimbic dopamine system, and other ways. Ghrelin activates neuropeptide Y (NPY) in the hypothalamic arcuate nucleus (ARC) through the GHSR. Ghrelin binds to neurons in the ventral tegmental area (VTA), where it promotes the activity of dopamine neurons in the nucleus accumbens (NAcs) in a GHSR-dependent way, increasing dopamine levels and the reward system. This article summarized the recent research progress of ghrelin in depression, anxiety, schizophrenia, anorexia nervosa (AN), and bulimia nervosa (BN), and emphasized its potential application for psychiatric disorders treatment.
Collapse
Affiliation(s)
- Qianshuo Mao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, Henan, China
| | - Jinjia Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, Henan, China
| | - Zihan Yang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, Henan, China
| | - Ruidong Ding
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, Henan, China
| | - Shuangyu Lv
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, Henan, China
- Department of Neurosurgery, The First Affiliated Hospital of Henan University, Henan University, Kaifeng 475001, Henan, China
| | - Xinying Ji
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, 6 Gong-Ming Road, Mazhai Town, Erqi District, Zhengzhou 450064, Henan, China
- Department of Medicine, Huaxian County People's Hospital, Huaxian 456400, Henan, China
| |
Collapse
|
6
|
Zhang M, Yang L, Jia J, Xu F, Gao S, Han F, Deng M, Wang J, Li V, Yu M, Sun Y, Yuan H, Zhou Y, Li N. Increased GHS-R1a expression in the hippocampus impairs memory encoding and contributes to AD-associated memory deficits. Commun Biol 2024; 7:1334. [PMID: 39415032 PMCID: PMC11484987 DOI: 10.1038/s42003-024-06914-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 09/17/2024] [Indexed: 10/18/2024] Open
Abstract
Growth hormone secretagogue receptor 1a (GHS-R1a), also known as the ghrelin receptor, is an important nutrient sensor and metabolic regulator in both humans and rodents. Increased GHS-R1a expression is observed in the hippocampus of both Alzheimer's disease (AD) patients and AD model mice. However, the causal relationship between GHS-R1a elevation in the hippocampus and AD memory deficits remains uncertain. Here, we find that increasing GHS-R1a expression in dCA1 pyramidal neurons impairs hippocampus-dependent memory formation, which is abolished by local administration of the endogenous antagonist LEAP2. GHS-R1a elevation in dCA1 pyramidal neurons suppresses excitability and blocks memory allocation in these neurons. Chemogenetic activation of those high GHS-R1a neurons during training rescues GHS-R1a overexpression-induced memory impairment. Moreover, we demonstrate that increasing GHS-R1a expression in dCA1 pyramidal neurons hampers these neurons' ability to encode spatial memory and reduces engram size in the dCA1 region. Finally, we show that GHS-R1a deletion mitigates spatial memory deficits in APP/PS1 mice with increased GHS-R1a expression in the hippocampus. Our findings reveal a negative, causal relationship between hippocampal GHS-R1a expression and memory encoding, and suggest that blocking the abnormal increase in GHS-R1a activity/expression may be a promising approach to improve memory and treat cognitive decline in AD.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266000, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
- College of Agriculture and Bioengineering, Heze University, Heze, Shandong, 274000, China
| | - Liu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Jiajia Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Fenghua Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Shanshan Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Fubing Han
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, China
| | - Mingru Deng
- Department of Neurology, Affiliated Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong, 266042, China
| | - Jiwei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Vincent Li
- Beverly Hills High School, Beverly Hills, CA, 90212, USA
| | - Ming Yu
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266000, China
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
| | - Haicheng Yuan
- Department of Neurology, Affiliated Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong, 266042, China
| | - Yu Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China.
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266000, China.
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China.
| | - Nan Li
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266000, China.
| |
Collapse
|
7
|
Chang L, He Y, Tian T, Li B. Nucleus accumbens ghrelin signaling controls anxiety-like behavioral response to acute stress. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2024; 20:18. [PMID: 38965529 PMCID: PMC11225390 DOI: 10.1186/s12993-024-00244-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/29/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Anxiety disorders are one of the most common mental disorders. Ghrelin is a critical orexigenic brain-gut peptide that regulates food intake and metabolism. Recently, the ghrelin system has attracted more attention for its crucial roles in psychiatric disorders, including depression and anxiety. However, the underlying neural mechanisms involved have not been fully investigated. METHODS In the present study, the effect and underlying mechanism of ghrelin signaling in the nucleus accumbens (NAc) core on anxiety-like behaviors were examined in normal and acute stress rats, by using immunofluorescence, qRT-PCR, neuropharmacology, molecular manipulation and behavioral tests. RESULTS We reported that injection of ghrelin into the NAc core caused significant anxiolytic effects. Ghrelin receptor growth hormone secretagogue receptor (GHSR) is highly localized and expressed in the NAc core neurons. Antagonism of GHSR blocked the ghrelin-induced anxiolytic effects. Moreover, molecular knockdown of GHSR induced anxiogenic effects. Furthermore, injection of ghrelin or overexpression of GHSR in the NAc core reduced acute restraint stress-induced anxiogenic effects. CONCLUSIONS This study demonstrates that ghrelin and its receptor GHSR in the NAc core are actively involved in modulating anxiety induced by acute stress, and raises an opportunity to treat anxiety disorders by targeting ghrelin signaling system.
Collapse
Affiliation(s)
- Leilei Chang
- Women and Children's Medical Research Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yecheng He
- Department of Preclinical Medicine, Suzhou Vocational Health College, Suzhou, 215009, China
| | - Tian Tian
- Department of Child Health Care, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Bin Li
- Women and Children's Medical Research Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
8
|
De Guia IL, Eslick S, Naismith SL, Kanduri S, Shah TM, Martins RN. The Crosstalk Between Amyloid-β, Retina, and Sleep for the Early Diagnosis of Alzheimer's Disease: A Narrative Review. J Alzheimers Dis Rep 2024; 8:1009-1021. [PMID: 39114553 PMCID: PMC11305848 DOI: 10.3233/adr-230150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 05/17/2024] [Indexed: 08/10/2024] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia, which is characterised by progressive memory loss and accumulation of hallmark markers amyloid-β (Aβ) and neurofibrillary tangles in the diseased brain. The current gold standard diagnostic methods have limitations of being invasive, costly, and not easily accessible. Thus, there is a need for new avenues, such as imaging the retina for early AD diagnosis. Sleep disruption is symptomatically frequent across preclinical and AD subjects. As circadian activity, such as the sleep-wake cycle, is linked to the retina, analysis of their association may be useful additions for achieving predictive AD diagnosis. In this narrative review, we provide an overview of human retina studies concerning the deposition of Aβ, the role of the retina in sleep-wake cycle, the disruption of sleep in AD, and to gather evidence for the associations between Aβ, the retina, and sleep. Understanding the mechanisms behind the associations between Aβ, retina, and sleep could assist in the interpretation of retinal changes accurately in AD.
Collapse
Affiliation(s)
| | - Shaun Eslick
- Macquarie University, North Ryde, NSW, Australia
| | - Sharon L. Naismith
- Faculty of Science, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | | | | | - Ralph N. Martins
- Macquarie University, North Ryde, NSW, Australia
- Edith Cowen University, Joondalup, WA, Australia
- Australian Alzheimer’s Research Foundation, Nedlands, WA, Australia
| |
Collapse
|
9
|
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
|
10
|
Örüm D, Korkmaz S, İlhan N, Örüm MH, Atmaca M. Leptin, Nesfatin-1, Orexin-A, and Total Ghrelin Levels in Drug-Naive Panic Disorder. Psychiatry Investig 2024; 21:142-150. [PMID: 38433413 PMCID: PMC10910167 DOI: 10.30773/pi.2023.0309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/06/2023] [Accepted: 11/09/2023] [Indexed: 03/05/2024] Open
Abstract
OBJECTIVE This study aimed to examine the changes in serum nesfatin-1, leptin, orexin-A, and total ghrelin levels of patients diagnosed with drug-naive panic disorder (PD) before and after six weeks of the treatment and to compare the findings with the healthy subjects. METHODS The neuropeptides were measured in venous blood samples taken from 32 patients and 32 healthy subjects. The blood samples of the patients who used paroxetine 20 mg/day plus alprazolam 0.5 mg/day were retaken again after six weeks. Measurements were performed with the Enzyme-Linked Immunosorbent Assay (ELISA) method. RESULTS Serum nesfatin-1, leptin, orexin-A and total ghrelin levels of the patient group were found to be significantly lower than the control group (p<0.001, p<0.001, p<0.001, and p<0.001, respectively). When the serum nesfatin-1, leptin, orexin-A and total ghrelin levels of the patient group were compared before and after treatment, significant differences were found in terms of orexin-A and total ghrelin levels (p=0.046, p<0.001, respectively). However, no significant differences were found in terms of nesfatin-1and leptin levels (p=0.205, p=0.988, respectively). CONCLUSION This study reports that PD, like other anxiety disorders, may affect serum nesfatin-1, leptin, orexin-A, and total ghrelin levels, and there may be a relationship between PD treatment and the levels of these neuropeptides. The variability of this relationship among the neuropeptides examined indicates that various factors other than treatment play a role in this process.
Collapse
Affiliation(s)
- Dilek Örüm
- Psychiatry, Elazığ Fethi Sekin City Hospital, Elazığ, Turkey
| | - Sevda Korkmaz
- Department of Psychiatry, Firat University, Faculty of Medicine, Elazığ, Turkey
| | - Nevin İlhan
- Department of Medical Biochemistry, Firat University, Faculty of Medicine, Elazığ, Turkey
| | - Mehmet Hamdi Örüm
- Psychiatry, Elazığ Mental Health and Diseases Hospital, Elazığ, Turkey
| | - Murad Atmaca
- Department of Psychiatry, Firat University, Faculty of Medicine, Elazığ, Turkey
| |
Collapse
|
11
|
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
|
12
|
Uner B, Ergin AD, Ansari IA, Macit-Celebi MS, Ansari SA, Kahtani HMA. Assessing the In Vitro and In Vivo Performance of L-Carnitine-Loaded Nanoparticles in Combating Obesity. Molecules 2023; 28:7115. [DOI: 15.https:/doi.org/10.3390/molecules28207115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2025] Open
Abstract
Addressing obesity is a critical health concern of the century, necessitating urgent attention. L-carnitine (LC), an essential water-soluble compound, plays a pivotal role in lipid breakdown via β-oxidation and facilitates the transport of long-chain fatty acids across mitochondrial membranes. However, LC’s high hydrophilicity poses challenges to its diffusion through bilayers, resulting in limited bioavailability, a short half-life, and a lack of storage within the body, mandating frequent dosing. In our research, we developed LC-loaded nanoparticle lipid carriers (LC-NLCs) using economically viable and tissue-localized nanostructured lipid carriers (NLCs) to address these limitations. Employing the central composite design model, we optimized the formulation, employing the high-pressure homogenization (HPH) method and incorporating Poloxamer® 407 (surfactant), Compritol® 888 ATO (solid lipid), and oleic acid (liquid oil). A comprehensive assessment of nanoparticle physical attributes was performed, and an open-field test (OFT) was conducted on rats. We employed immunofluorescence assays targeting CRP and PPAR-γ, along with an in vivo rat study utilizing an isolated fat cell line to assess adipogenesis. The optimal formulation, with an average size of 76.4 ± 3.4 nm, was selected due to its significant efficacy in activating the PPAR-γ pathway. Our findings from the OFT revealed noteworthy impacts of LC-NLC formulations (0.1 mg/mL and 0.2 mg/mL) on adipocyte cells, surpassing regular L-carnitine formulations’ effects (0.1 mg/mL and 0.2 mg/mL) by 169.26% and 156.63%, respectively (p < 0.05).
Collapse
Affiliation(s)
- Burcu Uner
- Department of Administrative and Pharmaceutical Sciences, University of Health Science and Pharmacy in St. Louis, St. Louis, MO 63110, USA
| | - Ahmet Dogan Ergin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Trakya University, 22030 Edirne, Turkey
- Department of Neuroscience, University of Turin, 10124 Turin, Italy
- Department of Pharmaceutical Nanotechnology, Institute of Health Sciences, Trakya University, 22030 Edirne, Turkey
| | - Irfan Aamer Ansari
- Department of Drug Science and Technology, University of Turin, 10124 Turin, Italy
| | - Melahat Sedanur Macit-Celebi
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ondokuz Mayıs University, 55270 Samsun, Turkey
| | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hamad M. Al Kahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
13
|
Uner B, Ergin AD, Ansari IA, Macit-Celebi MS, Ansari SA, Kahtani HMA. Assessing the In Vitro and In Vivo Performance of L-Carnitine-Loaded Nanoparticles in Combating Obesity. Molecules 2023; 28:7115. [PMID: 37894594 PMCID: PMC10609287 DOI: 10.3390/molecules28207115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/08/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023] Open
Abstract
Addressing obesity is a critical health concern of the century, necessitating urgent attention. L-carnitine (LC), an essential water-soluble compound, plays a pivotal role in lipid breakdown via β-oxidation and facilitates the transport of long-chain fatty acids across mitochondrial membranes. However, LC's high hydrophilicity poses challenges to its diffusion through bilayers, resulting in limited bioavailability, a short half-life, and a lack of storage within the body, mandating frequent dosing. In our research, we developed LC-loaded nanoparticle lipid carriers (LC-NLCs) using economically viable and tissue-localized nanostructured lipid carriers (NLCs) to address these limitations. Employing the central composite design model, we optimized the formulation, employing the high-pressure homogenization (HPH) method and incorporating Poloxamer® 407 (surfactant), Compritol® 888 ATO (solid lipid), and oleic acid (liquid oil). A comprehensive assessment of nanoparticle physical attributes was performed, and an open-field test (OFT) was conducted on rats. We employed immunofluorescence assays targeting CRP and PPAR-γ, along with an in vivo rat study utilizing an isolated fat cell line to assess adipogenesis. The optimal formulation, with an average size of 76.4 ± 3.4 nm, was selected due to its significant efficacy in activating the PPAR-γ pathway. Our findings from the OFT revealed noteworthy impacts of LC-NLC formulations (0.1 mg/mL and 0.2 mg/mL) on adipocyte cells, surpassing regular L-carnitine formulations' effects (0.1 mg/mL and 0.2 mg/mL) by 169.26% and 156.63%, respectively (p < 0.05).
Collapse
Affiliation(s)
- Burcu Uner
- Department of Administrative and Pharmaceutical Sciences, University of Health Science and Pharmacy in St. Louis, St. Louis, MO 63110, USA
| | - Ahmet Dogan Ergin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Trakya University, 22030 Edirne, Turkey
- Department of Neuroscience, University of Turin, 10124 Turin, Italy
- Department of Pharmaceutical Nanotechnology, Institute of Health Sciences, Trakya University, 22030 Edirne, Turkey
| | - Irfan Aamer Ansari
- Department of Drug Science and Technology, University of Turin, 10124 Turin, Italy;
| | - Melahat Sedanur Macit-Celebi
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ondokuz Mayıs University, 55270 Samsun, Turkey;
| | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.A.); (H.M.A.K.)
| | - Hamad M. Al Kahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.A.); (H.M.A.K.)
| |
Collapse
|
14
|
Gajewska A, Strzelecki D, Gawlik-Kotelnicka O. Ghrelin as a Biomarker of "Immunometabolic Depression" and Its Connection with Dysbiosis. Nutrients 2023; 15:3960. [PMID: 37764744 PMCID: PMC10537261 DOI: 10.3390/nu15183960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/08/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
Ghrelin, a gastrointestinal peptide, is an endogenous ligand of growth hormone secretagogue receptor 1a (GHSR1a), which is mainly produced by X/A-like cells in the intestinal mucosa. Beyond its initial description as a growth hormone (GH) secretagogue stimulator of appetite, ghrelin has been revealed to have a wide range of physiological effects, for example, the modulation of inflammation; the improvement of cardiac performance; the modulation of stress, anxiety, taste sensation, and reward-seeking behavior; and the regulation of glucose metabolism and thermogenesis. Ghrelin secretion is altered in depressive disorders and metabolic syndrome, which frequently co-occur, but it is still unknown how these modifications relate to the physiopathology of these disorders. This review highlights the increasing amount of research establishing the close relationship between ghrelin, nutrition, microbiota, and disorders such as depression and metabolic syndrome, and it evaluates the ghrelinergic system as a potential target for the development of effective pharmacotherapies.
Collapse
Affiliation(s)
- Agata Gajewska
- Faculty of Medicine, Medical University of Lodz, 92-216 Lodz, Poland;
| | - Dominik Strzelecki
- Department of Affective and Psychotic Disorders, Medical University of Lodz, 92-216 Lodz, Poland;
| | - Oliwia Gawlik-Kotelnicka
- Department of Affective and Psychotic Disorders, Medical University of Lodz, 92-216 Lodz, Poland;
| |
Collapse
|
15
|
Bouâouda H, Jha PK. Orexin and MCH neurons: regulators of sleep and metabolism. Front Neurosci 2023; 17:1230428. [PMID: 37674517 PMCID: PMC10478345 DOI: 10.3389/fnins.2023.1230428] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/07/2023] [Indexed: 09/08/2023] Open
Abstract
Sleep-wake and fasting-feeding are tightly coupled behavioral states that require coordination between several brain regions. The mammalian lateral hypothalamus (LH) is a functionally and anatomically complex brain region harboring heterogeneous cell populations that regulate sleep, feeding, and energy metabolism. Significant attempts were made to understand the cellular and circuit bases of LH actions. Rapid advancements in genetic and electrophysiological manipulation help to understand the role of discrete LH cell populations. The opposing action of LH orexin/hypocretin and melanin-concentrating hormone (MCH) neurons on metabolic sensing and sleep-wake regulation make them the candidate to explore in detail. This review surveys the molecular, genetic, and neuronal components of orexin and MCH signaling in the regulation of sleep and metabolism.
Collapse
Affiliation(s)
- Hanan Bouâouda
- Pharmacology Institute, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Pawan Kumar Jha
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
16
|
Li N, Li N, Yang L, Gu H, Ji J, Zhou H, Zhu Q, Yu M, Sun Y, Zhou Y. GHSR1a deficiency suppresses inhibitory drive on dCA1 pyramidal neurons and contributes to memory reinforcement. Cereb Cortex 2023; 33:2612-2625. [PMID: 35797708 DOI: 10.1093/cercor/bhac230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 11/12/2022] Open
Abstract
Growth hormone secretagogue receptor 1a (GHSR1a)-the receptor for orexigenic hormone ghrelin-is a G protein-coupled receptor that is widely distributed in the brain, including the hippocampus. Studies have demonstrated that genetic deletion of GHSR1a affects memory, suggesting the importance of ghrelin/GHSR1a signaling in cognitive control. However, current reports are controversial, and the mechanism underlying GHSR1a modulation of memory is uncertain. Here, we first report that global GHSR1a knockout enhances hippocampus-dependent memory, facilitates initial LTP in dorsal hippocampal Schaffer Collateral-CA1 synapses, and downregulates Akt activity in the hippocampus. Moreover, we show that the intrinsic excitability of GAD67+ interneurons-rather than neighboring pyramidal neurons in the dCA1-is suppressed by GHSR1a deletion, an effect that is antagonized by acute application of the Akt activator SC79. In addition, the inhibitory postsynaptic currents (IPSCs) on dCA1 pyramidal neurons are selectively reduced in mice with a GHSR1a deficiency. Finally, we demonstrate that selectively increasing the excitability of parvalbumin-expressing interneurons by hM3Dq-DREADDs increases IPSCs on dCA1 pyramidal neurons and normalizes memory in Ghsr1a KO mice. Our findings thus reveal a novel mechanism underlying memory enhancement of GHSR1a deficiency and herein support an adverse effect of GHSR1a signaling in hippocampus-dependent memory processes.
Collapse
Affiliation(s)
- Na Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
- Department of Medicine, Qingdao Binhai University, 425 West Jialing River Rd, Qingdao, Shandong, 266555, China
| | - Nan Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, 17 Shandong Rd, Qingdao, Shandong, 266000, China
| | - Liu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
| | - Huating Gu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
| | - Junjie Ji
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
| | - Hao Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
| | - Qianqian Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
| | - Ming Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, 17 Shandong Rd, Qingdao, Shandong, 266000, China
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, 750 Agronomy Rd, College Station, TX, 77843, United States
| | - Yu Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, 17 Shandong Rd, Qingdao, Shandong, 266000, China
- Department of Physiology, Institute of Brain Sciences and Related Disorders, Qingdao University, 308 Ningxia Rd., Qingdao, Shandong, 266071, China
- Department of rehabilitation medicine, Affiliated Hospital of Qingdao University, 16 Jiangsu Rd., Qingdao, Shandong, 266000, China
| |
Collapse
|
17
|
Fritz EM, Pierre A, De Bundel D, Singewald N. Ghrelin receptor agonist MK0677 and overnight fasting do not rescue deficient fear extinction in 129S1/SvImJ mice. Front Psychiatry 2023; 14:1094948. [PMID: 36846243 PMCID: PMC9947350 DOI: 10.3389/fpsyt.2023.1094948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/03/2023] [Indexed: 02/11/2023] Open
Abstract
The hunger hormone ghrelin has been implicated in the modulation of anxiety- and fear-related behaviors in rodents and humans, while its dysregulation may be associated with psychiatric illness. Along these lines, the ghrelin system has been suggested as a potential target to facilitate fear extinction, which is the main mechanism underlying cognitive behavioral therapy. So far, this hypothesis has not been tested in individuals that have difficulties to extinguish fear. Thus, we investigated pharmacological (ghrelin receptor agonist MK0677) and non-pharmacological (overnight fasting) strategies to target the ghrelin system in the 129S1/SvImJ (S1) mouse strain, which models the endophenotype of impaired fear extinction that has been associated with treatment resistance in anxiety and PTSD patients. MK0677 induced food intake and overnight fasting increased plasma ghrelin levels in S1 mice, suggesting that the ghrelin system is responsive in the S1 strain. However, neither systemic administration of MK0677 nor overnight fasting had an effect on fear extinction in S1 mice. Similarly, our groups previously reported that both interventions did not attenuate fear in extinction-competent C57BL/6J mice. In summary, our findings are in contrast to several studies reporting beneficial effects of GHSR agonism and overnight fasting on fear- and anxiety-related behaviors in rodents. Rather, our data agree with accumulating evidence of divergent behavioral effects of ghrelin system activation and underscore the hypothesis that potential benefits of targeting the ghrelin system in fear extinction may be dependent on factors (e.g., previous stress exposure) that are not yet fully understood.
Collapse
Affiliation(s)
- Eva Maria Fritz
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Innsbruck, Austria
| | - Anouk Pierre
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Dimitri De Bundel
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
18
|
The effect of Ramadan fasting on mental health and some hormonal levels in healthy males. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2023. [DOI: 10.1186/s41983-023-00623-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Abstract
Background
Millions of Muslims around the world fast during the holy month of Ramadan as a requirement of their religion Islam. Studies have reported varying effects of Ramadan fasting on mental health and various hormones. This study aimed to examine the effects of Ramadan fasting on mental health and plasma Leptin, Ghrelin, Neuropeptide Y (NPY), Growth Hormone levels in healthy individuals, and to evaluate the possible relationship between their hormone values and scale scores. Male healthcare professionals working at a university hospital without any psychiatric disease were included in the study. In the last week before Ramadan and in the first week after Ramadan, participants’ blood samples were taken at 8.00 in the morning after 12 h of fasting, taking into account the release pattern and pulsatile release of these hormones in order to measure plasma Leptin, Ghrelin, NPY and Growth Hormone levels. Simultaneously, a sociodemographic data form, the Brief Symptom Inventory (BSI) and the Scale of Dimensions of Interpersonal Relationships were applied to participants.
Results
The sample of the study included a total of 40 healthcare professionals. Participants’ BSI interpersonal sensitivity and phobic anxiety subscales scores and their general severity and positive symptom distress index scores decreased significantly after Ramadan compared to those measured before Ramadan. (p < 0.001, p = 0.020, p = 0.042, p = 0.006 respectively). Also participants’ ghrelin levels increased significantly after Ramadan compared to those measured before Ramadan (p < 0.001).
Conclusions
The effects of Ramadan fasting on mental health may be mediated by some psychoneuroendocrine mechanisms. In order to elucidate these mechanisms that mediate the effect of Ramadan fasting on mental health, there is a need for better-structured studies with larger samples and more variables.
Collapse
|
19
|
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
|
20
|
Harris BN, Roberts BR, DiMarco GM, Maldonado KA, Okwunwanne Z, Savonenko AV, Soto PL. Hypothalamic-pituitary-adrenal (HPA) axis activity and anxiety-like behavior during aging: A test of the glucocorticoid cascade hypothesis in amyloidogenic APPswe/PS1dE9 mice. Gen Comp Endocrinol 2023; 330:114126. [PMID: 36122793 PMCID: PMC10250074 DOI: 10.1016/j.ygcen.2022.114126] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/06/2022] [Accepted: 09/13/2022] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a progressive, dementing, whole-body disorder that presents with decline in cognitive, behavioral, and emotional functions, as well as endocrine dysregulation. The etiology of AD is not fully understood but stress- and anxiety-related hormones may play a role in its development and trajectory. The glucocorticoid cascade hypothesis posits that levels of glucocorticoids increase with age, leading to dysregulated negative feedback, further elevated glucocorticoids, and resulting neuropathology. We examined the impact of age (from 2 to 10 months) and stressor exposure (predator odor) on hormone levels (corticosterone and ghrelin), anxiety-like behavior (open field and light dark tests), and memory-related behavior (novel object recognition; NOR), and whether these various measures correlated with neuropathology (hippocampus and cortex amyloid beta, Aβ) in male and female APPswe/PS1dE9 transgenic and non-transgenic mice. Additionally, we performed exploratory analyses to probe if the open field and light dark test as commonly used tasks to assess anxiety levels were correlated. Consistent with the glucocorticoid cascade hypothesis, baseline corticosterone increased with age. Predator odor exposure elevated corticosterone at each age, but in contrast to the glucocorticoid cascade hypothesis, the magnitude of stressor-induced elevations in corticosterone levels did not increase with age. Overall, transgenic mice had higher post-stressor, but not baseline, corticosterone than non-transgenic mice, and across both genotypes, females consistently had higher (baseline and post-stressor) corticosterone than males. Behavior in the open field test primarily showed decreased locomotion with age, and this was pronounced in transgenic females. Anxiety-like behaviors in the light dark test were exacerbated following predator odor, and female transgenic mice were the most impacted. Compared to transgenic males, transgenic females had higher Aβ concentrations and showed more anxiety-like behavior. Performance on the NOR did not differ significantly between genotypes. Lastly, we did not find robust, statistically significant correlations among corticosterone, ghrelin, recognition memory, anxiety-like behaviors, or Aβ, suggesting outcomes are not strongly related on the individual level. Our data suggest that despite Aβ accumulation in the hippocampus and cortex, male and female APPswePS1dE9 transgenic mice do not differ robustly from their non-transgenic littermates in physiological, endocrine, and behavioral measures at the range of ages studied here.
Collapse
Affiliation(s)
- Breanna N Harris
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States.
| | - Breanna R Roberts
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
| | - Giuliana M DiMarco
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States; Center for Molecular and Behavioral Neuroscience, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | | | - Zenobia Okwunwanne
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
| | - Alena V Savonenko
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Paul L Soto
- Department of Psychology, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
21
|
Reich N, Hölscher C. Beyond Appetite: Acylated Ghrelin As A Learning, Memory and Fear Behavior-modulating Hormone. Neurosci Biobehav Rev 2022; 143:104952. [DOI: 10.1016/j.neubiorev.2022.104952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 04/27/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022]
|
22
|
Chen X, Mi J, Huang H, Wang J, Wu Y, Wu X, Zhang S. Ghrelin and ghrelin receptor (GHSR) in Chinese alligator, alligator sinensis: Molecular characterization, tissue distribution and mRNA expression changes during the active and hibernating periods. Gen Comp Endocrinol 2022; 327:114097. [PMID: 35853503 DOI: 10.1016/j.ygcen.2022.114097] [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: 04/22/2022] [Revised: 07/02/2022] [Accepted: 07/13/2022] [Indexed: 11/20/2022]
Abstract
The Chinese alligator (Alligator sinensis) is a freshwater crocodilian endemic to China. So far, the endocrine regulation of feeding and growth in Chinese alligator is poorly understood. In this study, the molecular structure and tissue expression profiles of ghrelin and its receptor GHSR in the Chinese alligator were characterized for the first time. The full-length cDNA of ghrelin was 1770 bp, including a 37 bp 5 '-UTR (untranslated region), a 435 bp ORF (open reading frame) and a 1298 bp 3 '-UTR. The ORF encodes a ghrelin precursor, which consists of 145 amino acid residues, including a signal peptide with 52 amino acid residues at the N-terminus, a mature peptide with 28 amino acid residues, and a possibly obestain at the C-terminus. The full-length cDNA of GHSR was 3961 bp, including a 5'-UTR of 375-bp, an ORF of 1059-bp and a 3' -UTR of 2527-bp. The ORF encodes a protein of 352 amino acid residues containing seven transmembrane domains, with multiple N glycosylation modification sites and conserved cysteine residue sites. The active core "GSSF" of Chinese alligator ghrelin was identical to that of mammals and birds, and the ghrelin binding site of GHSR was similar to that of mammals. The amino acid sequences of both ghrelin and GHSR share high identity with American alligator (Alligator mississippiensis) and birds. Ghrelin was highly expressed in cerebrum, mesencephalon, hypothalamus and multiple peripheral tissues, including lung, stomach and intestine, suggesting that it could play functions in paracrine and/or autocrine manners in addition to endocrine manner. GHSR expression level was higher in hypothalamus, epencephalon and medulla oblongata, and moderate in multiple peripheral tissues including lung, kindey, stomach and oviduct, implicating that ghrelin/GHSR system may participate in the regulation of energy balance, food intake, water and mineral balance, gastrointestinal motility, gastric acid secretion and reproduction. During hibernation, the expression of ghrelin and GHSR in the brain was significantly increased, while ghrelin was significantly decreased in heart, liver, lung, stomach, pancreas and ovary, and GHSR was significantly decreased in heart, liver, spleen, lung, kindey, stomach, ovary and oviduct. These temporal changes in ghrelin and GHSR expression could facilitate the physiological adaption to the hibernation of Chinese alligator. Our study could provide basic data for further studies on the regulation of feeding, physiological metabolism and reproduction of Chinese alligator, which could also be useful for the improvement of artificial breeding of this endangered species.
Collapse
Affiliation(s)
- Xianxian Chen
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China
| | - Jicong Mi
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China
| | - Hongbin Huang
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China
| | - Jing Wang
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China
| | - Yu Wu
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China
| | - Xiaobing Wu
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China
| | - Shengzhou Zhang
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China.
| |
Collapse
|
23
|
Yu M, Zhu QQ, Niu ML, Li N, Ren BQ, Yu TB, Zhou ZS, Guo JD, Zhou Y. Ghrelin infusion into the basolateral amygdala suppresses CTA memory formation in rats via the PI3K/Akt/mTOR and PLC/PKC signaling pathways. Acta Pharmacol Sin 2022; 43:2242-2252. [PMID: 35169271 PMCID: PMC9433413 DOI: 10.1038/s41401-022-00859-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022]
Abstract
Ghrelin is a circulating orexigenic hormone that promotes feeding behavior and regulates metabolism in humans and rodents. We previously reported that local infusion of ghrelin into the basolateral amygdala (BLA) blocked memory acquisition for conditioned taste aversion (CTA) by activating growth hormone secretagogue receptor 1a. In this study, we further explored the underlying mechanism and signaling pathways mediating ghrelin modulation of CTA memory in rats. Pharmacological agents targeting distinct signaling pathways were infused into the BLA during conditioning. We showed that preadministration of the PI3K inhibitor LY294002 abolished the repressive effect of ghrelin on CTA memory. Moreover, LY294002 pretreatment prevented ghrelin from inhibiting Arc and zif268 mRNA expression in the BLA triggered by CTA memory retrieval. Preadministration of rapamycin eliminated the repressive effect of ghrelin, while Gsk3 inhibitors failed to mimic ghrelin's effect. In addition, PLC and PKC inhibitors microinfused in the BLA blocked ghrelin's repression of CTA acquisition. These results demonstrate that ghrelin signaling in the BLA shapes CTA memory via the PI3K/Akt/mTOR and PLC/PKC pathways. We conducted in vivo multichannel recordings from mouse BLA neurons and found that microinjection of ghrelin (20 µM) suppressed intrinsic excitability. By means of whole-cell recordings from rat brain slices, we showed that bath application of ghrelin (200 nM) had no effect on basal synaptic transmission or synaptic plasticity of BLA pyramidal neurons. Together, this study reveals the mechanism underlying ghrelin-induced interference with CTA memory acquisition in rats, i.e., suppression of intrinsic excitability of BLA principal neurons via the PI3K/Akt/mTOR and PLC/PKC pathways.
Collapse
Affiliation(s)
- Ming Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China
| | - Qian-Qian Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China
| | - Ming-Lu Niu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China
| | - Nan Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China
| | - Bai-Qing Ren
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China
| | - Teng-Bo Yu
- Department of Sports Medicine, Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Zhi-Shang Zhou
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266071, China
| | - Ji-Dong Guo
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Yu Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China.
- Department of rehabilitation medicine, Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
24
|
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: 2.7] [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
|
25
|
Herrington JA, Guss Darwich J, Harshaw C, Brigande AM, Leif EB, Currie PJ. Elevated ghrelin alters the behavioral effects of perinatal acetaminophen exposure in rats. Dev Psychobiol 2022; 64:e22252. [DOI: 10.1002/dev.22252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/19/2021] [Accepted: 12/07/2021] [Indexed: 11/06/2022]
Affiliation(s)
- Joshua A. Herrington
- Department of Psychology Reed College 3203 SE Woodstock Blvd, Portland OR 97202, USA Portland Oregon USA
| | - Janet Guss Darwich
- Department of Psychology Reed College 3203 SE Woodstock Blvd, Portland OR 97202, USA Portland Oregon USA
| | - Christopher Harshaw
- Department of Psychology University of New Orleans New Orleans Louisiana USA
| | - Alev M. Brigande
- Department of Psychology Reed College 3203 SE Woodstock Blvd, Portland OR 97202, USA Portland Oregon USA
| | - Erica B. Leif
- Department of Psychology Reed College 3203 SE Woodstock Blvd, Portland OR 97202, USA Portland Oregon USA
| | - Paul J. Currie
- Department of Psychology Reed College 3203 SE Woodstock Blvd, Portland OR 97202, USA Portland Oregon USA
| |
Collapse
|
26
|
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: 1.0] [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
|
27
|
Li N, Xiao K, Mi X, Li N, Guo L, Wang X, Sun Y, Li GD, Zhou Y. Ghrelin signaling in dCA1 suppresses neuronal excitability and impairs memory acquisition via PI3K/Akt/GSK-3β cascades. Neuropharmacology 2022; 203:108871. [PMID: 34742928 DOI: 10.1016/j.neuropharm.2021.108871] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 12/22/2022]
Abstract
Ghrelin is a circulating peptide hormone that promotes feeding and regulates metabolism in humans and rodents. The action of ghrelin is mediated by the growth hormone secretagogue receptor type 1a (GHSR-1a) that is widely distributed in the brain, including the hippocampus. Studies have demonstrated the critical role of hippocampal ghrelin/GHS-R1a signaling in synaptic physiology and memory. However, those findings are controversial, and the mechanism underlying ghrelin modulation of learning and memory is uncertain. Here, we report that micro-infusion of ghrelin in the CA1 region of the dorsal hippocampus during training specifically impairs memory acquisition. The activation of GHS-R1a and the subsequent PI3K/Akt/GSK3β signaling cascades are involved in this process. Moreover, we report that bath application of ghrelin suppresses the intrinsic excitability of dCA1 pyramidal neurons through activating GHS-R1a, and PI3K inhibitor LY294002 blocks ghrelin's effect. However, LY294002 fails to rescue ghrelin-induced LTP impairment. Our findings support an adverse effect of ghrelin-dependent activation of GHS-R1a on memory acquisition, and suggest that PI3K/Akt/GSK3β signaling-dependent repression of neuronal intrinsic excitability is an important novel mechanism underlying memory inhibition of ghrelin in the hippocampus.
Collapse
Affiliation(s)
- Nan Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
| | - Kewei Xiao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
| | - Xue Mi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
| | - Na Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
| | - Li Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
| | - Xiaorong Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, United States
| | - Guo-Dong Li
- Department of Surgery, Valley Presbyterian Hospital, Van Nuys, CA, 91405, United States
| | - Yu Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China; Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China; Department of Rehabilitation Medicine, Affiliated Hospital of Qingdao University, Qingdao, Shangdong, 266000, China.
| |
Collapse
|
28
|
Abstract
The stomach hormone, ghrelin, which is released during food restriction, provides a link between circulating energy state and adaptive brain function. The maintenance of such homeostatic systems is essential for an organism to survive and thrive, and accumulating evidence points to ghrelin being a key regulator of adult hippocampal neurogenesis and memory function. Aberrant neurogenesis is linked to cognitive decline in aging and neurodegeneration. Therefore, identifying endogenous metabolic factors that regulate new adult-born neuron formation is an important objective in understanding the link between nutritional status and CNS function. Here, we review current developments in our understanding of ghrelin's role in regulating neurogenesis and memory function.
Collapse
Affiliation(s)
- Jeffrey S Davies
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, United Kingdom.
| |
Collapse
|
29
|
Ghrelin Is Effective on Passive Avoidance Memory by Altering the Expression of NMDAR and HTR1a Genes in the Hippocampus of Male Wistar Rats. Rep Biochem Mol Biol 2022; 10:380-386. [PMID: 34981014 DOI: 10.52547/rbmb.10.3.380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/18/2021] [Indexed: 11/18/2022]
Abstract
Background Memory-dependent psychological behaviors have an important role in life. Memory strengthening in adulthood to prevent its defects in aging is a significant issue. The ghrelin endogenous hormone improves memory by targeting glutamatergic and serotonergic circuits. Also, citicoline, a memory strengthening drug in aging, is not recommended to adults due to its side effects. The current study aims to test that ghrelin treatment, like citicoline, would improve passive avoidance memory via expression of the genes encoding the N-methyl-D-aspartate receptor (NMDAR1) and the serotonin receptor 1A (HTR1α) involved in this process. Methods Five groups of adult male rats received (1) saline (as control), (2) 0.5 mg/kg citicoline, or (3-5) 0.3, 1.5, and 3 nmol/μl ghrelin). The rats received the drugs via intra-hippocampal injection. Passive avoidance memory was determined using a shuttle box device. The latency to enter the dark chamber before (IL) and after (RL) injection and the total duration of the animal's presence in the light compartment (TLC) were evaluated. Then, the gene expression rates of NMDAR1 and HTR1α were measured by the Real-Time PCR. Results Ghrelin and citicoline had some similar and significant effects on passive avoidance memory, and both increased NMDAR1 and decreased HTR1α expression. Conclusion Ghrelin, like citicoline, improves passive avoidance learning by altering the NMDAR1 and HTR1α expression in the hippocampus.
Collapse
|
30
|
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
|
31
|
Farzin M, Babaei P, Rostampour M. Intrahippocampal Injections of Ghrelin and Aerobic Physical Exercise: Effects on Learning and Passive Avoidance Memory in Rats. NEUROPHYSIOLOGY+ 2021. [DOI: 10.1007/s11062-021-09912-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
32
|
Spiridon IA, Ciobanu DGA, Giușcă SE, Căruntu ID. Ghrelin and its role in gastrointestinal tract tumors (Review). Mol Med Rep 2021; 24:663. [PMID: 34296307 PMCID: PMC8335721 DOI: 10.3892/mmr.2021.12302] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 06/23/2021] [Indexed: 12/13/2022] Open
Abstract
Ghrelin, an orexigenic hormone, is a peptide that binds to the growth hormone secretagogue receptor; it is secreted mainly by enteroendocrine cells in the oxyntic glands of the stomach. Ghrelin serves a role in both local and systemic physiological processes, and is implicated in various pathologies, including neoplasia, with tissue expression in several types of malignancies in both in vitro and in vivo studies. However, the precise implications of the ghrelin axis in metastasis, invasion and cancer progression regulation has yet to be established. In the case of gastrointestinal (GI) tract malignancies, ghrelin has shown potential to become a prognostic factor or even a therapeutic target, although data in the literature are inconsistent and unsystematic, with reports untailored to a specific histological subtype of cancer or a particular localization. The evaluation of immunohistochemical expression shows a limited outlook owing to the low number of cases analyzed, and in vivo analyses have conflicting data regarding differences in ghrelin serum levels in patients with cancer. The aim of this review was to examine the relationship between ghrelin and GI tract malignancies to demonstrate the inconsistencies in current results and to highlight its clinical significance in the outcome of these patients.
Collapse
Affiliation(s)
- Irene Alexandra Spiridon
- Department of Pathology, 'Grigore T. Popa' University of Medicine and Pharmacy, Iași 700115, Romania
| | | | - Simona Eliza Giușcă
- Department of Pathology, 'Grigore T. Popa' University of Medicine and Pharmacy, Iași 700115, Romania
| | - Irina Draga Căruntu
- Department of Histology, 'Grigore T. Popa' University of Medicine and Pharmacy, Iași 700115, Romania
| |
Collapse
|
33
|
Beheshti S, Dehestani H. Differential expression levels of the hippocampal ghrelin and its receptor mRNA during memory consolidation. Behav Brain Res 2021; 408:113270. [PMID: 33811951 DOI: 10.1016/j.bbr.2021.113270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 03/23/2021] [Accepted: 03/28/2021] [Indexed: 12/30/2022]
Abstract
Ghrelin is a peptide, secreted mainly from the stomach. But, it is also produced in the brain. Studies have confirmed the positive impact of ghrelin on memory formation. However, the expression levels of ghrelin or its receptors were not measured in the brain during the process of memory formation. The probable alteration in the expression levels of ghrelin or its receptors in the brain during memory formation can be a reason for the contribution of its signaling in this process. We quantified the gene expression levels of ghrelin and its receptors in the hippocampus during fear and spatial memory consolidation. Thirty- nine adult male Wistar rats weighing 180-220 g were utilized. Memory consolidation was evaluated using the inhibitory avoidance task and Morris water maze. Rats were euthanized at different times (1, 3, and 24 h) post-training and their hippocampi were removed and freezed directly in liquid nitrogen. Quantitative real-time polymerize chain reaction (PCR) was used to quantify the messenger ribonucleic acid (mRNA) expression levels of the hippocampal ghrelin and its receptors. The mRNA levels of ghrelin exhibited a significant increase, 24 h post-training in the inhibitory avoidance task, while its receptor levels were down-regulated. Also, the mRNA expression levels of the hippocampal ghrelin were not changed significantly during memory consolidation in the Morris water maze, while its receptor showed a significant increase, 24 h post-training. The results show a differential profile of the expression levels of the hippocampal ghrelin or its receptor mRNA during fear or spatial memory consolidation. This proposes that a local increase in the hippocampal ghrelin or its receptor levels might be crucial for fear, and spatial memory consolidation. However, due to the small sample sizes, it is worth noting the preliminary nature of the conclusions in the present study.
Collapse
Affiliation(s)
- Siamak Beheshti
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Hadi Dehestani
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
34
|
Seitz BM, Tomiyama AJ, Blaisdell AP. Eating behavior as a new frontier in memory research. Neurosci Biobehav Rev 2021; 127:795-807. [PMID: 34087276 DOI: 10.1016/j.neubiorev.2021.05.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/15/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023]
Abstract
The study of memory is commonly associated with neuroscience, aging, education, and eyewitness testimony. Here we discuss how eating behavior is also heavily intertwined-and yet considerably understudied in its relation to memory processes. Both are influenced by similar neuroendocrine signals (e.g., leptin and ghrelin) and are dependent on hippocampal functions. While learning processes have long been implicated in influencing eating behavior, recent research has shown how memory of recent eating modulates future consumption. In humans, obesity is associated with impaired memory performance, and in rodents, dietary-induced obesity causes rapid decrements to memory. Lesions to the hippocampus disrupt memory but also induce obesity, highlighting a cyclic relationship between obesity and memory impairment. Enhancing memory of eating has been shown to reduce future eating and yet, little is known about what influences memory of eating or how memory of eating differs from memory for other behaviors. We discuss recent advancements in these areas and highlight fruitful research pursuits afforded by combining the study of memory with the study of eating behavior.
Collapse
|
35
|
Ghrelin restores memory impairment following olfactory bulbectomy in mice by activating hippocampal NMDA1 and MAPK1 gene expression. Behav Brain Res 2021; 410:113341. [PMID: 33964353 DOI: 10.1016/j.bbr.2021.113341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 12/21/2022]
Abstract
Ghrelin (Ghrl) is an orexigenic peptide with potential roles in the modulation of anxiety- and depressive-like symptoms induced by bilateral olfactory bulbectomy (OB) in rodents. In the present work, we assessed whether intrahippocampal Ghrl could reverse OB-induced depressive-like and amnesic effects by regulating molecular mechanisms related to neuroplasticity. Adult female albino Swiss mice were divided into sham and OB groups, and infused with saline (S) or Ghrl 0.03 nmol/μl, 0.3 nmol/μl, or 3 nmol/μl into the hippocampus before exposition to open-field test (OFT) and tail suspension test (TST) or immediately after training in the object recognition test (ORT). After test phase in ORT, animals were euthanized and their hippocampi were dissected to study the expression of genes related to memory. The OB-S animals presented hyperlocomotion in OFT, increased immobility in TST and memory impairment compared to sham-S (p < 0.05), but acute intrahippocampal infusion of Ghrl 0.3 nmol/μl produced an improvement on these parameters in OB animals (p < 0.05). In addition, this dose of Ghrl reversed OB-induced low expression of NMDA1 and MAPK1 iso1 and up-regulated the expression of CaMKIIa iso1 and iso2, and MAPK1 iso2 (p < 0.05). These results extend the existing literature regarding OB-induced behavioral and neurochemical changes, and provide mechanisms that could underlie the antidepressant effect of Ghrl in this model.
Collapse
|
36
|
Shi X, Guan K, Peng X, Xu B, Zhou X, Wang S, Xu S, Zheng M, Huang J, Wan X, Guan W, Su KP, Ye M, Gao X, Yin Z, Li X. Ghrelin modulates dopaminergic neuron formation and attention deficit hyperactivity disorder-like behaviors: From animals to human models. Brain Behav Immun 2021; 94:327-337. [PMID: 33412253 DOI: 10.1016/j.bbi.2020.12.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 10/22/2022] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is one of the most prevalent psychiatric disorders in children. The orexigenic hormone ghrelin is important in neuroprotection and neurodevelopment, which may play an important role in psychopathogenesis of ADHD. This study aimed to systematically investigate the genomic and pharmacological manipulations of ghrelin functioning in ADHD-like symptoms in zebrafish models and validated the effects of ghrelin polymorphisms in human subjects with ADHD. We firstly generated ghrelinΔ/Δ zebrafish mutant, which displayed hyperactive, attention deficit-like and impulsive-like behaviors, as well as endophenotypes, mimicking human ADHD. GhrelinΔ/Δ zebrafish exhibited downregulated expression levels of wnt1, wnt3a, wnt5a that are critical for dopaminergic neuron development to possibly regulate their number and spatial organization. Pharmacological blockade of wnt signaling with XAV939 induced a reduced moving activity and less dopaminergic neurons; whereas, wnt agonist SB415286 rescued hyperactivity and dopaminergic neuron loss in ghrelinΔ/Δ zebrafish. In addition, we further identified and validated a SNP, rs696217, on orexigenic hormone preproghrelin/ghrelin (T408T, Met72Met) to be associated with a higher risk of ADHD in a case-controlled association study with 248 subjects with ADHD and 208 subjects of healthy controls. Together, our results reveal a novel endogenous role for orexigenic hormone ghrelin in ADHD, which provides insights into genetic regulation and drug screens for the identification of novel treatments of ADHD.
Collapse
Affiliation(s)
- Xulai Shi
- The Affiliated Kangning Hospital of Wenzhou Medical University, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, PR China
| | - Kaiyu Guan
- The Affiliated Kangning Hospital of Wenzhou Medical University, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, PR China
| | - Xuyan Peng
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, PR China
| | - Bingru Xu
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, PR China
| | - Xianyong Zhou
- The Affiliated Kangning Hospital of Wenzhou Medical University, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, PR China
| | - Shao Wang
- The Affiliated Kangning Hospital of Wenzhou Medical University, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, PR China
| | - Shengnan Xu
- The Affiliated Kangning Hospital of Wenzhou Medical University, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, PR China
| | - Miaomiao Zheng
- The Affiliated Kangning Hospital of Wenzhou Medical University, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, PR China
| | - Jing Huang
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Xiaoyang Wan
- Institute of Infectious Liver Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Wanchun Guan
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, PR China
| | - Kuan-Pin Su
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; An-Nan Hospital, China Medical University, Tainan, Taiwan
| | - Minjie Ye
- The Affiliated Kangning Hospital of Wenzhou Medical University, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, PR China
| | - Xiang Gao
- Central Laboratory, Scientific Research Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Zhan Yin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, PR China.
| | - Xi Li
- The Affiliated Kangning Hospital of Wenzhou Medical University, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, PR China.
| |
Collapse
|
37
|
Stoyanova I, Lutz D. Ghrelin-Mediated Regeneration and Plasticity After Nervous System Injury. Front Cell Dev Biol 2021; 9:595914. [PMID: 33869167 PMCID: PMC8046019 DOI: 10.3389/fcell.2021.595914] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 02/24/2021] [Indexed: 12/17/2022] Open
Abstract
The nervous system is highly vulnerable to different factors which may cause injury followed by an acute or chronic neurodegeneration. Injury involves a loss of extracellular matrix integrity, neuronal circuitry disintegration, and impairment of synaptic activity and plasticity. Application of pleiotropic molecules initiating extracellular matrix reorganization and stimulating neuronal plasticity could prevent propagation of the degeneration into the tissue surrounding the injury. To find an omnipotent therapeutic molecule, however, seems to be a fairly ambitious task, given the complex demands of the regenerating nervous system that need to be fulfilled. Among the vast number of candidates examined so far, the neuropeptide and hormone ghrelin holds within a very promising therapeutic potential with its ability to cross the blood-brain barrier, to balance metabolic processes, and to stimulate neurorepair and neuroactivity. Compared with its well-established systemic effects in treatment of metabolism-related disorders, the therapeutic potential of ghrelin on neuroregeneration upon injury has received lesser appreciation though. Here, we discuss emerging concepts of ghrelin as an omnipotent player unleashing developmentally related molecular cues and morphogenic cascades, which could attenuate and/or counteract acute and chronic neurodegeneration.
Collapse
Affiliation(s)
- Irina Stoyanova
- Department of Anatomy and Cell Biology, Medical University Varna, Varna, Bulgaria
| | - David Lutz
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
38
|
Ribeiro LF, Catarino T, Carvalho M, Cortes L, Santos SD, Opazo PO, Ribeiro LR, Oliveiros B, Choquet D, Esteban JA, Peça J, Carvalho AL. Ligand-independent activity of the ghrelin receptor modulates AMPA receptor trafficking and supports memory formation. Sci Signal 2021; 14:14/670/eabb1953. [PMID: 33593997 DOI: 10.1126/scisignal.abb1953] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The biological signals of hunger, satiety, and memory are interconnected. The role of the hormone ghrelin in regulating feeding and memory makes ghrelin receptors attractive targets for associated disorders. We investigated the effects of the high ligand-independent activity of the ghrelin receptor GHS-R1a on the physiology of excitatory synapses in the hippocampus. Blocking this activity produced a decrease in the synaptic content of AMPA receptors in hippocampal neurons and a reduction in GluA1 phosphorylation at Ser845 Reducing the ligand-independent activity of GHS-R1a increased the surface diffusion of AMPA receptors and impaired AMPA receptor-dependent synaptic delivery induced by chemical long-term potentiation. Accordingly, we found that blocking this GHS-R1a activity impaired spatial and recognition memory in mice. These observations support a role for the ligand-independent activity of GHS-R1a in regulating AMPA receptor trafficking under basal conditions and in the context of synaptic plasticity that underlies learning.
Collapse
Affiliation(s)
- Luís F Ribeiro
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.
| | - Tatiana Catarino
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,University of Coimbra, IIIUC-Institute for Interdisciplinary Research, 3030-789 Coimbra, Portugal
| | - Mário Carvalho
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,MIT-Portugal Bioengineering Systems Doctoral Program, NOVA University of Lisbon, 1099-85, Lisboa, Portugal
| | - Luísa Cortes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,University of Coimbra, IIIUC-Institute for Interdisciplinary Research, 3030-789 Coimbra, Portugal
| | - Sandra D Santos
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,University of Coimbra, IIIUC-Institute for Interdisciplinary Research, 3030-789 Coimbra, Portugal
| | - Patricio O Opazo
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, 33000 Bordeaux, France.,CNRS, UMR 5297, 33000 Bordeaux, France
| | - Lyn Rosenbrier Ribeiro
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D AstraZeneca, Cambridge CB2 0SL, UK
| | - Bárbara Oliveiros
- Laboratory of Biostatistics and Medical Informatics (LBIM), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Daniel Choquet
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, 33000 Bordeaux, France.,CNRS, UMR 5297, 33000 Bordeaux, France.,Bordeaux Imaging Center, UMS 3420, CNRS-Bordeaux University, US4 INSERM, 33000 Bordeaux, France
| | - José A Esteban
- Department of Molecular Neurobiology, Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - João Peça
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,University of Coimbra, Department of Life Sciences, 3000-456 Coimbra, Portugal
| | - Ana Luísa Carvalho
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal. .,University of Coimbra, Department of Life Sciences, 3000-456 Coimbra, Portugal
| |
Collapse
|
39
|
Abstract
Eating food is one of the most complicated behaviours in mammals, especially humans. The primary function of ghrelin is regulation of the appetite level and its stimulation. It is also responsible for the body's energy balance and glucose homeostasis. Ghrelin has been shown to affect many brain structures, which confirms the presence of ghrelin receptors in the brain. Studies are also conducted to assess the possible role of ghrelin in anxiety states and in memory disorders and motor dysfunctions. Ghrelin has been found in saliva and salivary glands, teeth and gums, and in the taste buds of the tongue epithelium; it is also secreted by mucosal cells and gingival fibroblasts. The presence of ghrelin in developmental enamel, especially in odontoblasts and ameloblasts, may suggest its regulatory role in the development of teeth. Patients with chronic periodontitis have significantly higher concentrations of ghrelin in the peripheral blood serum, as compared to the control group. Ghrelin plays a special role in the proliferation of cancer cells and in the development of neoplastic metastases. The abundant presence of ghrelin receptors in cancer cells is considered an important target in the treatment of neoplasms. Ghrelin is a hormone whose multidirectional mechanism of action has not yet been fully understood. However, its ubiquitous occurrence in the human body and its very diverse participation in metabolic processes may prove to be a significant obstacle in achieving the expected clinical effect of ghrelin as an effective drug in selected disease units.
Collapse
|
40
|
Khelifa MS, Skov LJ, Holst B. Biased Ghrelin Receptor Signaling and the Dopaminergic System as Potential Targets for Metabolic and Psychological Symptoms of Anorexia Nervosa. Front Endocrinol (Lausanne) 2021; 12:734547. [PMID: 34646236 PMCID: PMC8503187 DOI: 10.3389/fendo.2021.734547] [Citation(s) in RCA: 2] [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: 07/01/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022] Open
Abstract
Anorexia Nervosa (AN) is a complex disease that impairs the metabolic, mental and physiological health of affected individuals in a severe and sometimes lethal way. Many of the common symptoms in AN patients, such as reduced food intake, anxiety, impaired gut motility or overexercising are connected to both the orexigenic gut hormone ghrelin and the dopaminergic system. Targeting the ghrelin receptor (GhrR) to treat AN seems a promising possibility in current research. However, GhrR signaling is highly complex. First, the GhrR can activate four known intracellular pathways Gαq, Gαi/o, Gα12/13 and the recruitment of β-arrestin. Biased signaling provides the possibility to activate or inhibit only one or a subset of the intracellular pathways of a pleiotropic receptor. This allows specific targeting of physiological functions without adverse effects. Currently little is known on how biased signaling could specifically modulate GhrR effects. Second, GhrR signaling has been shown to be interconnected with the dopaminergic system, particularly in the context of AN symptoms. This review highlights that a biased agonist for the GhrR may be a promising target for the treatment of AN, however extensive and systematic translational studies are still needed and the connection to the dopaminergic system has to be taken into account.
Collapse
|
41
|
Reich N, Hölscher C. Acylated Ghrelin as a Multi-Targeted Therapy for Alzheimer's and Parkinson's Disease. Front Neurosci 2020; 14:614828. [PMID: 33381011 PMCID: PMC7767977 DOI: 10.3389/fnins.2020.614828] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Much thought has been given to the impact of Amyloid Beta, Tau and Alpha-Synuclein in the development of Alzheimer's disease (AD) and Parkinson's disease (PD), yet the clinical failures of the recent decades indicate that there are further pathological mechanisms at work. Indeed, besides amyloids, AD and PD are characterized by the culminative interplay of oxidative stress, mitochondrial dysfunction and hyperfission, defective autophagy and mitophagy, systemic inflammation, BBB and vascular damage, demyelination, cerebral insulin resistance, the loss of dopamine production in PD, impaired neurogenesis and, of course, widespread axonal, synaptic and neuronal degeneration that leads to cognitive and motor impediments. Interestingly, the acylated form of the hormone ghrelin has shown the potential to ameliorate the latter pathologic changes, although some studies indicate a few complications that need to be considered in the long-term administration of the hormone. As such, this review will illustrate the wide-ranging neuroprotective properties of acylated ghrelin and critically evaluate the hormone's therapeutic benefits for the treatment of AD and PD.
Collapse
Affiliation(s)
- Niklas Reich
- Biomedical & Life Sciences Division, Lancaster University, Lancaster, United Kingdom
| | - Christian Hölscher
- Neurology Department, A Second Hospital, Shanxi Medical University, Taiyuan, China.,Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
42
|
Cornejo MP, Mustafá ER, Barrile F, Cassano D, De Francesco PN, Raingo J, Perello M. THE INTRIGUING LIGAND-DEPENDENT AND LIGAND-INDEPENDENT ACTIONS OF THE GROWTH HORMONE SECRETAGOGUE RECEPTOR ON REWARD-RELATED BEHAVIORS. Neurosci Biobehav Rev 2020; 120:401-416. [PMID: 33157147 DOI: 10.1016/j.neubiorev.2020.10.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023]
Abstract
The growth hormone secretagogue receptor (GHSR) is a G-protein-coupled receptor (GPCR) highly expressed in the brain, and also in some peripheral tissues. GHSR activity is evoked by the stomach-derived peptide hormone ghrelin and abrogated by the intestine-derived liver-expressed antimicrobial peptide 2 (LEAP2). In vitro, GHSR displays ligand-independent actions, including a high constitutive activity and an allosteric modulation of other GPCRs. Beyond its neuroendocrine and metabolic effects, cumulative evidence shows that GHSR regulates the activity of the mesocorticolimbic pathway and modulates complex reward-related behaviors towards different stimuli. Here, we review current evidence indicating that ligand-dependent and ligand-independent actions of GHSR enhance reward-related behaviors towards appetitive stimuli and drugs of abuse. We discuss putative neuronal networks and molecular mechanisms that GHSR would engage to modulate such reward-related behaviors. Finally, we briefly discuss imaging studies showing that ghrelin would also regulate reward processing in humans. Overall, we conclude that GHSR is a key regulator of the mesocorticolimbic pathway that influences its activity and, consequently, modulates reward-related behaviors via ligand-dependent and ligand-independent actions.
Collapse
Affiliation(s)
- María P Cornejo
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA). National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Emilio R Mustafá
- Laboratory of Electrophysiology of the IMBICE, 1900 La Plata, Buenos Aires, Argentina
| | - Franco Barrile
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA). National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Daniela Cassano
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA). National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Pablo N De Francesco
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA). National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Jesica Raingo
- Laboratory of Electrophysiology of the IMBICE, 1900 La Plata, Buenos Aires, Argentina
| | - Mario Perello
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA). National University of La Plata], 1900 La Plata, Buenos Aires, Argentina.
| |
Collapse
|
43
|
Fritz EM, Singewald N, De Bundel D. The Good, the Bad and the Unknown Aspects of Ghrelin in Stress Coping and Stress-Related Psychiatric Disorders. Front Synaptic Neurosci 2020; 12:594484. [PMID: 33192444 PMCID: PMC7652849 DOI: 10.3389/fnsyn.2020.594484] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022] Open
Abstract
Ghrelin is a peptide hormone released by specialized X/A cells in the stomach and activated by acylation. Following its secretion, it binds to ghrelin receptors in the periphery to regulate energy balance, but it also acts on the central nervous system where it induces a potent orexigenic effect. Several types of stressors have been shown to stimulate ghrelin release in rodents, including nutritional stressors like food deprivation, but also physical and psychological stressors such as foot shocks, social defeat, forced immobilization or chronic unpredictable mild stress. The mechanism through which these stressors drive ghrelin release from the stomach lining remains unknown and, to date, the resulting consequences of ghrelin release for stress coping remain poorly understood. Indeed, ghrelin has been proposed to act as a stress hormone that reduces fear, anxiety- and depression-like behaviors in rodents but some studies suggest that ghrelin may - in contrast - promote such behaviors. In this review, we aim to provide a comprehensive overview of the literature on the role of the ghrelin system in stress coping. We discuss whether ghrelin release is more than a byproduct of disrupted energy homeostasis following stress exposure. Furthermore, we explore the notion that ghrelin receptor signaling in the brain may have effects independent of circulating ghrelin and in what way this might influence stress coping in rodents. Finally, we examine how the ghrelin system could be utilized as a therapeutic avenue in stress-related psychiatric disorders (with a focus on anxiety- and trauma-related disorders), for example to develop novel biomarkers for a better diagnosis or new interventions to tackle relapse or treatment resistance in patients.
Collapse
Affiliation(s)
- Eva Maria Fritz
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Innsbruck, Austria
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Innsbruck, Austria
| | - Dimitri De Bundel
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
44
|
Iyer MR, Wood CM, Kunos G. Recent progress in the discovery of ghrelin O-acyltransferase (GOAT) inhibitors. RSC Med Chem 2020; 11:1136-1144. [PMID: 33479618 PMCID: PMC7651998 DOI: 10.1039/d0md00210k] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/03/2020] [Indexed: 12/29/2022] Open
Abstract
Ghrelin is a stomach-derived peptide hormone which stimulates appetite. For ghrelin to exert its orexigenic effect, octanoylation on the serine-3 residue of this gut-brain peptide is essential. The octanoylation of ghrelin is mediated by a unique acyltransferase enzyme known as ghrelin O-acyltransferase (GOAT). Thus modulating this enzyme offers viable approaches to alter feeding behaviors. Over the past decade, several small-molecule based approaches have appeared dealing with the discovery of compounds able to modulate this enzyme for the treatment of obesity and type 2 diabetes. Drug discovery efforts from academic groups and several pharmaceutical companies have fielded compounds having efficacy in altering acylated ghrelin levels in animal models but to date, compounds modulating the activity of the GOAT enzyme do not yet represent clinical options. This mini-review covers the drug discovery approaches of the last decade since the discovery of the GOAT enzyme.
Collapse
Affiliation(s)
- Malliga R Iyer
- Medicinal Chemistry Core and Laboratory of Physiologic Studies , National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health (NIAAA/NIH) , 5625 Fishers Lane , Rockville , MD 20852 , USA . ; Tel: +301 443 2807
| | - Casey M Wood
- Medicinal Chemistry Core and Laboratory of Physiologic Studies , National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health (NIAAA/NIH) , 5625 Fishers Lane , Rockville , MD 20852 , USA . ; Tel: +301 443 2807
| | - George Kunos
- Medicinal Chemistry Core and Laboratory of Physiologic Studies , National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health (NIAAA/NIH) , 5625 Fishers Lane , Rockville , MD 20852 , USA . ; Tel: +301 443 2807
| |
Collapse
|
45
|
Beheshti S, Sami M, Mirzabeh A, Yazdi A. D-Lys-3-GHRP-6 impairs memory consolidation and downregulates the hippocampal serotonin HT1A, HT7 receptors and glutamate GluA1 subunit of AMPA receptors. Physiol Behav 2020; 223:112969. [DOI: 10.1016/j.physbeh.2020.112969] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 04/27/2020] [Accepted: 05/12/2020] [Indexed: 01/08/2023]
|
46
|
Autio J, Stenbäck V, Gagnon DD, Leppäluoto J, Herzig KH. (Neuro) Peptides, Physical Activity, and Cognition. J Clin Med 2020; 9:jcm9082592. [PMID: 32785144 PMCID: PMC7464334 DOI: 10.3390/jcm9082592] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023] Open
Abstract
Regular physical activity (PA) improves cognitive functions, prevents brain atrophy, and delays the onset of cognitive decline, dementia, and Alzheimer’s disease. Presently, there are no specific recommendations for PA producing positive effects on brain health and little is known on its mediators. PA affects production and release of several peptides secreted from peripheral and central tissues, targeting receptors located in the central nervous system (CNS). This review will provide a summary of the current knowledge on the association between PA and cognition with a focus on the role of (neuro)peptides. For the review we define peptides as molecules with less than 100 amino acids and exclude myokines. Tachykinins, somatostatin, and opioid peptides were excluded from this review since they were not affected by PA. There is evidence suggesting that PA increases peripheral insulin growth factor 1 (IGF-1) levels and elevated serum IGF-1 levels are associated with improved cognitive performance. It is therefore likely that IGF-1 plays a role in PA induced improvement of cognition. Other neuropeptides such as neuropeptide Y (NPY), ghrelin, galanin, and vasoactive intestinal peptide (VIP) could mediate the beneficial effects of PA on cognition, but the current literature regarding these (neuro)peptides is limited.
Collapse
Affiliation(s)
- Juho Autio
- Institute of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, Oulu University Hospital, 90220 Oulu, Finland; (J.A.); (V.S.); (D.D.G.); (J.L.)
| | - Ville Stenbäck
- Institute of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, Oulu University Hospital, 90220 Oulu, Finland; (J.A.); (V.S.); (D.D.G.); (J.L.)
- Biocenter Oulu, 90220 Oulu, Finland
| | - Dominique D. Gagnon
- Institute of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, Oulu University Hospital, 90220 Oulu, Finland; (J.A.); (V.S.); (D.D.G.); (J.L.)
- Laboratory of Environmental Exercise Physiology, School of Human Kinetics, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Center of Research in Occupational Safety and Health, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Juhani Leppäluoto
- Institute of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, Oulu University Hospital, 90220 Oulu, Finland; (J.A.); (V.S.); (D.D.G.); (J.L.)
| | - Karl-Heinz Herzig
- Institute of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, Oulu University Hospital, 90220 Oulu, Finland; (J.A.); (V.S.); (D.D.G.); (J.L.)
- Department of Gastroenterology and Metabolism, Poznan University of Medical Sciences, 60-572 Poznan, Poland
- Correspondence:
| |
Collapse
|
47
|
Duriez P, Eddarkaoui S, Blum D, Dickson SL, Gorwood P, Tolle V, Viltart O. Does physical activity associated with chronic food restriction alleviate anxiety like behaviour, in female mice? Horm Behav 2020; 124:104807. [PMID: 32544401 DOI: 10.1016/j.yhbeh.2020.104807] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/17/2022]
Abstract
Anorexia nervosa (AN) is an eating disorder characterized by excessive weight loss, persistent food restriction and inappropriate physical activity relative to declining energy balance. The comorbidity with depression and/or anxiety disorders might contribute to the "chronicization" of the disease. We aimed here to question first the link between physical activity and anxiety from a clinical investigation of AN patients (n = 206). Then, using a rodent model mimicking numerous physiological and metabolic alterations commonly seen in AN patients, we examined whether 1) chronic food restriction increased anxiety-like behaviour and 2) physical activity plays a role in regulating anxiety levels. To this end, we exposed young female mice to a chronic food restriction (FR, n = 8) paradigm combined or not with access to a running wheel (FRW, n = 8) for two weeks. The mice were compared to a group of mice fed ad libitum without (AL, n = 6) or with running wheel access (ALW, n = 8). We explored anxiety-like behaviour of all mice in the following tests: hyponeophagia, marble burying, elevated plus maze, open field, and the light and dark box. On the last day, we used a restraint test of 30 min duration and measured their stress reactivity by assaying plasma corticosterone. In the open field and the elevated plus-maze, we found that FRW mice behaved similarly to AL and ALW mice whereas FR mice did not express anxiety-like behaviour. The FRW mice displayed the lowest latency to reach the food in the hyponeophagia test. Regarding stress reactivity, FRW mice exhibited corticosterone reactivity after acute stress that was similar to the control mice, while FR mice did not fully return to basal corticosterone at one hour after the restraint stress. Taken together, these data demonstrate a differential reactivity to acute stress in FR conditions and a beneficial effect of running wheel activity in ALW and FRW conditions. Moreover, we report the absence of a typical anxiety-like behaviour associated with the food restriction (FR and FRW groups). We conclude that this model (FR and FRW mice) did not express typical anxiety-like behaviour, but that physical activity linked to food restriction improved coping strategies in an anxiogenic context.
Collapse
Affiliation(s)
- Philibert Duriez
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP) INSERM U1266, "Vulnerability of Psychiatric and Addictive Disorders", F-75014 Paris, France; GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte-Anne, F-75014 Paris, France
| | - Sabiha Eddarkaoui
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France; Alzheimer & Tauopathies, LabEx DISTALZ, LiCEND, F-59000 Lille, France
| | - David Blum
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France; Alzheimer & Tauopathies, LabEx DISTALZ, LiCEND, F-59000 Lille, France
| | - Suzanne L Dickson
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, SE-405 30 Gothenburg, Sweden
| | - Philip Gorwood
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP) INSERM U1266, "Vulnerability of Psychiatric and Addictive Disorders", F-75014 Paris, France; GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte-Anne, F-75014 Paris, France
| | - Virginie Tolle
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP) INSERM U1266, "Vulnerability of Psychiatric and Addictive Disorders", F-75014 Paris, France
| | - Odile Viltart
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP) INSERM U1266, "Vulnerability of Psychiatric and Addictive Disorders", F-75014 Paris, France; Université de Lille, F-59650 Villeneuve d'Ascq, France.
| |
Collapse
|
48
|
Yadegary A, Nazari-Serenjeh F, Darbandi N. Synergistic improvement effect of nicotine-ghrelin co-injection into the anterior ventral tegmental area on morphine-induced amnesia. Neuropeptides 2020; 80:102025. [PMID: 32029269 DOI: 10.1016/j.npep.2020.102025] [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: 06/06/2019] [Revised: 01/15/2020] [Accepted: 01/29/2020] [Indexed: 11/15/2022]
Abstract
In the present study the effect of ghrelin or ghrelin/nicotine injection into the anterior ventral tegmental area (aVTA) on morphine-induced amnesia in passive avoidance learning have been evaluated. Also, the role of the aVTA nicotinic receptors in possible ghrelin-induced effects has been investigated. All animals were bilaterally implanted with chronic cannulas in the aVTA. A step-through type passive avoidance task was used for measurement of memory. We found that post-training subcutaneous (s.c.) injection of morphine (0.5-7.5 mg/kg) dose-dependently reduced the step-through latency, indicating morphine-induced amnesia. Post-training bilateral infusion of ghrelin (0.3, 1.5 and 3 nmol/μl) in a dose-dependent manner reversed amnesia induced by morphine (7.5 mg/kg, s.c.). Furthermore, reversal effect of ghrelin (3 nmol/μl) was blocked by pre-treatment of intra-aVTA administration of mecamylamine (1-3 μg/rat), a nicotinic acetylcholine receptor antagonist. Intra-aVTA administration of the higher dose of mecamylamine (3 μg/rat) into the aVTA by itself decreased the step-through latency and induced amnesia. In addition, post-training intra-aVTA administration of nicotine (0.25, 0.5, 1 μg/rat) which alone cannot affect memory consolidation, decreased significantly the amnesia induced by morphine (7.5 mg/kg, s.c.). Co-treatment of an ineffective dose of ghrelin (0.3 nmol/μl) with an ineffective dose of nicotine (0.25 μg/rat) significantly increased step-through latency of morphine (7.5 mg/kg, s.c.) treated animals, indicating the synergistic effect of the drugs. Taken together, our results suggest that intra-aVTA administration of ghrelin reversed morphine-induced amnesia and that ghrelin interacts synergistically with nicotine to mitigate morphine-induced amnesia.
Collapse
Affiliation(s)
- Atena Yadegary
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| | | | - Niloufar Darbandi
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| |
Collapse
|
49
|
Qi XR, Zhang L. The Potential Role of Gut Peptide Hormones in Autism Spectrum Disorder. Front Cell Neurosci 2020; 14:73. [PMID: 32296309 PMCID: PMC7136424 DOI: 10.3389/fncel.2020.00073] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/12/2020] [Indexed: 12/11/2022] Open
Abstract
Gut peptide hormones are one group of secretory factors produced from gastrointestinal endocrine cells with potent functions in modulating digestive functions. In recent decades, they have been found across different brain regions, many of which are involved in autism-related social, emotional and cognitive deficits. Clinical studies have revealed possible correlation between those hormones and autism spectrum disorder pathogenesis. In animal models, gut peptide hormones modulate neurodevelopment, synaptic transmission and neural plasticity, explaining their behavioral relevance. This review article will summarize major findings from both clinical and basic research showing the role of gut peptide hormones in mediating autism-related neurological functions, and their potential implications in autism pathogenesis. The pharmaceutical value of gut hormones in alleviating autism-associated behavioral syndromes will be discussed to provide new insights for future drug development.
Collapse
Affiliation(s)
- Xin-Rui Qi
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Li Zhang
- Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| |
Collapse
|
50
|
Méquinion M, Foldi CJ, Andrews ZB. The Ghrelin-AgRP Neuron Nexus in Anorexia Nervosa: Implications for Metabolic and Behavioral Adaptations. Front Nutr 2020; 6:190. [PMID: 31998738 PMCID: PMC6962137 DOI: 10.3389/fnut.2019.00190] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 12/11/2019] [Indexed: 11/13/2022] Open
Abstract
Anorexia Nervosa (AN) is viewed as primarily a psychiatric disorder owing to the considerable behavioral and genetic overlap with mood disorders and other psychiatric traits. However, the recent reconceptualization of AN as one of both psychiatric and metabolic etiology suggests that metabolic circuits conveying hunger, or sensitive to signals of hunger, may be a critical nexus linking metabolic dysfunction to mood disturbances. Within the brain, hunger is primarily percieved by Agouti-related (AgRP) neurons and hunger increases plasma concentrations of the hormone ghrelin, which targets ghrelin receptors on AgRP neurons to facilitate metabolic adaptations to low energy availability. However, beyond the fundamental role in maintaining hunger signaling, AgRP neurons regulate a diverse range of behaviors such as motivation, locomotor activity, negative reinforcement, anxiety, and obsession and a key factor involved in the manifestation of these behavioral changes in response to activation is the presence or absence of food availability. These changes can be considered adaptive in that they promote affective food-seeking strategies in environments with limited food availability. However, it also suggests that these neurons, so well-studied for their metabolic control, shape mood-related behaviors in a context-dependent manner and dysfunctional control leads not only to metabolic problems but also potentially mood-related problems. The purpose of this review is to underline the potential role of AgRP neurons and ghrelin signaling in both the metabolic and behavioral changes observed in anorexia nervosa. We aim to highlight the most recent studies on AgRP neurons and ghrelin signaling and integrate their metabolic and behavioral roles in normal function and highlight how dysfunction may contribute to the development of AN.
Collapse
Affiliation(s)
| | | | - Zane B. Andrews
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| |
Collapse
|