1
|
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
|
2
|
Nowacka A, Getz AM, Bessa-Neto D, Choquet D. Activity-dependent diffusion trapping of AMPA receptors as a key step for expression of early LTP. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230220. [PMID: 38853553 PMCID: PMC11343219 DOI: 10.1098/rstb.2023.0220] [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: 02/24/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 06/11/2024] Open
Abstract
This review focuses on the activity-dependent diffusion trapping of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) as a crucial mechanism for the expression of early long-term potentiation (LTP), a process central to learning and memory. Despite decades of research, the precise mechanisms by which LTP induction leads to an increase in AMPAR responses at synapses have been elusive. We review the different hypotheses that have been put forward to explain the increased AMPAR responsiveness during LTP. We discuss the dynamic nature of AMPAR complexes, including their constant turnover and activity-dependent modifications that affect their synaptic accumulation. We highlight a hypothesis suggesting that AMPARs are diffusively trapped at synapses through activity-dependent interactions with protein-based binding slots in the post-synaptic density (PSD), offering a potential explanation for the increased synaptic strength during LTP. Furthermore, we outline the challenges still to be addressed before we fully understand the functional roles and molecular mechanisms of AMPAR dynamic nanoscale organization in LTP. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Agata Nowacka
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, BordeauxF-33000, France
| | - Angela M. Getz
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, BordeauxF-33000, France
- University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, BordeauxF-33000, France
| | - Diogo Bessa-Neto
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, BordeauxF-33000, France
| | - Daniel Choquet
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, BordeauxF-33000, France
- University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, BordeauxF-33000, France
| |
Collapse
|
3
|
Wee RWS, Mishchanchuk K, AlSubaie R, Church TW, Gold MG, MacAskill AF. Internal-state-dependent control of feeding behavior via hippocampal ghrelin signaling. Neuron 2024; 112:288-305.e7. [PMID: 37977151 DOI: 10.1016/j.neuron.2023.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/13/2023] [Accepted: 10/12/2023] [Indexed: 11/19/2023]
Abstract
Hunger is an internal state that not only invigorates feeding but also acts as a contextual cue for higher-order control of anticipatory feeding-related behavior. The ventral hippocampus is crucial for differentiating optimal behavior across contexts, but how internal contexts such as hunger influence hippocampal circuitry is unknown. In this study, we investigated the role of the ventral hippocampus during feeding behavior across different states of hunger in mice. We found that activity of a unique subpopulation of neurons that project to the nucleus accumbens (vS-NAc neurons) increased when animals investigated food, and this activity inhibited the transition to begin eating. Increases in the level of the peripheral hunger hormone ghrelin reduced vS-NAc activity during this anticipatory phase of feeding via ghrelin-receptor-dependent increases in postsynaptic inhibition and promoted the initiation of eating. Together, these experiments define a ghrelin-sensitive hippocampal circuit that informs the decision to eat based on internal state.
Collapse
Affiliation(s)
- Ryan W S Wee
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Karyna Mishchanchuk
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Rawan AlSubaie
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Timothy W Church
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Matthew G Gold
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Andrew F MacAskill
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK.
| |
Collapse
|
4
|
Tian J, Guo L, Wang T, Jia K, Swerdlow RH, Zigman JM, Du H. Liver-expressed antimicrobial peptide 2 elevation contributes to age-associated cognitive decline. JCI Insight 2023; 8:166175. [PMID: 37212281 DOI: 10.1172/jci.insight.166175] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/31/2023] [Indexed: 05/23/2023] Open
Abstract
Elderly individuals frequently report cognitive decline, while various studies indicate hippocampal functional declines with advancing age. Hippocampal function is influenced by ghrelin through hippocampus-expressed growth hormone secretagogue receptor (GHSR). Liver-expressed antimicrobial peptide 2 (LEAP2) is an endogenous GHSR antagonist that attenuates ghrelin signaling. Here, we measured plasma ghrelin and LEAP2 levels in a cohort of cognitively normal individuals older than 60 and found that LEAP2 increased with age while ghrelin (also referred to in literature as "acyl-ghrelin") marginally declined. In this cohort, plasma LEAP2/ghrelin molar ratios were inversely associated with Mini-Mental State Examination scores. Studies in mice showed an age-dependent inverse relationship between plasma LEAP2/ghrelin molar ratio and hippocampal lesions. In aged mice, restoration of the LEAP2/ghrelin balance to youth-associated levels with lentiviral shRNA Leap2 downregulation improved cognitive performance and mitigated various age-related hippocampal deficiencies such as CA1 region synaptic loss, declines in neurogenesis, and neuroinflammation. Our data collectively suggest that LEAP2/ghrelin molar ratio elevation may adversely affect hippocampal function and, consequently, cognitive performance; thus, it may serve as a biomarker of age-related cognitive decline. Moreover, targeting LEAP2 and ghrelin in a manner that lowers the plasma LEAP2/ghrelin molar ratio could benefit cognitive performance in elderly individuals for rejuvenation of memory.
Collapse
Affiliation(s)
- Jing Tian
- Department of Pharmacology and Toxicology and
| | - Lan Guo
- Higuchi Biosciences Center, University of Kansas, Lawrence, Kansas, USA
| | - Tienju Wang
- Department of Pharmacology and Toxicology and
| | - Kun Jia
- Department of Pharmacology and Toxicology and
| | - Russell H Swerdlow
- Alzheimer's Disease Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jeffrey M Zigman
- Departments of Internal Medicine and Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Heng Du
- Department of Pharmacology and Toxicology and
- Higuchi Biosciences Center, University of Kansas, Lawrence, Kansas, USA
- Alzheimer's Disease Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
5
|
Bessa-Neto D, Choquet D. Molecular mechanisms of AMPAR reversible stabilization at synapses. Mol Cell Neurosci 2023; 125:103856. [PMID: 37105372 DOI: 10.1016/j.mcn.2023.103856] [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: 11/07/2022] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
In the central nervous system, glutamatergic synapses play a central role in the regulation of excitatory neuronal transmission. With the membrane-associated guanylate kinase (MAGUK) family of proteins as their structuring scaffold, glutamatergic receptors serve as the powerhouse of glutamatergic synapses. Glutamatergic receptors can be categorized as metabotropic and ionotropic receptors. The latter are then categorized into N-methyl-d-aspartate, kainate receptors, and α-amino-3-hydroxy-5-methyl-isoxazole-propionic acid receptors (AMPARs). Over the past two decades, genetic tagging technology and super-resolution microscopy have been of the utmost importance to unravel how the different receptors are organized at glutamatergic synapses. At the plasma membrane, receptors are highly mobile but show reduced mobility when at synaptic sites. This partial immobilization of receptors at synaptic sites is attributed to the stabilization/anchoring of receptors with the postsynaptic MAGUK proteins and auxiliary proteins, and presynaptic proteins. These partial immobilizations and localization of glutamatergic receptors within the synaptic sites are fundamental for proper basal transmission and synaptic plasticity. Perturbations of the stabilization of glutamatergic receptors are often associated with cognitive deficits. In this review, we describe the proposed mechanisms for synaptic localization and stabilization of AMPARs, the major players of fast excitatory transmission in the central nervous system.
Collapse
Affiliation(s)
- Diogo Bessa-Neto
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| | - Daniel Choquet
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France; Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, F-33000 Bordeaux, France.
| |
Collapse
|
6
|
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
|
7
|
The effect of ghrelin on antioxidant status in the rat's model of Alzheimer's disease induced by amyloid-beta. Biomedicine (Taipei) 2023; 12:44-54. [PMID: 36816173 PMCID: PMC9910231 DOI: 10.37796/2211-8039.1341] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 11/27/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with amyloid-beta (Aβ) plaque formation and oxidative stress in the brain. Ghrelin has been proven to exert antioxidant activity and neuroprotection in different neurological diseases. This study is going on to examine the effect of ghrelin on antioxidant status in the rat's model of AD induced by Aβ. Cognitive impairment was induced by intra-hippocampal administration of Aβ (10 μg) in Wistar rats and ghrelin (80 μg/kg) was administrated intraperitoneal for ten consecutive days. Behavior was assessed with Morris water maze and passive avoidance tests. Malondialdehyde (MDA) level as a marker of lipid peroxidation was assessed using the thiobarbituric acid. Catalase activity was assayed by the decomposition of H2O2. Antioxidant capacity was determined using the FRAP method. Treatment with ghrelin decreased the hippocampus and serum MDA levels in wild-type rodents and prevented an increase in hippocampal and serum MDA levels in animals receiving Aβ. There was no significant change in the serum catalase activity between the studied groups. Hippocampus catalase activity was reduced in the Aβ group and treatment with ghrelin increased it. The antioxidant capacity of the hippocampus and serum increased in the ghrelin-receiving control group. The hippocampus antioxidant capacity level decreased in the Aβ group, and treatment with ghrelin increased it, but there were no significant changes in the serum antioxidant capacity of animals receiving Aβ. These results provide evidence that the administration of ghrelin has antioxidant properties and protects against hippocampal lipid peroxidation in a rat model of AD.
Collapse
|
8
|
Hirono M, Nakata M. Ghrelin signaling in the cerebellar cortex enhances GABAergic transmission onto Purkinje cells. Sci Rep 2023; 13:2150. [PMID: 36750743 PMCID: PMC9905081 DOI: 10.1038/s41598-023-29226-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Ghrelin, an orexigenic peptide ligand for growth hormone secretagogue receptor 1a (GHS-R1a), occurs not only in the stomach but also in the brain, and modulates neuronal activity and synaptic efficacy. Previous studies showed that GHS-R1a exists in the cerebellum, and ghrelin facilitates spontaneous firing of Purkinje cells (PCs). However, the effects of ghrelin on cerebellar GABAergic transmission have yet to be elucidated. We found that ghrelin enhanced GABAergic transmission between molecular layer interneurons (MLIs) and PCs using electrophysiological recordings in mouse cerebellar slices. This finding was consistent with the possibility that blocking synaptic transmission enhanced the ghrelin-induced facilitation of PC firing. Ghrelin profoundly increased the frequency of spontaneous inhibitory postsynaptic currents (IPSCs) in PCs without affecting miniature or stimulation-evoked IPSCs, whereas it significantly facilitated spontaneous firing of MLIs. This facilitation of MLI spiking disappeared during treatments with blockers of GHS-R1a, type 1 transient receptor potential canonical (TRPC1) channels and KCNQ channels. These results suggest that both activating TRPC1 channels and inhibiting KCNQ channels occur downstream the ghrelin-GHS-R1a signaling pathway probably in somatodendritic sites of MLIs. Thus, ghrelin can control PC firing directly and indirectly via its modulation of GABAergic transmission, thereby impacting activity in cerebellar circuitry.
Collapse
Affiliation(s)
- Moritoshi Hirono
- Department of Physiology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-8509, Japan.
| | - Masanori Nakata
- Department of Physiology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-8509, Japan
| |
Collapse
|
9
|
Ghrelin system in Alzheimer's disease. Curr Opin Neurobiol 2023; 78:102655. [PMID: 36527939 PMCID: PMC10395051 DOI: 10.1016/j.conb.2022.102655] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia in seniors. Current efforts to understand the etiopathogenesis of this neurodegenerative disorder have brought forth questions about systemic factors in the development of AD. Ghrelin is a brain-gut peptide that is activated by ghrelin O-acyltransferase (GOAT) and signals via its receptor, growth hormone secretagogue receptor (GHSR). With increasing recognition of the neurotropic effects of ghrelin, the role of ghrelin system deregulation in the development of AD has been accentuated in recent years. In this review, we summarized recent research progress regarding the mechanisms of ghrelin signaling dysregulation and its contribution to AD brain pathology. In addition, we also discussed the therapeutic potential of strategies targeting ghrelin signaling for the treatment of this neurological disease.
Collapse
|
10
|
Tian J, Du E, Jia K, Wang T, Guo L, Zigman JM, Du H. Elevated Ghrelin Promotes Hippocampal Ghrelin Receptor Defects in Humanized Amyloid-β Knockin Mice During Aging. J Alzheimers Dis 2023; 96:1579-1592. [PMID: 38007666 PMCID: PMC10841720 DOI: 10.3233/jad-231002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
BACKGROUND Emerging evidence has revealed that dysregulation of the hormone ghrelin and its receptor, growth hormone secretagogue receptor (GHSR), contributes to the pathogenesis of Alzheimer's disease (AD). Specifically, defective GHSR function and resultant hippocampal ghrelin resistance are linked to hippocampal synaptic injury in AD paradigms. Also, AD patients exhibit elevated ghrelin activation. However, the detailed molecular mechanisms of hippocampal GHSR dysfunction and the relevance of ghrelin elevation to hippocampal ghrelin resistance in AD-relevant pathological settings are not fully understood. OBJECTIVE In the current study, we employed a recently established mouse line of AD risk [humanized amyloid beta knockin (hAβ KI mice), also referred to as a mouse model of late-onset AD in previous literature] to further define the role of ghrelin system dysregulation in the development of AD. METHODS We employed multidisciplinary techniques to determine the change of plasma ghrelin and the functional status of GHSR in hAβ KI mice as well as primary neuron cultures. RESULTS We observed concurrent plasma ghrelin elevation and hippocampal GHSR desensitization with disease progression. Further examination excluded the possibility that ghrelin elevation is a compensatory change in response to GHSR dysfunction. In contrast, further in vitro and in vivo results show that agonist-mediated overstimulation potentiates GHSR desensitization through enhanced GHSR internalization. CONCLUSIONS These findings suggest that circulating ghrelin elevation is a pathological event underlying hippocampal GHSR dysfunction, culminating in hippocampal ghrelin resistance and resultant synaptic injury in late-onset AD-related settings.
Collapse
Affiliation(s)
- Jing Tian
- Department of Pharmacology and Toxicology, The University of Kansas, Lawrence, KS, USA
| | - Eric Du
- Department of Pharmacology and Toxicology, The University of Kansas, Lawrence, KS, USA
- Blue Valley West High School, Overland Park, KS, USA
| | - Kun Jia
- Department of Pharmacology and Toxicology, The University of Kansas, Lawrence, KS, USA
| | - Tienju Wang
- Department of Pharmacology and Toxicology, The University of Kansas, Lawrence, KS, USA
| | - Lan Guo
- Department of Pharmacology and Toxicology, The University of Kansas, Lawrence, KS, USA
| | - Jeffrey M. Zigman
- Department of Internal Medicine, Center for Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Heng Du
- Department of Pharmacology and Toxicology, The University of Kansas, Lawrence, KS, USA
- Alzheimer’s disease Research Center (ADRC), Department of Neurology, The University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
11
|
Fadó R, Molins A, Rojas R, Casals N. Feeding the Brain: Effect of Nutrients on Cognition, Synaptic Function, and AMPA Receptors. Nutrients 2022; 14:nu14194137. [PMID: 36235789 PMCID: PMC9572450 DOI: 10.3390/nu14194137] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022] Open
Abstract
In recent decades, traditional eating habits have been replaced by a more globalized diet, rich in saturated fatty acids and simple sugars. Extensive evidence shows that these dietary factors contribute to cognitive health impairment as well as increase the incidence of metabolic diseases such as obesity and diabetes. However, how these nutrients modulate synaptic function and neuroplasticity is poorly understood. We review the Western, ketogenic, and paleolithic diets for their effects on cognition and correlations with synaptic changes, focusing mainly (but not exclusively) on animal model studies aimed at tracing molecular alterations that may contribute to impaired human cognition. We observe that memory and learning deficits mediated by high-fat/high-sugar diets, even over short exposure times, are associated with reduced arborization, widened synaptic cleft, narrowed post-synaptic zone, and decreased activity-dependent synaptic plasticity in the hippocampus, and also observe that these alterations correlate with deregulation of the AMPA-type glutamate ionotropic receptors (AMPARs) that are crucial to neuroplasticity. Furthermore, we explored which diet-mediated mechanisms modulate synaptic AMPARs and whether certain supplements or nutritional interventions could reverse deleterious effects, contributing to improved learning and memory in older people and patients with Alzheimer’s disease.
Collapse
Affiliation(s)
- Rut Fadó
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, E-08193 Cerdanyola del Vallès, Spain
- Correspondence: ; Tel.: +34-93-504-20-00
| | - Anna Molins
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain
| | - Rocío Rojas
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain
| | - Núria Casals
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
12
|
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: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022] Open
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
|
13
|
Ghosh-Swaby OR, Reichelt AC, Sheppard PAS, Davies J, Bussey TJ, Saksida LM. Metabolic hormones mediate cognition. Front Neuroendocrinol 2022; 66:101009. [PMID: 35679900 DOI: 10.1016/j.yfrne.2022.101009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 05/18/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022]
Abstract
Recent biochemical and behavioural evidence indicates that metabolic hormones not only regulate energy intake and nutrient content, but also modulate plasticity and cognition in the central nervous system. Disruptions in metabolic hormone signalling may provide a link between metabolic syndromes like obesity and diabetes, and cognitive impairment. For example, altered metabolic homeostasis in obesity is a strong determinant of the severity of age-related cognitive decline and neurodegenerative disease. Here we review the evidence that eating behaviours and metabolic hormones-particularly ghrelin, leptin, and insulin-are key players in the delicate regulation of neural plasticity and cognition. Caloric restriction and antidiabetic therapies, both of which affect metabolic hormone levels can restore metabolic homeostasis and enhance cognitive function. Thus, metabolic hormone pathways provide a promising target for the treatment of cognitive decline.
Collapse
Affiliation(s)
- Olivia R Ghosh-Swaby
- Schulich School of Medicine and Dentistry, Neuroscience Program, Western University, London, ON, Canada
| | - Amy C Reichelt
- Faculty of Health and Medical Sciences, Adelaide Medical School, Adelaide, Australia
| | - Paul A S Sheppard
- Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Jeffrey Davies
- Swansea University Medical School, Swansea University, Swansea, UK
| | - Timothy J Bussey
- Schulich School of Medicine and Dentistry, Neuroscience Program, Western University, London, ON, Canada; Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Lisa M Saksida
- Schulich School of Medicine and Dentistry, Neuroscience Program, Western University, London, ON, Canada; Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, ON, Canada.
| |
Collapse
|
14
|
Vints WAJ, Levin O, Fujiyama H, Verbunt J, Masiulis N. Exerkines and long-term synaptic potentiation: Mechanisms of exercise-induced neuroplasticity. Front Neuroendocrinol 2022; 66:100993. [PMID: 35283168 DOI: 10.1016/j.yfrne.2022.100993] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 03/03/2022] [Accepted: 03/06/2022] [Indexed: 01/30/2023]
Abstract
Physical exercise may improve cognitive function by modulating molecular and cellular mechanisms within the brain. We propose that the facilitation of long-term synaptic potentiation (LTP)-related pathways, by products induced by physical exercise (i.e., exerkines), is a crucial aspect of the exercise-effect on the brain. This review summarizes synaptic pathways that are activated by exerkines and may potentiate LTP. For a total of 16 exerkines, we indicated how blood and brain exerkine levels are altered depending on the type of physical exercise (i.e., cardiovascular or resistance exercise) and how they respond to a single bout (i.e., acute exercise) or multiple bouts of physical exercise (i.e., chronic exercise). This information may be used for designing individualized physical exercise programs. Finally, this review may serve to direct future research towards fundamental gaps in our current knowledge regarding the biophysical interactions between muscle activity and the brain at both cellular and system levels.
Collapse
Affiliation(s)
- Wouter A J Vints
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Sporto str. 6, LT-44221 Kaunas, Lithuania; Department of Rehabilitation Medicine Research School CAPHRI, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands; Centre of Expertise in Rehabilitation and Audiology, Adelante Zorggroep, P.O. Box 88, 6430 AB Hoensbroek, the Netherlands.
| | - Oron Levin
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Sporto str. 6, LT-44221 Kaunas, Lithuania; Movement Control & Neuroplasticity Research Group, Group Biomedical Sciences, Catholic University Leuven, Tervuursevest 101, 3001 Heverlee, Belgium.
| | - Hakuei Fujiyama
- Department of Psychology, Murdoch University, 90 South St., WA 6150 Perth, Australia; Centre for Healthy Ageing, Health Futures Institute, Murdoch University, 90 South St., WA 6150 Perth, Australia; Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, 90 South St., WA 6150 Perth, Australia.
| | - Jeanine Verbunt
- Department of Rehabilitation Medicine Research School CAPHRI, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands; Centre of Expertise in Rehabilitation and Audiology, Adelante Zorggroep, P.O. Box 88, 6430 AB Hoensbroek, the Netherlands.
| | - Nerijus Masiulis
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Sporto str. 6, LT-44221 Kaunas, Lithuania; Department of Rehabilitation, Physical and Sports Medicine, Institute of Health Science, Faculty of Medicine, Vilnius University, M. K. Čiurlionio Str. 21, LT-03101 Vilnius, Lithuania.
| |
Collapse
|
15
|
GHS-R1a activity suppresses synaptic function of primary cultured hippocampal neurons. Biochem Biophys Res Commun 2022; 602:91-97. [DOI: 10.1016/j.bbrc.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/01/2022] [Indexed: 11/18/2022]
|
16
|
Sassi M, Morgan AH, Davies JS. Ghrelin Acylation-A Post-Translational Tuning Mechanism Regulating Adult Hippocampal Neurogenesis. Cells 2022; 11:cells11050765. [PMID: 35269387 PMCID: PMC8909677 DOI: 10.3390/cells11050765] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/11/2022] [Accepted: 02/13/2022] [Indexed: 02/05/2023] Open
Abstract
Adult hippocampal neurogenesis—the generation of new functional neurones in the adult brain—is impaired in aging and many neurodegenerative disorders. We recently showed that the acylated version of the gut hormone ghrelin (acyl-ghrelin) stimulates adult hippocampal neurogenesis while the unacylated form of ghrelin inhibits it, thus demonstrating a previously unknown function of unacyl-ghrelin in modulating hippocampal plasticity. Analysis of plasma samples from Parkinson’s disease patients with dementia demonstrated a reduced acyl-ghrelin:unacyl-ghrelin ratio compared to both healthy controls and cognitively intact Parkinson’s disease patients. These data, from mouse and human studies, suggest that restoring acyl-ghrelin signalling may promote the activation of pathways to support memory function. In this short review, we discuss the evidence for ghrelin’s role in regulating adult hippocampal neurogenesis and the enzymes involved in ghrelin acylation and de-acylation as targets to treat mood-related disorders and dementia.
Collapse
|
17
|
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: 5.0] [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
|
18
|
Mustafá ER, Cordisco González S, Damian M, Cantel S, Denoyelle S, Wagner R, Schiöth HB, Fehrentz JA, Banères JL, Perelló M, Raingo J. LEAP2 Impairs the Capability of the Growth Hormone Secretagogue Receptor to Regulate the Dopamine 2 Receptor Signaling. Front Pharmacol 2021; 12:712437. [PMID: 34447311 PMCID: PMC8383165 DOI: 10.3389/fphar.2021.712437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
The growth hormone secretagogue receptor (GHSR) signals in response to ghrelin, but also acts via ligand-independent mechanisms that include either constitutive activation or interaction with other G protein-coupled receptors, such as the dopamine 2 receptor (D2R). A key target of GHSR in neurons is voltage-gated calcium channels type 2.2 (CaV2.2). Recently, the liver-expressed antimicrobial peptide 2 (LEAP2) was recognized as a novel GHSR ligand, but the mechanism of action of LEAP2 on GHSR is not well understood. Here, we investigated the role of LEAP2 on the canonical and non-canonical modes of action of GHSR on CaV2.2 function. Using a heterologous expression system and patch-clamp recordings, we found that LEAP2 impairs the reduction of CaV2.2 currents induced by ghrelin-evoked and constitutive GHSR activities, acting as a GHSR antagonist and inverse agonist, respectively. We also found that LEAP2 prevents GHSR from modulating the effects of D2R signaling on CaV2.2 currents, and that the GHSR-binding N-terminal region LEAP2 underlies these effects. Using purified labeled receptors assembled into lipid nanodiscs and Forster Resonance Energy Transfer (FRET) assessments, we found that the N-terminal region of LEAP2 stabilizes an inactive conformation of GHSR that is dissociated from Gq protein and, consequently, reverses the effect of GHSR on D2R-dependent Gi activation. Thus, our results provide critical molecular insights into the mechanism mediating LEAP2 modulation of GHSR.
Collapse
Affiliation(s)
- Emilio R Mustafá
- Laboratory of Electrophysiology 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 (UNLP)], La Plata, Argentina
| | - Santiago Cordisco González
- Laboratory of Electrophysiology 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 (UNLP)], La Plata, Argentina
| | - Marjorie Damian
- Institut des Biomolécules Max Mousseron (IBMM), Université Montpellier, CNRS, Montpellier, France
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron (IBMM), Université Montpellier, CNRS, Montpellier, France
| | - Severine Denoyelle
- Institut des Biomolécules Max Mousseron (IBMM), Université Montpellier, CNRS, Montpellier, France
| | - Renaud Wagner
- Plateforme IMPReSs, CNRS UMR7242, Biotechnologie et Signalisation Cellulaire, École Supérieure de Biotechnologie de Strasbourg, Strasbourg, France
| | - Helgi B Schiöth
- Department of Neuroscience, Uppsala University, Uppsala, Sweden.,Institute for Translational Medicine and Biothechnology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron (IBMM), Université Montpellier, CNRS, Montpellier, France
| | - Jean-Louis Banères
- Institut des Biomolécules Max Mousseron (IBMM), Université Montpellier, CNRS, Montpellier, France
| | - Mario Perelló
- 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 (UNLP)], La Plata, Argentina
| | - Jesica Raingo
- Laboratory of Electrophysiology 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 (UNLP)], La Plata, Argentina
| |
Collapse
|
19
|
Mampay M, Flint MS, Sheridan GK. Tumour brain: Pretreatment cognitive and affective disorders caused by peripheral cancers. Br J Pharmacol 2021; 178:3977-3996. [PMID: 34029379 DOI: 10.1111/bph.15571] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 04/12/2021] [Accepted: 05/05/2021] [Indexed: 12/20/2022] Open
Abstract
People that develop extracranial cancers often display co-morbid neurological disorders, such as anxiety, depression and cognitive impairment, even before commencement of chemotherapy. This suggests bidirectional crosstalk between non-CNS tumours and the brain, which can regulate peripheral tumour growth. However, the reciprocal neurological effects of tumour progression on brain homeostasis are not well understood. Here, we review brain regions involved in regulating peripheral tumour development and how they, in turn, are adversely affected by advancing tumour burden. Tumour-induced activation of the immune system, blood-brain barrier breakdown and chronic neuroinflammation can lead to circadian rhythm dysfunction, sleep disturbances, aberrant glucocorticoid production, decreased hippocampal neurogenesis and dysregulation of neural network activity, resulting in depression and memory impairments. Given that cancer-related cognitive impairment diminishes patient quality of life, reduces adherence to chemotherapy and worsens cancer prognosis, it is essential that more research is focused at understanding how peripheral tumours affect brain homeostasis.
Collapse
Affiliation(s)
- Myrthe Mampay
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Melanie S Flint
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Graham K Sheridan
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
20
|
Casas M, Fadó R, Domínguez JL, Roig A, Kaku M, Chohnan S, Solé M, Unzeta M, Miñano-Molina AJ, Rodríguez-Álvarez J, Dickson EJ, Casals N. Sensing of nutrients by CPT1C controls SAC1 activity to regulate AMPA receptor trafficking. J Cell Biol 2021; 219:152088. [PMID: 32931550 PMCID: PMC7659714 DOI: 10.1083/jcb.201912045] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/21/2020] [Accepted: 06/18/2020] [Indexed: 12/21/2022] Open
Abstract
Carnitine palmitoyltransferase 1C (CPT1C) is a sensor of malonyl-CoA and is located in the ER of neurons. AMPA receptors (AMPARs) mediate fast excitatory neurotransmission in the brain and play a key role in synaptic plasticity. In the present study, we demonstrate across different metabolic stress conditions that modulate malonyl-CoA levels in cortical neurons that CPT1C regulates the trafficking of the major AMPAR subunit, GluA1, through the phosphatidyl-inositol-4-phosphate (PI(4)P) phosphatase SAC1. In normal conditions, CPT1C down-regulates SAC1 catalytic activity, allowing efficient GluA1 trafficking to the plasma membrane. However, under low malonyl-CoA levels, such as during glucose depletion, CPT1C-dependent inhibition of SAC1 is released, facilitating SAC1’s translocation to ER-TGN contact sites to decrease TGN PI(4)P pools and trigger GluA1 retention at the TGN. Results reveal that GluA1 trafficking is regulated by CPT1C sensing of malonyl-CoA and provide the first report of a SAC1 inhibitor. Moreover, they shed light on how nutrients can affect synaptic function and cognition.
Collapse
Affiliation(s)
- Maria Casas
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Spain
| | - Rut Fadó
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Spain
| | - José Luis Domínguez
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Spain
| | - Aina Roig
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Spain
| | - Moena Kaku
- Department of Food and Life Science, Ibaraki University College of Agriculture, Ami, Ibaraki, Japan
| | - Shigeru Chohnan
- Department of Food and Life Science, Ibaraki University College of Agriculture, Ami, Ibaraki, Japan
| | - Montse Solé
- Biochemistry and Molecular Biology Department, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Mercedes Unzeta
- Biochemistry and Molecular Biology Department, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Alfredo Jesús Miñano-Molina
- Biochemistry and Molecular Biology Department, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - José Rodríguez-Álvarez
- Biochemistry and Molecular Biology Department, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain.,Department of Neuroscience, Albert Einstein College of Medicine, New York, NY
| | - Eamonn James Dickson
- Department of Physiology and Membrane Biology, University of California, School of Medicine, Davis, CA
| | - Núria Casals
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
21
|
Bioque M, González-Rodríguez A, Garcia-Rizo C, Cobo J, Monreal JA, Usall J, Soria V, Labad J. Targeting the microbiome-gut-brain axis for improving cognition in schizophrenia and major mood disorders: A narrative review. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110130. [PMID: 33045322 DOI: 10.1016/j.pnpbp.2020.110130] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/29/2020] [Accepted: 10/04/2020] [Indexed: 02/07/2023]
Abstract
Cognitive impairment has been consistently found to be a core feature of serious mental illnesses such as schizophrenia and major mood disorders (major depression and bipolar disorder). In recent years, a great effort has been made in elucidating the biological causes of cognitive deficits and the search for new biomarkers of cognition. Microbiome and gut-brain axis (MGB) hormones have been postulated to be potential biomarkers of cognition in serious mental illnesses. The main aim of this review was to synthesize current evidence on the association of microbiome and gut-brain hormones on cognitive processes in schizophrenia and major mood disorders and the association of MGB hormones with stress and the immune system. Our review underscores the role of the MGB axis on cognitive aspects of serious mental illnesses with the potential use of agents targeting the gut microbiota as cognitive enhancers. However, the current evidence for clinical trials focused on the MGB axis as cognitive enhancers in these clinical populations is scarce. Future clinical trials using probiotics, prebiotics, antibiotics, or faecal microbiota transplantation need to consider potential mechanistic pathways such as the HPA axis, the immune system, or gut-brain axis hormones involved in appetite control and energy homeostasis.
Collapse
Affiliation(s)
- Miquel Bioque
- Barcelona Clinic Schizophrenia Unit (BCSU), Neuroscience Institute, Hospital Clinic of Barcelona, University of Barcelona (UB), IDIBAPS, CIBERSAM, Barcelona, Spain
| | - Alexandre González-Rodríguez
- Department of Mental Health, Parc Tauli University Hospital, I3PT. Sabadell, Autonomous University of Barcelona (UAB), CIBERSAM, Barcelona, Spain
| | - Clemente Garcia-Rizo
- Barcelona Clinic Schizophrenia Unit (BCSU), Neuroscience Institute, Hospital Clinic of Barcelona, University of Barcelona (UB), IDIBAPS, CIBERSAM, Barcelona, Spain.
| | - Jesús Cobo
- Department of Mental Health, Parc Tauli University Hospital, I3PT. Sabadell, Autonomous University of Barcelona (UAB), CIBERSAM, Barcelona, Spain
| | - José Antonio Monreal
- Department of Mental Health, Parc Tauli University Hospital, I3PT. Sabadell, Autonomous University of Barcelona (UAB), CIBERSAM, Barcelona, Spain
| | - Judith Usall
- Parc Sanitari Sant Joan de Déu, Sant Boi de Llobregat, University of Barcelona (UB), CIBERSAM, Barcelona, Spain
| | - Virginia Soria
- Department of Psychiatry, Hospital Universitari Bellvitge, Hospitalet de Llobregat, University of Barcelona (UB), IDIBELL, CIBERSAM, Spain
| | | | - Javier Labad
- Department of Mental Health, Parc Tauli University Hospital, I3PT. Sabadell, Autonomous University of Barcelona (UAB), CIBERSAM, Barcelona, Spain
| |
Collapse
|
22
|
Zahiri H, Rostampour M, Khakpour B, Rohampour K. The effect of ghrelin injection in the CA1 region of hippocampus on the MK801- induced memory impairment in wistar rats. Behav Brain Res 2021; 405:113209. [PMID: 33639267 DOI: 10.1016/j.bbr.2021.113209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 02/20/2021] [Accepted: 02/21/2021] [Indexed: 01/28/2023]
Abstract
N-Methyl-D-Aspartate (NMDA) receptors are critically involved in the learning and memory formation and dizocilpine (MK-801) is an antagonist of NMDA receptor. Ghrelin plays a crucial role in learning and memory processes. The present study was conducted to the evaluation of ghrelin effect on passive avoidance memory impairment induced by MK801. In this experimental study, 24 male wistar rats were randomly distributed into 3 groups of 8 each. Passive avoidance tests of animals were evaluated using Shuttle Box apparatus. One week after the surgery, ghrelin (3 nmol) was injected intra-hippocampally, 5 min before the MK-801administration. MK-801 (0.15 mg/kg) was injected intraperitoneally (i.p.), 10 min before the test session. Pre-test injection of MK-801 significantly decreased STL (step through latency) at 24 h and 48 h (P < 0.001) and 10 days (P < 0.01) and increased TDC (time spent in dark compartment) at 24 h, 48 h and 10 days (P < 0.001) after training in comparison with control group. Pre-test injection of ghrelin + MK-801 significantly increased STL at 24 h (P < 0.01), 48 h and 10 days (P < 0.001) and decreased TDC at 24 h, 48 h and 10 days (P < 0.001) after training in comparison with MK-801 received group. It is concluded that pre-test injection of MK-801 impaired passive avoidance memory. Administration of ghrelin before MK-801 ameliorated memory impairment induced by MK-801. It is assumed that this compensative effect of ghrelin was mediated by NMDA receptor.
Collapse
Affiliation(s)
- Hamideh Zahiri
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Rostampour
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Behrooz Khakpour
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Kambiz Rohampour
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
23
|
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: 4.3] [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
|
24
|
Campanelli F, Laricchiuta D, Natale G, Marino G, Calabrese V, Picconi B, Petrosini L, Calabresi P, Ghiglieri V. Long-Term Shaping of Corticostriatal Synaptic Activity by Acute Fasting. Int J Mol Sci 2021; 22:ijms22041916. [PMID: 33671915 PMCID: PMC7918979 DOI: 10.3390/ijms22041916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 11/24/2022] Open
Abstract
Food restriction is a robust nongenic, nonsurgical and nonpharmacologic intervention known to improve health and extend lifespan in various species. Food is considered the most essential and frequently consumed natural reward, and current observations have demonstrated homeostatic responses and neuroadaptations to sustained intermittent or chronic deprivation. Results obtained to date indicate that food deprivation affects glutamatergic synapses, favoring the insertion of GluA2-lacking α-Ammino-3-idrossi-5-Metil-4-idrossazol-Propionic Acid receptors (AMPARs) in postsynaptic membranes. Despite an increasing number of studies pointing towards specific changes in response to dietary restrictions in brain regions, such as the nucleus accumbens and hippocampus, none have investigated the long-term effects of such practice in the dorsal striatum. This basal ganglia nucleus is involved in habit formation and in eating behavior, especially that based on dopaminergic control of motivation for food in both humans and animals. Here, we explored whether we could retrieve long-term signs of changes in AMPARs subunit composition in dorsal striatal neurons of mice acutely deprived for 12 hours/day for two consecutive days by analyzing glutamatergic neurotransmission and the principal forms of dopamine and glutamate-dependent synaptic plasticity. Overall, our data show that a moderate food deprivation in experimental animals is a salient event mirrored by a series of neuroadaptations and suggest that dietary restriction may be determinant in shaping striatal synaptic plasticity in the physiological state.
Collapse
Affiliation(s)
- Federica Campanelli
- Dipartmento di Medicina, Università di Perugia, 06129 Perugia, Italy; (F.C.); (G.N.); (G.M.); (V.C.)
- Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Daniela Laricchiuta
- Laboratorio di Neurofisiologia Sperimentale e del Comportamento, IRCCS Fondazione Santa Lucia c/o CERC, 00143 Rome, Italy; (D.L.); (L.P.)
| | - Giuseppina Natale
- Dipartmento di Medicina, Università di Perugia, 06129 Perugia, Italy; (F.C.); (G.N.); (G.M.); (V.C.)
- Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Gioia Marino
- Dipartmento di Medicina, Università di Perugia, 06129 Perugia, Italy; (F.C.); (G.N.); (G.M.); (V.C.)
- Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Valeria Calabrese
- Dipartmento di Medicina, Università di Perugia, 06129 Perugia, Italy; (F.C.); (G.N.); (G.M.); (V.C.)
- IRCCS San Raffaele Pisana, Rome 00176, Italy;
| | - Barbara Picconi
- IRCCS San Raffaele Pisana, Rome 00176, Italy;
- Università Telematica San Raffaele, 00166 Rome, Italy
| | - Laura Petrosini
- Laboratorio di Neurofisiologia Sperimentale e del Comportamento, IRCCS Fondazione Santa Lucia c/o CERC, 00143 Rome, Italy; (D.L.); (L.P.)
| | - Paolo Calabresi
- Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Clinica Neurologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Veronica Ghiglieri
- Laboratorio di Neurofisiologia Sperimentale e del Comportamento, IRCCS Fondazione Santa Lucia c/o CERC, 00143 Rome, Italy; (D.L.); (L.P.)
- Università Telematica San Raffaele, 00166 Rome, Italy
- Correspondence:
| |
Collapse
|
25
|
Xue F, Tian J, Yu C, Du H, Guo L. Type I interferon response-related microglial Mef2c deregulation at the onset of Alzheimer's pathology in 5×FAD mice. Neurobiol Dis 2021; 152:105272. [PMID: 33540048 PMCID: PMC7956132 DOI: 10.1016/j.nbd.2021.105272] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 01/15/2021] [Indexed: 02/08/2023] Open
Abstract
Alzheimer’s disease (AD) is a chronic neurodegenerative disorder with multifactorial etiology. The role of microglia in the pathogenesis of AD has been increasingly recognized in recent years; however, the detailed mechanisms shaping microglial phenotypes in AD-relevant pathological settings remain largely unresolved. Myocyte-specific enhancer factor 2C (Mef2C) is a transcription factor with versatile functions. Recent studies have attributed aging-related microglial changes to type I interferon (IFN-I)-associated Mef2C deregulation. In view of the close relationship between brain aging and AD, it is of great interest to determine microglial Mef2C changes in AD-related conditions. In this study, we have found that suppressed Mef2C nuclear translocation was an early and prominent microglial phenotype in a mouse model of brain amyloidosis (5×FAD mice), which exacerbated with age. Echoing the early Mef2C deregulation and its association with microglial activation, transcriptional data showed elicited IFN-I response in microglia from young 5×FAD mice. Amyloid beta 42 (Aβ42) in its oligomeric forms promoted Mef2C deregulation in microglia on acute organotypic brain slices with augmented microglial activation and synapse elimination via microglial phagocytosis. Importantly, these oligomeric Aβ42-mediated microglial changes were substantially attenuated by blocking IFN-I signaling. The simplest interpretation of the results is that Mef2C, concurring with activated IFN-I signaling, constitutes early microglial changes in AD-related conditions. In addition to the potential contribution of Mef2C deregulation to the development of microglial phenotypes in AD, Mef2C suppression in microglia may serve as a potential mechanistic pathway linking brain aging and AD.
Collapse
Affiliation(s)
- Feng Xue
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, KS 66045, United States; The Biological Science Department, University of Texas at Dallas, TX 75080, United States
| | - Jing Tian
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, KS 66045, United States; The Biological Science Department, University of Texas at Dallas, TX 75080, United States
| | - Chunxiao Yu
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, KS 66045, United States; The Biological Science Department, University of Texas at Dallas, TX 75080, United States
| | - Heng Du
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, KS 66045, United States; Higuchi Biosciences Center, University of Kansas, KS 66045, United States; The Biological Science Department, University of Texas at Dallas, TX 75080, United States.
| | - Lan Guo
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, KS 66045, United States; Higuchi Biosciences Center, University of Kansas, KS 66045, United States; The Biological Science Department, University of Texas at Dallas, TX 75080, United States.
| |
Collapse
|
26
|
Xiao X, Bi M, Jiao Q, Chen X, Du X, Jiang H. A new understanding of GHSR1a--independent of ghrelin activation. Ageing Res Rev 2020; 64:101187. [PMID: 33007437 DOI: 10.1016/j.arr.2020.101187] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/13/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022]
Abstract
Growth hormone secretagogue receptor 1a (GHSR1a), a member of the G protein-coupled receptor (GPCR) family, is a functional receptor of ghrelin. The expression levels and activities of GHSR1a are affected by various factors. In past years, it has been found that the ghrelin-GHSR1a system can perform biological functions such as anti-inflammation, anti-apoptosis, and anti-oxidative stress. In addition to mediating the effect of ghrelin, GHSR1a also has abnormally high constitutive activity; that is, it can still transmit intracellular signals without activation of the ghrelin ligand. This constitutive activity affects brain functions, growth and development of the body; therefore, it has profound impacts on neurodegenerative diseases and some other age-related diseases. In addition, GHSR1a can also form homodimers or heterodimers with other GPCRs, affecting the release of neurotransmitters, appetite regulation, cell proliferation and insulin release. Therefore, further understanding of the constitutive activities and dimerization of GHSR1a will enable us to better clarify the characteristics of GHSR1a and provide more therapeutic targets for drug development. Here, we focus on the roles of GHSR1a in various biological functions and provide a comprehensive summary of the current research on GHSR1a to provide broader therapeutic prospects for age-related disease treatment.
Collapse
Affiliation(s)
- Xue Xiao
- 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, 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, 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, China
| | - 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, 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, 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, China.
| |
Collapse
|
27
|
Tian J, Wang T, Wang Q, Guo L, Du H. MK0677, a Ghrelin Mimetic, Improves Neurogenesis but Fails to Prevent Hippocampal Lesions in a Mouse Model of Alzheimer's Disease Pathology. J Alzheimers Dis 2020; 72:467-478. [PMID: 31594237 DOI: 10.3233/jad-190779] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hippocampal lesions including synaptic injury, neuroinflammation, and impaired neurogenesis are featured pathology closely associated with neuronal stress and cognitive impairment in Alzheimer's disease (AD). Previous studies suggest that ghrelin and its receptor, growth hormone secretagogue receptor 1α (GHSR1α), promote hippocampal synaptic function and neurogenesis. GHSR1α activation thus holds the potential to be a therapeutic avenue for the treatment of hippocampal pathology in AD; however, a comprehensive study on the preventive effect of MK0677 on hippocampal lesions in AD-related conditions is still lacking. In this study, we treated a transgenic mouse model of AD-like amyloidosis (5xFAD mice) at the asymptomatic stage with MK0677, a potent ghrelin mimetic. We found that MK0677 fostered hippocampal neurogenesis in 5xFAD mice but observed little preventive function with regards to the development of hippocampal amyloid-β (Aβ) deposition, synaptic loss, microglial activation, or cognitive impairment. Furthermore, MK0677 at a dose of 3 mg/kg significantly increased 5xFAD mouse mortality. Despite enhanced hippocampal neurogenesis, MK0677 treatment has little beneficial effect to prevent hippocampal lesions or cognitive deficits against Aβ toxicity. This study, together with a failed large-scale clinical trial, suggests the ineffectiveness of MK0677 alone for AD prevention and treatment.
Collapse
Affiliation(s)
- Jing Tian
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Tienju Wang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Qi Wang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA.,Department of Neurology, Qianfoshan Hospital, Shandong First Medical University, Jinan, China
| | - Lan Guo
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Heng Du
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
28
|
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: 3.0] [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
|
29
|
Hornsby AK, Buntwal L, Carisi MC, Santos VV, Johnston F, Roberts LD, Sassi M, Mequinion M, Stark R, Reichenbach A, Lockie SH, Siervo M, Howell O, Morgan AH, Wells T, Andrews ZB, Burn DJ, Davies JS. Unacylated-Ghrelin Impairs Hippocampal Neurogenesis and Memory in Mice and Is Altered in Parkinson's Dementia in Humans. CELL REPORTS MEDICINE 2020; 1:100120. [PMID: 33103129 PMCID: PMC7575905 DOI: 10.1016/j.xcrm.2020.100120] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 05/20/2020] [Accepted: 09/16/2020] [Indexed: 12/20/2022]
Abstract
Blood-borne factors regulate adult hippocampal neurogenesis and cognition in mammals. We report that elevating circulating unacylated-ghrelin (UAG), using both pharmacological and genetic methods, reduced hippocampal neurogenesis and plasticity in mice. Spatial memory impairments observed in ghrelin-O-acyl transferase-null (GOAT−/−) mice that lack acyl-ghrelin (AG) but have high levels of UAG were rescued by acyl-ghrelin. Acyl-ghrelin-mediated neurogenesis in vitro was dependent on non-cell-autonomous BDNF signaling that was inhibited by UAG. These findings suggest that post-translational acylation of ghrelin is important to neurogenesis and memory in mice. To determine relevance in humans, we analyzed circulating AG:UAG in Parkinson disease (PD) patients diagnosed with dementia (PDD), cognitively intact PD patients, and controls. Notably, plasma AG:UAG was only reduced in PDD. Hippocampal ghrelin-receptor expression remained unchanged; however, GOAT+ cell number was reduced in PDD. We identify UAG as a regulator of hippocampal-dependent plasticity and spatial memory and AG:UAG as a putative circulating diagnostic biomarker of dementia. Circulating unacylated-ghrelin (UAG) reduces hippocampal neurogenesis Circulating acyl-ghrelin (AG) rescues spatial memory deficit in GOAT−/− mice UAG blocks the AG induced survival of newborn hippocampal cells Plasma AG:UAG and hippocampal GOAT+ cells are reduced in Parkinson’s dementia
Collapse
Affiliation(s)
- Amanda K.E. Hornsby
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Luke Buntwal
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Maria Carla Carisi
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Vanessa V. Santos
- Biomedical Discovery Institute, Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Fionnuala Johnston
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Luke D. Roberts
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Martina Sassi
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Mathieu Mequinion
- Biomedical Discovery Institute, Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Romana Stark
- Biomedical Discovery Institute, Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Alex Reichenbach
- Biomedical Discovery Institute, Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Sarah H. Lockie
- Biomedical Discovery Institute, Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Mario Siervo
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- School of Life Sciences, Queen's Medical Centre, The University of Nottingham Medical School, Nottingham NG7 2UH, UK
| | - Owain Howell
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Alwena H. Morgan
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Timothy Wells
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Zane B. Andrews
- Biomedical Discovery Institute, Department of Physiology, Monash University, Clayton, VIC, Australia
| | - David J. Burn
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Jeffrey S. Davies
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
- Corresponding author
| |
Collapse
|
30
|
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: 1.3] [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]
|
31
|
Tian J, Guo L, Sui S, Driskill C, Phensy A, Wang Q, Gauba E, Zigman JM, Swerdlow RH, Kroener S, Du H. Disrupted hippocampal growth hormone secretagogue receptor 1α interaction with dopamine receptor D1 plays a role in Alzheimer's disease. Sci Transl Med 2020; 11:11/505/eaav6278. [PMID: 31413143 PMCID: PMC6776822 DOI: 10.1126/scitranslmed.aav6278] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 06/24/2019] [Indexed: 12/12/2022]
Abstract
Hippocampal lesions are a defining pathology of Alzheimer's disease (AD). However, the molecular mechanisms that underlie hippocampal synaptic injury in AD have not been fully elucidated. Current therapeutic efforts for AD treatment are not effective in correcting hippocampal synaptic deficits. Growth hormone secretagogue receptor 1α (GHSR1α) is critical for hippocampal synaptic physiology. Here, we report that GHSR1α interaction with β-amyloid (Aβ) suppresses GHSR1α activation, leading to compromised GHSR1α regulation of dopamine receptor D1 (DRD1) in the hippocampus from patients with AD. The simultaneous application of the selective GHSR1α agonist MK0677 with the selective DRD1 agonist SKF81297 rescued Ghsr1α function from Aβ inhibition, mitigating hippocampal synaptic injury and improving spatial memory in an AD mouse model. Our data reveal a mechanism of hippocampal vulnerability in AD and suggest that a combined activation of GHSR1α and DRD1 may be a promising approach for treating AD.
Collapse
Affiliation(s)
- Jing Tian
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Lan Guo
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Shaomei Sui
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA.,Department of Neurology, Qianfoshan Hospital, Shandong First Medical University, Jinan, Shandong 250014, China
| | - Christopher Driskill
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Aarron Phensy
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Qi Wang
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA.,Department of Neurology, Qianfoshan Hospital, Shandong First Medical University, Jinan, Shandong 250014, China
| | - Esha Gauba
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Jeffrey M Zigman
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Russell H Swerdlow
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sven Kroener
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Heng Du
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA.
| |
Collapse
|
32
|
Carr KD. Modulatory Effects of Food Restriction on Brain and Behavioral Effects of Abused Drugs. Curr Pharm Des 2020; 26:2363-2371. [DOI: 10.2174/1381612826666200204141057] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/19/2019] [Indexed: 12/14/2022]
Abstract
Energy homeostasis is achieved, in part, by metabolic signals that regulate the incentive motivating
effects of food and its cues, thereby driving or curtailing procurement and consumption. The neural underpinnings
of these regulated incentive effects have been identified as elements within the mesolimbic dopamine pathway.
A separate line of research has shown that most drugs with abuse liability increase dopamine transmission in
this same pathway and thereby reinforce self-administration. Consequently, one might expect shifts in energy
balance and metabolic signaling to impact drug abuse risk. Basic science studies have yielded numerous examples
of drug responses altered by diet manipulation. Considering the prevalence of weight loss dieting in Western
societies, and the anorexigenic effects of many abused drugs themselves, we have focused on the CNS and behavioral
effects of food restriction in rats. Food restriction has been shown to increase the reward magnitude of diverse
drugs of abuse, and these effects have been attributed to neuroadaptations in the dopamine-innervated nucleus
accumbens. The changes induced by food restriction include synaptic incorporation of calcium-permeable
AMPA receptors and increased signaling downstream of D1 dopamine receptor stimulation. Recent studies suggest
a mechanistic model in which concurrent stimulation of D1 and GluA2-lacking AMPA receptors enables
increased stimulus-induced trafficking of GluA1/GluA2 AMPARs into the postsynaptic density, thereby increasing
the incentive effects of food, drugs, and associated cues. In addition, the established role of AMPA receptor
trafficking in enduring synaptic plasticity prompts speculation that drug use during food restriction may more
strongly ingrain behavior relative to similar use under free-feeding conditions.
Collapse
Affiliation(s)
- Kenneth D. Carr
- Departments of Psychiatry, Biochemistry and Molecular Pharmacology, New York University School of Medicine, 435 East 30th Street, New York, NY 10016, United States
| |
Collapse
|
33
|
Peng FZ, Fan J, Ge TT, Liu QQ, Li BJ. Rapid anti-depressant-like effects of ketamine and other candidates: Molecular and cellular mechanisms. Cell Prolif 2020; 53:e12804. [PMID: 32266752 PMCID: PMC7260066 DOI: 10.1111/cpr.12804] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/27/2022] Open
Abstract
Major depressive disorder takes at least 3 weeks for clinical anti‐depressants, such as serotonin selective reuptake inhibitors, to take effect, and only one‐third of patients remit. Ketamine, a kind of anaesthetic, can alleviate symptoms of major depressive disorder patients in a short time and is reported to be effective to treatment‐resistant depression patients. The rapid and strong anti‐depressant‐like effects of ketamine cause wide concern. In addition to ketamine, caloric restriction and sleep deprivation also elicit similar rapid anti‐depressant‐like effects. However, mechanisms about the rapid anti‐depressant‐like effects remain unclear. Elucidating the mechanisms of rapid anti‐depressant effects is the key to finding new therapeutic targets and developing therapeutic patterns. Therefore, in this review we summarize potential molecular and cellular mechanisms of rapid anti‐depressant‐like effects based on the pre‐clinical and clinical evidence, trying to provide new insight into future therapy.
Collapse
Affiliation(s)
- Fan Zhen Peng
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Jie Fan
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Tong Tong Ge
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Qian Qian Liu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Bing Jin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
34
|
SNAP-25 Puts SNAREs at Center Stage in Metabolic Disease. Neuroscience 2019; 420:86-96. [DOI: 10.1016/j.neuroscience.2018.07.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/12/2018] [Accepted: 07/19/2018] [Indexed: 12/20/2022]
|
35
|
Jeon SG, Hong SB, Nam Y, Tae J, Yoo A, Song EJ, Kim KI, Lee D, Park J, Lee SM, Kim JI, Moon M. Ghrelin in Alzheimer's disease: Pathologic roles and therapeutic implications. Ageing Res Rev 2019; 55:100945. [PMID: 31434007 DOI: 10.1016/j.arr.2019.100945] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/25/2019] [Accepted: 08/16/2019] [Indexed: 12/11/2022]
Abstract
Ghrelin, which has many important physiological roles, such as stimulating food intake, regulating energy homeostasis, and releasing insulin, has recently been studied for its roles in a diverse range of neurological disorders. Despite the several functions of ghrelin in the central nervous system, whether it works as a therapeutic agent for neurological dysfunction has been unclear. Altered levels and various roles of ghrelin have been reported in Alzheimer's disease (AD), which is characterized by the accumulation of misfolded proteins resulting in synaptic loss and cognitive decline. Interestingly, treatment with ghrelin or with the agonist of ghrelin receptor showed attenuation in several cases of AD-related pathology. These findings suggest the potential therapeutic implications of ghrelin in the pathogenesis of AD. In the present review, we summarized the roles of ghrelin in AD pathogenesis, amyloid beta (Aβ) homeostasis, tau hyperphosphorylation, neuroinflammation, mitochondrial deficit, synaptic dysfunction and cognitive impairment. The findings from this review suggest that ghrelin has a novel therapeutic potential for AD treatment. Thus, rigorously designed studies are needed to establish an effective AD-modifying strategy.
Collapse
|
36
|
Buntwal L, Sassi M, Morgan AH, Andrews ZB, Davies JS. Ghrelin-Mediated Hippocampal Neurogenesis: Implications for Health and Disease. Trends Endocrinol Metab 2019; 30:844-859. [PMID: 31445747 DOI: 10.1016/j.tem.2019.07.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/21/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022]
Abstract
There is a close relationship between cognition and nutritional status, however, the mechanisms underlying this relationship require elucidation. 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 thrive and survive, and accumulating evidence points to ghrelin being key in promoting adult hippocampal neurogenesis and memory. Aberrant neurogenesis is linked to cognitive decline in ageing and neurodegeneration. Therefore, identifying endogenous metabolic factors that regulate new adult-born neurone formation is an important objective in understanding the link between nutritional status and central nervous system (CNS) function. Here, we review current developments in our understanding of ghrelin's role in regulating neurogenesis and memory function.
Collapse
Affiliation(s)
- Luke Buntwal
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, SA2 8PP, UK
| | - Martina Sassi
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, SA2 8PP, UK
| | - Alwena H Morgan
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, SA2 8PP, UK
| | - Zane B Andrews
- Department of Physiology, Biomedical Discovery Unit, Monash University, Melbourne, Australia
| | - Jeffrey S Davies
- Molecular Neurobiology, Institute of Life Sciences, School of Medicine, Swansea University, SA2 8PP, UK.
| |
Collapse
|
37
|
Nazari-Serenjeh F, Darbandi N, Majidpour S, Moradi P. Ghrelin modulates morphine-nicotine interaction in avoidance memory: Involvement of CA1 nicotinic receptors. Brain Res 2019; 1720:146315. [DOI: 10.1016/j.brainres.2019.146315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 11/29/2022]
|
38
|
Mustafá ER, Cordisco Gonzalez S, Raingo J. Ghrelin Selectively Inhibits CaV3.3 Subtype of Low-Voltage-Gated Calcium Channels. Mol Neurobiol 2019; 57:722-735. [DOI: 10.1007/s12035-019-01738-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/16/2019] [Indexed: 01/01/2023]
|
39
|
Acute But Not Chronic Calorie Restriction Defends against Stress-Related Anxiety and Despair in a GHS-R1a-Dependent Manner. Neuroscience 2019; 412:94-104. [DOI: 10.1016/j.neuroscience.2019.05.067] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 12/27/2022]
|
40
|
Serrenho D, Santos SD, Carvalho AL. The Role of Ghrelin in Regulating Synaptic Function and Plasticity of Feeding-Associated Circuits. Front Cell Neurosci 2019; 13:205. [PMID: 31191250 PMCID: PMC6546032 DOI: 10.3389/fncel.2019.00205] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 04/24/2019] [Indexed: 12/21/2022] Open
Abstract
Synaptic plasticity of the neuronal circuits associated with feeding behavior is regulated by peripheral signals as a response to changes in the energy status of the body. These signals include glucose, free fatty acids, leptin and ghrelin and are released into circulation, being able to reach the brain. Ghrelin, a small peptide released from the stomach, is an orexigenic hormone produced in peripheral organs, and its action regulates food intake, body weight and glucose homeostasis. Behavioral studies show that ghrelin is implicated in the regulation of both hedonic and homeostatic feeding and of cognition. Ghrelin-induced synaptic plasticity has been described in neuronal circuits associated with these behaviors. In this review, we discuss the neuromodulatory mechanisms induced by ghrelin in regulating synaptic plasticity in three main neuronal circuits previously associated with feeding behaviors, namely hypothalamic (homeostatic feeding), ventral tegmental (hedonic and motivational feeding) and hippocampal (cognitive) circuits. Given the central role of ghrelin in regulating feeding behaviors, and the altered ghrelin levels associated with metabolic disorders such as obesity and anorexia, it is of paramount relevance to understand the effects of ghrelin on synaptic plasticity of neuronal circuits associated with feeding behaviors.
Collapse
Affiliation(s)
- Débora Serrenho
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal.,PhD Program in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra, Portugal
| | - Sandra D Santos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Ana Luísa Carvalho
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
41
|
Huang HJ, Chen XR, Han QQ, Wang J, Pilot A, Yu R, Liu Q, Li B, Wu GC, Wang YQ, Yu J. The protective effects of Ghrelin/GHSR on hippocampal neurogenesis in CUMS mice. Neuropharmacology 2019; 155:31-43. [PMID: 31103617 DOI: 10.1016/j.neuropharm.2019.05.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 04/27/2019] [Accepted: 05/12/2019] [Indexed: 12/18/2022]
Abstract
Ghrelin is an orexigenic hormone that also plays an important role in mood disorders. Our previous studies demonstrated that ghrelin administration could protect against depression-like behaviors of chronic unpredictable mild stress (CUMS) in rodents. However, the mechanism related to the effect of ghrelin on CUMS mice has yet to be revealed. This article shows that ghrelin (5 nmol/kg/day for 2 weeks, i.p.) decreased depression-like behaviors induced by CUMS and increased hippocampal integrity (neurogenesis and spine density) measured via Ki67, 5-bromo-2-deoxyuridine (BrdU), doublecortin (DCX) labeling and Golgi-cox staining, which were decreased under CUMS. The behavioral phenotypes of Growth hormone secretagogue receptor (Ghsr)-null and wild type (WT) mice were evaluated under no stress condition and after CUMS exposure to determine the effect of Ghsr knockout on the behavioral phenotypes and stress susceptibility of mice. Ghsr-null mice exhibited depression-like behaviors under no stress condition. CUMS induced similar depression- and anxiety-like behavioral manifestations in both Ghsr-null and WT mice. A similar pattern of behavioral changes was observed after hippocampal GHSR knockdown. Additionally, both Ghsr knockout as well as CUMS exhibited deleterious effects on neurogenesis and spine density in the dentate gyrus (DG). Besides, CCK8 assay and 5-Ethynyl-2'-deoxyuridine (EdU) incorporation assay showed that ghrelin has a proliferative effect on primary cultured hippocampal neural stem cells (NSCs) and this proliferation was blocked by D-Lys3-GHRP-6 (DLS, the antagonist of GHSR, 100 μM) pretreatment. Ghrelin-induced proliferation is associated with the inhibition of G1 arrest, and this inhibition was blocked by LY294002 (specific inhibitor of PI3K, 20 μM). Furthermore, the in vivo data displayed that LY294002 (50 nmol, i.c.v.) can significantly block the antidepressant-like action of exogenous ghrelin treatment. All these results suggest that ghrelin/GHSR signaling maintains the integrity of hippocampus and has an inherent neuroprotective effect whether facing stress or not.
Collapse
Affiliation(s)
- Hui-Jie Huang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiao-Rong Chen
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qiu-Qin Han
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Jing Wang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Adam Pilot
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rui Yu
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qiong Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, 200032, China
| | - Bing Li
- Center Laboratories, Jinshan Hospital of Fudan University, Shanghai, 201508, China
| | - Gen-Cheng Wu
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yan-Qing Wang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jin Yu
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
42
|
Guo L, Niu M, Yang J, Li L, Liu S, Sun Y, Zhou Z, Zhou Y. GHS-R1a Deficiency Alleviates Depression-Related Behaviors After Chronic Social Defeat Stress. Front Neurosci 2019; 13:364. [PMID: 31057357 PMCID: PMC6478702 DOI: 10.3389/fnins.2019.00364] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 03/29/2019] [Indexed: 12/13/2022] Open
Abstract
Ghrelin is an important orexigenic hormone that regulates feeding, metabolism and glucose homeostasis in human and rodents. Ghrelin functions by binding to its receptor, the growth hormone secretagogue receptor 1a (GHS-R1a), which is widely expressed inside and outside of the brain. Recent studies suggested that acyl-ghrelin, the active form of ghrelin, is a persistent biomarker for chronic stress exposure. However, how ghrelin/GHS-R1a signaling contributes to stress responses and mood regulation remains uncertain. In this study, we applied the chronic social defeat stress (CSDS) paradigm to both GHS-R1a knock-out (Ghsr-/-) mice and littermate control (Ghsr+/+) mice, and then measured their depression- and anxiety-related behaviors. We found that Ghsr+/+ mice, but not Ghsr-/- mice, displayed apparent anxiety and depression after CSDS, while two groups mice showed identical behaviors at baseline, non-stress state. By screening the central and peripheral responses of Ghsr-/- mice and Ghsr+/+ mice to chronic stress, we found similar elevations of total ghrelin and adrenocorticotropic hormone (ACTH) in the serum of Ghsr-/- mice and Ghsr+/+ mice after CSDS, but decreased interleukin-6 (IL-6) in the serum of defeated Ghsr-/- mice compared to defeated Ghsr+/+ mice. We also found increased concentration of brain derived neurotropic factor (BDNF) in the hippocampus of Ghsr-/- mice compared to Ghsr+/+ mice after CSDS. The basal levels of ghrelin, ACTH, IL-6, and BDNF were not different between Ghsr-/- mice and Ghsr+/+ mice. Our findings thus suggested that the differential expressions of BDNF and IL-6 after CSDS may contribute to less anxiety and less despair observed in GHS-R1a-deficient mice than in WT control mice. Therefore, ghrelin/GHS-R1a signaling may play a pro-anxiety and pro-depression effect in response to chronic stress, while GHS-R1a deficiency may provide resistance to depressive symptoms of CSDS.
Collapse
Affiliation(s)
- Li Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, China.,Department of Physiology, Binzhou Medical University, Yantai, China
| | - Minglu Niu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, China.,Department of Clinic Laboratory, PKU Care Luzhong Hospital, Zibo, China
| | - Jie Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, China.,Dongying No.1 Middle School, Dongying, China
| | - Li Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, China
| | - Shuhan Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, China
| | - Yuxiang Sun
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, United States
| | - Zhishang Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, China.,Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Yu Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, China.,Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China
| |
Collapse
|
43
|
Differential expression of the ghrelin-related mRNAs GHS-R1a, GHS-R1b, and MBOAT4 in Japanese patients with schizophrenia. Psychiatry Res 2019; 272:334-339. [PMID: 30597386 DOI: 10.1016/j.psychres.2018.12.135] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 11/26/2018] [Accepted: 12/24/2018] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Ghrelin regulates appetite and also plays important roles in cognition and may be involved in vulnerability to SCZ. METHODS In this study, we measured mRNA expression of the ghrelin-related molecules, growth hormone secretagogue receptor 1a (GHS-R1a) and 1b (GHS-R1b), and the ghrelin activator, membrane bound O-acyltransferase 4 (MBOAT4). Peripheral leukocytes from Japanese patients with SCZ (n = 49; 23 males, 26 females; age = 61.8 ± 13.3 years) and controls (n = 50; 25 males, 25 females; age = 62.0 ± 14.3 years) were recruited according to their clinical information. We also studied the DNA methylation rates of these genes in DNA from leukocytes. RESULTS The mRNA expression of GHS-R1a was significantly decreased in SCZ (SCZ vs. control: 0.35 ± 0.081 vs. 1.00 ± 0.059, respectively, p = 0.007), but expression levels of GHS-R1b and MBOAT4 were significantly increased in SCZ (SCZ vs. control: 2.02 ± 0.91 vs. 1.00 ± 0.32, p = 0.023, 1.37 ± 0.21 vs. 1.00 ± 0.11, respectively, p = 0.014). No differences in methylation rates for any genes were found. CONCLUSION We conclude that opposite expression of GHS-R1a and GHS-R1b, and elevated MBOAT4 mRNA expression may reflect the mechanisms of SCZ.
Collapse
|
44
|
Martínez Damonte V, Rodríguez SS, Raingo J. Growth hormone secretagogue receptor constitutive activity impairs voltage-gated calcium channel-dependent inhibitory neurotransmission in hippocampal neurons. J Physiol 2018; 596:5415-5428. [PMID: 30199095 DOI: 10.1113/jp276256] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 09/06/2018] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Presynaptic CaV 2 voltage-gated calcium channels link action potentials arriving at the presynaptic terminal to neurotransmitter release. Hence, their regulation is essential to fine tune brain circuitry. CaV 2 channels are highly sensitive to G protein-coupled receptor (GPCR) modulation. Our previous data indicated that growth hormone secretagogue receptor (GHSR) constitutive activity impairs CaV 2 channels by decreasing their surface density. We present compelling support for the impact of CaV 2.2 channel inhibition by agonist-independent GHSR activity exclusively on GABA release in hippocampal cultures. We found that this selectivity arises from a high reliance of GABA release on CaV 2.2 rather than on CaV 2.1 channels. Our data provide new information on the effects of the ghrelin-GHSR system on synaptic transmission, suggesting a putative physiological role of the constitutive signalling of a GPCR that is expressed at high levels in brain areas with restricted access to its natural agonist. ABSTRACT Growth hormone secretagogue receptor (GHSR) displays high constitutive activity, independent of its endogenous ligand, ghrelin. Unlike ghrelin-induced GHSR activity, the physiological role of GHSR constitutive activity and the mechanisms that underlie GHSR neuronal modulation remain elusive. We previously demonstrated that GHSR constitutive activity modulates presynaptic CaV 2 voltage-gated calcium channels. Here we postulate that GHSR constitutive activity-mediated modulation of CaV 2 channels could be relevant in the hippocampus since this brain area has high GHSR expression but restricted access to ghrelin. We performed whole-cell patch-clamp in hippocampal primary cultures from E16- to E18-day-old C57BL6 wild-type and GHSR-deficient mice after manipulating GHSR expression with lentiviral transduction. We found that GHSR constitutive activity impairs CaV 2.1 and CaV 2.2 native calcium currents and that CaV 2.2 basal impairment leads to a decrease in GABA but not glutamate release. We postulated that this selective effect is related to a higher CaV 2.2 over CaV 2.1 contribution to GABA release (∼40% for CaV 2.2 in wild-type vs. ∼20% in wild-type GHSR-overexpressing cultures). This effect of GHSR constitutive activity is conserved in hippocampal brain slices, where GHSR constitutive activity reduces local GABAergic transmission of the granule cell layer (intra-granule cell inhibitory postsynaptic current (IPSC) size ∼-67 pA in wild-type vs. ∼-100 pA in GHSR-deficient mice), whereas the glutamatergic output from the dentate gyrus to CA3 remains unchanged. In summary, we found that GHSR constitutive activity impairs IPSCs both in hippocampal primary cultures and in brain slices through a CaV 2-dependent mechanism without affecting glutamatergic transmission.
Collapse
Affiliation(s)
- Valentina Martínez Damonte
- Multidisciplinary Institute of Cell Biology (IMBICE), National Council of Science and Technology (CONICET), Buenos Aires Comision of Science (CIC) and La Plata University (UNLP), La Plata, Argentina
| | - Silvia Susana Rodríguez
- Multidisciplinary Institute of Cell Biology (IMBICE), National Council of Science and Technology (CONICET), Buenos Aires Comision of Science (CIC) and La Plata University (UNLP), La Plata, Argentina
| | - Jesica Raingo
- Multidisciplinary Institute of Cell Biology (IMBICE), National Council of Science and Technology (CONICET), Buenos Aires Comision of Science (CIC) and La Plata University (UNLP), La Plata, Argentina
| |
Collapse
|
45
|
Eslami M, Sadeghi B, Goshadrou F. Chronic ghrelin administration restores hippocampal long-term potentiation and ameliorates memory impairment in rat model of Alzheimer's disease. Hippocampus 2018; 28:724-734. [PMID: 30009391 DOI: 10.1002/hipo.23002] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 06/09/2018] [Accepted: 06/16/2018] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD), as a common age-related dementia, is a progressive manifestation of cognitive decline following synaptic failure resulted majorly by senile plaques composed of deposits of amyloid beta (Aβ). Ghrelin is a multifunctional peptide hormone with receptors present in various brain tissues including hippocampus and has been associated with neuroprotection, neuromodulation, and memory processing. Here, we investigated the neuroprotective and therapeutic effects of intracerebroventricular (icv) ghrelin infusion for 2 weeks on passive avoidance learning (PAL), memory retention, and synaptic plasticity in the hippocampal dentate gyrus (DG) and CA1 of both normal rats and Aβ1-42-induced neurotoxicity in AD model. Male Wistar rats were evaluated for their passive memory performance using a shuttle box while some groups had already received Aβ1-42 and/or chronic ghrelin. Using field potential recording, the induction of short- and long-term potentiation (STP and LTP) was studied in DG granule cells along with the LTP changes in CA1 pyramidal neurons through stimulation of the medial perforant path (mPP) and Schaffer collaterals (SCs), respectively. Our results demonstrated that chronic ghrelin treatment not only improved memory processing and retrieval in normal rats during the PAL task, but also promoted memory retention and alleviated memory loss by amelioration of Aβ1-42-induced synaptic plasticity impairment in AD subjects through augmentation of field excitatory postsynaptic potential (fEPSP) slope that led to LTP restitution in both the mPP-DG and the CA3-CA1 synapses. Meanwhile, STP was not significantly changed, meaning that although ghrelin enhanced postsynaptic excitability in DG, it did not change presynaptic transmitter release significantly. This suggests the involvement of postsynaptic mechanisms in long-term ghrelin-enhanced memory. In conclusion, it can be inferred that chronic ghrelin administration has an auspicious therapeutic value for impaired cognitive performance and memory deficits in AD-like neuropathology.
Collapse
Affiliation(s)
- Maryam Eslami
- Department of Physiology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahman Sadeghi
- Department of Physiology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Fatemeh Goshadrou
- Department of Physiology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Dos-Santos RC, Grover HM, Reis LC, Ferguson AV, Mecawi AS. Electrophysiological Effects of Ghrelin in the Hypothalamic Paraventricular Nucleus Neurons. Front Cell Neurosci 2018; 12:275. [PMID: 30210300 PMCID: PMC6121211 DOI: 10.3389/fncel.2018.00275] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/07/2018] [Indexed: 12/05/2022] Open
Abstract
The paraventricular nucleus (PVN) is involved in the control of sympathetic tone and the secretion of hormones, both functions known to be influenced by ghrelin, suggesting direct effect of ghrelin in this nucleus. However, the effects of ghrelin on the excitability of different PVN neuronal populations have not been demonstrated. This study assessed the effects of ghrelin on the activity of PVN neurons, correlating the responses to subpopulations of PVN neurons. We used a 64 multielectrode array to examine the effects of ghrelin administration on extracellular spike frequency in PVN neurons recorded in brain slices obtained from male Sprague-Dawley rats. Bath administration of 10 nM ghrelin increased (29/97, 30%) or decreased (37/97, 38%) spike frequency in PVN neurons. The GABAA and glutamate receptors antagonists abolish the decrease in spike frequency, without changes in the proportion of increases in spike frequency (23/53, 43%) induced by ghrelin. The results indicate a direct effect of ghrelin increasing PVN neurons activity and a synaptic dependent effect decreasing PVN neurons activity. The patch clamp recordings showed similar proportions of PVN neurons influenced by 10 nM ghrelin (33/95, 35% depolarized; 29/95, 30% hyperpolarized). Using electrophysiological fingerprints to identify specific subpopulations of PVN neurons we observed that the majority of pre-autonomic neurons (11/18 -61%) were depolarized by ghrelin, while both neuroendocrine (29% depolarizations, 40% hyperpolarizations), and magnocellular neurons (29% depolarizations, 21% hyperpolarizations) showed mixed responses. Finally, to correlate the electrophysiological response and the neurochemical phenotype of PVN neurons, cell cytoplasm was collected after recordings and RT-PCR performed to assess the presence of mRNA for vasopressin, oxytocin, thyrotropin (TRH) and corticotropin (CRH) releasing hormones. The single-cell RT-PCR showed that most TRH-expressing (4/5) and CRH-expressing (3/4) neurons are hyperpolarized in response to ghrelin. In conclusion, ghrelin either directly increases or indirectly decreases the activity of PVN neurons, this suggests that ghrelin acts on inhibitory PVN neurons that, in turn, decrease the activity of TRH-expressing and CRH-expressing neurons in the PVN.
Collapse
Affiliation(s)
- Raoni C Dos-Santos
- Department of Physiological Sciences, Institute of Biological and Health Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Brazil
| | - Hanna M Grover
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Luís C Reis
- Department of Physiological Sciences, Institute of Biological and Health Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Brazil
| | | | - André S Mecawi
- Department of Physiological Sciences, Institute of Biological and Health Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Brazil.,Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
47
|
Facilitation of hippocampal long-term potentiation and reactivation of latent HIV-1 via AMPK activation: Common mechanism of action linking learning, memory, and the potential eradication of HIV-1. Med Hypotheses 2018; 116:61-73. [DOI: 10.1016/j.mehy.2018.04.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 10/27/2017] [Accepted: 04/20/2018] [Indexed: 12/31/2022]
|
48
|
Jang JK, Kim WY, Cho BR, Lee JW, Kim JH. Locomotor sensitization is expressed by ghrelin and D1 dopamine receptor agonist in the nucleus accumbens core in amphetamine pre-exposed rat. Addict Biol 2018. [PMID: 28635134 DOI: 10.1111/adb.12533] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ghrelin modulates mesolimbic dopaminergic pathways in the brain in addition to its role in feeding. We investigated what roles ghrelin in the nucleus accumbens (NAcc) core may play in mediating locomotor activating effects of amphetamine (AMPH). First, when rats were administered with AMPH (1 mg/kg, i.p.) following a bilateral microinjection of ghrelin (0.1 or 0.5 μg/side) into the NAcc core, their locomotor activity was significantly enhanced, while these effects were blocked by co-microinjection of ghrelin receptor antagonist (0.5 μg/side) into this site. Second, we pre-exposed rats to saline or amphetamine (1 mg/kg, i.p.) every 2 to 3 days for a total of four times. After 2 weeks of drug-free withdrawal period, we examined the effect of saline, ghrelin (0.5 μg/side), D1 dopamine receptor agonist, SKF81297 (0.5 μg/side) or ghrelin (0.5 μg/side) + SKF81297 (0.5 μg/side) directly microinjected into the NAcc core on locomotor activity. When we measured rats' locomotor activity for 1 hour immediately following microinjections, only ghrelin + SKF81297 produces sensitized locomotor activity, while all others have no effects. These results suggest that ghrelin may have a distinct role in the NAcc core to provoke the sensitized locomotor activity induced by psychomotor stimulants, and further, it may produce these effects by interaction with D1 dopamine receptors.
Collapse
Affiliation(s)
- Ju Kyong Jang
- Department of Physiology, Brain Korea 21 Plus Project for Medical Science; Brain Research Institute, Yonsei University College of Medicine; Seoul South Korea
| | - Wha Young Kim
- Department of Physiology, Brain Korea 21 Plus Project for Medical Science; Brain Research Institute, Yonsei University College of Medicine; Seoul South Korea
| | - Bo Ram Cho
- Department of Physiology, Brain Korea 21 Plus Project for Medical Science; Brain Research Institute, Yonsei University College of Medicine; Seoul South Korea
| | - Jung Won Lee
- Department of Physiology, Brain Korea 21 Plus Project for Medical Science; Brain Research Institute, Yonsei University College of Medicine; Seoul South Korea
| | - Jeong-Hoon Kim
- Department of Physiology, Brain Korea 21 Plus Project for Medical Science; Brain Research Institute, Yonsei University College of Medicine; Seoul South Korea
| |
Collapse
|
49
|
Abstract
Ghrelin, a gastric-derived acylated peptide, regulates energy homeostasis by transmitting information about peripheral nutritional status to the brain, and is essential for protecting organisms against famine. Ghrelin operates brain circuits to regulate homeostatic and hedonic feeding. Recent research advances have shed new light on ghrelin's multifaceted roles in cellular homeostasis, which could maintain the internal environment and overcome metaflammation in metabolic organs. Here, we highlight our current understanding of the regulatory mechanisms of the ghrelin system in energy metabolism and cellular homeostasis and its clinical trials. Future studies of ghrelin will further elucidate how the stomach regulates systemic homeostasis.
Collapse
Affiliation(s)
- Shigehisa Yanagi
- Divisions of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Kiyotake, Miyazaki 889-1692, Japan
| | - Takahiro Sato
- Molecular Genetics, Institute of Life Science, Kurume University, Kurume 839-0864, Japan
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Masamitsu Nakazato
- Divisions of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Kiyotake, Miyazaki 889-1692, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan.
| |
Collapse
|
50
|
Li N, Song G, Wang Y, Zhu Q, Han F, Zhang C, Zhou Y. Blocking constitutive activity of GHSR1a in the lateral amygdala facilitates acquisition of conditioned taste aversion. Neuropeptides 2018; 68:22-27. [PMID: 29254662 DOI: 10.1016/j.npep.2017.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/05/2017] [Accepted: 12/05/2017] [Indexed: 11/28/2022]
Abstract
Ghrelin is a circulating peptide hormone promoting feeding and regulating energy metabolism in human and rodents. Ghrelin functions by binding to its receptor, the growth hormone secretagogue receptor 1a (GHSR1a), which are widely distributed throughout the brain including the amygdala, a brain region important for regulating valenced behavior, such as aversion. Interestingly, GHSR1a was once characterized by highly constitutive, ligand-independent activity. However, the physiological importance of such ligand-independent signaling on aversive memory processing has not been tested yet. Here, we applied [D-Arg1, D-Phe5, D-Trp7,9, Leu11]-Substance P (D-SP), a full inverse agonist for GHSR1a, into the lateral amygdala (LA) and investigated the effect of blocking GHSR1a constitutive activity on conditioned taste aversion (CTA) in rats. We found that intra-LA infusion of a single low dose of D-SP (8ng/0.5μl/side) facilitates CTA acquisition. Moreover, pre-administration of a high dose of D-SP into the LA abolishes the suppressive effect of exogenous ghrelin on CTA acquisition. In contrast, pre-administration of the same dose of D-SP does not affect the suppression of substance P, a potent neurokinin-1 (NK1) receptor ligand, on CTA. Therefore, our data indicated that the spontaneous or basal activity of GHSR1a signaling in the LA might interfere with CTA memory formation. D-SP decreases the constitutive activity of GHSR1a and thus facilitates CTA. Altogether, our present findings along with previous results support the idea that ghrelin/GHSR1a signaling in the LA circuit blocks conditioned taste aversion.
Collapse
Affiliation(s)
- Nan Li
- Department of Physiology, Medical College of Qingdao University, Qingdao 266071, Shandong, China
| | - Ge Song
- Department of Physiology, Medical College of Qingdao University, Qingdao 266071, Shandong, China
| | - Yaohui Wang
- Department of Physiology, Medical College of Qingdao University, Qingdao 266071, Shandong, China
| | - Qianqian Zhu
- Department of Physiology, Medical College of Qingdao University, Qingdao 266071, Shandong, China
| | - Fubing Han
- Department of Physiology, Medical College of Qingdao University, Qingdao 266071, Shandong, China
| | - Chonghui Zhang
- Department of Physiology, Medical College of Qingdao University, Qingdao 266071, Shandong, China
| | - Yu Zhou
- Department of Physiology, Medical College of Qingdao University, Qingdao 266071, Shandong, China.
| |
Collapse
|