1
|
Rea JJ, Liu CM, Hayes AMR, Bashaw AG, Schwartz GM, Ohan R, Décarie-Spain L, Kao AE, Klug ME, Phung KJ, Waldow AI, Wood RI, Kanoski SE. Hippocampus Oxytocin Signaling Promotes Prosocial Eating in Rats. Biol Psychiatry 2024:S0006-3223(24)01462-8. [PMID: 39038641 DOI: 10.1016/j.biopsych.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/24/2024]
Abstract
BACKGROUND Oxytocin (OT) is a hypothalamic neuropeptide involved in diverse physiological and behavioral functions, including social-based behavior and food intake control. The extent to which OT's role in regulating these 2 fundamental behaviors is interconnected is unknown, which is a critical gap in knowledge given that social factors have a strong influence on eating behavior in mammals. Here, we focus on OT signaling in the dorsal hippocampus (HPCd), a brain region recently linked to eating and social memory, as a candidate system where these functions overlap. METHODS HPCd OT signaling gain- and loss-of-function strategies were used in male Sprague Dawley rats that were trained in a novel social eating procedure to consume their first nocturnal meal under conditions that varied with regard to conspecific presence and familiarity. The endogenous role of HPCd OT signaling was also evaluated for olfactory-based social transmission of food preference learning, sociality, and social recognition memory. RESULTS HPCd OT administration had no effect on food intake under isolated conditions but significantly increased consumption in the presence of a familiar but not an unfamiliar conspecific. Supporting these results, chronic knockdown of HPCd OT receptor expression eliminated the food intake-promoting effects of a familiar conspecific. HPCd OT receptor knockdown also blocked social transmission of food preference learning and impaired social recognition memory without affecting sociality. CONCLUSIONS Collectively, the results of the current study identify endogenous HPCd OT signaling as a novel substrate in which OT synergistically influences eating and social behaviors, including the social facilitation of eating and the social transmission of food preference.
Collapse
Affiliation(s)
- Jessica J Rea
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California; Neuroscience Graduate Program, University of Southern California, Los Angeles, California
| | - Clarissa M Liu
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California; Neuroscience Graduate Program, University of Southern California, Los Angeles, California
| | - Anna M R Hayes
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Alexander G Bashaw
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California; Neuroscience Graduate Program, University of Southern California, Los Angeles, California
| | - Grace M Schwartz
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Rita Ohan
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Léa Décarie-Spain
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Alicia E Kao
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Molly E Klug
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Kenneth J Phung
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Alice I Waldow
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Ruth I Wood
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California; Department of Integrative Anatomical Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California; Neuroscience Graduate Program, University of Southern California, Los Angeles, California.
| |
Collapse
|
2
|
Yang M, Singh A, McDougle M, Décarie-Spain L, Kanoski S, de Lartigue G. Separate orexigenic hippocampal ensembles shape dietary choice by enhancing contextual memory and motivation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.09.561580. [PMID: 37873148 PMCID: PMC10592764 DOI: 10.1101/2023.10.09.561580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The hippocampus (HPC), traditionally known for its role in learning and memory, has emerged as a controller of food intake. While prior studies primarily associated the HPC with food intake inhibition, recent research suggests a critical role in appetitive processes. We hypothesized that orexigenic HPC neurons differentially respond to fats and/or sugars, potent natural reinforcers that contribute to obesity development. Results uncover previously-unrecognized, spatially-distinct neuronal ensembles within the dorsal HPC (dHPC) that are responsive to separate nutrient signals originating from the gut. Using activity-dependent genetic capture of nutrient-responsive HPC neurons, we demonstrate a causal role of both populations in promoting nutrient-specific preference through different mechanisms. Sugar-responsive neurons encode an appetitive spatial memory engram for meal location, whereas fat-responsive neurons selectively enhance the preference and motivation for fat intake. Collectively, these findings uncover a neural basis for the exquisite specificity in processing macronutrient signals from a meal that shape dietary choices.
Collapse
|
3
|
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
|
4
|
Troha R, Gowda M, Lee SLT, Markus E. Observational learning in rats: Interplay between demonstrator and observer behavior. J Neurosci Methods 2023; 388:109807. [PMID: 36731637 DOI: 10.1016/j.jneumeth.2023.109807] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 01/20/2023] [Accepted: 01/28/2023] [Indexed: 02/01/2023]
Abstract
BACKGROUND Observational learning is a vital skill for survival. This form of learning has been seen in humans and certain non-human animals. However, the neural circuitry underlying this form of learning is still poorly understood. NEW METHOD To better understand the factors underlying successful observation in rats, we employed a task where an observer must base its behavior on that of a demonstrator rat to identify a reward location. A comparison was made of behavior during a social and non-social observation condition. RESULTS Observers oriented more, responded faster and omitted less responses in the social compared to the non-social condition. Observer performance was also linked to initial orientation, proximity, and the manner in which the demonstrator rat performed the task. COMPARISON WITH EXISTING METHOD Previous work on observational learning encompassed multiple exposures to a single solution over days or weeks. The current method provides data from multiple individual novel observational learning trials, leading to much faster and more robust social learning. This method provides a clearly defined interval in which observation must take place. Allowing for precise tracking of both the observer and demonstrator behavior during the learning period. CONCLUSIONS This study highlights observer and demonstrator interplay in successful observational learning and provides a novel method for analyzing social behavior in rodents.
Collapse
Affiliation(s)
- Ryan Troha
- University of Connecticut, Storrs, CT, USA.
| | | | | | | |
Collapse
|
5
|
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
|
6
|
Xia ZD, Ma RX, Wen JF, Zhai YF, Wang YQ, Wang FY, Liu D, Zhao XL, Sun B, Jia P, Zheng XH. Pathogenesis, Animal Models, and Drug Discovery of Alzheimer's Disease. J Alzheimers Dis 2023; 94:1265-1301. [PMID: 37424469 DOI: 10.3233/jad-230326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Alzheimer's disease (AD), the most common cause of dementia, is a chronic neurodegenerative disease induced by multiple factors. The high incidence and the aging of the global population make it a growing global health concern with huge implications for individuals and society. The clinical manifestations are progressive cognitive dysfunction and lack of behavioral ability, which not only seriously affect the health and quality of life of the elderly, but also bring a heavy burden to the family and society. Unfortunately, almost all the drugs targeting the classical pathogenesis have not achieved satisfactory clinical effects in the past two decades. Therefore, the present review provides more novel ideas on the complex pathophysiological mechanisms of AD, including classical pathogenesis and a variety of possible pathogenesis that have been proposed in recent years. It will be helpful to find out the key target and the effect pathway of potential drugs and mechanisms for the prevention and treatment of AD. In addition, the common animal models in AD research are outlined and we examine their prospect for the future. Finally, Phase I, II, III, and IV randomized clinical trials or on the market of drugs for AD treatment were searched in online databases (Drug Bank Online 5.0, the U.S. National Library of Medicine, and Alzforum). Therefore, this review may also provide useful information in the research and development of new AD-based drugs.
Collapse
Affiliation(s)
- Zhao-Di Xia
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Ruo-Xin Ma
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Jin-Feng Wen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Yu-Fei Zhai
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Yu-Qi Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Feng-Yun Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Dan Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Xiao-Long Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Bao Sun
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
- Department of Pharmacy, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, PR China
| | - Pu Jia
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| | - Xiao-Hui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, PR China
| |
Collapse
|
7
|
Décarie-Spain L, Liu CM, Lauer LT, Subramanian K, Bashaw AG, Klug ME, Gianatiempo IH, Suarez AN, Noble EE, Donohue KN, Cortella AM, Hahn JD, Davis EA, Kanoski SE. Ventral hippocampus-lateral septum circuitry promotes foraging-related memory. Cell Rep 2022; 40:111402. [PMID: 36170832 PMCID: PMC9605732 DOI: 10.1016/j.celrep.2022.111402] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/27/2022] [Accepted: 08/31/2022] [Indexed: 11/30/2022] Open
Abstract
Remembering the location of a food or water source is essential for survival. Here, we reveal that spatial memory for food location is reflected in ventral hippocampus (HPCv) neuron activity and is impaired by HPCv lesion. HPCv mediation of foraging-related memory involves communication to the lateral septum (LS), as either reversible or chronic disconnection of HPCv-to-LS signaling impairs spatial memory retention for food or water location. This neural pathway selectively encodes appetitive spatial memory, as HPCv-LS disconnection does not affect spatial memory for escape location in a negative reinforcement procedure, food intake, or social and olfactory-based appetitive learning. Neural pathway tracing and functional mapping analyses reveal that LS neurons recruited during the appetitive spatial memory procedure are primarily GABAergic neurons that project to the lateral hypothalamus. Collective results emphasize that the neural substrates controlling spatial memory are outcome specific based on reinforcer modality.
Collapse
Affiliation(s)
- Léa Décarie-Spain
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Clarissa M Liu
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, University of Southern California, 3641Watt Way, Los Angeles, CA 90089, USA
| | - Logan Tierno Lauer
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Keshav Subramanian
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, University of Southern California, 3641Watt Way, Los Angeles, CA 90089, USA
| | - Alexander G Bashaw
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, University of Southern California, 3641Watt Way, Los Angeles, CA 90089, USA
| | - Molly E Klug
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Isabella H Gianatiempo
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Andrea N Suarez
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Emily E Noble
- Department of Foods and Nutrition, University of Georgia, 305 Sanford Drive, Athens, GA 30602, USA
| | - Kristen N Donohue
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Alyssa M Cortella
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Joel D Hahn
- Neurobiology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Elizabeth A Davis
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Scott E Kanoski
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, University of Southern California, 3641Watt Way, Los Angeles, CA 90089, USA.
| |
Collapse
|
8
|
Chen R, Cai G, Xu S, Sun Q, Luo J, Wang Y, Li M, Lin H, Liu J. Body mass index related to executive function and hippocampal subregion volume in subjective cognitive decline. Front Aging Neurosci 2022; 14:905035. [PMID: 36062154 PMCID: PMC9428252 DOI: 10.3389/fnagi.2022.905035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022] Open
Abstract
Objective This study aims to explore whether body mass index (BMI) level affects the executive function and hippocampal subregion volume of subjective cognitive decline (SCD). Materials and methods A total of 111 participants were included in the analysis, including SCD (38 of normal BMI, 27 of overweight and obesity) and normal cognitive control (NC) (29 of normal BMI, 17 of overweight and obesity). All subjects underwent the Chinese version of the Stroop Color-Word Test (SCWT) to measure the executive function and a high-resolution 3D T1 structural image acquisition. Two-way ANOVA was used to examine the differences in executive function and gray matter volume in hippocampal subregions under different BMI levels between the SCD and NC. Result The subdimensions of executive function in which different BMI levels interact with SCD and NC include inhibition control function [SCWT C-B reaction time(s): F (1,104) = 5.732, p = 0.018], and the hippocampal subregion volume of CA1 [F (1,99) = 8.607, p = 0.004], hippocampal tail [F (1,99) = 4.077, p = 0.046], and molecular layer [F (1,99) = 6.309, p = 0.014]. After correction by Bonferroni method, the population × BMI interaction only had a significant effect on the CA1 (p = 0.004). Further analysis found that the SCWT C-B reaction time of SCD was significantly longer than NC no matter whether it is at the normal BMI level [F (1,104) = 4.325, p = 0.040] or the high BMI level [F (1,104) = 21.530, p < 0.001], and the inhibitory control function of SCD was worse than that of NC. In the normal BMI group, gray matter volume in the hippocampal subregion (CA1) of SCD was significantly smaller than that of NC [F (1,99) = 4.938, p = 0.029]. For patients with SCD, the high BMI group had worse inhibitory control function [F (1,104) = 13.499, p < 0.001] and greater CA1 volume compared with the normal BMI group [F (1,99) = 7.619, p = 0.007]. Conclusion The BMI level is related to the inhibition control function and the gray matter volume of CA1 subregion in SCD. Overweight seems to increase the gray matter volume of CA1 in the elderly with SCD, but it is not enough to compensate for the damage to executive function caused by the disease. These data provide new insights into the relationship between BMI level and executive function of SCD from the perspective of imaging.
Collapse
Affiliation(s)
- Ruilin Chen
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Guiyan Cai
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Shurui Xu
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Qianqian Sun
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jia Luo
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yajun Wang
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Ming Li
- Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Hui Lin
- Department of Physical Education, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jiao Liu
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, China
- Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Key Laboratory of Orthopedics and Traumatology of Traditional Chinese Medicine and Rehabilitation, Ministry of Education, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
9
|
Huang Y, Yang Y, Zhao Y, Guo D, Chen L, Shi L, Xu G. DOCK4 regulates ghrelin production in gastric X/A-like cells. J Endocrinol Invest 2022; 45:1447-1454. [PMID: 35302184 DOI: 10.1007/s40618-022-01785-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/08/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Ghrelin, a gastric hormone, provides a hunger signal to the central nervous system to stimulate food intake. Ghrelin also modulates neuroinflammatory and apoptotic processes. Dedicator of cytokinesis 4 (DOCK4), a guanine nucleotide exchange factor (GEF), is involved in the regulation of neuronal polarization and axon regeneration. However, the effect of DOCK4 on ghrelin production has not been explored. METHODS The expression of DOCK4 in human and mouse stomach was examined by immunohistochemical staining. The synthesis and secretion of ghrelin in Dock4 null mice were evaluated by real-time quantitative PCR, Western blot and ELISA. The effects of DOCK4 on ghrelin production in mHypoE-42 cells were measured by real-time quantitative PCR and Western blot. RESULTS We showed that DOCK4 was expressed in both human and mouse gastric ghrelin cells. The mRNA and protein levels of gastric ghrelin, as well as ghrelin secretion, were remarkably diminished in Dock4 null mice. Furthermore, we showed that overexpression of Dock4 significantly stimulated ghrelin expression, while siRNA knockdown of endogenous Dock4 resulted in a marked decrease of ghrelin in mHypoE-N42 cells. CONCLUSIONS Our results identify DOCK4 as a critical regulator for ghrelin production in gastric X/A-like cells.
Collapse
Affiliation(s)
- Y Huang
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, 510632, Guangdong, China
| | - Y Yang
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Y Zhao
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, 510632, Guangdong, China
| | - D Guo
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - L Chen
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, 510632, Guangdong, China
| | - L Shi
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China.
| | - G Xu
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
10
|
Wellman M, Budin R, Woodside B, Abizaid A. Energetic demands of lactation produce an increase in the expression of growth hormone secretagogue receptor in the hypothalamus and ventral tegmental area of the rat despite a reduction in circulating ghrelin. J Neuroendocrinol 2022; 34:e13126. [PMID: 35365872 DOI: 10.1111/jne.13126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/07/2022] [Accepted: 02/08/2022] [Indexed: 12/26/2022]
Abstract
Lactating rats show changes in the secretion of hormones and brain signals that promote hyperphagia and facilitate the production of milk. Little is known, however, about the role of ghrelin in the mechanisms sustaining lactational hyperphagia. Here, we used Wistar female rats that underwent surgery to sever the galactophores to prevent milk delivery (GC rats) and decrease the energetic drain of milk delivery. We compared plasma acyl-ghrelin concentrations and growth hormone secretagogue receptor (GHSR) mRNA expression in different brain regions of GC rats with those of sham operated lactating and nonlactating rats. Additional lactating and nonlactating rats were implanted with cannulae aimed at the lateral ventricles and were used to compare feeding responses to central ghrelin or GHSR antagonist infusions to those of nonlactating rats receiving similar infusions on day 14-16 postpartum (pp). Results show lower plasma acyl-ghrelin concentrations on day 15 pp sham operated lactating rats compared to GC or nonlactating rats. These changes occur in association with increased GHSR mRNA expression in the hypothalamic arcuate nucleus (ARC) and ventral tegmental area (VTA) of sham operated lactating rats. Despite lactational hyperphagia, infusions of ghrelin (0.25 or 1 μg) resulted in similar increases in food intake in lactating and nonlactating rats. In addition, infusions of the GHSR antagonist JMV3002 (4 μg in 1 μl of vehicle) produced greater suppression of food intake in lactating rats than in nonlactating rats. These data suggest that, despite lower plasma ghrelin, the energetic drain of lactation increases sensitivity to the orexigenic effects of ghrelin in brain regions important for food intake and energy balance, and these events are associated with lactational hyperphagia.
Collapse
Affiliation(s)
- Martin Wellman
- Neuroscience Department, Carleton University, Ottawa, Ontario, Canada
| | - Radek Budin
- Centre for Studies in Behavioural Neurobiology, Psychology Department, Concordia University, Montreal, Quebec, Canada
| | - Barbara Woodside
- Centre for Studies in Behavioural Neurobiology, Psychology Department, Concordia University, Montreal, Quebec, Canada
| | - Alfonso Abizaid
- Neuroscience Department, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
11
|
Watts AG, Kanoski SE, Sanchez-Watts G, Langhans W. The physiological control of eating: signals, neurons, and networks. Physiol Rev 2022; 102:689-813. [PMID: 34486393 PMCID: PMC8759974 DOI: 10.1152/physrev.00028.2020] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Graciela Sanchez-Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Eidgenössische Technische Hochschule-Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
12
|
Perelló M, Cornejo MP, De Francesco PN, Fernandez G, Gautron L, Valdivia LS. The controversial role of the vagus nerve in mediating ghrelin´s actions: gut feelings and beyond. IBRO Neurosci Rep 2022; 12:228-239. [PMID: 35746965 PMCID: PMC9210457 DOI: 10.1016/j.ibneur.2022.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 12/26/2022] Open
Abstract
Ghrelin is a stomach-derived peptide hormone that acts via the growth hormone secretagogue receptor (GHSR) and displays a plethora of neuroendocrine, metabolic, autonomic and behavioral actions. It has been proposed that some actions of ghrelin are exerted via the vagus nerve, which provides a bidirectional communication between the central nervous system and peripheral systems. The vagus nerve comprises sensory fibers, which originate from neurons of the nodose and jugular ganglia, and motor fibers, which originate from neurons of the medulla. Many anatomical studies have mapped GHSR expression in vagal sensory or motor neurons. Also, numerous functional studies investigated the role of the vagus nerve mediating specific actions of ghrelin. Here, we critically review the topic and discuss the available evidence supporting, or not, a role for the vagus nerve mediating some specific actions of ghrelin. We conclude that studies using rats have provided the most congruent evidence indicating that the vagus nerve mediates some actions of ghrelin on the digestive and cardiovascular systems, whereas studies in mice resulted in conflicting observations. Even considering exclusively studies performed in rats, the putative role of the vagus nerve in mediating the orexigenic and growth hormone (GH) secretagogue properties of ghrelin remains debated. In humans, studies are still insufficient to draw definitive conclusions regarding the role of the vagus nerve mediating most of the actions of ghrelin. Thus, the extent to which the vagus nerve mediates ghrelin actions, particularly in humans, is still uncertain and likely one of the most intriguing unsolved aspects of the field.
Collapse
|
13
|
Espinoza García AS, Martínez Moreno AG, Reyes Castillo Z. The role of ghrelin and leptin in feeding behavior: Genetic and molecular evidence. ENDOCRINOL DIAB NUTR 2021; 68:654-663. [PMID: 34906346 DOI: 10.1016/j.endien.2020.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 10/02/2020] [Indexed: 06/14/2023]
Abstract
Feeding behavior is integrated within a wide variety of eating behaviors, which depend on psychosocial, biological and environmental factors. These types of behavior can cause nutrition-related diseases such as obesity, which affects more than 650 million people worldwide. Ghrelin and leptin are key hormones that regulate appetite, food intake and energy metabolism. Research in genetics suggests that genetic variants of both hormones are associated with complex forms of eating behavior, such as a preference for palatable food, making individuals susceptible to the modern obesogenic environment. This review analyses the scientific evidence around polymorphisms in the ghrelin and leptin genes and their association with eating behavior. The understanding of these mechanisms is relevant since it could impact on the objectives of pharmacological or behavioral interventions for their treatment.
Collapse
Affiliation(s)
- Astrid Selene Espinoza García
- Instituto de Investigaciones en Comportamiento Alimentario y Nutrición, Centro Universitario del Sur, Universidad de Guadalajara, Zapotlán el Grande, Jalisco, Mexico
| | - Alma Gabriela Martínez Moreno
- Instituto de Investigaciones en Comportamiento Alimentario y Nutrición, Centro Universitario del Sur, Universidad de Guadalajara, Zapotlán el Grande, Jalisco, Mexico
| | - Zyanya Reyes Castillo
- Instituto de Investigaciones en Comportamiento Alimentario y Nutrición, Centro Universitario del Sur, Universidad de Guadalajara, Zapotlán el Grande, Jalisco, Mexico.
| |
Collapse
|
14
|
Briggs SB, Hannapel R, Ramesh J, Parent MB. Inhibiting ventral hippocampal NMDA receptors and Arc increases energy intake in male rats. ACTA ACUST UNITED AC 2021; 28:187-194. [PMID: 34011515 PMCID: PMC8139633 DOI: 10.1101/lm.053215.120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/02/2021] [Indexed: 11/24/2022]
Abstract
Research into the neural mechanisms that underlie higher-order cognitive control of eating behavior suggests that ventral hippocampal (vHC) neurons, which are critical for emotional memory, also inhibit energy intake. We showed previously that optogenetically inhibiting vHC glutamatergic neurons during the early postprandial period, when the memory of the meal would be undergoing consolidation, caused rats to eat their next meal sooner and to eat more during that next meal when the neurons were no longer inhibited. The present research determined whether manipulations known to interfere with synaptic plasticity and memory when given pretraining would increase energy intake when given prior to ingestion. Specifically, we tested the effects of blocking vHC glutamatergic N-methyl-D-aspartate receptors (NMDARs) and activity-regulated cytoskeleton-associated protein (Arc) on sucrose ingestion. The results showed that male rats consumed a larger sucrose meal on days when they were given vHC infusions of the NMDAR antagonist APV or Arc antisense oligodeoxynucleotides than on days when they were given control infusions. The rats did not accommodate for that increase by delaying the onset of their next sucrose meal (i.e., decreased satiety ratio) or by eating less during the next meal. These data suggest that vHC NMDARs and Arc limit meal size and inhibit meal initiation.
Collapse
Affiliation(s)
- Sherri B Briggs
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303, USA
| | - Reilly Hannapel
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303, USA
| | - Janavi Ramesh
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303, USA
| | - Marise B Parent
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303, USA.,Department of Psychology, Georgia State University, Atlanta, Georgia 30303, USA
| |
Collapse
|
15
|
Contribution of growth hormone secretagogue receptor (GHSR) signaling in the ventral tegmental area (VTA) to the regulation of social motivation in male mice. Transl Psychiatry 2021; 11:230. [PMID: 33879778 PMCID: PMC8058340 DOI: 10.1038/s41398-021-01350-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/19/2021] [Accepted: 03/30/2021] [Indexed: 01/22/2023] Open
Abstract
Most psychiatric disorders are characterized by deficits in the ability to interact socially with others. Ghrelin, a hormone normally associated with the regulation of glucose utilization and appetite, is also implicated in the modulation of motivated behaviors including those associated with food and sex rewards. Here we hypothesized that deficits in ghrelin receptor (growth hormone secretagogue receptor; GHSR) signaling are also associated with deficits in social motivation in male mice. To test this hypothesis, we compared social motivation in male mice lacking GHSR or mice treated with the GHSR antagonist JMV2959 with that of WT or vehicle-treated mice. GHSR signaling in dopamine cells of the ventral tegmental area (VTA) has been implicated in the control of sexual behavior, thus we further hypothesized that GHSR signaling in the VTA is important for social motivation. Thus, we conducted studies where we delivered JMV2959 to block GHSR in the VTA of mice, and studies where we rescued the expression of GHSR in the VTA of GHSR knockout (KO) mice. Mice lacking GHSR or injected with JMV2959 peripherally for 3 consecutive days displayed lower social motivation as reflected by a longer latency to approach a novel conspecific and shorter interaction time compared to WT or vehicle-treated controls. Furthermore, intra-VTA infusion of JMV2959 resulted in longer latencies to approach a novel conspecific, whereas GHSR KO mice with partial rescue of the GHSR showed decreased latencies to begin a novel social interaction. Together, these data suggest that GHSR in the VTA facilitate social approach in male mice, and GHSR-signaling deficits within the VTA result in reduced motivation to interact socially.
Collapse
|
16
|
Espinoza García AS, Martínez Moreno AG, Reyes Castillo Z. The role of ghrelin and leptin in feeding behavior: Genetic and molecular evidence. ENDOCRINOL DIAB NUTR 2021; 68:S2530-0164(21)00047-1. [PMID: 33812908 DOI: 10.1016/j.endinu.2020.10.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 10/02/2020] [Accepted: 10/02/2020] [Indexed: 01/23/2023]
Abstract
Feeding behavior is integrated within a wide variety of eating behaviors, which depend on psychosocial, biological and environmental factors. These types of behavior can cause nutrition-related diseases such as obesity, which affects more than 650 million people worldwide. Ghrelin and leptin are key hormones that regulate appetite, food intake and energy metabolism. Research in genetics suggests that genetic variants of both hormones are associated with complex forms of eating behavior, such as a preference for palatable food, making individuals susceptible to the modern obesogenic environment. This review analyses the scientific evidence around polymorphisms in the ghrelin and leptin genes and their association with eating behavior. The understanding of these mechanisms is relevant since it could impact on the objectives of pharmacological or behavioral interventions for their treatment.
Collapse
Affiliation(s)
- Astrid Selene Espinoza García
- Instituto de Investigaciones en Comportamiento Alimentario y Nutrición, Centro Universitario del Sur, Universidad de Guadalajara, Zapotlán el Grande, Jalisco, México
| | - Alma Gabriela Martínez Moreno
- Instituto de Investigaciones en Comportamiento Alimentario y Nutrición, Centro Universitario del Sur, Universidad de Guadalajara, Zapotlán el Grande, Jalisco, México
| | - Zyanya Reyes Castillo
- Instituto de Investigaciones en Comportamiento Alimentario y Nutrición, Centro Universitario del Sur, Universidad de Guadalajara, Zapotlán el Grande, Jalisco, México.
| |
Collapse
|
17
|
Noble EE, Olson CA, Davis E, Tsan L, Chen YW, Schade R, Liu C, Suarez A, Jones RB, de La Serre C, Yang X, Hsiao EY, Kanoski SE. Gut microbial taxa elevated by dietary sugar disrupt memory function. Transl Psychiatry 2021; 11:194. [PMID: 33790226 PMCID: PMC8012713 DOI: 10.1038/s41398-021-01309-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/18/2021] [Accepted: 03/02/2021] [Indexed: 01/16/2023] Open
Abstract
Emerging evidence highlights a critical relationship between gut microbiota and neurocognitive development. Excessive consumption of sugar and other unhealthy dietary factors during early life developmental periods yields changes in the gut microbiome as well as neurocognitive impairments. However, it is unclear whether these two outcomes are functionally connected. Here we explore whether excessive early life consumption of added sugars negatively impacts memory function via the gut microbiome. Rats were given free access to a sugar-sweetened beverage (SSB) during the adolescent stage of development. Memory function and anxiety-like behavior were assessed during adulthood and gut bacterial and brain transcriptome analyses were conducted. Taxa-specific microbial enrichment experiments examined the functional relationship between sugar-induced microbiome changes and neurocognitive and brain transcriptome outcomes. Chronic early life sugar consumption impaired adult hippocampal-dependent memory function without affecting body weight or anxiety-like behavior. Adolescent SSB consumption during adolescence also altered the gut microbiome, including elevated abundance of two species in the genus Parabacteroides (P. distasonis and P. johnsonii) that were negatively correlated with hippocampal function. Transferred enrichment of these specific bacterial taxa in adolescent rats impaired hippocampal-dependent memory during adulthood. Hippocampus transcriptome analyses revealed that early life sugar consumption altered gene expression in intracellular kinase and synaptic neurotransmitter signaling pathways, whereas Parabacteroides microbial enrichment altered gene expression in pathways associated with metabolic function, neurodegenerative disease, and dopaminergic signaling. Collectively these results identify a role for microbiota "dysbiosis" in mediating the detrimental effects of early life unhealthy dietary factors on hippocampal-dependent memory function.
Collapse
Affiliation(s)
- Emily E. Noble
- grid.213876.90000 0004 1936 738XUniversity of Georgia, Athens, GA USA
| | - Christine A. Olson
- grid.19006.3e0000 0000 9632 6718University of California, Los Angeles, CA USA
| | - Elizabeth Davis
- grid.42505.360000 0001 2156 6853University of Southern California, Los Angeles, CA USA
| | - Linda Tsan
- grid.42505.360000 0001 2156 6853University of Southern California, Los Angeles, CA USA
| | - Yen-Wei Chen
- grid.19006.3e0000 0000 9632 6718University of California, Los Angeles, CA USA
| | - Ruth Schade
- grid.213876.90000 0004 1936 738XUniversity of Georgia, Athens, GA USA
| | - Clarissa Liu
- grid.42505.360000 0001 2156 6853University of Southern California, Los Angeles, CA USA
| | - Andrea Suarez
- grid.42505.360000 0001 2156 6853University of Southern California, Los Angeles, CA USA
| | - Roshonda B. Jones
- grid.42505.360000 0001 2156 6853University of Southern California, Los Angeles, CA USA
| | | | - Xia Yang
- grid.19006.3e0000 0000 9632 6718University of California, Los Angeles, CA USA
| | - Elaine Y. Hsiao
- grid.19006.3e0000 0000 9632 6718University of California, Los Angeles, CA USA
| | - Scott E. Kanoski
- grid.42505.360000 0001 2156 6853University of Southern California, Los Angeles, CA USA
| |
Collapse
|
18
|
Tsan L, Décarie-Spain L, Noble EE, Kanoski SE. Western Diet Consumption During Development: Setting the Stage for Neurocognitive Dysfunction. Front Neurosci 2021; 15:632312. [PMID: 33642988 PMCID: PMC7902933 DOI: 10.3389/fnins.2021.632312] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/19/2021] [Indexed: 01/18/2023] Open
Abstract
The dietary pattern in industrialized countries has changed substantially over the past century due to technological advances in agriculture, food processing, storage, marketing, and distribution practices. The availability of highly palatable, calorically dense foods that are shelf-stable has facilitated a food environment where overconsumption of foods that have a high percentage of calories derived from fat (particularly saturated fat) and sugar is extremely common in modern Westernized societies. In addition to being a predictor of obesity and metabolic dysfunction, consumption of a Western diet (WD) is related to poorer cognitive performance across the lifespan. In particular, WD consumption during critical early life stages of development has negative consequences on various cognitive abilities later in adulthood. This review highlights rodent model research identifying dietary, metabolic, and neurobiological mechanisms linking consumption of a WD during early life periods of development (gestation, lactation, juvenile and adolescence) with behavioral impairments in multiple cognitive domains, including anxiety-like behavior, learning and memory function, reward-motivated behavior, and social behavior. The literature supports a model in which early life WD consumption leads to long-lasting neurocognitive impairments that are largely dissociable from WD effects on obesity and metabolic dysfunction.
Collapse
Affiliation(s)
- Linda Tsan
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, United States.,Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, United States
| | - Léa Décarie-Spain
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, United States
| | - Emily E Noble
- Department of Foods and Nutrition, University of Georgia, Athens, GA, United States
| | - Scott E Kanoski
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, United States.,Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
19
|
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
|
20
|
Leblanc H, Ramirez S. Linking Social Cognition to Learning and Memory. J Neurosci 2020; 40:8782-8798. [PMID: 33177112 PMCID: PMC7659449 DOI: 10.1523/jneurosci.1280-20.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/16/2022] Open
Abstract
Many mammals have evolved to be social creatures. In humans, the ability to learn from others' experiences is essential to survival; and from an early age, individuals are surrounded by a social environment that helps them develop a variety of skills, such as walking, talking, and avoiding danger. Similarly, in rodents, behaviors, such as food preference, exploration of novel contexts, and social approach, can be learned through social interaction. Social encounters facilitate new learning and help modify preexisting memories throughout the lifespan of an organism. Moreover, social encounters can help buffer stress or the effects of negative memories, as well as extinguish maladaptive behaviors. Given the importance of such interactions, there has been increasing work studying social learning and applying its concepts in a wide range of fields, including psychotherapy and medical sociology. The process of social learning, including its neural and behavioral mechanisms, has also been a rapidly growing field of interest in neuroscience. However, the term "social learning" has been loosely applied to a variety of psychological phenomena, often without clear definition or delineations. Therefore, this review gives a definition for specific aspects of social learning, provides an overview of previous work at the circuit, systems, and behavioral levels, and finally, introduces new findings on the social modulation of learning. We contextualize such social processes in the brain both through the role of the hippocampus and its capacity to process "social engrams" as well as through the brainwide realization of social experiences. With the integration of new technologies, such as optogenetics, chemogenetics, and calcium imaging, manipulating social engrams will likely offer a novel therapeutic target to enhance the positive buffering effects of social experiences or to inhibit fear-inducing social stimuli in models of anxiety and post-traumatic stress disorder.
Collapse
Affiliation(s)
- Heloise Leblanc
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts, 02119
- Boston University School of Medicine, Boston, Massachusetts, 02118
| | - Steve Ramirez
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts, 02119
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02119
- Neurophotonics Center at Boston University, Boston, Massachusetts, 02119
- Center for Systems Neuroscience at Boston University, Boston, Massachusetts, 02119
| |
Collapse
|
21
|
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
|
22
|
Cornejo MP, Barrile F, Cassano D, Aguggia JP, García Romero G, Reynaldo M, Andreoli MF, De Francesco PN, Perello M. Growth hormone secretagogue receptor in dopamine neurons controls appetitive and consummatory behaviors towards high-fat diet in ad-libitum fed mice. Psychoneuroendocrinology 2020; 119:104718. [PMID: 32535402 DOI: 10.1016/j.psyneuen.2020.104718] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 01/10/2023]
Abstract
Growth hormone secretagogue receptor (GHSR), the receptor for ghrelin, is expressed in key brain nuclei that regulate food intake. The dopamine (DA) pathways have long been recognized to play key roles mediating GHSR effects on feeding behaviors. Here, we aimed to determine the role of GHSR in DA neurons controlling appetitive and consummatory behaviors towards high fat (HF) diet. For this purpose, we crossed reactivable GHSR-deficient mice with DA transporter (DAT)-Cre mice, which express Cre recombinase under the DAT promoter that is active exclusively in DA neurons, to generate mice with GHSR expression limited to DA neurons (DAT-GHSR mice). We found that DAT-GHSR mice show an increase of c-Fos levels in brain areas containing DA neurons after ghrelin treatment, in a similar fashion as seen in wild-type mice; however, they did not increase food intake or locomotor activity in response to systemically- or centrally-administered ghrelin. In addition, we found that satiated DAT-GHSR mice displayed both anticipatory activity to scheduled HF diet exposure and HF intake in a binge-like eating protocol similar to those in wild-type mice, whereas GHSR-deficient mice displayed impaired responses. We conclude that GHSR expression in DA neurons is sufficient to both mediate increased anticipatory activity to a scheduled HF diet exposure and fully orchestrate binge-like HF intake, but it is insufficient to restore the acute orexigenic or locomotor effects of ghrelin treatment. Thus, GHSR in DA neurons affects appetitive and consummatory behaviors towards HF diet that take place in the absence of caloric needs.
Collapse
Affiliation(s)
- María Paula 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
| | - 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
| | - Julieta Paola Aguggia
- 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
| | - Guadalupe García Romero
- 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
| | - Mirta Reynaldo
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - María Florencia Andreoli
- Laboratory of Experimental Neurodevelopment, Institute of Development and Pediatric Research (IDIP), La Plata Children's Hospital and Scientific Research Commission, Province of Buenos Aires (CIC-PBA)], La Plata, Buenos Aires, Argentina
| | - Pablo Nicolás 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
| | - 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
|
23
|
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
|
24
|
Quet E, Cassel JC, Cosquer B, Galloux M, Pereira De Vasconcelos A, Stéphan A. Ventral midline thalamus is not necessary for systemic consolidation of a social memory in the rat. Brain Neurosci Adv 2020; 4:2398212820939738. [PMID: 32954006 PMCID: PMC7479859 DOI: 10.1177/2398212820939738] [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: 02/28/2020] [Accepted: 06/02/2020] [Indexed: 11/18/2022] Open
Abstract
According to the standard theory of memory consolidation, recent memories
are stored in the hippocampus before their transfer to cortical
modules, a process called systemic consolidation. The ventral midline
thalamus (reuniens and rhomboid nuclei, ReRh) takes part in this
transfer as its lesion disrupts systemic consolidation of spatial and
contextual fear memories. Here, we wondered whether ReRh lesions would
also affect the systemic consolidation of another type of memory,
namely an olfaction-based social memory. To address this question we
focused on social transmission of food preference. Adult Long-Evans
rats were subjected to N-methyl-d-aspartate-induced,
fibre-sparing lesions of the ReRh nuclei or to a sham-operation, and
subsequently trained in a social transmission of food preference
paradigm. Retrieval was tested on the next day (recent memory,
nSham = 10, nReRh = 12) or after a 25-day
delay (remote memory, nSham = 10, nReRh = 10).
All rats, whether sham-operated or subjected to ReRh lesions, learned
and remembered the task normally, whatever the delay. Compared to our
former results on spatial and contextual fear memories (Ali et al.,
2017; Klein et al., 2019; Loureiro et al., 2012; Quet et al., 2020),
the present findings indicate that the ReRh nuclei might not be part
of a generic, systemic consolidation mechanism processing all kinds of
memories in order to make them persistent. The difference between
social transmission of food preference and spatial or contextual fear
memories could be explained by the fact that social transmission of
food preference is not hippocampus-dependent and that the persistence
of social transmission of food preference memory relies on different
circuits.
Collapse
Affiliation(s)
- Etienne Quet
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Centre National de la Recherche Scientifique (CNRS), Université de Strasbourg, Strasbourg, France
| | - Jean-Christophe Cassel
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Centre National de la Recherche Scientifique (CNRS), Université de Strasbourg, Strasbourg, France
| | - Brigitte Cosquer
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Centre National de la Recherche Scientifique (CNRS), Université de Strasbourg, Strasbourg, France
| | - Marine Galloux
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Centre National de la Recherche Scientifique (CNRS), Université de Strasbourg, Strasbourg, France
| | - Anne Pereira De Vasconcelos
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Centre National de la Recherche Scientifique (CNRS), Université de Strasbourg, Strasbourg, France
| | - Aline Stéphan
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Centre National de la Recherche Scientifique (CNRS), Université de Strasbourg, Strasbourg, France
| |
Collapse
|
25
|
Suarez AN, Liu CM, Cortella AM, Noble EE, Kanoski SE. Ghrelin and Orexin Interact to Increase Meal Size Through a Descending Hippocampus to Hindbrain Signaling Pathway. Biol Psychiatry 2020; 87:1001-1011. [PMID: 31836175 PMCID: PMC7188579 DOI: 10.1016/j.biopsych.2019.10.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/01/2019] [Accepted: 10/19/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Memory and cognitive processes influence the amount of food consumed during a meal, yet the neurobiological mechanisms mediating these effects are poorly understood. The hippocampus (HPC) has recently emerged as a brain region that integrates feeding-relevant biological signals with learning and memory processes to regulate feeding. We investigated whether the gut-derived hormone ghrelin acts in the ventral HPC (vHPC) to increase meal size through interactions with gut-derived satiation signaling. METHODS Interactions between vHPC ghrelin signaling, gut-derived satiation signaling, feeding, and interoceptive discrimination learning were assessed via rodent behavioral neuropharmacological approaches. Downstream neural pathways were identified using transsynaptic virus-based tracing strategies. RESULTS vHPC ghrelin signaling counteracted the food intake-reducing effects produced by various peripheral biological satiation signals, including cholecystokinin, exendin-4 (a glucagon-like peptide-1 receptor agonist), amylin, and mechanical distension of the stomach. Furthermore, vHPC ghrelin signaling produced interoceptive cues that generalized to a perceived state of energy deficit, thereby providing a potential mechanism for the attenuation of satiation processing. Neuroanatomical tracing identified a multiorder connection from vHPC neurons to lateral hypothalamic area orexin (hypocretin)-producing neurons that project to the laterodorsal tegmental nucleus in the hindbrain. Lastly, vHPC ghrelin signaling increased spontaneous meal size via downstream orexin receptor signaling in the laterodorsal tegmental nucleus. CONCLUSIONS vHPC ghrelin signaling increases meal size by counteracting the efficacy of various gut-derived satiation signals. These effects occur via downstream orexin signaling to the hindbrain laterodorsal tegmental nucleus, thereby highlighting a novel hippocampus-hypothalamus-hindbrain pathway regulating meal size control.
Collapse
Affiliation(s)
- Andrea N. Suarez
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California, USA.,Neuroscience Graduate Program, University of Southern California, Los Angeles, California, USA
| | - Clarissa M. Liu
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, USA
| | - Alyssa M. Cortella
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California, USA
| | - Emily E. Noble
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California, USA
| | - Scott E. Kanoski
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California, USA.,Neuroscience Graduate Program, University of Southern California, Los Angeles, California, USA,Correspondence: Dr. Scott E. Kanoski, Department of Biological Sciences, University of Southern California, 3560 Watt Way, PED 107, Los Angeles, CA 90089-0652, USA, Tel: +1 213 821 5762, Fax: +1 213 740 6159.
| |
Collapse
|
26
|
Berthoud HR, Morrison CD, Münzberg H. The obesity epidemic in the face of homeostatic body weight regulation: What went wrong and how can it be fixed? Physiol Behav 2020; 222:112959. [PMID: 32422162 DOI: 10.1016/j.physbeh.2020.112959] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 04/23/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022]
Abstract
Ever since the pioneering discoveries in the mid nineteen hundreds, the hypothalamus was recognized as a crucial component of the neural system controlling appetite and energy balance. The new wave of neuron-specific research tools has confirmed this key role of the hypothalamus and has delineated many other brain areas to be part of an expanded neural system sub serving these crucial functions. However, despite significant progress in defining this complex neural circuitry, many questions remain. One of the key questions is why the sophisticated body weight regulatory system is unable to prevent the rampant obesity epidemic we are experiencing. Why are pathologically obese body weight levels defended, and what can we do about it? Here we try to find answers to these questions by 1) reminding the reader that the neural controls of ingestive behavior have evolved in a demanding, restrictive environment and encompass much of the brain's major functions, far beyond the hypothalamus and brainstem, 2) hypothesizing that the current obesogenic environment impinges mainly on a critical pathway linking hypothalamic areas with the motivational and reward systems to produce uncompensated hyperphagia, and 3) proposing adequate strategies for prevention and treatment.
Collapse
Affiliation(s)
- Hans-Rudolf Berthoud
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA.
| | - Christopher D Morrison
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Heike Münzberg
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| |
Collapse
|
27
|
Observational learning of a shifting goal location in rats: Impact of distance, observed performance, familiarity, and delay. J Neurosci Methods 2020; 335:108617. [PMID: 32014445 DOI: 10.1016/j.jneumeth.2020.108617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 01/22/2020] [Accepted: 01/29/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND Observational learning allows for learning without direct exposure to danger or energetic demands. This form of learning is seen in humans, other primates, and other species such as rodents. The neurobiology behind social learning has been studied mostly in rats, specifically focusing on social transmission of food preference and fear. However, less is known regarding the neural circuitry behind social learning of a foraging scenario. NEW METHOD The current study examined observational learning in a working memory Tmaze task. The food location changed daily such that the observing animal had to learn the correct location anew each day. This delineated the time frame when an animal learned by observation, making the phenomenon easier to study. RESULTS Rats learned the location of a food reward by observing a conspecific. Furthermore, the distance of the rats from the maze affected performance. Additionally, performance was affected by whether the performer made mistakes. This memory could persist for at least five minutes. Lastly, performance was not affected by observer-demonstrator familiarity COMPARISON WITH EXISTING METHODS: Previous rodent foraging studies typically exposed observers to the same behavior over many observation sessions. In this scenario, it is difficult to determine when and how an animal learns through observation. The current task delineates the period of observation in each session, allowing manipulations during the observation period. CONCLUSIONS The current paradigm allows for repeated examinations of observational learning and provides an alternative method for neurobiological studies of social learning.
Collapse
|
28
|
Behavioural characterization of ghrelin ligands, anamorelin and HM01: Appetite and reward-motivated effects in rodents. Neuropharmacology 2020; 168:108011. [PMID: 32067989 DOI: 10.1016/j.neuropharm.2020.108011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/14/2022]
Abstract
The ghrelinergic system has been steadily investigated as a therapeutic target in the treatment of metabolic disorders and modulation of appetite. While endogenous ghrelin activates the full complement of the growth hormone secretagogue receptor (GHSR-1a) pathways, synthetic GHSR-1a ligands display biased signalling and functional selectivity, which have a significant impact on the intended and indeed, unintended, therapeutic effects. The widespread expression of the GHSR-1a receptor in vivo also necessitates an imperative consideration of the biodistribution of GHSR-1a ligands. Here, we investigate anamorelin and HM01, two recently described synthetic GHSR-1a ligands which have shown promising effects on food intake in preclinical and clinical studies. We compare the downstream signalling pathways in cellular in vitro assays, including calcium mobilization, IP-one, internalization and β-arrestin recruitment assays. We describe a novel divergent activation of central reward circuitry by anamorelin and HM01 using c-Fos immunostaining as well as behavioural effects in food intake and reward paradigms. Interestingly, we found a paradoxical reduction in reward-related behaviour for anamorelin and HM01 treated animals in our chosen paradigms. The work highlights the critical importance to consider signalling bias in relation to future ghrelin-based therapies. In addition, central access of GHSR-1a ligands, particularly to reward areas of the brain, remains a crucial factor in eliciting potent appetite-stimulating effects. The precise characterization of downstream ghrelinergic signalling and biodistribution of novel GHSR-1a ligands will be decisive in their successful development and will allow predictive modelling and design of future synthetic ligands to combat metabolic and appetite disorders involving the ghrelinergic system. This article is part of the special issue on 'Neuropeptides'.
Collapse
|
29
|
Sun Z, Wang ZT, Sun FR, Shen XN, Xu W, Ma YH, Dong Q, Tan L, Yu JT. Late-life obesity is a protective factor for prodromal Alzheimer's disease: a longitudinal study. Aging (Albany NY) 2020; 12:2005-2017. [PMID: 31986486 PMCID: PMC7053604 DOI: 10.18632/aging.102738] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 12/31/2019] [Indexed: 12/20/2022]
Abstract
Higher body mass index (BMI) in late-life has recently been considered as a possible protective factor for Alzheimer's disease (AD), which yet remains conflicting. To test this hypothesis, we have evaluated the cross-sectional and longitudinal associations of BMI categories with CSF biomarkers, brain β-amyloid (Aβ) load, brain structure, and cognition and have assessed the effect of late-life BMI on AD risk in a large sample (n = 1,212) of non-demented elderly from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database. At baseline, higher late-life BMI categories were associated with higher levels of CSF Aβ42 (p=0.037), lower levels of CSF total-tau (t-tau, p=0.026) and CSF t-tau/Aβ42 (p=0.008), lower load of Aβ in the right hippocampus (p=0.030), as well as larger volumes of hippocampus (p<0.0001), entorhinal cortex (p=0.009) and middle temporal lobe (p=0.040). But no association was found with CSF phosphorylated-tau (p-tau) or CSF p-tau/Aβ42. Longitudinal studies showed that higher BMI individuals experienced a slower decline in cognitive function. In addition, Kaplan–Meier survival analysis revealed that higher late-life BMI had a reduced risk of progression to AD over time (p = 0.009). Higher BMI in late-life decreased the risk of AD, and this process may be driven by AD-related biomarkers.
Collapse
Affiliation(s)
- Zhen Sun
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Zuo-Teng Wang
- College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China
| | - Fu-Rong Sun
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xue-Ning Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Xu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China.,College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China.,Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | -
- Data used in preparation of this article were obtained from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database (adni.loni.usc.edu). As such, the investigators within the ADNI contributed to the design and implementation of ADNI and/or provided data but did not participate in analysis or writing of this report. A complete listing of ADNI investigators can be found at: http://adni.loni.usc.edu/wp-content/uploads/how_to_apply/ADNI_Acknowledgement_List.pdf
| |
Collapse
|
30
|
Fan L, Mao C, Hu X, Zhang S, Yang Z, Hu Z, Sun H, Fan Y, Dong Y, Yang J, Shi C, Xu Y. New Insights Into the Pathogenesis of Alzheimer's Disease. Front Neurol 2020; 10:1312. [PMID: 31998208 PMCID: PMC6965067 DOI: 10.3389/fneur.2019.01312] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 11/27/2019] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD), a common neurodegenerative disease in the elderly and the most prevalent cause of dementia, is characterized by progressive cognitive impairment. The prevalence of AD continues to increase worldwide, becoming a great healthcare challenge of the twenty-first century. In the more than 110 years since AD was discovered, many related pathogenic mechanisms have been proposed, and the most recognized hypotheses are the amyloid and tau hypotheses. However, almost all clinical trials targeting these mechanisms have not identified any effective methods to treat AD. Scientists are gradually moving away from the simple assumption, as proposed in the original amyloid hypothesis, to new theories of pathogenesis, including gamma oscillations, prion transmission, cerebral vasoconstriction, growth hormone secretagogue receptor 1α (GHSR1α)-mediated mechanism, and infection. To place these findings in context, we first reviewed the neuropathology of AD and further discussed new insights in the pathogenesis of AD.
Collapse
Affiliation(s)
- Liyuan Fan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chengyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xinchao Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shuo Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhihua Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhengwei Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Huifang Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yu Fan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yali Dong
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
31
|
Pilarzyk K, Klett J, Pena EA, Porcher L, Smith AJ, Kelly MP. Loss of Function of Phosphodiesterase 11A4 Shows that Recent and Remote Long-Term Memories Can Be Uncoupled. Curr Biol 2019; 29:2307-2321.e5. [PMID: 31303492 DOI: 10.1016/j.cub.2019.06.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 03/06/2019] [Accepted: 06/07/2019] [Indexed: 12/19/2022]
Abstract
Systems consolidation is a process by which memories initially require the hippocampus for recent long-term memory (LTM) but then become increasingly independent of the hippocampus and more dependent on the cortex for remote LTM. Here, we study the role of phosphodiesterase 11A4 (PDE11A4) in systems consolidation. PDE11A4, which degrades cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP), is preferentially expressed in neurons of CA1, the subiculum, and the adjacently connected amygdalohippocampal region. In male and female mice, deletion of PDE11A enhances remote LTM for social odor recognition and social transmission of food preference (STFP) despite eliminating or silencing recent LTM for those same social events. Measurement of a surrogate marker of neuronal activation (i.e., Arc mRNA) suggests the recent LTM deficits observed in Pde11 knockout mice correspond with decreased activation of ventral CA1 relative to wild-type littermates. In contrast, the enhanced remote LTM observed in Pde11a knockout mice corresponds with increased activation and altered functional connectivity of anterior cingulate cortex, frontal association cortex, parasubiculum, and the superficial layer of medial entorhinal cortex. The apparent increased neural activation observed in prefrontal cortex of Pde11a knockout mice during remote LTM retrieval may be related to an upregulation of the N-methyl-D-aspartate receptor subunits NR1 and NR2A. Viral restoration of PDE11A4 to vCA1 alone is sufficient to rescue both the LTM phenotypes and upregulation of NR1 exhibited by Pde11a knockout mice. Together, our findings suggest remote LTM can be decoupled from recent LTM, which may have relevance for cognitive deficits associated with aging, temporal lobe epilepsy, or transient global amnesia.
Collapse
Affiliation(s)
- Katy Pilarzyk
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Road, Columbia, SC 29209, USA
| | - Jennifer Klett
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Road, Columbia, SC 29209, USA
| | - Edsel A Pena
- Department of Statistics, University of South Carolina, 1523 Green Street, Columbia, SC 29201, USA
| | - Latarsha Porcher
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Road, Columbia, SC 29209, USA
| | - Abigail J Smith
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Road, Columbia, SC 29209, USA
| | - Michy P Kelly
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Road, Columbia, SC 29209, USA.
| |
Collapse
|
32
|
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
|
33
|
Suarez AN, Noble EE, Kanoski SE. Regulation of Memory Function by Feeding-Relevant Biological Systems: Following the Breadcrumbs to the Hippocampus. Front Mol Neurosci 2019; 12:101. [PMID: 31057368 PMCID: PMC6482164 DOI: 10.3389/fnmol.2019.00101] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/03/2019] [Indexed: 12/15/2022] Open
Abstract
The hippocampus (HPC) controls fundamental learning and memory processes, including memory for visuospatial navigation (spatial memory) and flexible memory for facts and autobiographical events (declarative memory). Emerging evidence reveals that hippocampal-dependent memory function is regulated by various peripheral biological systems that are traditionally known for their roles in appetite and body weight regulation. Here, we argue that these effects are consistent with a framework that it is evolutionarily advantageous to encode and recall critical features surrounding feeding behavior, including the spatial location of a food source, social factors, post-absorptive processing, and other episodic elements of a meal. We review evidence that gut-to-brain communication from the vagus nerve and from feeding-relevant endocrine systems, including ghrelin, insulin, leptin, and glucagon-like peptide-1 (GLP-1), promote hippocampal-dependent spatial and declarative memory via neurotrophic and neurogenic mechanisms. The collective literature reviewed herein supports a model in which various stages of feeding behavior and hippocampal-dependent memory function are closely linked.
Collapse
Affiliation(s)
| | | | - Scott E. Kanoski
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
34
|
Davidson TL, Jones S, Roy M, Stevenson RJ. The Cognitive Control of Eating and Body Weight: It's More Than What You "Think". Front Psychol 2019; 10:62. [PMID: 30814963 PMCID: PMC6381074 DOI: 10.3389/fpsyg.2019.00062] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 01/10/2019] [Indexed: 12/18/2022] Open
Abstract
Over the past decade, a great deal of research has established the importance of cognitive processes in the control of energy intake and body weight. The present paper begins by identifying several of these cognitive processes. We then summarize evidence from human and nonhuman animal models, which shows how excess intake of obesity-promoting Western diet (WD) may have deleterious effects on these cognitive control processes. Findings that these effects may be manifested as early-life deficits in cognitive functioning and may also be associated with the emergence of serious late-life cognitive impairment are described. Consistent with these possibilities, we review evidence, obtained primarily from rodent models, that consuming a WD is associated with the emergence of pathophysiologies in the hippocampus, an important brain substrate for learning, memory, and cognition. The implications of this research for mechanism are discussed within the context of a “vicious-cycle model,” which describes how eating a WD could impair hippocampal function, producing cognitive deficits that promote increased WD intake and body weight gain, which could contribute to further hippocampal dysfunction, cognitive decline, and excess eating and weight gain.
Collapse
Affiliation(s)
- Terry L Davidson
- Center for Behavioral Neuroscience, Department of Psychology, American University, Washington, DC, United States
| | - Sabrina Jones
- Center for Behavioral Neuroscience, Department of Psychology, American University, Washington, DC, United States
| | - Megan Roy
- Center for Behavioral Neuroscience, Department of Psychology, American University, Washington, DC, United States
| | | |
Collapse
|
35
|
Abtahi S, Howell E, Salvucci JT, Bastacky JMR, Dunn DP, Currie PJ. Exendin-4 antagonizes the metabolic action of acylated ghrelinergic signaling in the hypothalamic paraventricular nucleus. Gen Comp Endocrinol 2019; 270:75-81. [PMID: 30336120 PMCID: PMC6886705 DOI: 10.1016/j.ygcen.2018.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 09/19/2018] [Accepted: 10/14/2018] [Indexed: 12/11/2022]
Abstract
In the current study we investigated the interaction of hypothalamic paraventricular nucleus (PVN) glucagon-like peptide-1 (GLP-1) and ghrelin signaling in the control of metabolic function. We first demonstrated that acylated ghrelin injected directly into the PVN reliably altered the respiratory exchange ratio (RER) of adult male Sprague Dawley rats. All testing was carried out during the initial 2 h of the nocturnal cycle using an indirect open circuit calorimeter. Results indicated that acylated ghrelin induced a robust increase in RER representing a shift toward enhanced carbohydrate oxidation and reduced lipid utilization. In contrast, treatment with comparable dosing of des-acyl ghrelin failed to significantly impact metabolic activity. In separate groups of rats we subsequently investigated the ability of exendin-4 (Ex-4), a GLP-1 analogue, to alter acylated ghrelin's metabolic effects. Rodents were treated with either systemic or direct PVN Ex-4 followed by acyl ghrelin microinjection. While our results showed that both systemic and PVN administration of Ex-4 significantly reduced RER, importantly, Ex-4 pretreatment itself reliably inhibited the impact of ghrelin on RER. Overall, these findings provide increasingly compelling evidence that GLP-1 and ghrelin signaling interact in the neural control of metabolic function within the PVN.
Collapse
Affiliation(s)
- Shayan Abtahi
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, United States
| | - Erin Howell
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, United States
| | - Jack T Salvucci
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, United States
| | - Joshua M R Bastacky
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, United States
| | - David P Dunn
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, United States
| | - Paul J Currie
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, United States.
| |
Collapse
|
36
|
Han JE, Frasnelli J, Zeighami Y, Larcher K, Boyle J, McConnell T, Malik S, Jones-Gotman M, Dagher A. Ghrelin Enhances Food Odor Conditioning in Healthy Humans: An fMRI Study. Cell Rep 2018; 25:2643-2652.e4. [DOI: 10.1016/j.celrep.2018.11.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/19/2018] [Accepted: 11/02/2018] [Indexed: 01/02/2023] Open
|