1
|
Dong Y, Goodwin-Groen S, Ma J, Kim E, Giudice SD, Santos M, Aoki C. Mechanisms underlying sustained resilience against anorexia nervosa from sub-anesthetic ketamine: a review and new research based on electron microscopic analyses of synapses using a mouse model. Physiol Behav 2025:114956. [PMID: 40403996 DOI: 10.1016/j.physbeh.2025.114956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/18/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
The activity-based anorexia (ABA) animal model captures key maladaptive behaviors of anorexia nervosa - starvation-evoked hyperactivity, voluntary food restriction, severe weight loss and elevated anxiety-like behavior. By repeating ABA induction, the model reveals an animal's gain of resilience against ABA relapses and concomitant synaptic plasticity. We review findings on the efficacy of sub-anesthetic ketamine administered during ABA in mid-adolescence in gaining resilience against ABA relapses, and the molecular changes evoked at medial prefrontal cortex (mPFC) synapses. GluN2B-containing NMDA receptors are significantly greater at excitatory synapses on dendritic spines of pyramidal cells. Drebrin, an F-actin binding protein that promotes activity-dependent trafficking of NMDA receptors to synaptic membranes, also increases at excitatory synapses on GABA-interneurons and pyramidal cells. These changes are at sites very near (<1 μm) but clearly not at the post-synaptic plasma membrane of excitatory synapses, enabling rapid strengthening of synapses through receptor trafficking to the plasma membrane without increasing steady-state excitability. We propose that these changes underlie the ketamine-evoked gain of resilience against anorexia-like behaviors >15 days post-injection, during ABA relapse in late adolescence. Ketamine treatment during ABA in late-adolescence ameliorates ABA relapse in adulthood >15 days later but to a lesser extent. A new EM analysis revealed that wheel running promotes GABAergic inhibitory synapse formation on pyramidal cells of the hippocampus and that ketamine augments GABAergic inhibition's contribution towards suppression of the most maladaptive behavior - starvation-evoked hyperactivity - while also augmenting food consumption, as reflected by weight gain at the end of food availability periods.
Collapse
Affiliation(s)
- Yiru Dong
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003.
| | | | - Jessie Ma
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003.
| | - Esther Kim
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003.
| | - Sophia Del Giudice
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003.
| | - Michael Santos
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003.
| | - Chiye Aoki
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003; Neuroscience Institute, NYU Langone, New York, NY 10003.
| |
Collapse
|
2
|
Alcantara-Gonzalez D, Kennedy M, Criscuolo C, Botterill J, Scharfman HE. Increased excitability of dentate gyrus mossy cells occurs early in life in the Tg2576 model of Alzheimer's disease. Alzheimers Res Ther 2025; 17:105. [PMID: 40375112 PMCID: PMC12079945 DOI: 10.1186/s13195-025-01747-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/22/2025] [Indexed: 05/18/2025]
Abstract
BACKGROUND Hyperexcitability in Alzheimer's disease (AD) is proposed to emerge early and contribute to disease progression. The dentate gyrus (DG) and its primary cell type, granule cells (GCs) are implicated in hyperexcitability in AD. Hence, we hypothesized that mossy cells (MCs), important regulators of GC excitability, contribute to early hyperexcitability in AD. Indeed, MCs and GCs are linked to hyperexcitability in epilepsy. METHODS Using the Tg2576 model of AD and WT mice (~ 1 month-old), we compared MCs and GCs electrophysiologically and morphologically, assessed the activity marker c-Fos, Aβ expression and a hippocampal- and MC-dependent memory task that is impaired at 3-4 months of age in Tg2576 mice. RESULTS Tg2576 MCs had increased spontaneous excitatory events (sEPSP/Cs) and decreased spontaneous inhibitory currents (sIPSCs), increasing the excitation/inhibition ratio. Additionally, Tg2576 MC intrinsic excitability was enhanced. Consistent with in vitro results, Tg2576 MCs showed enhanced c-Fos protein expression. Tg2576 MCs had increased intracellular Aβ expression, suggesting a reason for increased excitability. GCs showed increased excitatory and inhibitory input without changes in intrinsic properties, consistent with effects of increased MC activity. In support, increased GC activity was normalized by an antagonist of MC input to GCs. Also in support, Tg2576 MC axons showed sprouting to the area of GC dendrites. These effects occurred before an impairment in the memory task, suggesting they are extremely early alterations. CONCLUSIONS Alterations in Tg2576 MCs and GCs early in life suggest an early role for MCs in increased GC excitability. MCs may be a novel target to intervene in AD pathophysiology at early stages.
Collapse
Affiliation(s)
- David Alcantara-Gonzalez
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd. Bldg. 39, Orangeburg, NY, 10962, USA.
- Department of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York University Langone Health, New York City, NY, 10016, USA.
| | - Meghan Kennedy
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd. Bldg. 39, Orangeburg, NY, 10962, USA
| | - Chiara Criscuolo
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd. Bldg. 39, Orangeburg, NY, 10962, USA
- Department of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York University Langone Health, New York City, NY, 10016, USA
| | - Justin Botterill
- College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Helen E Scharfman
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd. Bldg. 39, Orangeburg, NY, 10962, USA.
- Department of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York University Langone Health, New York City, NY, 10016, USA.
- Neuroscience Institute, New York University Langone Health, New York City, NY, 10016, USA.
| |
Collapse
|
3
|
Kaya E, Wegienka E, Akhtarzandi-Das A, Do H, Eban-Rothschild A, Rothschild G. Food intake enhances hippocampal sharp wave-ripples. eLife 2025; 14:RP105059. [PMID: 40227932 PMCID: PMC11996173 DOI: 10.7554/elife.105059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025] Open
Abstract
Effective regulation of energy metabolism is critical for survival. Metabolic control involves various nuclei within the hypothalamus, which receive information about the body's energy state and coordinate appropriate responses to maintain homeostasis, such as thermogenesis, pancreatic insulin secretion, and food-seeking behaviors. It has recently been found that the hippocampus, a brain region traditionally associated with memory and spatial navigation, is also involved in metabolic regulation. Specifically, hippocampal sharp wave-ripples (SWRs), which are high-frequency neural oscillations supporting memory consolidation and foraging decisions, have been shown to reduce peripheral glucose levels. However, whether SWRs are enhanced by recent feeding-when the need for glucose metabolism increases, and if so, whether feeding-dependent modulation of SWRs is communicated to other brain regions involved in metabolic regulation-remains unknown. To address these gaps, we recorded SWRs from the dorsal CA1 region of the hippocampus of mice during sleep sessions before and after consumption of meals of varying caloric values. We found that SWRs occurring during sleep are significantly enhanced following food intake, with the magnitude of enhancement being dependent on the caloric content of the meal. This pattern occurred under both food-deprived and ad libitum feeding conditions. Moreover, we demonstrate that GABAergic neurons in the lateral hypothalamus, which are known to regulate food intake, exhibit a robust SWR-triggered increase in activity. These findings identify the satiety state as a factor modulating SWRs and suggest that hippocampal-lateral hypothalamic communication is a potential mechanism by which SWRs could modulate peripheral metabolism and food intake.
Collapse
Affiliation(s)
- Ekin Kaya
- Department of Psychology, University of MichiganAnn ArborUnited States
| | - Evan Wegienka
- Department of Psychology, University of MichiganAnn ArborUnited States
| | | | - Hanh Do
- Department of Psychology, University of MichiganAnn ArborUnited States
| | | | - Gideon Rothschild
- Department of Psychology, University of MichiganAnn ArborUnited States
- Kresge Hearing Research Institute and Department of Otolaryngology, Head and Neck Surgery, University of MichiganAnn ArborUnited States
| |
Collapse
|
4
|
Mendez-Hernandez R, Braga I, Bali A, Yang M, de Lartigue G. Vagal Sensory Gut-Brain Pathways That Control Eating-Satiety and Beyond. Compr Physiol 2025; 15:e70010. [PMID: 40229922 DOI: 10.1002/cph4.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
The vagus nerve is the body's primary sensory conduit from gut to brain, traditionally viewed as a passive relay for satiety signals. However, emerging evidence reveals a far more complex system-one that actively encodes diverse aspects of meal-related information, from mechanical stretch to nutrient content, metabolic state, and even microbial metabolites. This review challenges the view of vagal afferent neurons (VANs) as simple meal-termination sensors and highlights their specialized subpopulations, diverse sensory modalities, and downstream brain circuits, which shape feeding behavior, metabolism, and cognition. We integrate recent advances from single-cell transcriptomics, neural circuit mapping, and functional imaging to examine how VANs contribute to gut-brain communication beyond satiety, including their roles in food reward and memory formation. By synthesizing the latest research and highlighting emerging directions for the field, this review provides a comprehensive update on vagal sensory pathways and their role as integrators of meal information.
Collapse
Affiliation(s)
- Rebeca Mendez-Hernandez
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Isadora Braga
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Avnika Bali
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mingxin Yang
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Guillaume de Lartigue
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
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 2025; 97:540-549. [PMID: 39038641 PMCID: PMC11743826 DOI: 10.1016/j.biopsych.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [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
|
6
|
Kaya E, Wegienka E, Akhtarzandi-Das A, Do H, Eban-Rothschild A, Rothschild G. Food intake enhances hippocampal sharp wave-ripples. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.08.617304. [PMID: 39416018 PMCID: PMC11482785 DOI: 10.1101/2024.10.08.617304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Effective regulation of energy metabolism is critical for survival. Metabolic control involves various nuclei within the hypothalamus, which receive information about the body's energy state and coordinate appropriate responses to maintain homeostasis, such as thermogenesis, pancreatic insulin secretion, and food-seeking behaviors. It has recently been found that the hippocampus, a brain region traditionally associated with memory and spatial navigation, is also involved in metabolic regulation. Specifically, hippocampal sharp wave ripples (SWRs), which are high-frequency neural oscillations supporting memory consolidation and foraging decisions, have been shown to reduce peripheral glucose levels. However, whether SWRs are enhanced by recent feeding-when the need for glucose metabolism increases, and if so, whether feeding-dependent modulation of SWRs is communicated to other brain regions involved in metabolic regulation, remains unknown. To address these gaps, we recorded SWRs from the dorsal CA1 region of the hippocampus of mice during sleep sessions before and after consumption of meals of varying caloric values. We found that SWRs occurring during sleep are significantly enhanced following food intake, with the magnitude of enhancement being dependent on the caloric content of the meal. This pattern occurred under both food-deprived and ad libitum feeding conditions. Moreover, we demonstrate that GABAergic neurons in the lateral hypothalamus, which are known to regulate food intake, exhibit a robust SWR-triggered increase in activity. These findings identify the satiety state as a factor modulating SWRs and suggest that hippocampal-lateral hypothalamic communication is a potential mechanism by which SWRs could modulate peripheral metabolism and food intake.
Collapse
Affiliation(s)
- Ekin Kaya
- Department of Psychology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Evan Wegienka
- Department of Psychology, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Hanh Do
- Department of Psychology, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Gideon Rothschild
- Department of Psychology, University of Michigan, Ann Arbor, MI, 48109, USA
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
7
|
Smith W, Azevedo EP. Hunger Games: A Modern Battle Between Stress and Appetite. J Neurochem 2025; 169:e70006. [PMID: 39936619 DOI: 10.1111/jnc.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 02/13/2025]
Abstract
Stress, an evolutionarily adaptive mechanism, has become a pervasive challenge in modern life, significantly impacting feeding-relevant circuits that play a role in the development and pathogenesis of eating disorders (EDs). Stress activates the hypothalamic-pituitary-adrenal (HPA) axis, disrupts specific neural circuits, and dysregulates key brain regions, including the hypothalamus, hippocampus, and lateral septum. These particular structures are interconnected and key in integrating stress and feeding signals, modulating hunger, satiety, cognition, and emotional coping behaviors. Here we discuss the interplay between genetic predispositions and environmental factors that may exacerbate ED vulnerability. We also highlight the most commonly used animal models to study the mechanisms driving EDs and recent rodent studies that emphasize the discovery of novel cellular and molecular mechanisms integrating stress and feeding signals within the hippocampus-lateral septum-hypothalamus axis. In this review, we discuss the role of gut microbiome, an emerging area of research in the field of EDs and unanswered questions that persist and hinder the scientific progress, such as why some individuals remain resilient to stress while others become at high risk for the development of EDs. We finally discuss the need for future research delineating the impact of specific stressors on neural circuits, clarifying the relevance and functionality of hippocampal-septal-hypothalamic connectivity, and investigating the role of key neuropeptides such as CRH, oxytocin, and GLP-1 in human ED pathogenesis. Emerging tools like single-cell sequencing and advanced human imaging could uncover cellular and circuit-level changes in brain areas relevant for feeding in ED patients. Ultimately, by integrating basic and clinical research, science offers promising avenues for developing personalized, mechanism-based treatments targeting maladaptive eating behavior for patients suffering from EDs.
Collapse
Affiliation(s)
- Whitnei Smith
- Laboratory of Neurobiology of Behavior, Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Estefania P Azevedo
- Laboratory of Neurobiology of Behavior, Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
8
|
Yang M, Singh A, de Araujo A, McDougle M, Ellis H, Décarie-Spain L, Kanoski SE, de Lartigue G. Separate orexigenic hippocampal ensembles shape dietary choice by enhancing contextual memory and motivation. Nat Metab 2025; 7:276-296. [PMID: 39815079 PMCID: PMC11860247 DOI: 10.1038/s42255-024-01194-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/28/2024] [Indexed: 01/18/2025]
Abstract
The hippocampus (HPC) has emerged as a critical player in the control of food intake, beyond its well-known role in memory. While previous studies have primarily associated the HPC with food intake inhibition, recent research suggests a role in appetitive processes. Here we identified spatially distinct neuronal populations within the dorsal HPC (dHPC) that respond to either fats or sugars, potent natural reinforcers that contribute to obesity development. Using activity-dependent genetic capture of nutrient-responsive dHPC neurons, we demonstrate a causal role of both populations in promoting nutrient-specific intake through different mechanisms. Sugar-responsive neurons encoded spatial memory for sugar location, whereas fat-responsive neurons selectively enhanced the preference and motivation for fat intake. Importantly, stimulation of either nutrient-responsive dHPC neurons increased food intake, while ablation differentially impacted obesogenic diet consumption and prevented diet-induced weight gain. Collectively, these findings uncover previously unknown orexigenic circuits underlying macronutrient-specific consumption and provide a foundation for developing potential obesity treatments.
Collapse
Affiliation(s)
- Mingxin Yang
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arashdeep Singh
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alan de Araujo
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Molly McDougle
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hillary Ellis
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Léa Décarie-Spain
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Scott E Kanoski
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Guillaume de Lartigue
- Monell Chemical Senses Center, Philadelphia, PA, USA.
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Puska G, Szendi V, Egyed M, Dimén D, Cservenák M, Dobolyi Á. Maternally activated connections of the ventral lateral septum reveal input from the posterior intralaminar thalamus. Brain Struct Funct 2025; 230:27. [PMID: 39775138 DOI: 10.1007/s00429-024-02870-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 11/01/2024] [Indexed: 01/11/2025]
Abstract
The lateral septum (LS) demonstrates activation in response to pup exposure in mothers, and its lesions eliminate maternal behaviors suggesting it is part of the maternal brain circuitry. This study shows that the density of pup-activated neurons in the ventral subdivision of the LS (LSv) is nearly equivalent to that in the medial preoptic area (MPOA), the major regulatory site of maternal behavior in rat dams. However, when somatosensory inputs including suckling were not allowed, pup-activation was markedly reduced in the LSv. Retrograde tract tracing identified various brain regions potentially influencing LSv neuronal activation through their projections. Among all, anterograde tract tracing confirmed that the posterior intralaminar thalamic nucleus (PIL), implicated in processing touch-related stimuli, targets the pup-activated region of the LSv. Moreover, nerve terminals containing the maternally induced PIL neuropeptide parathyroid hormone 2 (PTH2), were found to form synaptic connections with c-Fos activated LSv neurons using electron microscopy. Confirmation of PTH2 + PIL fibers projecting to LSv was achieved by retrograde tract tracing methods. Furthermore, double retrograde injections revealed that neurons within the PIL can project to both LSv and MPOA, suggesting their simultaneous regulation by PIL input. We also established that septal neurons activated by the pups in the mother are GABAergic and send inhibitory projections to the MPOA and other components of the maternal brain circuitry. This implies that the LSv and MPOA form an interconnected subcircuit in the maternal brain network, which is primarily driven by somatosensory input from the pups via the PIL PTH2 + neurons.
Collapse
Affiliation(s)
- Gina Puska
- Department of Zoology, University of Veterinary Medicine Budapest, Budapest, Hungary
- Department of Physiology and Neurobiology, Laboratory of Molecular and Systems Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Vivien Szendi
- Department of Physiology and Neurobiology, Laboratory of Molecular and Systems Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Máté Egyed
- Department of Physiology and Neurobiology, Laboratory of Molecular and Systems Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Diána Dimén
- Department of Physiology and Neurobiology, Laboratory of Molecular and Systems Neurobiology, Eötvös Loránd University, Budapest, Hungary
- Department of Psychological and Brain Sciences, Addiction and Neuroplasticity Laboratory, Indiana University, Bloomington, IN, USA
| | - Melinda Cservenák
- Department of Physiology and Neurobiology, Laboratory of Molecular and Systems Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Árpád Dobolyi
- Department of Physiology and Neurobiology, Laboratory of Molecular and Systems Neurobiology, Eötvös Loránd University, Budapest, Hungary.
- Department of Anatomy, Histology and Embryology, Laboratory of Neuromorphology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
10
|
Scharfman HE. Towards an Understanding of the Dentate Gyrus Hilus. Hippocampus 2025; 35:e23677. [PMID: 39721944 DOI: 10.1002/hipo.23677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024]
Abstract
For many years, the hilus of the dentate gyrus (DG) was a mystery because anatomical data suggested a bewildering array of cells without clear organization. Moreover, some of the anatomical information led to more questions than answers. For example, it had been identified that one of the major cell types in the hilus, the mossy cell, innervates granule cells (GCs). However, mossy cells also targeted local GABAergic neurons. Furthermore, it was not yet clear if mossy cells were glutamatergic or GABAergic. This led to many debates about the role of mossy cells. However, it was clear that hilar neurons, including mossy cells, were likely to have very important functions because they provided strong input to GCs. Hilar neurons also attracted attention in epilepsy because pathological studies showed that hilar neurons were often lost, but GCs remained. Vulnerability of hilar neurons also occurred after traumatic brain injury and ischemia. These observations fueled an interest to understand hilar neurons and protect them, an interest that continues to this day. This article provides a historical and personal perspective into the ways that I sought to contribute to resolving some of the debates and moving the field forward. Despite several technical challenges the outcomes of the studies have been worth the effort with some surprising findings along the way. Given the growing interest in the hilus, and the advent of multiple techniques to selectively manipulate hilar neurons, there is a great opportunity for future research.
Collapse
Affiliation(s)
- Helen E Scharfman
- Department of Child and Adolescent Psychology, Neuroscience & Physiology, and Psychiatry and the Neuroscience Institute, New York University Grossman School of Medicine, New York University Langone Health, New York, New York, USA
- Center for Dementia Research, The Nathan S. Kline Institute of Psychiatric Research, Orangeburg, New York, USA
| |
Collapse
|
11
|
Bernstein HL, Lu YL, Botterill JJ, Duffy ÁM, LaFrancois JJ, Scharfman HE. Field EPSPs of Dentate Gyrus Granule Cells Studied by Selective Optogenetic Activation of Hilar Mossy Cells in Hippocampal Slices. Hippocampus 2025; 35:e23652. [PMID: 39665517 DOI: 10.1002/hipo.23652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/05/2024] [Accepted: 11/19/2024] [Indexed: 12/13/2024]
Abstract
Glutamatergic dentate gyrus (DG) mossy cells (MCs) innervate the primary DG cell type, granule cells (GCs). Numerous MC synapses are on GC proximal dendrites in the inner molecular layer (IML). However, field recordings of the GC excitatory postsynaptic potential (fEPSPs) have not been used to study this pathway selectively. Here we describe methods to selectively activate MC axons in the IML using mice with Cre recombinase expressed in MCs. Slices were made after injecting adeno-associated virus (AAV) encoding channelrhodopsin (ChR2) in the DG. In these slices, we show that fEPSPs could be recorded reliably in the IML in response to optogenetic stimulation of MC axons. Furthermore, fEPSPs were widespread across the septotemporal axis. However, fEPSPs were relatively weak because they were small in amplitude and did not elicit a significant population spike in GCs. They also showed little paired pulse facilitation. We confirmed the extracellular findings with patch clamp recordings of GCs despite different recording chambers and other differences in methods. Together the results provide a simple method for studying MC activation of GCs and add to the evidence that this input is normally weak but widespread across the GC population.
Collapse
Affiliation(s)
- Hannah L Bernstein
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, and the Neuroscience Institute, New York University Langone Health, New York, New York, USA
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, New York State Office of Mental Health, Orangeburg, New York, USA
| | - Yi-Ling Lu
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, and the Neuroscience Institute, New York University Langone Health, New York, New York, USA
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, New York State Office of Mental Health, Orangeburg, New York, USA
| | - Justin J Botterill
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, and the Neuroscience Institute, New York University Langone Health, New York, New York, USA
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, New York State Office of Mental Health, Orangeburg, New York, USA
| | - Áine M Duffy
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, and the Neuroscience Institute, New York University Langone Health, New York, New York, USA
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, New York State Office of Mental Health, Orangeburg, New York, USA
| | - John J LaFrancois
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, and the Neuroscience Institute, New York University Langone Health, New York, New York, USA
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, New York State Office of Mental Health, Orangeburg, New York, USA
| | - Helen E Scharfman
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, and the Neuroscience Institute, New York University Langone Health, New York, New York, USA
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, New York State Office of Mental Health, Orangeburg, New York, USA
| |
Collapse
|
12
|
Bernstein HL, Lu YL, Botterill JJ, Duffy ÁM, LaFrancois JJ, Scharfman HE. Field EPSPs of dentate gyrus granule cells studied by selective optogenetic activation of hilar mossy cells in hippocampal slices. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.08.622679. [PMID: 39574724 PMCID: PMC11580982 DOI: 10.1101/2024.11.08.622679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Glutamatergic dentate gyrus (DG) mossy cells (MCs) innervate the primary DG cell type, granule cells (GCs). Numerous MC synapses are on GC proximal dendrites in the inner molecular layer (IML). However, field recordings of the GC excitatory postsynaptic potential (fEPSPs) have not been used to study this pathway selectively. Here we describe methods to selectively activate MC axons in the IML using mice with Cre recombinase expressed in MCs. Slices were made after injecting adeno-associated virus (AAV) encoding channelrhodopsin (ChR2) in the DG. In these slices, we show that fEPSPs could be recorded reliably in the IML in response to optogenetic stimulation of MC axons. Furthermore, fEPSPs were widespread across the septotemporal axis. However, fEPSPs were relatively weak because they were small in amplitude and did not elicit a significant population spike in GCs. They also showed little paired pulse facilitation. We confirmed the extracellular findings with patch clamp recordings of GCs despite different recording chambers and other differences in methods. Together the results provide a simple method for studying MC activation of GCs and add to the evidence that this input is normally weak but widespread across the GC population. KEY POINTS We describe a method to activate the MC input to GCs selectively using optogenetics in hippocampal slicesMC excitation is weakly excitatory but so common among GCs that a field EPSP is generated at the site of MC synapses on GCsMC excitation of GCs is consistent across the septotemporal axis and contralaterallyUsing the characteristics of optogenetically-evoked fEPSPs, electrical stimulation of the MC input to GCs can be optimized.
Collapse
|
13
|
Kendig MD, Corbit LH. Cue-potentiated feeding in rodents: Implications for weight regulation in obesogenic environments. Neurobiol Learn Mem 2024; 215:107984. [PMID: 39265925 DOI: 10.1016/j.nlm.2024.107984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/16/2024] [Accepted: 09/06/2024] [Indexed: 09/14/2024]
Abstract
Cue-potentiated feeding (CPF) describes instances where food intake is increased by exposure to conditioned cues associated with food, often in the absence of hunger. CPF effects have been reported in a range of experimental protocols developed by researchers working across diverse fields spanning behavioural neuroscience, social psychology and ecology. Here we review the evolution of research on cue-potentiated feeding in animal models to identify important behavioural parameters and key neural circuits and pharmacological systems underlying the effect. Overall, evidence indicates that social, discrete and contextual stimuli can be used to elicit CPF effects across multiple species, though effects are often subtle and sensitive to procedural variables. While regular exposure to food cues is thought to be a key risk factor for overeating in so-called 'obesogenic' environments, further work is needed to identify whether CPF promotes positive energy balance and weight gain over the longer term. We suggest several methodological and conceptual areas for inquiry to elucidate the contribution of CPF to the regulation of food choice and energy intake.
Collapse
Affiliation(s)
- Michael D Kendig
- School of Life Sciences, University of Technology Sydney, Australia.
| | - Laura H Corbit
- Department of Psychology and Department of Cell and Systems Biology, University of Toronto, Canada.
| |
Collapse
|
14
|
Mohammadkhani A, Mitchell C, James MH, Borgland SL, Dayas CV. Contribution of hypothalamic orexin (hypocretin) circuits to pathologies of motivation. Br J Pharmacol 2024; 181:4430-4449. [PMID: 39317446 PMCID: PMC11458361 DOI: 10.1111/bph.17325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 06/17/2024] [Accepted: 06/28/2024] [Indexed: 09/26/2024] Open
Abstract
The orexin (also known as hypocretin) system, consisting of neuropeptides orexin-A and orexin-B, was discovered over 25 years ago and was immediately identified as a central regulator of sleep and wakefulness. These peptides interact with two G-protein coupled receptors, orexin 1 (OX1) and orexin 2 (OX2) receptors which are capable of coupling to all heterotrimeric G-protein subfamilies, but primarily transduce increases in calcium signalling. Orexin neurons are regulated by a variety of transmitter systems and environmental stimuli that signal reward availability, including food and drug related cues. Orexin neurons are also activated by anticipation, stress, cues predicting motivationally relevant information, including those predicting drugs of abuse, and engage neuromodulatory systems, including dopamine neurons of the ventral tegmental area (VTA) to respond to these signals. As such, orexin neurons have been characterized as motivational activators that coordinate a range of functions, including feeding and arousal, that allow the individual to respond to motivationally relevant information, critical for survival. This review focuses on the role of orexins in appetitive motivation and highlights a role for these neuropeptides in pathologies characterized by inappropriately high levels of motivated arousal (overeating, anxiety and substance use disorders) versus those in which motivation is impaired (depression).
Collapse
Affiliation(s)
- Aida Mohammadkhani
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, Alberta, Canada
| | - Caitlin Mitchell
- School of Biomedical Sciences and Pharmacy, University of Newcastle, University Drive, Callaghan, New South Wales, Australia
- The Hunter Medical Research, New Lambton Heights, New South Wales, Australia
| | - Morgan H James
- Department of Psychiatry and Brain Health Institute, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, Alberta, Canada
| | - Christopher V Dayas
- School of Biomedical Sciences and Pharmacy, University of Newcastle, University Drive, Callaghan, New South Wales, Australia
- The Hunter Medical Research, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
15
|
Goode TD, Alipio JB, Besnard A, Pathak D, Kritzer-Cheren MD, Chung A, Duan X, Sahay A. A dorsal hippocampus-prodynorphinergic dorsolateral septum-to-lateral hypothalamus circuit mediates contextual gating of feeding. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606427. [PMID: 39149322 PMCID: PMC11326193 DOI: 10.1101/2024.08.02.606427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Adaptive regulation of feeding depends on linkage of internal states and food outcomes with contextual cues. Human brain imaging has identified dysregulation of a hippocampal-lateral hypothalamic area (LHA) network in binge eating, but mechanistic instantiation of underlying cell-types and circuitry is lacking. Here, we identify an evolutionary conserved and discrete Prodynorphin (Pdyn)-expressing subpopulation of Somatostatin (Sst)-expressing inhibitory neurons in the dorsolateral septum (DLS) that receives primarily dorsal, but not ventral, hippocampal inputs. DLS(Pdyn) neurons inhibit LHA GABAergic neurons and confer context- and internal state-dependent calibration of feeding. Viral deletion of Pdyn in the DLS mimicked effects seen with optogenetic silencing of DLS Pdyn INs, suggesting a potential role for DYNORPHIN-KAPPA OPIOID RECEPTOR signaling in contextual regulation of food-seeking. Together, our findings illustrate how the dorsal hippocampus has evolved to recruit an ancient LHA feeding circuit module through Pdyn DLS inhibitory neurons to link contextual information with regulation of food consumption.
Collapse
Affiliation(s)
- Travis D Goode
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Jason Bondoc Alipio
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Antoine Besnard
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Devesh Pathak
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Michael D Kritzer-Cheren
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Ain Chung
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Xin Duan
- Department of Ophthalmology, University of California, San Francisco, CA
- Department of Physiology, University of California, San Francisco, CA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA
| | - Amar Sahay
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| |
Collapse
|
16
|
Puska G, Szendi V, Dobolyi A. Lateral septum as a possible regulatory center of maternal behaviors. Neurosci Biobehav Rev 2024; 161:105683. [PMID: 38649125 DOI: 10.1016/j.neubiorev.2024.105683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 04/09/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Abstract
The lateral septum (LS) is involved in controlling anxiety, aggression, feeding, and other motivated behaviors. Lesion studies have also implicated the LS in various forms of caring behaviors. Recently, novel experimental tools have provided a more detailed insight into the function of the LS, including the specific role of distinct cell types and their neuronal connections in behavioral regulations, in which the LS participates. This article discusses the regulation of different types of maternal behavioral alterations using the distributions of established maternal hormones such as prolactin, estrogens, and the neuropeptide oxytocin. It also considers the distribution of neurons activated in mothers in response to pups and other maternal activities, as well as gene expressional alterations in the maternal LS. Finally, this paper proposes further research directions to keep up with the rapidly developing knowledge on maternal behavioral control in other maternal brain regions.
Collapse
Affiliation(s)
- Gina Puska
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary; Department of Zoology, University of Veterinary Medicine Budapest, Budapest, Hungary
| | - Vivien Szendi
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Arpád Dobolyi
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary; Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
17
|
Tan B, Browne CJ, Nöbauer T, Vaziri A, Friedman JM, Nestler EJ. Drugs of abuse hijack a mesolimbic pathway that processes homeostatic need. Science 2024; 384:eadk6742. [PMID: 38669575 PMCID: PMC11077477 DOI: 10.1126/science.adk6742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 02/26/2024] [Indexed: 04/28/2024]
Abstract
Drugs of abuse are thought to promote addiction in part by "hijacking" brain reward systems, but the underlying mechanisms remain undefined. Using whole-brain FOS mapping and in vivo single-neuron calcium imaging, we found that drugs of abuse augment dopaminoceptive ensemble activity in the nucleus accumbens (NAc) and disorganize overlapping ensemble responses to natural rewards in a cell type-specific manner. Combining FOS-Seq, CRISPR-perturbation, and single-nucleus RNA sequencing, we identified Rheb as a molecular substrate that regulates cell type-specific signal transduction in NAc while enabling drugs to suppress natural reward consumption. Mapping NAc-projecting regions activated by drugs of abuse revealed input-specific effects on natural reward consumption. These findings characterize the dynamic, molecular and circuit basis of a common reward pathway, wherein drugs of abuse interfere with the fulfillment of innate needs.
Collapse
Affiliation(s)
- Bowen Tan
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University; New York, NY 10065, USA
| | - Caleb J. Browne
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health; Toronto, ON, M5T 1R8, Canada
| | - Tobias Nöbauer
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University; New York, NY 10065, USA
| | - Alipasha Vaziri
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University; New York, NY 10065, USA
- The Kavli Neural Systems Institute, The Rockefeller University; New York, NY 10065, USA
| | - Jeffrey M. Friedman
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University; New York, NY 10065, USA
| | - Eric J. Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| |
Collapse
|
18
|
Xue Y, Zhang YN, Wang M, Fu HY, Mao YC, Hu M, Sun MT, Guo HG, Cao L, Feng CZ. Prolonged oral intake of green tea polyphenols attenuates delirium-like behaviors in mice induced by anesthesia/surgery. Heliyon 2024; 10:e26200. [PMID: 38495146 PMCID: PMC10943306 DOI: 10.1016/j.heliyon.2024.e26200] [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: 06/15/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 03/19/2024] Open
Abstract
Postoperative delirium (POD) is a severe postoperative complication characterized by delirium-like symptoms. So far, no effective preventable strategy for POD prevention has been identified. Reports show that the consumption of green tea polyphenols (GTP) is associated with better cognitive function by modulating the composition of gut microbiota. Whether GTP also play a role in alleviating POD through gut microbiota is unknown. Herein, we studied the effect of prolonged (eight weeks) GTP intake on postoperative delirium in C57BL/6 mice with laparotomies under isoflurane anesthesia (anesthesia/surgery). We subsequently investigated anesthesia/surgery caused behavioral changes and increased the expression of malondialdehyde (MAD), an oxidative stress marker, and the activities of superoxide dismutase (SOD), an antioxidant marker, in the mice at 6 h after anesthesia/surgery. However, GTP administration reversed these changes and alleviated anesthesia/surgery-induced decrease in the abundance of gut bacterial genera, Roseburia. Further, fecal microbiota transplant demonstrated that compared with mice in the control group, treatment of C57BL/6 mice with feces from GTP-treated mice had a slight effect on the behavioral changes of mice. These data suggest that daily consumption of GTP could protect against anesthesia/surgery-induced behavioral changes, which is closely associated with gut microbiota modification by GTP.
Collapse
Affiliation(s)
- Yao Xue
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Yan-Na Zhang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Man Wang
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Hui-Yuan Fu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Ying-Chao Mao
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Min Hu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Mei-Tao Sun
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Hong-Gang Guo
- Center of Animal Research, Hangzhou Medical College, Hangzhou, China
| | - Lin Cao
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chen-Zhuo Feng
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
19
|
Wee RWS, Mishchanchuk K, AlSubaie R, Church TW, Gold MG, MacAskill AF. Internal-state-dependent control of feeding behavior via hippocampal ghrelin signaling. Neuron 2024; 112:288-305.e7. [PMID: 37977151 DOI: 10.1016/j.neuron.2023.10.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/13/2023] [Accepted: 10/12/2023] [Indexed: 11/19/2023]
Abstract
Hunger is an internal state that not only invigorates feeding but also acts as a contextual cue for higher-order control of anticipatory feeding-related behavior. The ventral hippocampus is crucial for differentiating optimal behavior across contexts, but how internal contexts such as hunger influence hippocampal circuitry is unknown. In this study, we investigated the role of the ventral hippocampus during feeding behavior across different states of hunger in mice. We found that activity of a unique subpopulation of neurons that project to the nucleus accumbens (vS-NAc neurons) increased when animals investigated food, and this activity inhibited the transition to begin eating. Increases in the level of the peripheral hunger hormone ghrelin reduced vS-NAc activity during this anticipatory phase of feeding via ghrelin-receptor-dependent increases in postsynaptic inhibition and promoted the initiation of eating. Together, these experiments define a ghrelin-sensitive hippocampal circuit that informs the decision to eat based on internal state.
Collapse
Affiliation(s)
- Ryan W S Wee
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Karyna Mishchanchuk
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Rawan AlSubaie
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Timothy W Church
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Matthew G Gold
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Andrew F MacAskill
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK.
| |
Collapse
|
20
|
Bénac N, Ezequiel Saraceno G, Butler C, Kuga N, Nishimura Y, Yokoi T, Su P, Sasaki T, Petit-Pedrol M, Galland R, Studer V, Liu F, Ikegaya Y, Sibarita JB, Groc L. Non-canonical interplay between glutamatergic NMDA and dopamine receptors shapes synaptogenesis. Nat Commun 2024; 15:27. [PMID: 38167277 PMCID: PMC10762086 DOI: 10.1038/s41467-023-44301-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024] Open
Abstract
Direct interactions between receptors at the neuronal surface have long been proposed to tune signaling cascades and neuronal communication in health and disease. Yet, the lack of direct investigation methods to measure, in live neurons, the interaction between different membrane receptors at the single molecule level has raised unanswered questions on the biophysical properties and biological roles of such receptor interactome. Using a multidimensional spectral single molecule-localization microscopy (MS-SMLM) approach, we monitored the interaction between two membrane receptors, i.e. glutamatergic NMDA (NMDAR) and G protein-coupled dopamine D1 (D1R) receptors. The transient interaction was randomly observed along the dendritic tree of hippocampal neurons. It was higher early in development, promoting the formation of NMDAR-D1R complexes in an mGluR5- and CK1-dependent manner, favoring NMDAR clusters and synaptogenesis in a dopamine receptor signaling-independent manner. Preventing the interaction in the neonate, and not adult, brain alters in vivo spontaneous neuronal network activity pattern in male mice. Thus, a weak and transient interaction between NMDAR and D1R plays a structural and functional role in the developing brain.
Collapse
Affiliation(s)
- Nathan Bénac
- Univ. Bordeaux, CNRS, IINS, UMR 5297, F-33000, Bordeaux, France
| | | | - Corey Butler
- Univ. Bordeaux, CNRS, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Nahoko Kuga
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-0033, Japan
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-aoba, Sendai, Miyagi, 980-8578, Japan
| | - Yuya Nishimura
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Taiki Yokoi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-aoba, Sendai, Miyagi, 980-8578, Japan
| | - Ping Su
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Canada
| | - Takuya Sasaki
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-0033, Japan
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-aoba, Sendai, Miyagi, 980-8578, Japan
| | | | - Rémi Galland
- Univ. Bordeaux, CNRS, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Vincent Studer
- Univ. Bordeaux, CNRS, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Fang Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Canada
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-0033, Japan
- Center for Information and Neural Networks, Suita City, Osaka, 565-0871, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, 113-0033, Japan
| | | | - Laurent Groc
- Univ. Bordeaux, CNRS, IINS, UMR 5297, F-33000, Bordeaux, France.
| |
Collapse
|
21
|
Noguchi H, Arela JC, Ngo T, Cocas L, Pleasure S. Shh from mossy cells contributes to preventing NSC pool depletion after seizure-induced neurogenesis and in aging. eLife 2023; 12:RP91263. [PMID: 38079471 PMCID: PMC10712957 DOI: 10.7554/elife.91263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
Epileptic seizures induce aberrant neurogenesis from resident neural stem cells (NSCs) in the dentate gyrus of the adult mouse hippocampus, which has been implicated in depletion of the NSC pool and impairment of hippocampal function. However, the mechanisms regulating neurogenesis after seizures remain unknown. Here, we demonstrate that Sonic hedgehog (Shh) from mossy cells is a major source of Shh signaling activity after seizures, by which mossy cells contribute to seizure-induced neurogenesis and maintenance of the NSC pool. Deletion of Shh from mossy cells attenuates seizure-induced neurogenesis. Moreover, in the absence of Shh from mossy cells, NSCs pool are prematurely depleted after seizure-induced proliferation, and NSCs have impaired self-renewal. Likewise, lack of Shh from mossy cells accelerates age-related decline of the NSC pool with accompanying reduction of self-renewal of NSCs outside the context of pathology such as seizures. Together, our findings indicate that Shh from mossy cells is critical to maintain NSCs and to prevent exhaustion from excessive consumption in aging and after seizures.
Collapse
Affiliation(s)
- Hirofumi Noguchi
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
| | - Jessica Chelsea Arela
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
| | - Thomas Ngo
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
| | - Laura Cocas
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
- Santa Clara University, Biology Department, Neuroscience ProgramSanta ClaraUnited States
| | - Samuel Pleasure
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
- Programs in Neuroscience and Developmental & Stem Cell Biology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San FranciscoSan FranciscoUnited States
| |
Collapse
|
22
|
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
|
23
|
Yang R, Zhang Y, Deng Y, Yang Y, Zhong W, Zhu L. 2-Ethylhexyl Diphenyl Phosphate Causes Obesity in Zebrafish by Stimulating Overeating via Inhibition of Dopamine Receptor D2. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:14162-14172. [PMID: 37704188 DOI: 10.1021/acs.est.3c04070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Obesity is a popular public health problem worldwide and is mainly caused by overeating, but little is known about the impacts of synthetic chemicals on obesity. Herein, we evaluated the obesogenic effect caused by 2-ethylhexyl diphenyl phosphate (EHDPHP) on zebrafish. Adult zebrafish were exposed to 5, 35, and 245 μg/L of EHDPHP for 21 days. Results showed that EHDPHP exposure significantly promoted the feeding behavior of zebrafish, as evidenced by shorter reaction time, increased average food intake, feeding rate, and intake frequency (p < 0.05). Transcriptomic, real-time quantitative PCR, and neurotransmitter analyses revealed that the dopamine (DA) receptor D2 (DRD2) was inhibited, which interfered with the DA neural reward regulation system, thus stimulating food addiction to zebrafish. This was further verified by the restored DRD2 after 7 days of Halo (a DRD2 agonist) treatment. A strong interaction between EHDPHP and DRD2 was identified via molecular docking. As a consequence of the abnormal feeding behavior, the exposed fish exhibited significant obesity evidenced by increased body weight, body mass index, plasma total cholesterol, triglyceride, and body fat content. Additionally, the pathways linked to Parkinson's disease, alcoholism, and cocaine addiction were also disrupted, implying that EHDPHP might cause other neurological disorders via the disrupted DA system.
Collapse
Affiliation(s)
- Rongyan Yang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Yuan Zhang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Yun Deng
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Yi Yang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Wenjue Zhong
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Lingyan Zhu
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| |
Collapse
|
24
|
Tan B, Browne CJ, Nöbauer T, Vaziri A, Friedman JM, Nestler EJ. Drugs of abuse hijack a mesolimbic pathway that processes homeostatic need. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.03.556059. [PMID: 37732251 PMCID: PMC10508763 DOI: 10.1101/2023.09.03.556059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Addiction prioritizes drug use over innate needs by "hijacking" brain circuits that direct motivation, but how this develops remains unclear. Using whole-brain FOS mapping and in vivo single-neuron calcium imaging, we find that drugs of abuse augment ensemble activity in the nucleus accumbens (NAc) and disorganize overlapping ensemble responses to natural rewards in a cell-type-specific manner. Combining "FOS-Seq", CRISPR-perturbations, and snRNA-seq, we identify Rheb as a shared molecular substrate that regulates cell-type-specific signal transductions in NAc while enabling drugs to suppress natural reward responses. Retrograde circuit mapping pinpoints orbitofrontal cortex which, upon activation, mirrors drug effects on innate needs. These findings deconstruct the dynamic, molecular, and circuit basis of a common reward circuit, wherein drug value is scaled to promote drug-seeking over other, normative goals.
Collapse
Affiliation(s)
- Bowen Tan
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
- These authors contributed equally
| | - Caleb J. Browne
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- These authors contributed equally
| | - Tobias Nöbauer
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA
| | - Alipasha Vaziri
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA
- The Kavli Neural Systems Institute, The Rockefeller University, New York, NY 10065, USA
| | - Jeffrey M. Friedman
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Eric J. Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
25
|
Barbosa DAN, Gattas S, Salgado JS, Kuijper FM, Wang AR, Huang Y, Kakusa B, Leuze C, Luczak A, Rapp P, Malenka RC, Hermes D, Miller KJ, Heifets BD, Bohon C, McNab JA, Halpern CH. An orexigenic subnetwork within the human hippocampus. Nature 2023; 621:381-388. [PMID: 37648849 PMCID: PMC10499606 DOI: 10.1038/s41586-023-06459-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 07/20/2023] [Indexed: 09/01/2023]
Abstract
Only recently have more specific circuit-probing techniques become available to inform previous reports implicating the rodent hippocampus in orexigenic appetitive processing1-4. This function has been reported to be mediated at least in part by lateral hypothalamic inputs, including those involving orexigenic lateral hypothalamic neuropeptides, such as melanin-concentrating hormone5,6. This circuit, however, remains elusive in humans. Here we combine tractography, intracranial electrophysiology, cortico-subcortical evoked potentials, and brain-clearing 3D histology to identify an orexigenic circuit involving the lateral hypothalamus and converging in a hippocampal subregion. We found that low-frequency power is modulated by sweet-fat food cues, and this modulation was specific to the dorsolateral hippocampus. Structural and functional analyses of this circuit in a human cohort exhibiting dysregulated eating behaviour revealed connectivity that was inversely related to body mass index. Collectively, this multimodal approach describes an orexigenic subnetwork within the human hippocampus implicated in obesity and related eating disorders.
Collapse
Affiliation(s)
- Daniel A N Barbosa
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sandra Gattas
- Department of Electrical Engineering and Computer Science, University of California, Irvine, Irvine, CA, USA
| | - Juliana S Salgado
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Fiene Marie Kuijper
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
- Université Paris Cité, Paris, France
- Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Allan R Wang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuhao Huang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Bina Kakusa
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Christoph Leuze
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Artur Luczak
- Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Paul Rapp
- Department of Military & Emergency Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Robert C Malenka
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Dora Hermes
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Kai J Miller
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, USA
| | - Boris D Heifets
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Cara Bohon
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Jennifer A McNab
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Casey H Halpern
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Surgery, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA.
| |
Collapse
|
26
|
Noguchi H, Arela JC, Ngo TT, Cocas L, Pleasure SJ. Shh from mossy cells contributes to preventing NSC pool depletion after seizure-induced neurogenesis and in aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.21.554173. [PMID: 37662214 PMCID: PMC10473584 DOI: 10.1101/2023.08.21.554173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Epileptic seizures induce aberrant neurogenesis from resident neural stem cells (NSCs) in the dentate gyrus of the adult mouse hippocampus, which has been implicated in depletion of the NSC pool and impairment of hippocampal function. However, the mechanisms regulating neurogenesis after seizures remain unknown. Here we demonstrate that Shh from mossy cells is a major source of Shh signaling activity after seizures, by which mossy cells contribute to seizure-induced neurogenesis and maintenance of the NSC pool. Deletion of Shh from mossy cells attenuates seizure-induced neurogenesis. Moreover, in the absence of Shh from mossy cells, NSCs pool are prematurely depleted after seizure-induced proliferation, and NSCs have impaired self-renewal. Likewise, lack of Shh from mossy cells accelerates age-related decline of the NSC pool with accompanying reduction of self-renewal of NSCs outside the context of pathology such as seizures. Together, our findings indicate that Shh from mossy cells is critical to maintain NSCs and to prevent exhaustion from excessive consumption in aging and after seizures.
Collapse
|
27
|
Han Y, He Y, Harris L, Xu Y, Wu Q. Identification of a GABAergic neural circuit governing leptin signaling deficiency-induced obesity. eLife 2023; 12:e82649. [PMID: 37043384 PMCID: PMC10097419 DOI: 10.7554/elife.82649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 03/24/2023] [Indexed: 04/13/2023] Open
Abstract
The hormone leptin is known to robustly suppress food intake by acting upon the leptin receptor (LepR) signaling system residing within the agouti-related protein (AgRP) neurons of the hypothalamus. However, clinical studies indicate that leptin is undesirable as a therapeutic regiment for obesity, which is at least partly attributed to the poorly understood complex secondary structure and key signaling mechanism of the leptin-responsive neural circuit. Here, we show that the LepR-expressing portal neurons send GABAergic projections to a cohort of α3-GABAA receptor expressing neurons within the dorsomedial hypothalamic nucleus (DMH) for the control of leptin-mediated obesity phenotype. We identified the DMH as a key brain region that contributes to the regulation of leptin-mediated feeding. Acute activation of the GABAergic AgRP-DMH circuit promoted food intake and glucose intolerance, while activation of post-synaptic MC4R neurons in the DMH elicited exactly opposite phenotypes. Rapid deletion of LepR from AgRP neurons caused an obesity phenotype which can be rescued by blockage of GABAA receptor in the DMH. Consistent with behavioral results, these DMH neurons displayed suppressed neural activities in response to hunger or hyperglycemia. Furthermore, we identified that α3-GABAA receptor signaling within the DMH exerts potent bi-directional regulation of the central effects of leptin on feeding and body weight. Together, our results demonstrate a novel GABAergic neural circuit governing leptin-mediated feeding and energy balance via a unique α3-GABAA signaling within the secondary leptin-responsive neural circuit, constituting a new avenue for therapeutic interventions in the treatment of obesity and associated comorbidities.
Collapse
Affiliation(s)
- Yong Han
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of MedicineHoustonUnited States
| | - Yang He
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of MedicineHoustonUnited States
| | - Lauren Harris
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of MedicineHoustonUnited States
| | - Yong Xu
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of MedicineHoustonUnited States
| | - Qi Wu
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
28
|
Farhadi DS, Estofan L, Privitera M. Anorexia and the hippocampus: A case report. Epilepsy Behav Rep 2022; 21:100577. [PMID: 36590373 PMCID: PMC9800335 DOI: 10.1016/j.ebr.2022.100577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/30/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
Eating disorders have been shown to be associated with epilepsy, typically associated with the temporal lobe and usually of non-dominant hemisphere origin. We report the case of a 56-year-old woman with drug resistant epilepsy, localized to the dominant left hippocampus. She experienced an increasing frequency of seizures over a two-year period associated with loss of appetite and substantial weight loss independent of antiseizure medication changes. Extensive workup eliminated gastrointestinal and paraneoplastic etiologies. There was no history of psychiatric illness, including anorexia nervosa. Pre-surgical workup showed mesial temporal sclerosis on MRI and video-EEG was consistent with ipsilateral medial temporal seizure onset. The patient underwent laser interstitial ablation of the left amygdala and hippocampus, which resulted in a cessation of seizures. Within 24 h of the laser ablation, her appetite returned to normal and, within 8 months she regained 26 lbs. To our knowledge, this is the first case report of a patient with dominant temporal lobe epilepsy with anorexia that was temporally associated with escalating seizure frequency and stopped with treatment and cessation of seizures, suggesting a causal and pathogenic relationship.
Collapse
Affiliation(s)
- Dara S. Farhadi
- University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
| | - Leonel Estofan
- Department of Neurology, University of Cincinnati, Gardner Neuroscience Institute, United States
| | - Michael Privitera
- Department of Neurology, University of Cincinnati, Gardner Neuroscience Institute, United States,Corresponding author at: 260 Stetson St, Suite 2300, Cincinnati, OH 45267-0525, United States.
| |
Collapse
|
29
|
Steiner A, Owen BM, Bauer JP, Seanez L, Kwon S, Biddinger JE, Huffman R, Ayala JE, Nobis WP, Lewis AS. Glucagon-like peptide-1 receptor differentially controls mossy cell activity across the dentate gyrus longitudinal axis. Hippocampus 2022; 32:797-807. [PMID: 36063105 PMCID: PMC9675713 DOI: 10.1002/hipo.23469] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/27/2022] [Accepted: 08/18/2022] [Indexed: 01/07/2023]
Abstract
Understanding the role of dentate gyrus (DG) mossy cells (MCs) in learning and memory has rapidly evolved due to increasingly precise methods for targeting MCs and for in vivo recording and activity manipulation in rodents. These studies have shown MCs are highly active in vivo, strongly remap to contextual manipulation, and that their inhibition or hyperactivation impairs pattern separation and location or context discrimination. Less well understood is how MC activity is modulated by neurohormonal mechanisms, which might differentially control the participation of MCs in cognitive functions during discrete states, such as hunger or satiety. In this study, we demonstrate that glucagon-like peptide-1 (GLP-1), a neuropeptide produced in the gut and the brain that regulates food consumption and hippocampal-dependent mnemonic function, might regulate MC function through expression of its receptor, GLP-1R. RNA-seq demonstrated that most, though not all, Glp1r in hippocampal principal neurons is expressed in MCs, and in situ hybridization revealed strong expression of Glp1r in hilar neurons. Glp1r-ires-Cre mice crossed with Ai14D reporter mice followed by co-labeling for the MC marker GluR2/3 revealed that almost all MCs in the ventral DG expressed Glp1r and that almost all Glp1r-expressing hilar neurons were MCs. However, only ~60% of dorsal DG MCs expressed Glp1r, and Glp1r was also expressed in small hilar neurons that were not MCs. Consistent with this expression pattern, peripheral administration of the GLP-1R agonist exendin-4 (5 μg/kg) increased cFos expression in ventral but not dorsal DG hilar neurons. Finally, whole-cell patch-clamp recordings from ventral MCs showed that bath application of exendin-4 (200 nM) depolarized MCs and increased action potential firing. Taken together, this study adds to known MC activity modulators a neurohormonal mechanism that may preferentially affect ventral DG physiology and may potentially be targetable by several GLP-1R pharmacotherapies already in clinical use.
Collapse
Affiliation(s)
- Alex Steiner
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Benjamin M. Owen
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James P. Bauer
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Leann Seanez
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sam Kwon
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jessica E. Biddinger
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ragan Huffman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Julio E. Ayala
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - William P. Nobis
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alan S. Lewis
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
30
|
Abstract
The nucleus accumbens (NAc) is a canonical reward center that regulates feeding and drinking but it is not known whether these behaviors are mediated by same or different neurons. We employed two-photon calcium imaging in awake, behaving mice and found that during the appetitive phase, both hunger and thirst are sensed by a nearly identical population of individual D1 and D2 neurons in the NAc that respond monophasically to food cues in fasted animals and water cues in dehydrated animals. During the consummatory phase, we identified three distinct neuronal clusters that are temporally correlated with action initiation, consumption, and cessation shared by feeding and drinking. These dynamic clusters also show a nearly complete overlap of individual D1 neurons and extensive overlap among D2 neurons. Modulating D1 and D2 neural activities revealed analogous effects on feeding versus drinking behaviors. In aggregate, these data show that a highly overlapping set of D1 and D2 neurons in NAc detect food and water reward and elicit concordant responses to hunger and thirst. These studies establish a general role of this mesolimbic pathway in mediating instinctive behaviors by controlling motivation-associated variables rather than conferring behavioral specificity.
Collapse
|
31
|
Parent MB. Using Postmeal Measures and Manipulations to Investigate Hippocampal Mnemonic Control of Eating Behavior. Neuroscience 2022; 497:228-238. [PMID: 34998891 PMCID: PMC9256844 DOI: 10.1016/j.neuroscience.2021.12.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/27/2021] [Accepted: 12/30/2021] [Indexed: 10/19/2022]
Abstract
Episodic meal-related memories provide the brain with a powerful mechanism for tracking and controlling eating behavior because they contain a detailed record of recent energy intake that likely outlasts the physiological signals generated by feeding bouts. This review briefly summarizes evidence from human participants showing that episodic meal-related memory limits later eating behavior and then describes our research aimed at investigating whether hippocampal neurons mediate the inhibitory effects of meal-related memory on subsequent feeding. Our approach has been inspired by pioneering work conducted by Ivan Izquierdo and others who used posttraining manipulations to investigate memory consolidation. This review describes the rationale and value of posttraining manipulations, how Izquierdo used them to demonstrate that dorsal hippocampal (dHC) neurons are critical for memory consolidation, and how we have adapted this strategy to investigate whether dHC neurons are necessary for mnemonic control of energy intake. I describe our evidence showing that ingestion activates the molecular processes necessary for synaptic plasticity and memory during the early postprandial period, when the memory of the meal would be undergoing consolidation, and then summarize our findings showing that neural activity in dHC neurons is critical during the early postprandial period for limiting future intake. Collectively, our evidence supports the hypothesis that dHC neurons mediate the inhibitory effects of ingestion-related memory on future intake and demonstrates that post-experience memory modulation is not confined to artificial laboratory memory tasks.
Collapse
Affiliation(s)
- M B Parent
- Neuroscience Institute & Department of Psychology, Georgia State University, PO Box 5030, Atlanta, GA 30303, USA.
| |
Collapse
|
32
|
Bhatti DL, Medrihan L, Chen MX, Jin J, McCabe KA, Wang W, Azevedo EP, Ledo JH, Kim Y. Molecular and Cellular Adaptations in Hippocampal Parvalbumin Neurons Mediate Behavioral Responses to Chronic Social Stress. Front Mol Neurosci 2022; 15:898851. [PMID: 35813065 PMCID: PMC9268684 DOI: 10.3389/fnmol.2022.898851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Parvalbumin-expressing interneurons (PV neurons) maintain inhibitory control of local circuits implicated in behavioral responses to environmental stressors. However, the roles of molecular and cellular adaptations in PV neurons in stress susceptibility or resilience have not been clearly established. Here, we show behavioral outcomes of chronic social defeat stress (CSDS) are mediated by differential neuronal activity and gene expression in hippocampal PV neurons in mice. Using in vivo electrophysiology and chemogenetics, we find increased PV neuronal activity in the ventral dentate gyrus is required and sufficient for behavioral susceptibility to CSDS. PV neuron-selective translational profiling indicates mitochondrial oxidative phosphorylation is the most significantly altered pathway in stress-susceptible versus resilient mice. Among differentially expressed genes associated with stress-susceptibility and resilience, we find Ahnak, an endogenous regulator of L-type calcium channels which are implicated in the regulation of mitochondrial function and gene expression. Notably, Ahnak deletion in PV neurons impedes behavioral susceptibility to CSDS. Altogether, these findings indicate behavioral effects of chronic stress can be controlled by selective modulation of PV neuronal activity or a regulator of L-type calcium signaling in PV neurons.
Collapse
Affiliation(s)
- Dionnet L. Bhatti
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
- Program in Neuroscience, Harvard Medical School, Boston, MA, United States
| | - Lucian Medrihan
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Michelle X. Chen
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Junghee Jin
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Kathryn A. McCabe
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Wei Wang
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
- Bioinformatics Resource Center, The Rockefeller University, New York, NY, United States
| | - Estefania P. Azevedo
- Laboratory of Molecular Genetics, The Rockefeller University, New York, NY, United States
| | - Jose H. Ledo
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Yong Kim
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States
- Brain Health Institute, Rutgers University, Piscataway, NJ, United States
- *Correspondence: Yong Kim,
| |
Collapse
|
33
|
Sagarkar S, Bhat N, Sapre M, Dudhabhate B, Kokare DM, Subhedar NK, Sakharkar AJ. TET1-induced DNA demethylation in dentate gyrus is important for reward conditioning and reinforcement. Mol Neurobiol 2022; 59:5426-5442. [PMID: 35705787 DOI: 10.1007/s12035-022-02917-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 06/05/2022] [Indexed: 10/18/2022]
Abstract
Neuroadaptations in neurocircuitry of reward memories govern the persistent and compulsive behaviors. The study of the role of hippocampus in processing of reward memory and its retrieval is critical to our understanding of addiction and relapse. The aim of this study is to probe the epigenetic mechanisms underlying reward memory in the frame of dentate gyrus (DG). To that end, the rats conditioned to the food baited arm of a Y-maze and subjected to memory probe trial. The hippocampus of conditioned rats displayed higher mRNA levels of Ten-eleven translocase 1 (Tet1) and brain-derived neurotrophic factor (Bdnf) after memory probe trial. The DNA hydroxymethylation and TET1 occupancy at the Bdnf promoters showed concomitant increase. Stereotactic administration of Tet1 siRNA in the DG before and after conditioning inhibited reward memory formation and recall, respectively. Administration of Tet1 siRNA impaired the reward memory recall that was reinstated following administration of exogenous BDNF peptide or after wash-off period of 8 days. Infusion of a MEK/ERK inhibitor, U0126 in the DG inhibited reward memory retrieval. The TET1-induced DNA demethylation at the Bdnf promoters raised BDNF levels in the hippocampus, thereby setting the stage for reward memory retrieval. The study underscores the causative role of TET1 in the DG for reward memory formation and recall.
Collapse
Affiliation(s)
- Sneha Sagarkar
- Department of Zoology, Savitribai Phule Pune University, Pune, 411 007, India.
| | - Nagashree Bhat
- Department of Zoology, Savitribai Phule Pune University, Pune, 411 007, India
| | - Madhura Sapre
- Department of Zoology, Savitribai Phule Pune University, Pune, 411 007, India
| | - Biru Dudhabhate
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, 440 033, India
| | - Dadasaheb M Kokare
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, 440 033, India
| | - Nishikant K Subhedar
- Indian Institute of Science Education and Research (IISER), Pune, 411 008, India
| | - Amul J Sakharkar
- Department of Biotechnology, Savitribai Phule Pune University, Pune, 411 007, India.
| |
Collapse
|
34
|
Azevedo EP, Ivan VJ, Friedman JM, Stern SA. Higher-Order Inputs Involved in Appetite Control. Biol Psychiatry 2022; 91:869-878. [PMID: 34593204 PMCID: PMC9704062 DOI: 10.1016/j.biopsych.2021.07.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/15/2021] [Accepted: 07/17/2021] [Indexed: 01/01/2023]
Abstract
The understanding of the neural control of appetite sheds light on the pathogenesis of eating disorders such as anorexia nervosa and obesity. Both diseases are a result of maladaptive eating behaviors (overeating or undereating) and are associated with life-threatening health problems. The fine regulation of appetite involves genetic, physiological, and environmental factors, which are detected and integrated in the brain by specific neuronal populations. For centuries, the hypothalamus has been the center of attention in the scientific community as a key regulator of appetite. The hypothalamus receives and sends axonal projections to several other brain regions that are important for the integration of sensory and emotional information. These connections ensure that appropriate behavioral decisions are made depending on the individual's emotional state and environment. Thus, the mechanisms by which higher-order brain regions integrate exteroceptive information to coordinate feeding is of great importance. In this review, we will focus on the functional and anatomical projections connecting the hypothalamus to the limbic system and higher-order brain centers in the cortex. We will also address the mechanisms by which specific neuronal populations located in higher-order centers regulate appetite and how maladaptive eating behaviors might arise from altered connections among cortical and subcortical areas with the hypothalamus.
Collapse
Affiliation(s)
- Estefania P Azevedo
- Laboratory of Molecular Genetics, The Rockefeller University, New York, New York.
| | - Violet J Ivan
- Laboratory of Molecular Genetics, The Rockefeller University, New York, New York
| | - Jeffrey M Friedman
- Laboratory of Molecular Genetics, The Rockefeller University, New York, New York; Howard Hughes Medical Institute, New York, New York
| | - Sarah A Stern
- Integrative Neural Circuits and Behavior Research Group, Max Planck Florida Institute for Neuroscience, Jupiter, Florida.
| |
Collapse
|
35
|
Haddad-Tóvolli R, Ramírez S, Muñoz-Moreno E, Milà-Guasch M, Miquel-Rio L, Pozo M, Chivite I, Altirriba J, Obri A, Gómez-Valadés AG, Toledo M, Eyre E, Bortolozzi A, Valjent E, Soria G, Claret M. Food craving-like episodes during pregnancy are mediated by accumbal dopaminergic circuits. Nat Metab 2022; 4:424-434. [PMID: 35379970 DOI: 10.1038/s42255-022-00557-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 02/22/2022] [Indexed: 12/22/2022]
Abstract
Preparation for motherhood requires a myriad of physiological and behavioural adjustments throughout gestation to provide an adequate environment for proper embryonic development1. Cravings for highly palatable foods are highly prevalent during pregnancy2 and contribute to the maintenance and development of gestational overweight or obesity3. However, the neurobiology underlying the distinct ingestive behaviours that result from craving specific foods remain unknown. Here we show that mice, similarly to humans, experience gestational food craving-like episodes. These episodes are associated with a brain connectivity reorganization that affects key components of the dopaminergic mesolimbic circuitry, which drives motivated appetitive behaviours and facilitates the perception of rewarding stimuli. Pregnancy engages a dynamic modulation of dopaminergic signalling through neurons expressing dopamine D2 receptors in the nucleus accumbens, which directly modulate food craving-like events. Importantly, persistent maternal food craving-like behaviour has long-lasting effects on the offspring, particularly in males, leading to glucose intolerance, increased body weight and increased susceptibility to develop eating disorders and anxiety-like behaviours during adulthood. Our results reveal the cognitively motivated nature of pregnancy food cravings and advocates for moderating emotional eating during gestation to prevent deterioration of the offspring's neuropsychological and metabolic health.
Collapse
Affiliation(s)
- Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Sara Ramírez
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Emma Muñoz-Moreno
- Experimental 7T MRI Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria Milà-Guasch
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lluis Miquel-Rio
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain
| | - Macarena Pozo
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Iñigo Chivite
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jordi Altirriba
- Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Arnaud Obri
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Alicia G Gómez-Valadés
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Miriam Toledo
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Elena Eyre
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Analia Bortolozzi
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain
| | | | - Guadalupe Soria
- Experimental 7T MRI Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Laboratory of Surgical Neuroanatomy, Faculty of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- CIBER de Bioingeniera, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
- School of Medicine, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
36
|
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: 69] [Impact Index Per Article: 23.0] [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
|
37
|
GoodSmith D, Kim SH, Puliyadi V, Ming GL, Song H, Knierim JJ, Christian KM. Flexible encoding of objects and space in single cells of the dentate gyrus. Curr Biol 2022; 32:1088-1101.e5. [PMID: 35108522 PMCID: PMC8930604 DOI: 10.1016/j.cub.2022.01.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/12/2021] [Accepted: 01/10/2022] [Indexed: 01/05/2023]
Abstract
The hippocampus is involved in the formation of memories that require associations among stimuli to construct representations of space and the items and events within that space. Neurons in the dentate gyrus (DG), an initial input region of the hippocampus, have robust spatial tuning, but it is unclear how nonspatial information may be integrated with spatial activity in this region. We recorded from the DG of 21 adult mice as they foraged for food in an environment that contained discrete objects. We found DG cells with multiple firing fields at a fixed distance and direction from objects (landmark vector cells) and cells that exhibited localized changes in spatial firing when objects in the environment were manipulated. By classifying recorded DG cells into putative dentate granule cells and mossy cells, we examined how the addition or displacement of objects affected the spatial firing of these DG cell types. Object-related activity was detected in a significant proportion of mossy cells. Although few granule cells with responses to object manipulations were recorded, likely because of the sparse nature of granule cell firing, there was generally no significant difference in the proportion of granule cells and mossy cells with object responses. When mice explored a second environment with the same objects, DG spatial maps completely reorganized, and a different subset of cells responded to object manipulations. Together, these data reveal the capacity of DG cells to detect small changes in the environment while preserving a stable spatial representation of the overall context.
Collapse
Affiliation(s)
- Douglas GoodSmith
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA; Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA; Department of Neurobiology and Neuroscience Institute, University of Chicago, 5801 S Ellis Avenue, Chicago, IL 60637, USA
| | - Sang Hoon Kim
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Vyash Puliyadi
- Department of Psychological and Brain Sciences, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA; Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA; The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| | - James J Knierim
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA.
| | - Kimberly M Christian
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| |
Collapse
|
38
|
Cong S, Yao X, Xie L, Yan J, Shen L, and the Alzheimer’s Disease Neuroimaging Initiative. Genetic Influence Underlying Brain Connectivity Phenotype: A Study on Two Age-Specific Cohorts. Front Genet 2022; 12:782953. [PMID: 35237294 PMCID: PMC8884108 DOI: 10.3389/fgene.2021.782953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/16/2021] [Indexed: 11/29/2022] Open
Abstract
Background: Human brain structural connectivity is an important imaging quantitative trait for brain development and aging. Mapping the network connectivity to the phenotypic variation provides fundamental insights in understanding the relationship between detailed brain topological architecture, function, and dysfunction. However, the underlying neurobiological mechanism from gene to brain connectome, and to phenotypic outcomes, and whether this mechanism changes over time, remain unclear. Methods: This study analyzes diffusion-weighted imaging data from two age-specific neuroimaging cohorts, extracts structural connectome topological network measures, performs genome-wide association studies of the measures, and examines the causality of genetic influences on phenotypic outcomes mediated via connectivity measures. Results: Our empirical study has yielded several significant findings: 1) It identified genetic makeup underlying structural connectivity changes in the human brain connectome for both age groups. Specifically, it revealed a novel association between the minor allele (G) of rs7937515 and the decreased network segregation measures of the left middle temporal gyrus across young and elderly adults, indicating a consistent genetic effect on brain connectivity across the lifespan. 2) It revealed rs7937515 as a genetic marker for body mass index in young adults but not in elderly adults. 3) It discovered brain network segregation alterations as a potential neuroimaging biomarker for obesity. 4) It demonstrated the hemispheric asymmetry of structural network organization in genetic association analyses and outcome-relevant studies. Discussion: These imaging genetic findings underlying brain connectome warrant further investigation for exploring their potential influences on brain-related complex diseases, given the significant involvement of altered connectivity in neurological, psychiatric and physical disorders.
Collapse
Affiliation(s)
- Shan Cong
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Xiaohui Yao
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Linhui Xie
- Department of Electrical and Computer Engineering, School of Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, United States
| | - Jingwen Yan
- Department of BioHealth Informatics, School of Informatics and Computing, Indiana University Purdue University Indianapolis, Indianapolis, IN, United States
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | |
Collapse
|
39
|
Parent MB, Higgs S, Cheke LG, Kanoski SE. Memory and eating: A bidirectional relationship implicated in obesity. Neurosci Biobehav Rev 2022; 132:110-129. [PMID: 34813827 PMCID: PMC8816841 DOI: 10.1016/j.neubiorev.2021.10.051] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 10/17/2021] [Accepted: 10/28/2021] [Indexed: 01/03/2023]
Abstract
This paper reviews evidence demonstrating a bidirectional relationship between memory and eating in humans and rodents. In humans, amnesia is associated with impaired processing of hunger and satiety cues, disrupted memory of recent meals, and overconsumption. In healthy participants, meal-related memory limits subsequent ingestive behavior and obesity is associated with impaired memory and disturbances in the hippocampus. Evidence from rodents suggests that dorsal hippocampal neural activity contributes to the ability of meal-related memory to control future intake, that endocrine and neuropeptide systems act in the ventral hippocampus to provide cues regarding energy status and regulate learned aspects of eating, and that consumption of hypercaloric diets and obesity disrupt these processes. Collectively, this evidence indicates that diet-induced obesity may be caused and/or maintained, at least in part, by a vicious cycle wherein excess intake disrupts hippocampal functioning, which further increases intake. This perspective may advance our understanding of how the brain controls eating, the neural mechanisms that contribute to eating-related disorders, and identify how to treat diet-induced obesity.
Collapse
Affiliation(s)
- Marise B Parent
- Neuroscience Institute & Department of Psychology, Georgia State University, Box 5030, Atlanta, GA 30303-5030, United States.
| | - Suzanne Higgs
- School of Psychology, University of Birmingham, Edgbaston, Birmingham, BI5 2TT, United Kingdom.
| | - Lucy G Cheke
- Department of Psychology, University of Cambridge, Downing Street, Cambridge, CB2 3EB, United Kingdom.
| | - Scott E Kanoski
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, 90089-0371, United States.
| |
Collapse
|
40
|
Putting Together Pieces of the Lateral Septum: Multifaceted Functions and Its Neural Pathways. eNeuro 2021; 8:ENEURO.0315-21.2021. [PMID: 34764187 PMCID: PMC8647703 DOI: 10.1523/eneuro.0315-21.2021] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 01/01/2023] Open
Abstract
The lateral septum (LS) is implicated as a hub that regulates a variety of affects, such as reward, feeding, anxiety, fear, sociability, and memory. However, it remains unclear how the LS, previously treated as a structure of homogeneity, exhibits such multifaceted functions. Emerging evidence suggests that different functions of the LS are mediated largely by its diverse input and output connections. It has also become clear that the LS is a heterogeneous region, where its dorsal and ventral poles play dissociable and often opposing roles. This functional heterogeneity can often be explained by distinct dorsal and ventral hippocampal inputs along the LS dorsoventral axis, as well as antagonizing connections between LS subregions. Similarly, outputs from LS subregions to respective downstream targets, such as hypothalamic, preoptic, and tegmental areas, also account for this functional heterogeneity. In this review, we provide an updated perspective on LS subregion classification, connectivity, and functions. We also identify key questions that have yet to be addressed in the field.
Collapse
|
41
|
Wang KY, Wu JW, Cheng JK, Chen CC, Wong WY, Averkin RG, Tamás G, Nakazawa K, Lien CC. Elevation of hilar mossy cell activity suppresses hippocampal excitability and avoidance behavior. Cell Rep 2021; 36:109702. [PMID: 34525354 DOI: 10.1016/j.celrep.2021.109702] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/09/2021] [Accepted: 08/20/2021] [Indexed: 12/30/2022] Open
Abstract
Modulation of hippocampal dentate gyrus (DG) excitability regulates anxiety. In the DG, glutamatergic mossy cells (MCs) receive the excitatory drive from principal granule cells (GCs) and mediate the feedback excitation and inhibition of GCs. However, the circuit mechanism by which MCs regulate anxiety-related information routing through hippocampal circuits remains unclear. Moreover, the correlation between MC activity and anxiety states is unclear. In this study, we first demonstrate, by means of calcium fiber photometry, that MC activity in the ventral hippocampus (vHPC) of mice increases while they explore anxiogenic environments. Next, juxtacellular recordings reveal that optogenetic activation of MCs preferentially recruits GABAergic neurons, thereby suppressing GCs and ventral CA1 neurons. Finally, chemogenetic excitation of MCs in the vHPC reduces avoidance behaviors in both healthy and anxious mice. These results not only indicate an anxiolytic role of MCs but also suggest that MCs may be a potential therapeutic target for anxiety disorders.
Collapse
Affiliation(s)
- Kai-Yi Wang
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Jei-Wei Wu
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Jen-Kun Cheng
- Department of Medicine, Mackay Medical College, New Taipei 252, Taiwan; Department of Anesthesiology, Mackay Memorial Hospital, Taipei 104, Taiwan
| | | | - Wai-Yi Wong
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Robert G Averkin
- ELKH-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy and Neuroscience, University of Szeged, Közép fasor 52, Szeged 6726, Hungary
| | - Gábor Tamás
- ELKH-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy and Neuroscience, University of Szeged, Közép fasor 52, Szeged 6726, Hungary
| | - Kazu Nakazawa
- Department of Neuroscience, Southern Research, Birmingham, AL 35205, USA; Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Cheng-Chang Lien
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; Brain Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan.
| |
Collapse
|
42
|
Ma Y, Bayguinov PO, McMahon SM, Scharfman HE, Jackson MB. Direct synaptic excitation between hilar mossy cells revealed with a targeted voltage sensor. Hippocampus 2021; 31:1215-1232. [PMID: 34478219 DOI: 10.1002/hipo.23386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/09/2021] [Accepted: 08/21/2021] [Indexed: 12/18/2022]
Abstract
The dentate gyrus not only gates the flow of information into the hippocampus, it also integrates and processes this information. Mossy cells (MCs) are a major type of excitatory neuron strategically located in the hilus of the dentate gyrus where they can contribute to this processing through networks of synapses with inhibitory neurons and dentate granule cells. Some prior work has suggested that MCs can form excitatory synapses with other MCs, but the role of these synapses in the network activity of the dentate gyrus has received little attention. Here, we investigated synaptic inputs to MCs in mouse hippocampal slices using a genetically encoded hybrid voltage sensor (hVOS) targeted to MCs by Cre-lox technology. This enabled optical recording of voltage changes from multiple MCs simultaneously. Stimulating granule cells and CA3 pyramidal cells activated well-established inputs to MCs and elicited synaptic responses as expected. However, the weak blockade of MC responses to granule cell layer stimulation by DCG-IV raised the possibility of another source of excitation. To evaluate synapses between MCs as this source, single MCs were stimulated focally. Stimulation of one MC above its action potential threshold evoked depolarizing responses in neighboring MCs that depended on glutamate receptors. Short latency responses of MCs to other MCs did not depend on release from granule cell axons. However, granule cells did contribute to the longer latency responses of MCs to stimulation of other MCs. Thus, MCs transmit their activity to other MCs both through direct synaptic coupling and through polysynaptic coupling with dentate granule cells. MC-MC synapses can redistribute information entering the dentate gyrus and thus shape and modulate the electrical activity underlying hippocampal functions such as navigation and memory, as well as excessive excitation during seizures.
Collapse
Affiliation(s)
- Yihe Ma
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Peter O Bayguinov
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Shane M McMahon
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Helen E Scharfman
- New York University Langone Health and the Nathan Kline Institute for Psychiatric Research, Orangeburg, New Jersey, USA
| | - Meyer B Jackson
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
43
|
Dopamine signaling regulates hematopoietic stem and progenitor cell function. Blood 2021; 138:2051-2065. [PMID: 34370827 DOI: 10.1182/blood.2020010419] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 07/25/2021] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic stem and progenitor cell (HSPC) function in bone marrow (BM) is controlled by stroma-derived signals, but the identity and interplay of these signals remain incompletely understood. Here, we show that sympathetic nerve-derived dopamine directly controls HSPC behavior through D2-subfamily dopamine receptors. Blockade of dopamine synthesis as well as pharmacological or genetic inactivation of D2-subfamily dopamine receptors lead to reduced HSPC frequency, inhibition of proliferation and low BM transplantation efficiency. Conversely, treatment with a D2-type receptor agonist increases BM regeneration and transplantation efficiency. Mechanistically, dopamine controls expression of the kinase Lck, which, in turn, regulates mitogen-activated protein kinase-mediated signaling triggered by stem cell factor in HSPCs. Our work reveals critical functional roles of dopamine in HSPCs, which may open up new therapeutic options for improved BM transplantation and other conditions requiring the rapid expansion of HSPCs.
Collapse
|
44
|
A neural circuit for excessive feeding driven by environmental context in mice. Nat Neurosci 2021; 24:1132-1141. [PMID: 34168339 DOI: 10.1038/s41593-021-00875-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/13/2021] [Indexed: 02/05/2023]
Abstract
Despite notable genetic influences, obesity mainly results from the overconsumption of food, which arises from the interplay of physiological, cognitive and environmental factors. In patients with obesity, eating is determined more by external cues than by internal physiological needs. However, how environmental context drives non-homeostatic feeding is elusive. Here, we identify a population of somatostatin (TNSST) neurons in the mouse hypothalamic tuberal nucleus that are preferentially activated by palatable food. Activation of TNSST neurons enabled a context to drive non-homeostatic feeding in sated mice and required inputs from the subiculum. Pairing a context with palatable food greatly potentiated synaptic transmission between the subiculum and TNSST neurons and drove non-homeostatic feeding that could be selectively suppressed by inhibiting TNSST neurons or the subiculum but not other major orexigenic neurons. These results reveal how palatable food, through a specific hypothalamic circuit, empowers environmental context to drive non-homeostatic feeding.
Collapse
|
45
|
Abstract
An organism's survival can depend on its ability to recall and navigate to spatial locations associated with rewards, such as food or a home. Accumulating research has revealed that computations of reward and its prediction occur on multiple levels across a complex set of interacting brain regions, including those that support memory and navigation. However, how the brain coordinates the encoding, recall and use of reward information to guide navigation remains incompletely understood. In this Review, we propose that the brain's classical navigation centres - the hippocampus and the entorhinal cortex - are ideally suited to coordinate this larger network by representing both physical and mental space as a series of states. These states may be linked to reward via neuromodulatory inputs to the hippocampus-entorhinal cortex system. Hippocampal outputs can then broadcast sequences of states to the rest of the brain to store reward associations or to facilitate decision-making, potentially engaging additional value signals downstream. This proposal is supported by recent advances in both experimental and theoretical neuroscience. By discussing the neural systems traditionally tied to navigation and reward at their intersection, we aim to offer an integrated framework for understanding navigation to reward as a fundamental feature of many cognitive processes.
Collapse
|
46
|
Abstract
The gut microbiota has the capacity to affect host appetite via intestinal satiety pathways, as well as complex feeding behaviors. In this Review, we highlight recent evidence that the gut microbiota can modulate food preference across model organisms. We discuss effects of the gut microbiota on the vagus nerve and brain regions including the hypothalamus, mesolimbic system, and prefrontal cortex, which play key roles in regulating feeding behavior. Crosstalk between commensal bacteria and the central and peripheral nervous systems is associated with alterations in signaling of neurotransmitters and neuropeptides such as dopamine, brain-derived neurotrophic factor (BDNF), and glucagon-like peptide-1 (GLP-1). We further consider areas for future research on mechanisms by which gut microbes may influence feeding behavior involving these neural pathways. Understanding roles for the gut microbiota in feeding regulation will be important for informing therapeutic strategies to treat metabolic and eating disorders.
Collapse
|
47
|
Botterill JJ, Gerencer KJ, Vinod KY, Alcantara‐Gonzalez D, Scharfman HE. Dorsal and ventral mossy cells differ in their axonal projections throughout the dentate gyrus of the mouse hippocampus. Hippocampus 2021; 31:522-539. [PMID: 33600026 PMCID: PMC8247909 DOI: 10.1002/hipo.23314] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 01/24/2021] [Accepted: 01/30/2021] [Indexed: 11/14/2022]
Abstract
Glutamatergic hilar mossy cells (MCs) have axons that terminate both near and far from their cell body but stay within the DG, making synapses primarily in the molecular layer. The long-range axons are considered the primary projection, and extend throughout the DG ipsilateral to the soma, and project to the contralateral DG. The specificity of MC axons for the inner molecular layer (IML) has been considered to be a key characteristic of the DG. In the present study, we made the surprising finding that dorsal MC axons are an exception to this rule. We used two mouse lines that allow for Cre-dependent viral labeling of MCs and their axons: dopamine receptor D2 (Drd2-Cre) and calcitonin receptor-like receptor (Crlr-Cre). A single viral injection into the dorsal DG to label dorsal MCs resulted in labeling of MC axons in both the IML and middle molecular layer (MML). Interestingly, this broad termination of dorsal MC axons occurred throughout the septotemporal DG. In contrast, long-range axons of ventral MCs terminated in the IML, consistent with the literature. Taken together, these results suggest that dorsal and ventral MCs differ significantly in their axonal projections. Since MC projections in the ML are thought to terminate primarily on GCs, the results suggest a dorsal-ventral difference in MC activation of GCs. The surprising difference in dorsal and ventral MC projections should therefore be considered when evaluating dorsal-ventral differences in DG function.
Collapse
Affiliation(s)
- Justin J. Botterill
- Center for Dementia ResearchThe Nathan Kline Institute for Psychiatric ResearchOrangeburgNew YorkUSA
| | - Kathleen J. Gerencer
- Center for Dementia ResearchThe Nathan Kline Institute for Psychiatric ResearchOrangeburgNew YorkUSA
| | - K. Yaragudri Vinod
- Department of Analytical PsychopharmacologyThe Nathan Kline Institute for Psychiatric ResearchOrangeburgNew YorkUSA
- Emotional Brain InstituteThe Nathan Kline Institute for Psychiatric ResearchOrangeburgNew YorkUSA
- Department of Child & Adolescent Psychiatry, Neuroscience & Physiology and Psychiatry and the New York University Neuroscience InstituteNew York University Langone HealthNew YorkNew YorkUSA
| | - David Alcantara‐Gonzalez
- Center for Dementia ResearchThe Nathan Kline Institute for Psychiatric ResearchOrangeburgNew YorkUSA
| | - Helen E. Scharfman
- Center for Dementia ResearchThe Nathan Kline Institute for Psychiatric ResearchOrangeburgNew YorkUSA
- Department of Child & Adolescent Psychiatry, Neuroscience & Physiology and Psychiatry and the New York University Neuroscience InstituteNew York University Langone HealthNew YorkNew YorkUSA
| |
Collapse
|
48
|
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: 11.8] [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
|
49
|
Bidirectional Regulation of Cognitive and Anxiety-like Behaviors by Dentate Gyrus Mossy Cells in Male and Female Mice. J Neurosci 2021; 41:2475-2495. [PMID: 33472828 DOI: 10.1523/jneurosci.1724-20.2021] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 12/04/2020] [Accepted: 01/04/2021] [Indexed: 02/08/2023] Open
Abstract
The dentate gyrus (DG) of the hippocampus is important for cognition and behavior. However, the circuits underlying these functions are unclear. DG mossy cells (MCs) are potentially important because of their excitatory synapses on the primary cell type, granule cells (GCs). However, MCs also activate GABAergic neurons, which inhibit GCs. We used viral delivery of designer receptors exclusively activated by designer drugs (DREADDs) in mice to implement a gain- and loss-of-function study of MCs in diverse behaviors. Using this approach, manipulations of MCs could bidirectionally regulate behavior. The results suggest that inhibiting MCs can reduce anxiety-like behavior and improve cognitive performance. However, not all cognitive or anxiety-related behaviors were influenced, suggesting specific roles of MCs in some, but not all, types of cognition and anxiety. Notably, several behaviors showed sex-specific effects, with females often showing more pronounced effects than the males. We also used the immediate early gene c-Fos to address whether DREADDs bidirectionally regulated MC or GC activity. We confirmed excitatory DREADDs increased MC c-Fos. However, there was no change in GC c-Fos, consistent with MC activation leading to GABAergic inhibition of GCs. In contrast, inhibitory DREADDs led to a large increase in GC c-Fos, consistent with a reduction in MC excitation of GABAergic neurons, and reduced inhibition of GCs. Together, these results suggest that MCs regulate anxiety and cognition in specific ways. We also raise the possibility that cognitive performance may be improved by reducing anxiety.SIGNIFICANCE STATEMENT The dentate gyrus (DG) has many important cognitive roles as well as being associated with affective behavior. This study addressed how a glutamatergic DG cell type called mossy cells (MCs) contributes to diverse behaviors, which is timely because it is known that MCs regulate the activity of the primary DG cell type, granule cells (GCs), but how MC activity influences behavior is unclear. We show, surprisingly, that activating MCs can lead to adverse behavioral outcomes, and inhibiting MCs have an opposite effect. Importantly, the results appeared to be task-dependent and showed that testing both sexes was important. Additional experiments indicated what MC and GC circuitry was involved. Together, the results suggest how MCs influence behaviors that involve the DG.
Collapse
|
50
|
Kobayashi Y, Okada T, Miki D, Sekino Y, Koganezawa N, Shirao T, Diniz GB, Saito Y. Properties of primary cilia in melanin-concentrating hormone receptor 1-bearing hippocampal neurons in vivo and in vitro. Neurochem Int 2020; 142:104902. [PMID: 33197527 DOI: 10.1016/j.neuint.2020.104902] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/02/2020] [Accepted: 11/10/2020] [Indexed: 12/31/2022]
Abstract
The primary cilium is a solitary organelle that organizes a sensitive signaling hub in a highly ordered microenvironment. Cilia are plastic structures, changing their length in response to bioactive substances, and ciliary length may be regulated to ensure efficient signaling capacity. Mammalian brain neurons possess primary cilia that are enriched in a set of G protein-coupled receptors (GPCRs), including the feeding-related melanin-concentrating hormone (MCH) receptor 1 (MCHR1). We previously demonstrated a novel biological phenomenon, ciliary MCHR1-mediated cilia length shortening through Gi/o and Akt signaling, using a simple cell culture model of human retinal pigmented epithelial RPE1 cells exogenously expressing MCHR1. In the present study, we characterized the properties of endogenous MCHR1-expressing primary cilia in hippocampal neurons in rodents. Using cultured dissociated rat hippocampal neurons in vitro, we showed that MCH triggered cilia length reduction involved in MCHR1-Gi/o and -Akt signaling. In rat hippocampal slice cultures with preservation of the cytoarchitecture and cell populations, ciliary MCHR1 was abundantly located in the CA1 and CA3 regions, but not in the dentate gyrus. Notably, treatment of slice cultures with MCH induced Gi/o- and Akt-dependent cilia shortening in the CA1 region without influencing cilia length in the CA3 region. Regarding the in vivo mouse brain, we observed higher levels of ciliary MCHR1 in the CA1 and CA3 regions as well as in slice cultures. In the starved state mice, a marked increase in MCH mRNA expression was detected in the lateral hypothalamus. Furthermore, MCHR1-positive cilia length in the hippocampal CA1 region was significantly shortened in fasted mice compared with fed mice. The present findings focused on the hippocampus provide a potential approach to investigate how MCHR1-driven cilia shortening regulates neuronal activity and physiological function toward feeding and memory tasks.
Collapse
Affiliation(s)
- Yuki Kobayashi
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan
| | - Tomoya Okada
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan
| | - Daisuke Miki
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan
| | - Yuko Sekino
- Endowed Laboratory of Human Cell-Based Drug Discovery, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Noriko Koganezawa
- Department of Neurobiology and Behavior, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Tomoaki Shirao
- Department of Neurobiology and Behavior, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan; AlzMed,Inc., UT South Clinical Research Building, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8485, Japan
| | - Giovanne B Diniz
- Department of Neurosurgery, Yale School of Medicine, 310 Cedar St, New Haven, CT, 06520, USA
| | - Yumiko Saito
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan.
| |
Collapse
|