1
|
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
|
2
|
Li C, Yan Y, Stork O, Shen R, Behnisch T. The E3 Ubiquitin Ligase PRAJA1: A Key Regulator of Synaptic Dynamics and Memory Processes with Implications for Alzheimer's Disease. Int J Mol Sci 2025; 26:2909. [PMID: 40243483 PMCID: PMC11988436 DOI: 10.3390/ijms26072909] [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: 02/05/2025] [Revised: 03/13/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
The precise regulation of synaptic function by targeted protein degradation is fundamental to learning and memory, yet the roles of many brain-enriched E3 ubiquitin ligases in this process remain elusive. Here, we uncover a critical and previously unappreciated role for the E3 ubiquitin ligase PRAJA1 in orchestrating synaptic plasticity and hippocampus-dependent memory. Utilizing C57BL/6 and 5xFAD male mice and employing a multi-faceted approach including protein biochemistry, molecular biology, in vitro electrophysiology, and behavioral assays, we demonstrate that long-term potentiation (LTP) induction triggers a rapid, proteasome-dependent downregulation of PRAJA1 within the CA1 region of the hippocampus. Critically, selective knockdown of PRAJA1 in vivo profoundly enhanced both object recognition and spatial memory, while disrupting normal exploratory behavior. Mechanistically, we reveal that PRAJA1 acts as a key regulator of synaptic architecture and transmission: its downregulation leads to a reduction in key synaptic proteins and spine density, influencing the excitatory/inhibitory balance and facilitating synaptic plasticity. Conversely, increased PRAJA1 expression potentiates GABAergic transmission. Furthermore, we identify spinophilin as a novel substrate of PRAJA1, suggesting a direct molecular link between PRAJA1 and synaptic remodeling. Strikingly, our findings implicate dysregulation of PRAJA1 in the pathogenesis of Alzheimer's disease, positioning PRAJA1 as a potential therapeutic target for cognitive enhancement in neurodegenerative conditions. These results unveil PRAJA1 as a critical molecular brake on synaptic plasticity and memory formation, offering a promising new avenue for understanding and potentially treating memory impairment.
Collapse
Affiliation(s)
- Chuhan Li
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yan Yan
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Oliver Stork
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany;
| | - Ruling Shen
- Shanghai Laboratory Animal Research Center, Shanghai 201203, China
| | - Thomas Behnisch
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
3
|
Raven F, Medina AV, Schmidt K, He A, Vankampen AA, Balendran V, Aton SJ. Brief sleep disruption alters synaptic structures among hippocampal and neocortical somatostatin-expressing interneurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.22.604591. [PMID: 39211205 PMCID: PMC11360998 DOI: 10.1101/2024.07.22.604591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Study objectives Brief sleep loss alters cognition and synaptic structures of principal neurons in hippocampus and neocortex. However, while in vivo recording and bioinformatic data suggest that inhibitory interneurons are more strongly affected by sleep loss, it is unclear how sleep and sleep deprivation affect interneurons' synapses. Disruption of the SST+ interneuron population seems to be a critical early sign of neuropathology in Alzheimer's dementia, schizophrenia, and bipolar disorder - and the risk of developing all three is increased by habitual sleep loss. We aimed to test how the synaptic structures of SST+ interneurons in various brain regions are affected by brief sleep disruption. Methods We used Brainbow 3.0 to label SST+ interneurons in the dorsal hippocampus, prefrontal cortex, and visual cortex of male SST-CRE transgenic mice, then compared synaptic structures in labeled neurons after a 6-h period of ad lib sleep, or gentle handling sleep deprivation (SD) starting at lights on. Results Dendritic spine density among SST+ interneurons in both hippocampus and neocortex was altered in a subregion-specific manner, with increased overall and thin spine density in CA1, dramatic increases in spine volume and surface area in CA3, and small but significant changes (primarily decreases) in spine size in CA1, PFC and V1. Conclusions Our suggest that the synaptic connectivity of SST+ interneurons is significantly altered in a brain region-specific manner by a few hours of sleep loss. This suggests a cell type-specific mechanism by which sleep loss disrupts cognition and alters excitatory-inhibitory balance in brain networks.
Collapse
Affiliation(s)
- Frank Raven
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Alexis Vega Medina
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Kailynn Schmidt
- University of New England College of Osteopathic Medicine, Biddeford, ME 04005
| | - Annie He
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Anna A. Vankampen
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Vinodh Balendran
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Sara J. Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| |
Collapse
|
4
|
Krause GM, Chirich Barreira LM, Albrecht A. Spatial mRNA expression patterns of orexin receptors in the dorsal hippocampus. Sci Rep 2024; 14:24788. [PMID: 39433837 PMCID: PMC11494061 DOI: 10.1038/s41598-024-76237-9] [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: 04/19/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
Orexins are wake-promoting neuropeptides that originate from hypothalamic neurons projecting to widespread brain areas throughout the central nervous system. They modulate various physiological functions via their orexin 1 (OXR1) and 2 (OXR2) receptors, including sleep-wake rhythm but also cognitive functions such as memory formation. Here, we provide a detailed analysis of OXR1 and OXR2 mRNA expression profiles in the dorsal hippocampus as a key region for memory formation, using RNAscope multiplex in situ hybridization. Interconnected subareas relevant for cognition and memory such as the medial prefrontal cortex and the nucleus reuniens of the thalamus were assessed as well. Both receptor types display distinct profiles, with the highest percentage of OXR1 mRNA-positive cells in the hilus of the dentate gyrus. Here, the content of OXR1 mRNA per cell was slightly modulated at selected time points over a 12 h light/ 12 dark light phase. Using RNAScope and quantitative polymerase chain reaction approaches, we began to address a cell-type specific expression of OXR1 in hilar GABAergic interneurons. The distinct expression profiles of both receptor subtypes within hippocampal subareas and circuits provide an interesting basis for future interventional studies on orexin receptor function in spatial and contextual memory.
Collapse
Affiliation(s)
- Gina Marie Krause
- Institute of Anatomy, Otto-von-Guericke-University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | | | - Anne Albrecht
- Institute of Anatomy, Otto-von-Guericke-University, Leipziger Str. 44, 39120, Magdeburg, Germany.
- Center for Behavioral Brain Sciences (CBBS), Universitätsplatz 2, 39106, Magdeburg, Germany.
- German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Magdeburg, Germany.
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany.
| |
Collapse
|
5
|
Cheng L, Xiao L, Lin W, Li M, Liu J, Qiu X, Li M, Zheng Y, Xu C, Wang Y, Chen Z. Histamine H 1 receptors in dentate gyrus-projecting cholinergic neurons of the medial septum suppress contextual fear retrieval in mice. Nat Commun 2024; 15:5805. [PMID: 38987240 PMCID: PMC11237085 DOI: 10.1038/s41467-024-50042-4] [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: 10/25/2023] [Accepted: 06/28/2024] [Indexed: 07/12/2024] Open
Abstract
Fear memory is essential for survival and adaptation, yet excessive fear memories can lead to emotional disabilities and mental disorders. Despite previous researches have indicated that histamine H1 receptor (H1R) exerts critical and intricate effects on fear memory, the role of H1R is still not clarified. Here, we show that deletion of H1R gene in medial septum (MS) but not other cholinergic neurons selectively enhances contextual fear memory without affecting cued memory by differentially activating the dentate gyrus (DG) neurons in mice. H1R in cholinergic neurons mediates the contextual fear retrieval rather than consolidation by decreasing acetylcholine release pattern in DG. Furthermore, selective knockdown of H1R in the MS is sufficient to enhance contextual fear memory by manipulating the retrieval-induced neurons in DG. Our results suggest that H1R in MS cholinergic neurons is critical for contextual fear retrieval, and could be a potential therapeutic target for individuals with fear-related disorders.
Collapse
Affiliation(s)
- Li Cheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Ling Xiao
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenkai Lin
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Minzhu Li
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiaying Liu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaoyun Qiu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Menghan Li
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Yanrong Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Cenglin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
6
|
Chartampila E, Elayouby KS, Leary P, LaFrancois JJ, Alcantara-Gonzalez D, Jain S, Gerencer K, Botterill JJ, Ginsberg SD, Scharfman HE. Choline supplementation in early life improves and low levels of choline can impair outcomes in a mouse model of Alzheimer's disease. eLife 2024; 12:RP89889. [PMID: 38904658 PMCID: PMC11192536 DOI: 10.7554/elife.89889] [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: 06/22/2024] Open
Abstract
Maternal choline supplementation (MCS) improves cognition in Alzheimer's disease (AD) models. However, the effects of MCS on neuronal hyperexcitability in AD are unknown. We investigated the effects of MCS in a well-established mouse model of AD with hyperexcitability, the Tg2576 mouse. The most common type of hyperexcitability in Tg2576 mice are generalized EEG spikes (interictal spikes [IIS]). IIS also are common in other mouse models and occur in AD patients. In mouse models, hyperexcitability is also reflected by elevated expression of the transcription factor ∆FosB in the granule cells (GCs) of the dentate gyrus (DG), which are the principal cell type. Therefore, we studied ΔFosB expression in GCs. We also studied the neuronal marker NeuN within hilar neurons of the DG because reduced NeuN protein expression is a sign of oxidative stress or other pathology. This is potentially important because hilar neurons regulate GC excitability. Tg2576 breeding pairs received a diet with a relatively low, intermediate, or high concentration of choline. After weaning, all mice received the intermediate diet. In offspring of mice fed the high choline diet, IIS frequency declined, GC ∆FosB expression was reduced, and hilar NeuN expression was restored. Using the novel object location task, spatial memory improved. In contrast, offspring exposed to the relatively low choline diet had several adverse effects, such as increased mortality. They had the weakest hilar NeuN immunoreactivity and greatest GC ΔFosB protein expression. However, their IIS frequency was low, which was surprising. The results provide new evidence that a diet high in choline in early life can improve outcomes in a mouse model of AD, and relatively low choline can have mixed effects. This is the first study showing that dietary choline can regulate hyperexcitability, hilar neurons, ΔFosB, and spatial memory in an animal model of AD.
Collapse
Affiliation(s)
- Elissavet Chartampila
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Karim S Elayouby
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Paige Leary
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Department of Neuroscience and Physiology, New York University Grossman School of MedicineNew YorkUnited States
| | - John J LaFrancois
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
| | - David Alcantara-Gonzalez
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
| | - Swati Jain
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Kasey Gerencer
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Justin J Botterill
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Stephen D Ginsberg
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Department of Neuroscience and Physiology, New York University Grossman School of MedicineNew YorkUnited States
- Department of Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
- NYU Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Helen E Scharfman
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Department of Neuroscience and Physiology, New York University Grossman School of MedicineNew YorkUnited States
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
- Department of Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
- NYU Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
7
|
Chartampila E, Elayouby KS, Leary P, LaFrancois JJ, Alcantara-Gonzalez D, Jain S, Gerencer K, Botterill JJ, Ginsberg SD, Scharfman HE. Choline supplementation in early life improves and low levels of choline can impair outcomes in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.12.540428. [PMID: 37214805 PMCID: PMC10197642 DOI: 10.1101/2023.05.12.540428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Maternal choline supplementation (MCS) improves cognition in Alzheimer's disease (AD) models. However, effects of MCS on neuronal hyperexcitability in AD are unknown. We investigated effects of MCS in a well-established mouse model of AD with hyperexcitability, the Tg2576 mouse. The most common type of hyperexcitability in Tg2576 mice are generalized EEG spikes (interictal spikes; IIS). IIS also are common in other mouse models and occur in AD patients. Im mouse models, hyperexcitability is also reflected by elevated expression of the transcription factor ΔFosB in the granule cells (GCs) of the dentate gyrus (DG), which are the principal cell type. Therefore we studied ΔFosB expression in GCs. We also studied the the neuronal marker NeuN within hilar neurons of the DG because other studies have reduced NeuN protein expression is a sign of oxidative stress or other pathology. This is potentially important because hilar neurons regulate GC excitability. Tg2576 breeding pairs received a diet with a relatively low, intermediate or high concentration of choline. After weaning, all mice received the intermediate diet. In offspring of mice fed the high choline diet, IIS frequency declined, GC ΔFosB expression was reduced, and NeuN expression was restored. Using the novel object location task, spatial memory improved. In contrast, offspring exposed to the relatively low choline diet had several adverse effects, such as increased mortality. They had the weakest hilar NeuN immunoreactivity and greatest GC ΔFosB protein expression. However, their IIS frequency was low, which was surprising. The results provide new evidence that a diet high in choline in early life can improve outcomes in a mouse model of AD, and relatively low choline can have mixed effects. This is the first study showing that dietary choline can regulate hyperexcitability, hilar neurons, ΔFosB and spatial memory in an animal model of AD.
Collapse
Affiliation(s)
- Elissavet Chartampila
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Current address:Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27510
| | - Karim S. Elayouby
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Current address: Department of Neurology, Mount Sinai School of Medicine, New York, NY 10029
| | - Paige Leary
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY 100016
| | - John J. LaFrancois
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Department of Child and Adolescent Psychiatry , New York University Grossman School of Medicine, New York, NY 10016
| | - David Alcantara-Gonzalez
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Department of Child and Adolescent Psychiatry , New York University Grossman School of Medicine, New York, NY 10016
| | - Swati Jain
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - Kasey Gerencer
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Current address: Department of Psychology, University of Maine, Orono, ME 04469
| | - Justin J. Botterill
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Current address: Department of Anatomy, Physiology, & Pharmacology, College of Medicine, Saskatoon, SK S7N 5E5
| | - Stephen D. Ginsberg
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY 100016
- Department of Psychiatry, New York University Grossman School of Medicine New York, NY 10016
- NYU Neuroscience Institute,, New York University Grossman School of Medicine, New York, NY 10016
| | - Helen E. Scharfman
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY 100016
- Department of Child and Adolescent Psychiatry , New York University Grossman School of Medicine, New York, NY 10016
- Department of Psychiatry, New York University Grossman School of Medicine New York, NY 10016
- NYU Neuroscience Institute,, New York University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
8
|
Arrabal-Gómez C, Beltran-Casanueva R, Hernández-García A, Bayolo-Guanche JV, Barbancho-Fernández MA, Serrano-Castro PJ, Narváez M. Enhancing Cognitive Functions and Neuronal Growth through NPY1R Agonist and Ketamine Co-Administration: Evidence for NPY1R-TrkB Heteroreceptor Complexes in Rats. Cells 2024; 13:669. [PMID: 38667284 PMCID: PMC11049095 DOI: 10.3390/cells13080669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
This study investigates the combined effects of the neuropeptide Y Y1 receptor (NPY1R) agonist [Leu31-Pro34]NPY at a dose of 132 µg and Ketamine at 10 mg/Kg on cognitive functions and neuronal proliferation, against a backdrop where neurodegenerative diseases present an escalating challenge to global health systems. Utilizing male Sprague-Dawley rats in a physiological model, this research employed a single-dose administration of these compounds and assessed their impact 24 h after treatment on object-in-place memory tasks, alongside cellular proliferation within the dorsal hippocampus dentate gyrus. Methods such as the in situ proximity ligation assay and immunohistochemistry for proliferating a cell nuclear antigen (PCNA) and doublecortin (DCX) were utilized. The results demonstrated that co-administration significantly enhanced memory consolidation and increased neuronal proliferation, specifically neuroblasts, without affecting quiescent neural progenitors and astrocytes. These effects were mediated by the potential formation of NPY1R-TrkB heteroreceptor complexes, as suggested by receptor co-localization studies, although further investigation is required to conclusively prove this interaction. The findings also highlighted the pivotal role of brain-derived neurotrophic factor (BDNF) in mediating these effects. In conclusion, this study presents a promising avenue for enhancing cognitive functions and neuronal proliferation through the synergistic action of the NPY1R agonist and Ketamine, potentially via NPY1R-TrkB heteroreceptor complex formation, offering new insights into therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Carlos Arrabal-Gómez
- NeuronLab, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain; (C.A.-G.); (M.A.B.-F.)
- Facultad de Psicología, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain
- Unit of Neurology, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga, 29010 Málaga, Spain
- Vithas Málaga, Grupo Hospitalario Vithas, 29016 Málaga, Spain
| | - Rasiel Beltran-Casanueva
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; (R.B.-C.); (A.H.-G.); (J.V.B.-G.)
- Receptomics and Brain Disorders Lab, Edificio Lopez-Peñalver, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain
| | - Aracelis Hernández-García
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; (R.B.-C.); (A.H.-G.); (J.V.B.-G.)
- Receptomics and Brain Disorders Lab, Edificio Lopez-Peñalver, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain
| | - Juan Vicente Bayolo-Guanche
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; (R.B.-C.); (A.H.-G.); (J.V.B.-G.)
- Receptomics and Brain Disorders Lab, Edificio Lopez-Peñalver, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain
| | - Miguel Angel Barbancho-Fernández
- NeuronLab, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain; (C.A.-G.); (M.A.B.-F.)
| | - Pedro Jesús Serrano-Castro
- NeuronLab, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain; (C.A.-G.); (M.A.B.-F.)
- Unit of Neurology, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga, 29010 Málaga, Spain
- Vithas Málaga, Grupo Hospitalario Vithas, 29016 Málaga, Spain
| | - Manuel Narváez
- NeuronLab, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain; (C.A.-G.); (M.A.B.-F.)
- Unit of Neurology, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga, 29010 Málaga, Spain
- Vithas Málaga, Grupo Hospitalario Vithas, 29016 Málaga, Spain
| |
Collapse
|
9
|
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. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.09.579729. [PMID: 38645244 PMCID: PMC11027210 DOI: 10.1101/2024.02.09.579729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
INTRODUCTION Hyperexcitability in Alzheimer's disease (AD) emerge early and contribute to disease progression. The dentate gyrus (DG) is implicated in hyperexcitability in AD. We hypothesized that mossy cells (MCs), regulators of DG excitability, contribute to early hyperexcitability in AD. Indeed, MCs generate hyperexcitability in epilepsy. METHODS Using the Tg2576 model and WT mice (∼1month-old), we compared MCs electrophysiologically, assessed c-Fos activity marker, Aβ expression and mice performance in a hippocampal-dependent memory task. RESULTS Tg2576 MCs exhibit increased spontaneous excitatory events and decreased inhibitory currents, increasing the charge transfer excitation/inhibition ratio. Tg2576 MC intrinsic excitability was enhanced, and showed higher c-Fos, intracellular Aβ expression, and axon sprouting. Granule cells only showed changes in synaptic properties, without intrinsic changes. The effects occurred before a memory task is affected. DISCUSSION Early electrophysiological and morphological alterations in Tg2576 MCs are consistent with enhanced excitability, suggesting an early role in DG hyperexcitability and AD pathophysiology. HIGHLIGHTS ∘ MCs from 1 month-old Tg2576 mice had increased spontaneous excitatory synaptic input. ∘ Tg2576 MCs had reduced spontaneous inhibitory synaptic input. ∘ Several intrinsic properties were abnormal in Tg2576 MCs. ∘ Tg2576 GCs had enhanced synaptic excitation but no changes in intrinsic properties. ∘ Tg2576 MCs exhibited high c-Fos expression, soluble Aβ and axonal sprouting.
Collapse
|
10
|
Satchell M, Fry B, Noureddine Z, Simmons A, Ognjanovski NN, Aton SJ, Zochowski MR. Neuromodulation via muscarinic acetylcholine pathway can facilitate distinct, complementary, and sequential roles for NREM and REM states during sleep-dependent memory consolidation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.19.541465. [PMID: 38293183 PMCID: PMC10827095 DOI: 10.1101/2023.05.19.541465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Across vertebrate species, sleep consists of repeating cycles of NREM followed by REM. However, the respective functions of NREM, REM, and their stereotypic cycling pattern are not well understood. Using a simplified biophysical network model, we show that NREM and REM sleep can play differential and critical roles in memory consolidation primarily regulated, based on state-specific changes in cholinergic signaling. Within this network, decreasing and increasing muscarinic acetylcholine (ACh) signaling during bouts of NREM and REM, respectively, differentially alters neuronal excitability and excitatory/inhibitory balance. During NREM, deactivation of inhibitory neurons leads to network-wide disinhibition and bursts of synchronized activity led by firing in engram neurons. These features strengthen connections from the original engram neurons to less-active network neurons. In contrast, during REM, an increase in network inhibition suppresses firing in all but the most-active excitatory neurons, leading to competitive strengthening/pruning of the memory trace. We tested the predictions of the model against in vivo recordings from mouse hippocampus during active sleep-dependent memory storage. Consistent with modeling results, we find that functional connectivity between CA1 neurons changes differentially at transition from NREM to REM sleep during learning. Returning to the model, we find that an iterative sequence of state-specific activations during NREM/REM cycling is essential for memory storage in the network, serving a critical role during simultaneous consolidation of multiple memories. Together these results provide a testable mechanistic hypothesis for the respective roles of NREM and REM sleep, and their universal relative timing, in memory consolidation. Significance statement Using a simplified computational model and in vivo recordings from mouse hippocampus, we show that NREM and REM sleep can play differential roles in memory consolidation. The specific neurophysiological features of the two sleep states allow for expansion of memory traces (during NREM) and prevention of overlap between different memory traces (during REM). These features are likely essential in the context of storing more than one new memory simultaneously within a brain network.
Collapse
|
11
|
Kruse P, Brandes G, Hemeling H, Huang Z, Wrede C, Hegermann J, Vlachos A, Lenz M. Synaptopodin Regulates Denervation-Induced Plasticity at Hippocampal Mossy Fiber Synapses. Cells 2024; 13:114. [PMID: 38247806 PMCID: PMC10814840 DOI: 10.3390/cells13020114] [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: 11/21/2023] [Revised: 12/17/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
Neurological diseases can lead to the denervation of brain regions caused by demyelination, traumatic injury or cell death. The molecular and structural mechanisms underlying lesion-induced reorganization of denervated brain regions, however, are a matter of ongoing investigation. In order to address this issue, we performed an entorhinal cortex lesion (ECL) in mouse organotypic entorhino-hippocampal tissue cultures of both sexes and studied denervation-induced plasticity of mossy fiber synapses, which connect dentate granule cells (dGCs) with CA3 pyramidal cells (CA3-PCs) and play important roles in learning and memory formation. Partial denervation caused a strengthening of excitatory neurotransmission in dGCs, CA3-PCs and their direct synaptic connections, as revealed by paired recordings (dGC-to-CA3-PC). These functional changes were accompanied by ultrastructural reorganization of mossy fiber synapses, which regularly contain the plasticity-regulating protein synaptopodin and the spine apparatus organelle. We demonstrate that the spine apparatus organelle and synaptopodin are related to ribosomes in close proximity to synaptic sites and reveal a synaptopodin-related transcriptome. Notably, synaptopodin-deficient tissue preparations that lack the spine apparatus organelle failed to express lesion-induced synaptic adjustments. Hence, synaptopodin and the spine apparatus organelle play a crucial role in regulating lesion-induced synaptic plasticity at hippocampal mossy fiber synapses.
Collapse
Affiliation(s)
- Pia Kruse
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Gudrun Brandes
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Hanna Hemeling
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Zhong Huang
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Christoph Wrede
- Institute of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany
- Research Core Unit Electron Microscopy, Hannover Medical School, 30625 Hannover, Germany
| | - Jan Hegermann
- Institute of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany
- Research Core Unit Electron Microscopy, Hannover Medical School, 30625 Hannover, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, 79104 Freiburg, Germany
| | - Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
12
|
Klinger K, del Ángel M, Çalışkan G, Stork O. Increasing NPYergic transmission in the hippocampus rescues aging-related deficits of long-term potentiation in the mouse dentate gyrus. Front Aging Neurosci 2023; 15:1283581. [PMID: 38020778 PMCID: PMC10673643 DOI: 10.3389/fnagi.2023.1283581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Loss of neuropeptide Y (NPY)-expressing interneurons in the hippocampus and decaying cholinergic neuromodulation are thought to contribute to impaired cognitive function during aging. However, the interaction of these two neuromodulatory systems in maintaining hippocampal synaptic plasticity during healthy aging has not been explored so far. Here we report profound sex differences in the Neuropeptide-Y (NPY) levels in the dorsal dentate gyrus (DG) with higher NPY concentrations in the male mice compared to their female counterparts and a reduction of NPY levels during aging specifically in males. This change in aged males is accompanied by a deficit in theta burst-induced long-term potentiation (LTP) in the medial perforant path-to-dorsal DG (MPP-DG) synapse, which can be rescued by enhancing cholinergic activation with the acetylcholine esterase blocker, physostigmine. Importantly, NPYergic transmission is required for this rescue of LTP. Moreover, exogenous NPY application alone is sufficient to recover LTP induction in aged male mice, even in the absence of the cholinergic stimulator. Together, our results suggest that in male mice NPYergic neurotransmission is a critical factor for maintaining dorsal DG LTP during aging.
Collapse
Affiliation(s)
- Katharina Klinger
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University, Magdeburg, Germany
- Research Group “Synapto-Oscillopathies”, Institute of Biology, Otto-von-Guericke-University, Magdeburg, Germany
| | - Miguel del Ángel
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University, Magdeburg, Germany
| | - Gürsel Çalışkan
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University, Magdeburg, Germany
- Research Group “Synapto-Oscillopathies”, Institute of Biology, Otto-von-Guericke-University, Magdeburg, Germany
- Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Oliver Stork
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences, Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Magdeburg, Germany
- German Center for Mental Health (DZPG), Magdeburg, Germany
| |
Collapse
|
13
|
Kloc ML, Chen Y, Daglian JM, Holmes GL, Baram TZ, Barry JM. Spatial learning impairments and discoordination of entorhinal-hippocampal circuit coding following prolonged febrile seizures. Hippocampus 2023; 33:970-992. [PMID: 37096324 PMCID: PMC10529121 DOI: 10.1002/hipo.23541] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 03/30/2023] [Accepted: 04/08/2023] [Indexed: 04/26/2023]
Abstract
How the development and function of neural circuits governing learning and memory are affected by insults in early life remains poorly understood. The goal of this study was to identify putative changes in cortico-hippocampal signaling mechanisms that could lead to learning and memory deficits in a clinically relevant developmental pathophysiological rodent model, Febrile status epilepticus (FSE). FSE in both pediatric cases and the experimental animal model, is associated with enduring physiological alterations of the hippocampal circuit and cognitive impairment. Here, we deconstruct hippocampal circuit throughput by inducing slow theta oscillations in rats under urethane anesthesia and isolating the dendritic compartments of CA1 and dentate gyrus subfields, their reception of medial and lateral entorhinal cortex inputs, and the efficacy of signal propagation to each somatic cell layer. We identify FSE-induced theta-gamma decoupling at cortical synaptic input pathways and altered signal phase coherence along the CA1 and dentate gyrus somatodendritic axes. Moreover, increased DG synaptic activity levels are predictive of poor cognitive outcomes. We propose that these alterations in cortico-hippocampal coordination interfere with the ability of hippocampal dendrites to receive, decode and propagate neocortical inputs. If this frequency-specific syntax is necessary for cortico-hippocampal coordination and spatial learning and memory, its loss could be a mechanism for FSE cognitive comorbidities.
Collapse
Affiliation(s)
- Michelle L. Kloc
- Epilepsy Cognition and Development Group, Department of Neurological Sciences, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| | - Yuncai Chen
- Departments of Pediatrics, University California-Irvine, Irvine, California, USA
- Departments of Anatomy/Neurobiology, University California-Irvine, Irvine, California, USA
| | - Jennifer M. Daglian
- Departments of Pediatrics, University California-Irvine, Irvine, California, USA
| | - Gregory L. Holmes
- Epilepsy Cognition and Development Group, Department of Neurological Sciences, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| | - Tallie Z. Baram
- Departments of Pediatrics, University California-Irvine, Irvine, California, USA
- Departments of Anatomy/Neurobiology, University California-Irvine, Irvine, California, USA
- Departments of Neurology, University California-Irvine, Irvine, California, USA
| | - Jeremy M. Barry
- Epilepsy Cognition and Development Group, Department of Neurological Sciences, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| |
Collapse
|
14
|
Zernov N, Veselovsky AV, Poroikov VV, Melentieva D, Bolshakova A, Popugaeva E. New Positive TRPC6 Modulator Penetrates Blood-Brain Barrier, Eliminates Synaptic Deficiency and Restores Memory Deficit in 5xFAD Mice. Int J Mol Sci 2022; 23:13552. [PMID: 36362339 PMCID: PMC9653995 DOI: 10.3390/ijms232113552] [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: 10/05/2022] [Revised: 11/01/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022] Open
Abstract
Synapse loss in the brain of Alzheimer's disease patients correlates with cognitive dysfunctions. Drugs that limit synaptic loss are promising pharmacological agents. The transient receptor potential cation channel, subfamily C, member 6 (TRPC6) regulates the formation of an excitatory synapse. Positive regulation of TRPC6 results in increased synapse formation and enhances learning and memory in animal models. The novel selective TRPC6 agonist, 3-(3-,4-Dihydro-6,7-dimethoxy-3,3-dimethyl-1-isoquinolinyl)-2H-1-benzopyran-2-one, has recently been identified. Here we present in silico, in vitro, ex vivo, pharmacokinetic and in vivo studies of this compound. We demonstrate that it binds to the extracellular agonist binding site of the human TRPC6, protects hippocampal mushroom spines from amyloid toxicity in vitro, efficiently recovers synaptic plasticity in 5xFAD brain slices, penetrates the blood-brain barrier and recovers cognitive deficits in 5xFAD mice. We suggest that C20 might be recognized as the novel TRPC6-selective drug suitable to treat synaptic deficiency in Alzheimer's disease-affected hippocampal neurons.
Collapse
Affiliation(s)
- Nikita Zernov
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia
| | - Alexander V. Veselovsky
- Department of Bioinformatics, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia
| | - Vladimir V. Poroikov
- Department of Bioinformatics, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia
| | - Daria Melentieva
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia
| | - Anastasia Bolshakova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia
| | - Elena Popugaeva
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia
| |
Collapse
|
15
|
Perrenoud Q, Leclerc C, Geoffroy H, Vitalis T, Richetin K, Rampon C, Gallopin T. Molecular and electrophysiological features of GABAergic neurons in the dentate gyrus reveal limited homology with cortical interneurons. PLoS One 2022; 17:e0270981. [PMID: 35802727 PMCID: PMC9269967 DOI: 10.1371/journal.pone.0270981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
GABAergic interneurons tend to diversify into similar classes across telencephalic regions. However, it remains unclear whether the electrophysiological and molecular properties commonly used to define these classes are discriminant in the hilus of the dentate gyrus. Here, using patch-clamp combined with single cell RT-PCR, we compare the relevance of commonly used electrophysiological and molecular features for the clustering of GABAergic interneurons sampled from the mouse hilus and primary sensory cortex. While unsupervised clustering groups cortical interneurons into well-established classes, it fails to provide a convincing partition of hilar interneurons. Statistical analysis based on resampling indicates that hilar and cortical GABAergic interneurons share limited homology. While our results do not invalidate the use of classical molecular marker in the hilus, they indicate that classes of hilar interneurons defined by the expression of molecular markers do not exhibit strongly discriminating electrophysiological properties.
Collapse
Affiliation(s)
- Quentin Perrenoud
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Clémence Leclerc
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, France
| | - Hélène Geoffroy
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Tania Vitalis
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Kevin Richetin
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, France
| | - Claire Rampon
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, France
| | - Thierry Gallopin
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
- * E-mail:
| |
Collapse
|
16
|
Probing the Skin–Brain Axis: New Vistas Using Mouse Models. Int J Mol Sci 2022; 23:ijms23137484. [PMID: 35806489 PMCID: PMC9267936 DOI: 10.3390/ijms23137484] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 12/10/2022] Open
Abstract
Inflammatory diseases of the skin, including atopic dermatitis and psoriasis, have gained increasing attention with rising incidences in developed countries over the past decades. While bodily properties, such as immunological responses of the skin, have been described in some detail, interactions with the brain via different routes are less well studied. The suggested routes of the skin–brain axis comprise the immune system, HPA axis, and the peripheral and central nervous system, including microglia responses and structural changes. They provide starting points to investigate the molecular mechanisms of neuropsychiatric comorbidities in AD and psoriasis. To this end, mouse models exist for AD and psoriasis that could be tested for relevant behavioral entities. In this review, we provide an overview of the current mouse models and assays. By combining an extensive behavioral characterization and state-of-the-art genetic interventions with the investigation of underlying molecular pathways, insights into the mechanisms of the skin–brain axis in inflammatory cutaneous diseases are examined, which will spark further research in humans and drive the development of novel therapeutic strategies.
Collapse
|
17
|
Albrecht A, Müller I, Weiglein A, Pollali E, Çalışkan G, Stork O. Choosing memory retrieval strategies: A critical role for inhibition in the dentate gyrus. Neurobiol Stress 2022; 20:100474. [PMID: 35958670 PMCID: PMC9357949 DOI: 10.1016/j.ynstr.2022.100474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/30/2022] [Accepted: 07/20/2022] [Indexed: 11/20/2022] Open
Abstract
Remembering the location of food is essential for survival. Rodents and humans employ mainly hippocampus-dependent spatial strategies, but when being stressed they shift to striatum-mediated stimulus-based strategies. To investigate underlying brain circuits, we tested mice with a heightened stress susceptibility due to a lack of the GABA-synthetizing enzyme GAD65 (GAD65−/− mice) in a dual solution task. Here, GAD65−/− mice preferred to locate a food reward in an open field via a proximal cue, while their wildtype littermates preferred a spatial strategy. The analysis of cFos co-activation across brain regions and of stress-induced mRNA expression changes of GAD65 pointed towards the hippocampal dorsal dentate gyrus (dDG) as a central structure for mediating stress effects on strategy choices via GAD65. Reducing the GAD65 expression locally in the dDG by a shRNA mediated knock down was sufficient to replicate the phenotype of the global GAD65 knock out and to increase dDG excitability. Using DREADD vectors to specifically interfere with dDG circuit activity during dual solution retrieval but not learning confirmed that the dDG modulates strategy choices and that a balanced excitability of this structure is necessary to establish spatial strategy preference. These data highlight the dDG as a critical hub for choosing between spatial and non-spatial foraging strategies. Stress reduces spatial memory preferences for locating rewards in an open field. GAD65 deficient mice show reduced preferences for spatial memory strategy. Dorsal dentate gyrus knock down of GAD65 is sufficient to reduce spatial strategies. Excitability in the dorsal dentate gyrus modulates retrieval strategy choices.
Collapse
Affiliation(s)
- Anne Albrecht
- Institute of Anatomy, Otto-von-Guericke-University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Corresponding author.
| | - Iris Müller
- Institute of Biology, Otto-von-Guericke-University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Aliće Weiglein
- Institute of Anatomy, Otto-von-Guericke-University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Evangelia Pollali
- Institute of Biology, Otto-von-Guericke-University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Gürsel Çalışkan
- Institute of Biology, Otto-von-Guericke-University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Oliver Stork
- Institute of Biology, Otto-von-Guericke-University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Universitätsplatz 2, 39106, Magdeburg, Germany
| |
Collapse
|
18
|
Mu R, Tang S, Han X, Wang H, Yuan D, Zhao J, Long Y, Hong H. A cholinergic medial septum input to medial habenula mediates generalization formation and extinction of visual aversion. Cell Rep 2022; 39:110882. [PMID: 35649349 DOI: 10.1016/j.celrep.2022.110882] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 12/07/2021] [Accepted: 05/06/2022] [Indexed: 12/28/2022] Open
Abstract
Generalization of visual aversion is a critical function of the brain that supports survival, but the underlying neurobiological mechanisms are unclear. We establish a rapid generalization procedure for inducing visual aversion by dynamic stripe images. By using fiber photometry, apoptosis, chemogenetic and optogenetic techniques, and behavioral tests, we find that decreased cholinergic neurons' activity in the medial septum (MS) leads to generalization loss of visual aversion. Strikingly, we identify a projection from MS cholinergic neurons to the medial habenula (MHb) and find that inhibition of the MS→MHb cholinergic circuit disrupts aversion-generalization formation while its continuous activation disrupts subsequent extinction. Further studies show that MS→MHb cholinergic projections modulate the generalization of visual aversion possibly via M1 muscarinic acetylcholine receptors (mAChRs) of downstream neurons coreleasing glutamate and acetylcholine. These findings reveal that the MS→MHb cholinergic circuit is a critical node in aversion-generalization formation and extinction and potentially provides insight into the pathogenesis of affective disorders.
Collapse
Affiliation(s)
- Ronghao Mu
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China
| | - Susu Tang
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaomeng Han
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China
| | - Hao Wang
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China
| | - Danhua Yuan
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China
| | - Jiajia Zhao
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China
| | - Yan Long
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China.
| | - Hao Hong
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
19
|
Chen Y, Zhou Y, Li XC, Ma X, Mi WL, Chu YX, Wang YQ, Mao-Ying QL. Neuronal GRK2 regulates microglial activation and contributes to electroacupuncture analgesia on inflammatory pain in mice. Biol Res 2022; 55:5. [PMID: 35115050 PMCID: PMC8812183 DOI: 10.1186/s40659-022-00374-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 01/22/2022] [Indexed: 12/30/2022] Open
Abstract
Background G protein coupled receptor kinase 2 (GRK2) has been demonstrated to play a crucial role in the development of chronic pain. Acupuncture is an alternative therapy widely used for pain management. In this study, we investigated the role of spinal neuronal GRK2 in electroacupuncture (EA) analgesia. Methods The mice model of inflammatory pain was built by subcutaneous injection of Complete Freund’s Adjuvant (CFA) into the plantar surface of the hind paws. The mechanical allodynia of mice was examined by von Frey test. The mice were subjected to EA treatment (BL60 and ST36 acupuncture points) for 1 week. Overexpression and downregulation of spinal neuronal GRK2 were achieved by intraspinal injection of adeno associated virus (AAV) containing neuron-specific promoters, and microglial activation and neuroinflammation were evaluated by real-time PCR. Results Intraplantar injection with CFA in mice induced the decrease of GRK2 and microglial activation along with neuroinflammation in spinal cord. EA treatment increased the spinal GRK2, reduced neuroinflammation, and significantly decreased CFA-induced mechanical allodynia. The effects of EA were markedly weakened by non-cell-specific downregulation of spinal GRK2. Further, intraspinal injection of AAV containing neuron-specific promoters specifically downregulated neuronal GRK2, and weakened the regulatory effect of EA on CFA-induced mechanical allodynia and microglial activation. Meanwhile, overexpression of spinal neuronal GRK2 decreased mechanical allodynia. All these indicated that the neuronal GRK2 mediated microglial activation and neuroinflammation, and subsequently contributed to CFA-induced inflammatory pain. Conclusion The restoration of the spinal GRK2 and subsequent suppression of microglial activation and neuroinflammation might be an important mechanism for EA analgesia. Our findings further suggested that the spinal GRK2, especially neuronal GRK2, might be the potential target for EA analgesia and pain management, and we provided a new experimental basis for the EA treatment of pain. Supplementary Information The online version contains supplementary material available at 10.1186/s40659-022-00374-6.
Collapse
Affiliation(s)
- Yu Chen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yang Zhou
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xiao-Chen Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xue Ma
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, People's Republic of China
| | - Wen-Li Mi
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yu-Xia Chu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yan-Qing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, People's Republic of China.,State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, People's Republic of China.,Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, 200433, People's Republic of China
| | - Qi-Liang Mao-Ying
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, People's Republic of China. .,Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
20
|
Yavas E, Trott JM, Fanselow MS. Sexually dimorphic muscarinic acetylcholine receptor modulation of contextual fear learning in the dentate gyrus. Neurobiol Learn Mem 2021; 185:107528. [PMID: 34607024 PMCID: PMC8849609 DOI: 10.1016/j.nlm.2021.107528] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/16/2021] [Accepted: 09/24/2021] [Indexed: 11/28/2022]
Abstract
Contextual fear conditioning, where the prevailing situational cues become associated with an aversive unconditional stimulus such as electric shock, is sexually dimorphic. Males typically show higher levels of fear than females. There are two components to contextual fear conditioning. First the multiple cues that encompass the context must be integrated into a coherent representation, a process that requires the hippocampus. The second is that representation must be communicated to the basolateral amygdala where it can be associated with shock. If there is inadequate time for forming the representation prior to shock poor conditioning results and this is called the immediate shock deficit. One can isolate the contextual processing component, as well as alleviate the deficit, by providing an opportunity to explore the context without shock prior to the conditioning session. The purpose of the present study was to determine the extent to which cholinergic processes within the dentate gyrus of the hippocampus during contextual processing contribute to the sexual dimorphism. Clozapine-n-oxide (CNO) is a putatively inactive compound that acts only upon synthetic genetically engineered receptors. However, we found that CNO infused into the dentate gyrus prior to exploration eliminated the sexual dimorphism by selectively decreasing freezing in males to the level of females. Biological activity of CNO is usually attributed to metabolism of CNO to clozapine and we found that clozapine, and the muscarinic cholinergic antagonist, scopolamine, produced results similar to CNO, preferentially affecting males. On the other hand, the muscarinic agonist oxotremorine selectively impaired conditioning in females. Overall, the current experiments reveal significant off-target effects of CNO and implicate muscarinic cholinergic receptors in the dentate gyrus as a significant mediator of the sexual dimorphism in contextual fear conditioning.
Collapse
Affiliation(s)
- Ersin Yavas
- Staglin Center for Brain and Behavioral Health, Department of Psychology, Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Jeremy M Trott
- Staglin Center for Brain and Behavioral Health, Department of Psychology, Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Michael S Fanselow
- Staglin Center for Brain and Behavioral Health, Department of Psychology, Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
21
|
Palacios-Filardo J, Udakis M, Brown GA, Tehan BG, Congreve MS, Nathan PJ, Brown AJH, Mellor JR. Acetylcholine prioritises direct synaptic inputs from entorhinal cortex to CA1 by differential modulation of feedforward inhibitory circuits. Nat Commun 2021; 12:5475. [PMID: 34531380 PMCID: PMC8445995 DOI: 10.1038/s41467-021-25280-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 07/21/2021] [Indexed: 02/08/2023] Open
Abstract
Acetylcholine release in the hippocampus plays a central role in the formation of new memory representations. An influential but largely untested theory proposes that memory formation requires acetylcholine to enhance responses in CA1 to new sensory information from entorhinal cortex whilst depressing inputs from previously encoded representations in CA3. Here, we show that excitatory inputs from entorhinal cortex and CA3 are depressed equally by synaptic release of acetylcholine in CA1. However, feedforward inhibition from entorhinal cortex exhibits greater depression than CA3 resulting in a selective enhancement of excitatory-inhibitory balance and CA1 activation by entorhinal inputs. Entorhinal and CA3 pathways engage different feedforward interneuron subpopulations and cholinergic modulation of presynaptic function is mediated differentially by muscarinic M3 and M4 receptors, respectively. Thus, our data support a role and mechanisms for acetylcholine to prioritise novel information inputs to CA1 during memory formation.
Collapse
Affiliation(s)
- Jon Palacios-Filardo
- Center for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, UK
| | - Matt Udakis
- Center for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, UK
| | - Giles A Brown
- Sosei Heptares, Steinmetz Building, Granta Park, Great Abingdon, Cambridge, UK
- OMass Therapeutics Ltd, The Schrödinger Building, Oxford, UK
| | - Benjamin G Tehan
- Sosei Heptares, Steinmetz Building, Granta Park, Great Abingdon, Cambridge, UK
- OMass Therapeutics Ltd, The Schrödinger Building, Oxford, UK
| | - Miles S Congreve
- Sosei Heptares, Steinmetz Building, Granta Park, Great Abingdon, Cambridge, UK
| | - Pradeep J Nathan
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Alastair J H Brown
- Sosei Heptares, Steinmetz Building, Granta Park, Great Abingdon, Cambridge, UK
| | - Jack R Mellor
- Center for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, UK.
| |
Collapse
|
22
|
Raven F, Aton SJ. The Engram's Dark Horse: How Interneurons Regulate State-Dependent Memory Processing and Plasticity. Front Neural Circuits 2021; 15:750541. [PMID: 34588960 PMCID: PMC8473837 DOI: 10.3389/fncir.2021.750541] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 08/26/2021] [Indexed: 12/15/2022] Open
Abstract
Brain states such as arousal and sleep play critical roles in memory encoding, storage, and recall. Recent studies have highlighted the role of engram neurons-populations of neurons activated during learning-in subsequent memory consolidation and recall. These engram populations are generally assumed to be glutamatergic, and the vast majority of data regarding the function of engram neurons have focused on glutamatergic pyramidal or granule cell populations in either the hippocampus, amygdala, or neocortex. Recent data suggest that sleep and wake states differentially regulate the activity and temporal dynamics of engram neurons. Two potential mechanisms for this regulation are either via direct regulation of glutamatergic engram neuron excitability and firing, or via state-dependent effects on interneuron populations-which in turn modulate the activity of glutamatergic engram neurons. Here, we will discuss recent findings related to the roles of interneurons in state-regulated memory processes and synaptic plasticity, and the potential therapeutic implications of understanding these mechanisms.
Collapse
Affiliation(s)
| | - Sara J. Aton
- Department of Molecular, Cellular, and Developmental Biology, College of Literature, Sciences, and the Arts, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
23
|
Ogando MB, Pedroncini O, Federman N, Romano SA, Brum LA, Lanuza GM, Refojo D, Marin-Burgin A. Cholinergic modulation of dentate gyrus processing through dynamic reconfiguration of inhibitory circuits. Cell Rep 2021; 36:109572. [PMID: 34433032 DOI: 10.1016/j.celrep.2021.109572] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 06/28/2021] [Accepted: 07/30/2021] [Indexed: 10/20/2022] Open
Abstract
The dentate gyrus (DG) of the hippocampus plays a key role in memory formation, and it is known to be modulated by septal projections. By performing electrophysiology and optogenetics, we evaluated the role of cholinergic modulation in the processing of afferent inputs in the DG. We show that mature granule cells (GCs), but not adult-born immature neurons, have increased responses to afferent perforant path stimuli upon cholinergic modulation. This is due to a highly precise reconfiguration of inhibitory circuits, differentially affecting Parvalbumin and Somatostatin interneurons, resulting in a nicotinic-dependent perisomatic disinhibition of GCs. This circuit reorganization provides a mechanism by which mature GCs could escape the strong inhibition they receive, creating a window of opportunity for plasticity. Indeed, coincident activation of perforant path inputs with optogenetic release of acetylcholine produces a long-term potentiated response in GCs, essential for memory formation.
Collapse
Affiliation(s)
- Mora B Ogando
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society Godoy Cruz 2390, C1425FQD Buenos Aires, Argentina.
| | - Olivia Pedroncini
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society Godoy Cruz 2390, C1425FQD Buenos Aires, Argentina
| | - Noel Federman
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society Godoy Cruz 2390, C1425FQD Buenos Aires, Argentina
| | - Sebastián A Romano
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society Godoy Cruz 2390, C1425FQD Buenos Aires, Argentina
| | - Luciano A Brum
- Fundación Instituto Leloir-Instituto de Investigaciones Bioquímicas de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Patricias Argentinas 435, Buenos Aires C1405BWE, Argentina
| | - Guillermo M Lanuza
- Fundación Instituto Leloir-Instituto de Investigaciones Bioquímicas de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Patricias Argentinas 435, Buenos Aires C1405BWE, Argentina
| | - Damian Refojo
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society Godoy Cruz 2390, C1425FQD Buenos Aires, Argentina
| | - Antonia Marin-Burgin
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society Godoy Cruz 2390, C1425FQD Buenos Aires, Argentina.
| |
Collapse
|
24
|
Delorme J, Wang L, Kuhn FR, Kodoth V, Ma J, Martinez JD, Raven F, Toth BA, Balendran V, Vega Medina A, Jiang S, Aton SJ. Sleep loss drives acetylcholine- and somatostatin interneuron-mediated gating of hippocampal activity to inhibit memory consolidation. Proc Natl Acad Sci U S A 2021; 118:e2019318118. [PMID: 34344824 PMCID: PMC8364159 DOI: 10.1073/pnas.2019318118] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Sleep loss disrupts consolidation of hippocampus-dependent memory. To characterize effects of learning and sleep loss, we quantified activity-dependent phosphorylation of ribosomal protein S6 (pS6) across the dorsal hippocampus of mice. We find that pS6 is enhanced in dentate gyrus (DG) following single-trial contextual fear conditioning (CFC) but is reduced throughout the hippocampus after brief sleep deprivation (SD; which disrupts contextual fear memory [CFM] consolidation). To characterize neuronal populations affected by SD, we used translating ribosome affinity purification sequencing to identify cell type-specific transcripts on pS6 ribosomes (pS6-TRAP). Cell type-specific enrichment analysis revealed that SD selectively activated hippocampal somatostatin-expressing (Sst+) interneurons and cholinergic and orexinergic hippocampal inputs. To understand the functional consequences of SD-elevated Sst+ interneuron activity, we used pharmacogenetics to activate or inhibit hippocampal Sst+ interneurons or cholinergic input from the medial septum. The activation of either cell population was sufficient to disrupt sleep-dependent CFM consolidation by gating activity in granule cells. The inhibition of either cell population during sleep promoted CFM consolidation and increased S6 phosphorylation among DG granule cells, suggesting their disinhibition by these manipulations. The inhibition of either population across post-CFC SD was insufficient to fully rescue CFM deficits, suggesting that additional features of sleeping brain activity are required for consolidation. Together, our data suggest that state-dependent gating of DG activity may be mediated by cholinergic input and local Sst+ interneurons. This mechanism could act as a sleep loss-driven inhibitory gate on hippocampal information processing.
Collapse
Affiliation(s)
- James Delorme
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Lijing Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Femke Roig Kuhn
- Program in Behavioural and Cognitive Neurosciences, University of Groningen, 9700 AB Groningen, The Netherlands
| | - Varna Kodoth
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Jingqun Ma
- Bioinformatics Core, Biomedical Research Core Facilities, University of Michigan, Ann Arbor, MI 48019
| | - Jessy D Martinez
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Frank Raven
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Brandon A Toth
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Vinodh Balendran
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Alexis Vega Medina
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Sha Jiang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Sara J Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019;
| |
Collapse
|
25
|
Johnston S, Parylak SL, Kim S, Mac N, Lim C, Gallina I, Bloyd C, Newberry A, Saavedra CD, Novak O, Gonçalves JT, Gage FH, Shtrahman M. AAV ablates neurogenesis in the adult murine hippocampus. eLife 2021; 10:e59291. [PMID: 34259630 PMCID: PMC8331179 DOI: 10.7554/elife.59291] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/13/2021] [Indexed: 12/14/2022] Open
Abstract
Recombinant adeno-associated virus (rAAV) has been widely used as a viral vector across mammalian biology and has been shown to be safe and effective in human gene therapy. We demonstrate that neural progenitor cells (NPCs) and immature dentate granule cells (DGCs) within the adult murine hippocampus are particularly sensitive to rAAV-induced cell death. Cell loss is dose dependent and nearly complete at experimentally relevant viral titers. rAAV-induced cell death is rapid and persistent, with loss of BrdU-labeled cells within 18 hr post-injection and no evidence of recovery of adult neurogenesis at 3 months post-injection. The remaining mature DGCs appear hyperactive 4 weeks post-injection based on immediate early gene expression, consistent with previous studies investigating the effects of attenuating adult neurogenesis. In vitro application of AAV or electroporation of AAV2 inverted terminal repeats (ITRs) is sufficient to induce cell death. Efficient transduction of the dentategyrus (DG)- without ablating adult neurogenesis- can be achieved by injection of rAAV2-retro serotyped virus into CA3. rAAV2-retro results in efficient retrograde labeling of mature DGCs and permits in vivo two-photon calcium imaging of dentate activity while leaving adult neurogenesis intact. These findings expand on recent reports implicating rAAV-linked toxicity in stem cells and other cell types and suggest that future work using rAAV as an experimental tool in the DG and as a gene therapy for diseases of the central nervous system should be carefully evaluated.
Collapse
Affiliation(s)
- Stephen Johnston
- Neurosciences Graduate Program, University of California, San DiegoLa JollaUnited States
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
| | - Sarah L Parylak
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
| | - Stacy Kim
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
- Department of Neurosciences, University of California, San DiegoLa JollaUnited States
| | - Nolan Mac
- Department of Biology, University of California, San DiegoLa JollaUnited States
| | - Christina Lim
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
| | - Iryna Gallina
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
| | - Cooper Bloyd
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
| | - Alexander Newberry
- Department of Physics, University of California, San DiegoLa JollaUnited States
| | - Christian D Saavedra
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
| | - Ondrej Novak
- Laboratory of Experimental Epileptology, Department of Physiology, Second Faculty of Medicine, Charles UniversityPragueUnited Kingdom
| | - J Tiago Gonçalves
- Ruth L. and David S. Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of MedicineBronxUnited States
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of MedicineBronxUnited States
| | - Fred H Gage
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
| | - Matthew Shtrahman
- Department of Neurosciences, University of California, San DiegoLa JollaUnited States
| |
Collapse
|
26
|
Reducing glutamic acid decarboxylase in the dorsal dentate gyrus attenuates juvenile stress induced emotional and cognitive deficits. Neurobiol Stress 2021; 15:100350. [PMID: 34150959 PMCID: PMC8193143 DOI: 10.1016/j.ynstr.2021.100350] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/11/2021] [Accepted: 05/28/2021] [Indexed: 11/22/2022] Open
Abstract
A high degree of regional, temporal and molecular specificity is evident in the regulation of GABAergic signaling in stress-responsive circuitry, hampering the use of systemic GABAergic modulators for the treatment of stress-related psychopathology. Here we investigated the effectiveness of local intervention with the GABA synthetic enzymes GAD65 and GAD67 in the dorsal dentate gyrus (dDG) vs ventral DG (vDG) to alleviate anxiety-like behavior and stress-induced symptoms in the rat. We induced shRNA-mediated knock down of either GAD65 or GAD67 with lentiviral vectors microinjected into the dDG or vDG of young adult male rats and examined anxiety behavior, learning and memory performance. Subsequently we tested whether reducing GAD65 expression in the dDG would also confer resilience against juvenile stress-induced behavioral and physiological symptoms in adulthood. While knock down of either isoform in the vDG increased anxiety levels in the open field and the elevated plus maze tests, the knock down of GAD65, but not GAD67, in the dDG conferred a significant reduction in anxiety levels. Strikingly, this manipulation also attenuated juvenile stress evoked anxiety behavior, cognitive and synaptic plasticity impairments. Local GABAergic circuitry in the DG plays an important and highly region-specific role in control of emotional behavior and stress responding. Reduction of GAD65 expression in the dDG appears to provide resilience to juvenile stress-induced emotional and cognitive deficits, opening a new direction towards addressing a significant risk factor for developing stress and trauma-related psychopathologies later in life. GAD67/65 in the dorsal/ventral dentate gyrus differentially modulate anxiety. Reduced GAD65 expression in the dorsal dentate gyrus supports stress resilience. The dorsal dentate gyrus plays a key role in stress resilience.
Collapse
|
27
|
Comeras LB, Hörmer N, Mohan Bethuraj P, Tasan RO. NPY Released From GABA Neurons of the Dentate Gyrus Specially Reduces Contextual Fear Without Affecting Cued or Trace Fear. Front Synaptic Neurosci 2021; 13:635726. [PMID: 34122036 PMCID: PMC8187774 DOI: 10.3389/fnsyn.2021.635726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/20/2021] [Indexed: 11/15/2022] Open
Abstract
Disproportionate, maladapted, and generalized fear are essential hallmarks of posttraumatic stress disorder (PTSD), which develops upon severe trauma in a subset of exposed individuals. Among the brain areas that are processing fear memories, the hippocampal formation exerts a central role linking emotional-affective with cognitive aspects. In the hippocampus, neuronal excitability is constrained by multiple GABAergic interneurons with highly specialized functions and an extensive repertoire of co-released neuromodulators. Neuropeptide Y (NPY) is one of these co-transmitters that significantly affects hippocampal signaling, with ample evidence supporting its fundamental role in emotional, cognitive, and metabolic circuitries. Here we investigated the role of NPY in relation to GABA, both released from the same interneurons of the dorsal dentate gyrus (DG), in different aspects of fear conditioning. We demonstrated that activation of dentate GABA neurons specifically during fear recall reduced cue-related as well as trace-related freezing behavior, whereas inhibition of the same neurons had no significant effects. Interestingly, concomitant overexpression of NPY in these neurons did not further modify fear recall, neither under baseline conditions nor upon chemogenetic stimulation. However, potentially increased co-release of NPY substantially reduced contextual fear, promoted extinction learning, and long-term suppression of fear in a foreground context–conditioning paradigm. Importantly, NPY in the dorsal DG was not only expressed in somatostatin neurons, but also in parvalbumin-positive basket cells and axoaxonic cells, indicating intense feedback and feedforward modulation of hippocampal signaling and precise curtailing of neuronal engrams. Thus, these findings suggest that co-release of NPY from specific interneuron populations of the dorsal DG modifies dedicated aspects of hippocampal processing by sharpening the activation of neural engrams and the consecutive fear response. Since inappropriate and generalized fear is the major impediment in the treatment of PTSD patients, the dentate NPY system may be a suitable access point to ameliorate PTSD symptoms and improve the inherent disease course.
Collapse
Affiliation(s)
- Lucas B Comeras
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Noa Hörmer
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | | | - Ramon O Tasan
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
28
|
Alcantara-Gonzalez D, Chartampila E, Criscuolo C, Scharfman HE. Early changes in synaptic and intrinsic properties of dentate gyrus granule cells in a mouse model of Alzheimer's disease neuropathology and atypical effects of the cholinergic antagonist atropine. Neurobiol Dis 2021; 152:105274. [PMID: 33484828 DOI: 10.1016/j.nbd.2021.105274] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/09/2021] [Accepted: 01/16/2021] [Indexed: 12/19/2022] Open
Abstract
It has been reported that hyperexcitability occurs in a subset of patients with Alzheimer's disease (AD) and hyperexcitability could contribute to the disease. Several studies have suggested that the hippocampal dentate gyrus (DG) may be an important area where hyperexcitability occurs. Therefore, we tested the hypothesis that the principal DG cell type, granule cells (GCs), would exhibit changes at the single-cell level which would be consistent with hyperexcitability and might help explain it. We used the Tg2576 mouse, where it has been shown that hyperexcitability is robust at 2-3 months of age. GCs from 2 to 3-month-old Tg2576 mice were compared to age-matched wild type (WT) mice. Effects of muscarinic cholinergic antagonism were tested because previously we found that Tg2576 mice exhibited hyperexcitability in vivo that was reduced by the muscarinic cholinergic antagonist atropine, counter to the dogma that in AD one needs to boost cholinergic function. The results showed that GCs from Tg2576 mice exhibited increased frequency of spontaneous excitatory postsynaptic potentials/currents (sEPSP/Cs) and reduced frequency of spontaneous inhibitory synaptic events (sIPSCs) relative to WT, increasing the excitation:inhibition (E:I) ratio. There was an inward NMDA receptor-dependent current that we defined here as a novel synaptic current (nsC) in Tg2576 mice because it was very weak in WT mice. Intrinsic properties were distinct in Tg2576 GCs relative to WT. In summary, GCs of the Tg2576 mouse exhibit early electrophysiological alterations that are consistent with increased synaptic excitation, reduced inhibition, and muscarinic cholinergic dysregulation. The data support previous suggestions that the DG contributes to hyperexcitability and there is cholinergic dysfunction early in life in AD mouse models.
Collapse
Affiliation(s)
- David Alcantara-Gonzalez
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | - Elissavet Chartampila
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | - Chiara Criscuolo
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | - Helen E Scharfman
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; Department of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York University Langone Health, New York, NY 10016, USA; Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|
29
|
Fujihara K, Sato T, Miyasaka Y, Mashimo T, Yanagawa Y. Genetic deletion of the 67-kDa isoform of glutamate decarboxylase alters conditioned fear behavior in rats. FEBS Open Bio 2020; 11:340-353. [PMID: 33325157 PMCID: PMC7876494 DOI: 10.1002/2211-5463.13065] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/24/2020] [Accepted: 12/14/2020] [Indexed: 12/31/2022] Open
Abstract
The GABAergic system is thought to play an important role in the control of cognition and emotion, such as fear, and is related to the pathophysiology of psychiatric disorders. For example, the expression of the 67‐kDa isoform of glutamate decarboxylase (GAD67), a GABA‐producing enzyme, is downregulated in the postmortem brains of patients with major depressive disorder and schizophrenia. However, knocking out the Gad1 gene, which encodes GAD67, is lethal in mice, and thus, the association between Gad1 and cognitive/emotional functions is unclear. We recently developed Gad1 knockout rats and found that some of them can grow into adulthood. Here, we performed fear‐conditioning tests in adult Gad1 knockout rats to assess the impact of the loss of Gad1 on fear‐related behaviors and the formation of fear memory. In a protocol assessing both cued and contextual memory, Gad1 knockout rats showed a partial antiphase pattern of freezing during training and significantly excessive freezing during the contextual test compared with wild‐type rats. However, Gad1 knockout rats did not show any synchronous increase in freezing with auditory tones in the cued test. On the other hand, in a contextual memory specialized protocol, Gad1 knockout rats exhibited comparable freezing behavior to wild‐type rats, while their fear extinction was markedly impaired. These results suggest that GABA synthesis by GAD67 has differential roles in cued and contextual fear memory.
Collapse
Affiliation(s)
- Kazuyuki Fujihara
- Departments of Genetic and Behavioral Neuroscience, Graduate School of Medicine, Gunma University, Maebashi, Japan.,Department of Psychiatry and Neuroscience, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Takumi Sato
- Departments of Genetic and Behavioral Neuroscience, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Yoshiki Miyasaka
- Institute of Experimental Animal Sciences, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tomoji Mashimo
- Laboratory Animal Research Center, Institute of Medical Science, the University of Tokyo, Japan
| | - Yuchio Yanagawa
- Departments of Genetic and Behavioral Neuroscience, Graduate School of Medicine, Gunma University, Maebashi, Japan
| |
Collapse
|
30
|
Vergara P, Sakaguchi M. Mechanisms Underlying Memory Consolidation by Adult-Born Neurons During Sleep. Front Cell Neurosci 2020; 14:594401. [PMID: 33324167 PMCID: PMC7726349 DOI: 10.3389/fncel.2020.594401] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/04/2020] [Indexed: 12/17/2022] Open
Abstract
The mammalian hippocampus generates new neurons that incorporate into existing neuronal networks throughout the lifespan, which bestows a unique form of cellular plasticity to the memory system. Recently, we found that hippocampal adult-born neurons (ABNs) that were active during learning reactivate during subsequent rapid eye movement (REM) sleep and provided causal evidence that ABN activity during REM sleep is necessary for memory consolidation. Here, we describe the potential underlying mechanisms by highlighting distinct characteristics of ABNs including decoupled firing from local oscillations and ability to undergo profound synaptic remodeling in response to experience. We further discuss whether ABNs constitute the conventional definition of engram cells by focusing on their active and passive roles in the memory system. This synthesis of evidence helps advance our thinking on the unique mechanisms by which ABNs contribute to memory consolidation.
Collapse
Affiliation(s)
- Pablo Vergara
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Masanori Sakaguchi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
31
|
Albrecht A, Redavide E, Regev-Tsur S, Stork O, Richter-Levin G. Hippocampal GABAergic interneurons and their co-localized neuropeptides in stress vulnerability and resilience. Neurosci Biobehav Rev 2020; 122:229-244. [PMID: 33188820 DOI: 10.1016/j.neubiorev.2020.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/05/2020] [Accepted: 11/01/2020] [Indexed: 12/13/2022]
Abstract
Studies in humans and rodents suggest a critical role for the hippocampal formation in cognition and emotion, but also in the adaptation to stressful events. Successful stress adaptation promotes resilience, while its failure may lead to stress-induced psychopathologies such as depression and anxiety disorders. Hippocampal architecture and physiology is shaped by its strong control of activity via diverse classes of inhibitory interneurons that express typical calcium binding proteins and neuropeptides. Celltype-specific opto- and chemogenetic intervention strategies that take advantage of these biochemical markers have bolstered our understanding of the distinct role of different interneurons in anxiety, fear and stress adaptation. Moreover, some of the signature proteins of GABAergic interneurons have a potent impact on emotion and cognition on their own, making them attractive targets for interventions. In particular, neuropeptide Y is a promising endogenous agent for mediating resilience against severe stress. In this review, we evaluate the role of the major types of interneurons across hippocampal subregions in the adaptation to chronic and acute stress and to emotional memory formation.
Collapse
Affiliation(s)
- Anne Albrecht
- Institute of Anatomy, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany; Center for Behavioral Brain Science, Universitätsplatz 2, 39106 Magdeburg, Germany.
| | - Elisa Redavide
- Institute of Anatomy, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany; Center for Behavioral Brain Science, Universitätsplatz 2, 39106 Magdeburg, Germany; Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany; Institute of Pharmacology and Toxicology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany.
| | - Stav Regev-Tsur
- Sagol Department of Neurobiology, University of Haifa, 199 Aba-Hushi Avenue, 3498838 Haifa, Israel; The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, 199 Aba-Hushi Avenue, 3498838 Haifa, Israel.
| | - Oliver Stork
- Center for Behavioral Brain Science, Universitätsplatz 2, 39106 Magdeburg, Germany; Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany.
| | - Gal Richter-Levin
- Sagol Department of Neurobiology, University of Haifa, 199 Aba-Hushi Avenue, 3498838 Haifa, Israel; The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, 199 Aba-Hushi Avenue, 3498838 Haifa, Israel; Psychology Department, University of Haifa199 Aba-Hushi Avenue, 3498838 Haifa, Israel.
| |
Collapse
|
32
|
Goode TD, Tanaka KZ, Sahay A, McHugh TJ. An Integrated Index: Engrams, Place Cells, and Hippocampal Memory. Neuron 2020; 107:805-820. [PMID: 32763146 PMCID: PMC7486247 DOI: 10.1016/j.neuron.2020.07.011] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/17/2020] [Accepted: 07/13/2020] [Indexed: 01/10/2023]
Abstract
The hippocampus and its extended network contribute to encoding and recall of episodic experiences. Drawing from recent anatomical, physiological, and behavioral studies, we propose that hippocampal engrams function as indices to mediate memory recall. We broaden this idea to discuss potential relationships between engrams and hippocampal place cells, as well as the molecular, cellular, physiological, and circuit determinants of engrams that permit flexible routing of information to intra- and extrahippocampal circuits for reinstatement, a feature critical to memory indexing. Incorporating indexing into frameworks of memory function opens new avenues of study and even therapies for hippocampal dysfunction.
Collapse
Affiliation(s)
- Travis D Goode
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Kazumasa Z Tanaka
- Memory Research Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Kunigami-gun, Okinawa, Japan
| | - Amar Sahay
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| | - Thomas J McHugh
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Wakoshi, Saitama, Japan.
| |
Collapse
|
33
|
Méndez-Couz M, Manahan-Vaughan D, Silva AP, González-Pardo H, Arias JL, Conejo NM. Metaplastic contribution of neuropeptide Y receptors to spatial memory acquisition. Behav Brain Res 2020; 396:112864. [PMID: 32827566 DOI: 10.1016/j.bbr.2020.112864] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/21/2022]
Abstract
Neuropeptide Y (NPY) is highly abundant in the brain and is released as a co-transmitter with plasticity-related neurotransmitters such as glutamate, GABA and noradrenaline. Functionally, its release is associated with appetite, anxiety, and stress regulation. NPY acting on Y2 receptors (Y2R), facilitates fear extinction, suggesting a role in associative memory. Here, we explored to what extent NPY action at Y2R contributes to hippocampus-dependent spatial memory and found that dorsal intrahippocampal receptor antagonism improved spatial reference memory acquired in a water maze in rats, without affecting anxiety levels, or spontaneous motor activity. Water maze training resulted in an increase of Y2R, but not Y1R expression in the hippocampus. By contrast, in the prefrontal cortex there was a decrease in Y2R, and an increase of Y1R expression. Our results indicate that neuropeptide Y2R are significantly involved in hippocampus-dependent spatial memory and that receptor expression is dynamically regulated by this learning experience. Effects are consistent with a metaplastic contribution of NPY receptors to cumulative spatial learning.
Collapse
Affiliation(s)
- Marta Méndez-Couz
- Laboratory of Neuroscience, Department of Psychology, Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Spain; Ruhr University Bochum, Medical Faculty, Dept. Neurophysiology, Bochum, Germany.
| | | | - Ana Paula Silva
- Laboratory of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal
| | - Héctor González-Pardo
- Laboratory of Neuroscience, Department of Psychology, Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Spain
| | - Jorge Luis Arias
- Laboratory of Neuroscience, Department of Psychology, Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Spain
| | - Nélida María Conejo
- Laboratory of Neuroscience, Department of Psychology, Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Spain
| |
Collapse
|
34
|
Regev-Tsur S, Demiray YE, Tripathi K, Stork O, Richter-Levin G, Albrecht A. Region-specific involvement of interneuron subpopulations in trauma-related pathology and resilience. Neurobiol Dis 2020; 143:104974. [PMID: 32561495 DOI: 10.1016/j.nbd.2020.104974] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 02/12/2020] [Accepted: 06/13/2020] [Indexed: 01/22/2023] Open
Abstract
Only a minority of trauma-exposed individuals develops Posttraumatic stress disorder (PTSD) and active processes may support trauma resilience. Individual behavioral profiling allows investigating neurobiological alterations related to resilience or pathology in animal models of PTSD and is utilized here to examine the activation of different interneuron subpopulations of the dentate gyrus-amygdala system associated with trauma resilience or pathology. To model PTSD, rats were exposed to juvenile stress combined with underwater trauma (UWT) in adulthood. Four weeks later, individual anxiety levels were assessed in the elevated plus maze test for classifying rats as highly anxious 'affected' vs. 'non-affected', i.e. behaving as control animals. Analyzing the activation of specific interneuron subpopulations in the dorsal and ventral dentate gyrus (DG), the basolateral (BLA) and central amygdala by immunohistochemical double-labeling for cFos and different interneuron markers, revealed an increased activation of cholecystokinin (CCK)-positive interneurons in the ventral DG, together with increased activation of parvalbumin- and CCK-positive interneurons in the BLA of affected trauma-exposed rats. By contrast, increased activation of neuropeptide Y (NPY)-positive interneurons was observed in the dorsal DG of trauma-exposed, but non-affected rats. To test for a direct contribution of NPY in the dorsal DG to trauma resilience, a local shRNA-mediated knock down was performed after UWT. Such a treatment significantly reduced the prevalence of resilient animals. Our results suggest that distinct interneuron populations are associated with resilience or pathology in PTSD with high regional specificity. NPY within the dorsal DG was found to significantly contribute to trauma resilience.
Collapse
Affiliation(s)
- Stav Regev-Tsur
- Sagol Department of Neurobiology, University of Haifa, Israel; The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, Israel; Psychology Department, University of Haifa, Israel
| | - Yunus Emre Demiray
- Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Germany
| | | | - Oliver Stork
- Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Germany; Center for Behavioral Brain Science, Magdeburg, Germany.
| | - Gal Richter-Levin
- Sagol Department of Neurobiology, University of Haifa, Israel; The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, Israel; Psychology Department, University of Haifa, Israel.
| | - Anne Albrecht
- Sagol Department of Neurobiology, University of Haifa, Israel; The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, Israel; Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Germany; Center for Behavioral Brain Science, Magdeburg, Germany
| |
Collapse
|
35
|
Dail MB, Leach CA, Meek EC, Olivier AK, Pringle RB, Green CE, Chambers JE. Novel Brain-Penetrating Oxime Acetylcholinesterase Reactivators Attenuate Organophosphate-Induced Neuropathology in the Rat Hippocampus. Toxicol Sci 2020; 169:465-474. [PMID: 30835286 DOI: 10.1093/toxsci/kfz060] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Organophosphate (OP) anticholinesterases cause excess acetylcholine leading to seizures which, if prolonged, result in neuronal damage in the rodent brain. Novel substituted phenoxyalkyl pyridinium oximes have previously shown evidence of penetrating the rat blood-brain barrier (BBB) in in vivo tests with a sarin surrogate (nitrophenyl isopropyl methylphosphonate, NIMP) or the active metabolite of the insecticide parathion, paraoxon (PXN), by reducing the time to cessation of seizure-like behaviors and accumulation of glial fibrillary acidic protein, whereas 2-PAM did not. The neuroprotective ability of our lead oximes (15, 20, and 55) was tested using NeuN, Nissl, and Fluoro-Jade B staining in the rat hippocampus. Following lethal-level subcutaneous challenge with NIMP or PXN, rats were intramuscularly administered a novel oxime or 2-PAM plus atropine and euthanized at 4 days. There were statistically significant increases in the median damage scores of the NeuN-stained NIMP, NIMP/2-PAM, and NIMP/Oxime 15 groups compared with the control whereas the scores of the NIMP/Oxime 20 and NIMP/Oxime 55 were not significantly different from the control. The same pattern of statistical significance was observed with PXN. Nissl staining provided a similar pattern, but without statistical differences. Fluoro-Jade B indicated neuroprotection from PXN with novel oximes but not with 2-PAM. The longer blood residence times of Oximes 20 and 55 compared with Oxime 15 might have contributed to their greater efficacy. These results suggest that novel oximes 20 and 55 were able to penetrate the BBB and attenuate neuronal damage after NIMP and PXN exposure, indicating potential broad-spectrum usefulness.
Collapse
Affiliation(s)
- Mary B Dail
- *Department of Basic Sciences, Center for Environmental Health Sciences
| | - Charles A Leach
- *Department of Basic Sciences, Center for Environmental Health Sciences
| | - Edward C Meek
- *Department of Basic Sciences, Center for Environmental Health Sciences
| | - Alicia K Olivier
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi 39762
| | - Ronald B Pringle
- *Department of Basic Sciences, Center for Environmental Health Sciences
| | - Carol E Green
- Biosciences Division, SRI International, Menlo Park, California 94025
| | - Janice E Chambers
- *Department of Basic Sciences, Center for Environmental Health Sciences
| |
Collapse
|
36
|
Lissek S, Klass A, Tegenthoff M. Left Inferior Frontal Gyrus Participates in Mediating the Renewal Effect Irrespective of Context Salience. Front Behav Neurosci 2020; 14:43. [PMID: 32292332 PMCID: PMC7118360 DOI: 10.3389/fnbeh.2020.00043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/13/2020] [Indexed: 11/13/2022] Open
Abstract
The renewal effect of extinction demonstrates the context-dependency of extinction learning. It is defined as the recovery of an extinguished response occurring when the contexts of extinction and recall differ. Behavioral studies showed that modulating context relevance can strengthen context-specific responses. In our fMRI study, we investigated to what extent a modulation of context salience can alter renewal levels and provide additional information about the neural basis for renewal. In a within-subjects design, participants completed two sessions of an associative learning task in randomized order. In the salient condition (SAL), a context was presented alone at the start of each trial, before being presented together with the stimulus. The regular condition (REG) contained no context-alone phase. In about one-third of participants (SWITCH), the context salience modulation significantly increased renewal rates in the SAL compared to the REG condition. The other participants showed either renewal (REN) or no renewal (NoREN) in both conditions. The modulation did not significantly affect learning performance during the initial forming of associations or extinction learning. In the SWITCH group, activation in left opercular inferior frontal gyrus (iFG) during the recall phase was associated with a renewal effect, together with activity in the bilateral posterior hippocampus and ventromedial prefrontal cortex (vmPFC). Also during the extinction phase, left opercular iFG activation was higher in groups exhibiting renewal in recall, irrespective of the context salience modulation. Besides confirming the participation of vmPFC in extinction recall, our findings provide novel insights regarding an as yet undetected, potentially important role for renewal-supporting processes in left iFG during extinction learning and recall, which are presumably based on the region's proposed function of evaluating competing response options under conditions of ambiguity.
Collapse
Affiliation(s)
- Silke Lissek
- Department of Neurology, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | | | | |
Collapse
|
37
|
Çalışkan G, Raza SA, Demiray YE, Kul E, Sandhu KV, Stork O. Depletion of dietary phytoestrogens reduces hippocampal plasticity and contextual fear memory stability in adult male mouse. Nutr Neurosci 2019; 24:951-962. [PMID: 31814540 DOI: 10.1080/1028415x.2019.1698826] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction: Phytoestrogens are non-steroidal estrogen analogues and are found primarily in soy products. They have received increasing attention as dietary supplements for estrogen deficiency and as modulators of endogenous estrogen functions, including cognition and emotion. In addition to modifying the levels of circulating sex hormones, phytoestrogens also exert direct effects on estrogen and androgen receptors in the brain and thus effectively modulate the neural circuit functions.Objective: The aim of this study was to investigate the long-term effects of low phytoestrogen intake (∼6 weeks) on the hippocampal plasticity and hippocampus-dependent memory formation in the adult C57BL/6 male mice.Methods and Results: In comparison to mice on a diet with normal phytoestrogen content, mice on low phytoestrogen diet showed a significant reduction in the phosphorylation of NR2B subunit, a molecular correlate of plasticity in the Schaffer collateral-CA1 synapse. We observed a profound decrease in long-term potentiation (LTP) in the ventral hippocampus, whereas no effect on plasticity was evident in its dorsal portion. Furthermore, we demonstrated that acute perfusion of slices with an estrogen analogue equol, an isoflovane metabolized from daidzein produced by the bacterial flora in the gut, was able to rescue the observed LTP deficit. Examining potential behavioral correlates of the plasticity attenuation, we found that mice on phytoestrogen-free diet display decreased contextual fear memory at remote but not at recent time points after training.Conclusions: Our data suggests that nutritional phytoestrogens have profound effects on the plasticity in the ventral hippocampus and ventral hippocampus-dependent memory.
Collapse
Affiliation(s)
- Gürsel Çalışkan
- Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Syed Ahsan Raza
- Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Yunus E Demiray
- Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Emre Kul
- Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Kiran V Sandhu
- Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Oliver Stork
- Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| |
Collapse
|
38
|
Christian KM, Ming GL, Song H. Adult neurogenesis and the dentate gyrus: Predicting function from form. Behav Brain Res 2019; 379:112346. [PMID: 31722241 DOI: 10.1016/j.bbr.2019.112346] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/05/2019] [Accepted: 11/05/2019] [Indexed: 12/11/2022]
Abstract
Hypotheses about the functional properties of the dentate gyrus and adult dentate neurogenesis have been shaped by early observations of the anatomy of this region, mostly in rodents. This has led to the development of a few core propositions that have guided research over the past several years, including the predicted role of this region in pattern separation and the local transformation of inputs from the entorhinal cortex. We now have the opportunity to review these predictions and update these anatomical observations based on recently developed techniques that reveal the complex structure, connectivity, and dynamic properties of distinct cell populations in the dentate gyrus at a higher resolution. Cumulative evidence suggests that the dentate gyrus and adult-born granule cells play a role in some forms of behavioral discriminations, but there are still many unanswered questions about how the dentate gyrus processes information to support the disambiguation of stimuli.
Collapse
Affiliation(s)
- Kimberly M Christian
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, 19104, USA; Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Guo-Li Ming
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, 19104, USA; Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Developmental and Cell Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, 19104, USA; Institute for Epigenetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongjun Song
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, 19104, USA; Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Developmental and Cell Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Institute for Epigenetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
39
|
Role of adult-born granule cells in the hippocampal functions: Focus on the GluN2B-containing NMDA receptors. Eur Neuropsychopharmacol 2019; 29:1065-1082. [PMID: 31371103 DOI: 10.1016/j.euroneuro.2019.07.135] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/19/2019] [Accepted: 07/15/2019] [Indexed: 02/06/2023]
Abstract
Adult-born granule cells constitute a small subpopulation of the dentate gyrus (DG) in the hippocampus. However, they greatly influence several hippocampus-dependent behaviors, suggesting that adult-born granule cells have specific roles that influence behavior. In order to understand how exactly these adult-born granule cells contribute to behavior, it is critical to understand the underlying electrophysiology and neurochemistry of these cells. Here, this review simultaneously focuses on the specific electrophysiological properties of adult-born granule cells, relying on the GluN2B subunit of NMDA glutamate receptors, and how it influences neurochemistry throughout the brain. Especially in a critical age from 4 to 6 weeks post-division during which they modulate hippocampal functions, adult-born granule cells exhibit a higher intrinsic excitability and an enhanced long-term potentiation. Their stimulation decreases the overall excitation/inhibition balance of the DG via recruitment of local interneurons, and in the CA3 region of the hippocampus. However, the link between neurochemical effects of adult-born granule cells and behavior remain to be further examined.
Collapse
|
40
|
Li D, Jing D, Liu Z, Chen Y, Huang F, Behnisch T. Enhanced Expression of Secreted α-Klotho in the Hippocampus Alters Nesting Behavior and Memory Formation in Mice. Front Cell Neurosci 2019; 13:133. [PMID: 31001090 PMCID: PMC6454015 DOI: 10.3389/fncel.2019.00133] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/18/2019] [Indexed: 12/30/2022] Open
Abstract
The klotho gene family consists of α-, β-, and γ-Klotho, which encode type I single-pass transmembrane proteins with large extracellular domains. α-Klotho exists as a full-length membrane-bound and as a soluble form after cleavage of the extracellular domain. Due to gene splicing, a short extracellular Klotho form can be expressed and secreted. Inactivation of α-Klotho leads to a phenotype that resembles accelerated aging, as the expression level of the α-Klotho protein in the hippocampal formation of mice decreases with age. Here, we show that intrahippocampal viral expression of secreted human α-Klotho alters social behavior and memory formation. Interestingly, overexpression of secreted human α-Klotho in the CA1 changed the nest-building behavior and improved object recognition, object location and passive avoidance memory. Moreover, α-Klotho overexpression increased hippocampal synaptic transmission in response to standardized stimulation strengths, altered paired-pulse facilitation of synaptic transmission, and enhanced activity-dependent synaptic plasticity. These results indicate that memory formation benefits from an augmented level of secreted α-Klotho.
Collapse
Affiliation(s)
- Dongxue Li
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Dongqing Jing
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ziyang Liu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ying Chen
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Fang Huang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Thomas Behnisch
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Çalışkan G, Stork O. Hippocampal network oscillations at the interplay between innate anxiety and learned fear. Psychopharmacology (Berl) 2019; 236:321-338. [PMID: 30417233 DOI: 10.1007/s00213-018-5109-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 11/05/2018] [Indexed: 12/14/2022]
Abstract
The hippocampus plays a central role as a hub for episodic memory and as an integrator of multimodal sensory information in time and space. Thereby, it critically determines contextual setting and specificity of episodic memories. It is also a key site for the control of innate anxiety states and involved in psychiatric diseases with heightened anxiety and generalized fear memory such as post-traumatic stress disorder (PTSD). Expression of both innate "unlearned" anxiety and "learned" fear requires contextual processing and engagement of a brain-wide network including the hippocampus together with the amygdala and medial prefrontal cortex. Strikingly, the hippocampus is also the site of emergence of oscillatory rhythms that coordinate information processing and filtering in this network. Here, we review data on how the hippocampal network oscillations and their coordination with amygdalar and prefrontal oscillations are engaged in innate threat evaluation. We further explore how such innate oscillatory communication might have an impact on contextualization and specificity of "learned" fear. We illustrate the partial overlap of fear and anxiety networks that are built by the hippocampus in conjunction with amygdala and prefrontal cortex. We further propose that (mal)-adaptive interplay via (dis)-balanced oscillatory communication between the anxiety network and the fear network may determine the strength of fear memories and their resistance to extinction.
Collapse
Affiliation(s)
- Gürsel Çalışkan
- Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, Universitätsplatz 2, 39106, Magdeburg, Germany.
| | - Oliver Stork
- Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Universitätsplatz 2, 39106, Magdeburg, Germany
| |
Collapse
|
42
|
Richter-Levin G, Stork O, Schmidt MV. Animal models of PTSD: a challenge to be met. Mol Psychiatry 2019; 24:1135-1156. [PMID: 30816289 PMCID: PMC6756084 DOI: 10.1038/s41380-018-0272-5] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 08/13/2018] [Accepted: 09/11/2018] [Indexed: 02/07/2023]
Abstract
Recent years have seen increased interest in psychopathologies related to trauma exposure. Specifically, there has been a growing awareness to posttraumatic stress disorder (PTSD) in part due to terrorism, climate change-associated natural disasters, the global refugee crisis, and increased violence in overpopulated urban areas. However, notwithstanding the increased awareness to the disorder, the increasing number of patients, and the devastating impact on the lives of patients and their families, the efficacy of available treatments remains limited and highly unsatisfactory. A major scientific effort is therefore devoted to unravel the neural mechanisms underlying PTSD with the aim of paving the way to developing novel or improved treatment approaches and drugs to treat PTSD. One of the major scientific tools used to gain insight into understanding physiological and neuronal mechanisms underlying diseases and for treatment development is the use of animal models of human diseases. While much progress has been made using these models in understanding mechanisms of conditioned fear and fear memory, the gained knowledge has not yet led to better treatment options for PTSD patients. This poor translational outcome has already led some scientists and pharmaceutical companies, who do not in general hold opinions against animal models, to propose that those models should be abandoned. Here, we critically examine aspects of animal models of PTSD that may have contributed to the relative lack of translatability, including the focus on the exposure to trauma, overlooking individual and sex differences, and the contribution of risk factors. Based on findings from recent years, we propose research-based modifications that we believe are required in order to overcome some of the shortcomings of previous practice. These modifications include the usage of animal models of PTSD which incorporate risk factors and of the behavioral profiling analysis of individuals in a sample. These modifications are aimed to address factors such as individual predisposition and resilience, thus taking into consideration the fact that only a fraction of individuals exposed to trauma develop PTSD. We suggest that with an appropriate shift of practice, animal models are not only a valuable tool to enhance our understanding of fear and memory processes, but could serve as effective platforms for understanding PTSD, for PTSD drug development and drug testing.
Collapse
Affiliation(s)
- Gal Richter-Levin
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel. .,The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, Haifa, Israel. .,Psychology Department, University of Haifa, Haifa, Israel.
| | - Oliver Stork
- 0000 0001 1018 4307grid.5807.aDepartment of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany ,grid.452320.2Center for Behavioral Brain Sciences, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Mathias V. Schmidt
- 0000 0000 9497 5095grid.419548.5Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
43
|
Hippocampal NPY Y2 receptors modulate memory depending on emotional valence and time. Neuropharmacology 2018; 143:20-28. [DOI: 10.1016/j.neuropharm.2018.09.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/29/2018] [Accepted: 09/11/2018] [Indexed: 12/13/2022]
|
44
|
Pilly PK, Howard MD, Bhattacharyya R. Modeling Contextual Modulation of Memory Associations in the Hippocampus. Front Hum Neurosci 2018; 12:442. [PMID: 30473660 PMCID: PMC6237880 DOI: 10.3389/fnhum.2018.00442] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/12/2018] [Indexed: 11/13/2022] Open
Abstract
We present a computational model of how memories can be contextually acquired and recalled in the hippocampus. Our adaptive contextual memory model comprises the lateral entorhinal cortex (LEC), the dentate gyrus (DG) and areas CA3 and CA1 in the hippocampus, and assumes external inputs about context that originate in the prefrontal cortex (PFC). Specifically, we propose that there is a top-down bias on the excitability of cells in the DG of the hippocampus that recruits a sub-population of cells to differentiate contexts, independent of experienced stimuli, expanding the "pattern separation" role typically attributed to the DG. It has been demonstrated in rats that if PFC is inactivated, both acquisition and recall of memory associations are impaired. However, PFC inactivation during acquisition of one set of memory associations surprisingly leads to subsequent facilitation of the acquisition of a conflicting set of memory associations in the same context under normal PFC operation. We provide here the first computational and algorithmic account of how the absence or presence of the top-down contextual biases on the excitability of DG cells during different learning phases of these experiments explains these data. Our model simulates PFC inactivation as the loss of inhibitory control on DG, which leads to full or partial activation of DG cells related to conflicting memory associations previously acquired in different contexts. This causes context-inappropriate memory traces to become active in the CA3 recurrent network and thereby the output CA1 area within the hippocampus. We show that these incongruous memory patterns proactively interfere with and slow the acquisition of new memory associations. Further, we demonstrate that pattern completion within CA3 in response to a partial cue for the recall of previously acquired memories is also impaired by PFC inactivation for the same reason. Pre-training the model with interfering memories in contexts different from those used in the experiments, simulating a lifetime of experiences, was crucial to reproduce the rat behavioral data. Finally, we made several testable predictions based on the model that suggest future experiments to deepen our understanding of brain-wide memory processes.
Collapse
Affiliation(s)
- Praveen K Pilly
- Center for Human-Machine Collaboration, Information and Systems Sciences Laboratory, HRL Laboratories Malibu, CA, United States
| | - Michael D Howard
- Center for Human-Machine Collaboration, Information and Systems Sciences Laboratory, HRL Laboratories Malibu, CA, United States
| | - Rajan Bhattacharyya
- Center for Human-Machine Collaboration, Information and Systems Sciences Laboratory, HRL Laboratories Malibu, CA, United States
| |
Collapse
|
45
|
Çalışkan G, Stork O. Hippocampal network oscillations as mediators of behavioural metaplasticity: Insights from emotional learning. Neurobiol Learn Mem 2018; 154:37-53. [PMID: 29476822 DOI: 10.1016/j.nlm.2018.02.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 02/13/2018] [Accepted: 02/19/2018] [Indexed: 01/15/2023]
Abstract
Behavioural metaplasticity is evident in experience-dependent changes of network activity patterns in neuronal circuits that connect the hippocampus, amygdala and medial prefrontal cortex. These limbic regions are key structures of a brain-wide neural network that translates emotionally salient events into persistent and vivid memories. Communication in this network by-and-large depends on behavioural state-dependent rhythmic network activity patterns that are typically generated and/or relayed via the hippocampus. In fact, specific hippocampal network oscillations have been implicated to the acquisition, consolidation and retrieval, as well as the reconsolidation and extinction of emotional memories. The hippocampal circuits that contribute to these network activities, at the same time, are subject to both Hebbian and non-Hebbian forms of plasticity during memory formation. Further, it has become evident that adaptive changes in the hippocampus-dependent network activity patterns provide an important means of adjusting synaptic plasticity. We here summarise our current knowledge on how these processes in the hippocampus in interaction with amygdala and medial prefrontal cortex mediate the formation and persistence of emotional memories.
Collapse
Affiliation(s)
- Gürsel Çalışkan
- Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany.
| | - Oliver Stork
- Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Universitätsplatz 2, 39106 Magdeburg, Germany
| |
Collapse
|