1
|
Kim SB, Mun BR, Kim SY, Elangovan M, Park EJ, Choi WS, Park WJ. Therapeutic effects of a novel synthetic α-secretase. Front Aging Neurosci 2024; 16:1383905. [PMID: 38912519 PMCID: PMC11191342 DOI: 10.3389/fnagi.2024.1383905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/08/2024] [Indexed: 06/25/2024] Open
Abstract
Excessive accumulation of amyloid-β (Aβ) has been associated with the pathogenesis of Alzheimer's disease (AD). Clinical studies have further proven that elimination of Aβ can be a viable therapeutic option. In the current study, we conceptualized a fusion membrane protein, referred to as synthetic α-secretase (SAS), that can cleave amyloid precursor protein (APP) and Aβ specifically at the α-site. In mammalian cells, SAS indeed cleaved APP and Aβ at the α-site. Overexpression of SAS in the hippocampus was achieved by direct injection of recombinant adeno-associated virus serotype 9 (AAV9) that expresses SAS (AAV9-SAS) into the bilateral ventricles of mouse brains. SAS enhanced the non-amyloidogenic processing of APP, thus reducing the levels of soluble Aβ and plaques in the 5xFAD mice. In addition, SAS significantly attenuated the cognitive deficits in 5xFAD mice, as demonstrated by novel object recognition and Morris water maze tests. Unlike other Aβ-cleaving proteases, SAS has highly strict substrate specificity. We propose that SAS can be an efficient modality to eliminate excessive Aβ from diseased brains.
Collapse
Affiliation(s)
- Sung Bin Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Bo-Ram Mun
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Sung Yoon Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Muthukumar Elangovan
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Euy Jun Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Won-Seok Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Woo Jin Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| |
Collapse
|
2
|
Vasudevan K, Hassell JE, Maren S. Hippocampal Engrams and Contextual Memory. ADVANCES IN NEUROBIOLOGY 2024; 38:45-66. [PMID: 39008010 PMCID: PMC12006847 DOI: 10.1007/978-3-031-62983-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Memories are not formed in a vacuum and often include rich details about the time and place in which events occur. Contextual stimuli promote the retrieval of events that have previously occurred in the encoding context and limit the retrieval of context-inappropriate information. Contexts that are associated with traumatic or harmful events both directly elicit fear and serve as reminders of aversive events associated with trauma. It has long been appreciated that the hippocampus is involved in contextual learning and memory and is central to contextual fear conditioning. However, little is known about the underlying neuronal mechanisms underlying the encoding and retrieval of contextual fear memories. Recent advancements in neuronal labeling methods, including activity-dependent tagging of cellular ensembles encoding memory ("engrams"), provide unique insight into the neural substrates of memory in the hippocampus. Moreover, these methods allow for the selective manipulation of memory ensembles. Attenuating or erasing fear memories may have considerable therapeutic value for patients with post-traumatic stress disorder or other trauma- or stressor-related conditions. In this chapter, we review the role of the hippocampus in contextual fear conditioning in rodents and explore recent work implicating hippocampal ensembles in the encoding and retrieval of aversive memories.
Collapse
Affiliation(s)
- Krithika Vasudevan
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, College Station, TX, USA
| | - James E Hassell
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, College Station, TX, USA
| | - Stephen Maren
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
3
|
Mansk LMZ, Jaimes LF, Dias TL, Pereira GS. Social recognition memory differences between mouse strains: On the effects of social isolation, adult neurogenesis, and environmental enrichment. Brain Res 2023; 1819:148535. [PMID: 37595660 DOI: 10.1016/j.brainres.2023.148535] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 07/25/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
Remembering conspecifics is paramount for the establishment and maintenance of groups. Here we asked whether the variability in social behavior caused by different breeding strategies affects social recognition memory (SRM). We tested the hypothesis that the inbred Swiss and the outbred C57BL/6 mice behave differently on SRM. Social memory in C57BL/6 mice endured at least 14 days, while in Swiss mice lasted 24 h but not ten days. We showed previously that an enriched environment enhanced the persistence of SRM in Swiss mice. Here we reproduced this result and added that it also increases the survival of adult-born neurons in the hippocampus. Next, we tested whether prolonged SRM observed in C57BL/6 mice could be changed by diminishing the trial duration or using an interference stimulus after learning. Neither short acquisition time nor interference during consolidation affected it. However, social isolation impaired SRM in C57BL/6 mice, similar to what was previously observed in Swiss mice. Our results demonstrate that SRM expression can vary according to the mouse strain, which shows the importance of considering this variable when choosing the most suitable model to answer specific questions about this memory system. We also demonstrate the suitability of both C57BL/6 and Swiss strains for exploring the impact of environmental conditions and adult neurogenesis on social memory.
Collapse
Affiliation(s)
- Lara M Z Mansk
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Laura F Jaimes
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thomaz L Dias
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Grace S Pereira
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
4
|
Froula JM, Hastings SD, Krook-Magnuson E. The little brain and the seahorse: Cerebellar-hippocampal interactions. Front Syst Neurosci 2023; 17:1158492. [PMID: 37034014 PMCID: PMC10076554 DOI: 10.3389/fnsys.2023.1158492] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
There is a growing appreciation for the cerebellum beyond its role in motor function and accumulating evidence that the cerebellum and hippocampus interact across a range of brain states and behaviors. Acute and chronic manipulations, simultaneous recordings, and imaging studies together indicate coordinated coactivation and a bidirectional functional connectivity relevant for various physiological functions, including spatiotemporal processing. This bidirectional functional connectivity is likely supported by multiple circuit paths. It is also important in temporal lobe epilepsy: the cerebellum is impacted by seizures and epilepsy, and modulation of cerebellar circuitry can be an effective strategy to inhibit hippocampal seizures. This review highlights some of the recent key hippobellum literature.
Collapse
Affiliation(s)
- Jessica M. Froula
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | | | | |
Collapse
|
5
|
Eiden LE, Hernández VS, Jiang SZ, Zhang L. Neuropeptides and small-molecule amine transmitters: cooperative signaling in the nervous system. Cell Mol Life Sci 2022; 79:492. [PMID: 35997826 PMCID: PMC11072502 DOI: 10.1007/s00018-022-04451-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/15/2022] [Accepted: 06/23/2022] [Indexed: 12/17/2022]
Abstract
Neuropeptides are expressed in cell-specific patterns throughout mammalian brain. Neuropeptide gene expression has been useful for clustering neurons by phenotype, based on single-cell transcriptomics, and for defining specific functional circuits throughout the brain. How neuropeptides function as first messengers in inter-neuronal communication, in cooperation with classical small-molecule amine transmitters (SMATs) is a current topic of systems neurobiology. Questions include how neuropeptides and SMATs cooperate in neurotransmission at the molecular, cellular and circuit levels; whether neuropeptides and SMATs always co-exist in neurons; where neuropeptides and SMATs are stored in the neuron, released from the neuron and acting, and at which receptors, after release; and how neuropeptides affect 'classical' transmitter function, both directly upon co-release, and indirectly, via long-term regulation of gene transcription and neuronal plasticity. Here, we review an extensive body of data about the distribution of neuropeptides and their receptors, their actions after neuronal release, and their function based on pharmacological and genetic loss- and gain-of-function experiments, that addresses these questions, fundamental to understanding brain function, and development of neuropeptide-based, and potentially combinatorial peptide/SMAT-based, neurotherapeutics.
Collapse
Affiliation(s)
- Lee E Eiden
- Section On Molecular Neuroscience, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 49 Convent Drive, Room 5A38, Bethesda, MD, 20892, USA.
| | - Vito S Hernández
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Sunny Z Jiang
- Section On Molecular Neuroscience, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 49 Convent Drive, Room 5A38, Bethesda, MD, 20892, USA
| | - Limei Zhang
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico.
| |
Collapse
|
6
|
Super-resolved 3D-STED microscopy identifies a layer-specific increase in excitatory synapses in the hippocampal CA1 region of Neuroligin-3 KO mice. Biochem Biophys Res Commun 2021; 582:144-149. [PMID: 34715405 DOI: 10.1016/j.bbrc.2021.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/02/2021] [Indexed: 12/18/2022]
Abstract
The chemical synapse is one type of cell-adhesion system that transmits information from a neuron to another neuron in the complex neuronal network in the brain. Synaptic transmission is the rate-limiting step during the information processing in the neuronal network and its plasticity is involved in cognitive functions. Thus, morphological and electrophysiological analyses of synapses are of particular importance in neuroscience research. In the current study, we applied super-resolved three-dimensional stimulated emission depletion (3D-STED) microscopy for the morphological analyses of synapses. This approach allowed us to estimate the precise number of excitatory and inhibitory synapses in the mouse hippocampal tissue. We discovered a region-specific increase in excitatory synapses in a model mouse of autism spectrum disorder, Neuroligin-3 KO, with this method. This type of analysis will open a new field in developmental neuroscience in the future.
Collapse
|
7
|
Distinct functions of ventral CA1 and dorsal CA2 in social memory. Curr Opin Neurobiol 2021; 68:29-35. [PMID: 33421771 DOI: 10.1016/j.conb.2020.12.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW For animals that live in social groups, the ability to recognize conspecifics is essential. Recent studies of both human patients and animal models have vigorously sought to discern the precise mechanisms by which hippocampal neurons and neural circuits contribute to the encoding, consolidation, storage, and retrieval of social memory. In particular, optogenetic manipulation enables us to investigate the presence of memory engrams. RECENT FINDINGS We recently revealed the presence of social memory engrams in hippocampal ventral CA1 neurons, using optogenetic manipulation and calcium (Ca2+) imaging. SUMMARY In the present manuscript, we discuss the current viewpoints on two hippocampal subregions in regards to social memory representation, namely dorsal CA2 for information processing and ventral CA1 for the storage of social memory, specifically from the perspectives of behavioral neuroscience and neurophysiology.
Collapse
|
8
|
Silkis IG, Markevich VA. Possible Mechanisms of the Influence of the Supramillary Nucleus on the Functioning of the Dentate Gyrus and the CA2 Field of the Hippocamsus (Role of Disinhibition). NEUROCHEM J+ 2020. [DOI: 10.1134/s181971242004011x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
9
|
Navarro JF, Croteau DL, Jurek A, Andrusivova Z, Yang B, Wang Y, Ogedegbe B, Riaz T, Støen M, Desler C, Rasmussen LJ, Tønjum T, Galas MC, Lundeberg J, Bohr VA. Spatial Transcriptomics Reveals Genes Associated with Dysregulated Mitochondrial Functions and Stress Signaling in Alzheimer Disease. iScience 2020; 23:101556. [PMID: 33083725 PMCID: PMC7522123 DOI: 10.1016/j.isci.2020.101556] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/24/2020] [Accepted: 09/09/2020] [Indexed: 12/15/2022] Open
Abstract
Alzheimer disease (AD) is a devastating neurological disease associated with progressive loss of mental skills and cognitive and physical functions whose etiology is not completely understood. Here, our goal was to simultaneously uncover novel and known molecular targets in the structured layers of the hippocampus and olfactory bulbs that may contribute to early hippocampal synaptic deficits and olfactory dysfunction in AD mice. Spatially resolved transcriptomics was used to identify high-confidence genes that were differentially regulated in AD mice relative to controls. A diverse set of genes that modulate stress responses and transcription were predominant in both hippocampi and olfactory bulbs. Notably, we identify Bok, implicated in mitochondrial physiology and cell death, as a spatially downregulated gene in the hippocampus of mouse and human AD brains. In summary, we provide a rich resource of spatially differentially expressed genes, which may contribute to understanding AD pathology. Spatial transcriptomics identifies differentially expressed genes with spatial patterns Early application of spatial transcriptomics to olfactory bulbs from AD models Bok gene is spatially differentially expressed in AD mouse and patient brains Paip1 and Homer1 genes are regulated in a PolB-dependent manner
Collapse
Affiliation(s)
- José Fernández Navarro
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, 17165 Stockholm, Sweden
| | - Deborah L Croteau
- Laboratory of Molecular Gerontology, National Institute on Aging, Baltimore, MD 21224, USA
| | - Aleksandra Jurek
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, 17165 Stockholm, Sweden
| | - Zaneta Andrusivova
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, 17165 Stockholm, Sweden
| | - Beimeng Yang
- Laboratory of Molecular Gerontology, National Institute on Aging, Baltimore, MD 21224, USA
| | - Yue Wang
- Laboratory of Molecular Gerontology, National Institute on Aging, Baltimore, MD 21224, USA
| | - Benjamin Ogedegbe
- Laboratory of Molecular Gerontology, National Institute on Aging, Baltimore, MD 21224, USA
| | - Tahira Riaz
- Unit for Genome Dynamics, Department of Microbiology, University of Oslo and Oslo University Hospital, 0372 Oslo, Norway
| | - Mari Støen
- Unit for Genome Dynamics, Department of Microbiology, University of Oslo and Oslo University Hospital, 0372 Oslo, Norway
| | - Claus Desler
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Lene Juel Rasmussen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Tone Tønjum
- Unit for Genome Dynamics, Department of Microbiology, University of Oslo and Oslo University Hospital, 0372 Oslo, Norway
| | - Marie-Christine Galas
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, 59000 Lille, France
| | - Joakim Lundeberg
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, 17165 Stockholm, Sweden
| | - Vilhelm A Bohr
- Laboratory of Molecular Gerontology, National Institute on Aging, Baltimore, MD 21224, USA.,Unit for Genome Dynamics, Department of Microbiology, University of Oslo and Oslo University Hospital, 0372 Oslo, Norway
| |
Collapse
|
10
|
Abstract
Contemporary brain research seeks to understand how cognition is reducible to neural activity. Crucially, much of this effort is guided by a scientific paradigm that views neural activity as essentially driven by external stimuli. In contrast, recent perspectives argue that this paradigm is by itself inadequate and that understanding patterns of activity intrinsic to the brain is needed to explain cognition. Yet, despite this critique, the stimulus-driven paradigm still dominates-possibly because a convincing alternative has not been clear. Here, we review a series of findings suggesting such an alternative. These findings indicate that neural activity in the hippocampus occurs in one of three brain states that have radically different anatomical, physiological, representational, and behavioral correlates, together implying different functional roles in cognition. This three-state framework also indicates that neural representations in the hippocampus follow a surprising pattern of organization at the timescale of ∼1 s or longer. Lastly, beyond the hippocampus, recent breakthroughs indicate three parallel states in the cortex, suggesting shared principles and brain-wide organization of intrinsic neural activity.
Collapse
Affiliation(s)
- Kenneth Kay
- Howard Hughes Medical Institute, Kavli Institute for Fundamental Neuroscience, Department of Physiology, University of California San Francisco, San Francisco, California
| | - Loren M Frank
- Howard Hughes Medical Institute, Kavli Institute for Fundamental Neuroscience, Department of Physiology, University of California San Francisco, San Francisco, California
| |
Collapse
|
11
|
Cinar E, Yalcin-Cakmakli G, Saka E, Ulusoy A, Yuruker S, Elibol B, Tel BC. Modelling cognitive deficits in Parkinson's disease: Is CA2 a gateway for hippocampal synucleinopathy? Exp Neurol 2020; 330:113357. [PMID: 32437708 DOI: 10.1016/j.expneurol.2020.113357] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 04/01/2020] [Accepted: 05/05/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Cognitive dysfunction is one of the most disabling non-motor symptoms of Parkinson's disease (PD), though its pathological correlates still remain elusive. Hippocampal Lewy pathology has recently been correlated by compelling evidence from post-mortem and imaging studies. Animal models recapitulating cognitive impairment in PD are essential to better understand the underlying pathophysiology. To investigate the hippocampal involvement in cognitive dysfunction of PD, we generated an experimental model by inducing midbrain and hippocampal α-synuclein pathology simultaneously. METHODS Rats were injected either with human α-synuclein or green fluorescent protein (GFP) expressing adeno-associated viral vectors (AAV), or saline bilaterally into substantia nigra (SN) and dentate gyrus (DG). A group of untreated animals were used as naïve controls. Cognitive and behavioral changes were evaluated with tests probing for spatial learning, short-term memory, anxiety and hedonistic behavior. Immunohistochemical staining, immunoblotting and stereological analysis were performed for pathological characterization. RESULTS Bilateral α-synuclein overexpression in SN and DG led to mild but significant motor impairment as well as dysfunctions in short-term memory and spatial learning. There was no hedonistic deficit, whereas a hypo-anxious state was induced. While stereological analysis revealed no significant neuronal loss in any sectors of cornu ammonis, there was considerable decrease (43%) in TH+-neurons in SN pars compacta supporting the well-known vulnerability of nigral dopaminergic neurons to α-synuclein mediated neurodegeneration. On the other hand, synaptophysin levels decreased in similar amounts both in striatum and hippocampus, suggesting comparable synaptic loss in target areas. Interestingly, phosphorylated-S129-α-synuclein staining revealed significant expression in CA2 characterized by more mature and dense cellular accumulations compared to CA1-CA3 sub-regions displaying more diffuse grain-like aggregates, suggesting preferential susceptibility of CA2 to produce α-synuclein induced pathology. CONCLUSION Bilateral α-synuclein overexpression in DG and SN reproduced partial motor and hippocampus related cognitive deficits. Using this model, we showed a predisposition of CA2 for pathological α-synuclein accumulation, which may provide further insights for future experimental and clinical studies.
Collapse
Affiliation(s)
- Elif Cinar
- Department of Pharmacology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.
| | | | - Esen Saka
- Department of Neurology, Hacettepe University, Sihhiye, Ankara, Turkey
| | - Ayse Ulusoy
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Sinan Yuruker
- Usak University Faculty of Medicine, Department of Histology and Embryology, Usak, Turkey
| | - Bulent Elibol
- Department of Neurology, Hacettepe University, Sihhiye, Ankara, Turkey
| | - Banu C Tel
- Department of Pharmacology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
12
|
Dasgupta A, Lim YJ, Kumar K, Baby N, Pang KLK, Benoy A, Behnisch T, Sajikumar S. Group III metabotropic glutamate receptors gate long-term potentiation and synaptic tagging/capture in rat hippocampal area CA2. eLife 2020; 9:e55344. [PMID: 32310084 PMCID: PMC7170650 DOI: 10.7554/elife.55344] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/08/2020] [Indexed: 12/17/2022] Open
Abstract
Metabotropic glutamate receptors (mGluRs) play an important role in synaptic plasticity and memory and are largely classified based on amino acid sequence homology and pharmacological properties. Among group III metabotropic glutamate receptors, mGluR7 and mGluR4 show high relative expression in the rat hippocampal area CA2. Group III metabotropic glutamate receptors are known to down-regulate cAMP-dependent signaling pathways via the activation of Gi/o proteins. Here, we provide evidence that inhibition of group III mGluRs by specific antagonists permits an NMDA receptor- and protein synthesis-dependent long-lasting synaptic potentiation in the apparently long-term potentiation (LTP)-resistant Schaffer collateral (SC)-CA2 synapses. Moreover, long-lasting potentiation of these synapses transforms a transient synaptic potentiation of the entorhinal cortical (EC)-CA2 synapses into a stable long-lasting LTP, in accordance with the synaptic tagging/capture hypothesis (STC). Furthermore, this study also sheds light on the role of ERK/MAPK protein signaling and the downregulation of STEP protein in the group III mGluR inhibition-mediated plasticity in the hippocampal CA2 region, identifying them as critical molecular players. Thus, the regulation of group III mGluRs provides a conducive environment for the SC-CA2 synapses to respond to events that could lead to activity-dependent synaptic plasticity.
Collapse
Affiliation(s)
- Ananya Dasgupta
- Department of Physiology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Life Sciences Institute Neurobiology Programme, National University of SingaporeSingaporeSingapore
| | - Yu Jia Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
| | - Krishna Kumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Life Sciences Institute Neurobiology Programme, National University of SingaporeSingaporeSingapore
| | - Nimmi Baby
- Department of Physiology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Life Sciences Institute Neurobiology Programme, National University of SingaporeSingaporeSingapore
| | - Ka Lam Karen Pang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Life Sciences Institute Neurobiology Programme, National University of SingaporeSingaporeSingapore
| | - Amrita Benoy
- Department of Physiology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Life Sciences Institute Neurobiology Programme, National University of SingaporeSingaporeSingapore
| | - Thomas Behnisch
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan UniversityShanghaiChina
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Life Sciences Institute Neurobiology Programme, National University of SingaporeSingaporeSingapore
| |
Collapse
|
13
|
Tzakis N, Holahan MR. Social Memory and the Role of the Hippocampal CA2 Region. Front Behav Neurosci 2019; 13:233. [PMID: 31632251 PMCID: PMC6779725 DOI: 10.3389/fnbeh.2019.00233] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/17/2019] [Indexed: 01/02/2023] Open
Abstract
The CA2 region of the hippocampus is a somewhat obscure area lacking in an understanding of its form and function. Until recently, the CA2 has been thought of as merely an extension of the CA3, with some referring to it as the CA3a region. Recent investigations into this area have not only delineated the CA2, but also defined it as a region distinct from both CA1 and CA3, with a unique set of cortical inputs and outputs and contributions to cognitive processes. One such process that has been shown to depend on the CA2 is the ability to recognize a novel or familiar conspecific, known as social recognition memory. Here, we review these findings and discuss the parallels between CA2 dysfunction and social impairments.
Collapse
Affiliation(s)
- Nikolaos Tzakis
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | | |
Collapse
|
14
|
Yang J, Zhang L, Cao LL, Qi J, Li P, Wang XP, Sun XL. MicroRNA-99a is a Potential Target for Regulating Hypothalamic Synaptic Plasticity in the Peri/Postmenopausal Depression Model. Cells 2019; 8:cells8091081. [PMID: 31540304 PMCID: PMC6769887 DOI: 10.3390/cells8091081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 12/28/2022] Open
Abstract
Accumulating evidence has demonstrated that there is a growing trend of menopausal women suffering from depression. However, the pathogenesis of menopausal depression still remains unclear. Hence, this paper aims to reveal the pathological mechanisms involved in postmenopausal depression by using a novel peri- to postmenopausal depression model induced by a two-step ovariectomy plus chronic mild stress (CMS). The results of metabolic chambers and serum hormone/cytokine determination revealed that peri/postmenopausal depressive mice exhibited endocrine and metabolic disorders. Electrophysiological recordings indicated that the hippocampal synaptic transmission was compromised. Compared to the sham group, the microRNA-99a (miR-99a) level decreased significantly in the hypothalamus, and its target FK506-binding protein 51 (FKBP51) enormously increased; in contrast, the nuclear translocation of the progesterone receptor (PR) decreased in hypothalamic paraventricular nucleus (PVN) in the peri/postmenopausal depression mouse model. Additionally, synaptic proteins, including postsynaptic density protein 95 (PSD-95) and synaptophysin (SYN), showed a similar decrease in the hypothalamus. Accordingly, the present work suggests that miR-99a may be involved in the regulation of hypothalamic synaptic plasticity and that it might be a potential therapeutic target for peri/postmenopausal depression.
Collapse
Affiliation(s)
- Jin Yang
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China.
| | - Ling Zhang
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China.
| | - Lu-Lu Cao
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China.
| | - Jun Qi
- Jiangsu Vocational College of Nursing, Huaian 223001, China.
| | - Ping Li
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China.
| | - Xi-Peng Wang
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China.
| | - Xiu-Lan Sun
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China.
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
15
|
Charlton PE, Schatz KC, Burke K, Paul MJ, Dent ML. Sex differences in auditory brainstem response audiograms from vasopressin-deficient Brattleboro and wild-type Long-Evans rats. PLoS One 2019; 14:e0222096. [PMID: 31469871 PMCID: PMC6716658 DOI: 10.1371/journal.pone.0222096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/21/2019] [Indexed: 11/18/2022] Open
Abstract
Rats are highly social creatures that produce ultrasonic vocalizations (USVs) during social interactions. Brattleboro rats, a Long-Evans derived rat that lacks vasopressin (AVP) due to a mutation in the Avp gene, exhibit atypical social behavior, including fewer USVs with altered spectrotemporal characteristics during social interactions. It is unclear why Brattleboro rats produce atypical USVs, but one factor could be differences in auditory acuity between them and wild-type Long Evans rats with functional vasopressin. Previous studies have suggested a link between increased levels of AVP and auditory processing. Additionally, few studies have investigated sex differences in auditory perception by Long-Evans rats. Sex differences in auditory acuity have been found throughout the animal kingdom, but have not yet been demonstrated in rat audiograms. This study aimed to measure auditory brainstem response (ABR) derived audiograms for frequencies ranging from 1 to 64 kHz in male and female homozygous Brattleboro (Hom), heterozygous Brattleboro (Het), and wild-type (WT) Long-Evans rats to better understand the role of AVP and sex differences in auditory processing by these rats. We failed to detect significant differences between the ABR audiograms of Hom, Het, and WT Long-Evans rats, suggesting that varying levels of AVP do not affect auditory processing. Interestingly, males and females of all genotypes did differ in their ABR thresholds, with males exhibiting higher thresholds than females. The sex differences in auditory acuity were significant at the lowest and highest frequencies, possibly affecting the perception of USVs. These are the first known sex differences in rat audiograms.
Collapse
Affiliation(s)
- Payton E. Charlton
- Department of Psychology, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Kelcie C. Schatz
- Department of Psychology, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Kali Burke
- Department of Psychology, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Matthew J. Paul
- Department of Psychology, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Micheal L. Dent
- Department of Psychology, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
16
|
Amidfar M, Woelfer M, Réus GZ, Quevedo J, Walter M, Kim YK. The role of NMDA receptor in neurobiology and treatment of major depressive disorder: Evidence from translational research. Prog Neuropsychopharmacol Biol Psychiatry 2019; 94:109668. [PMID: 31207274 DOI: 10.1016/j.pnpbp.2019.109668] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/24/2019] [Accepted: 06/11/2019] [Indexed: 12/16/2022]
Abstract
There is accumulating evidence demonstrating that dysfunction of glutamatergic neurotransmission, particularly via N-methyl-d-aspartate (NMDA) receptors, is involved in the pathophysiology of major depressive disorder (MDD). Several studies have revealed an altered expression of NMDA receptor subtypes and impaired NMDA receptor-mediated intracellular signaling pathways in brain circuits of patients with MDD. Clinical studies have demonstrated that NMDA receptor antagonists, particularly ketamine, have rapid antidepressant effects in treatment-resistant depression, however, neurobiological mechanisms are not completely understood. Growing body of evidence suggest that signal transduction pathways involved in synaptic plasticity play critical role in molecular mechanisms underlying rapidly acting antidepressant properties of ketamine and other NMDAR antagonists in MDD. Discovering the molecular mechanisms underlying the unique antidepressant actions of ketamine will facilitate the development of novel fast acting antidepressants which lack undesirable effects of ketamine. This review provides a critical examination of the NMDA receptor involvement in the neurobiology of MDD including analyses of alterations in NMDA receptor subtypes and their interactive signaling cascades revealed by postmortem studies. Furthermore, to elucidate mechanisms underlying rapid-acting antidepressant properties of NMDA receptor antagonists we discussed their effects on the neuroplasticity, mostly based on signaling systems involved in synaptic plasticity of mood-related neurocircuitries.
Collapse
Affiliation(s)
| | - Marie Woelfer
- Clinical Affective Neuroimaging Laboratory, University Magdeburg, Germany; New Jersey Institute of Technology, Newark, NJ, USA
| | - Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil; Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Martin Walter
- Clinical Affective Neuroimaging Laboratory, University Magdeburg, Germany; Department of Psychiatry, University Tuebingen, Germany
| | - Yong-Ku Kim
- Department of Psychiatry, College of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
17
|
Chaaya N, Jacques A, Belmer A, Beecher K, Ali SA, Chehrehasa F, Battle AR, Johnson LR, Bartlett SE. Contextual Fear Conditioning Alter Microglia Number and Morphology in the Rat Dorsal Hippocampus. Front Cell Neurosci 2019; 13:214. [PMID: 31139053 PMCID: PMC6527886 DOI: 10.3389/fncel.2019.00214] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 04/29/2019] [Indexed: 12/11/2022] Open
Abstract
Contextual fear conditioning is a Pavlovian conditioning paradigm capable of rapidly creating fear memories to contexts, such as rooms or chambers. Contextual fear conditioning protocols have long been utilized to evaluate how fear memories are consolidated, maintained, expressed, recalled, and extinguished within the brain. These studies have identified the lateral portion of the amygdala and the dorsal portion of the hippocampus as essential for contextual fear memory consolidation. The current study was designed to evaluate how two different contextual fear memories alter amygdala and hippocampus microglia, brain derived neurotrophic factor (BDNF), and phosphorylated cyclic-AMP response element binding (pCREB). We find rats provided with standard contextual fear conditioning to have more microglia and more cells expressing BDNF in the dentate gyrus as compared to a context only control group. Additionally, standard contextual fear conditioning altered microglia morphology to become amoeboid in shape – a common response to central nervous system insult, such as traumatic brain injury, infection, ischemia, and more. The unpaired fear conditioning procedure (whereby non-reinforced and non-overlapping auditory tones were provided at random intervals during conditioning), despite producing equivalent levels of fear as the standard procedure, did not alter microglia, BDNF or pCREB number in any dorsal hippocampus or lateral amygdala brain regions. Despite this, the unpaired fear conditioning protocol produced some alterations in microglia morphology, but less compared to rats provided with standard contextual fear conditioning. Results from this study demonstrate that contextual fear conditioning is capable of producing large alterations to dentate gyrus plasticity and microglia, whereas unpaired fear conditioning only produces minor changes to microglia morphology. These data show, for the first time, that Pavlovian fear conditioning protocols can induce similar responses as trauma, infection or other insults within the central nervous system.
Collapse
Affiliation(s)
- Nicholas Chaaya
- School of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Angela Jacques
- School of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Arnauld Belmer
- School of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kate Beecher
- School of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Syed A Ali
- School of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Fatemeh Chehrehasa
- Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia.,School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Andrew R Battle
- Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia.,School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Luke R Johnson
- Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia.,School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD, Australia.,Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University School of Medicine, Bethesda, MD, United States
| | - Selena E Bartlett
- School of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
18
|
Jacob S, Sumathi T. Extenuation of in utero toxic effects of MeHg in the developing neurons by Fisetin via modulating the expression of synaptic transmission and plasticity regulators in hippocampus of the rat offspring. Chem Biol Interact 2019; 305:3-10. [PMID: 30890323 DOI: 10.1016/j.cbi.2019.03.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/13/2019] [Indexed: 12/27/2022]
Abstract
The neurotoxic environmental contaminant, methylmercury (MeHg), has shown to have detrimental effects on the developing brain when exposed during gestation. We have shown in our earlier studies that gestational administration of 3,3',4',7-Tetrahydroxyflavone or Fisetin reduces the toxic effects of MeHg in the developing rat brain. The current study has pivoted to study the mechanism behind the mitigating action of Fisetin against prenatal MeHg exposure induced neurotoxicity. Negligible data is available about the toxicity targets of MeHg in the developing brain. Studies have exhibited that MeHg exposure cause toxic effects on synaptic transmission and plasticity in the offspring brain. Hence, we aimed to study the effect of Fisetin on MeHg induced alterations in the expressions of regulatory genes and proteins involved in synaptic plasticity and transmission. Pregnant rats were grouped according to the type of oral administration as, (i) Control, (ii) MeHg (1.5 mg/kg b. w.), (iii) MeHg + Fisetin (30 mg/kg b. w.) and (iv) Fisetin (30 mg/kg b. w). Maternal administration of Fisetin prevented MeHg exposure induced downregulation of neurogranin (Nrgn), dendrin (Ddn), Syntaxin 1 A (Stx1a), Lin-7 homolog A (Lin7a), Complexin-2 (Cplx2) and Exocyst complex component 8 (Exoc8) genes in the offspring rat. Fisetin also prevented MeHg exposure induced downregulation of brain derived neurotrophic factor (BDNF), Glial-cell derived neurotrophic factor (GDNF) protein expressions and hampered reactive astrogliosis in the hippocampus of F1 generation rats. Hence, through this study, we conclude that Fisetin modulates the expression of regulatory genes and proteins involved in synaptic transmission and plasticity and extenuates MeHg neurotoxicity in the developing rat brain.
Collapse
Affiliation(s)
- Sherin Jacob
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, 600113, Tamil Nadu, India
| | - Thangarajan Sumathi
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, 600113, Tamil Nadu, India.
| |
Collapse
|
19
|
Muñoz MD, Solís JM. Characterisation of the mechanisms underlying the special sensitivity of the CA2 hippocampal area to adenosine receptor antagonists. Neuropharmacology 2019; 144:9-18. [DOI: 10.1016/j.neuropharm.2018.10.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/17/2018] [Accepted: 10/12/2018] [Indexed: 12/28/2022]
|
20
|
Lopatina OL, Komleva YK, Gorina YV, Olovyannikova RY, Trufanova LV, Hashimoto T, Takahashi T, Kikuchi M, Minabe Y, Higashida H, Salmina AB. Oxytocin and excitation/inhibition balance in social recognition. Neuropeptides 2018; 72:1-11. [PMID: 30287150 DOI: 10.1016/j.npep.2018.09.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 09/18/2018] [Accepted: 09/18/2018] [Indexed: 12/15/2022]
Abstract
Social recognition is the sensitive domains of complex behavior critical for identification, interpretation and storage of socially meaningful information. Social recognition develops throughout childhood and adolescent, and is affected in a wide variety of psychiatric disorders. Recently, new data appeared on the molecular mechanisms of these processes, particularly, the excitatory-inhibitory (E/I) ratio which is modified during development, and then E/I balance is established in the adult brain. While E/I imbalance has been proposed as a mechanism for schizophrenia, it also seems to be the common mechanism in autism spectrum disorder (ASD). In addition, there is a strong suggestion that the oxytocinergic system is related to GABA-mediated E/I control in the context of brain socialization. In this review, we attempt to summarize the underpinning molecular mechanisms of E/I balance and its imbalance, and related biomarkers in the brain in healthiness and pathology. In addition, because there are increasing interest on oxytocin in the social neuroscience field, we will pay intensive attention to the role of oxytocin in maintaining E/I balance from the viewpoint of its effects on improving social impairment in psychiatric diseases, especially in ASD.
Collapse
Affiliation(s)
- Olga L Lopatina
- Depatment of Biochemistry, Medical, Pharmaceutical & Toxicological Chemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk 660022, Russia; Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa 920-8640, Japan
| | - Yulia K Komleva
- Depatment of Biochemistry, Medical, Pharmaceutical & Toxicological Chemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk 660022, Russia
| | - Yana V Gorina
- Depatment of Biochemistry, Medical, Pharmaceutical & Toxicological Chemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk 660022, Russia
| | - Raisa Ya Olovyannikova
- Depatment of Biochemistry, Medical, Pharmaceutical & Toxicological Chemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk 660022, Russia
| | - Lyudmila V Trufanova
- Depatment of Biochemistry, Medical, Pharmaceutical & Toxicological Chemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk 660022, Russia
| | - Takanori Hashimoto
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa 920-8640, Japan
| | - Tetsuya Takahashi
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa 920-8640, Japan
| | - Mitsuru Kikuchi
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa 920-8640, Japan
| | - Yoshio Minabe
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa 920-8640, Japan
| | - Haruhiro Higashida
- Depatment of Biochemistry, Medical, Pharmaceutical & Toxicological Chemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk 660022, Russia; Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa 920-8640, Japan
| | - Alla B Salmina
- Depatment of Biochemistry, Medical, Pharmaceutical & Toxicological Chemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk 660022, Russia; Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa 920-8640, Japan.
| |
Collapse
|
21
|
Pang CCC, Kiecker C, O'Brien JT, Noble W, Chang RCC. Ammon's Horn 2 (CA2) of the Hippocampus: A Long-Known Region with a New Potential Role in Neurodegeneration. Neuroscientist 2018; 25:167-180. [PMID: 29865938 DOI: 10.1177/1073858418778747] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The hippocampus has a critical role in cognition and human memory and is one of the most studied structures in the brain. Despite more than 400 years of research, little is known about the Ammon's horn region cornu ammonis 2 (CA2) subfield in comparison to other subfield regions (CA1, CA3, and CA4). Recent findings have shown that CA2 plays a bigger role than previously thought. Here, we review understanding of hippocampus and CA2 ontogenesis, together with basic and clinical findings about the potential role of this region in neurodegenerative disease. The CA2 has widespread anatomical connectivity, unique signaling molecules, and intrinsic electrophysiological properties. Experimental studies using in vivo models found that the CA2 region has a role in cognition, especially in social memory and object recognition. In models of epilepsy and hypoxia, the CA2 exhibits higher resilience to cell death and hypoxia in comparison with neighboring regions, and while hippocampal atrophy remains poorly understood in Parkinson's disease (PD) and dementia with Lewy bodies (DLB), findings from postmortem PD brain demonstrates clear accumulation of α-synuclein pathology in CA2, and the CA2-CA3 region shows relatively more atrophy compared with other hippocampal subfields. Taken together, there is a growing body of evidence suggesting that the CA2 can be an ideal hallmark with which to differentiate different neurodegenerative stages of PD. Here, we summarize these recent data and provide new perspectives/ideas for future investigations to unravel the contribution of the CA2 to neurodegenerative diseases.
Collapse
Affiliation(s)
- Cindy Chi-Ching Pang
- 1 Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,2 Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Clemens Kiecker
- 3 Department of Developmental Neurobiology, King's College London, London, UK
| | - John T O'Brien
- 4 Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Wendy Noble
- 2 Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Raymond Chuen-Chung Chang
- 1 Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,5 State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
22
|
Evans PR, Parra-Bueno P, Smirnov MS, Lustberg DJ, Dudek SM, Hepler JR, Yasuda R. RGS14 Restricts Plasticity in Hippocampal CA2 by Limiting Postsynaptic Calcium Signaling. eNeuro 2018; 5:ENEURO.0353-17.2018. [PMID: 29911178 PMCID: PMC6001268 DOI: 10.1523/eneuro.0353-17.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 04/06/2018] [Accepted: 04/30/2018] [Indexed: 01/17/2023] Open
Abstract
Pyramidal neurons in hippocampal area CA2 are distinct from neighboring CA1 in that they resist synaptic long-term potentiation (LTP) at CA3 Schaffer collateral synapses. Regulator of G protein signaling 14 (RGS14) is a complex scaffolding protein enriched in CA2 dendritic spines that naturally blocks CA2 synaptic plasticity and hippocampus-dependent learning, but the cellular mechanisms by which RGS14 gates LTP are largely unexplored. A previous study has attributed the lack of plasticity to higher rates of calcium (Ca2+) buffering and extrusion in CA2 spines. Additionally, a recent proteomics study revealed that RGS14 interacts with two key Ca2+-activated proteins in CA2 neurons: calcium/calmodulin and CaMKII. Here, we investigated whether RGS14 regulates Ca2+ signaling in its host CA2 neurons. We found that the nascent LTP of CA2 synapses caused by genetic knockout (KO) of RGS14 in mice requires Ca2+-dependent postsynaptic signaling through NMDA receptors, CaMK, and PKA, revealing similar mechanisms to those in CA1. We report that RGS14 negatively regulates the long-term structural plasticity of dendritic spines of CA2 neurons. We further show that wild-type (WT) CA2 neurons display significantly attenuated spine Ca2+ transients during structural plasticity induction compared with the Ca2+ transients from CA2 spines of RGS14 KO mice and CA1 controls. Finally, we demonstrate that acute overexpression of RGS14 is sufficient to block spine plasticity, and elevating extracellular Ca2+ levels restores plasticity to RGS14-expressing neurons. Together, these results demonstrate for the first time that RGS14 regulates plasticity in hippocampal area CA2 by restricting Ca2+ elevations in CA2 spines and downstream signaling pathways.
Collapse
Affiliation(s)
- Paul R. Evans
- Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322
| | | | | | - Daniel J. Lustberg
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Serena M. Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - John R. Hepler
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322
| | - Ryohei Yasuda
- Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458
| |
Collapse
|
23
|
Okuyama T. Social memory engram in the hippocampus. Neurosci Res 2018; 129:17-23. [DOI: 10.1016/j.neures.2017.05.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/21/2017] [Accepted: 05/25/2017] [Indexed: 01/10/2023]
|
24
|
Zhang Y, Fang X, Fan W, Tang W, Cai J, Song L, Zhang C. Brain-derived neurotrophic factor as a biomarker for cognitive recovery in acute schizophrenia: 12-week results from a prospective longitudinal study. Psychopharmacology (Berl) 2018; 235:1191-1198. [PMID: 29392373 DOI: 10.1007/s00213-018-4835-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 01/14/2018] [Indexed: 12/22/2022]
Abstract
RATIONALE It is generally accepted that impaired cognitive function is a core feature of schizophrenia. There is evidence for the role of brain-derived neurotrophic factor (BDNF) in cognitive function. Olanzapine was reported to yield cognitive improvement in patients with schizophrenia. OBJECTIVES In this study, we performed a prospective, open-label, 12-week observation trial to investigate whether peripheral BDNF may represent a potential biomarker for the effect of cognitive improvement induced by olanzapine in patients with schizophrenia. METHODS In total, 95 patients with acute schizophrenia were enrolled in the study. We also recruited 72 healthy individuals for a control group. The Positive and Negative Syndrome Scale (PANSS) was used to evaluate symptom severity and treatment response. Cognitive function was evaluated using the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS). Plasma BDNF levels were measured with an enzyme-linked immunosorbent assay. RESULTS Of the 95 patients consented into the study, 68 completed the 12-week follow up. Our results showed that schizophrenia patients with acute exacerbation had significantly poorer performance than that of the controls (Ps < 0.01). A significantly decreased plasma level of BDNF in patients was observed compared with the controls (F = 7.77, P = 0.006). A significant improvement in each PANSS subscore and total score was observed when the patients completed this study (Ps < 0.01). Additionally, 12-week olanzapine treatment exhibited significant improvements in RBANS immediate memory, attention, and total scores (P = 0.018, 0.001, and 0.007, respectively). Along with the clinical improvement, plasma BDNF levels after 12-week olanzapine monotherapy (4.67 ± 1.74 ng/ml) were also significantly increased compared with those at baseline (3.38 ± 2.11 ng/ml) (P < 0.01). Spearman's correlation analysis showed that the increase in plasma levels of BDNF is significantly correlated with the change in the RBANS total scores (r = 0.28, P = 0.02) but not with the change in the PANSS total scores (r = - 0.18, P = 0.13). There is a significant correlation of BDNF increase with the change of RBANS attention subscore (r = 0.27, P = 0.028). CONCLUSIONS Our findings suggest that olanzapine improves psychiatric symptoms and cognitive dysfunction, particularly attention and immediate memory, in patients with acute schizophrenia, in parallel with increased plasma BDNF levels. Plasma BDNF levels may be a potential biomarker for cognitive recovery in acute schizophrenia.
Collapse
Affiliation(s)
- Yi Zhang
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyu Fang
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weixing Fan
- Department of Psychiatry, Jinhua Second Hospital, Jinhua, Zhejiang, China
| | - Wei Tang
- Department of Psychiatry, Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jun Cai
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lisheng Song
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Zhang
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
25
|
Hippocampal area CA2: properties and contribution to hippocampal function. Cell Tissue Res 2018; 373:525-540. [PMID: 29335778 DOI: 10.1007/s00441-017-2769-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 12/07/2017] [Indexed: 12/30/2022]
Abstract
This review focuses on area CA2 of the hippocampus, as recent results have revealed the unique properties and surprising role of this region in encoding social, temporal and contextual aspects of memory. Originally identified and described by Lorente de No, in 1934, this region of the hippocampus has unique intra-and extra-hippocampal connectivity, sending and receiving input to septal and hypothalamic regions. Recent in vivo studies have indicated that CA2 pyramidal neurons encode spatial information during immobility and play an important role in the generation of sharp-wave ripples. Furthermore, CA2 neurons act to control overall excitability in the hippocampal network and have been found to be consistently altered in psychiatric diseases, indicating that normal function of this region is necessary for normal cognition. With its unique role, area CA2 has a unique molecular profile, interneuron density and composition. Furthermore, this region has an unusual manifestation of synaptic plasticity that does not occur post-synaptically at pyramidal neuron dendrities but through the local network of inhibitory neurons. While much progress has recently been made in understanding the large contribution of area CA2 to social memory formation, much still needs to be learned.
Collapse
|
26
|
Akinyemi RO, Allan LM, Oakley A, Kalaria RN. Hippocampal Neurodegenerative Pathology in Post-stroke Dementia Compared to Other Dementias and Aging Controls. Front Neurosci 2017; 11:717. [PMID: 29311794 PMCID: PMC5742173 DOI: 10.3389/fnins.2017.00717] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 12/08/2017] [Indexed: 12/14/2022] Open
Abstract
Neuroimaging evidence from older stroke survivors in Nigeria and Northeast England showed medial temporal lobe atrophy (MTLA) to be independently associated with post-stroke cognitive impairment and dementia. Given the hypothesis ascribing MTLA to neurodegenerative processes, we assessed Alzheimer pathology in the hippocampal formation and entorhinal cortex of autopsied brains from of post-stroke demented and non-demented subjects in comparison with controls and other dementias. We quantified markers of amyloid β (total Aβ, Aβ-40, Aβ-42, and soluble Aβ) and hyperphosphorylated tau in the hippocampal formation and entorhinal cortex of 94 subjects consisting of normal controls (n = 12), vascular dementia, VaD (17), post-stroke demented, PSD (n = 15), and post-stroke non-demented, PSND (n = 23), Alzheimer's disease, AD (n = 14), and mixed AD and vascular dementia, AD_VAD (n = 13) using immunohistochemical techniques. We found differential expression of amyloid and tau across the disease groups, and across hippocampal sub-regions. Among amyloid markers, the pattern of Aβ-42 immunoreactivity was similar to that of total Aβ. Tau immunoreactivity showed highest expression in the AD and mixed AD and vascular dementia, AD_VaD, which was higher than in control, post - stroke and VaD groups (p < 0.05). APOE ε4 allele positivity was associated with higher expression of amyloid and tau pathology in the subiculum and entorhinal cortex of post-stroke cases (p < 0.05). Comparison between PSND and PSD revealed higher total Aβ immunoreactivity in PSND compared to PSD in the CA1, subiculum and entorhinal cortex (p < 0.05) but no differences between PSND and PSD in Aβ-42, Aβ-40, soluble Aβ or tau immunoreactivities (p > 0.05). Correlation of MMSE and CAMCOG scores with AD pathological measures showed lack of correlation with amyloid species although tau immunoreactivity demonstrated correlation with memory scores (p < 0.05). Our findings suggest hippocampal AD pathology does not necessarily differ between demented and non-demented post-stroke subjects. The dissociation of cognitive performance with hippocampal AD pathological burden suggests more dominant roles for non-Alzheimer neurodegenerative and / or other non-neurodegenerative substrates for dementia following stroke.
Collapse
Affiliation(s)
- Rufus O Akinyemi
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Oyo, Nigeria.,Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Louise M Allan
- Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Arthur Oakley
- Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rajesh N Kalaria
- Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
27
|
Perineuronal Nets Suppress Plasticity of Excitatory Synapses on CA2 Pyramidal Neurons. J Neurosci 2017; 36:6312-20. [PMID: 27277807 DOI: 10.1523/jneurosci.0245-16.2016] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 05/02/2016] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Long-term potentiation of excitatory synapses on pyramidal neurons in the stratum radiatum rarely occurs in hippocampal area CA2. Here, we present evidence that perineuronal nets (PNNs), a specialized extracellular matrix typically localized around inhibitory neurons, also surround mouse CA2 pyramidal neurons and envelop their excitatory synapses. CA2 pyramidal neurons express mRNA transcripts for the major PNN component aggrecan, identifying these neurons as a novel source for PNNs in the hippocampus. We also found that disruption of PNNs allows synaptic potentiation of normally plasticity-resistant excitatory CA2 synapses; thus, PNNs play a role in restricting synaptic plasticity in area CA2. Finally, we found that postnatal development of PNNs on CA2 pyramidal neurons is modified by early-life enrichment, suggesting that the development of circuits containing CA2 excitatory synapses are sensitive to manipulations of the rearing environment. SIGNIFICANCE STATEMENT Perineuronal nets (PNNs) are thought to play a major role in restricting synaptic plasticity during postnatal development, and are altered in several models of neurodevelopmental disorders, such as schizophrenia and Rett syndrome. Although PNNs have been predominantly studied in association with inhibitory neurons throughout the brain, we describe a dense expression of PNNs around excitatory pyramidal neurons in hippocampal area CA2. We also provide insight into a previously unrecognized role for PNNs in restricting plasticity at excitatory synapses and raise the possibility of an early critical period of hippocampal plasticity that may ultimately reveal a key mechanism underlying learning and memory impairments of PNN-associated neurodevelopmental disorders.
Collapse
|
28
|
Ko J. Neuroanatomical Substrates of Rodent Social Behavior: The Medial Prefrontal Cortex and Its Projection Patterns. Front Neural Circuits 2017; 11:41. [PMID: 28659766 PMCID: PMC5468389 DOI: 10.3389/fncir.2017.00041] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/29/2017] [Indexed: 12/30/2022] Open
Abstract
Social behavior encompasses a number of distinctive and complex constructs that form the core elements of human imitative culture, mainly represented as either affiliative or antagonistic interactions with conspecifics. Traditionally considered in the realm of psychology, social behavior research has benefited from recent advancements in neuroscience that have accelerated identification of the neural systems, circuits, causative genes and molecular mechanisms that underlie distinct social cognitive traits. In this review article, I summarize recent findings regarding the neuroanatomical substrates of key social behaviors, focusing on results from experiments conducted in rodent models. In particular, I will review the role of the medial prefrontal cortex (mPFC) and downstream subcortical structures in controlling social behavior, and discuss pertinent future research perspectives.
Collapse
Affiliation(s)
- Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST)Daegu, South Korea
| |
Collapse
|
29
|
Differential Expression and Cell-Type Specificity of Perineuronal Nets in Hippocampus, Medial Entorhinal Cortex, and Visual Cortex Examined in the Rat and Mouse. eNeuro 2017; 4:eN-NWR-0379-16. [PMID: 28593193 PMCID: PMC5461557 DOI: 10.1523/eneuro.0379-16.2017] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 05/18/2017] [Accepted: 05/18/2017] [Indexed: 12/21/2022] Open
Abstract
Perineuronal nets (PNNs) are specialized extracellular matrix (ECM) structures that condense around the soma and proximal dendrites of subpopulations of neurons. Emerging evidence suggests that they are involved in regulating brain plasticity. However, the expression of PNNs varies between and within brain areas. A lack of quantitative studies describing the distribution and cell-specificity of PNNs makes it difficult to reveal the functional roles of PNNs. In the current study, we examine the distribution of PNNs and the identity of PNN-enwrapped neurons in three brain areas with different cognitive functions: the dorsal hippocampus, medial entorhinal cortex (mEC) and primary visual cortex (V1). We compared rats and mice as knowledge from these species are often intermingled. The most abundant expression of PNNs was found in the mEC and V1, while dorsal hippocampus showed strikingly low levels of PNNs, apart from dense expression in the CA2 region. In hippocampus we also found apparent species differences in expression of PNNs. While we confirm that the PNNs enwrap parvalbumin-expressing (PV+) neurons in V1, we found that they mainly colocalize with excitatory CamKII-expressing neurons in CA2. In mEC, we demonstrate that in addition to PV+ cells, the PNNs colocalize with reelin-expressing stellate cells. We also show that the maturation of PNNs in mEC coincides with the formation of grid cell pattern, while PV+ cells, unlike in other cortical areas, are present from early postnatal development. Finally, we demonstrate considerable effects on the number of PSD-95-gephyrin puncta after enzymatic removal of PNNs.
Collapse
|
30
|
Zemla R, Basu J. Hippocampal function in rodents. Curr Opin Neurobiol 2017; 43:187-197. [PMID: 28477511 DOI: 10.1016/j.conb.2017.04.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 04/11/2017] [Accepted: 04/12/2017] [Indexed: 11/16/2022]
Abstract
The hippocampus is crucial for the formation and recall of long-term memories about people, places, objects, and events. Capitalizing on high-resolution microscopy, in vivo electrophysiology, and genetic manipulation, recent research in rodents provides evidence for hippocampal ensemble coding on the spatial, episodic, and contextual dimensions. Here we highlight the functional contribution of newly described long-range connections between hippocampus and cortical areas, and the relative impact of inhibitory and excitatory dynamics in generating behaviorally relevant population activity. Our goal is to provide an integrated view of hippocampal circuit function to understand mnemonic computations at the systems and cellular levels that underlie adaptive learned behaviors.
Collapse
Affiliation(s)
- Roland Zemla
- Neuroscience Institute, New York University School of Medicine, USA; Medical Scientist Training Program, New York University School of Medicine, USA
| | - Jayeeta Basu
- Neuroscience Institute, New York University School of Medicine, USA; Department of Neuroscience and Physiology, New York University School of Medicine, USA.
| |
Collapse
|
31
|
Jagannath V, Marinova Z, Monoranu CM, Walitza S, Grünblatt E. Expression of D-Amino Acid Oxidase ( DAO/ DAAO) and D-Amino Acid Oxidase Activator ( DAOA/G72) during Development and Aging in the Human Post-mortem Brain. Front Neuroanat 2017; 11:31. [PMID: 28428746 PMCID: PMC5382383 DOI: 10.3389/fnana.2017.00031] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 03/24/2017] [Indexed: 12/30/2022] Open
Abstract
In the brain, D-amino acid oxidase (DAO/DAAO) mainly oxidizes D-serine, a co-agonist of the N-methyl-D-aspartate (NMDA) receptors. Thus, DAO can regulate the function of NMDA receptors via D-serine breakdown. Furthermore, DAO activator (DAOA)/G72 has been reported as both DAOA and repressor. The co-expression of DAO and DAOA genes and proteins in the human brain is not yet elucidated. The aim of this study was to understand the regional and age span distribution of DAO and DAOA (mRNA and protein) in a concomitant manner. We determined DAO and DAOA mRNA and protein expression across six brain regions in normal human post-mortem brain samples (16 weeks of gestation to 91 years) using quantitative real-time reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay. We found higher expression of DAO mRNA in the cerebellum, whereas lower expression of DAO protein in the cerebellum compared to the other brain regions studied, which suggests post-transcriptional regulation. We detected DAOA protein but not DAOA mRNA in all brain regions studied, suggesting a tightly regulated expression. To understand this regulation at the transcriptional level, we analyzed DNA methylation levels at DAO and DAOA CpG sites in the cerebellum and frontal cortex of control human post-mortem brain obtained from Gene Expression Omnibus datasets. Indeed, DAO and DAOA CpG sites in the cerebellum were significantly more methylated than those in the frontal cortex. While investigating lifespan effects, we found that DAO mRNA levels were positively correlated with age <2 years in the cerebellum and amygdala. We also detected a significant positive correlation (controlled for age) between DAO and DAOA protein in all of the brain regions studied except for the frontal cortex. In summary, DAO and DAOA expression in the human brain are both age and brain region dependent.
Collapse
Affiliation(s)
- Vinita Jagannath
- Molecular and Neurobiochemistry Laboratory, Centre for Child and Adolescent Psychiatry Research, Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of ZurichZurich, Switzerland
| | - Zoya Marinova
- Molecular and Neurobiochemistry Laboratory, Centre for Child and Adolescent Psychiatry Research, Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of ZurichZurich, Switzerland
| | - Camelia-Maria Monoranu
- Department of Neuropathology, Institute of Pathology, University of WürzburgWürzburg, Germany
| | - Susanne Walitza
- Molecular and Neurobiochemistry Laboratory, Centre for Child and Adolescent Psychiatry Research, Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of ZurichZurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH ZurichZurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of ZurichZurich, Switzerland
| | - Edna Grünblatt
- Molecular and Neurobiochemistry Laboratory, Centre for Child and Adolescent Psychiatry Research, Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of ZurichZurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH ZurichZurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of ZurichZurich, Switzerland
| |
Collapse
|
32
|
Guarana (Paullinia cupana) ameliorates memory impairment and modulates acetylcholinesterase activity in Poloxamer-407-induced hyperlipidemia in rat brain. Physiol Behav 2017; 168:11-19. [DOI: 10.1016/j.physbeh.2016.10.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 09/21/2016] [Accepted: 10/04/2016] [Indexed: 01/18/2023]
|
33
|
Hippocampal α-Synuclein in Dementia with Lewy Bodies Contributes to Memory Impairment and Is Consistent with Spread of Pathology. J Neurosci 2016; 37:1675-1684. [PMID: 28039370 DOI: 10.1523/jneurosci.3047-16.2016] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/07/2016] [Accepted: 12/11/2016] [Indexed: 11/21/2022] Open
Abstract
Despite considerable research to uncover them, the anatomic and neuropathologic correlates of memory impairment in dementia with Lewy bodies (DLB) remain unclear. While some studies have implicated Lewy bodies in the neocortex, others have pointed to α-synuclein pathology in the hippocampus. We systematically examined hippocampal Lewy pathology and its distribution in hippocampal subfields in 95 clinically and neuropathologically characterized human cases of DLB, finding that α-synuclein pathology was highest in two hippocampal-related subregions: the CA2 subfield and the entorhinal cortex (EC). While the EC had numerous classic somatic Lewy bodies, CA2 contained mainly Lewy neurites in presumed axon terminals, suggesting the involvement of the EC → CA2 circuitry in the pathogenesis of DLB symptoms. Clinicopathological correlations with measures of verbal and visual memory supported a role for EC Lewy pathology, but not CA2, in causing these memory deficits. Lewy pathology in CA1-the main output region for CA2-correlated best with results from memory testing despite a milder pathology. This result indicates that CA1 may be more functionally relevant than CA2 in the context of memory impairment in DLB. These correlations remained significant after controlling for several factors, including concurrent Alzheimer's pathology (neuritic plaques and neurofibrillary tangles) and the interval between time of testing and time of death. Our data suggest that although hippocampal Lewy pathology in DLB is predominant in CA2 and EC, memory performance correlates most strongly with CA1 burden.SIGNIFICANCE STATEMENT This study provides a detailed neuropathologic analysis of hippocampal Lewy pathology in human patients with autopsy-confirmed dementia with Lewy bodies. The approach-informed by regional molecular markers, concurrent Alzheimer's pathology analysis, and relevant clinical data-helps tease out the relative contribution of Lewy pathology to memory dysfunction in the disease. Levels of Lewy pathology were found to be highest in the hippocampal CA2 subregion and entorhinal cortex, implicating a potentially overlooked circuit in disease pathogenesis. However, correlation with memory performance was strongest with CA1. This unexpected finding suggests that Lewy pathology must reach a critical burden across hippocampal circuitry to contribute to memory dysfunction beyond that related to other factors, notably coexisting Alzheimer's disease tau pathology.
Collapse
|
34
|
Saxena K, Morris RGM. Social memory goes viral. Science 2016; 353:1496-1497. [DOI: 10.1126/science.aai7788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
A precisely mapped brain network discriminates and remembers friends and strangers
Collapse
Affiliation(s)
- Kapil Saxena
- Centre for Brain Development and Repair, INSTEM, Bangalore 560065, India
- Patrick Wilde Centre and Centre for Cognitive and Neural Systems, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Richard G. M. Morris
- Centre for Brain Development and Repair, INSTEM, Bangalore 560065, India
- Patrick Wilde Centre and Centre for Cognitive and Neural Systems, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| |
Collapse
|
35
|
Okuyama T, Kitamura T, Roy DS, Itohara S, Tonegawa S. Ventral CA1 neurons store social memory. Science 2016; 353:1536-1541. [PMID: 27708103 PMCID: PMC5493325 DOI: 10.1126/science.aaf7003] [Citation(s) in RCA: 457] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 07/29/2016] [Indexed: 01/09/2023]
Abstract
The medial temporal lobe, including the hippocampus, has been implicated in social memory. However, it remains unknown which parts of these brain regions and their circuits hold social memory. Here, we show that ventral hippocampal CA1 (vCA1) neurons of a mouse and their projections to nucleus accumbens (NAc) shell play a necessary and sufficient role in social memory. Both the proportion of activated vCA1 cells and the strength and stability of the responding cells are greater in response to a familiar mouse than to a previously unencountered mouse. Optogenetic reactivation of vCA1 neurons that respond to the familiar mouse enabled memory retrieval and the association of these neurons with unconditioned stimuli. Thus, vCA1 neurons and their NAc shell projections are a component of the storage site of social memory.
Collapse
Affiliation(s)
- Teruhiro Okuyama
- RIKEN-MIT Center for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - Takashi Kitamura
- RIKEN-MIT Center for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - Dheeraj S Roy
- RIKEN-MIT Center for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | | | - Susumu Tonegawa
- RIKEN-MIT Center for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA. Brain Science Institute, RIKEN, Saitama, 351-0198, Japan. Howard Hughes Medical Institute at MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
36
|
Matsumoto N, Okamoto K, Takagi Y, Ikegaya Y. 3-Hz subthreshold oscillations of CA2 neurons In vivo. Hippocampus 2016; 26:1570-1578. [PMID: 27650674 DOI: 10.1002/hipo.22657] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2016] [Indexed: 12/12/2022]
Abstract
The CA2 region is unique in the hippocampus; it receives direct synaptic innervations from several hypothalamic nuclei and expresses various receptors of neuromodulators, including adenosine, vasopressin, and oxytocin. Furthermore, the CA2 region may have distinct brain functions, such as the control of instinctive and social behaviors; however, little is known about the dynamics of the subthreshold membrane potentials of CA2 neurons in vivo. We conducted whole-cell current-clamp recordings from CA2 pyramidal cells in urethane-anesthetized mice and monitored the intrinsic fluctuations in their membrane potentials. The CA2 pyramidal cells emitted spontaneous action potentials at mean firing rates of ∼0.8 Hz. In approximately half of the neurons, the subthreshold membrane potential oscillated at ∼3 Hz. In two neurons, we obtained simultaneous recordings of local field potentials from the CA1 stratum radiatum and demonstrated that the 3-Hz oscillations of CA2 neurons were not correlated with CA1 field potentials. In tetrodotoxin-perfused acute hippocampal slices, the membrane potentials of CA2 pyramidal cells were not preferentially entrained to 3-Hz sinusoidal current inputs, which suggest that intracellular 3-Hz oscillations reflect the neuronal dynamics of the surrounding networks. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nobuyoshi Matsumoto
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kazuki Okamoto
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yuki Takagi
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.,Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita City, Osaka, Japan
| |
Collapse
|
37
|
Abstract
Hippocampal area CA2 has several features that distinguish it from CA1 and CA3, including a unique gene expression profile, failure to display long-term potentiation and relative resistance to cell death. A recent increase in interest in the CA2 region, combined with the development of new methods to define and manipulate its neurons, has led to some exciting new discoveries on the properties of CA2 neurons and their role in behaviour. Here, we review these findings and call attention to the idea that the definition of area CA2 ought to be revised in light of gene expression data.
Collapse
|
38
|
Kay K, Sosa M, Chung JE, Karlsson MP, Larkin MC, Frank LM. A hippocampal network for spatial coding during immobility and sleep. Nature 2016; 531:185-90. [PMID: 26934224 PMCID: PMC5037107 DOI: 10.1038/nature17144] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 01/13/2016] [Indexed: 12/11/2022]
Abstract
How does an animal know where it is when it stops moving? Hippocampal place cells fire at discrete locations as subjects traverse space, thereby providing an explicit neural code for current location during locomotion. In contrast, during awake immobility, the hippocampus is thought to be dominated by neural firing representing past and possible future experience. The question of whether and how the hippocampus constructs a representation of current location in the absence of locomotion has stood unresolved. Here we report that a distinct population of hippocampal neurons, located in the CA2 subregion, signals current location during immobility, and furthermore does so in association with a previously unidentified hippocampus-wide network pattern. In addition, signaling of location persists into brief periods of desynchronization prevalent in slow-wave sleep. The hippocampus thus generates a distinct representation of current location during immobility, pointing to mnemonic processing specific to experience occurring in the absence of locomotion.
Collapse
Affiliation(s)
- Kenneth Kay
- UCSF Center for Integrative Neuroscience and Department of Physiology, University of California San Francisco, California 94158, USA
| | - Marielena Sosa
- UCSF Center for Integrative Neuroscience and Department of Physiology, University of California San Francisco, California 94158, USA
| | - Jason E Chung
- UCSF Center for Integrative Neuroscience and Department of Physiology, University of California San Francisco, California 94158, USA
| | - Mattias P Karlsson
- UCSF Center for Integrative Neuroscience and Department of Physiology, University of California San Francisco, California 94158, USA
| | - Margaret C Larkin
- UCSF Center for Integrative Neuroscience and Department of Physiology, University of California San Francisco, California 94158, USA
| | - Loren M Frank
- UCSF Center for Integrative Neuroscience and Department of Physiology, University of California San Francisco, California 94158, USA.,Howard Hughes Medical Institute, University of California San Francisco, California 94158, USA
| |
Collapse
|
39
|
Faraji J, Soltanpour N, Moeeini R, Hosseini SA, Pakdel S, Moharrerie A, Arjang K, Soltanpour N, Metz GA. Regional vulnerability of the hippocampus to repeated motor activity deprivation. Behav Brain Res 2016; 301:178-89. [DOI: 10.1016/j.bbr.2015.12.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/11/2015] [Accepted: 12/18/2015] [Indexed: 12/12/2022]
|
40
|
Neurons in Vulnerable Regions of the Alzheimer's Disease Brain Display Reduced ATM Signaling. eNeuro 2016; 3:eN-NWR-0124-15. [PMID: 27022623 PMCID: PMC4770009 DOI: 10.1523/eneuro.0124-15.2016] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 02/03/2016] [Accepted: 02/06/2016] [Indexed: 01/30/2023] Open
Abstract
Ataxia telangiectasia (A-T) is a multisystemic disease caused by mutations in the ATM (A-T mutated) gene. It strikes before 5 years of age and leads to dysfunctions in many tissues, including the CNS, where it leads to neurodegeneration, primarily in cerebellum. Alzheimer's disease (AD), by contrast, is a largely sporadic neurodegenerative disorder that rarely strikes before the 7th decade of life with primary neuronal losses in hippocampus, frontal cortex, and certain subcortical nuclei. Despite these differences, we present data supporting the hypothesis that a failure of ATM signaling is involved in the neuronal death in individuals with AD. In both, partially ATM-deficient mice and AD mouse models, neurons show evidence for a loss of ATM. In human AD, three independent indices of reduced ATM function-nuclear translocation of histone deacetylase 4, trimethylation of histone H3, and the presence of cell cycle activity-appear coordinately in neurons in regions where degeneration is prevalent. These same neurons also show reduced ATM protein levels. And though they represent only a fraction of the total neurons in each affected region, their numbers significantly correlate with disease stage. This previously unknown role for the ATM kinase in AD pathogenesis suggests that the failure of ATM function may be an important contributor to the death of neurons in AD individuals.
Collapse
|
41
|
Alexander GM, Farris S, Pirone JR, Zheng C, Colgin LL, Dudek SM. Social and novel contexts modify hippocampal CA2 representations of space. Nat Commun 2016; 7:10300. [PMID: 26806606 PMCID: PMC4737730 DOI: 10.1038/ncomms10300] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 11/27/2015] [Indexed: 01/01/2023] Open
Abstract
The hippocampus supports a cognitive map of space and is critical for encoding declarative memory (who, what, when and where). Recent studies have implicated hippocampal subfield CA2 in social and contextual memory but how it does so remains unknown. Here we find that in adult male rats, presentation of a social stimulus (novel or familiar rat) or a novel object induces global remapping of place fields in CA2 with no effect on neuronal firing rate or immediate early gene expression. This remapping did not occur in CA1, suggesting this effect is specific for CA2. Thus, modification of existing spatial representations might be a potential mechanism by which CA2 encodes social and novel contextual information.
Collapse
Affiliation(s)
- Georgia M. Alexander
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Drive, Mail Drop F2-04, Research Triangle Park, North Carolina 27709, USA
| | - Shannon Farris
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Drive, Mail Drop F2-04, Research Triangle Park, North Carolina 27709, USA
| | - Jason R. Pirone
- Social and Scientific Systems, Inc., 1009 Slater Road Suite 120, Durham, North Carolina 27703, USA
| | - Chenguang Zheng
- Center for Learning and Memory, The University of Texas at Austin, 1 University Station Stop C7000, NMS 4.104, Austin, Texas 78712, USA
- Department of Neuroscience, The University of Texas at Austin, Austin, Texas 78712, USA
| | - Laura L. Colgin
- Center for Learning and Memory, The University of Texas at Austin, 1 University Station Stop C7000, NMS 4.104, Austin, Texas 78712, USA
- Department of Neuroscience, The University of Texas at Austin, Austin, Texas 78712, USA
| | - Serena M. Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Drive, Mail Drop F2-04, Research Triangle Park, North Carolina 27709, USA
| |
Collapse
|
42
|
Prabsattroo T, Wattanathorn J, Somsapt P, Sritragool O. Positive Modulation of Pink Nelumbo nucifera Flowers on Memory Impairment, Brain Damage, and Biochemical Profiles in Restraint Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:5789857. [PMID: 26858824 PMCID: PMC4686682 DOI: 10.1155/2016/5789857] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/30/2015] [Accepted: 08/31/2015] [Indexed: 12/29/2022]
Abstract
Due to the crucial role of oxidative stress in the stress-induced memory deficit, the benefit of substance possessing antioxidant effect is focused. Since no data are available, we aimed to determine the effect of Nelumbo nucifera flowers extract on spatial memory and hippocampal damage in stressed rats. Male Wistar rats, weighing 250-350 g, were orally given N. nucifera extract at doses of 10, 10, and 200 mg·kg(-1) 45 minutes before the exposure to 12-hour restraint stress. The spatial memory and serum corticosterone were assessed at 7 and 14 days of study period. At the end of study, acetylcholinesterase (AChE), monoamine oxidase type A and monoamine oxidase type B (MAO-A and MAO-B), oxidative stress status, neuron density, and Ki67 expression in hippocampus were also assessed. The results showed that N. nucifera extract decreased memory deficit and brain damage, serum corticosterone, oxidative stress status, AChE, and MAO-A and MAO-B activities but increased neuron density and Ki67 expression in hippocampus. These suggested that the improved oxidative stress status, adult neurogenesis, and cholinergic and monoaminergic functions might be responsible for the protective effect against stress-related brain damage and dysfunction of the extract. Therefore, N. nucifera extract is the potential neuroprotective and memory enhancing agent. However, further researches are still required.
Collapse
Affiliation(s)
- Thawatchai Prabsattroo
- Department of Physiology and Graduate School (Neuroscience Program), Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- Integrative Complementary and Alternative Medicine Research and Development Center, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Jintanaporn Wattanathorn
- Integrative Complementary and Alternative Medicine Research and Development Center, Khon Kaen University, Khon Kaen 40002, Thailand
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Pichet Somsapt
- Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40000, Thailand
| | - Opass Sritragool
- Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40000, Thailand
| |
Collapse
|
43
|
Basu J, Siegelbaum SA. The Corticohippocampal Circuit, Synaptic Plasticity, and Memory. Cold Spring Harb Perspect Biol 2015; 7:7/11/a021733. [PMID: 26525152 DOI: 10.1101/cshperspect.a021733] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Synaptic plasticity serves as a cellular substrate for information storage in the central nervous system. The entorhinal cortex (EC) and hippocampus are interconnected brain areas supporting basic cognitive functions important for the formation and retrieval of declarative memories. Here, we discuss how information flow in the EC-hippocampal loop is organized through circuit design. We highlight recently identified corticohippocampal and intrahippocampal connections and how these long-range and local microcircuits contribute to learning. This review also describes various forms of activity-dependent mechanisms that change the strength of corticohippocampal synaptic transmission. A key point to emerge from these studies is that patterned activity and interaction of coincident inputs gives rise to associational plasticity and long-term regulation of information flow. Finally, we offer insights about how learning-related synaptic plasticity within the corticohippocampal circuit during sensory experiences may enable adaptive behaviors for encoding spatial, episodic, social, and contextual memories.
Collapse
Affiliation(s)
- Jayeeta Basu
- Department of Neuroscience and Physiology, NYU Neuroscience Institute, New York University School of Medicine, New York, New York 10016
| | - Steven A Siegelbaum
- Kavli Institute for Brain Science, Columbia University, New York, New York 10032 Department of Neuroscience, Columbia University, New York, New York 10032 Department of Pharmacology, Columbia University, New York, New York 10032
| |
Collapse
|
44
|
Knierim JJ, Neunuebel JP. Tracking the flow of hippocampal computation: Pattern separation, pattern completion, and attractor dynamics. Neurobiol Learn Mem 2015; 129:38-49. [PMID: 26514299 DOI: 10.1016/j.nlm.2015.10.008] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 10/04/2015] [Accepted: 10/21/2015] [Indexed: 10/22/2022]
Abstract
Classic computational theories of the mnemonic functions of the hippocampus ascribe the processes of pattern separation to the dentate gyrus (DG) and pattern completion to the CA3 region. Until the last decade, the large majority of single-unit studies of the hippocampus in behaving animals were from the CA1 region. The lack of data from the DG, CA3, and the entorhinal inputs to the hippocampus severely hampered the ability to test these theories with neurophysiological techniques. The past ten years have seen a major increase in the recordings from the CA3 region and the medial entorhinal cortex (MEC), with an increasing (but still limited) number of experiments from the lateral entorhinal cortex (LEC) and DG. This paper reviews a series of studies in a local-global cue mismatch (double-rotation) experiment in which recordings were made from cells in the anterior thalamus, MEC, LEC, DG, CA3, and CA1 regions. Compared to the standard cue environment, the change in the DG representation of the cue-mismatch environment was greater than the changes in its entorhinal inputs, providing support for the theory of pattern separation in the DG. In contrast, the change in the CA3 representation of the cue-mismatch environment was less than the changes in its entorhinal and DG inputs, providing support for a pattern completion/error correction function of CA3. The results are interpreted in terms of continuous attractor network models of the hippocampus and the relationship of these models to pattern separation and pattern completion theories. Whereas DG may perform an automatic pattern separation function, the attractor dynamics of CA3 allow it to perform a pattern separation or pattern completion function, depending on the nature of its inputs and the relative strength of the internal attractor dynamics.
Collapse
Affiliation(s)
- James J Knierim
- Krieger Mind/Brain Institute and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, United States.
| | - Joshua P Neunuebel
- Dept. of Psychological and Brain Sciences, University of Delaware, United States
| |
Collapse
|
45
|
Lee H, Wang C, Deshmukh SS, Knierim JJ. Neural Population Evidence of Functional Heterogeneity along the CA3 Transverse Axis: Pattern Completion versus Pattern Separation. Neuron 2015; 87:1093-105. [PMID: 26298276 DOI: 10.1016/j.neuron.2015.07.012] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 06/25/2015] [Accepted: 07/17/2015] [Indexed: 10/23/2022]
Abstract
Classical theories of associative memory model CA3 as a homogeneous attractor network because of its strong recurrent circuitry. However, anatomical gradients suggest a functional diversity along the CA3 transverse axis. We examined the neural population coherence along this axis, when the local and global spatial reference frames were put in conflict with each other. Proximal CA3 (near the dentate gyrus), where the recurrent collaterals are the weakest, showed degraded representations, similar to the pattern separation shown by the dentate gyrus. Distal CA3 (near CA2), where the recurrent collaterals are the strongest, maintained coherent representations in the conflict situation, resembling the classic attractor network system. CA2 also maintained coherent representations. This dissociation between proximal and distal CA3 provides strong evidence that the recurrent collateral system underlies the associative network functions of CA3, with a separate role of proximal CA3 in pattern separation.
Collapse
Affiliation(s)
- Heekyung Lee
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Cheng Wang
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sachin S Deshmukh
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA
| | - James J Knierim
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
46
|
Mankin EA, Diehl GW, Sparks FT, Leutgeb S, Leutgeb JK. Hippocampal CA2 activity patterns change over time to a larger extent than between spatial contexts. Neuron 2015; 85:190-201. [PMID: 25569350 DOI: 10.1016/j.neuron.2014.12.001] [Citation(s) in RCA: 200] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2014] [Indexed: 11/24/2022]
Abstract
The hippocampal CA2 subregion has a different anatomical connectivity pattern within the entorhino-hippocampal circuit than either the CA1 or CA3 subregion. Yet major differences in the neuronal activity patterns of CA2 compared with the other CA subregions have not been reported. We show that standard spatial and temporal firing patterns of individual hippocampal principal neurons in behaving rats, such as place fields, theta modulation, and phase precession, are also present in CA2, but that the CA2 subregion differs substantially from the other CA subregions in its population coding. CA2 ensembles do not show a persistent code for space or for differences in context. Rather, CA2 activity patterns become progressively dissimilar over time periods of hours to days. The weak coding for a particular context is consistent with recent behavioral evidence that CA2 circuits preferentially support social, emotional, and temporal rather than spatial aspects of memory.
Collapse
Affiliation(s)
- Emily A Mankin
- Neurobiology Section and Center for Neural Circuits and Behavior, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Geoffrey W Diehl
- Neurobiology Section and Center for Neural Circuits and Behavior, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Fraser T Sparks
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Stefan Leutgeb
- Neurobiology Section and Center for Neural Circuits and Behavior, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jill K Leutgeb
- Neurobiology Section and Center for Neural Circuits and Behavior, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
47
|
Evans PR, Dudek SM, Hepler JR. Regulator of G Protein Signaling 14: A Molecular Brake on Synaptic Plasticity Linked to Learning and Memory. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 133:169-206. [PMID: 26123307 DOI: 10.1016/bs.pmbts.2015.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The regulators of G protein signaling (RGS) proteins are a diverse family of proteins that function as central components of G protein and other signaling pathways. In the brain, regulator of G protein signaling 14 (RGS14) is enriched in neurons in the hippocampus where the mRNA and protein are highly expressed. This brain region plays a major role in processing learning and forming new memories. RGS14 is an unusual RGS protein that acts as a multifunctional scaffolding protein to integrate signaling events and pathways essential for synaptic plasticity, including conventional and unconventional G protein signaling, mitogen-activated protein kinase, and, possibly, calcium signaling pathways. Within the hippocampus of primates and rodents, RGS14 is predominantly found in the enigmatic CA2 subfield. Principal neurons within the CA2 subfield differ from neighboring hippocampal regions in that they lack a capacity for long-term potentiation (LTP) of synaptic transmission, which is widely viewed as the cellular substrate of learning and memory formation. RGS14 was recently identified as a natural suppressor of LTP in hippocampal CA2 neurons as well as forms of learning and memory that depend on the hippocampus. Although CA2 has only recently been studied, compelling recent evidence implicates area CA2 as a critical component of hippocampus circuitry with functional roles in mediating certain types of learning and memory. This review will highlight the known functions of RGS14 in cell signaling and hippocampus physiology, and discuss potential roles for RGS14 in human cognition and disease.
Collapse
Affiliation(s)
- Paul R Evans
- Department of Pharmacology, Emory University School of Medicine, Rollins Research Center, Atlanta, Georgia, USA
| | - Serena M Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - John R Hepler
- Department of Pharmacology, Emory University School of Medicine, Rollins Research Center, Atlanta, Georgia, USA.
| |
Collapse
|
48
|
Hunting increases phosphorylation of calcium/calmodulin-dependent protein kinase type II in adult barn owls. Neural Plast 2015; 2015:819257. [PMID: 25789177 PMCID: PMC4348593 DOI: 10.1155/2015/819257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/18/2014] [Indexed: 11/18/2022] Open
Abstract
Juvenile barn owls readily adapt to prismatic spectacles, whereas adult owls living under standard aviary conditions do not. We previously demonstrated that phosphorylation of the cyclic-AMP response element-binding protein (CREB) provides a readout of the instructive signals that guide plasticity in juveniles. Here we investigated phosphorylation of calcium/calmodulin-dependent protein kinase II (pCaMKII) in both juveniles and adults. In contrast to CREB, we found no differences in pCaMKII expression between prism-wearing and control juveniles within the external nucleus of the inferior colliculus (ICX), the major site of plasticity. For prism-wearing adults that hunted live mice and are capable of adaptation, expression of pCaMKII was increased relative to prism-wearing adults that fed passively on dead mice and are not capable of adaptation. This effect did not bear the hallmarks of instructive information: it was not localized to rostral ICX and did not exhibit a patchy distribution reflecting discrete bimodal stimuli. These data are consistent with a role for CaMKII as a permissive rather than an instructive factor. In addition, the paucity of pCaMKII expression in passively fed adults suggests that the permissive default setting is "off" in adults.
Collapse
|
49
|
Bester-Meredith JK, Fancher AP, Mammarella GE. Vasopressin Proves Es-sense-tial: Vasopressin and the Modulation of Sensory Processing in Mammals. Front Endocrinol (Lausanne) 2015; 6:5. [PMID: 25705203 PMCID: PMC4319160 DOI: 10.3389/fendo.2015.00005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 01/12/2015] [Indexed: 11/29/2022] Open
Abstract
As mammals develop, they encounter increasing social complexity in the surrounding world. In order to survive, mammals must show appropriate behaviors toward their mates, offspring, and same-sex conspecifics. Although the behavioral effects of the neuropeptide arginine vasopressin (AVP) have been studied in a variety of social contexts, the effects of this neuropeptide on multimodal sensory processing have received less attention. AVP is widely distributed through sensory regions of the brain and has been demonstrated to modulate olfactory, auditory, gustatory, and visual processing. Here, we review the evidence linking AVP to the processing of social stimuli in sensory regions of the brain and explore how sensory processing can shape behavioral responses to these stimuli. In addition, we address the interplay between hormonal and neural AVP in regulating sensory processing of social cues. Because AVP pathways show plasticity during development, early life experiences may shape life-long processing of sensory information. Furthermore, disorders of social behavior such as autism and schizophrenia that have been linked with AVP also have been linked with dysfunctions in sensory processing. Together, these studies suggest that AVP's diversity of effects on social behavior across a variety of mammalian species may result from the effects of this neuropeptide on sensory processing.
Collapse
Affiliation(s)
- Janet K. Bester-Meredith
- Department of Biology, Seattle Pacific University, Seattle, WA, USA
- *Correspondence: Janet K. Bester-Meredith, Department of Biology, Seattle Pacific University, 3307 3rd Avenue W, Seattle, WA 98119, USA e-mail:
| | | | | |
Collapse
|
50
|
Zhang Y, Brownstein AJ, Buonora M, Niikura K, Ho A, Correa da Rosa J, Kreek MJ, Ott J. Self administration of oxycodone alters synaptic plasticity gene expression in the hippocampus differentially in male adolescent and adult mice. Neuroscience 2014; 285:34-46. [PMID: 25446355 DOI: 10.1016/j.neuroscience.2014.11.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/19/2014] [Accepted: 11/04/2014] [Indexed: 11/16/2022]
Abstract
Abuse and addiction to prescription opioids such as oxycodone (a short-acting Mu opioid receptor (MOP-r) agonist) in adolescence is a pressing public health issue. We have previously shown differences in oxycodone self-administration behaviors between adolescent and adult C57BL/6J mice and expression of striatal neurotransmitter receptor genes, in areas involved in reward. In this study, we aimed to determine whether oxycodone self-administration differentially affects genes regulating synaptic plasticity in the hippocampus of adolescent compared to adult mice, since the hippocampus may be involved in learning aspects associated with chronic drug self administration. Hippocampus was isolated for mRNA analysis from mice that had self administered oxycodone (0.25 mg/kg/infusion) 2h/day for 14 consecutive days or from yoked saline controls. Gene expression was analyzed with real-time polymerase chain reaction (PCR) using a commercially available "synaptic plasticity" PCR array containing 84 genes. We found that adolescent and adult control mice significantly differed in the expression of several genes in the absence of oxycodone exposure, including those coding for mitogen-activated protein kinase, calcium/calmodulin-dependent protein kinase II gamma subunit, glutamate receptor, ionotropic AMPA2 and metabotropic 5. Chronic oxycodone self administration increased proviral integration site 1 (Pim1) and thymoma viral proto-oncogene 1 mRNA levels compared to controls in both age groups. Both Pim1 and cadherin 2 mRNAs showed a significant combined effect of Drug Condition and Age × Drug Condition. Furthermore, the mRNA levels of both cadherin 2 and cAMP response element modulators showed an experiment-wise significant difference between oxycodone and saline control in adult but not in adolescent mice. Overall, this study demonstrates for the first time that chronic oxycodone self-administration differentially alters synaptic plasticity gene expression in the hippocampus of adolescent and adult mice.
Collapse
Affiliation(s)
- Y Zhang
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA.
| | - A J Brownstein
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - M Buonora
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - K Niikura
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - A Ho
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - J Correa da Rosa
- Laboratory of Investigative Dermatology, The Rockefeller University, New York, NY 10065, USA
| | - M J Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - J Ott
- Institute of Psychology, Chinese Academy of Sciences, Beijing, China; The Laboratory of Statistical Genetics, The Rockefeller University, New York, NY, USA
| |
Collapse
|